1
|
Ko YS, Won JY, Jin H, Nguyen NB, Won Y, Nsanzimana V, Yun SP, Park SW, Kim HJ. ABCG8‑mediated sterol efflux increases cancer cell progression via the LRP6/Wnt/β‑catenin signaling pathway in radiotherapy‑resistant MDA‑MB‑231 triple‑negative breast cancer cells. Int J Mol Med 2025; 55:80. [PMID: 40116083 PMCID: PMC11964413 DOI: 10.3892/ijmm.2025.5521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/03/2025] [Indexed: 03/23/2025] Open
Abstract
Expression levels of ATP‑binding cassette (ABC) transporters are known to be increased in various tumor cells, including in breast cancer, and they are responsible for mediating drug resistance, leading to treatment failure. In the present study, gene expression array analysis revealed that among ABC transporter subtypes, ABC subfamily G member 8 (ABCG8) was one of the most increased in radiotherapy‑resistant triple‑negative breast cancer (RT‑R‑TNBC) cells compared with in TNBC cells. ABCG8 is involved in sterol efflux; however, its role in cancer is not well known. Therefore, the present study investigated the effect of ABCG8 on tumor progression in RT‑R‑TNBC cells. Gene expression profiling was conducted using the QuantiSeq 3' mRNA‑Seq Service, followed by western blotting to confirm protein levels. Loss‑of‑function assays using small interfering RNA (si) transfection were performed to assess the roles of ABCG8 and its regulatory signaling pathways. RT‑R‑MDA‑MB‑231 cells exhibited increased cholesterol levels in both cells and the surrounding media via induction of sterol regulatory element binding protein 1 (mature form) and fatty acid synthase. siABCG8 transfection increased intracellular cholesterol levels but decreased cholesterol levels in the media, indicating an accumulation of cholesterol inside cells. Additionally, RT‑R‑MDA‑MB‑231 cells exhibited increased levels of β‑catenin compared with MDA‑MB‑231 cells, which was significantly reduced by ABCG8 knockdown. Furthermore, ABCG8 knockdown led to cell cycle arrest in the G2/M phase in RT‑R‑MDA‑MB‑231 cells by reducing Polo‑like kinase 1 (PLK1) and Cyclin B1 expression. RT‑R‑MDA‑MB‑231 cells also exhibited increased phosphorylated‑low‑density lipoprotein (LDL) receptor‑related protein 6 (LRP6) levels compared with MDA‑MB‑231 cells, and these were decreased by siABCG8 transfection. LRP6 siRNA transfection decreased β‑catenin, PLK1 and Cyclin B1 expression. In addition, feedback mechanisms such as liver X receptor and inducible degrader of LDL were decreased in RT‑R‑MDA‑MB‑231 cells under normal conditions compared with in MDA‑MB‑231 cells. To the best of our knowledge, the present study was the first to suggest that the cholesterol exported by ABCG8, not inside the cells, may affect cancer progression via the LRP6/Wnt/β‑catenin signaling pathway in RT‑R‑TNBC. The regulation of this pathway may offer a potential therapeutic strategy for the treatment of RT‑R‑TNBC.
