1
|
Earla JR, Kponee-Shovein K, Kurian AW, Mahendran M, Song Y, Hua Q, Hilts A, Sun Y, Hirshfield KM, Mejia JA. Real-world perioperative treatment patterns and economic burden of recurrence in early-stage HER2-negative breast cancer: a SEER-Medicare study. J Med Econ 2025; 28:54-69. [PMID: 39648858 DOI: 10.1080/13696998.2024.2439228] [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: 10/16/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/10/2024]
Abstract
AIM This study aimed to describe treatment patterns and quantify the economic impact of recurrence in early-stage human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC). MATERIALS & METHODS Medicare beneficiaries with stages I-III HER2-negative BC and lumpectomy or partial/total mastectomy were identified from SEER-Medicare data (2010-2019). Perioperative therapies were reported in the neoadjuvant and adjuvant setting. Locoregional recurrence and distant metastasis were identified using a claims-based algorithm developed with clinical input and consisting of a diagnosis-based and treatment-based indicator. All-cause and BC-related healthcare resource utilization (HRU) per-patient-month and monthly healthcare costs were estimated from the recurrence date for patients with recurrence and from an imputed index date for patients without recurrence using frequency matching. HRU and costs were compared between groups stratified by hormone receptor-positive (HR+) or triple negative BC (TNBC) using multivariable regression models. RESULTS Of 28,655 patients, 8.5% experienced recurrence, 90.4% had HR+ disease, and 5.6% received neoadjuvant therapy. Relative to patients without recurrence, patients with recurrence had more advanced disease (stage II/III: 73.7% vs. 34.0%) and higher-grade tumors (Grade 3/4: 40.6% vs. 18.0%) at diagnosis. Recurrence in HR+/HER2-negative BC and TNBC was associated with higher rates of all-cause hospitalizations (incidence rate ratio [IRR]: 2.84 and 3.65), emergency department (ED) visits (IRR: 1.75 and 2.00), and outpatient visits (IRR: 1.46 and 1.55; all p < 0.001). Similarly, recurrence was associated with higher rates of BC-related HRU, particularly for ED visits in HR+/HER2-negative BC (IRR: 4.24; p < 0.001) and hospitalizations in TNBC (IRR: 11.71; p < 0.001). Patients with HR+/HER2-negative BC and TNBC recurrence incurred higher monthly all-cause (cost difference [CD]: $3988 and $4651) and BC-related healthcare costs (CD: $3743 and $5819). CONCLUSIONS Our findings highlight the considerable economic burden of recurrence in early-stage HER2-negative BC and underscore the unmet need for optimization of therapies that reduce recurrence in this population.
Collapse
Affiliation(s)
| | | | | | | | - Yan Song
- Analysis Group, Inc, Boston, MA, USA
| | - Qi Hua
- Analysis Group, Inc, Boston, MA, USA
| | | | | | | | | |
Collapse
|
2
|
Gui A, Cao X, Meng F, Chen Y, Ma S, Chen H. Protein lactylation within the nucleus independently predicts the prognosis of non‑specific triple‑negative breast cancer. Oncol Lett 2025; 29:72. [PMID: 39628828 PMCID: PMC11612721 DOI: 10.3892/ol.2024.14818] [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: 08/07/2024] [Accepted: 11/05/2024] [Indexed: 12/06/2024] Open
Abstract
Protein lactylation represents a pervasive post-translational modification prevalent in histones and diverse proteins, fostering tumor initiation and progression. Nonetheless, the impact of protein lactylation on the prognosis of non-specific triple-negative breast cancer (TNBC) remains uncertain. In the present study, the pan-lysine lactylation (panKlac) levels in cytoplasmic and nuclear compartments were semi-quantitatively examined using a tissue microarray encompassing 77 non-specific TNBC tissues. The association of the prognosis of patients with the panKlac levels in the cytoplasmic and nuclear compartments or other tumor attributes was assessed using Kaplan-Meier and Cox regression analyses. Furthermore, the molecular pathways involved in the promotional effect of lactylation on cell proliferation were determined through a transcriptomic analysis. The results indicated that the panKlac levels were markedly higher in tumor tissues than in para-tumor mammary regions and showed no significant correlations with various clinicopathological parameters, such as tumor dimension, lymph node involvement or histological grading. Notably, high panKlac levels within the nucleus served as an independent predictor of recurrence-free survival, whereas high cytoplasmic panKlac levels were a protective factor for patient survival. The panKlac levels were also markedly elevated in the TNBC cell line, MDA-MB-231. Additionally, glycolysis inhibition significantly reduced the global panKlac levels and concurrently diminished cell proliferation. According to the comprehensive transcriptomic analysis results, pathways related to ribosomal subunit biosynthesis/assembly and aminoacyl-tRNA biosynthesis were involved in the tumor-promoting mechanisms of lactylation. Further results revealed the oncogenic propensity of tyrosyl-tRNA synthetase 1 (YARS1) and its association with lactate production. Overall, Klac levels within the nucleus are an independent prognostic indicator for patients with non-specific TNBC. It is imperative to delve deeper into the roles and mechanisms of nuclear protein lactylation and YARS1 in non-specific TNBC.
Collapse
Affiliation(s)
- Anping Gui
- Breast Center, People's Hospital of Zhongshan City, Zhongshan, Guangdong 528400, P.R. China
| | - Xiaoshan Cao
- Department of Pathology, People's Hospital of Zhongshan City, Zhongshan, Guangdong 528400, P.R. China
| | - Fengjiao Meng
- Department of Pathology, People's Hospital of Zhongshan City, Zhongshan, Guangdong 528400, P.R. China
| | - Yingzhi Chen
- Department of Pathology, People's Hospital of Zhongshan City, Zhongshan, Guangdong 528400, P.R. China
| | - Shihui Ma
- Breast Center, People's Hospital of Zhongshan City, Zhongshan, Guangdong 528400, P.R. China
| | - Hong Chen
- Department of Oncological Surgery, People's Hospital of Zhongshan City, Zhongshan, Guangdong 528400, P.R. China
| |
Collapse
|
3
|
Zhang J, Lu L, Zhang W, Miao Y, Du H, Xia H, Tao Z, Du Z, Tang Y, Fang Q. Gadolinium ion-loaded mesoporous organosilica nanoplatform for enhanced radiotherapy in breast tumor treatment. Colloids Surf B Biointerfaces 2025; 246:114374. [PMID: 39541910 DOI: 10.1016/j.colsurfb.2024.114374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype with limited therapeutic options, often exhibiting resistance to standard radiotherapy (RT) and chemotherapy. Recent advancements in nanomedicine provide an opportunity to enhance treatment efficacy through innovative drug delivery systems and radiosensitizers. In this study, we present a novel nanotheranostic platform, MOs-G@DOX, engineered to enhance the therapeutic efficacy of RT in the treatment of TNBC. This platform consists of gadolinium-containing mesoporous organosilica nanoparticles (MOs-G) that serve a dual function as a drug carrier and a radiosensitizer. The MOs-G were synthesized via a surfactant-mediated sol-gel process, followed by gadolinium incorporation through nanoprecipitation. The antitumor drug doxorubicin (DOX) was subsequently loaded into the mesoporous structure, forming the MOs-G@DOX nanoplatform. Comprehensive in vitro and in vivo studies demonstrated that MOs-G@DOX exhibits excellent biocompatibility and significantly enhances the radiosensitivity of TNBC cells, leading to superior tumor growth inhibition compared to conventional treatments. The stability of MOs-G, with minimal gadolinium ion leakage, further underscores its potential as a safe and effective nanomedicine. Additionally, the combination of MOs-G@DOX with RT showed a marked increase in reactive oxygen species (ROS) generation and tumor cell apoptosis, which were confirmed through histological analyses. These findings suggest that MOs-G@DOX is a promising candidate for advancing cancer therapy, particularly in the context of RT for TNBC.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| | - Li Lu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Wenqing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Yuchen Miao
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hengda Du
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hui Xia
- Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Zhiyong Tao
- Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Zhaofeng Du
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Yulong Tang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Qiang Fang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| |
Collapse
|
4
|
Wang T, Chen T, Li D, Hang X, Zhang S, Yi H, Jiang T, Ding D, Zhang X. Core-shell vector-mediated co-delivery of CRISPR/Cas9 system and hydrophobic drugs against triple-negative breast cancer stem cells. J Control Release 2025; 378:1080-1091. [PMID: 39733911 DOI: 10.1016/j.jconrel.2024.12.062] [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: 09/26/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 12/31/2024]
Abstract
Cancer stem cells (CSCs) play an important role in the development of triple-negative breast cancer (TNBC), including metastasis, invasion, tumorigenicity, and drug resistance. Moreover, non-CSCs can spontaneously transform into CSCs in special tumor microenvironments, thereby leading to poor prognosis or even failed treatments. Therefore, reversing CSCs into normal tumor cells in a sustained-acting manner is a promising strategy. It has been reported that down-regulation of FBXO44 protein expression inhibits tumor cell stemness. Moreover, CRISPR/Cas9 technology, a well-known precise gene editing tool, was adopted to permanently block FBXO44 within the genome upon its successful implementation. Given this, a core-shell nanoparticle (NP) consisting of amphiphilic polymer core and crosslinked-hyaluronic acid shell (nDOX-PL/pFBXO44 NPs) is developed in this work to concurrently deliver FBXO44-targeted CRISPR/Cas9 plasmids (pFBXO44) and doxorubicin (DOX) for combinational CSC reprogramming and chemotherapy of TNBC, which exhibits tumor cell targeting, endosomal escape, and reduction responsiveness to release DOX and plasmids in the cytoplasma. CRISPR/Cas9-mediated downregulation of FBXO44 expression could convert CSC into normal tumor cells, and effectively inhibit tumor growth without obvious side effects in vivo after combining with chemotherapy. In summary, we developed an intelligent system to co-deliver genetic and hydrophobic drugs, achieving effective cancer stemness reversal and synergistic suppression of contractable TNBC.
Collapse
Affiliation(s)
- Tong Wang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, DuShuHu High Education Zone, Suzhou, Jiangsu Province 215123, China
| | - Tianyi Chen
- College of Life and Health, Nanjing Polytechnic Institute, 625 Geguan Road, Nanjing, Jiangsu Province 210048, China
| | - Dazhao Li
- Clinical Medical Research Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xiaoxing Hang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, DuShuHu High Education Zone, Suzhou, Jiangsu Province 215123, China
| | - Shuangshuang Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, DuShuHu High Education Zone, Suzhou, Jiangsu Province 215123, China
| | - Han Yi
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China.
| | - Dawei Ding
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| | - Xuenong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, DuShuHu High Education Zone, Suzhou, Jiangsu Province 215123, China.
| |
Collapse
|
5
|
Bhutta ZA, Choi KC. Canine mammary tumors as a promising adjunct preclinical model for human breast cancer research: similarities, opportunities, and challenges. Arch Pharm Res 2025:10.1007/s12272-024-01524-y. [PMID: 39752109 DOI: 10.1007/s12272-024-01524-y] [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: 07/23/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Despite significant progress in the field of human breast cancer research and treatment, there is a consistent increase in the incidence rate of 0.5 percent annually, posing challenges in the development of effective novel therapeutic strategies. The failure rate of drugs in clinical trials stands at approximately 95%, primarily attributed to the limitations and lack of reliability of existing preclinical models, such as mice, which do not mimic human tumor biology. This article examines the potential utility of canine mammary tumors as an adjunct preclinical model for investigating human breast cancer. Given the numerous similarities between canine and human breast cancer, canines present a promising alternative model. The discussion delves into the intricate molecular and clinical aspects of human breast cancer and canine mammary tumors, shedding light on the tumors' molecular profiles, identifying specific molecular markers, and the application of radiological imaging modalities. Furthermore, the manuscript addresses the current constraints of preclinical cancer studies, the benefits of using canines as models, and the obstacles linked to the canine mammary tumors model. By concentrating on these elements, this review aims to highlight the viability of canine models in enhancing our understanding and management of human breast cancer.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
6
|
Zhao G, Liu Y, Yin S, Cao R, Zhao Q, Fu Y, Du Y. FOSL1 transcriptionally dictates the Warburg effect and enhances chemoresistance in triple-negative breast cancer. J Transl Med 2025; 23:1. [PMID: 39748430 PMCID: PMC11697476 DOI: 10.1186/s12967-024-06014-9] [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: 10/23/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Dysregulated energy metabolism has emerged as a defining hallmark of cancer, particularly evident in triple-negative breast cancer (TNBC). Distinct from other breast cancer subtypes, TNBC exhibits heightened glycolysis and aggressiveness. However, the transcriptional mechanisms of aerobic glycolysis in TNBC remains poorly understood. METHODS The Cancer Genome Atlas (TCGA) cohort was utilized to identify genes associated with glycolysis. The role of FOSL1 in glycolysis and tumor growth in TNBC cells was confirmed through both loss-of-function and gain-of-function experiments. The subcutaneous xenograft model was established to evaluate the therapeutic potential of targeting FOSL1 in TNBC. Additionally, chromatin immunoprecipitation and luciferase reporter assays were employed to investigate the transcriptional regulation of glycolytic genes mediated by FOSL1. RESULTS FOSL1 is identified as a pivotal glycolysis-related transcription factor in TNBC. Functional verification shows that FOSL1 enhances the glycolytic metabolism of TNBC cells, as evidenced by glucose uptake, lactate production, and extracellular acidification rates. Notably, FOSL1 promotes tumor growth in TNBC in a glycolysis-dependent manner, as inhibiting glycolysis with 2-Deoxy-D-glucose markedly diminishes the oncogenic effects of FOSL1 in TNBC. Mechanistically, FOSL1 transcriptionally activates the expression of genes such as SLC2A1, ENO1, and LDHA, which further accelerate the glycolytic flux. Moreover, FOSL1 is highly expressed in doxorubicin (DOX)-resistant TNBC cells and clinical samples from cases of progressive disease following neoadjuvant chemotherapy. Targeting FOSL1 proves effective in overcoming chemoresistance in DOX-resistant MDA-MB-231 cells. CONCLUSION In summary, FOSL1 establishes a robust link between aerobic glycolysis and carcinogenesis, positioning it as a promising therapeutic target, especially in the context of TNBC chemotherapy.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun City, Jilin Province, P.R. China
| | - Yutong Liu
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun City, Jilin Province, P.R. China
| | - Shiqi Yin
- Anhui University of Science and Technology Affiliated Fengxian Hospital, Shanghai, China
| | - Runxiang Cao
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun City, Jilin Province, P.R. China
| | - Qian Zhao
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun City, Jilin Province, P.R. China
| | - Yifan Fu
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun City, Jilin Province, P.R. China
| | - Ye Du
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, No.71, Xinmin Street, Changchun City, Jilin Province, P.R. China.
