1
|
Lin YT, Hong ZJ, Liao GS, Dai MS, Chao TK, Tsai WC, Sung YK, Chiu CH, Chang CK, Yu JC. Unexpected contralateral axillary lymph node metastasis without ipsilateral involvement in triple-negative breast cancer: A case report and review of literature. World J Clin Cases 2025; 13:103571. [DOI: 10.12998/wjcc.v13.i18.103571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Breast cancer is a leading cause of cancer-related mortality among women worldwide, with invasive ductal carcinoma (IDC) being the most prevalent subtype. Lymph node metastasis is the primary prognostic indicator, typically evaluated via biopsy of the ipsilateral sentinel or axillary lymph nodes. Contralateral axillary metastasis (CAM) without ipsilateral involvement is exceedingly rare, particularly in early-stage breast cancer. This report presents a case of CAM in a patient with triple-negative breast cancer (TNBC), underscoring diagnostic and therapeutic complexities.
CASE SUMMARY A 73-year-old female presented with left-sided early-stage IDC in February 2023. Despite a modified radical mastectomy and pathologically negative ipsilateral lymph nodes, a postoperative positron emission tomography (PET) scan detected fluorodeoxyglucose-avid nodes in the contralateral axilla. Biopsy confirmed metastatic ductal carcinoma with triple-negative status, resulting in an upstaged diagnosis of metastatic breast cancer, stage IV, M1. The patient underwent six cycles of adjuvant chemotherapy, with follow-up PET imaging showing regression of the contralateral lesion. This case highlights the importance of advanced imaging in TNBC for precise staging and treatment optimization.
CONCLUSION This case highlights the aggressive nature of TNBC and the need for advanced imaging to ensure accurate staging and effective management.
Collapse
Affiliation(s)
- Yun-Ting Lin
- Department of General Medicine, Tri Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Zhi-Jie Hong
- Division of Traumatology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Guo-Shiou Liao
- Division of General Surgery, Department of Surgery, Tri Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ming-Shen Dai
- Division of Hematology/Oncology, Department of Internal Medicine, National Defense Medical Center, Taipei 114, Taiwan
| | - Tai-Kuang Chao
- Department of Pathology and Parasitology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Wen-Chiuan Tsai
- Department of Pathology, National Defense Medical Center, Taipei 114, Taiwan
| | - Yu-Kai Sung
- Department of Pathology and Parasitology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chuang-Hsin Chiu
- Department of Nuclear Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Cheng-Kuang Chang
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Jyh-Cherng Yu
- Division of General Surgery, Department of Surgery, Tri Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
2
|
Altundag-Erdogan Ö, Çelebi-Saltik B. Niclosamide Treatment Suppressed Metastatic, Apoptotic, and Proliferative Characteristics of MDA-MB-231 Cancer Stem Cells. ACS OMEGA 2025; 10:23629-23638. [PMID: 40521512 PMCID: PMC12163758 DOI: 10.1021/acsomega.5c02545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025]
Abstract
This study evaluated the efficacy of niclosamide after the invasion of aggressive TNBC breast CSCs into a 3D bone-mimicking model. Initially, the optional dose required for triggering apoptosis was determined for MDA-MB-231 CSCs (CD44+ and CD24-). Our findings revealed that approximately 50% of the cells showed apoptotic properties, as assessed with Annexin V/7AAD assay and WST-1 at IC50 = 100 μM (6 h). Additionally, this treatment suppressed p-STAT3 protein levels and increased Bax levels (p < 0.05), as determined by Western Blot. The expression of genes associated with metastasis and cell migration (CXCR4, MMP2, MMP9), drug resistance (ABCG1, ABCG2), stemness (OCT4, NANOG) and cell cycle and proliferation (CYCLIN D1) was found to be significantly suppressed (p < 0.05). Therefore, after validating the efficacy of the 100 μM dose on CSCs, cell cycle, ELISA, Western Blot, and RT-qPCR analyses were conducted in the 3D model. It was found that the cells were arrested in the G0-G1 phase (p < 0.05). 100 μM Niclosamide suppressed the levels of EMT markers, Vimentin (p > 0.05) and ZEB1 (p < 0.05). Additionally, RT-qPCR results indicated a significant downregulation of CXCR4, ABCG1, ABCG2, MMP2, OCT4, CCND1, AXIN2, and LGR5 gene expressions following niclosamide treatment in both CD133+ and CD133- groups (p < 0.05). The increase in the Bax protein, a key player in apoptosis induction, along with the decrease in the anti-apoptotic protein Bcl-2, suggests the activation of cell death mechanisms. Notably, its targeted impact on the CD44+/CD24- population suggests that niclosamide could enhance the sensitivity of CSCs to treatment, thereby preventing tumor recurrence.
Collapse
Affiliation(s)
- Özlem Altundag-Erdogan
- Hacettepe
University, Graduate School of Health Sciences
Department of Stem Cell Sciences, Sihhiye, Ankara, TR06100, Turkey
- Hacettepe
University, Center for Stem Cell Research
and Development, Sihhiye, Ankara, TR06100, Turkey
| | - Betül Çelebi-Saltik
- Hacettepe
University, Graduate School of Health Sciences
Department of Stem Cell Sciences, Sihhiye, Ankara, TR06100, Turkey
- Hacettepe
University, Center for Stem Cell Research
and Development, Sihhiye, Ankara, TR06100, Turkey
| |
Collapse
|
3
|
An Y, Lan J, Tang J, Luo N. PTPN2 inhibits TG-induced ERS-initiated TNBC apoptosis through the mitochondrial pathway. Sci Rep 2025; 15:19896. [PMID: 40481083 PMCID: PMC12144175 DOI: 10.1038/s41598-025-04312-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Triple negative breast cancer (TNBC) is the most malignant subtype of breast cancer that portends a poor prognosis and limited treatment. PTPN2 is a member of the non-receptor protein tyrosine phosphatase family that regulates biological processes by dephosphorylating various signaling molecules. Endoplasmic reticulum stress (ERS) plays a dual regulatory role by promoting both survival and apoptosis. This study aims to elucidate the role of PTPN2 in mediating the pro-apoptotic effects of ERS induced by Thapsigargin (TG), and its influence on the fate of TNBC cells, utilizing both loss-of-function and gain-of-function methodologies. Our findings indicate that PTPN2 modulates TG-induced ERS via the IRE1-XBP1 and PERK/EIF2α/ATF-4 signaling pathways. Furthermore, PTPN2 mitigates the TG-induced reduction in cell proliferation and the concomitant increase in apoptosis. Specifically, PTPN2 appears to inhibit several facets of TG-induced apoptosis, including: (1) Ca2+ elevation in mitochondria, (2) the production of reactive oxygen species (ROS), and (3) Bax/Bcl-2 augmentation which dictates mitochondria-mediated apoptosis. Additionally, we observed that the knockdown of PTPN2 enhances TG-induced autophagy; however, our results suggest that autophagy may serve a protective role against TG-induced apoptosis. Consequently, targeting PTPN2 in conjunction with ERS-inducing agents may represent a promising therapeutic strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Yanhe An
- Department of Anatomy and Histology, School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jinxin Lan
- Department of Anatomy and Histology, School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jiaping Tang
- Department of Anatomy and Histology, School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Na Luo
- Department of Anatomy and Histology, School of Medicine, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
4
|
Liu L, Lin J, Hu W, Zhao T, Fu L, Ding H. Clinicopathological and Ultrasonographic Characteristics of Breast Cancer in Young Women. J Adolesc Young Adult Oncol 2025; 14:238-243. [PMID: 39229762 DOI: 10.1089/jayao.2024.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
Purpose: The early detection of breast cancer in women under the age of 40 has posed significant challenges. This can be attributed in part to the limited research conducted on the breast cancer in this age group, particularly with regards to large sample sizes. We aimed to address this gap by analyzing and comparing the ultrasound imaging and pathological characteristics of breast cancer in women aged under 40 and those aged 40 and above. Methods: A retrospective assessment was conducted to examine the ultrasound imaging and clinicopathologic characteristics of 555 women with surgically confirmed breast cancers. The patient cohort consisted of 160 individuals below the age of 40 and 395 individuals aged 40 years and above. Results: Our study identified the breast cancer in patients under 40 years was more likely to show regular shape (p = 0.043) compared with tumors in patients who were 40 years and over. Furthermore, in young female patients (<40 years), irregular shape was correlated with the HER2-enriched type (p = 0.02), circumscribed margin (p = 0.001), and a lack of calcifications (p = 0.02) were associated with the triple-negative type. In another group (≥40 years), only a lack of calcifications (p = 0.003) were associated with the triple-negative type. Conclusion: Breast cancer in women under the age of 40 exhibits distinct ultrasonographic characteristics patterns that vary across different immunophenotypes, which may provide certain predictive information for physicians.
Collapse
Affiliation(s)
- Lu Liu
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Lin
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenjie Hu
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Ting Zhao
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | - Lina Fu
- Department of Ultrasound, Huashan Hospital, Fudan University, Shanghai, China
| | | |
Collapse
|
5
|
Márquez-Mendoza JM, Baranda-Ávila N, Lizano M, Langley E. Micro-RNAs targeting the estrogen receptor alpha involved in endocrine therapy resistance in breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167783. [PMID: 40057206 DOI: 10.1016/j.bbadis.2025.167783] [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: 09/13/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Endocrine therapy resistance (ETR) in breast cancer (BC) is a multicausal phenomenon with diverse alterations in the tumor cell interactome. Within these alterations, non-coding RNAs (ncRNAs) such as micro-RNAs (miRNAs) modulate the expression of tumor suppressor genes and proto-oncogenes, such as the ESR1 gene encoding estrogen receptor alpha (ERα). This work aims to review the effects of miRNAs targeting ERα mRNA and their mechanisms related to ETR in BC. A thorough review of the literature and an in silico study were carried out to elucidate the involvement of each miRNA, thus contributing to the understanding of ETR in BC.
Collapse
Affiliation(s)
- J M Márquez-Mendoza
- Programa de Doctorado en Ciencias Biomédicas, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - N Baranda-Ávila
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico
| | - M Lizano
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City 04510, Mexico
| | - E Langley
- Unidad de Investigación Biomédica en Cáncer, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico.
| |
Collapse
|
6
|
Chisaki Y, Nakamura N, Komuro T, Nyuji H, Harano M, Kitada N. Cost-Effectiveness Analysis of Pembrolizumab Plus Chemotherapy Compared with Chemotherapy as First-Line Treatment for Advanced PD-L1-Positive Triple-Negative Breast Cancer from a Japanese Healthcare Perspective. Clin Drug Investig 2025; 45:317-326. [PMID: 40317386 DOI: 10.1007/s40261-025-01445-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND AND OBJECTIVES Pembrolizumab has been approved for the immunotherapy of programmed death ligand 1 (PD-L1)-positive triple-negative breast cancer (TNBC) based on the KEYNOTE-355 trial. However, cost-effectiveness evidence is limited. The purpose of this study was to evaluate the cost-effectiveness of pembrolizumab plus chemotherapy compared with chemotherapy alone for patients with PD-L1-positive inoperable or metastatic TNBC from a Japanese healthcare perspective. METHODS The cost-effectiveness analysis was performed for pembrolizumab, of which the drug price was determined at 214,498 Japanese yen (JPY), or 1631 US dollars (USD) (1 USD = 131.5 JPY) for KEYTRUDA® (100 mg), using a partition survival model based on the KEYNOTE-355 trial subgroup analysis in Japan. The comparison was made using quality-adjusted life years (QALYs) and the incremental cost-effectiveness ratio (ICER). One-way deterministic and probabilistic sensitivity analyses (PSA), which evaluate the impact of parameter uncertainty, were performed to assess the robustness and calculate the acceptable probability, defined as the probability of the ICER being below the willingness-to-pay (WTP). RESULTS Pembrolizumab plus chemotherapy provided an additional 0.676 QALYs at an incremental cost of 8,503,072 JPY. The ICER for pembrolizumab plus chemotherapy compared with conventional chemotherapy was 12,577,178 JPY (95,644 USD) per QALY. The ICER per QALY was below the willingness-to-pay threshold of 15,000,000 JPY. PSAs revealed that the acceptable probability was 83.9% at 15,000,000 JPY. CONCLUSIONS The pembrolizumab plus chemotherapy is likely to be a cost-effective option compared with conventional chemotherapy for patients with PD-L1-positive inoperable or metastatic TNBC in a Japanese medical environment from a healthcare system.
Collapse
Affiliation(s)
- Yugo Chisaki
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan.
| | - Nobuhiko Nakamura
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Takako Komuro
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Hirokatsu Nyuji
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Mai Harano
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Noriaki Kitada
- Education and Research Center for Clinical Pharmacy, Kyoto Pharmaceutical University, 5-Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto, 607-8414, Japan
| |
Collapse
|
7
|
Garcia-Murillas I, Abbott CW, Cutts RJ, Boyle SM, Pugh J, Keough KC, Li B, Pyke RM, Navarro FCP, Chen RO, Dunne K, Bunce C, Johnston SRD, Ring A, Russell S, Evans A, Skene A, Smith IE, Turner NC. Whole genome sequencing-powered ctDNA sequencing for breast cancer detection. Ann Oncol 2025; 36:673-681. [PMID: 39914664 DOI: 10.1016/j.annonc.2025.01.021] [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: 09/15/2024] [Revised: 12/20/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Circulating tumour DNA (ctDNA)-based detection of molecular residual disease (MRD) presents a strategy to identify patients at high risk of relapse. In this article, we profile early breast cancer patients with an ultrasensitive, whole genome sequencing (WGS)-based, tumour-informed ctDNA platform. MATERIALS AND METHODS We analysed 617 plasma samples (median 8, range 2-14) from 78 patients (23 triple-negative breast cancer, 35 human epidermal growth factor receptor 2-positive, 18 hormone receptor-positive, and 2 unknown). Samples were collected at diagnosis before therapy, cycle 2 of neoadjuvant chemotherapy, post-surgery after neoad'juvant therapy if administered, every 3 months during the first year, and every 6 months thereafter. Plasma DNA was analysed using the NeXT Personal MRD platform, a tumour-informed WGS approach to produce personalized ctDNA sequencing panels tracking a median of 1451 variants per patient. MRD detection was correlated with clinical outcomes. RESULTS ctDNA was detected at levels ranging from 2.19 parts per million (PPM) to 204 900 PPM (median 405 PPM), with 39% of all ctDNA detections in the ultra-low range <100 PPM. Of patients with samples at diagnosis, 98% (49/50) had ctDNA detected before treatment. At a median follow-up of 76 months (range 5-118 months), detection of ctDNA was associated with high risk of future relapse (P < 0.0001; log-rank test) and shortened overall survival (P < 0.0001) with a median lead time from ctDNA detection to clinical relapse of 15 months (range 0.9-61.5 months). MRD was identified in 100% (11/11) of patients who relapsed, with a median level of ctDNA at first MRD detection of 13.1 PPM. No ctDNA-undetected patients relapsed throughout follow-up (64/64). Comparison with exome-powered MRD detection assays showed improved sensitivity and lead time. CONCLUSIONS A whole genome-powered MRD assay detected breast cancer relapse with a long lead time over clinical relapse, and was strongly associated with relapse-free survival. Rates of ctDNA detection at diagnosis were higher than those reported with exome-based tumour-informed assays.