Collapse
Affiliation(s)
- Young Shin Ko
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Ju Yeong Won
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Hana Jin
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Nam Binh Nguyen
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Yaeram Won
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Vedaste Nsanzimana
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, College of Medicine, Institute of Medical Sciences, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Gyeongsangnam-do 52727, Republic of Korea
| |
Collapse
|
2
|
Dong C, Sun Y, Xu X, Li H, Song X, Wei W, Jiao C, Xu H, Liu Y, Mierzhakenmu Z, Li L, Ma B. c-Myc knockdown restores tamoxifen sensitivity in triple-negative breast cancer by reactivating the expression of ERα: the central role of miR-152 and miR-148a. Breast Cancer 2025; 32:529-542. [PMID: 40029493 DOI: 10.1007/s12282-025-01683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Poor prognosis of triple-negative breast cancer (TNBC) is owing to its intrinsic heterogeneity and lack of targeted therapies. Emerging evidence has characterized that targeting c-Myc might be a promising way to treat TNBC. METHODS c-Myc knocked down TNBC cells were generated and the tamoxifen sensitivity was determined. Methylation-specific PCR analysis was used to detect the methylation status of ERα promoter, and c-Myc-mediated miRNA transcription was examined using chromatin immunoprecipitation and dual-luciferase reporter assays. The in vivo tamoxifen sensitivity was determined by mouse xenograft model. RESULTS c-Myc knockdown in TNBC cells leads to the reactivation of ERα and consequent acquisition its sensitivity to tamoxifen. c-Myc depletion decreased the methylation in the promoter of ERα and DNMT1 was identified as the main executor. c-Myc knockdown-induced tamoxifen sensitivity was reversed by DNMT1 overexpression. The expression of miR-152-3p and miR-148a-3p was largely induced in c-Myc knockdown TNBC cells, and both miR-152-3p and miR-148a-3p could target DNMT1 to regulate its expression. c-Myc binds to the promoter regions of miR-152-3p and miR-148a-3p to exert transcriptional suppression. By suppressing miR-152-3p or miR-148a-3p expression using inhibitors, enhanced sensitivity to tamoxifen induced by c-Myc knockdown was partially reversed. In vivo xenograft tumor model demonstrated that c-Myc knockdown mildly inhibits the growth of tumor, and a dramatic decline was observed when administrated with tamoxifen combined with c-Myc knockdown. CONCLUSION Our study first illustrated that c-Myc knockdown in TNBC cells reactivate ERα expression in a miR-152/miR-148a-DNMT1-dependent manner, and brought new sights into treating TNBC using hormonal therapies.
Collapse
Affiliation(s)
- Chao Dong
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Yonghong Sun
- Department of Central Operating Room, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiaoli Xu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Huiling Li
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xinyu Song
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Wenxin Wei
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Chong Jiao
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Haoyi Xu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Yuanjing Liu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Zuliyaer Mierzhakenmu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Li Li
- Department of Gynecological Oncology (First Ward), The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| | - Binlin Ma
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
3
|
Aparajay P, Madhyastha H, Altamimi MA, Dev A, Hussain A, Bhowmik S. Functionalized Niosomes for Co-Delivery of Curcumin and Imatinib Mesylate to Treat Breast Cancer: In Vitro and In Vivo Investigations. AAPS PharmSciTech 2025; 26:119. [PMID: 40301277 DOI: 10.1208/s12249-025-03102-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/29/2025] [Indexed: 05/01/2025] Open
Abstract
Breast cancer is notable for its aggressive mutations, high resistance, and delayed diagnosis. Traditional treatments often cause severe side effects, highlighting the need for targeted therapies. This study developed a targeted delivery system using folic acid and Arginylglycylaspartic acid (RGD)-modified niosomes to deliver hydrophilic imatinib mesylate (IM) and hydrophobic curcumin (C) to treat breast cancer. The formulations were prepared and characaterized for size, zet potential, polydispersity index, % entrapment efficiency, and morphology. Moreover, FTIR (Fourier Transform Infrared) study negated incompatibility. In vitro drug release study was carried out at two different pH. In vitro cytotoxicity (dose dependent and ROS activity) and in vivo bioavailability studies were conducted to generate a proof of concept. The dual drug-loaded niosomal vesicles (R-F-PL64oxNS@IM-C) were designed for effective delivery of IM and C having particle size (< 300 nm) with high zeta potential (- 18 mV). The formulation achieved high entrapment efficiency (>70%) for both drugs with sustained release over 36 h at the explored two pH. In vitro results using MCF- 7 cells revealed significant cell death by R-F-PL64oxNS@IM-C as compared to pure drugs (IM & C) through upregulation and downregulation of proapoptotic and antiapoptotic genes, respectively. In vivo studies showed approximately 1.9- and 5-fold higher biodistribution of C and IM, respectively using targeted niosomal systems as compared to pure drugs. The pharmacokinetic analysis revealed that Cmax and AUC of IM from R-F-PL64oxNS@IM and C from R-F-PL64oxNS@IM-C were significantly higher compared to pure IM and curcumin. Moreover, the Tmax had also increased for both IM (3 h) and C (3 h) using RGD and folic acid guided niosomal formulation suggesting its enhanced retention in systemic circulation leading to more bioavailability as compared to IM (0.5 h) and C (0.5 h). The targeted delivery also led to significant reduction in TNF-α levels, indicating improved therapeutic potential. The developed R-F-PL64oxNS@IM-C shown more precisely killing of breast cancer cell than pure IM and C.