| |
Collapse
|
7
|
Chen Y, Shi L, Yin W, Xia H, Lin C. PD‑1/PD‑L1 inhibitor‑based immunotherapy in locally advanced or metastatic triple‑negative breast cancer: A meta‑analysis. Oncol Lett 2025; 29:57. [PMID: 39606565 PMCID: PMC11600704 DOI: 10.3892/ol.2024.14803] [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: 05/29/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is negative for oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 expression. Locally advanced and metastatic TNBC not only have a worse prognosis and are more invasive than TNBC, but are also the most immunogenic subtypes of breast cancer. There is still a lack of clarity regarding the optimal treatment of locally advanced or metastatic TNBC. The present study aimed to assess the efficacy and safety of programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitor-based immunotherapy [i.e., immune checkpoint inhibitors (ICIs)] alone or in combination with other therapies for the treatment of locally advanced or metastatic TNBC. The PubMed, Cochrane Library, Embase and MEDLINE databases were searched up to July 19, 2023 to identify studies that examined the efficacy and safety of ICIs for treating TNBC. The primary outcomes were progression-free survival (PFS) and overall survival (OS). The secondary outcomes were safety and adverse events. The data were analysed using Review Manager 5.4. A total of 8 studies (3,338 patients) were included in the present meta-analysis. Compared with other therapies, ICIs had a significantly different effect on OS [hazard ratio (HR)=0.83; 95% confidence interval (CI)=0.69-1.00; P<0.05; I2=59%] in patients with locally advanced or metastatic TNBC. In addition, ICIs significantly prolonged PFS compared with other therapies (intent-to-treat: HR=0.81; 95% CI=0.75-0.88; P<0.00001; I2=0%). Immunotherapy based on PD-1/PD-L1 inhibitors showed variable efficacy on OS and PFS in TNBC, while a significant improvement was observed for PD-L1(+). Future studies should focus on PD-L1 subgroup status, which may help optimize personalized treatment regimens for TNBC.
Collapse
Affiliation(s)
- Yonghui Chen
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
| | - Liji Shi
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
| | - Weihua Yin
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
- Department of Oncology, Baoan Central Hospital of Shenzhen, Shenzhen, Guangdong 518102, P.R. China
| | - Hongmei Xia
- Department of Oncology, Baoan Central Hospital of Shenzhen, Shenzhen, Guangdong 518102, P.R. China
| | - Canling Lin
- School of Chemical and Biological Engineering, Yichun College, Yichun, Jiangxi 336000, P.R. China
| |
Collapse
|
8
|
Wang Y, Wang X, Sun H, Zhang Z, Gu J. LncRNA MCM3AP-AS1 promotes chemoresistance in triple-negative breast cancer through the miR-524-5p/RBM39 axis. Mol Cell Biochem 2025; 480:371-384. [PMID: 38472681 DOI: 10.1007/s11010-023-04908-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 11/25/2023] [Indexed: 03/14/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most lethal subtype of BC, with unfavorable treatment outcomes. Evidence suggests the engagement of lncRNA MCM3AP-AS1 in BC development. This study investigated the action of MCM3AP-AS1 in chemoresistance of TNBC cells. Drug-resistant TNBC cell lines SUM159PTR and MDA-MB-231R were constructed by exposure to increasing concentrations of doxorubicin/docetaxel (DOX/DXL). MCM3AP-AS1 and miR-524-5p expression levels were determined by RT-qPCR. RNA binding motif 39 (RBM39) level was measured using Western blot. Cell viability and apoptosis were assessed by CCK-8 assay and flow cytometry. The targeted binding of miR-524-5p with MCM3AP-AS1 or RBM39 was predicted by ECORI database and validated by dual-luciferase assays. The gain-and-loss of function assays were conducted in cells to investigate the interactions among MCM3AP-AS1, miR-524-5p, and RBM39. TNBC xenograft mouse models were established through subcutaneous injection of MCM3AP-AS1-silencing MDA-MB-231R cells and intraperitoneally administrated with DOX/DXL to verify the role of MCM3AP-AS1 in vivo. MCM3AP-AS1 was upregulated in drug-resistant TNBC cells, and MCM3AP-AS1 silencing could sensitize drug-resistant TNBC cells to chemotherapeutic drugs by promoting apoptosis. MCM3AP-AS1 targeted miR-524-5p. After DOX/DXL treatment, miR-524-5p inhibition partially reversed the effect of MCM3AP-AS1 silencing on inhibiting chemoresistance and promoting apoptosis of drug-resistant TNBC cells. miR-524-5p targeted RBM39. Silencing MCM3AP-AS1 promoted apoptosis via the miR-524-5p/RBM39 axis, thereby enhancing chemosensitivity of drug-resistant TNBC cells. MCM3AP-AS1 knockdown upregulated miR-524-5p, downregulated RBM39, and restrained tumor development in vivo. MCM3AP-AS1 silencing potentiates apoptosis of drug-resistant TNBC cells by upregulating miR-524-5p and downregulating RBM39, thereby suppressing chemoresistance in TNBC.
Collapse
Affiliation(s)
- Yueping Wang
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China
- Department of Molecular and Cellular Biology, University of Connecticut, Storrs, CT, 06269, USA
| | - Xuedong Wang
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China.
| | - Haiyi Sun
- School of Clinical Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ziyun Zhang
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China
| | - Juan Gu
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China
| |
Collapse
|
9
|
Shu Y, Lan J, Luo H, Fu H, Xiao X, Yang L. FOS-Mediated PLCB1 Induces Radioresistance and Weakens the Antitumor Effects of CD8 + T Cells in Triple-Negative Breast Cancer. Mol Carcinog 2025; 64:162-175. [PMID: 39451071 DOI: 10.1002/mc.23834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024]
Abstract
Radioresistance and immune evasion are interactive and crucial events leading to treatment failure and progression of human malignancies. This research studies the role of phospholipase C beta 1 (PLCB1) in these events in triple-negative breast cancer (TNBC) and the regulatory mechanism. PLCB1 was bioinformatically predicted as a dysregulated gene potentially linked to radioresistance in TNBC. Parental TNBC cell lines were exposed to fractionated radiation for 6 weeks. PLCB1 expression was decreased in the first 2 weeks but gradually increased from Week 3. PLCB1 knockdown increased the radiosensitivity of the cells, as manifested by a decreased half-inhibitory dose of irradiation, reduced cell proliferation, apoptosis resistance, mobility, and tumorigenesis in mice. The FOS transcription factor promoted PLCB1 transcription and activated the PI3K/AKT signaling. Knockdown of FOS similarly reduced radioresistance and T cells-mediated immune evasion. However, the radiosensitivity of TNBC cells and the antitumor effects of CD8+ T cells could be affected by a PI3K/AKT activator or by the PLCB1 upregulation. The PLCB1 or FOS knockdown also suppressed radioresistance and tumorigenesis of the TNBC cells in mice. In conclusion, FOS-mediated PLCB1 induces radioresistance and weakens the antitumor effects of CD8+ T cells in TNBC by activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yuxian Shu
- Department of Breast Comprehensive Radiotherapy, Jiangxi Cancer Hospital, Nanchang, Jiangxi, People's Republic of China
| | - Jun Lan
- First Department of General Surgery, Jiangxi Gao'an People's Hospital, Gao'an, Jiangxi, People's Republic of China
| | - Huijing Luo
- Department of Oncology, Taihe County People's Hospital, Ji'an, Jiangxi, People's Republic of China
| | - Huiying Fu
- Department of Oncology, No.908 Hospital, Joint Logistics Support Force, Nanchang, Jiangxi, People's Republic of China
| | - Xuhuang Xiao
- Department of Oncology, Taihe County Traditional Chinese Medicine Hospital, Ji'an, Jiangxi, People's Republic of China
| | - Liping Yang
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
10
|
Peg V, Abengozar-Muela M, Acosta J, Andrés L, García-Rojo M, Hardisson D, Nicolau MJ, Ramos-Oliver I, Rodrigo M, Sánchez-Bernal ML, Sanz J, Garrote L, Ramírez I, Rojo F. New Approach in the Interpretation of Complex Triple-negative Breast Cancer Immunohistochemistry Specimens Processed With VENTANA PD-L1 (SP142) Assay. Appl Immunohistochem Mol Morphol 2025; 33:15-21. [PMID: 39636314 DOI: 10.1097/pai.0000000000001237] [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: 06/18/2024] [Accepted: 10/13/2024] [Indexed: 12/07/2024]
Abstract
Triple-negative breast cancer (TNBC) is challenging to treat because of its lack of specific molecular targets. The IMMUNOPEG study aimed to evaluate a novel structured method for interpreting TNBC immunohistochemistry specimens processed with VENTANA PD-L1 (SP142) assay. The study involved 10 pathologists who evaluated 50 different immunohistochemistry specimens of TNBC with programmed death ligand 1 (PD-L1) expression considered challenging and that were previously evaluated by the scientific committee, using the NAVIFY Digital Pathology platform. Initially, the overall percent agreement (OPA) was 74%, with a negative percent agreement (NPA) of 68.2% for samples classified as negative, and a positive percent agreement (PPA) of 94.5% for positive samples. After training on the method, the OPA improved significantly to 81.6%, with the NPA increasing to 80.5% and the PPA decreasing to 85.5%. The mean percentage of the tumor area occupied by PD-L1-stained immune cells decreased from 2.5% to 1.6% post-training, approaching to the scientific committee's consensus of 1.029%. The study found that the pathologists' confidence in their assessments increased significantly when using the structured method, which was found to be easy to use by 9 out of 10 pathologists. All pathologists agreed that the structured method was useful for assessing PD-L1 expression. The study suggests that this method has potential value in interpreting challenging cases of PD-L1 immunohistochemistry (IHC) in TNBC. Further refinement and a training protocol may be necessary to enhance the method's efficiency. The potential for generalizing this structured method to other IHC procedures and pathologies warrants additional research.
Collapse
Affiliation(s)
- Vicente Peg
- Department of Pathology, Vall d'Hebron University Hospital
- Universidad Autónoma de Barcelona, Barcelona
| | | | - Jesús Acosta
- Department of Anatomic Pathology, Hospital General Universitario Santa Lucía, Cartagena
| | - Leire Andrés
- Department of Pathology, Hospital Universitario Cruces, Bilbao
| | - Marcial García-Rojo
- Department of Pathology, Hospital Universitario de Jerez, Jerez de la Frontera
| | | | - María Jesús Nicolau
- Department of Pathology, Hospital General Universitario de Castellón, Castellón
| | - Irma Ramos-Oliver
- Department of Pathology, Hospital Universitari Doctor Josep Trueta, Girona
| | | | | | - Julián Sanz
- Department of Pathology, Clínica Universidad de Navarra, Madrid
| | - Leia Garrote
- Medical Department, Roche Farma S.A., Madrid, Spain
| | | | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz, Madrid, Spain
| |
Collapse
|
11
|
Xiao H, Liu L, Huang S. STK32C modulates doxorubicin resistance in triple-negative breast cancer cells via glycolysis regulation. Mol Cell Biochem 2025; 480:459-471. [PMID: 38507019 DOI: 10.1007/s11010-024-04989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 03/09/2024] [Indexed: 03/22/2024]
Abstract
Understanding the mechanisms underlying doxorubicin resistance in triple-negative breast cancer (TNBC) holds paramount clinical significance. In our study, we investigate the potential of STK32C, a little-explored kinase, to impact doxorubicin sensitivity in TNBC cells. Our findings reveal elevated STK32C expression in TNBC specimens, associated with unfavorable prognosis in doxorubicin-treated TNBC patients. Subsequent experiments highlighted that STK32C depletion significantly augmented the sensitivity of doxorubicin-resistant TNBC cells to doxorubicin. Mechanistically, we unveiled that the cytoplasmic subset of STK32C plays a pivotal role in mediating doxorubicin sensitivity, primarily through the regulation of glycolysis. Furthermore, the kinase activity of STK32C proved to be essential for its mediation of doxorubicin sensitivity, emphasizing its role as a kinase. Our study suggests that targeting STK32C may represent a novel therapeutic approach with the potential to improve doxorubicin's efficacy in TNBC treatment.
Collapse
Affiliation(s)
- Huawei Xiao
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Lei Liu
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China
| | - Shaoyan Huang
- Department of Medical Oncology, Yantaishan Hospital, Yantai, Shandong Province, China.
| |
Collapse
|
12
|
Wang X, Wang X, Gu J, Wei Y, Wang Y. circUBR5 promotes ribosome biogenesis and induces docetaxel resistance in triple-negative breast cancer cell lines via the miR-340-5p/CMTM6/c-MYC axis. Neoplasia 2025; 59:101062. [PMID: 39672097 PMCID: PMC11697786 DOI: 10.1016/j.neo.2024.101062] [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: 03/10/2024] [Revised: 09/11/2024] [Accepted: 09/21/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE Docetaxel (DTX) represents an effective chemotherapeutic agent for treating triple-negative breast cancer (TNBC), but the efficacy is strongly limited by drug resistance. c-MYC-mediated ribosome biogenesis is considered a feasible strategy to confront chemoresistance in BC. We elucidated the impact of CMTM6 on TNBC DTX chemoresistance by governing c-MYC-mediated ribosome biogenesis, and its upstream ceRNA regulatory pathways. METHODS DTX-resistant TNBC cells MDA-MB-231R and HCC1937R were generated by exposing sensitive cells MDA-MB-231 and HCC1937 to escalating doses of DTX. The expression patterns of CMTM6 and c-MYC were assessed by Western blot. The relationships between CMTM6 and miR-340-5p, circUBR5 and miR-340-5p were determined using bioinformatics analysis, luciferase assay, RIP, RNA in situ hybridization and biotin-labeled miR co-precipitation assay. Following ectopic expression and depletion experiments in DTX-resistant cells, cell chemoresistance, apoptosis, colony formation and nascent protein synthesis were evaluated. RESULTS CMTM6 expression was elevated in DTX-resistant TNBC cells. CMTM6 knockdown enhanced apoptosis of DTX-resistant TNBC cells and increased their sensitivity to DTX by blocking c-MYC-mediated ribosome biogenesis. Mechanistically, miR-340-5p targeted CMTM6 and negatively regulated the expression of CMTM6 in DTX-resistant TNBC cells. Moreover, circUBR5 attenuated the repression on CMTM6 expression as a ceRNA for miR-340-5p. circUBR5 knockdown inactivated c-MYC-mediated ribosome biogenesis, and therefore enhanced DTX efficacy by promoting miR-340-5p binding to CMTM6. CONCLUSION circUBR5 knockdown facilitated miR-340-5p-targeted CMTM6 via a ceRNA mechanism, thereby reducing c-MYC-mediated ribosome biogenesis and accelerating chemosensitization of DTX-resistant TNBC cells, which offered a theoretical guideline for clinical research on the feasibility of inhibiting ribosome biogenesis to reduce TNBC chemoresistance.