Collapse
Affiliation(s)
- I Garcia-Murillas
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - R J Cutts
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - J Pugh
- Personalis Inc., Fremont, USA
| | | | - B Li
- Personalis Inc., Fremont, USA
| | | | | | | | - K Dunne
- The Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - C Bunce
- Clinical Trials Unit, Royal Marsden Hospital, London, UK
| | | | - A Ring
- Breast Unit, Royal Marsden Hospital, London, UK
| | - S Russell
- Hinchingbrooke Hospital, Hinchingbrooke Park, Huntingdon, UK
| | - A Evans
- Poole General Hospital, Dorset, UK
| | - A Skene
- Royal Bournemouth Hospital, Bournemouth, UK
| | - I E Smith
- Breast Unit, Royal Marsden Hospital, London, UK
| | - N C Turner
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK; The Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK; Breast Unit, Royal Marsden Hospital, London, UK.
| |
Collapse
|
8
|
Ma Y, Zhu M, Zhang J, Jiao D, Hou Y, Chen X, Liu Z. Efficacy of adjuvant capecitabine in triple-negative breast cancer with residual disease after neoadjuvant therapy: a real-world study. Breast 2025; 81:104477. [PMID: 40250161 PMCID: PMC12036065 DOI: 10.1016/j.breast.2025.104477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 04/08/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE To determine the beneficiaries of capecitabine in patients with triple-negative breast cancer (TNBC) who failed to achieve pathological complete response (pCR) by analyzing the efficacy of the drug in different HER2 statuses and TNM stages. METHODS The Kaplan-Meier survival curve was plotted to estimate the effect of capecitabine therapy on disease-free survival (DFS) and overall survival (OS). Furthermore, the Cox proportional hazards model was used to analyze the factors that influence DFS and OS. RESULTS A total of 296 patients with TNBC who had non-pCR after undergoing neoadjuvant therapy (NAT) were included in this study. There were 152 patients (51.4 %) in the capecitabine group and 144 patients (48.6 %) in the no-capecitabine group. The 3-year DFS and OS rates of the capecitabine group were better than those of the no-capecitabine group (DFS 80.0 % vs. 68.0 % p = 0.012, OS 95.9 % vs. 86.9 % p = 0.011). In addition, the capecitabine group exhibited significantly better DFS and OS than the no-capecitabine group in the HER2-low (DFS p = 0.004, OS p = 0.009) and stage III (DFS p = 0.004, OS p = 0.008) populations but not in the HER2-0 or stage II population. CONCLUSION Adjuvant capecitabine therapy significantly improved the prognosis of patients with TNBC who had residual disease after NAT, and the improvements in the outcomes were significant in patients with HER2-low expression and stage III disease. Other effective treatment methods should be explored for patients with HER2-0 expression or stage II disease.
Collapse
Affiliation(s)
- Youzhao Ma
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Mingda Zhu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Jingyang Zhang
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Dechuang Jiao
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Yangyang Hou
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Xiuchun Chen
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China
| | - Zhenzhen Liu
- Department of Breast Disease, Henan Breast Cancer Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, No.127, Dongming Road, Zhengzhou, 450000, China.
| |
Collapse
|
9
|
Wu Q, Siddharth S, Verma D, Parida S, Sharma D. TRIM29 upregulation contributes to chemoresistance in triple negative breast cancer via modulating S100P-β-catenin axis. Cell Commun Signal 2025; 23:244. [PMID: 40420099 DOI: 10.1186/s12964-025-02233-9] [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: 11/20/2024] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Triple negative breast cancer, an inherently aggressive disease, is further impaired by the limited therapeutic options and chemotherapy-resistance; hence, elucidating the signaling nodes underlying chemotherapy resistance is of major interest. Focusing on the differentially expressed genes in recurrent TNBC, we identified TRIM29, a ubiquitin ligase belonging to TRIM family, as a uniquely enriched protein in chemoresistant TNBC. Here, we demonstrate that chemoresistant TNBC cells are inherently aggressive, exhibiting elevated growth and migration potential compared to chemosensitive cells, and in particular, they possess higher TRIM29 expression whose expression level modulation results in altered chemosensitivity. TRIM29 overexpression reduces chemotherapy response whereas TRIM29 knockout not only increases chemosensitivity but also reduces TNBC tumor growth. Tumor-dissociated cells maintain TRIM29 knockout status as well as exhibit similar functional alterations as chemoresistant TNBC cells. Mechanistically, RNA-sequencing of parental-chemosensitive, chemoresistant-inherently overexpressing TRIM29 and chemoresistant-TRIM29 knockout TNBC cells reveals a unique set of genes (S100P, SERPINB3, SERPINB4, CEACAM5, CEACAM6 and CDH6) showing significant upregulation with the acquisition of chemoresistance and downregulation with the TRIM29 knockout. Furthermore, an enrichment of β-catenin pathway in chemoresistant TNBC cells is observed. We uncovered a functional network where S100P, a metastasis inducing secretory factor, bidirectionally interacts with TRIM29, and modulates the expression of SERPINB3, SERPINB4, CEACAM5, CEACAM6 as well as β-catenin pathway genes. Showing the functional importance, S100P inhibitor reduces the growth and mammosphere formation in chemoresistant TNBC. Moreover, combining β-catenin inhibitor with chemotherapy shows synergistic inhibition of chemoresistant TNBC cells. Indeed, higher expression of TRIM29, S100P and β-catenin associates with reduced recurrence free survival. This work proposes TRIM29 as an important node that modulates a unique gene network in chemoresistant TNBC and whose biological impact is mediated by modulation of S100P and β-catenin.
Collapse
Affiliation(s)
- Qitong Wu
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Sumit Siddharth
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| | - Deepak Verma
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Sheetal Parida
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Dipali Sharma
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| |
Collapse
|
10
|
Zhang H, Gong L, Yu L, Xian C, Ma Z, Wang X, Xia R. Emerging roles of non-coding RNA derived from extracellular vesicles in regulating PD-1/PD-L1 pathway: insights into cancer immunotherapy and clinical applications. Cancer Cell Int 2025; 25:188. [PMID: 40410719 PMCID: PMC12103061 DOI: 10.1186/s12935-025-03809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
Numerous studies have demonstrated that extracellular vesicles (EVs) carry a variety of noncoding RNAs (ncRNAs), which can be taken up by neighboring cells or transported to distant sites via bodily fluids, thereby facilitating intercellular communication and regulating multiple cellular functions. Within the tumor microenvironment, EV-ncRNA, on the one hand, regulate the expression of PD-L1, thereby influencing tumor immune evasion, promoting tumor cell proliferation, and enhancing tumor growth, invasion, and metastasis in vivo. On the other hand, these specific EV-ncRNAs can also modulate the functions of immune cells (such as CD8 + T cells, macrophages, and NK cells) through various molecular mechanisms, inducing an immunosuppressive microenvironment and promoting resistance to anti-PD-1 therapy. Therefore, delving into the molecular mechanisms underlying EV-ncRNA regulation of immune checkpoints presents compelling therapeutic prospects for strategies that selectively target EV-ncRNAs. In this review, we elaborate on the cutting-edge research progress related to EV-ncRNAs in the context of cancer and dissect their pivotal roles in the PD-1/PD-L1 immune checkpoint pathway. We also highlight the promising clinical applications of EV-ncRNAs in anti-PD-1/PD-L1 immunotherapy, bridging basic research with practical clinical applications.
Collapse
Affiliation(s)
- Haixia Zhang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Lianfeng Gong
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Li Yu
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
- Department of Urology, General Hospital of The Yangtze River Shipping, Wuhan, 430010, China
| | - Chenge Xian
- Naidong District People's Hospital, Shannan, 856004, Tibet Autonomous Region, China
| | - Zhaowu Ma
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| | - Xianwang Wang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
- Shannan Maternal and Child Health Hospital, Shannan, 856099, Tibet Autonomous Region, China.
| | - Ruohan Xia
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| |
Collapse
|
11
|
Anand V, El-Dana F, Baran N, Borgman J, Yin Z, Zhao H, Wong ST, Andreeff M, Battula VL. GD3 synthase drives resistance to p53-induced apoptosis in breast cancer by modulating mitochondrial function. Oncogene 2025:10.1038/s41388-025-03432-x. [PMID: 40382494 DOI: 10.1038/s41388-025-03432-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 04/21/2025] [Accepted: 04/29/2025] [Indexed: 05/20/2025]
Abstract
TP53 mutations are common in breast cancer (BC) and are associated with poor prognosis. GD3 synthase (GD3S/ST8SIA1), a gene associated with breast cancer stem cells, is upregulated in tumors with p53 mutations. However, the functional relationship between GD3S and p53 is unknown. Here, we show that GD3S levels are highest in breast tumors with specific p53 mutations. Functional studies revealed that wild-type (WT) p53 inhibits GD3S expression, whereas mutation in p53 enhances GD3S expression by upregulating GD3S promoter activity. Moreover, we found that GD3S inhibits wild-type p53-induced apoptosis in BC cells, while BC cells harboring gain-of-function p53 mutations are dependent on GD3S for their growth. Mechanistic insights indicate that GD3S strengthens mitochondrial function by regulating their oxygen consumption rate and membrane polarity. Our findings demonstrate that specific GOF p53 mutations rely on GD3S to exert their tumor-promoting effects and that GD3S is a novel anti-apoptotic factor in BC cells. Stabilizing WT p53 and reducing mutant p53 levels downregulates GD3S expression, thereby augmenting apoptosis. GD3S overexpression counteracts the cell death triggered by WT p53 stabilization in BC cells, as well as that triggered by p53 knockdown in cells with specific GOF p53 mutations, which suggests that GD3S helps confer apoptosis resistance.
Collapse
Affiliation(s)
- Vivek Anand
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fouad El-Dana
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalia Baran
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Hematology and Central Hematological Laboratory, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jenny Borgman
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zheng Yin
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Weill Cornell Medicine, Houston, TX, USA
| | - Hong Zhao
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Weill Cornell Medicine, Houston, TX, USA
| | - Stephen T Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Neal Cancer Center, Weill Cornell Medicine, Houston, TX, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - V Lokesh Battula
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Internal Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
12
|
Song EJ, Kim WS, Han Y, Lee C, Moon EJ, Kim HJ, Kang NS. Discovery of a transforming growth factor-β1 inhibitory peptide, Charis 1000 to enhance the therapeutic efficacy of paclitaxel in triple-negative breast cancer. Int J Biol Macromol 2025; 314:144216. [PMID: 40379179 DOI: 10.1016/j.ijbiomac.2025.144216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and invasive subtype of breast cancer for which chemotherapy, such as paclitaxel (PTX), remains a primary treatment option. However, resistance to chemotherapy poses a significant challenge, necessitating the development of novel therapeutic strategies. This study aimed to address PTX resistance in TNBC by developing a peptide drug, Charis 1000 (C1K), designed to target transforming growth factor beta (TGF-β) signaling. C1K was synthesized using standard solid-phase peptide synthesis and optimized for enhanced stability. Molecular docking predicted the binding interactions between C1K and TGF-β1, and surface plasmon resonance (SPR) confirmed a moderate binding affinity. The therapeutic potential of C1K was evaluated in TNBC cell lines (4T1, MDA-MB-231, and PTX-resistant MDA-MB-231) and in vivo using a syngeneic 4T1 mouse model. Functional assays demonstrated that C1K inhibited TGF-β-mediated signaling, reduced autophagy, a key mechanism underlying PTX resistance, and significantly enhanced PTX-induced apoptosis. In vivo studies further revealed synergistic effects of C1K and PTX, resulting in enhanced apoptosis in both sensitive and PTX-resistant TNBC cells. These findings suggest that C1K effectively targets TGF-β to inhibit autophagy and potentiate the apoptotic effects of PTX, as a promising combinatorial therapeutic agent for improving treatment outcomes in TNBC patients.
Collapse
Affiliation(s)
- Eun Ju Song
- Graduate School of New Drug Discovery and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Won-Sam Kim
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Yunhee Han
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Cheolmin Lee
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea; Graduate School of Chemistry, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeonbuk State, 54896 Republic of Korea
| | - Eun-Joung Moon
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea
| | - Hae-Jin Kim
- Department of New Drug Research Institute, Ensol Biosciences Inc., Techno 10-ro 51, Yuseong-gu, Daejeon 34036, Republic of Korea.
| | - Nam Sook Kang
- Graduate School of New Drug Discovery and Development, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
13
|
Majmudar PR, Keri RA. The neural stem cell gene PAFAH1B1 controls cell cycle progression, DNA integrity, and paclitaxel sensitivity of triple-negative breast cancer cells. J Biol Chem 2025:110235. [PMID: 40378956 DOI: 10.1016/j.jbc.2025.110235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/19/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive disease with limited approved therapeutic options. The rapid growth and genomic instability of TNBC cells makes mitosis a compelling target, and a current mainstay of treatment is paclitaxel (Ptx), a taxane that stabilizes microtubules during mitosis. While initially effective, acquired resistance to Ptx is common, and other antimitotic therapies can be similarly rendered ineffective due to the development of resistance or systemic toxicity underscoring the need for new therapeutic approaches. Interrogating CRISPR essentiality screens in TNBC cell lines, we identified PAFAH1B1 (LIS1) as a potential vulnerability in this disease. PAFAH1B1 regulates mitotic spindle orientation, proliferation, and cell migration during neurodevelopment, yet little is known regarding its function in breast cancer. We found that suppressing PAFAH1B1 expression in TNBC cells reduces cell number, while non-malignant cells remain unaffected. PAFAH1B1 suppression alters cell cycle dynamics, increasing mitotic duration and accumulation of cells in the G2/M phase. The suppression of PAFAH1B1 expression also increases DNA double-strand breaks, indicating a requirement for sustained PAFAH1B1 expression to maintain the genomic integrity of TNBC cells. Lastly, PAFAH1B1 silencing substantially enhances these defects in cells that are taxane-resistant and sensitizes both parental and Ptx-resistant TNBC cells to Ptx. These results indicate that LIS1/PAFAH1B1 may be a novel target for the development of new anti-mitotic agents for treating TNBC, particularly in the context of paclitaxel resistance.