Collapse
Affiliation(s)
- Priyadarshi Aparajay
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki, 8891692, Japan
| | - Mohammad A Altamimi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| | - Afzal Hussain
- Department of Pharmaceutics, College of Pharmacy, King Saud University, 11451, Riyadh, Saudi Arabia.
| | - Shuvadip Bhowmik
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| |
Collapse
|
4
|
Kim M, Lee M, Lee A, Choi BO, Park WC, Kim SH, Lee J, Kang J. Correlating p53 immunostaining patterns with somatic TP53 mutation and functional properties of mutant p53 in triple-negative breast cancer. Histopathology 2025. [PMID: 40162573 DOI: 10.1111/his.15453] [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: 08/12/2024] [Revised: 01/21/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025]
Abstract
AIMS Immunohistochemical (IHC) staining of p53 is a potential marker for TP53 mutations in various cancers. However, criteria for predicting TP53 mutations in triple-negative breast cancer (TNBC) using p53 IHC staining are not yet established. We aim to correlate p53 IHC expression patterns with TP53 mutation status in TNBC. METHODS AND RESULTS A total of 113 TNBC cases were analysed for p53 IHC staining pattern and somatic TP53 mutation using whole-exome sequencing. Functional properties of TP53 mutations were determined using the National Cancer Institute (NCI) TP53 database. P53 IHC patterns were categorized as nuclear overexpression (n = 58), null pattern (n = 40), wildtype (n = 15), cytoplasmic (n = 5), and subclonal (n = 5). The cutoff for predictive p53 nuclear overexpression was determined to be 80%, which strongly correlated with TP53 mutations. Notably, p53 overexpression had a positive predictive value (PPV) of 83% for missense or in-frame mutations, while the null pattern showed a PPV of 85% for detecting nonsense, frameshift, or splicing mutations. P53 overexpression was significantly linked to missense mutations within the DNA-binding domain (DBD) exhibiting gain-of-function (GOF) or dominant-negative effect (DNE). Cases exhibiting cytoplasmic expression correlated with nonsense or frameshift mutations in the DBD, nuclear localization signal (NLS), or splice sites. Cases with subclonal p53 staining patterns were associated with TP53 mutations. CONCLUSION Our study proposes newly defined criteria for interpreting p53 immunostaining patterns in TNBC, potentially allowing for the prediction of TP53 mutation types and their functional implications.