Collapse
Affiliation(s)
- Xuedong Wang
- School of Medicine, Anhui University of Science & Technology, Huainan, Anhui, 232001, China
| | - Xinping Wang
- School of Medicine, Anhui University of Science & Technology, Huainan, Anhui, 232001, China
- Center for Precision Medicine, Anhui No.2 Provincial People's Hospital, Hefei, Anhui, 230041, China
| | - Juan Gu
- School of Medicine, Anhui University of Science & Technology, Huainan, Anhui, 232001, China
| | - Yilei Wei
- Lingbi Hospital, Anhui No.2 Provincial People's Hospital, Lingbi, Anhui, 234200, China
| | - Yueping Wang
- School of Medicine, Anhui University of Science & Technology, Huainan, Anhui, 232001, China
- Lingbi Hospital, Anhui No.2 Provincial People's Hospital, Lingbi, Anhui, 234200, China
| |
Collapse
|
13
|
Yan Z, Zhong Z, Shi C, Feng M, Feng X, Liu T. The prognostic marker KRT81 is involved in suppressing CD8 + T cells and predicts immunotherapy response for triple-negative breast cancer. Cancer Biol Ther 2024; 25:2355705. [PMID: 38778753 PMCID: PMC11123506 DOI: 10.1080/15384047.2024.2355705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
Triple-negative breast Cancer (TNBC) is an aggressive subtype lacking estrogen, progesterone, and HER2 receptors. Known for limited targeted therapies, it poses challenges and requires personalized treatment strategies. Differential analysis revealed a significant decrease in keratin 81 (KRT81) expression in non-TNBC samples and an increase in TNBC samples, lower KRT81 expression correlated with better TNBC patient outcomes. It emerged as an independent predictive factor for TNBC, with associations found between its expression and clinically relevant features. We further developed a nomogram for survival probability assessment based on Cox regression results, demonstrating its accuracy through calibration curves. Gene annotation analysis indicated that KRT81 is involved in immune-related pathways and tumor cell adhesion. KRT81 is associated with immune cell infiltration of Follicular helper T cells (Tfh) and CD8 + T cells, suggesting its potential impact on the immunological microenvironment. The study delved into KRT81's predictive value for immunotherapy responses, high expression of KRT81 was associated with greater potential for immune evasion. Single-cell RNA sequencing analysis pinpointed KRT81 expression within a specific malignant subtype which was a risk factor for TNBC. Furthermore, KRT81 promoted TNBC cell proliferation, migration, invasion, and adhesion was confirmed by gene knockout or overexpression assay. Co-culture experiments further indicated KRT81's potential role in inhibiting CD8 + T cells, and correlation analysis implied KRT81 was highly correlated with immune checkpoint CD276, providing insights into its involvement in the immune microenvironment via CD276. In conclusion, this comprehensive study positions KRT81 as a promising prognostic marker for predicting tumor progression and immunotherapy responses in TNBC.
Collapse
Affiliation(s)
- Zhideng Yan
- Department of General Surgery, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Zhihui Zhong
- Center of Stem Cell and Regenerative Medicine, Gaozhou People’s Hospital, Gaozhou, Guangdong, China
| | - Chuanke Shi
- Department of General Surgery, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Muyin Feng
- Department of Pathology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People’s Hospital, Gaozhou, Guangdong, China
| | - Tong Liu
- Department of General Surgery, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan, Guangdong, China
| |
Collapse
|
14
|
Wang M, Lan S, Song M, Zhang R, Zhang W, Sun X, Liu G. Synthesis of Zinc Oxide-Doped Carbon Dots for Treatment of Triple-Negative Breast Cancer. Int J Nanomedicine 2024; 19:13949-13971. [PMID: 39742095 PMCID: PMC11687324 DOI: 10.2147/ijn.s494262] [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: 09/13/2024] [Accepted: 12/18/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction The anti-cancer properties of zinc oxide-doped carbon dots (CDs/ZnO) in inhibiting triple-negative breast cancer (TNBC) progression merit more investigation. Methods With citric acid as the carbon source, urea applied as the nitrogen source, and zinc oxide (ZnO) used as a reactive dopant, CDs/ZnO were synthesized by microwave heating in the current study, followed by the characterization and biocompatibility assessments. Subsequently, the anti-cancer capabilities of CDs/ZnO against TNBC progression were evaluated by various biochemical and molecular techniques, including viability, proliferation, migration, invasion, adhesion, clonogenicity, cell cycle distribution, apoptosis, redox homeostasis, metabolome, and transcriptome assays of MDA-MB-231 cells. Additionally, the in vivo anti-cancer potentials of CDs/ZnO against TNBC progression were analyzed using TNBC xenograft mouse models. Results The biocompatibility of CDs/ZnO was supported by the non-significant changes in the pathological and physiological parameters in the CDs/ZnO treated mice, alongside a non-cytotoxic effect of CDs/ZnO on the proliferation of normal cells. Notably, the CDs/ZnO treatments effectively decreased the viability, proliferation, migration, invasion, adhesion, and clonogenicity of MDA-MB-231 cells. Furthermore, the CDs/ZnO treatments induced cell cycle arrest, apoptosis, redox imbalance, metabolome disturbances, and transcriptomic alterations of MDA-MB-231 cells by regulating the MAPK signaling pathway. Additionally, the CDs/ZnO treatments markedly suppressed the in vivo tumor growth in the TNBC xenograft mouse models. Conclusion In this study, we synthesized CDs/ZnO via microwave heating, using citric acid as the carbon source, urea as the nitrogen source, and ZnO as a reactive dopant. We confirmed the biosafety and potent anti-cancer efficacy of CDs/ZnO in inhibiting TNBC progression by disrupting malignant cell behaviors through modulation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Mengqi Wang
- College of Life Science and Oceanography, Weifang University, Weifang, Shandong, People’s Republic of China
| | - Shuting Lan
- Key Laboratory of Medical Cell Biology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Mingjun Song
- College of Chemical Engineering and Environmental Chemistry, Weifang University, Weifang, Shandong, People’s Republic of China
| | - Rongrong Zhang
- Key Laboratory of Medical Cell Biology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Wenqi Zhang
- Key Laboratory of Medical Cell Biology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Xiaomei Sun
- Key Laboratory of Medical Cell Biology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Gang Liu
- Key Laboratory of Medical Cell Biology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, People’s Republic of China
| |
Collapse
|
15
|
Hermán-Sánchez N, G-García ME, Jiménez-Vacas JM, Yubero-Serrano EM, López-Sánchez LM, Romero-Martín S, Raya-Povedano JL, Álvarez-Benito M, Castaño JP, Luque RM, Gahete MD. The splicing machinery is dysregulated and represents a therapeutic vulnerability in breast cancer. Cell Mol Life Sci 2024; 82:18. [PMID: 39725737 DOI: 10.1007/s00018-024-05515-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/27/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024]
Abstract
Breast cancer (BCa) is a highly prevalent pathological condition (̴30% in women) with limited and subtype-dependent prognosis and therapeutic options. Therefore, BCa management might benefit from the identification of novel molecular elements with clinical potential. Since splicing process is gaining a great relevance in cancer, this work analysed the expression of multiple Spliceosome Components (SCs = 17) and Splicing Factors (SFs = 26) and found a drastic dysregulation in BCa (n = 69) vs. control (negative biopsies; n = 50) samples. Among all the components analysed, we highlight the upregulation of ESRP1 and down-regulation of PRPF8 and NOVA1 in BCa vs. control samples. Indeed, ESRP1 was specially overexpressed in triple-negative BCa (TNBCa) and associated with worse prognosis (i.e., higher BCa grade and lower overall survival), suggesting an association of ESRP1 with BCa aggressiveness. On the other hand, PRPF8 expression was generally downregulated in BCa with no associations to clinical characteristics, while NOVA1 expression was lower in TNBCa patients and highly aggressive tumours. Consistently, NOVA1 overexpression in vitro reduced functional parameters of aggressiveness in ER-/PR- cell lines (MDA-MB-231 and BT-549) but not in ER+/PR+ cells (MCF7), suggesting a critical role of NOVA1 in subtype-specific BCa. Finally, the in vitro pharmacological inhibition of splicing machinery using pladienolide B decreased aggressiveness features in all the BCa cell lines, showing a subtype-independent inhibitory potential, but being relatively innocuous in normal-like breast cells. These results demonstrate the profound dysregulation of the splicing machinery in BCa and their potential as source of promising diagnosis/prognosis markers, as well as valuable therapeutic targets for BCa.
Collapse
Affiliation(s)
- Natalia Hermán-Sánchez
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
| | - Miguel E G-García
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
| | - Juan M Jiménez-Vacas
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
| | - Elena M Yubero-Serrano
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Lipids and Atherosclerosis Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Laura M López-Sánchez
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
| | - Sara Romero-Martín
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- Mammary Gland Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Jose L Raya-Povedano
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- Mammary Gland Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Marina Álvarez-Benito
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain
- Reina Sofía University Hospital, Córdoba, 14004, Spain
- Mammary Gland Unit, Reina Sofía University Hospital, Córdoba, Spain
| | - Justo P Castaño
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain.
- Reina Sofía University Hospital, Córdoba, 14004, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain.
| | - Raúl M Luque
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain.
- Reina Sofía University Hospital, Córdoba, 14004, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain.
| | - Manuel D Gahete
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), IMIBIC building. Av. Menéndez Pidal s/n, Córdoba, 14004, Spain.
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, 14004, Spain.
- Reina Sofía University Hospital, Córdoba, 14004, Spain.
- CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain.
| |
Collapse
|
16
|
Aishajiang R, Liu Z, Liang Y, Du P, Wei Y, Zhuo X, Liu S, Lei P, Wang T, Yu D. Concurrent Amplification of Ferroptosis and Immune System Activation Via Nanomedicine-Mediated Radiosensitization for Triple-Negative Breast Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407833. [PMID: 39721034 DOI: 10.1002/advs.202407833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Radiation therapy (RT) is one of the core therapies for current cancer management. However, the emergence of radioresistance has become a major cause of radiotherapy failure and disease progression. Therefore, overcoming radioresistance to achieve highly effective treatment for refractory tumors is significant yet challenging. Here, pH-responsive DSPE-PEoz modified hollow Bi2Se3-RSL3/diABZi (DP-HBN/RA) nanomedicine is designed as a radiation sensitizer for efficient treatment of triple-negative breast cancer by simultaneously amplifying ferroptosis and immune system activation. DP-HBN/RA can efficiently concentrate X-ray radiation energy inside the tumor, thereby promoting precise ionizing radiation exposure in tumor cells to produce large amounts of reactive oxygen species (ROS), leading to lipid peroxidation-induced ferroptosis. Meanwhile, ferroptotic cell death is intensified through the inactivation of GPX4 by RSL3 released from DP-HBN/RA to acidic conditions in the tumor microenvironment. Additionally, DP-HBN/RA enhances RT efficacy to exacerbate unrepairable DNA damage and release DNA fragments that activate the cGAS-STING signal pathway, evoking a systematic immune response. Ingeniously, the released diABZi reinforces cGAS-STING activation to boost the immunology antitumor effect. This work links the induction of ferroptosis and the initiation of systematic immune response to achieve highly effective tumor suppression, which opens up new avenues for future treatments of refractory tumors.
Collapse
Affiliation(s)
- Reyida Aishajiang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130022, China
| | - Zhongshan Liu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130022, China
| | - Yuan Liang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Pengye Du
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Yi Wei
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Xiqian Zhuo
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130022, China
| | - Shuyu Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Pengpeng Lei
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Tiejun Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130022, China
| | - Duo Yu
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, 130022, China
| |
Collapse
|
17
|
Wang X, Wang W, Zeng H, Hu X, Chen F, Shen L, Tao J. Molecular structure of polysaccharide mediated autophagy markers KIF23 and PRC1 proteins and their regulatory role in triple negative cancer through the p53 signaling pathway. Int J Biol Macromol 2024; 291:139155. [PMID: 39725112 DOI: 10.1016/j.ijbiomac.2024.139155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 12/28/2024]
Abstract
As a process of intracellular degradation and recycling of its own components, abnormal regulation of autophagy has been strongly associated with the development of multiple cancer types, including triple-negative breast cancer. The amino acid sequences of KIF23 and PRC1 proteins were analyzed by bioinformatics method, their three-dimensional structures were predicted, and their interactions with polysaccharides were studied by molecular docking technology. The localization and expression patterns of KIF23 and PRC1 in cells were studied by cell biology techniques. By constructing breast cancer cell lines that stably overexpress or knock down KIF23 and PRC1, we evaluated the effect of these proteins on autophagy activity. Finally, molecular biological methods such as Western blot and real-time quantitative PCR were used to detect the expression changes of proteins related to p53 signaling pathway and the levels of autophagy markers such as LC3 and p62, thereby revealing the regulatory effects of KIF23 and PRC1 on autophagy of triple-negative breast cancer cells through p53 signaling pathway. The study found that the KIF23 and PRC1 proteins have complex three-dimensional structures, and their interactions with polysaccharides may affect their function during cell division and autophagy. In triple-negative breast cancer cells, overexpression of KIF23 and PRC1 significantly enhanced autophagy activity, while knockdown of these proteins inhibited autophagy. Further experiments showed that KIF23 and PRC1 regulate the expression of autophagy related proteins by influencing the activity of p53 signaling pathway. Overexpression of KIF23 and PRC1 led to inhibition of the p53 signaling pathway, while knocking down these proteins activated the p53 signaling pathway, which was consistent with reduced autophagy activity.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Clinical Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Wei Wang
- GCP Center, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Hanling Zeng
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China
| | - Xinru Hu
- Department of Public Health, School of Medicine and Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Fangyi Chen
- Department of Public Health, School of Medicine and Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Shen
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China.
| | - Jing Tao
- Department of General Surgery, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210031, China.
| |
Collapse
|
18
|
Zhang Y, Tang X, Wang Y, Shi F, Gao X, Guo Y, Liu Q, Ma W. Recent advances targeting chemokines for breast cancer. Int Immunopharmacol 2024; 146:113865. [PMID: 39718056 DOI: 10.1016/j.intimp.2024.113865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024]
Abstract
Breast cancer (BC) is a complex and heterogeneous disease, and its onset and progression involve the interplay of multiple molecular mechanisms. Chemokines and their receptors are key regulators of cell migration and immune responses and contribute significantly to the pathophysiology of BC. This article reviews the classification, functions, and mechanisms of chemokines and their receptors in the proliferation, migration, invasion, and angiogenesis of BC cells. This study explores the regulatory roles of chemokines and their receptors in the immune microenvironment of BC, particularly the ways they influence the infiltration, polarization, and antitumor immune responses of immune cells. Finally, this article summarizes the current treatment strategies for breast cancer that utilize chemokines and their receptors and provides insights into future research directions and trends in this field.