Collapse
Affiliation(s)
- Parth R Majmudar
- Department of Pharmacology, Case Western Reserve University School of Medicine, 2109 Adelbert Road, Cleveland, OH 44106, United States; Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States
| | - Ruth A Keri
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, United States.
| |
Collapse
|
14
|
Zhang S, Liu J, Yang Y, Tao R, Ren X, Zhou X, Liu S. Valproic acid induces ferroptosis and suppresses the proliferation of MDA-MB-231 cells by targeting FDFT1. Front Pharmacol 2025; 16:1540667. [PMID: 40438588 PMCID: PMC12116638 DOI: 10.3389/fphar.2025.1540667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Valproic acid (VPA) constitutes a branched-chain, short-chain fatty acid that serves as an antiepileptic medication. It has been increasingly recognized that VPA has presented potential anti-tumor properties, including breast cancer. However, the exploration of novel breast cancer treatment methods necessitates a more comprehensive and in-depth understanding of the novel mechanism of VPA inhibition of breast cancer. It has been proven that farnesyl-diphosphate farnesyltransferase 1 (FDFT1) participate in oncogenesis and development of cancers. However, the effect of FDFT1 on breast cancer is still obscure. Thus, it is important to investigate the potential of VPA to trigger ferroptosis in breast cancer cells via targeting FDFT1. Methods In this study, the underlying mechanisms of VPA on ferroptosis in breast cancer cells were explored in vitro and vivo. Initially, the effects of VPA on the proliferation of breast cancer cells were assessed utilizing the Cell Counting Kit-8, cell counting, and colony formation assays. Subsequently, the ferroptosis in breast cancer cells treated with VPA were determined through the use of the Lipid Peroxidation malondialdehyde Assay Kit, reduced glutathione and oxidized glutathione disulfide Assay Kit, flow cytometry, transmission electron microscopy, and western bloting. To explore the impact of VPA in combination with ferrostatin-1, Erastin or RSL3, on MDA-MB-231 cell proliferation and ferroptosis, respective CCK-8, colony formation and WB assays were conducted. Thereafter, we assessed whether VPA facilitated ferroptosis in MDA-MB-231 cells by modulating the expression of FDFT1. Finally, the anti-breast cancer effects of VPA in vivo were validated through a xenograft mouse model, and histological examination via hematoxylin-eosin staining and immunohistochemistry staining were employed to delve into the underlying mechanisms of VPA's inhibitory effects on breast cancer cells in vivo. Results and Discussion The assay outcomes indicated that VPA impedes the proliferation of breast cancer cells. The findings from the ferroptosis index demonstrated that MDA-MB-231 cells are more sensitive to VPA induced ferroptosis than MCF-7 cells. Subsequent to the introduction of ferrostatin-1 (Fer-1), Erastin or RSL3, it was observed that Fer-1 reversed the ferroptosis facilitated by VPA, whereas Erastin or RSL3, in conjunction with VPA, respectively, induced ferroptosis in MDA-MB-231 cells. We revealed that the downregulation of FDFT1 enhanced proliferation and inhibited ferroptosis of MDA-MB-231 cells. Additionally, we discovered that VPA may facilitate ferroptosis in MDA-MB-231 cells by negatively modulating the levels of the solute carrier family 7 member 11 (SLC7A11) protein through the upregulation of FDFT1 expression. In conclusion, this study elucidated that VPA induced the ferroptosis of MDA-MB-231 cells via targeting FDFT1, representing a novel mechanism underlying its efficacy in potentially inhibiting breast cancer.
Collapse
Affiliation(s)
- Shuxian Zhang
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Jiazhuo Liu
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Yisong Yang
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Ran Tao
- Department of Anatomy, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xin Ren
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Xingzhi Zhou
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| | - Shuangping Liu
- Engineering Technology Research Center for Functional Component Utilization of Organic Natural Products, Medical College, Dalian University, Dalian, Liaoning, China
| |
Collapse
|
15
|
Rosenbaum SR, Hughes CJ, Fields KM, Purdy SC, Gustafson AL, Wolin A, Hampton D, Shrivastava NM, Turner N, Danis E, Ebmeier C, Spoelstra N, Richer J, Jedlicka P, Costello JC, Zhao R, Ford HL. EYA3 regulation of NF-κB and CCL2 suppresses cytotoxic NK cells in the premetastatic niche to promote TNBC metastasis. SCIENCE ADVANCES 2025; 11:eadt0504. [PMID: 40333987 PMCID: PMC12057687 DOI: 10.1126/sciadv.adt0504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
Triple-negative breast cancer cells must evade immune surveillance to metastasize to distant sites, yet this process is not well understood. The Eyes absent (EYA) family of proteins, which are crucial for embryonic development, become dysregulated in cancer, where they have been shown to mediate proliferation, migration, and invasion. Our study reveals an unusual mechanism by which EYA3 reduces the presence of cytotoxic natural killer (NK) cells in the premetastatic niche (PMN) to enhance metastasis, independent of its effects on the primary tumor. We find that EYA3 up-regulates nuclear factor κB signaling to enhance CCL2 expression, which, in contrast to previous findings, suppresses cytotoxic NK cell activation in vitro and their infiltration into the PMN in vivo. These findings uncover an unexpected role for CCL2 in inhibiting NK cell responses at the PMN and suggest that targeting EYA3 could be an effective strategy to reactivate antitumor immune responses to inhibit metastasis.
Collapse
Affiliation(s)
- Sheera R. Rosenbaum
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Connor J. Hughes
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Kaiah M. Fields
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Stephen Connor Purdy
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Annika L. Gustafson
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Arthur Wolin
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Drake Hampton
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Natasha M. Shrivastava
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Nicholas Turner
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Etienne Danis
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Christopher Ebmeier
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Nicole Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Jennifer Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Paul Jedlicka
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - James C. Costello
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Rui Zhao
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
| | - Heide L. Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Pharmacology and Molecular Medicine Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Molecular Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus (AMC), Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| |
Collapse
|
16
|
Yu Y, Hu J, Wang W, Lei H, Xi Z, Zhang P, Zhao E, Lu C, Chen H, Liu C, Li L. Targeting PSMD14 combined with arachidonic acid induces synthetic lethality via FADS1 m 6A modification in triple-negative breast cancer. SCIENCE ADVANCES 2025; 11:eadr3173. [PMID: 40344056 PMCID: PMC12063657 DOI: 10.1126/sciadv.adr3173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Dysregulation of deubiquitination is essential for cancer growth. However, the role of 26S proteasome non-ATPase regulatory subunit 14 (PSMD14) in the progression of triple-negative breast cancer (TNBC) remains to be determined. Gain- and loss-of-function experiments showed that silencing PSMD14 notably attenuated the growth, invasion, and metastasis of TNBC cells in vitro and in vivo. Overexpression of PSMD14 produced the opposite results. Mechanistically, PSMD14 decreased K63-linked ubiquitination on SF3B4 protein to de-ubiquitin and stabilize SF3B4 protein. Then, SF3B4/HNRNPC complex bound to FADS1 mRNA and promoted exon inclusion in the target mRNA through m6A site on FADS1 mRNA recognized by HNRNPC, thereby up-regulating the expression of FADS1 and activating Akt/mTOR signaling. Exogenous arachidonic acid supplementation combined with PSMD14 knockdown induced synthetic lethality, which was further confirmed in TNBC organoid (PDO) and TNBC patient-derived xenograft (PDX) mouse models. Overall, our findings reveal an oncogenic role of PSMD14 in TNBC progression, which indicates a potential biomarker and ferroptosis-mediated therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Yuanhang Yu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jin Hu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenwen Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hao Lei
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laboratory of Tropical Translational Medicine of Ministry of Education and School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Zihan Xi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiyi Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ende Zhao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chong Lu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hengyu Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laboratory of Tropical Translational Medicine of Ministry of Education and School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Li
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
17
|
Wang XD, Liu YS, Liang ZL, Hu MH. Mitochondrial DNA-targeted triphenylamine-thiophene (TPATP)-derived ligands boost type-I/II photodynamic therapy for triple-negative breast cancer. Eur J Med Chem 2025; 289:117489. [PMID: 40064143 DOI: 10.1016/j.ejmech.2025.117489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025]
Abstract
Triple-negative breast cancer (TNBC) is aggressive with poor prognosis. Current strategies include chemotherapy, surgery, and radiotherapy, but face challenges like suboptimal outcomes, low survival, and drug resistance. Thus, novel TNBC therapies are crucial. Activity-based photodynamic therapy (PDT) is a highly regarded cancer treatment strategy known for its spatiotemporal precision, making it a promising option for the treatment of TNBC. In this study, we designed and synthesized three triphenylamine-thiophene (TPATP)-derived ligands binding to mitochondrial DNA G4 (mtG4), which were able to label mitochondria in TNBC cells under red-light excitation, and demonstrated significant phototoxicity through type-I/II process under white-light irradiation, hinting at dual-functional potential for PDT and imaging. The optimal ligand, TP2, was demonstrated to disrupt mitochondrial functions under white-light irradiation, leading to MMP loss, ATP reduction, ROS increase, which further triggered significant apoptosis in TNBC cells.
Collapse
Affiliation(s)
- Xiao-Dong Wang
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Yong-Si Liu
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Zhi-Ling Liang
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Ming-Hao Hu
- Nation-Regional Engineering Lab for Synthetic Biology of Medicine, International Cancer Center, School of Pharmacy, Shenzhen University Medical School, Shenzhen, 518060, China.
| |
Collapse
|
18
|
Wang W, Chaudhary R, Szpendyk J, El Khalki L, Yousafzai NA, Chan R, Desai A, Sossey-Alaoui K. Kindlin-2-Mediated Hematopoiesis Remodeling Regulates Triple-Negative Breast Cancer Immune Evasion. Mol Cancer Res 2025; 23:450-462. [PMID: 39918417 DOI: 10.1158/1541-7786.mcr-24-0698] [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: 07/23/2024] [Revised: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 05/03/2025]
Abstract
Triple-negative breast cancer (TNBC) presents significant clinical challenges because of its limited treatment options and aggressive behavior, often associated with poor prognosis. This study focuses on kindlin-2, an adapter protein, and its role in TNBC progression, particularly in hematopoiesis-mediated immune evasion. TNBC tumors expressing high levels of kindlin-2 induce a notable reshaping of hematopoiesis, promoting the expansion of myeloid cells in the bone marrow and spleen. This shift correlated with increased levels of neutrophils and monocytes in tumor-bearing mice over time. Conversely, genetic knockout (KO) of kindlin-2 mitigated this myeloid bias and fostered T-cell infiltration within the tumor microenvironment, indicating the pivotal role of kindlin-2 in immune modulation. Further investigations revealed that kindlin-2 deficiency led to reduced expression of PD-L1, a critical immune checkpoint inhibitor, in TNBC tumors. This molecular change sensitized kindlin-2-deficient tumors to host antitumor immune responses, resulting in enhanced tumor suppression in immunocompetent mouse models. Single-cell RNA sequencing, bulk RNA sequencing, and IHC data supported these findings by highlighting enriched immune-related pathways and increased infiltration of immune cells in kindlin-2-deficient tumors. Therapeutically, targeting PD-L1 in kindlin-2-expressing TNBC tumors effectively inhibited tumor growth, akin to the effects observed with genetic kindlin-2 KO or PD-L1 KO. Our data underscore kindlin-2 as a promising therapeutic target in combination with immune checkpoint blockade to bolster antitumor immunity and counteract resistance mechanisms typical of TNBC and other immune-evasive solid tumors. Implications: Kindlin-2 regulates tumor immune evasion through the systemic modulation of hematopoiesis and PD-L1 expression, which warrants therapeutic targeting of kindlin-2 in patients with TNBC.
Collapse
Affiliation(s)
- Wei Wang
- MetroHealth System, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | | | | | - Lamyae El Khalki
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Neelum Aziz Yousafzai
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Ricky Chan
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Amar Desai
- Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Khalid Sossey-Alaoui
- MetroHealth System, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
- Case Comprehensive Cancer Center, Cleveland, Ohio
| |
Collapse
|
19
|
Tafenzi HA, Choulli F, Essaadi I, Belbaraka R. Real-World Outcomes of Combination Anthracycline and Taxane Adjuvant Therapies in Early Triple-Negative Breast Cancer: A Moroccan Retrospective Analysis. JCO Glob Oncol 2025; 11:e2400650. [PMID: 40344550 DOI: 10.1200/go-24-00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
PURPOSE Neoadjuvant chemoimmunotherapy followed by adjuvant immunotherapy is the gold standard for treating patients with higher risk early triple-negative breast cancer (TNBC); however, in some cases, these patients undergo surgery followed by chemotherapy-based anthracyclines and taxanes without adhering to the guidelines. METHODS Patients with previously untreated stage I, II, and III TNBC who received adjuvant therapy with either doxorubicin and cyclophosphamide (AC) + docetaxel (AC-D), AC + weekly paclitaxel (AC-WP), epirubicin and cyclophosphamide (EC) + docetaxel (EC-D), or EC + WP (EC-WP); older than 18 years; and diagnosed between January 1st, 2011, and December 31st, 2022, were eligible for the study. Disease-free survival (DFS) is the primary reported end point. Overall survival (OS) and safety were the secondary end points. RESULTS We included 272 female patients. At a prespecified event-driven data cutoff, with a median follow-up of 26.3 months, the 5-year DFS was 49% (95% CI, 38 to 63) in the AC-D group, 45% (95% CI, 29 to 70) in the AC-WP group, 73% (95% CI, 61 to 100) in the EC-D group, and 72% (95% CI, 44 to 100) in the EC-WP group (hazard ratio [HR], 0.2 [95% CI, 0.06 to 0.67]; P = .08). The 7-year OS was 52% (95% CI, 32 to 83) in the AC-D group, 88% (95% CI, 78 to 99) in the AC-WP group, 95% (95% CI, 88 to 100) in the EC-D group, and 83% (95% CI, 58 to 100) in the EC-WP group (HR, 0.19 [95% CI, 0.06 to 0.66]; P = .03). Most of the grade 3-4 adverse events occurred in the AC-D group, primarily neutropenia, nausea-vomiting, and alopecia. CONCLUSION EC-D was linked to a slightly longer survival free of invasive, noninvasive, or distant disease and a significantly longer OS with fewer adverse events. Further studies are needed to confirm and establish long-term clinical benefits.