Collapse
Affiliation(s)
- Meejeong Kim
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Miseon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Byung-Ock Choi
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Chan Park
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Hun Kim
- Department of Radiology, Seoul Saint Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
5
|
Ding R, Kan Q, Wang T, Xiao R, Song Y, Li D. Ginsenoside Rh2 regulates triple-negative breast cancer proliferation and apoptosis via the IL-6/JAK2/STAT3 pathway. Front Pharmacol 2025; 15:1483896. [PMID: 39845783 PMCID: PMC11751231 DOI: 10.3389/fphar.2024.1483896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/05/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is the most challenging subtype of breast cancer to treat. While previous studies have demonstrated that ginsenoside Rh2 induces apoptosis in TNBC cells, the specific molecular targets and underlying mechanisms remain poorly understood. This study aims to uncover the molecular mechanisms through which ginsenoside Rh2 regulates apoptosis and proliferation in TNBC, offering new insights into its therapeutic potential. Methods Network analysis and transcriptome sequencing were utilized to explore the potential mechanisms of ginsenoside Rh2 in treating TNBC. In vivo imaging and immunohistochemistry were employed to examine the effects of ginsenoside Rh2 in a TNBC mouse model. Functional assays were conducted to assess the impact of ginsenoside Rh2 on TNBC cell behavior. Additionally, ELISA, Western blot, and quantitative real-time PCR were used to further investigate the mechanisms of ginsenoside Rh2-induced apoptosis in TNBC cells. Results Through network analysis, 47 common targets were identified, and Gene Ontology (GO) enrichment analysis suggested that ginsenoside Rh2 may exert therapeutic effects in TNBC by influencing apoptosis, cell proliferation, and protein kinase activity. Both transcriptomic analysis and network analysis revealed the JAK/STAT signaling pathway as a key mechanism. Ginsenoside Rh2 inhibited tumor growth in TNBC mice and reduced the expression of IL- 6, IL-6R, STAT3, Bcl-2, and Bcl-xL in tumor tissues. The ability of ginsenoside Rh2 to inhibit TNBC cell proliferation was further confirmed by attenuating the activation of the IL-6/JAK2/STAT3 apoptosis pathway and reducing the expression of protein kinases AMPK-α1 and PKA-Cα. Conclusion Based on network analysis and experimental validation, our findings demonstrate that ginsenoside Rh2 regulates TNBC proliferation and apoptosis through suppression of the IL-6/JAK2/STAT3 pathway, both in vitro and in vivo. This comprehensive approach represents a significant advancement in understanding the therapeutic potential of ginsenoside Rh2 in treating TNBC.
Collapse
Affiliation(s)
| | | | | | | | | | - Duolu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Zhang Z, Gao J, Jia L, Kong S, Zhai M, Wang S, Li W, Wang S, Su Y, Li W, Zhu C, Wang W, Lu Y, Li W. Excessive glutathione intake contributes to chemotherapy resistance in breast cancer: a propensity score matching analysis. World J Surg Oncol 2024; 22:345. [PMID: 39709466 DOI: 10.1186/s12957-024-03626-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND We aim to explore the impact of excessive glutathione (GSH) intake on chemotherapy sensitivity in breast cancer. METHODS Clinicopathological data were collected from 460 breast cancer patients who underwent adjuvant chemotherapy from January 2016 to December 2019 from Zhengzhou University People's Hospital. The clinicopathological characteristics following GSH treatment were collected and compared with those in Non-GSH group after 1:2 propensity score matching (PSM). Intracellular GSH levels and the expression of antioxidant enzymes (NRF2, GPX4 and SOD1) were evaluated in tumor tissues in 51 patients receiving neoadjuvant chemotherapy. RESULTS The recurrence rate after adjuvant chemotherapy was significantly higher in the GSH group (n = 28, 31.8%) than that in the Non-GSH group (n = 39, 22.2%; P = 0.010). Additionally, patients in the HGSH group (high GSH intake, ≥ 16 days) exhibited an elevated recurrence rate compared to that in the LGSH group (low GSH intake, < 16 days) (n = 15 (46.8%) vs. n = 52 (22.4%); P = 0.003). Cox regression revealed that High GSH intake, Ki67 ≥ 30%, Triple negative and Lymphovascular invasion were independent risk factors of progression after adjuvant chemotherapy. Among patients receiving neoadjuvant chemotherapy, intracellular GSH levels and the expression levels of antioxidant enzymes (NRF2, GPX4 and SOD1) in the resistant patients were substantially higher (P < 0.001). CONCLUSIONS Excessive GSH intake may contribute to chemotherapy resistance in breast cancer, and the levels of intracellular GSH and antioxidant enzymes are elevated in resistant patients after neoadjuvant chemotherapy, indicating that the standardization of GSH intake may assist in reducing chemotherapy resistance.