Collapse
Affiliation(s)
- Yanan Zhang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Xiufeng Tang
- Department of Pharmacy and Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, China.
| | - Ying Wang
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Fengcui Shi
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Xing Gao
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Yingxin Guo
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Qian Liu
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China.
| | - Wenjian Ma
- College of Biological and Chemical Engineering, Qilu Institute of Technology, Shandong, China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China.
| |
Collapse
|
19
|
Lu W, Yang S. METTL3/IGF2BP1 promotes the development of triple-negative breast cancer by mediating m6A methylation modification of PRMT7. Tissue Cell 2024; 93:102690. [PMID: 39709713 DOI: 10.1016/j.tice.2024.102690] [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: 09/03/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND PRMT7 is upregulated in breast cancer and promotes tumor metastasis. Here we aimed to explore the function and mechanism of PRMT7 in triple-negative breast cancer (TNBC). METHODS The expression of PRMT7, METTL3 and IGF2BP1 was detected by immunohistochemistry (IHC), qRT-PCR and western blot. Cell viability and proliferation were measured using MTT and EdU assay. Flow cytometry and TUNEL assays were used to evaluate apoptosis. Invasion and migration were assessed by transwell and wound healing assays, respectively. Glucose consumption and lactate production were measured to assess glycolysis. In addition, the interaction between METTL3 and PRMT was verified by methylated RNA immunoprecipitation. The roles of METTL3 and PRMT in vivo were investigated through a xenograft model. RESULTS PRMT7 was upregulated in TNBC tissues and cells, and the knockdown of PRMT7 inhibited cell proliferation, invasion, migration and glycolysis, but induced apoptosis in TNBC cells. METTL3/IGF2BP1 enhanced PRMT7 expression by mediating the m6A methylation modification of PRMT7. Besides, METTL3 knockdown suppressed the progression of TNBC cells and regulated the WNT/β-catenin pathway via PRMT7. Moreover, silencing METTL3 restrained TNBC tumor growth in vivo through regulating PRMT7. CONCLUSION METTL3/IGF2BP1 facilitates the progression of TNBC by mediating m6A methylation modification of PRMT7.
Collapse
Affiliation(s)
- Wanli Lu
- Department of General Surgery, Qinghai University Affiliated Hospital, Xining 810000, China
| | - Shenghu Yang
- Department of General Surgery, Qinghai University Affiliated Hospital, Xining 810000, China.
| |
Collapse
|
20
|
Zhuo Z, Lu W, Zhang L, Zhang D, Cui Y, Wu X, Mei H, Chang L, Song Q. Transcriptomic analysis reveals potential crosstalk genes and immune relationship between triple-negative breast cancer and depression. Discov Oncol 2024; 15:762. [PMID: 39692924 DOI: 10.1007/s12672-024-01562-4] [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: 04/03/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024] Open
Abstract
TNBC, the most aggressive form of breast cancer, lacks accurate and effective therapeutic targets. Immunotherapy presents a promising approach for addressing TNBC. Anxiety and depression are frequently concurrent symptoms in TNBC patients. MDD affects the tumor immune microenvironment of TNBC, with its characteristic genes affecting the pathophysiology of MDD and potentially increasing the risk of TNBC recurrence and metastasis. This study reveals significant differences in T lymphocyte infiltration between high-risk and low-risk TNBC groups based on MDD feature genes. This finding aids in identifying TNBC patients who may benefit from immunotherapy, providing new insights for future TNBC immunotherapy strategies. Our aim is to identify MDD-related genes involved in the pathogenesis of TNBC and to provide predictive biomarkers for TNBC immunotherapy.
Collapse
Affiliation(s)
- Zhili Zhuo
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Wenping Lu
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China.
| | - Ling Zhang
- Department of pathology, China Academy of Chinese Medical Sciences Guang' anmen Hospital, Beijing, 100053, China
| | - Dongni Zhang
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Yongjia Cui
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Xiaoqing Wu
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Heting Mei
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Lei Chang
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| | - Qingya Song
- Oncology Department, China Academy of Chinese Medical Sciences Guang'anmen Hospital, No.5 Beixiange, Xicheng District, Beijing, 100053, China
| |
Collapse
|
21
|
Earla JR, Kurian AW, Kponee-Shovein K, Mahendran M, Song Y, Hua Q, Hilts A, Sun Y, Hirshfield KM, Robson M, Mejia JA. Correlation Between Disease-Free Survival Endpoints and Overall Survival in Elderly Patients with Early-Stage HER2-Negative Breast Cancer: A SEER-Medicare Analysis. Adv Ther 2024:10.1007/s12325-024-03074-7. [PMID: 39680314 DOI: 10.1007/s12325-024-03074-7] [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] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION Recent trial-level meta-analyses have established disease-free survival (DFS) as a valid surrogate for overall survival (OS) in human epidermal growth factor receptor 2-negative (HER2-) breast cancer (BC), irrespective of disease stage, and in early-stage hormone receptor-positive (HR+)/HER2- BC. To advance the understanding of the association between additional DFS endpoints and OS, this study assessed the patient-level correlations between DFS and OS, invasive DFS (IDFS) and OS, and distant DFS (DDFS) and OS in Medicare beneficiaries with early-stage HER2- BC, overall and in subgroups of patients with HR+/HER2- BC and triple-negative BC (TNBC). METHODS Patients with stages I-III HER2- BC aged ≥ 66 years were identified from SEER-Medicare data (2010-2019). DFS, IDFS, DDFS, and OS were assessed using Kaplan-Meier analyses. Normal scores rank correlation was estimated between each DFS endpoint and OS, overall and separately in patients with HR+/HER2- BC and TNBC. RESULTS Of 28,655 patients, 90.4% had HR+/HER2- BC and 9.6% had TNBC (median follow-up 4 years). Median DFS, IDFS, and DDFS were 4.5, 5.9, and 6.3 years, respectively, in HR+/HER2- BC and 3.0, 3.8, and 4.4 years, respectively, in TNBC. Median OS was not reached (5-year OS, HR+/HER2- BC 83.7%; TNBC 67.7%). A significant positive correlation was observed between each DFS endpoint and OS across cohorts, with the strongest correlation observed between DDFS and OS in HR+/HER2- BC (correlation coefficient 0.60; 95% confidence interval 0.57-0.62; p < 0.001) and in TNBC (0.69; 0.65-0.71; p < 0.001). CONCLUSION We observed significant positive patient-level correlations between DFS and OS, IDFS and OS, and DDFS and OS in early-stage HER2- BC. Our IDFS and DDFS findings advance the understanding of the role of these DFS endpoints as predictors of OS, and their potential utility as surrogate endpoints in clinical trials of early-stage HER2- BC, given additional validation in trial-level meta-analyses.
Collapse
Affiliation(s)
| | | | | | | | - Yan Song
- Analysis Group, Inc, Boston, MA, USA
| | - Qi Hua
- Analysis Group, Inc, Boston, MA, USA
| | | | - Yezhou Sun
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| | | | - Mark Robson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaime A Mejia
- Merck & Co., Inc., 90 E Scott Ave, Rahway, NJ, 07065, USA
| |
Collapse
|
22
|
Ba L, Zhao Z, Zhang C, Chu Y, Wu C. Expression and prognostic impact of hypoxia- and immune escape-related genes in triple-negative breast cancer: A comprehensive analysis. Int Immunopharmacol 2024; 146:113810. [PMID: 39689602 DOI: 10.1016/j.intimp.2024.113810] [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: 09/19/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that lacks effective therapeutic options. Hypoxia and immune escape are critical factors that contribute to the progression of and resistance to therapy in patients with TNBC. Nevertheless, few studies have comprehensively analyzed hypoxia and immune escape in patients with TNBC. This study aimed to examine the expression of hypoxia- and immune escape-related genes in TNBC and their influence on prognosis. TNBC datasets were downloaded and processed from The Cancer Genome Atlas and Gene Expression Omnibus. Differential expression analysis identified 4949 differentially expressed genes, between TNBC and normal tissues. The intersection yielded 116 hypoxia- and immune escape-related differentially expressed genes (H&IERDEGs), including KIF4A, BIRC5, and BUB1. Enrichment analyses indicated that H&IERDEGs were significantly enriched in biological processes, including cell chemotaxis, leukocyte migration, and cytokine-cytokine receptor interaction. Subsequently, weighted gene co-expression network analysis identified 43 module genes that were found to define two TNBC subtypes. We constructed a prognostic risk model consisting of eight signature genes, which demonstrated a high predictive performance to predict the overall survival (OS) of patients with TNBC with an area under the curve (AUC) exceeding 0.9 at 1 year survival. This indicates that the model effectively differentiates between outcomes, reflecting its robust performance. This study investigated the roles and potential mechanisms of hypoxia- and immune escape-related genes in TNBC and constructed a prognostic risk model with a high predictive performance. These findings offer novel molecular markers and potential therapeutic targets for TNBC.
Collapse
Affiliation(s)
- Li Ba
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Zhiyu Zhao
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Laboratory of Medical Genetics, Harbin Medical University, Harbin 150001, PR China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin 150001, PR China
| | - Chunmei Zhang
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Yinzhu Chu
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Changjun Wu
- Department of Ultrasound, First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China.
| |
Collapse
|
23
|
Wang M, Liu Y, Li Y, Lu T, Wang M, Cheng Z, Chen L, Wen T, Pan M, Hu G. Tumor Microenvironment-Responsive Nanoparticles Enhance IDO1 Blockade Immunotherapy by Remodeling Metabolic Immunosuppression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2405845. [PMID: 39661740 DOI: 10.1002/advs.202405845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/25/2024] [Indexed: 12/13/2024]
Abstract
The clinical efficacy of immune checkpoint blockade (ICB) therapy is significantly compromised in the metabolically disordered tumor microenvironment (TME), posing a formidable challenge that cannot be ignored in current antitumor strategies. In this study, TME-responsive nanoparticles (HMP1G NPs) loaded with 1-methyltryptophan (1-MT; an indoleamine 2,3-dioxygenase 1 [IDO1] inhibitor,) and S-nitrosoglutathione (GSNO; a nitric oxide donor) is developed to enhance the therapeutic efficacy of 1-MT-mediated ICB. The HMP1G NPs responded to H+ and glutathione in the TME, releasing Mn2+, GSNO, and 1-MT. The released Mn2+ catalyzed the production of abundant reactive oxygen species and nitric oxide from hydrogen peroxide and GSNO, and the generated nitric oxide, synergistically with 1-MT, inhibited the accumulation of kynurenine mediated by IDO1 in the tumor. Mechanistically, HMP1G NPs downregulated tumor cell-derived IDO1 via the aryl hydrocarbon receptor/signal transducer and activator of transcription 3/interleukin signaling axis to improve kynurenine/tryptophan metabolism and immunosuppression. In a murine breast cancer model, treatment with HMP1G NPs elicited effective antitumor immunity and enhanced survival outcomes. This study highlights a novel nano-platform that simultaneously improves metabolism and enhances ICB efficacy to achieve a new and efficient antitumor strategy.
Collapse
Affiliation(s)
- Mengna Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yuhong Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yanshi Li
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Tao Lu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Min Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Zhaobo Cheng
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Lin Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Tongling Wen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- The First Clinical College, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Min Pan
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Guohua Hu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
24
|
Galappaththi SPL, Smith KR, Alsatari ES, Hunter R, Dyess DL, Turbat-Herrera EA, Dasgupta S. The Genomic and Biologic Landscapes of Breast Cancer and Racial Differences. Int J Mol Sci 2024; 25:13165. [PMID: 39684874 DOI: 10.3390/ijms252313165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Breast cancer is a significant health challenge worldwide and is the most frequently diagnosed cancer among women globally. This review provides a comprehensive overview of breast cancer biology, genomics, and microbial dysbiosis, focusing on its various subtypes and racial differences. Breast cancer is primarily classified into carcinomas and sarcomas, with carcinomas constituting most cases. Epidemiology and breast cancer risk factors are important for public health intervention. Staging and grading, based on the TNM and Nottingham grading systems, respectively, are crucial to determining the clinical outcome and treatment decisions. Histopathological subtypes include in situ and invasive carcinomas, such as invasive ductal carcinoma (IDC) and invasive lobular carcinoma (ILC). The review explores molecular subtypes, including Luminal A, Luminal B, Basal-like (Triple Negative), and HER2-enriched, and delves into breast cancer's histological and molecular progression patterns. Recent research findings related to nuclear and mitochondrial genetic alterations, epigenetic reprogramming, and the role of microbiome dysbiosis in breast cancer and racial differences are also reported. The review also provides an update on breast cancer's current diagnostics and treatment modalities.