Collapse
Affiliation(s)
- Hassan Abdelilah Tafenzi
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | - Farah Choulli
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| | - Ismail Essaadi
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
- Medical Oncology Department, Avicenna Military Hospital of Marrakech, Marrakech, Morocco
| | - Rhizlane Belbaraka
- Medical Oncology Department, Mohammed VI University Hospital of Marrakech, Marrakech, Morocco
- Biosciences and Health Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
20
|
Zhang W, Wang S, Xing Y, Luo X, Wang R, Yu F. Bioorthogonal SERS-bioluminescence dual-modal imaging for real-time tracking of triple-negative breast cancer metastasis. Acta Biomater 2025; 197:431-443. [PMID: 40101869 DOI: 10.1016/j.actbio.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, characterized by early metastasis and a poor prognosis. Traditional imaging modalities often lack the sensitivity and molecular specificity required for the early detection of metastatic lesions. In this study, we developed a dual-modal imaging strategy that integrates surface-enhanced Raman scattering (SERS) and bioluminescence imaging probes, utilizing bioorthogonal labeling to track TNBC organ metastasis. The SERS probes were encapsulated with azide-labeled macrophage membranes to extend circulation time and enhance targeting efficiency. Additionally, bioorthogonal metabolic glycolengineering was employed to modify luciferase-labeled tumor cells (4T1-Luc) with bicyclo[6.1.0]nonyne (BCN) groups, facilitating precise binding between the probes and 4T1-Luc cells through click chemistry reactions. This dual-modal imaging approach enabled real-time monitoring of small metastatic lesions with high sensitivity, providing a non-invasive and accurate method for assessing tumor metastasis and therapeutic response in vivo. Our findings indicate that the dual-modal imaging technique, combining SERS and bioluminescence with bioorthogonal labeling, holds significant potential for advanced applications in oncology. STATEMENT OF SIGNIFICANCE: This study devised a surface-enhanced Raman scattering (SERS) and bioluminescence dual-modal imaging strategy integrated with a bioorthogonal label to address the challenge of tracking the metastasis of aggressive triple-negative breast cancer (TNBC). In contrast to conventional methods, this approach facilitated real-time, whole-body monitoring of tumor dissemination through bioluminescence. Simultaneously, it achieved the detection of micro-metastases in organs using SERS, thereby exceeding the sensitivity limitations of existing imaging techniques. Clinical validation with human samples further demonstrated its potential for non-invasive therapeutic assessment and early intervention. By bridging preclinical innovation and clinical requirements, this research offered a transformative tool for precision oncology. It is expected to attract the interest of researchers in the fields of biomedicine, nanotechnology, and cancer therapeutics.
Collapse
Affiliation(s)
- Wei Zhang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Sisi Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China; Department of Breast and Thyroid Surgery, The Second Affiliated Hospital, Hainan Medical University, Haikou 571199, PR China
| | - Yanlong Xing
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Xianzhu Luo
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China
| | - Rui Wang
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China.
| | - Fabiao Yu
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, Key Laboratory of Haikou Trauma, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, PR China; Engineering Research Center for Hainan Bio-Smart Materials and Bio-Medical Devices, Key Laboratory of Hainan Functional Materials and Molecular Imaging, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, PR China.
| |
Collapse
|
21
|
Alonso-Valenteen F, Mikhael S, Wang H, Sims J, Taguiam M, Teh J, Sances S, Wong M, Miao T, Srinivas D, Gonzalez-Almeyda N, Cho RH, Sanchez R, Nguyenle K, Serrano E, Ondatje B, Benhaghnazar RL, Gray HB, Gross Z, Yu J, Svendsen CN, Abrol R, Medina-Kauwe LK. Systemic HER3 ligand-mimicking nanobioparticles enter the brain and reduce intracranial tumour growth. NATURE NANOTECHNOLOGY 2025; 20:683-696. [PMID: 39984637 PMCID: PMC12095042 DOI: 10.1038/s41565-025-01867-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/14/2025] [Indexed: 02/23/2025]
Abstract
Crossing the blood-brain barrier (BBB) and reaching intracranial tumours is a clinical challenge for current targeted interventions including antibody-based therapies, contributing to poor patient outcomes. Increased cell surface density of human epidermal growth factor receptor 3 (HER3) is associated with a growing number of metastatic tumour types and is observed on tumour cells that acquire resistance to a growing number of clinical targeted therapies. Here we describe the evaluation of HER3-homing nanobiological particles (nanobioparticles (NBPs)) on such tumours in preclinical models and our discovery that systemic NBPs could be found in the brain even in the absence of such tumours. Our subsequent studies described here show that HER3 is prominently associated with both mouse and human brain endothelium and with extravasation of systemic NBPs in mice and in human-derived BBB chips in contrast to non-targeted agents. In mice, systemically delivered NBPs carrying tumoricidal agents reduced the growth of intracranial triple-negative breast cancer cells, which also express HER3, with improved therapeutic profile compared to current therapies and compared to agents using traditional BBB transport routes. As HER3 associates with a growing number of metastatic tumours, the NBPs described here may offer targeted efficacy especially when such tumours localize to the brain.
Collapse
Affiliation(s)
| | - Simoun Mikhael
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - HongQiang Wang
- Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jessica Sims
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael Taguiam
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - James Teh
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sam Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michelle Wong
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Tianxin Miao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dustin Srinivas
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Ryan H Cho
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Kimngan Nguyenle
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Erik Serrano
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Briana Ondatje
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Harry B Gray
- California Institute of Technology, Pasadena, CA, USA
| | - Zeev Gross
- Technion-Israel Institute, Haifa, Israel
| | - John Yu
- Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ravinder Abrol
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- California State University, Northridge, CA, USA
| | - Lali K Medina-Kauwe
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Linstra R, Stappenbelt C, Bakker FJ, Everts M, Bhattacharya A, Yu S, van Bergen SD, van der Vegt B, Wisman GBA, Fehrmann RSN, de Bruyn M, van Vugt MATM. MYC controls STING levels to downregulate inflammatory signaling in breast cancer cells upon DNA damage. J Biol Chem 2025; 301:108560. [PMID: 40311680 PMCID: PMC12166436 DOI: 10.1016/j.jbc.2025.108560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Amplification of the MYC proto-oncogene is frequently observed in various cancer types, including triple-negative breast cancer (TNBC). Emerging evidence suggests that suppression of local antitumor immune responses by MYC, at least in part, explains the tumor-promoting effects of MYC. Specifically, MYC upregulation was demonstrated to suppress the tumor-cell intrinsic activation of a type I interferon response and thereby hamper innate inflammatory signaling, which may contribute to the disappointing response to immunotherapy in patients with TNBC. In this study, we show that MYC interferes with protein expression and functionality of the STING pathway. MYC-mediated STING downregulation in BT-549 and MDA-MB-231 TNBC cell lines requires the DNA-binding ability of MYC and is independent of binding of MYC to its co-repressor MIZ1. Both STAT1 and STAT3 promote the steady-state expression levels of STING, and STAT3 cooperates with MYC in regulating STING. Conversely, MYC-mediated downregulation of STING affects protein levels of STAT1 and downstream chemokine production. Furthermore, we show that MYC overexpression hampers immune cell activation triggered by DNA damage through etoposide or irradiation treatment and specifically impedes the activation of natural killer cells. Collectively, these results show that MYC controls STING levels and thereby regulates tumor cell-intrinsic inflammatory signaling. These results contribute to our understanding of how MYC suppresses inflammatory signaling in TNBC and may explain why a large fraction of patients with TNBC do not benefit from immunotherapy.
Collapse
Affiliation(s)
- Renske Linstra
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Chantal Stappenbelt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Femke J Bakker
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marieke Everts
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Arkajyoti Bhattacharya
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Shibo Yu
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stella D van Bergen
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
23
|
Zhao Q, Pramanik J, Lu Y, Homer NZM, Imianowski CJ, Zhang B, Iqbal M, Shaji SK, Morris AC, Roychoudhuri R, Okkenhaug K, Qiu P, Mahata B. Perturbing local steroidogenesis to improve breast cancer immunity. Nat Commun 2025; 16:3945. [PMID: 40287432 PMCID: PMC12033260 DOI: 10.1038/s41467-025-59356-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Breast cancer, particularly triple-negative breast cancer (TNBC), evades the body's immune defences, in part by cultivating an immunosuppressive tumour microenvironment. Here, we show that suppressing local steroidogenesis can augment anti-tumour immunity against TNBC. Through targeted metabolomics of steroids coupled with immunohistochemistry, we profiled the existence of immunosuppressive steroids in TNBC patient tumours and discerned the steroidogenic activity in immune-infiltrating regions. In mouse, genetic inhibition of immune cell steroidogenesis restricted TNBC tumour progression with a significant reduction in immunosuppressive components such as tumour associated macrophages. Steroidogenesis inhibition appears to bolster anti-tumour immune responses in dendritic and T cells by impeding glucocorticoid signalling. Undertaking metabolic modelling of the single-cell transcriptomics and targeted tumour-steroidomics, we pinpointed the predominant steroidogenic cells. Inhibiting steroidogenesis pharmacologically using a identified drug, posaconazole, curtailed tumour expansion in a humanised TNBC mouse model. This investigation paves the way for targeting steroidogenesis and its signalling pathways in breast cancer affected by immune-steroid maladaptation.
Collapse
Affiliation(s)
- Qiuchen Zhao
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Cancer Research UK Cambridge Centre and Department of Oncology, University of Cambridge, Cambridge, CB2 0XZ, UK
| | - Jhuma Pramanik
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Yongjin Lu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Natalie Z M Homer
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Baojie Zhang
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Muhammad Iqbal
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | | | | | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Pengfei Qiu
- Breast Cancer Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- The Precision Breast Cancer Institute, Addenbrookes Hospital, Department of Oncology, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Bidesh Mahata
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
| |
Collapse
|
24
|
Ta TM, Reed VL, Gupta SRR, Khan M, Chandra N, Dwyer N, Fultang N, Singh IK, Peethambaran B. Novel macromolecule CPD4 suppresses cell proliferation and metastasis of triple-negative breast cancer by targeting ROR1 protein. Int J Biol Macromol 2025; 310:143301. [PMID: 40274160 DOI: 10.1016/j.ijbiomac.2025.143301] [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: 02/25/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Breast cancer is the second most common cancer among women in the US, with triple-negative breast cancer (TNBC) accounting for 15-20 % of new diagnoses. TNBC cells lack estrogen and progesterone receptors, and human epidermal growth factor receptor 2, which makes them resistant to standard hormone treatments. Current therapies like chemotherapy and radiation often harm both cancerous and healthy cells, underscoring the need for developing new targeted treatments. ROR1, an oncoprotein that is overexpressed in various cancers, including breast cancer, is minimally present in normal tissues. Targeting ROR1 signaling has been shown to trigger apoptosis and reduce TNBC cell proliferation. A novel macromolecule compound, CPD4, was discovered through in-silico docking for its ability to bind and inhibit the pseudokinase domain of ROR1. In vitro evidence revealed that CPD4 decreases cell viability and induces apoptosis in TNBC cell lines at concentrations of 2-10 μM, while leaving normal breast cells unharmed. CPD4 also blocks migration, invasion, and causes G2/M-phase arrest in TNBC cells. Its mechanism of action involves reducing key downstream markers of ROR1 signaling, particularly the phosphorylation of AKT/GSK3β. In 3D spheroid cultures, CPD4 reduces the size of TNBC spheroids. Moreover, the combination treatment of CPD4 and the standard chemotherapy docetaxel exhibits synergistic efficacy against different TNBC cell lines with a combination index below 0.01. These results suggest that CPD4 holds promise as a therapeutic option for TNBC and could potentially benefit other cancers with ROR1 overexpression.
Collapse
Affiliation(s)
- Tram M Ta
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Victoria L Reed
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Shradheya R R Gupta
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
| | - Maryam Khan
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Nikhil Chandra
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Nick Dwyer
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Norman Fultang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Indrakant K Singh
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India; Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi, India
| | - Bela Peethambaran
- Department of Biology, Saint Joseph's University, Philadelphia, PA 19131, USA.
| |
Collapse
|
25
|
Luo L, Yuan F, Palovcak A, Li F, Yuan Q, Calkins D, Manalo Z, Li Y, Wang D, Zhou M, Zhou C, Li M, Tan YD, Bai F, Ban Y, Mason C, Roberts E, Bilbao D, Liu ZJ, Briegel K, Welford SM, Pei XH, Daunert S, Liu W, Zhang Y. Oncogenic properties of wild-type DNA repair gene FANCA in breast cancer. Cell Rep 2025; 44:115480. [PMID: 40146775 DOI: 10.1016/j.celrep.2025.115480] [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: 09/23/2024] [Revised: 01/10/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
FANCA is one of the 23 genes whose deficiencies lead to defective DNA interstrand crosslink repair and cancer-prone Fanconi anemia disease. Beyond its functions in DNA repair and tumor suppression, we report that high FANCA expression is strongly associated with breast cancer development. Overexpression of WT-FANCA significantly promotes breast cancer cell proliferation and tumor growth both in vitro and in vivo, while FANCA deficiency severely compromises the proliferation of breast cancer cells, but not non-tumorigenic breast epithelial cells. Heterozygous knockout of FANCA in breast cancer mouse models is sufficient to cause significant reduction of breast tumor growth in vivo. Furthermore, we have shown that high FANCA expression in breast cancer correlates with promoter hypomethylation in a TET-dependent manner, and TET inhibition recapitulates the proliferation defects caused by FANCA deficiency. Our study identifies the oncogenic properties of WT-FANCA and shows that FANCA is a promising target for breast cancer intervention.
Collapse
Affiliation(s)
- Liang Luo
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fenghua Yuan
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anna Palovcak
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fang Li
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Qingqi Yuan
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Calkins
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zoe Manalo
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yan Li
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dazhi Wang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mike Zhou
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Catherine Zhou
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matthew Li
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yuan-De Tan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Feng Bai
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yuguang Ban
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Evan Roberts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Pathology & Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zhao-Jun Liu
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karoline Briegel
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Scott M Welford
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xin-Hai Pei
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Sylvia Daunert
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wenjun Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yanbin Zhang
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
26
|
Castellano I, Rousset S, Casella D, Capella G, Borella F, Rosa MD, Cassoni P, Catalano A, Ferrante G, Giordano L. Early detection of triple-negative breast cancer: evidence of a favourable prognostic impact in a comparative analysis of screen-detected versus symptomatic cases. BMC Cancer 2025; 25:730. [PMID: 40251506 PMCID: PMC12007119 DOI: 10.1186/s12885-025-14067-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
PURPOSE Mammographic screening is effective in reducing breast cancer mortality, but the impact of screening on triple-negative breast cancers (TNBCs) outcomes remains debated. This study aims to determine if screen detection is an independent prognostic factor for TNBCs and to analyse the radiological and pathological differences between screen-detected and symptomatic TNBCs. METHODS This retrospective cohort study analysed 353 histologically confirmed TNBC cases diagnosed between 2013 and 2020 at a single institution in Turin, Italy. Cases were categorized into screen-detected and symptomatic groups based on initial presentation. Clinical, radiological and pathological characteristics as well as disease-free survival (DFS) and overall survival (OS) were compared between groups. Statistical analyses included Kaplan-Meier survival curves and Cox proportional hazard models, adjusting for several clinical and biological variables. RESULTS 50.1% of cases were screen-detected and 49.9% were symptomatic. Screen-detected cases were more commonly smaller (T1 or T2) (96.6%) than symptomatic cases (75%) (p < 0.001). Also, compared to symptomatic tumours, screen-detected ones were more often node negative (62.4% vs. 48%, p = 0.007) and diagnosed at a lower stage (85.4% vs. 63.8%, p < 0.001), with better DFS and OS. Detection method was not an independent prognostic factor, while stage at diagnosis, vascular invasion, histologic type and tumour-infiltrating lymphocytes (TILS) were more significant predictors of prognosis. Radiological and biological features were similar between the two groups. CONCLUSIONS TNBCs correlate with favourable pathological features and improved survival outcomes in univariate analyses, but these benefits diminish when accounting for traditional prognostic factors. Hence, the better prognosis observed among screen-detected cases is more likely due to stage shift rather than tumour biology.