Collapse
Affiliation(s)
- Zhiyuan Zhang
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Jiaru Gao
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Linjiao Jia
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Shuxin Kong
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Maosen Zhai
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Shuai Wang
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Wenwen Li
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Shoukai Wang
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Yuqing Su
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Wanyue Li
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, 450003, China
| | - Changzheng Zhu
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanxiang Lu
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China.
| | - Wentao Li
- Department of Breast Surgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, No.7 Weiwu Road, Zhengzhou, Henan, 450003, China.
- Henan Provincial Engineering Research Center of Breast Cancer Precise Prevention and Treatment, Zhengzhou, Henan, 450003, China.
| |
Collapse
|
7
|
Vashisth P, Smith CL, Amarasekara DL, Dasanyake GS, Singh G, Chism CM, Hamadani CM, Shaikh T, Grovich N, Gamboa B, Fitzkee NC, Hammer NI, Tanner EEL. Choline Carboxylic Acid Ionic Liquid-stabilized Anisotropic Gold Nanoparticles for Photothermal Therapy. ACS APPLIED NANO MATERIALS 2024; 7:26332-26343. [PMID: 39935958 PMCID: PMC11810124 DOI: 10.1021/acsanm.3c04645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Gold nanoparticles (AuNPs) are commonly used in cancer research due to their unique physical and optical properties. However, current AuNP synthesis methods often involve cytotoxic cationic surfactants like cetyltrimethyl ammonium bromide (CTAB). Tedious CTAB replacement methodologies have been used to increase the biocompatibility, further increasing the complexity of synthesizing biocompatible AuNPs limiting their biomedical applications. To address this issue, we explore cholinium decanoate (CADA) ionic liquid (IL) as a biocompatible stabilizing agent by replacing CTAB using a simple modified seeded method for synthesizing anisotropic AuNPs for photothermal therapy of triple-negative breast cancer cells (MDA-MB-231). The prepared CADA AuNPs showed quasi-spherical morphology, confirmed by transmission electron microscopy (TEM) and a broad plasmonic absorption band using Vis-NIR spectroscopy. CADA AuNPs exhibited excellent in-vitro biocompatibility with both MCF-10A (healthy human mammary cells) and MDA-MB-231 cells. We evaluate their in-vitro photothermal efficacy against MDA-MB-231 cancer cells, demonstrating significant cell death even at low AuNP concentration (20 μg/mL), low laser power density (0.6 W/cm2, 808 nm continuous laser), and short irradiation time of 5 minutes, primarily through apoptosis. Overall, this work represents the first effort in using a modified seeded method for synthesizing biocompatible IL-based anisotropic AuNPs for photothermal therapy, offering a promising avenue for future cancer treatment research using ILs.