Collapse
Affiliation(s)
- Sapthala P Loku Galappaththi
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Kelly R Smith
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Enas S Alsatari
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Rachel Hunter
- Department of Surgery, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Donna L Dyess
- Department of Surgery, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Elba A Turbat-Herrera
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
| | - Santanu Dasgupta
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36604, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36688, USA
- Department of Biochemistry and Molecular Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| |
Collapse
|
25
|
Lu Y, Zhen Y, Li Z, Luo B, Yin B, Zhang L. Discovery of a novel Fam20C inhibitor for treatment of triple-negative breast cancer. Int J Biol Macromol 2024; 286:138398. [PMID: 39647747 DOI: 10.1016/j.ijbiomac.2024.138398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/30/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Sequence similarity 20 family member C (Fam20C), a Golgi casein kinase, has a gradually elucidated mechanism in triple-negative breast cancer (TNBC) and is considered a possible target for therapeutic intervention. In this study, we combined virtual screening and chemical synthesis methods to obtain a new small-molecule Fam20C inhibitor, compound 5k, which possesses desirable kinase inhibitory activity against Fam20C and significant anti-proliferative activity against MDA-MB-231 and BT-549 cells. Subsequently, cellular thermal shift assay (CETSA), molecular docking, and molecular dynamics (MD) simulations revealed that compound 5k binds to Fam20C. Moreover, compound 5k showed favorable antitumor efficacy in TNBC cells and xenograft models by promoting apoptosis and inhibiting migration. Mechanistically, compound 5k can inhibit the proliferation, promote apoptosis, and inhibit migration of TNBC cells by targeting Fam20C, thereby inhibiting the deterioration of TNBC and preventing its progression. Taken together, these results suggest that compound 5k can be utilized as a novel Fam20C inhibitor, laying a foundation for the discovery of more small-molecule drugs for the treatment of TNBC in the future.
Collapse
Affiliation(s)
- Yingying Lu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yongqi Zhen
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Boqin Luo
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Yin
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
26
|
Cortés A, López-Miranda E, Fernández-Ortega A, Carañana V, Servitja S, Urruticoechea A, Lema-Roso L, Márquez A, Lazaris A, Alcalá-López D, Mina L, Gener P, Rodríguez-Morató J, Antonarelli G, Llombart-Cussac A, Pérez-García J, Cortés J. Olaparib monotherapy in advanced triple-negative breast cancer patients with homologous recombination deficiency and without germline mutations in BRCA1/2: The NOBROLA phase 2 study. Breast 2024; 78:103834. [PMID: 39520738 PMCID: PMC11585816 DOI: 10.1016/j.breast.2024.103834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE To evaluate olaparib in advanced triple negative breast cancer (TNBC) patients with homologous recombination deficiency (HRD) and no germline BRCA1/2 mutations (gBRCA1/2mut). METHODS NOBROLA (NCT03367689) is a single-arm, open-label, multicenter, phase IIa trial, enrolling adult patients with advanced TNBC without gBRCA1/2mut and with HRD, who were treated with olaparib. The primary endpoint was clinical benefit rate (CBR) per RECIST v.1.1. RESULTS Six of 114 patients were eligible and received olaparib. Median follow up was 8.5 months. CBR and overall response rate (ORR) were 50 % (95 % CI, 11.8-88.2). CONCLUSIONS The observed results could prompt further investigation. TRIAL ClinicalTrials.gov identifier NCT03367689.
Collapse
Affiliation(s)
| | - Elena López-Miranda
- Hospital Universitario Ramón y Cajal, Madrid, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA
| | | | - Vicente Carañana
- Department of Medical Oncology, Hospital Arnau de Vilanova, Valencia, Spain
| | | | | | | | - Antonia Márquez
- UGCI Oncología Médica, Hospital Universitario Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Alexandros Lazaris
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA
| | - Daniel Alcalá-López
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA
| | - Leonardo Mina
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA
| | - Petra Gener
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA
| | - Jose Rodríguez-Morató
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA
| | - Gabriele Antonarelli
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy; Division of Early Drug Development for Innovative Therapy, European Institute of Oncology, IRCCS, Milan, Italy
| | - Antonio Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA; Department of Medical Oncology, Hospital Arnau de Vilanova, Valencia, Spain.
| | - José Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA; International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
| | - Javier Cortés
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Ridgewood, New Jersey, USA; International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain; Universidad Europea de Madrid, Faculty of Biomedical and Health Sciences, Department of Medicine, Madrid, Spain
| |
Collapse
|
27
|
van den Driest L, Kelly P, Marshall A, Johnson CH, Lasky-Su J, Lannigan A, Rattray Z, Rattray NJ. A gap analysis of UK biobank publications reveals SNPs associated with intrinsic subtypes of breast cancer. Comput Struct Biotechnol J 2024; 23:2200-2210. [PMID: 38817965 PMCID: PMC11137368 DOI: 10.1016/j.csbj.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 06/01/2024] Open
Abstract
Breast cancer is a multifaceted disease and a leading cause of cancer morbidity and mortality in females across the globe. In 2020 alone, 2.3 million women were diagnosed and 685,000 died of breast cancer worldwide. With the number of diagnoses projected to increase to 3 million per year by 2040 it is essential that new methods of detection and disease stratification are sought to decrease this global cancer burden. Although significant improvements have been made in breast cancer diagnosis and treatment, the prognosis of breast cancer remains poor in some patient groups (i.e. triple negative breast cancer), necessitating research into better patient stratification, diagnosis and drug discovery. The UK Biobank, a comprehensive biomedical and epidemiological database with a wide variety of multiomics data (genomics, proteomics, metabolomics) offers huge potential to uncover groundbreaking discoveries in breast cancer research leading to improved patient stratification. Combining genomic, proteomic, and metabolic profiles of breast cancer in combination with histological classification, can aid treatment decisions through accurate diagnosis and prognosis prediction of tumor behaviour. Here, we systematically reviewed PubMed publications reporting the analysis of UK Biobank data in breast cancer research. Our analysis of UK Biobank studies in the past five years identified 125 publications, of which 76 focussed on genomic data analysis. Interestingly, only two studies reported the analysis of metabolomics and proteomics data, with none performing multiomics analysis of breast cancer. A meta-analysis of the 76 publications identified 2870 genetic variants associated with breast cancer across 445 genes. Subtype analysis revealed differential genetic alteration in 13 of the 445 genes and the identification of 59 well-established breast cancer genes. in differential pathways. Pathway interaction analyses illuminated their involvement in general cancer biomolecular pathways (e.g. DNA damage repair, Gene expression). While our meta-analysis only measured genetic differences in breast cancer due to current usage of UK Biobank data, minimal multi-omics analyses have been performed and the potential for harnessing multi-omics strategies within the UK Biobank cohort holds promise for unravelling the biological signatures of distinct breast cancer subtypes further in the future.
Collapse
Affiliation(s)
- Lisa van den Driest
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Patricia Kelly
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Alan Marshall
- School of Social and Political Science, University of Edinburgh, Chrystal Macmillan Building, George Square, Edinburgh EH8 9LD, UK
| | - Caroline H. Johnson
- Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06510, USA
| | - Jessica Lasky-Su
- Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA 02115, USA
| | - Alison Lannigan
- NHS Lanarkshire, Lanarkshire, Scotland, UK
- Wishaw General Hospital, NHS Lanarkshire, 50 Netherton St, Wishaw ML2 0DP, UK
| | - Zahra Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
- NHS Lanarkshire, Lanarkshire, Scotland, UK
| | - Nicholas J.W. Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
- NHS Lanarkshire, Lanarkshire, Scotland, UK
| |
Collapse
|
28
|
Record SM, Thomas SM, Dalton J, van den Bruele AB, Chiba A, DiLalla G, DiNome ML, Rosenberger LH, Woriax HE, Hwang ES, Plichta JK. Triple-Negative Breast Cancer in Older Patients: Does SLNB Guide Therapy? Ann Surg Oncol 2024; 31:8802-8812. [PMID: 39198350 DOI: 10.1245/s10434-024-16106-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Older breast cancer patients represent a heterogeneous population. Studies demonstrate that sentinel lymph node biopsy (SLNB) omission may be appropriate in some clinical scenarios, yet patients with triple-negative breast cancer (TNBC) are often excluded from these studies. This study evaluated differences in treatment and survival for older patients with TNBC based on SLNB receipt and result. METHODS Patients 70 years old or older with a diagnosis of cT1-2/cN0/M0 TNBC (2010-2019) who underwent surgery were selected from the National Cancer Database. Logistic regression estimated the association of SLNB with therapy, and Cox proportional hazards models estimated the association of SLNB with overall survival (OS) after adjustment for select factors. RESULTS Of the 15,167 patients included in the study (median age, 77 years), 13.02% did not undergo SLNB, 5.14% had pN1 disease, 0.12% had pN2 disease, and 0.01% had pN3 disease. Most of the patients (83.9%) underwent surgery first, and 16.1% received neoadjuvant chemotherapy. Of those who underwent surgery first and SLNB, 6.2% had pN+ disease. Receipt of SLNB was associated with a higher likelihood of chemotherapy (odds ratio [OR] 1.85; 95% confidence interval [CI] 1.55-2.21), regardless of pN status. Compared with those who did not undergo a SLNB, a negative SLNB was significantly associated with lower mortality (hazard ratio [HR] 0.68; 95% CI 0.63-0.75), although there was no difference for a positive SLNB (HR 1.14; 95% CI 0.98-1.34). The patients receiving chemotherapy first showed no difference in survival based on SLNB receipt or result (p = 0.23). CONCLUSIONS Most older patients with TNBC do not have nodal involvement and do not receive chemotherapy. The receipt and results of SLNB may be associated with outcomes for some who undergo surgery first, but not for those who receive neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Sydney M Record
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Samantha M Thomas
- Duke Cancer Institute, Duke University, Durham, NC, USA
- Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Juliet Dalton
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Astrid Botty van den Bruele
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Akiko Chiba
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Gayle DiLalla
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Maggie L DiNome
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Laura H Rosenberger
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Hannah E Woriax
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Jennifer K Plichta
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
- Duke Cancer Institute, Duke University, Durham, NC, USA.
- Department of Population Health Sciences, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
29
|
Liu J, Lyu Q, Wu M, Zhou Y, Wang T, Zhang Y, Fan N, Yang C, Wang W. Integrating mTOR Inhibition and Photodynamic Therapy Based on Carrier-Free Nanodrugs for Breast Cancer Immunotherapy. Adv Healthc Mater 2024; 13:e2402357. [PMID: 39235716 PMCID: PMC11650419 DOI: 10.1002/adhm.202402357] [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: 06/27/2024] [Revised: 08/07/2024] [Indexed: 09/06/2024]
Abstract
Conventional photodynamic therapy (PDT) in cancer treatment needs to utilize oxygen to produce reactive oxygen species to eliminate malignant tissues. However, oxygen consumption in tumor microenvironment exacerbates cancer cell hypoxia and may promote vasculature angiogenesis. Since the mammalian target of rapamycin (mTOR) signaling pathway plays a vital role in endothelial cell proliferation and fibrosis, mTOR inhibitor drugs hold the potential to reverse hypoxia-evoked angiogenesis for improved PDT effect. In this study, a carrier-free nanodrug formulation composed of Torin 1 as mTORC1/C2 dual inhibitor and Verteporfin as a photosensitizer and Yes-associated protein inhibitor is developed. These two drug molecules can self-assemble into stable nanoparticles through π-π stacking and hydrophobic interactions with good long-term stability. The nanodrugs can prompt synergistic apoptosis, combinational anti-angiogenesis, and strong immunogenic cell death effects upon near-infrared light irradiation in vitro. Furthermore, the nanosystem also exhibits improved antitumor effect, anti-cancer immune response, and distant tumor inhibition through tumor microenvironment remodeling in vivo. In this way, the nanodrugs can reverse PDT-elicited angiogenesis and promote cancer immunotherapy to eliminate tumor tissues and prevent metastasis. This nanosystem provides insights into integrating mTOR inhibitors and photosensitizers for safe and effective breast cancer treatment in clinical settings.
Collapse
Affiliation(s)
- Jinzhao Liu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Qingyang Lyu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Meicen Wu
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Yang Zhou
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Tianyi Wang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Yichi Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Ni Fan
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Chang Yang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical BiotechnologyThe University of Hong KongHong Kong999077China
- Department of Pharmacology and PharmacyLi Ka Shing Faculty of MedicineThe University of Hong KongHong Kong999077China
- Dr. Li Dak‐Sum Research CentreThe University of Hong KongHong Kong999077China
| |
Collapse
|
30
|
Singh MT, Thaggikuppe Krishnamurthy P, Magham SV. Harnessing the synergistic potential of NK1R antagonists and selective COX-2 inhibitors for simultaneous targeting of TNBC cells and cancer stem cells. J Drug Target 2024; 32:258-269. [PMID: 38252517 DOI: 10.1080/1061186x.2024.2309568] [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: 09/26/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
Triple-negative breast cancer (TNBC) lacks the expression of oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), rendering it unresponsive to endocrine therapy and HER2 targeted treatments. Though certain chemotherapeutics targeting the cell cycle have shown efficacy to a certain extent, the presence of chemotherapy-resistant cancer stem cells (CSCs) presents a significant challenge in tackling TNBC. Multiple lines of evidence suggest the upregulation of neuropeptide Substance P (SP), its NK-1 receptor (NK1R) and the Cyclooxygenase-2 (COX-2) enzyme in TNBC patients. Upregulation of the SP/NK1R system and COX-2 influences major signalling pathways involved in cell proliferation, growth, survival, angiogenesis, inflammation, metastasis and stem cell activity. The simultaneous activation and crosstalk between the pathways activated by SP/NK1R and COX-2 consequently increase the levels of key regulators of self-renewal pathways in CSCs, promoting stemness. The combination therapy with NK1R antagonists and COX-2 inhibitors can simultaneously target TNBC cells and CSCs, thereby enhancing treatment efficacy and reducing the risk of recurrence and relapse. This review discusses the rationale for combining NK1R antagonists and COX-2 inhibitors for the better management of TNBC and a novel strategy to deliver drug cargo precisely to the tumour site to address the challenges associated with off-target binding.