Collapse
Affiliation(s)
| | - Stefano Rousset
- Department of Public Health and Pediatrics, Post Graduate School of Medical Statistics, University of Turin & CPO Piemonte, Turin, Italy.
| | - Denise Casella
- SSD Epidemiologia Screening, CPO-AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Giulia Capella
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fulvio Borella
- Obstetrics and Gynecology Unit 1, Department of Surgical Sciences, Sant'Anna Hospital, University of Turin, Turin, Italy
| | - Martina Di Rosa
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Alberto Catalano
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Centre for Biostatistics, Epidemiology, and Public Health, Department of Clinical and Biological Sciences, University of Turin, Orbassano, TO, Italy
| | - Gianluigi Ferrante
- SSD Epidemiologia Screening, CPO-AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Livia Giordano
- SSD Epidemiologia Screening, CPO-AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| |
Collapse
|
27
|
Schmid P, Oliveira M, O’Shaughnessy J, Cristofanilli M, Graff SL, Im SA, Loi S, Saji S, Wang S, Cescon DW, Hovey T, Nawrot A, Tse K, Vukovic P, Curigliano G. TROPION-Breast05: a randomized phase III study of Dato-DXd with or without durvalumab versus chemotherapy plus pembrolizumab in patients with PD-L1-high locally recurrent inoperable or metastatic triple-negative breast cancer. Ther Adv Med Oncol 2025; 17:17588359251327992. [PMID: 40297626 PMCID: PMC12035291 DOI: 10.1177/17588359251327992] [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: 11/08/2024] [Accepted: 02/28/2025] [Indexed: 04/30/2025] Open
Abstract
Background Standard of care (SoC) for patients with advanced triple-negative breast cancer (TNBC) whose tumors express PD-L1 (combined positive score ⩾ 10) is chemotherapy plus anti-PD-(L)1 inhibitors; however, prognosis and survival for most patients is poor. Datopotamab deruxtecan (Dato-DXd), a novel antibody-drug conjugate comprising a humanized anti-TROP2 IgG1 monoclonal antibody conjugated to a potent topoisomerase I inhibitor payload via a plasma-stable, cleavable, tetrapeptide-based linker, has shown preliminary activity as mono or combination therapy in advanced/metastatic TNBC. Objectives TROPION-Breast05 is an ongoing randomized, open-label, multicenter phase III study. The primary objective is to demonstrate the superiority of Dato-DXd in combination with durvalumab (an anti-PD-L1 antibody) versus SoC treatment in patients with PD-L1-high locally recurrent inoperable or metastatic TNBC. Methods and design Patients (⩾18 years) will be randomized 1:1 to receive Dato-DXd (6 mg/kg intravenously (IV) every 3 weeks (Q3W)) plus durvalumab (1120 mg IV Q3W) or investigator's choice of chemotherapy (ICC; paclitaxel, nab-paclitaxel, or gemcitabine plus carboplatin) plus pembrolizumab (200 mg IV Q3W). In selected countries, patients will also be randomized (1:1:1) to a third arm of Dato-DXd monotherapy. The primary study endpoint is progression-free survival (PFS) per blinded independent central review (Dato-DXd plus durvalumab arm vs ICC plus pembrolizumab arm). Overall survival is a key secondary endpoint; other secondary endpoints include PFS (investigator-assessed), objective response rate, duration of response, clinical benefit rate at Week 24 (all assessed in the Dato-DXd plus durvalumab arm vs ICC plus pembrolizumab arm), patient-reported outcomes, and safety. Ethics The study is approved by independent ethics committees or institutional review boards at each study site. All patients will provide written informed consent. Discussion TROPION-Breast05 will assess the potential role of Dato-DXd with or without durvalumab in patients with PD-L1-high advanced or metastatic TNBC. The findings of this trial could lead to a new treatment option for these patients. Trial registration ClinicalTrials.gov identifier: NCT06103864 (Date of registration: 27 October 2023).
Collapse
Affiliation(s)
- Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
| | - Mafalda Oliveira
- Vall d’Hebron University Hospital, Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | | | - Stephanie L. Graff
- Brown University Health Cancer Institute, Legorreta Cancer Center, Brown University, Providence, RI, USA
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | - Shusen Wang
- Department of Medical Oncology, Cancer Center of Sun Yat-sen University, Guangzhou, China
| | - David W. Cescon
- Department of Medical Oncology, Princess Margaret Cancer Centre/UHN, Toronto, ON, Canada
| | - Tina Hovey
- Biostatistics, Phastar UK (under contract to AstraZeneca), London, UK
| | | | - Karson Tse
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Giuseppe Curigliano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
28
|
Tzikas AK, Holmberg E, Parris TZ, Kovács A, Lovmar L, Karlsson P. Survival outcomes in hormone receptor-negative breast cancer among BRCA carriers versus noncarriers in western Sweden. Acta Oncol 2025; 64:550-557. [PMID: 40235390 PMCID: PMC12016662 DOI: 10.2340/1651-226x.2025.43109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND AND PURPOSE BRCA-related hormone receptor (HR)-negative breast cancers (BC) are reported to have aggressive tumor biology but also exhibit chemosensitivity. However, the impact of BRCA1/2 pathogenetic variants (PV) on BC outcomes remains unclear. This study compares survival outcomes for HR-negative BC between BRCA carriers and noncarriers. PATIENTS/MATERIAL AND METHODS From 489 female BRCA-carriers prospectively registered in western Sweden (1996-2017), those with primary HR-negative BC who underwent breast surgery until 2019 were included in the BRCA cohort. For each BRCA-carrier, three BRCA-noncarriers with HR-negative BC were matched based on age, time of diagnosis, and follow-up duration. Overall survival (OS) was analyzed using Kaplan‑Meier estimates and Cox proportional hazard ratios after adjustment for stage, chemotherapy, and surgical technique. A sensitivity analysis was performed to investigate the effect of HER2 status on HR-negative BC diagnosed after 2007. RESULTS Among the 106 BRCA carriers, 101 (95%) had a BRCA1 and 5 (5%) a BRCA2 PV. Most of the BRCA-carriers (89/106, 84%) were diagnosed with BC prior to genetic screening. Surgical techniques were similar between BRCA-carriers (n = 106) and noncarriers (n = 318). Chemotherapy was more common among BRCA-carriers (87% vs. 72%, p < 0.001). No significant difference in OS was found between BRCA-carriers and noncarriers among patients with HR-negative BC (adjusted HR: 0.81 [95% confidence interval [CI]: 0.43-1.53], p = 0.51) or considering HER2 status (adjusted HR 0.95 [95% CI: 0.43-2.07], p = 0.89). INTERPRETATION This study suggests that BRCA1/2 pathogenic variants do not independently impact survival outcomes in HR-negative BC. However, a moderate association between BRCA status and OS cannot be ruled out.
Collapse
Affiliation(s)
- Anna-Karin Tzikas
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, NU hospital group, Department of Oncology, Uddevalla, Sweden.
| | - Erik Holmberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anikó Kovács
- d. Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Pathology, Gothenburg, Sweden
| | - Lovisa Lovmar
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Genetics and Genomics, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Oncology, Gothenburg, Sweden
| |
Collapse
|
29
|
Zhao G, Zeng Y, Cheng W, Karkampouna S, Papadopoulou P, Hu B, Zang S, Wezenberg E, Forn-Cuní G, Lopes-Bastos B, Julio MKD, Kros A, Snaar-Jagalska BE. Peptide-Modified Lipid Nanoparticles Boost the Antitumor Efficacy of RNA Therapeutics. ACS NANO 2025; 19:13685-13704. [PMID: 40176316 PMCID: PMC12004924 DOI: 10.1021/acsnano.4c14625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
RNA therapeutics offer a promising approach to cancer treatment by precisely regulating cancer-related genes. While lipid nanoparticles (LNPs) are currently the most advanced nonviral clinically approved vectors for RNA therapeutics, their antitumor efficacy is limited by their unspecific hepatic accumulation after systemic administration. Thus, there is an urgent need to enhance the delivery efficiency of LNPs to target tumor-residing tissues. Here, we conjugated the cluster of differentiation 44 (CD44)-specific targeting peptide A6 (KPSSPPEE) to the cholesterol of LNPs via PEG, named AKPC-LNP, enabling specific tumor delivery. This modification significantly improved delivery to breast cancer cells both in vitro and in vivo, as shown by flow cytometry and confocal microscopy. We further used AKPC-siYT to codeliver siRNAs targeting the transcriptional coactivators YAP and TAZ, achieving potent gene silencing and increased cell death in both 2D cultures and 3D tumor spheroids, outperforming unmodified LNPs. In a breast tumor cell xenografted zebrafish model, systemically administered AKPC-siYT induced robust silencing of YAP/TAZ and downstream genes and significantly enhanced tumor suppression compared to unmodified LNPs. Additionally, AKPC-siYT effectively reduced proliferation in prostate cancer organoids and tumor growth in a patient-derived xenograft (PDX) model. Overall, we developed highly efficient AKPC-LNPs carrying RNA therapeutics for targeted cancer therapy.
Collapse
Affiliation(s)
- Gangyin Zhao
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
- Shenzhen
Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 51800, China
| | - Ye Zeng
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Wanli Cheng
- Urology
Research Laboratory, Department for BioMedical Research, University of Bern, Bern 3010, Switzerland
| | - Sofia Karkampouna
- Urology
Research Laboratory, Department for BioMedical Research, University of Bern, Bern 3010, Switzerland
- Department
of Urology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Panagiota Papadopoulou
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Bochuan Hu
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Shuya Zang
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Emma Wezenberg
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Gabriel Forn-Cuní
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Bruno Lopes-Bastos
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - Marianna Kruithof-de Julio
- Department
of Urology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Alexander Kros
- Department
of Supramolecular & Biomaterials Chemistry, Leiden Institute of
Chemistry, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| | - B. Ewa Snaar-Jagalska
- Department
of Cellular Tumor Biology, Leiden Institute of Biology, Leiden University, Einsteinweg 55, Leiden 2333 CC, the Netherlands
| |
Collapse
|
30
|
Chang YT, Wu IT, Lee CH, Hung CC. Phyto-Sesquiterpene Lactones Prevent the Development of Multidrug Resistance in TNBC via ABC Transporters Inhibition and STAT3/MYC Signaling. Cancers (Basel) 2025; 17:1321. [PMID: 40282497 PMCID: PMC12026016 DOI: 10.3390/cancers17081321] [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: 03/04/2025] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Multidrug resistance (MDR) in triple-negative breast cancer (TNBC) leads to treatment failure and tumor recurrence. Dysregulation of the MYC oncogene is associated with the pathogenesis of TNBC and the development of chemoresistance via overexpression of ATP-binding cassette (ABC) transporters. Therefore, in the present study, we aimed to identify molecules from a natural product origin that prevent the development of MDR in TNBC by targeting the MYC signaling. METHODS The cell viability of TNBC was evaluated using sulforhodamine assay. Protein levels were detected by western blots or enzyme-linked immunosorbent assays. Intracellular calcein and hoechst33342 accumulation assay aimed to evaluate the inhibitory ability of phytocompounds on drug-efflux functions of ABCB1 and ABCG2 transporters. The Cancer Genome Atlas (TCGA) database was used to explore clinical genomic data. Furthermore, the zebrafish xenotransplantation model bearing Dil-labeled TNBC cells was applied to testify the in vivo effects of phyto-sesquiterpene lactones. RESULTS The results of the present study demonstrated that the phyto-sesquiterpene lactones exhibited an MDR prevention effect by repressing efflux activities of ABCB1 and ABCG2 transporters. Mechanistic studies showed that phyto-sesquiterpene lactones inducted TNBC cell apoptosis and cell cycle G2/M arrested by blocking the STAT3/MYC pathway. Clinical genomic data demonstrated that the percentages of MYC amplification and mRNA were upregulated approximately two-fold higher in the TNBC patients than the non-TNBC breast cancer patients. The survival of patients with an alteration in MYC was significantly lower in TNBC as compared to other subtypes. Moreover, the results of the zebrafish xenograft model confirmed that phyto-sesquiterpene lactones exerted stronger inhibitory effects on TNBC tumor growth in vivo. CONCLUSIONS In conclusion, these three phyto-sesquiterpene lactones were promising candidates for TNBC treatment and shed light on the prevention of developing MDR TNBC.
Collapse
Affiliation(s)
- Ying-Tzu Chang
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; (Y.-T.C.); (I.-T.W.)
| | - I-Ting Wu
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; (Y.-T.C.); (I.-T.W.)
| | - Chien-Hsing Lee
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Chin-Chuan Hung
- Department of Pharmacy, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; (Y.-T.C.); (I.-T.W.)
- Department of Pharmacy, China Medical University Hospital, Taichung 404327, Taiwan
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
31
|
Li C, Wang X, Shi D, Yang M, Yang W, Chen L. RFX5 promotes the progression of triple-negative breast cancer through transcriptional activation of JAG1. Hum Cell 2025; 38:86. [PMID: 40220043 DOI: 10.1007/s13577-025-01216-9] [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: 10/22/2024] [Accepted: 04/01/2025] [Indexed: 04/14/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype characterized by high recurrence rates, low survival rates, and a lack of therapeutic targets. Regulatory Factor X5 (RFX5) is a critical transcription factor during tumor progression. However, the role of RFX5 involving breast cancer or TNBC has not been studied. This study obtained 60 tumor samples of TNBC for analysis and ascertained that RFX5 is linked with the severe stage. We constructed RFX5 knockdown and overexpression models involving TNBC cells. RFX5 overexpression enhanced TNBC cell proliferation by detecting cell vitality and replication of DNA and analyzing cell cycle data. RFX5 facilitated cell migration and invasion, which were determined by wound healing and Transwell assays. The anti-apoptotic RFX5 properties were confirmed with Hoechst staining and Annexin V/PI apoptosis assays. The Notch pathway was activated in TNBC, and Jagged canonical Notch ligand 1 (JAG1) could enhance TNBC growth and metastasis. RFX5 upregulation elevated JAG1 mRNA and protein levels. Chromatin immunoprecipitation and luciferase reporter assays indicated that RFX5 promoted the transcriptional activation of JAG1 by binding the promoter (- 1890/+ 15 or - 1359/+ 15 area). JAG1 knockdown reduced RFX5-induced expression of Notch signaling-related factors Notch1, NICD, and Hes1. This paper indicated that RFX5 is a transcription factor for JAG1 and established that RFX5 could activate the Notch pathway via transcriptional activation of JAG1 and promote TNBC progression. Targeting RFX5 could be a promising therapeutic approach against TNBC.
Collapse
Affiliation(s)
- Chenhao Li
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China.
| | - Xin Wang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Dongliang Shi
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Meng Yang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Wenhua Yang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| | - Liang Chen
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, No.16 Xinhua West Road, Cangzhou, Hebei, China
| |
Collapse
|
32
|
Yin Y, Fan Y, Ouyang Q, Song L, Wang X, Li W, Li M, Yan X, Wang S, Sun T, Teng Y, Tang X, Tong Z, Sun Z, Ge J, Jin X, Diao Y, Liu G, Xu B. Sacituzumab tirumotecan in previously treated metastatic triple-negative breast cancer: a randomized phase 3 trial. Nat Med 2025:10.1038/s41591-025-03630-w. [PMID: 40217078 DOI: 10.1038/s41591-025-03630-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/04/2025] [Indexed: 04/25/2025]
Abstract
Chemotherapy remains a standard treatment option for metastatic triple-negative breast cancer (TNBC) but is associated with limited survival. Although some targeted antibody-drug conjugates have demonstrated clinical benefits and are considered standard therapy, persistent unmet medical needs remain due to varying accessibility. The OptiTROP-Breast01 phase 3 trial assessed sacituzumab tirumotecan (sac-TMT) versus chemotherapy in patients with locally recurrent or metastatic TNBC who had received two or more prior therapies, including at least one for metastatic disease. Patients were randomized to sac-TMT (n = 130) or chemotherapy (n = 133). The primary endpoint of progression-free survival (PFS) by blinded independent central review (BICR) was met based on the protocol-specified interim analysis. At final analysis, the median PFS by BICR was 6.7 (95% confidence interval (CI), 5.5-8.0) months with sac-TMT and 2.5 (95% CI, 1.7-2.7) months with chemotherapy (hazard ratio (HR), 0.32; 95% CI, 0.24-0.44; P < 0.00001). Concurrently, at the protocol-specified interim analysis for overall survival (OS), the median OS was not reached (95% CI, 11.2 months to not estimable (NE)) with sac-TMT and 9.4 (95% CI, 8.5-11.7) months with chemotherapy (HR, 0.53; 95% CI, 0.36-0.78; P = 0.0005). The percentage of patients with an objective response was 45.4% with sac-TMT and 12.0% with chemotherapy. The median duration of response was 7.1 (95% CI, 5.6-NE) months with sac-TMT and 3.0 (95% CI, 2.5-NE) months with chemotherapy. The most common treatment-related adverse event with sac-TMT was hematologic toxicity. Sac-TMT demonstrated statistically significant and clinically meaningful improvements in PFS compared to chemotherapy, with a manageable safety profile. The study findings support sac-TMT as an additional effective treatment option for pretreated metastatic TNBC. ClinicalTrials.gov identifier: NCT05347134 .
Collapse
Affiliation(s)
- Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Fan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | - Wei Li
- The First Hospital of Jilin University, Changchun, China
| | - Man Li
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Xi Yan
- West China Hospital of Sichuan University, Chengdu, China
| | - Shusen Wang
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tao Sun
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Yuee Teng
- The First Hospital of China Medical University, Shenyang, China
| | - Xianjun Tang
- Chongqing University Cancer Hospital, Chongqing, China
| | - Zhongsheng Tong
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | | | - Junyou Ge
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China
- National Engineering Research Center of Targeted Biologics, Chengdu, China
| | - Xiaoping Jin
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China
| | - Yina Diao
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China
| | - Gesha Liu
- Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd., Chengdu, China
| | - Binghe Xu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
33
|
Park WK, Nam SJ, Kim SW, Yu J, Lee SK, Ryu JM, Chae BJ, Shin DS, Ahn JS, Park YH, Kim JY, Shin J, Ahn HK, Cho EY, Lee H, Lee JE. Real-world evidence of the efficacy of neoadjuvant pembrolizumab in triple-negative breast cancer: A surgeon's point of view. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110011. [PMID: 40233520 DOI: 10.1016/j.ejso.2025.110011] [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: 02/18/2025] [Revised: 03/11/2025] [Accepted: 04/02/2025] [Indexed: 04/17/2025]
Abstract
PURPOSE Neoadjuvant pembrolizumab has shown efficacy in improving pathologic complete response (pCR) rates and survival outcomes in triple-negative breast cancer (TNBC). However, real-world data on its clinical efficacy, safety, and surgical outcomes remain limited. MATERIALS AND METHODS This retrospective observational study included 331 TNBC patients treated at a single institution from July 2022 to December 2023. Patients received weekly paclitaxel with carboplatin followed by doxorubicin and cyclophosphamide (AC), with or without pembrolizumab. Primary outcomes included pCR rates and surgical de-escalation in the breast and axilla. Secondary outcomes included event-free survival (EFS), overall survival (OS), surgical complications, and persistent immune-related adverse events (irAEs). RESULTS The pCR rate was higher in the pembrolizumab group (57.7 %) compared to the control group (49.1 %), with an estimated difference of 8.5 % (p = 0.120). Pembrolizumab enabled 32.7 % of patients initially planned for mastectomy to undergo breast-conserving surgery (BCS). Axillary lymph node dissection (ALND) was avoided in 91 % of pembrolizumab-treated patients versus 86.9 % in the control group. Persistent irAEs were observed in 10.9 % of the pembrolizumab group, with thyroid dysfunction being the most common. Surgical complications were similar between groups. Short-term oncologic outcomes, including EFS and OS, did not differ significantly after a median follow-up of 21 months. CONCLUSION Pembrolizumab demonstrated potential benefits in increasing pCR rates and facilitating surgical de-escalation while maintaining comparable safety to conventional regimens. Persistent irAEs underscore the need for preoperative management and vigilant monitoring. Future studies should identify predictive biomarkers to optimize pembrolizumab's use and assess its long-term impact on survival outcomes.
Collapse
Affiliation(s)
- Woong Ki Park
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seok Jin Nam
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Seok Won Kim
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jonghan Yu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Se Kyung Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jai Min Ryu
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byung Joo Chae
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Dong Seung Shin
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Junghoon Shin
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hee Kyung Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Eun Yoon Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyunwoo Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong Eon Lee
- Division of Breast Surgery, Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
34
|
Chintalaramulu N, Singh DP, Sapkota B, Raman D, Alahari S, Francis J. Caveolin-1: an ambiguous entity in breast cancer. Mol Cancer 2025; 24:109. [PMID: 40197489 PMCID: PMC11974173 DOI: 10.1186/s12943-025-02297-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/07/2025] [Indexed: 04/10/2025] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer in women and the second leading cause of death from cancer among women. Metastasis is the major cause of BC-associated mortality. Accumulating evidence implicates Caveolin-1 (Cav-1), a structural protein of plasma membrane caveolae, in BC metastasis. Cav-1 exhibits a dual role, as both a tumor suppressor and promoter depending on the cellular context and BC subtype. This review highlights the role of Cav-1 in modulating glycolytic metabolism, tumor-stromal interactions, apoptosis, and senescence. Additionally, stromal Cav-1's expression is identified as a potential prognostic marker, offering insights into its contrasting roles in tumor suppression and progression. Furthermore, Cav-1's context-dependent effects are explored in BC subtypes including hormone receptor-positive, HER2-positive, and triple-negative BC (TNBC). The review further delves into the role of Cav-1 in regulating the metastatic cascade including extracellular matrix interactions, cell migration and invasion, and premetastatic niche formation. The later sections discuss the therapeutic targeting of Cav-1 by metabolic inhibitors such as betulinic acid and Cav-1 modulating compounds. While Cav-1 may be a potential biomarker and therapeutic target, its heterogeneous expression and context-specific activity necessitates further research to develop precise interventions. Future studies investigating the mechanistic role of Cav-1 in metastasis may pave the way for effective treatment of metastatic BC.
Collapse
Affiliation(s)
- Naveen Chintalaramulu
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | | | - Biplov Sapkota
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Dayanidhi Raman
- Department of Cell and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, USA
| | | | - Joseph Francis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
35
|
Woo SH, Kim DH, Karapurkar JK, Kim SJ, Jang HY, Jang JY, Han BW, Kim JS, Park YJ, Choi MJ, Ramakrishna S, Kim KS. AXL kinase inhibitor exhibits antitumor activity by inducing apoptotic cell death in triple-negative breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119928. [PMID: 40044045 DOI: 10.1016/j.bbamcr.2025.119928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/06/2025] [Accepted: 02/25/2025] [Indexed: 04/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer associated with a poor prognosis and decreased patient survival. It is intimately linked to AXL overexpression and AXL hyperactivation. Here, we explored the therapeutic potential of AX-0085, a small molecule AXL inhibitor. While AX-0085 was previously characterized in the context of lung adenocarcinoma, this study demonstrates its application in triple-negative breast cancer (TNBC) models. AX-0085 exhibited high binding affinity to the ATP binding site located beneath the conserved glycine-rich loop (P-loop) that links the β1 and β2 strands of the AXL kinase domain. Furthermore, it was demonstrated that the benzamide group of AX-0085 and LyS567's Nζ atom could generate a hydrogen bond. AX-0085 efficiently suppressed the AXL/GAS6 signaling pathway activation in TNBC cells in vitro, which in turn prevented AXL/GAS6 signaling-dependent pro-cancerous behavior like cell proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT). In TNBC, an AX-0085-induced cell cycle arrest that took place during the G1 phase reduced the expression of CYCLIN E and CDK2. Additionally, AX-0085 facilitated apoptotic cell death in TNBC. Treatment of AX-0085 on in vivo mouse xenografts transplanted with 4 T1 cells showed a significant tumor reduction. Thus, our findings demonstrate that AX-0085 has an effective therapeutic role in TNBC by inhibiting AXL activation.
Collapse
Affiliation(s)
- Sang Hyeon Woo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Dong Ha Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | | | - Su Jin Kim
- Axceso Biopharma Co., Ltd., Yongin, Republic of Korea
| | - Hae Yeon Jang
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea; Ewha Research Center for Systems Biology, Ewha Womans University, Seoul, Republic of Korea
| | - Jun Young Jang
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Byung Woo Han
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Sang Kim
- Department of Life Science, Ewha Womans University, Seoul, Republic of Korea; Ewha Research Center for Systems Biology, Ewha Womans University, Seoul, Republic of Korea
| | | | | | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea; College of Medicine, Hanyang University, Seoul, Republic of Korea.
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea; College of Medicine, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Qi M, Gao S, Zhang Z, Lang R, Huang Z, Wang J, Qian X, Chen K, Liu H. Secretory breast carcinoma: a multicenter clinicopathologic study of 80 cases with emphasis on prognostic analysis and chemotherapy benefit. Breast Cancer Res Treat 2025; 210:451-461. [PMID: 39730784 DOI: 10.1007/s10549-024-07583-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 12/11/2024] [Indexed: 12/29/2024]
Abstract
PURPOSE To investigate clinicopathologic characteristics and prognosis in secretory breast carcinoma (SBC) and to determine chemotherapy benefits stratified by different subgroups. METHODS SBCs and triple-negative invasive ductal carcinoma patients (TN-IDCs) were enrolled from three cancer centers between January 2011 and December 2020. SBCs were further divided into two subgroups: those with triple negativity (TN-SBCs) and those without (non-TN-SBCs). Clinicopathologic characteristics were thoroughly compared among the three subgroups associated with triple negativity. Kaplan-Meier estimates and Cox proportional hazard models were performed for survival analysis. RESULTS A total of 80 SBCs and 310 TN-IDCs were included in the study. The TN-SBC subgroup consisted of 35 individuals (43.75%) with mild clinical behaviors and a satisfying prognosis in comparison to non-TN-SBCs and TN-IDCs. In SBCs, N stage (N1 vs. N0: HR = 11.176, 95% CI 0.843-148.132, p = 0.067; N2-3 vs. N0: HR = 30.409, 95% CI 1.378-671.169, p = 0.031), LNR (HR = 23.894, 95% CI 1.614-353.835, p = 0.021), and histological grade (HR = 28.634, 95% CI 2.745-298.703, p = 0.005) were significantly correlated with disease-free survival (DFS). Patients in high LNR group receiving chemotherapy achieved a prolonged DFS (p = 0.025), while chemotherapy did not confer a survival benefit in TN-SBCs of our interest (p = 0.12). CONCLUSION TN-SBC is a unique entity with low malignant potential. Advanced N stage, high LNR, and advanced histological grade are adverse determinants of DFS in SBC. Adjuvant chemotherapy provides superior DFS in high LNR SBCs rather than TN-SBCs, hence it is recommended for high LNR SBCs.
Collapse
Affiliation(s)
- Mengyang Qi
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, West Huanhu Road, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Shuang Gao
- Department of Gastrointestinal Surgery, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin, 301800, China
| | - Zhe Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ronggang Lang
- Department of Breast Pathology and Research Lab, Department of Breast Oncology, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Zhidong Huang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, West Huanhu Road, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Jinhui Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, West Huanhu Road, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China
| | - Xiaolong Qian
- Department of Breast Pathology and Research Lab, Department of Breast Oncology, National Clinical Research Center for Cancer, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Kuisheng Chen
- Department of Pathology, Henan Key Laboratory of Tumor Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, West Huanhu Road, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
37
|
Anampa JD, Alvarez Soto A, Bernal AM, Acuna-Villaorduna A. Racial disparities in treatment and outcomes between Hispanic and non-Hispanic black women with triple-negative breast cancer. Breast Cancer Res Treat 2025; 210:307-317. [PMID: 39589609 DOI: 10.1007/s10549-024-07565-7] [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: 09/06/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is an aggressive breast cancer (BC) subtype with higher incidence and mortality rates in non-Hispanic Black (NHB) women than non-Hispanic Whites. Studies assessing disparities between NHB and Hispanic women, the two largest US racial/ethnic minorities, are lacking. This study evaluates disparities in the treatment and outcomes between NHB and Hispanic women with non-metastatic TNBC. METHODS This observational, population-based study using the SEER database included adult, female patients diagnosed with non-metastatic TNBC between 2010 and 2015 and identified as NHB or Hispanic. Logistic regression analysis was used to examine the adjusted odds of receiving breast cancer-directed treatment. Kaplan-Meier and cumulative incidence of death curves were plotted to assess overall survival (OS) and risk of breast cancer-related death, respectively. Multivariate regression analyses with Cox and Fine-Gray methods were calculated to assess factors associated with OS and breast cancer-related death, respectively. RESULTS There were 3426 Hispanic and 5419 NHB patients with non-metastatic TNBC. Hispanics had better 5-year OS relative to NHB (76% vs. 72%). No differences in the odds of receiving chemotherapy or surgery between cohorts was seen. However, the odds of undergoing breast-conserving surgery (BCS) and receiving radiation was higher in NHB than Hispanics, (OR, 1.22; 95% CI, 1.10-1.36) and (OR, 1.50; 95% CI, 1.36-1.66), respectively. Lack of radiation therapy was associated with increased BC-related death in NHB relative to Hispanics (sHR, 1.40; 95% CI, 1.19-1.65). Nevertheless, this difference was not seen when radiation was given, (sHR, 1.03; 95% CI, 0.87-1.23). CONCLUSIONS We found racial disparities in treatment and outcomes between NHB and Hispanics. NHB were more likely to receive radiation therapy and have BCS. Still, after adjusting for demographic and treatment-related factors, NHB had worse OS and BCSS relative to Hispanics. Additional research is needed to understand the drivers of these disparities.