Collapse
Affiliation(s)
- Madhu Tanya Singh
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Sai Varshini Magham
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
31
|
Huang F, Wang F, Hu Q, Li Y, Jiang D. PTGR1-mediated immune evasion mechanisms in late-stage triple-negative breast cancer: mechanisms of M2 macrophage infiltration and CD8 + T cell suppression. Apoptosis 2024; 29:2002-2024. [PMID: 39068625 DOI: 10.1007/s10495-024-01991-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by metabolic dysregulation. Tumor cell immune escape plays an indispensable role in the development of TNBC tumors. Furthermore, in the abstract, we explicitly mention the techniques used and enhance the clarity and impact of our findings. "Based on bioinformatics analysis results, we utilized CRISPR/Cas9 technology to knockout the target gene and established a mouse model of breast cancer. Through experiments such as CCK8, scratch assay, and Transwell assay, we further investigated the impact of target gene knockout on the malignant behavior of tumor cells. Subsequently, we conducted immunohistochemistry and Western Blot experiments to study the expression of macrophage polarization and infiltration-related markers and evaluate the effect of the target gene on macrophage polarization. Next, through co-culture experiments, we simulated the tumor microenvironment and used immunohistochemistry staining to observe and analyze the distribution and activation status of M2 macrophages and CD8+ T cells in the co-culture system. We validated in vivo experiments the molecular mechanism by which the target gene regulates immune cell impact on TNBC progression.
Collapse
Affiliation(s)
- Fang Huang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China
| | - Fuhe Wang
- Department of General surgery, Hebei Yiling Hospital, Shijiazhuang, 050000, P. R. China
| | - Qilu Hu
- Department of Radiotherapy, Heze Traditional Chinese Medicine Hospital, Heze, 274008, P. R. China
| | - Ying Li
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China
| | - Da Jiang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China.
| |
Collapse
|
32
|
Bhamidipati P, Nagaraju GP, Malla R. Immunoglobulin-binding protein and Toll-like receptors in immune landscape of breast cancer. Life Sci 2024; 358:123196. [PMID: 39481836 DOI: 10.1016/j.lfs.2024.123196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/29/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Breast cancer (BC) is a complex disease exhibiting significant heterogeneity and encompassing various molecular subtypes. Among these, triple-negative breast cancer (TNBC) stands out as one of the most challenging types, characterized by its aggressive nature and poor prognosis. This review embarks on a comprehensive exploration of the immune landscape of BC, with a primary focus on the functional and structural characterization of immunoglobulin-binding protein (BiP) and its pivotal role in regulating the unfolded response (UPR) pathway of proteins. Moreover, we unravel the multifaceted functions of BiP in BC, with a special emphasis on the involvement of cell surface BiP in TNBC metastasis, drug resistance, and its contribution to the formation of the tumor microenvironment (TME). We also provide mechanistic insights into how ER-resident BiP mediates the sensitization of drug-resistant BC to different treatment strategies, thereby offering promising avenues for therapeutic intervention. We also delve into the role of Toll-like receptors (TLRs), shedding light on their diverse expression patterns across BC and their influence on modulating the tumor immune response. Understanding the interplay between BiP, TLRs, and the immune response, especially in TNBC, opens avenues for novel immunotherapies. Future research should focus on developing targeted therapies that activate ER-resident BiP or inhibit cell surface BiP, and modulate TLR signaling. Moreover, exploring BiP as a biomarker for TNBC diagnosis, prognosis, and treatment response will be crucial for personalized medicine.
Collapse
Affiliation(s)
- Priyamvada Bhamidipati
- Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - RamaRao Malla
- Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India.
| |
Collapse
|
33
|
Sheng Y, Mills G, Zhao X. Identifying therapeutic strategies for triple-negative breast cancer via phosphoproteomics. Expert Rev Proteomics 2024:1-17. [PMID: 39588933 DOI: 10.1080/14789450.2024.2432477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Given the poor prognosis of patients with TNBC, it is urgent to identify new biomarkers and therapeutic targets to enable personalized treatment strategies and improve patient survival. Comprehensive insights beyond genomic and transcriptomic analysis are crucial to improved outcomes for patients. As proteins are the workhorses of cellular function with their activity primarily regulated by phosphorylation, advanced phosphoproteomics techniques, such as mass spectrometry and antibody arrays, are essential for elucidating kinase signaling pathways that drive TNBC progression and contribute to therapy resistance. AREA COVERED This review discusses the critical need to integrate phosphoproteomics into TNBC research, evaluates commonly used technologies and their applications, and explores their advantages and limitations. We highlight significant findings from phosphoproteomic analyses in TNBC and address the challenges of implementing these technologies into clinical practice. EXPERT OPINION Rapid advances in phosphoproteomics analysis facilitate subtype stratification, adaptive response monitoring, and identification of biomarkers and therapeutic targets in TNBC. However, challenges in analyzing protein phosphorylation, especially in deep spatially resolved analysis of malignant cells and the tumor ecosystem, hinder the translation of phosphoproteomics to the CLIA setting. Nonetheless, phosphoproteomics offers a powerful tool that, when integrated into routine clinical practice, has the potential to revolutionize patient care.
Collapse
Affiliation(s)
- Yuhan Sheng
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gordon Mills
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Xuejiao Zhao
- Division of Oncological Sciences Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
34
|
Ramadan WS, Alseksek RK, Mouffak S, Talaat IM, Saber-Ayad MM, Menon V, Ilce BY, El-Awady R. Impact of HDAC6-mediated progesterone receptor expression on the response of breast cancer cells to hormonal therapy. Eur J Pharmacol 2024; 983:177001. [PMID: 39284403 DOI: 10.1016/j.ejphar.2024.177001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/12/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Modulation of estrogen receptor (ER) and progesterone receptor (PR) expression, as well as their emerging functional crosstalk, remains a potential approach for enhancing the response to hormonal therapy in breast cancer. Aberrant epigenetic alterations induced by histone deacetylases (HDACs) were massively implicated in dysregulating the function of hormone receptors in breast cancer. Although much is known about the regulation of ER signaling by HDAC, the precise role of HDAC in modulating the expression of PR and its impact on the outcomes of hormonal therapy is poorly defined. Here, we demonstrate the involvement of HDAC6 in regulating PR expression in breast cancer cells. The correlation between HDAC6 and hormone receptors was investigated in patients' tissues by immunohistochemistry (n = 80) and publicly available data (n = 3260) from breast cancer patients. We explored the effect of modulating the expression of HDAC6 as well as its catalytic inhibition on the level of hormone receptors by a variety of molecular analyses, including Western blot, immunofluorescence, Real-time PCR, RNA-seq analysis and chromatin immunoprecipitation. Based on our in-silico and immunohistochemistry analyses, HDAC6 levels were negatively correlated with PR status in breast cancer tissues. The downregulation of HDAC6 enhanced the expression of PR-B in hormone receptor-positive and triple-negative breast cancer (TNBC) cells. The selective targeting of HDAC6 by tubacin resulted in the enrichment of the H3K9 acetylation mark at the PGR-B gene promoter region and enhanced the expression of PR-B. Additionally, transcriptomic analysis of tubacin-treated cells revealed enhanced activity of acetyltransferase and growth factor signaling pathways, along with the enrichment of transcription factors involved in the transcriptional activity of ER, underscoring the crucial role of HDAC6 in regulating hormone receptors. Notably, the addition of HDAC6 inhibitor potentiated the effects of anti-ER and anti-PR drugs mainly in TNBC cells. Together, these data highlight the role of HDAC6 in regulating PR expression and provide a promising therapeutic approach for boosting breast cancer sensitivity to hormonal therapy.
Collapse
Affiliation(s)
- Wafaa S Ramadan
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Rahma K Alseksek
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University City Road, Sharjah, 27272, United Arab Emirates
| | - Soraya Mouffak
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Iman M Talaat
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; Clinical Sciences Department, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; Pathology Department, Faculty of Medicine, Alexandria University, Champollion Street, Alexandria, 21131, Egypt
| | - Maha M Saber-Ayad
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; Clinical Sciences Department, College of Medicine, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Varsha Menon
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Burcu Yener Ilce
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates
| | - Raafat El-Awady
- Research Institute for Medical and Health Sciences, University of Sharjah, University City Road, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University City Road, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
35
|
Xu G, Liang Q, Gao L, Xu S, Luo W, Wu Q, Li J, Zhang Z, Liang H, Yang F. Developing an Arene Binuclear Ruthenium(II) Complex to Induce Ferroptosis and Activate the cGAS-STING Pathway: Targeted Inhibiting Growth and Metastasis of Triple Negative Breast Cancer. J Med Chem 2024; 67:19573-19585. [PMID: 39436824 DOI: 10.1021/acs.jmedchem.4c01908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
To effectively inhibit the growth and metastasis of triple-negative breast cancer (TNBC), we developed a high-efficiency and low-toxicity arene ruthenium (Ru) complex based on apoferritin (AFt). To achieve this, we optimized a series of Ru(II) 1,10-phenanthroline-2,9-diformaldehyde thiosemicarbazone complexes by studying their structure-activity relationships to obtain an arene binuclear Ru(II) complex (C5) with significant cytotoxicity and high accumulation in the mitochondria of tumor cells. Subsequently, a C5-AFt nanoparticle (NPs) delivery system was constructed. We found that the C5/C5-AFt NPs effectively inhibited TNBC growth and metastasis with few side effects. The C5-AFt NPs improved the anticancer and targeting abilities of C5 in vivo. Moreover, we confirmed the mechanism by which C5/C5-AFt NPs inhibit tumor growth and metastasis via mitochondrial damage-mediated ferroptosis and activation of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Qiongyue Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Lijuan Gao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shihang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Weicong Luo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Qiuming Wu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Jingyuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
- School of Pharmaceutical Sciences, Jiangxi Normal University, Nanchang, Jiangxi 330022, China
| |
Collapse
|
36
|
Xu L, Sun J, Guo J, Guo S, Li J, Tang Y, Liu X. Transcriptional factor KLF9 overcomes 5-fluorouracil resistance in breast cancer via PTEN-dependent regulation of aerobic glycolysis. J Chemother 2024:1-12. [PMID: 39491797 DOI: 10.1080/1120009x.2024.2421701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
The emergence of resistance to 5-Fluorouracil (5-FU) is a staple in breast cancer chemotherapy. This paper delves into the role of PTEN in breast cancer resistance to 5-FU and examines the underlying molecular pathways. PTEN expression was detected in bioinformatics databases and upstream transcription factors (TFs) were identified. PTEN mRNA and protein levels, aerobic glycolysis proteins, lactate production, glucose consumption, and cell viability were measured. Binding interactions were confirmed, and cell proliferation assessed. In breast cancer cells, PTEN expression was downregulated. PTEN overexpression counteracted 5-FU resistance through the suppression of aerobic glycolysis. KLF9, as a TF upstream of PTEN, enhanced the levels of PTEN. In conclusion, the TF KLF9 inhibits the aerobic glycolysis level of breast cancer cells by up-regulating PTEN expression, thereby reducing their resistance to 5-FU. The discovery of this mechanism provides a new theoretical basis for the treatment of breast cancer.
Collapse
Affiliation(s)
- Liang Xu
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Jing Sun
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Junlan Guo
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Shengnan Guo
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Jiangli Li
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Yijun Tang
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Xiaohui Liu
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| |
Collapse
|
37
|
Ye X, Cen Y, Li Q, Zhang Y, Li Q, Li J. Immunosuppressive SOX9-AS1 Resists Triple-Negative Breast Cancer Senescence Via Regulating Wnt Signalling Pathway. J Cell Mol Med 2024; 28:e70208. [PMID: 39550706 PMCID: PMC11569622 DOI: 10.1111/jcmm.70208] [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: 03/20/2024] [Revised: 10/07/2024] [Accepted: 11/02/2024] [Indexed: 11/18/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) are involved in the regulation of triple-negative breast cancer (TNBC) senescence, while pro-carcinogenic lncRNAs resist senescence onset leading to the failure of therapy-induced senescence (TIS) strategy, urgently identifying the key senescence-related lncRNAs (SRlncRNAs). We mined seven SRlncRNAs (SOX9-AS1, LINC01152, AC005152.3, RP11-161 M6.2, RP5-968 J1.1, RP11-351 J23.1 and RP11-666A20.3) by bioinformatics, of which SOX9-AS1 was reported to be pro-carcinogenic. In vitro experiments revealed the highest expression of SOX9-AS1 in MDA-MD-231 cells. SOX9-AS1 knockdown inhibited cell growth (proliferation, cycle and apoptosis) and malignant phenotypes (migration and invasion), while SOX9-AS1 overexpression rescued these effects. Additionally, SOX9-AS1 knockdown facilitated tamoxifen-induced cellular senescence and the transcription of senescence-associated secretory phenotype (SASP) factors (IL-1α, IL-1β, IL-6 and IL-8) mechanistically by resisting senescence-induced Wnt signal (GSK-3β/β-catenin) activation. Immune infiltration analysis revealed that low SOX9-AS1 expression was accompanied by a high infiltration of naïve B cells, CD8+ T cells and γδ T cells. In conclusion, SOX9-AS1 resists TNBC senescence via regulating the Wnt signalling pathway and inhibits immune infiltration. Targeted inhibition of SOX9-AS1 enhances SASP and thus mobilises immune infiltration to adjunct TIS strategy.
Collapse
Affiliation(s)
- Xuan Ye
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical UniversityGuangdong Provincial Clinical Research Center for Child HealthGuangzhouPR China
| | - Yi Cen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory DiseaseGuangzhou Medical UniversityGuangzhouPR China
| | - Quan Li
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical UniversityGuangdong Provincial Clinical Research Center for Child HealthGuangzhouPR China
| | - Yuan‐Ping Zhang
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical UniversityGuangdong Provincial Clinical Research Center for Child HealthGuangzhouPR China
| | - Qian Li
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical UniversityGuangdong Provincial Clinical Research Center for Child HealthGuangzhouPR China
| | - Jie Li
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical UniversityGuangdong Provincial Clinical Research Center for Child HealthGuangzhouPR China
| |
Collapse
|
38
|
Heinemann FS, Gershon PD. Differential Abundance of DNA Damage Sensors and Innate Immune Signaling Proteins in Inositol Polyphosphate 4-Phosphatase Type II-Negative Triple-Negative Breast Cancer Classified by Immunotype. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2212-2232. [PMID: 39147237 DOI: 10.1016/j.ajpath.2024.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024]
Abstract
The influence of neoplastic cells on the tumor microenvironment is poorly understood. In this study, eight patient samples representing two immunotypes of triple-negative breast cancer (TNBC), defined by quantitative histologic criteria as T-cell desert and T-cell infiltrated (TCI), were compared via label-free quantitative protein mass spectrometry of material extracted directly from targeted regions of formalin-fixed, paraffin-embedded tissue sections. Of 2934 proteins quantitated, 439 were significantly differentially abundant, among which 361 were overabundant in TCI-TNBC. The 361-protein group included proteins involved in major histocompatibility complex-I antigen processing and presentation, viral defense, DNA damage response, and innate immune signaling. Immunohistochemical validation of selected proteins showed good positive correlation between neoplastic cell histoscores and label-free quantitation. Extension of immunohistochemical analysis to a total of 58 inositol polyphosphate 4-phosphatase type II-negative TNBC confirmed elevated levels of the DNA damage sensor interferon-γ-inducible protein 16, inflammasome adaptor apoptosis-associated speck-like protein containing a CARD (ASC), and pore-forming protein gasdermin D in TCI-TNBC neoplastic cells. By contrast, cGMP-AMP synthase inhibitor barrier to autointegration factor (BAF) was elevated in the neoplastic cells of T-cell desert TNBC. These findings demonstrate a previously unknown correlation between the degree of T-cell infiltration in inositol polyphosphate 4-phosphatase type II-negative TNBC and the levels, in cognate neoplastic cells, of proteins that modulate innate immune signaling in response to DNA damage.