Collapse
Affiliation(s)
- Jesus D Anampa
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, 1695 Eastchester Rd, 2nd Floor, Bronx, NY, 10461, USA.
| | - Alvaro Alvarez Soto
- Department of Medicine, Hematology/Oncology, Carole and Ray Neag Comprehensive Cancer Center, UCONN Health, Farmington, CT, USA
| | - Ana M Bernal
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, 1695 Eastchester Rd, 2nd Floor, Bronx, NY, 10461, USA
| | - Ana Acuna-Villaorduna
- Department of Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
38
|
Su X, Luo Y, Wang Y, Qu P, Liu J, Han S, Ma C, Deng S, Liang Q, Qi X, Cheng P, Hou L. A select inhibitor of MORC2 encapsulated by chimeric membranecoated DNA nanocage target alleviation TNBC progression. Mater Today Bio 2025; 31:101497. [PMID: 39906202 PMCID: PMC11791359 DOI: 10.1016/j.mtbio.2025.101497] [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: 11/14/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant type of breast cancer and lacks effective targeted therapeutic drugs, resulting in a high recurrence rate and worse outcome. In this study, bioinformatic analysis and a series of experiments demonstrated that MOCR2 was highly expressed in TNBC and closely associated with poor prognosis, indicating that MOCR2 may be a potential therapeutic target for TNBC. Subsequently, Angoline was identified as an inhibitor of MORC2 protein by high-throughput screening and can significantly kill the TNBC cells by blocking cell cycle and inducing apoptosis. Furthermore, the biomimetic nanodrug delivery system (PMD) was designed by encapsulating tetrahedral DNA nanostructures with biomimetic cell membrane, and it can efficiently evade the phagocytosis of immune system and target TNBC tissue. Additionally, PMD can markedly enhance the killing effect of Angoline on TNBC tumors. Therefore, PMD-enveloped Angoline provide a highly effective targeted therapeutic regimen for TNBC and may improve the outcome for patients with TNBC.
Collapse
Affiliation(s)
- Xiaohan Su
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Breast Surgery, Mianyang 404 hospital, Mianyang, China
| | - Yunbo Luo
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yali Wang
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Peng Qu
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jun Liu
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shiqi Han
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Cui Ma
- Department of Mathematics, Army Medical University, Chongqing, China
| | - Shishan Deng
- Department of Breast and Thyroid Surgery, Biological Targeting Laboratory of Breast Cancer, Academician (expert) Workstation, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qi Liang
- Department of Laboratory Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaowei Qi
- Department of Breast Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, China
| | - Lingmi Hou
- Breast Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
39
|
Solaimani M, Hosseinzadeh S, Abasi M. Non-coding RNAs, a double-edged sword in breast cancer prognosis. Cancer Cell Int 2025; 25:123. [PMID: 40170036 PMCID: PMC11959806 DOI: 10.1186/s12935-025-03679-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/06/2025] [Indexed: 04/03/2025] Open
Abstract
Cancer is a rising issue worldwide, and numerous studies have focused on understanding the underlying reasons for its occurrence and finding proper ways to defeat it. By applying technological advances, researchers are continuously uncovering and updating treatments in cancer therapy. Their vast functions in the regulation of cell growth and proliferation and their significant role in the progression of diseases, including cancer. This review provides a comprehensive analysis of ncRNAs in breast cancer, focusing on long non-coding RNAs such as HOTAIR, MALAT1, and NEAT1, as well as microRNAs such as miR-21, miR-221/222, and miR-155. These ncRNAs are pivotal in regulating cell proliferation, metastasis, drug resistance, and apoptosis. Additionally, we discuss experimental approaches that are useful for studying them and highlight the advantages and challenges of each method. We then explain the results of these clinical trials and offer insights for future studies by discussing major existing gaps. On the basis of an extensive number of studies, this review provides valuable insights into the potential of ncRNAs in cancer therapy. Key findings show that even though the functions of ncRNAs are vast and undeniable in cancer, there are still complications associated with their therapeutic use. Moreover, there is an absence of sufficient experiments regarding their application in mouse models, which is an area to work on. By emphasizing the crucial role of ncRNAs, this review underscores the need for innovative approaches and further studies to explore their potential in cancer therapy.
Collapse
Affiliation(s)
- Maryam Solaimani
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Sahar Hosseinzadeh
- Faculty of Pharmacy and Medical Biotechnology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mozhgan Abasi
- Immunogenetics Research Center, Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, PO Box: 48175/861, Sari, Iran.
| |
Collapse
|
40
|
Li Y, Wang Y, Jing Y, Zhu Y, Huang X, Wang J, Dilraba E, Guo C. Visualization analysis of breast cancer-related ubiquitination modifications over the past two decades. Discov Oncol 2025; 16:431. [PMID: 40163091 PMCID: PMC11958930 DOI: 10.1007/s12672-025-02032-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Ubiquitination is a type of post-translational modification, referring to the process in which the small molecular protein ubiquitin covalently binds to target proteins under the catalysis of a series of enzymes. The process of ubiquitination is vital in the onset and progression of breast cancer. The use of the ubiquitin-protease system is expected to be a new way to treat human breast cancer. This research aimed to investigate the evolution patterns, key areas of interest, and future directions of ubiquitination in breast cancer via bibliometric analysis. METHODS Research articles on ubiquitination modifications in breast cancer were sourced from the Web of Science Core Collection database and analyzed via Microsoft Excel 2021, Bibliometrix, VOSviewer, and Citespace software for thorough bibliometrics. RESULTS From 2005-2024, 1850 English articles published in 405 journals by 1842 institutions/universities from 61 countries were included in the study. Keywords, research fields, co-cited literature and other information were included. Research on ubiquitination modifications has focused on breast cancer, expression, protein, activation, degradation, ubiquitination, phosphorylation, etc. Notably, the keywords that broke out in the past five years have focused on "triple-negative breast cancer", "promotion", and "metabolism". These findings suggest that key areas of current research are metabolism, immunity, survival, and prognosis in triple-negative breast cancer. CONCLUSIONS Our findings indicate that research on triple-negative breast cancer, as well as its immunological and metabolic aspects, is a burgeoning and promising area. Our work offers valuable guidance and fresh perspectives on the relationship between breast cancer and ubiquitin modification.
Collapse
Affiliation(s)
- Yongxiang Li
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yiyang Wang
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Yubo Jing
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Youseng Zhu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Xinzhu Huang
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - JunYi Wang
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Elihamu Dilraba
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Chenming Guo
- Department of Breast Surgery, Center of Digestive and Vascular Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China.
| |
Collapse
|
41
|
Cieslak SG, Shahbazi R. Gamma delta T cells and their immunotherapeutic potential in cancer. Biomark Res 2025; 13:51. [PMID: 40148988 PMCID: PMC11951843 DOI: 10.1186/s40364-025-00762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Gamma-delta (γδ) T cells are a unique subset of T lymphocytes that play diverse roles in immune responses, bridging innate and adaptive immunity. With growing interest in their potential for cancer immunotherapy, a comprehensive and inclusive exploration of γδ T cell families, their development, activation mechanisms, functions, therapeutic implications, and current treatments is essential. This review aims to provide an inclusive and thorough discussion of these topics. Through our discussion, we seek to uncover insights that may harbinger innovative immunotherapeutic strategies. Beginning with an overview of γδ T cell families including Vδ1, Vδ2, and Vδ3, this review highlights their distinct functional properties and contributions to anti-tumor immunity. Despite γδ T cells exhibiting both anti-tumor and pro-tumor activities, our review elucidates strategies to harness the anti-tumor potential of γδ T cells for therapeutic benefit. Moreover, our paper discusses the structural intricacies of the γδ T cell receptor and its significance in tumor recognition. Additionally, this review examines conventional and emerging γδ T cell therapies, encompassing both non-engineered and engineered approaches, with a focus on their efficacy and safety profiles in clinical trials. From multifunctional capabilities to diverse tissue distribution, γδ T cells play a pivotal role in immune regulation and surveillance. By analyzing current research findings, this paper offers insights into the dynamic landscape of γδ T cell-based immunotherapies, underscoring their promise as a potent armamentarium against cancer. Furthermore, by dissecting the complex biology of γδ T cells, we learn valuable information about the anti-cancer contributions of γδ T cells, as well as potential targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Stephen G Cieslak
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA.
- Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Brown Center for Immunotherapy, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
42
|
Battogtokh G, Akala EO. Development of Multifunctional Targeted Dual-Loaded Polymeric Nanoparticles for Triple-Negative Breast Cancer Treatment. Pharmaceutics 2025; 17:425. [PMID: 40284424 PMCID: PMC12030066 DOI: 10.3390/pharmaceutics17040425] [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: 03/04/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Triple-negative breast cancer (TNBC) is a subtype of breast cancer that accounts for 15-20% of all breast cancer cases. TNBC is very difficult to treat with conventional treatment modalities such as chemotherapy, radiotherapy, and surgery; Methods: In this study, we developed a dual-loaded targeted nanotherapeutics against TNBC to solve the challenging problems associated with TNBC treatment: lack of efficacy, toxicity, and poor site-specific drug delivery; PEGylated methacrylate-polylactide copolymer containing cisplatin was synthesized and characterized; Results: The copolymer was used to fabricate nanoparticles (NPs) in the presence of paclitaxel with 1.33% drug loading. The nanoparticles were homogenous, with an average particle size of 198 nm and a negative zeta potential (-41.3 mV). Cetuximab (CTX), a monoclonal antibody that binds to the epidermal growth factor receptor (EGFR), was attached to the NP's surface to enhance the targetability to TNBC. In vitro studies including cell uptake and cytotoxicity in MDA-MB-231 cells confirmed that CTX-targeted NPs have the potential for treating TNBC. The IC50 of CTX-NPs after 96 h of incubation was 0.1 μM, which was significantly lower than those of p-NPs (0.49 μM) and free drugs (PTX + cPt: 0.57 μM); Conclusions: In summary, this research shows that CTX-targeted polymeric NPs containing cisplatin and paclitaxel are effective in treating TNBC in vivo investigations are ongoing.
Collapse
Affiliation(s)
| | - Emmanuel O. Akala
- Center for Drug Research and Development, Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA;
| |
Collapse
|
43
|
Pinto RJ, Ferreira D, Salamanca P, Miguel F, Borges P, Barbosa C, Costa V, Lopes C, Santos LL, Pereira L. Coding and regulatory somatic profiling of triple-negative breast cancer in Sub-Saharan African patients. Sci Rep 2025; 15:10325. [PMID: 40133516 PMCID: PMC11937512 DOI: 10.1038/s41598-025-94707-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
The burden of triple-negative breast cancer (TNBC) may be shaped by genetic factors, particularly inherited and somatic mutation profiles. However, data on this topic remain limited, especially for the African continent, where a higher TNBC incidence is observed. In the age of precision medicine, cataloguing TNBC diversity in African patients becomes imperative. We performed whole exome sequencing, including untranslated regions, on 30 samples from Angola and Cape Verde, which allowed to ascertain on potential regulatory mutations in TNBC for the first time. A high somatic burden was observed for the African cohort, with 86% of variants being so far unreported. Recurring to predictive functional algorithms, 17% of the somatic single nucleotide variants were predicted to be deleterious at the protein level, and 20% overlapped with candidate cis-regulatory elements controlling gene expression. Several of these somatic functionally-impactful mutations and copy number variation (mainly in 1q, 8q, 6 and 10p) occur in known BC- and all cancer-driver genes, enriched for several cancer mechanisms, including response to radiation and related DNA repair mechanisms. TP53 is the top of these known BC-driver genes, but our results identified possible novel TNBC driver genes that may play a main role in the African context, as TTN, CEACAM7, DEFB132, COPZ2 and GAS1. These findings emphasize the need to expand cancer omics screenings across the African continent, the region of the globe with highest genomic diversity, accelerating the discovery of new somatic mutations and cancer-related pathways.
Collapse
Affiliation(s)
- Ricardo J Pinto
- i3S, Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP, Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Dylan Ferreira
- Research Center of IPO-Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal
| | | | | | - Pamela Borges
- Hospital Universitário Agostinho Neto, Praia, Cabo Verde
| | - Carla Barbosa
- Hospital Universitário Agostinho Neto, Praia, Cabo Verde
| | - Vitor Costa
- Hospital Universitário Agostinho Neto, Praia, Cabo Verde
| | - Carlos Lopes
- Unilabs | Laboratório Anatomia Patológica, Porto, Portugal
| | - Lúcio Lara Santos
- Research Center of IPO-Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal
- FP-I3ID, University Fernando Pessoa, Porto, Portugal
- Department of Surgical Oncology, Portuguese Oncology Institute of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences, University Fernando Pessoa, Gondomar, Portugal
| | - Luisa Pereira
- i3S, Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal.
- IPATIMUP, Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal.
| |
Collapse
|
44
|
Nava-Ochoa A, Mertens-Talcott SU, Talcott ST, Noratto GD. Dark Sweet Cherry ( Prunus avium L.) Juice Phenolics Rich in Anthocyanins Exhibit Potential to Inhibit Drug Resistance Mechanisms in 4T1 Breast Cancer Cells via the Drug Metabolism Pathway. Curr Issues Mol Biol 2025; 47:213. [PMID: 40136467 PMCID: PMC11941269 DOI: 10.3390/cimb47030213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025] Open
Abstract
Anthocyanins (ACNs) from dark sweet cherries (DSCs) have shown efficacy against breast cancer (BC) cells, particularly triple-negative breast cancer (TNBC) cells, without affecting normal breast cells. This study investigated the impact of ACNs on TNBC cells, focusing on drug resistance mechanisms involving drug metabolism and transport enzymes. Specifically, it was examined whether ACNs influenced Doxorubicin (DOX) metabolism by targeting drug metabolism enzymes (phase I metabolism) and drug transport enzymes (phase III metabolism) in TNBC cells. 4T1 TNBC cells were treated with ACNs, DOX, and the combination of both (ACN-DOX). Results showed a synergistic inhibition of cell viability by ACNs and DOX. In addition, the modulation of phase I drug-metabolizing enzymes was exerted by ACNs, reducing the activity of cytochrome P450 (CYP) enzymes induced by DOX. A reduction of drug efflux by ACNs was shown by decreasing P-glycoprotein (P-gp) activity, leading to a higher intracellular accumulation of DOX. These effects were confirmed using CYP and P-gp inducers and inhibitors, showing their impact on cell viability. In conclusion, the combination of ACNs with DOX has the potential to lower DOX doses, enhance its efficacy, and possibly reduce side effects, offering a promising approach for TNBC treatment.