Collapse
Affiliation(s)
- F Scott Heinemann
- Department of Pathology, Hoag Memorial Hospital Presbyterian, Newport Beach, California.
| | - Paul D Gershon
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California.
| |
Collapse
|
39
|
Wang C, Fan P, Wang Q. Evolving therapeutics and ensuing cardiotoxicities in triple-negative breast cancer. Cancer Treat Rev 2024; 130:102819. [PMID: 39216183 DOI: 10.1016/j.ctrv.2024.102819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Defined as scarce expression of hormone receptors and human epidermal growth factor receptor 2, triple-negative breast cancer (TNBC) is labeled as the most heterogeneous subtype of breast cancer with poorest prognosis. Despite rapid advancements in precise subtyping and tailored therapeutics, the ensuing cancer therapy-related cardiovascular toxicity (CTR-CVT) could exert detrimental impacts to TNBC survivors. Nowadays, this interdisciplinary issue is incrementally concerned by cardiologists, oncologists and other pertinent experts, propelling cardio-oncology as a booming field focusing on the whole-course management of cancer patients with potential cardiovascular threats. Here in this review, we initially profile the evolving molecular subtyping and therapeutic landscape of TNBC. Further, we introduce various monitoring approaches of CTR-CVT. In the main body, we elaborate on typical cardiotoxicities ensuing anti-TNBC treatments in detail, ranging from chemotherapy (especially anthracyclines), surgery, anesthetics, radiotherapy to immunotherapy, with future perspectives on promising directions in the era of artificial intelligence and traditional Chinese medicine.
Collapse
Affiliation(s)
- Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong 226007, Jiangsu, China
| | - Pinchao Fan
- The First Clinical Medical College, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211112, Jiangsu, China
| | - Qingqing Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
40
|
Wu S, Ge A, Deng X, Liu L, Wang Y. Evolving immunotherapeutic solutions for triple-negative breast carcinoma. Cancer Treat Rev 2024; 130:102817. [PMID: 39154410 DOI: 10.1016/j.ctrv.2024.102817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Triple-negative breast carcinoma (TNBC) remains a formidable clinical hurdle owing to its high aggressiveness and scant therapeutic options. Nonetheless, the evolving landscape of immunotherapeutic strategies opens up promising avenues for tackling this hurdle. This review discusses the advancing immunotherapy for TNBC, accentuating personalized interventions due to tumor microenvironment (TME) diversity. Immune checkpoint inhibitors (ICIs) hold pivotal significance, both as single-agent therapies and when administered alongside cytotoxic agents. Moreover, the concurrent inhibition of multiple immune checkpoints represents a potent approach to augment the efficacy of cancer immunotherapy. Synergistic effects have been observed when ICIs are combined with targeted treatments like PARP inhibitors, anti-angiogenics, and ADCs (antibody-drug conjugates). Emerging tactics include tumor vaccines, cellular immunotherapy, and oncolytic viruses, leveraging the immune system's ability for selective malignant cell destruction. This review offers an in-depth examination of the diverse landscape of immunotherapy development for TNBC, furnishing meticulous insights into various advancements within this field. In addition, immunotherapeutic interventions offer hope for TNBC, needing further research for optimization.
Collapse
Affiliation(s)
- Shiting Wu
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Anqi Ge
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Xianguang Deng
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Lifang Liu
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China
| | - Yue Wang
- Department of Galactophore, the First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province 410007, China.
| |
Collapse
|
41
|
Sun P, Huang H, Ma JC, Feng B, Zhang Y, Qin G, Zeng W, Cui ZK. Repurposing propofol for breast cancer therapy through promoting apoptosis and arresting cell cycle. Oncol Rep 2024; 52:155. [PMID: 39364744 PMCID: PMC11465104 DOI: 10.3892/or.2024.8814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/17/2024] [Indexed: 10/05/2024] Open
Abstract
Breast cancer is the most prevalent cancer among women worldwide, characterized by a high mortality rate and propensity for metastasis. Although surgery is the standard treatment for breast cancer, there is still no effective method to inhibit tumor metastasis and improve the prognosis of patients with breast cancer after surgery. Propofol, one of the most widely used intravenous anesthetics in surgery, has exhibited a positive association with improved survival outcomes in patients with breast cancer post‑surgery. However, the underlying molecular mechanism remains to be elucidated. The present study revealed that triple negative breast cancer cells, MDA‑MB‑231 and 4T1, exposed to propofol exhibited a significant decrease in cell viability. Notably, propofol exhibited minimal cytotoxic effects on HUVECs under the same conditions. Furthermore, propofol significantly inhibited the migration and invasion ability of MDA‑MB‑231 and 4T1 cells. Propofol promoted apoptosis in 4T1 cells through upregulation of Bax and cleaved caspase 3, while downregulating B‑cell lymphoma‑extra large. Concomitantly, propofol induced cell cycle arrest of 4T1 cells by downregulating cyclin E2 and phosphorylated cell division cycle 6. Furthermore, propofol exhibited excellent anticancer efficacy in a 4T1 breast cancer allograft mouse model. The present study sheds light on the potential of propofol as an old medicine with a novel use for breast cancer treatment.
Collapse
Affiliation(s)
- Peng Sun
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Hanqing Huang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jian-Chao Ma
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Binyang Feng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yiqing Zhang
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zhong-Kai Cui
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
42
|
Lu B, Qiu R, Wei J, Wang L, Zhang Q, Li M, Zhan X, Chen J, Hsieh IY, Yang C, Zhang J, Sun Z, Zhu Y, Jiang T, Zhu H, Li J, Zhao W. Phase separation of phospho-HDAC6 drives aberrant chromatin architecture in triple-negative breast cancer. NATURE CANCER 2024; 5:1622-1640. [PMID: 39198689 DOI: 10.1038/s43018-024-00816-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/30/2024] [Indexed: 09/01/2024]
Abstract
How dysregulated liquid-liquid phase separation (LLPS) contributes to the oncogenesis of female triple-negative breast cancer (TNBC) remains unknown. Here we demonstrate that phosphorylated histone deacetylase 6 (phospho-HDAC6) forms LLPS condensates in the nuclei of TNBC cells that are essential for establishing aberrant chromatin architecture. The disordered N-terminal domain and phosphorylated residue of HDAC6 facilitate effective LLPS, whereas nuclear export regions exert a negative dominant effect. Through phase-separation-based screening, we identified Nexturastat A as a specific disruptor of phospho-HDAC6 condensates, which effectively suppresses tumor growth. Mechanistically, importin-β interacts with phospho-HDAC6, promoting its translocation to the nucleus, where 14-3-3θ mediates the condensate formation. Disruption of phospho-HDAC6 LLPS re-established chromatin compartments and topologically associating domain boundaries, leading to disturbed chromatin loops. The phospho-HDAC6-induced aberrant chromatin architecture affects chromatin accessibility, histone acetylation, RNA polymerase II elongation and transcriptional profiles in TNBC. This study demonstrates phospho-HDAC6 LLPS as an emerging mechanism underlying the dysregulation of chromatin architecture in TNBC.
Collapse
Affiliation(s)
- Bing Lu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ru Qiu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jiatian Wei
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Li Wang
- Department of Anesthesiology, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qinkai Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Mingsen Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Xiudan Zhan
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jian Chen
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - I-Yun Hsieh
- Shunde Hospital (The First People's Hospital of Shunde), Southern Medical University, Foshan, China
| | - Ciqiu Yang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Zhang
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zicheng Sun
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Yifan Zhu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Tao Jiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Han Zhu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.
- Department of Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | - Wei Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
43
|
Ali AA, Belali TM, Abu-Alghayth MH, Alyahyawi Y, Abalkhail A, Hazazi A, Nassar SA, Khan FR, Shmrany HA, Syed SM. Non-coding RNAs and estrogen receptor signaling in breast cancer: Nanotechnology-based therapeutic approaches. Pathol Res Pract 2024; 263:155568. [PMID: 39288475 DOI: 10.1016/j.prp.2024.155568] [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: 05/21/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024]
Abstract
This review investigates the regulatory role of non-coding RNAs (ncRNAs) in estrogen receptor (ER) signaling pathways, particularly in the context of breast cancer therapy, with an emphasis on the emerging potential of nanotechnology for drug delivery. The information was obtained from reputable databases, including PubMed, Elsevier, Springer, Wiley, Taylor, and Francis, which contain past and present research. Breast cancer remains the most prevalent cancer among women worldwide, and ER signaling mechanisms heavily influence its progression. Treatment options have traditionally encompassed surgery, chemotherapy, radiation therapy, targeted therapy, and hormone therapy. In recent decades, nanomedicine has emerged as a promising approach to breast cancer treatment. By passively targeting tumor cells and reducing toxicity, nanodrugs can overcome the challenges of conventional chemotherapy. Additionally, nanocarriers can stimulate tumor cells, enhancing treatment efficacy. Recent advancements in nanomedicine offer promising approaches for targeted cancer therapy, potentially overcoming the limitations of conventional treatments. This review explores the interactions between long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) with ER pathways, their impact on breast cancer progression, and how these interactions can be leveraged to enhance therapeutic efficacy through nanotechnology-based drug delivery systems.
Collapse
Affiliation(s)
- Amer Al Ali
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, University of Bisha, 255, Al Nakhil, Bisha 67714, Saudi Arabia
| | - Tareg M Belali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia
| | - Mohammed H Abu-Alghayth
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia
| | - Yara Alyahyawi
- Department of Medical Laboratory Technology, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, Buraydah 51452, P.O. Box 6666, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Somia A Nassar
- Department of Laboratory Medical Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Saudi Arabia; Department of Parasitology & Animal Diseases, National Research Centre, 33 Bohouth St., Dokki, Giza 12622, Egypt
| | - Farhan R Khan
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al- Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Humood Al Shmrany
- Department of Laboratory Medical Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Saudi Arabia.
| | - Shoaeb Mohammad Syed
- Department of Pharmaceutics, Dayanand College of Pharmacy, Barshi Road, Latur, MS, 413531, India.
| |
Collapse
|
44
|
García-Hernández AP, Corona DN, Carlos-Reyes Á, Sierra-Martínez M, Acosta-Altamirano G, Cisneros-Villanueva M, Pérez-Navarro Y, Ibarra-Sierra E, Marchat LA, López-Camarillo C. The lncRNA AFAP1-AS1 is upregulated in metastatic triple-negative breast tumors and controls hypoxia-activated vasculogenic mimicry and angiogenesis. BMC Cancer 2024; 24:1332. [PMID: 39472818 PMCID: PMC11523880 DOI: 10.1186/s12885-024-13019-6] [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: 06/18/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Vasculogenic mimicry (VM) is an alternative intratumoral microcirculation system that depends on the capacity of tumor cells to reorganize and grow in three-dimensional (3D) channel architectures like the capillaries formed by endothelial cells. Both VM and angiogenesis may coordinately function to feed cancer cells, allowing tumor growth. Long noncoding RNAs (lncRNAs) regulate critical cellular functions in cancer cells, including cell proliferation, apoptosis, angiogenesis, invasion, and metastasis. The lncRNA, known as actin filament-associated protein 1-antisense RNA 1 (AFAP1-AS1), has been described as an oncogene in diverse types of cancers. However, its role in VM and metastasis in triple-negative breast cancer (TNBC) is unknown. METHODS Reverse transcription and quantitative polymerase chain reaction (RT‒qPCR) experiments were performed to evaluate the expression of 10 selected lncRNAs from literature in metastatic and nonmetastatic biopsies from TNBC patients. The expression of AFAP1-AS1 was analyzed in Genotype-Tissue Expression Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) datasets. The AFAP1-AS1 expression was knocked in TNBC Hs578T cells by transfection of specific siRNAs. Channel-like formation assays were performed using 3D cultures over Matrigel in hypoxia-treated Hs578T cancer cells with diminished expression of AFAP1-AS1. The angiogenesis tests were conducted using human umbilical vein endothelial cells (HUVECs) and AFAP1-AS1- silenced Hs578T cells on 3D cell cultures. The presence of VM (CD31-/PAS+) in tumor tissues from TNBC patients with and without metastasis was assessed through immunohistochemistry using endothelial marker CD31 antibodies and periodic acid-Schiff (PAS) staining. RESULTS Compared with normal mammary tissues, AFAP1-AS1 expression was higher in breast cancer tissues. Moreover, AFAP1-AS1 expression was upregulated in the TNBC subtype compared to receptor-positive breast tumors. In addition, the expression of AFAP1-AS1 was correlated with the expression of the thirteen genes characteristic of a previously reported hypoxia signature. Interestingly, AFAP1-AS1 was upregulated in primary TNBC tumors from patients who developed metastasis compared with the nonmetastatic group. Functional analysis revealed that the knockdown of AFAP1-AS1 in Hs578T cells significantly impaired the hypoxia-induced VM, accompanied by a decrease in the development of 3D channel networks. Similarly, AFAP1-AS1 knockdown counteracts the angiogenic potential of cancer cells, as indicated by a reduction in the number of polygons, sprouting cells, and nodes in HUVEC cells. Remarkably, an increase in CD31-/PAS + staining of 3D channel networks in primary breast tumors from metastatic patients was found compared with the nonmetastatic group. Finally, we found that the number of blood vessels increased in the nonmetastatic group more than in the metastatic cohort. CONCLUSIONS Our data suggested that AFAP1-AS1 controls both VM and angiogenesis in Hs578T breast cancer cells and that increased metastasis is associated with VM in TNBC patients.