Collapse
Affiliation(s)
| | | | | | - Giuliana D. Noratto
- Department of Food Science and Technology, Texas A&M University, College Station, TX 77843, USA; (A.N.-O.); (S.U.M.-T.); (S.T.T.)
| |
Collapse
|
45
|
Jiang R, Yang L, Liu X, Xu Y, Han L, Chen Y, Gao G, Wang M, Su T, Li H, Fang L, Sun N, Du H, Zheng J, Wang G. Genetically engineered macrophages reverse the immunosuppressive tumor microenvironment and improve immunotherapeutic efficacy in TNBC. Mol Ther 2025:S1525-0016(25)00198-4. [PMID: 40119517 DOI: 10.1016/j.ymthe.2025.03.024] [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: 09/20/2024] [Revised: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 03/24/2025] Open
Abstract
The main challenges in current immunotherapy for triple-negative breast cancer (TNBC) lie in the immunosuppressive tumor microenvironment (TME). Considering tumor-associated macrophages (TAMs) are the most abundant immune cells in the TME, resetting TAMs is a promising strategy for ameliorating the immunosuppressive TME. Here, we developed genetically engineered macrophages (GEMs) with gene-carrying adenoviruses, to maintain the M1-like phenotype and directly deliver the immune regulators interleukin-12 and CXCL9 into local tumors, thereby reversing the immunosuppressive TME. In tumor-bearing mice, GEMs demonstrated targeted enrichment in tumors and successfully reprogramed TAMs to M1-like macrophages. Moreover, GEMs significantly enhanced the accumulation, proliferation, and activation of CD8+ T cells, mature dendritic cells, and natural killer cells within tumors, while diminishing M2-like macrophages, immunosuppressive myeloid-derived suppressor cells, and regulatory T cells. This treatment efficiently suppressed tumor growth. In addition, combination therapy with GEMs and anti-programmed cell death protein 1 further improved interferon-γ+CD8+ T cell percentages and tumor inhibition efficacy in an orthotopic murine TNBC model. Therefore, this study provides a novel strategy for reversing the immunosuppressive TME and improving immunotherapeutic efficacy through live macrophage-mediated gene delivery.
Collapse
Affiliation(s)
- Ranran Jiang
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Liechi Yang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Xin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Department of Urology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China
| | - Yujun Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuxin Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Tong Su
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Nan Sun
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Hongwei Du
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Department of Oncology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
46
|
Ye Z, Yuan J, Hong D, Xu P, Liu W. Multimodal diagnostic models and subtype analysis for neoadjuvant therapy in breast cancer. Front Immunol 2025; 16:1559200. [PMID: 40170854 PMCID: PMC11958217 DOI: 10.3389/fimmu.2025.1559200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Breast cancer, a heterogeneous malignancy, comprises multiple subtypes and poses a substantial threat to women's health globally. Neoadjuvant therapy (NAT), administered prior to surgery, is integral to breast cancer treatment strategies. It aims to downsize tumors, optimize surgical outcomes, and evaluate tumor responsiveness to treatment. However, accurately predicting NAT efficacy remains challenging due to the disease's complexity and the diverse responses across different molecular subtypes. Methods In this study, we harnessed multimodal data, including proteomic, genomic, MRI imaging, and clinical information, sourced from multiple cohorts such as I-SPY2, TCGA-BRCA, GSE161529, and METABRIC. Post data preprocessing, Lasso regression was utilized for feature extraction and selection. Five machine learning algorithms were employed to construct diagnostic models, with pathological complete response (pCR) as the predictive endpoint. Results Our results revealed that the multi-omics Ridge regression model achieved the optimal performance in predicting pCR, with an AUC of 0.917. Through unsupervised clustering using the R package MOVICS and nine clustering algorithms, we identified four distinct multimodal breast cancer subtypes associated with NAT. These subtypes exhibited significant differences in proteomic profiles, hallmark cancer gene sets, pathway activities, tumor immune microenvironments, transcription factor activities, and clinical characteristics. For instance, CS1 subtype, predominantly ER-positive, had a low pCR rate and poor response to chemotherapy drugs, while CS4 subtype, characterized by high immune infiltration, showed a better response to immunotherapy. At the single-cell level, we detected significant heterogeneity in the tumor microenvironment among the four subtypes. Malignant cells in different subtypes displayed distinct copy number variations, differentiation levels, and evolutionary trajectories. Cell-cell communication analysis further highlighted differential interaction patterns among the subtypes, with implications for tumor progression and treatment response. Conclusion Our multimodal diagnostic model and subtype analysis provide novel insights into predicting NAT efficacy in breast cancer. These findings hold promise for guiding personalized treatment strategies. Future research should focus on experimental validation, in-depth exploration of the underlying mechanisms, and extension of these methods to other cancers and treatment modalities.
Collapse
Affiliation(s)
- Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
47
|
Ding R, Tang L, Zeng D, Li J, Jia Y, Yan X, Zhang C, Wu L. Discovery of novel JQ1 derivatives as dual ferroptosis and apoptosis inducers for the treatment of triple-negative breast cancer. Eur J Med Chem 2025; 286:117275. [PMID: 39826487 DOI: 10.1016/j.ejmech.2025.117275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
The activation of ferroptosis in refractory cancers may enhances their sensitivity to apoptosis-based chemotherapy, resulting in a synergistic effect via combination therapy. To enhance the anticancer effect of JQ1, a known BRD4 inhibitor with a significant antiproliferative effect on triple-negative breast cancer (TNBC), various new JQ1 derivatives as dual ferroptosis and apoptosis inducers were designed and synthesized. Among them, compound BG11 revealed a remarkable inhibitory activity against TNBC cells and obviously suppressed BRD4 and GPX4 expression and activities. Further studies suggested that BG11 induced cell ferroptosis through promoting Fe2+ and intracellular lipid peroxide deposition. In addition, BG11 could induce apoptosis through increasing Bax (apoptotic protein) expression and decreasing Bcl-2 (anti-apoptotic protein) expression within MDA-MB-231 cells. Surprisingly, BG11 significantly inhibited tumor proliferation in the MDA-MB-231 xenograft model without obvious toxicity. Based on the above findings, BG11 may be the candidate dual ferroptosis and apoptosis inducers for treating TNBC.
Collapse
Affiliation(s)
- Ran Ding
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Lijie Tang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Dexin Zeng
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China; Quanzhou Hospital of Traditional Chinese Medicine, Quanzhou, 362000, China
| | - Jian Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yingdong Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiqing Yan
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
48
|
Xu Z, Zhou Z, Son JB, Feng H, Adrada BE, Moseley TW, Candelaria RP, Guirguis MS, Patel MM, Whitman GJ, Leung JWT, Le-Petross HTC, Mohamed RM, Panthi B, Lane DL, Chen H, Wei P, Tripathy D, Litton JK, Valero V, Huo L, Hunt KK, Korkut A, Thompson A, Yang W, Yam C, Rauch GM, Ma J. Deep Learning Models Based on Pretreatment MRI and Clinicopathological Data to Predict Responses to Neoadjuvant Systemic Therapy in Triple-Negative Breast Cancer. Cancers (Basel) 2025; 17:966. [PMID: 40149299 PMCID: PMC11940201 DOI: 10.3390/cancers17060966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
PURPOSE To develop deep learning models for predicting the pathologic complete response (pCR) to neoadjuvant systemic therapy (NAST) in patients with triple-negative breast cancer (TNBC) based on pretreatment multiparametric breast MRI and clinicopathological data. METHODS The prospective institutional review board-approved study [NCT02276443] included 282 patients with stage I-III TNBC who had multiparametric breast MRI at baseline and underwent NAST and surgery during 2016-2021. Dynamic contrast-enhanced MRI (DCE), diffusion-weighted imaging (DWI), and clinicopathological data were used for the model development and internal testing. Data from the I-SPY 2 trial (2010-2016) were used for external testing. Four variables with a potential impact on model performance were systematically investigated: 3D model frameworks, tumor volume preprocessing, tumor ROI selection, and data inputs. RESULTS Forty-eight models with different variable combinations were investigated. The best-performing model in the internal testing dataset used DCE, DWI, and clinicopathological data with the originally contoured tumor volume, the tight bounding box of the tumor mask, and ResNeXt50, and achieved an area under the receiver operating characteristic curve (AUC) of 0.76 (95% CI: 0.60-0.88). The best-performing models in the external testing dataset achieved an AUC of 0.72 (95% CI: 0.57-0.84) using only DCE images (originally contoured tumor volume, enlarged bounding box of tumor mask, and ResNeXt50) and an AUC of 0.72 (95% CI: 0.56-0.86) using only DWI images (originally contoured tumor volume, enlarged bounding box of tumor mask, and ResNet18). CONCLUSIONS We developed 3D deep learning models based on pretreatment data that could predict pCR to NAST in TNBC patients.
Collapse
Affiliation(s)
- Zhan Xu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Z.X.)
| | - Zijian Zhou
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Z.X.)
| | - Jong Bum Son
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Z.X.)
| | - Haonan Feng
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Beatriz E. Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Tanya W. Moseley
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Rosalind P. Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Mary S. Guirguis
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Miral M. Patel
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Gary J. Whitman
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Jessica W. T. Leung
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Huong T. C. Le-Petross
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Rania M. Mohamed
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Bikash Panthi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Z.X.)
| | - Deanna L. Lane
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Huiqin Chen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Jennifer K. Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Kelly K. Hunt
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Anil Korkut
- Department of Bioinformatics & Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Alastair Thompson
- Section of Breast Surgery, Baylor College of Medicine, 7200 Cambridge St., Houston, TX 77030, USA
| | - Wei Yang
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Gaiane M. Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Jingfei Ma
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (Z.X.)
| |
Collapse
|
49
|
Punie K, Kurian AW, Ntalla I, Sjekloca N, Estrin A, Dabrowski EC, Lai C, Hurvitz S. Unmet need for previously untreated metastatic triple-negative breast cancer: a real-world study of patients diagnosed from 2011 to 2022 in the United States. Oncologist 2025; 30:oyaf034. [PMID: 40163689 PMCID: PMC11957248 DOI: 10.1093/oncolo/oyaf034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND This real-world study describes the treatment landscape evolution after targeted therapy approval and associated survival outcomes for previously untreated metastatic triple-negative breast cancer (mTNBC) in the United States. PATIENTS AND METHODS This retrospective analysis used de-identified electronic health record-derived data of patients diagnosed with mTNBC (January 2011-July 2022; index date was first-line [1L] treatment start date). Patient characteristics, treatment patterns, real-world overall survival (rwOS), and time to next treatment or death (TTNTD) were determined. Outcomes before (2011-2017, early cohort) and after (2018-2022, late cohort) targeted therapy approval were evaluated. RESULTS Among 2004 eligible patients, 21% were classified as Black, 13% had Eastern Cooperative Oncology Group performance status ≥2, and 63% were diagnosed with recurrent disease; median age was 60 years. First-line chemotherapy-only (single- and multiple-agent chemotherapy) use decreased with the introduction of targeted therapies from 96% before 2018 to 65% between 2019 and 2022. From 2019, 33% of patients received programmed death-(ligand) 1 inhibitor-based regimen; ~2% received poly (ADP-ribose) polymerase inhibitors. Median 1L treatment duration was 2.6 months and this did not change over time. Of all 1L patients, 34% died before second-line (2L) and 51% subsequently received 2L treatment. Median (95% CI) 1L rwOS and TTNTD were 11.3 (10.7-12.0) months and 4.3 (4.1-4.6) months, respectively. Median 1L 5-year survival [95% CI] showed statistically significant but small improvement from the early (10.9 [10.3-11.6] months) to late cohort (11.9 [10.7-13.1] months; HR [95% CI], 0.87 [0.78-0.96]). CONCLUSION This analysis demonstrated that, despite changes in care over time, survival improvements were not clinically meaningful; thus, a substantial unmet need for more efficacious treatments in previously untreated patients with mTNBC remains.
Collapse
Affiliation(s)
- Kevin Punie
- Department of Medical Oncology, Oncology Centre Antwerp, Ziekenhuis aan de Stroom, Antwerp, 2610, Belgium
| | - Allison W Kurian
- Departments of Medicine and of Epidemiology and Population Health, Stanford University, Stanford, CA, 94305, United States
| | - Ioanna Ntalla
- Gilead Sciences Europe, LTD, Uxbridge, UB11 1AF, United Kingdom
| | | | | | | | - Catherine Lai
- Gilead Sciences, Inc., Foster City, CA 94404, United States
| | - Sara Hurvitz
- Clinical Research Division, Fred Hutchinson Cancer Center; Department of Medicine/Division of Hematology Oncology, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
50
|
Zhang Y, Chen H, Mo H, Zhao N, Sun X, Liu B, Gao R, Xu B, Zhang Z, Liu Z, Ma F. Distinct cellular mechanisms underlie chemotherapies and PD-L1 blockade combinations in triple-negative breast cancer. Cancer Cell 2025; 43:446-463.e7. [PMID: 39919737 DOI: 10.1016/j.ccell.2025.01.007] [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: 06/01/2024] [Revised: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Combining immune checkpoint blockade (ICB) with chemotherapy shows promise for treating triple-negative breast cancer (TNBC), though the mechanisms remain incompletely understood. Here, we integrate published and new single-cell RNA sequencing (scRNA-seq) data to investigate the tumor immune microenvironment (TIME) in TNBC patients treated with paclitaxel (PTX), nab-paclitaxel (Nab-PTX), and their combinations with the anti-PD-L1 antibody atezolizumab (ATZ). Compared to ATZ plus PTX, ATZ plus Nab-PTX rewires TCF7+ stem-like effector memory CD8+ T cells (Tsem) and CD4+ T follicular helper (Tfh) cells. Nab-paclitaxel, unlike PTX, also reshapes the myeloid compartment, expanding mast cells and pro-inflammatory macrophages. Our analyses in human TNBC and murine models underscore the crucial role of mast cells in orchestrating anti-tumor immune responses, likely by promoting the recruitment and activation of T and B cells. In vivo experiments demonstrate that activating mast cells alongside PD-L1 blockade attenuates TNBC progression, suggesting mast cells as a promising adjunct for enhancing ICB therapy efficacy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Hongyan Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hongnan Mo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing, ChaoYang District, Beijing 100005, China
| | - Baolin Liu
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ranran Gao
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Binghe Xu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Fei Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|