Collapse
Affiliation(s)
- Alejandra Paola García-Hernández
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290. Col. Del Valle, Ciudad de México, 03100, México
| | - David Núñez Corona
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290. Col. Del Valle, Ciudad de México, 03100, México
| | - Ángeles Carlos-Reyes
- Laboratorio de Onco-inmunobiologia, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Ciudad de México, México
| | - Mónica Sierra-Martínez
- Unidad de Investigación en Salud del Hospital Regional de Alta Especialidad de Ixtapaluca, IMSS- Bienestar, Ixtapaluca Estado de México, Ciudad de México, México
| | - Gustavo Acosta-Altamirano
- Dirección de Investigación, Hospital General de México "Dr. Eduardo Liceaga", Ciudad de México, México
| | | | - Yussel Pérez-Navarro
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290. Col. Del Valle, Ciudad de México, 03100, México
| | - Eloisa Ibarra-Sierra
- Departamento de Investigación, Instituto Estatal de Cancerología "Dr. Arturo Beltrán Ortega", Acapulco, Guerrero, México
| | - Laurence A Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de México, México
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, San Lorenzo 290. Col. Del Valle, Ciudad de México, 03100, México.
| |
Collapse
|
45
|
Pérez-Bermejo M, Caballero-Pascual M, Legidos-García ME, Martínez-Peris M, Casaña-Mohedo J, Llorca-Colomer F, Ventura I, Tomás-Aguirre F, Asins-Cubells A, Murillo-Llorente MT. Sacituzumab Govitecan in Triple Negative Breast Cancer: A Systematic Review of Clinical Trials. Cancers (Basel) 2024; 16:3622. [PMID: 39518062 PMCID: PMC11545346 DOI: 10.3390/cancers16213622] [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: 09/12/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Triple-negative breast cancer is difficult to treat due to the absence of hormone receptors and Her2neu. Sacituzumab govitecan is a new therapeutic approach that uses an antibody directed against the Trop-2 antigen present in solid epithelial tumors, linked to the active metabolite SN-38, similar to irinotecan, to specifically target cancer cells while minimizing damage to healthy cells. The objective of the present review was to evaluate the efficacy and safety of sacituzumab govitecan as a single treatment in patients with triple-negative breast cancer and to compare its results with the standard conventional chemotherapy regimen currently used in this disease. METHODS A systematic review of randomized clinical trials of sacituzumab govitecan was performed. The search was performed in Medline (PubMed), Web of Science, and Cochrane from September 2022 to January 2024. RESULTS Thirty-eight articles are included and evaluated according to inclusion and exclusion criteria corresponding to the two most relevant clinical trials, including specific analyses of cohorts and subgroup study arms within these trials. Data from more recent clinical trials are also reviewed. CONCLUSIONS The efficacy results showed a significantly greater clinical benefit with sacituzumab govitecan compared to standard chemotherapy in patients with triple-negative breast cancer. This drug will become a treatment of substantial impact in future treatment guidelines for this type of cancer.
Collapse
Affiliation(s)
- Marcelino Pérez-Bermejo
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| | - Mónica Caballero-Pascual
- School of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain;
| | - María Ester Legidos-García
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| | - Miriam Martínez-Peris
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| | - Jorge Casaña-Mohedo
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| | - Francisco Llorca-Colomer
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| | - Ignacio Ventura
- Molecular and Mitochondrial Medicine Research Group, School of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain;
| | - Francisco Tomás-Aguirre
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| | - Adalberto Asins-Cubells
- Centro de Salud de L’Eliana, Departamento Arnau de Vilanova-Lliria, C/Rosales, 23, L’Eliana, 46183 Valencia, Spain;
| | - María Teresa Murillo-Llorente
- SONEV Research Group, Faculty of Medicine and Health Sciences, Catholic University of Valencia, C/Quevedo Nº 2, 46001 Valencia, Spain; (M.E.L.-G.); (M.M.-P.); (J.C.-M.); (F.L.-C.); (F.T.-A.); (M.T.M.-L.)
| |
Collapse
|
46
|
Lu J, Zhu P, Zhang X, Zeng L, Xu B, Zhou P. tRNA-derived fragments: Unveiling new roles and molecular mechanisms in cancer progression. Int J Cancer 2024; 155:1347-1360. [PMID: 38867475 DOI: 10.1002/ijc.35041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
tRNA-derived fragments (tRFs) are novel small noncoding RNAs (sncRNAs) that range from approximately 14 to 50 nt. They are generated by the cleavage of mature tRNAs or precursor tRNAs (pre-tRNAs) at specific sites. Based on their origin and length, tRFs can be classified into three categories: (1) tRF-1 s; (2) tRF-3 s, tRF-5 s, and internal tRFs (i-tRFs); and (3) tRNA halves. They play important roles in stress response, signal transduction, and gene expression processes. Recent studies have identified differential expression of tRFs in various tumors. Aberrantly expressed tRFs have critical clinical value and show promise as new biomarkers for tumor diagnosis and prognosis and as therapeutic targets. tRFs regulate the malignant progression of tumors via various mechanisms, primarily including modulation of noncoding RNA biogenesis, global chromatin organization, gene expression regulation, modulation of protein translation, regulation of epigenetic modification, and alternative splicing regulation. In conclusion, tRF-mediated regulatory pathways could present new avenues for tumor treatment, and tRFs could serve as promising therapeutic targets for cancer therapy.
Collapse
Affiliation(s)
- Jingjing Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Clinical Medical Research Center, Affiliated Hospital of Nantong University, Nantong, China
| | - Ping Zhu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiufen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Oncology Institute, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Linzi Zeng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Bujie Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ping Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Pachuau L, Lalremmawia H, Ralte L, Vanlalpeka J, Pautu JL, Chenkual S, Zomuana T, Lalruatfela ST, Zohmingthanga J, Chhakchhuak L, Varma AK, Kumar NS. Uncovering novel pathogenic variants and pathway mutations in triple-negative breast cancer among the endogamous mizo tribe. Breast Cancer Res Treat 2024:10.1007/s10549-024-07501-9. [PMID: 39384723 DOI: 10.1007/s10549-024-07501-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024]
Abstract
PURPOSE The incidence of triple-negative breast cancer (TNBC) in India is higher compared to Western populations. The objective of this study is to identify novel and less reported variants in TNBC in Mizoram, a state with a high cancer incidence in India. METHODS We analysed whole exome sequencing data from triple-negative breast cancer (TNBC) patients in the Mizo population to identify key and novel variants. Moreover, we analysed reported breast cancer-related genes and pathway alterations. RESULTS Somatic mutation analysis revealed that TP53 was the most frequently mutated gene and TP53, CACNA1E, IGSF3, RYR1, and FAM155A as significantly mutated driver genes. Based on the ACMG guidelines, we identified a rare pathogenic germline variant of BRCA1 (p.C1697R) in 13% and a likely pathogenic frameshift insertion in RBMX (p.P106Ffs) in 73% of the patients. We also found that the ATM, STK11, and CDKN2A genes were significantly mutated in germline TNBC samples compared to healthy samples. Moreover, we identified novel somatic variants in CHEK2 (p.K182M) and NF1 (p.C245X), and novel germline variants RB1 (p.D111G), CDH1 (p.A10Gfs), CDKN2A (p.V96G), CDKN2A (p.S12Afs*22), MAP3K1 (CAAdelins0), MSH6 (p.L1226_L1230del), and PMS2 (TTCdelins0). Pathway analysis revealed that most somatic mutations were highly associated with PI3K-Akt signalling pathway and MAPK signalling pathways in TNBC. CONCLUSIONS These findings identified novel variants and key genes contributing to disease development and progression. Further analysis of less studied genes, including RBMX, MRC1, ATM, CTNNB1, and CDKN2A, in TNBC may reveal new potential genes for targeted therapeutic strategies and contribute to clinical advancements in the treatment of TNBC.
Collapse
Affiliation(s)
- Lalawmpuii Pachuau
- Department of Biotechnology, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
- Department of Pathology, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, Mizoram, 796001, India
| | - H Lalremmawia
- Department of Biotechnology, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Lalengkimi Ralte
- Department of Biotechnology, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Johan Vanlalpeka
- Department of Medicine, Zoram Medical College, Falkawn, Aizawl, Mizoram, 796 005, India
| | - Jeremy Lalrinsanga Pautu
- Department of Medical Oncology, Mizoram State Cancer Institute, Zemabawk, Aizawl, Mizoram, 796017, India
| | - Saia Chenkual
- Department of Surgery, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, 796001, Mizoram, India
- Zoram Medical College, Falkawn, Aizawl, Mizoram, 796 005, India
| | - Thomas Zomuana
- Department of Surgery, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, 796001, Mizoram, India
| | - Sailo Tlau Lalruatfela
- Department of Surgery, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, 796001, Mizoram, India
| | | | - Lalchhandama Chhakchhuak
- Department of Pathology, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, Mizoram, 796001, India
| | - Ashok K Varma
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Anushaktinagar, Maharastra, 400094, Mumbai, India
| | | |
Collapse
|
48
|
Chen RY, Ding LJ, Liu YJ, Shi JJ, Yu J, Li CY, Lu JF, Yang GJ, Chen J. Marine Staurosporine Analogues: Activity and Target Identification in Triple-Negative Breast Cancer. Mar Drugs 2024; 22:459. [PMID: 39452867 PMCID: PMC11509616 DOI: 10.3390/md22100459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with high mortality and drug resistance and no targeted drug available at present. Compound 4, a staurosporine alkaloid derived from Streptomyces sp. NBU3142 in a marine sponge, exhibits potent anti-TNBC activity. This research investigated its impact on MDA-MB-231 cells and their drug-resistant variants. The findings highlighted that compound 4 inhibits breast cancer cell migration, induces apoptosis, arrests the cell cycle, and promotes cellular senescence in both regular and paclitaxel-resistant MDA-MB-231 cells. Additionally, this study identified mitogen-activated protein kinase kinase kinase 11 (MAP3K11) as a target of compound 4, implicating its role in breast tumorigenesis by affecting cell proliferation, migration, and cell cycle progression.
Collapse
Affiliation(s)
- Ru-Yi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Li-Jian Ding
- School of Pharmacy, Health Science Center, Ningbo University, Ningbo 315211, China;
| | - Yan-Jun Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Jin-Jin Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Jing Yu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo 315211, China; (R.-Y.C.); (Y.-J.L.); (J.-J.S.); (J.Y.); (C.-Y.L.); (J.-F.L.)
| |
Collapse
|
49
|
Yang Y, Li H, Yang W, Shi Y. Improving efficacy of TNBC immunotherapy: based on analysis and subtyping of immune microenvironment. Front Immunol 2024; 15:1441667. [PMID: 39430759 PMCID: PMC11487198 DOI: 10.3389/fimmu.2024.1441667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/10/2024] [Indexed: 10/22/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive type of breast cancer that encompasses several distinct subtypes. Recent advances in immunotherapy offer a promising future for the treatment of these highly heterogeneous and readily metastatic tumors. Despite advancements, the efficacy of immunotherapy remains limited as shown by unimproved efficacy of PD-L1 biomarker and limited patient benefit. To enhance the effectiveness of TNBC immunotherapy, we conducted investigation on the microenvironment, and corresponding therapeutic interventions of TNBC and recommended further investigation into the identification of additional biomarkers that can facilitate the subtyping of TNBC for more targeted therapeutic approaches. TNBC is a highly aggressive subtype with dismal long-term survival due to the lack of opportunities for traditional endocrine and targeted therapies. Recent advances in immunotherapy have shown promise, but response rates can be limited due to the heterogeneous tumor microenvironments and developed therapy resistance, especially in metastatic cases. In this review, we will investigate the tumor microenvironment of TNBC and corresponding therapeutic interventions. We will summarize current subtyping strategies and available biomarkers for TNBC immunotherapy, with a particular emphasis on the need for further research to identify additional prognostic markers and refine tailored therapies for specific TNBC subtypes. These efforts aim to improve treatment sensitivity and ultimately enhance survival outcomes for advanced-stage TNBC patients.
Collapse
Affiliation(s)
- Yalan Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Haifeng Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yanxia Shi
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
50
|
Sahu P, Camarillo IG, Dettin M, Zamuner A, Teresa Conconi M, Barozzi M, Giri P, Sundararajan R, Sieni E. Electroporation enhances cell death in 3D scaffold-based MDA-MB-231 cells treated with metformin. Bioelectrochemistry 2024; 159:108734. [PMID: 38762949 DOI: 10.1016/j.bioelechem.2024.108734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
Triple-negative breast cancer (TNBC), the most aggressive subtype of breast cancer lacks estrogen, progesterone, and HER2 receptors and hence, is therapeutically challenging. Towards this, we studied an alternate therapy by repurposing metformin (FDA-approved type-2 diabetic drug with anticancer properties) in a 3D-scaffold culture, with electrical pulses. 3D cell culture was used to simulate the tumor microenvironment more closely and MDA-MB-231, human TNBC cells, treated with both 5 mM metformin (Met) and 8 electrical pulses at 2500 V/cm, 10 µs (EP1) and 800 V/cm, 100 µs (EP2) at 1 Hz were studied in 3D and 2D. They were characterized using cell viability, reactive oxygen species (ROS), glucose uptake, and lactate production assays at 24 h. Cell viability, as low as 20 % was obtained with EP1 + 5 mM Met. They exhibited 1.65-fold lower cell viability than 2D with EP1 + 5 mM Met. ROS levels indicated a 2-fold increase in oxidative stress for EP1 + 5 mM Met, while the glucose uptake was limited to only 9 %. No significant change in the lactate production indicated glycolytic arrest and a non-conducive environment for MDA-MB-231 growth. Our results indicate that 3D cell culture, with a more realistic tumor environment that enhances cell death using metformin and electrical pulses could be a promising approach for TNBC therapeutic intervention studies.
Collapse
Affiliation(s)
- Praveen Sahu
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Ignacio G Camarillo
- Deptartment of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
| | - Monica Dettin
- Department of Industrial Engineering, University of Padova, Padova 35122, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padova, Padova 35122, Italy; Department of Civil, Environmental, and Architectural Engineering, University of Padova, Italy
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| | - Marco Barozzi
- Department of Theoretical and Applied Sciences, University of Insubria, Varese 21100, Italy
| | - Pragatheiswar Giri
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Elisabetta Sieni
- Department of Theoretical and Applied Sciences, University of Insubria, Varese 21100, Italy.
| |
Collapse
|