Collapse
Affiliation(s)
- Priyavrat Vashisth
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Cameron L. Smith
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Dhanush L. Amarasekara
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Gaya S. Dasanyake
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Gagandeep Singh
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Claylee M. Chism
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Christine M. Hamadani
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Tanveer Shaikh
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Noah Grovich
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Briana Gamboa
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Nicholas C. Fitzkee
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Nathan I. Hammer
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Eden E. L. Tanner
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| |
Collapse
|
8
|
Panagopoulou M, Panou T, Gkountakos A, Tarapatzi G, Karaglani M, Tsamardinos I, Chatzaki E. BRCA1 & BRCA2 methylation as a prognostic and predictive biomarker in cancer: Implementation in liquid biopsy in the era of precision medicine. Clin Epigenetics 2024; 16:178. [PMID: 39643918 PMCID: PMC11622545 DOI: 10.1186/s13148-024-01787-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND BReast CAncer gene 1 (BRCA1) and BReast CAncer gene 2 (BRCA2) encode for tumor suppressor proteins which are critical regulators of the Homologous Recombination (HR) pathway, the most precise and important DNA damage response mechanism. Dysfunctional HR proteins cannot repair double-stranded DNA breaks in mammalian cells, a situation called HR deficiency. Since their identification, pathogenic variants and other alterations of BRCA1 and BRCA2 genes have been associated with an increased risk of developing mainly breast and ovarian cancer. Interestingly, HR deficiency is also detected in tumors not carrying BRCA1/2 mutations, a condition termed "BRCAness". MAIN TEXT One of the main mechanisms causing the BRCAness phenotype is the methylation of the BRCA1/2 promoters, and this epigenetic modification is associated with carcinogenesis and poor prognosis mainly among patients with breast and ovarian cancer. BRCA1 promoter methylation has been suggested as an emerging biomarker of great predictive significance, especially concerning Poly (ADP-ribose) Polymerase inhibitors (PARP inhibitor-PARPi) responsiveness, along with or beyond BRCA1/2 mutations. However, as its clinical exploitation is still insufficient, the impact of BRCA1/2 promoter methylation status needs to be further evaluated. The current review aims to gather the latest findings about the mechanisms that underline BRCA1/2 function as well as the molecular characteristics of tumors associated with BRCA1/2 defects, by focusing on DNA methylation. Furthermore, we critically analyze their translational meaning and the validity of BRCA methylation biomarkers in predicting treatment response. CONCLUSIONS We believe that BRCA1/2 methylation alone or combined with other biomarkers in a clinical setting is expected to change the scenery in prognosis and predicting treatment response in multiple cancer types and is worthy of further attention. The quantitative BRCA1 promoter methylation assessment might predict treatment response in PARPi and analysis of BRCA1/2 methylation in liquid biopsy might define patient subgroups at different time points that may benefit from PARPi. Finally, we suggest a pipeline that could be implemented in liquid biopsy to aid precision pharmacotherapy in BRCA-associated tumors.
Collapse
Grants
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
- TAEDR-0535850 European Union- Next-Generation EU, Greece 2.0 National Recovery and Resilience plan, National Flagship Initiative "Health and Pharmaceuticals"
Collapse
Affiliation(s)
- Maria Panagopoulou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece.
- Institute of Agri-Food and Life Sciences, University Research and Innovation Centre, Hellenic Mediterranean University, 71003, Heraklion, Greece.
| | - Theodoros Panou
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Anastasios Gkountakos
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Gesthimani Tarapatzi
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
| | - Makrina Karaglani
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
- Institute of Agri-Food and Life Sciences, University Research and Innovation Centre, Hellenic Mediterranean University, 71003, Heraklion, Greece
| | - Ioannis Tsamardinos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 70013, Heraklion, Greece
- Department of Computer Science, University of Crete, Voutes Campus, 70013, Heraklion, Greece
- Institute of Applied and Computational Mathematics, 70013, Heraklion, Greece
- JADBio Gnosis Data Analysis (DA) S.A., Science and Technology Park of Crete (STEPC), 70013, Heraklion, Greece
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, 68100, Alexandroupolis, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 70013, Heraklion, Greece
| |
Collapse
|
9
|
Bhattarai S, Rupji M, Chao HP, Xu Q, Saini G, Rida P, Aleskandarany MA, Green AR, Ellis IO, Janssen EA, Jonsdottir K, Rakha E, Kowalski J, Aneja R. Cell cycle traverse rate predicts long-term outcomes in a multi-institutional cohort of patients with triple-negative breast cancer. BJC REPORTS 2024; 2:87. [PMID: 39537757 PMCID: PMC11561184 DOI: 10.1038/s44276-024-00097-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/02/2024] [Accepted: 08/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Ki67 index (KI) and mitotic index (MI) are proliferation markers with established prognostic value in breast carcinomas. While KI is evaluated immunohistochemically and reported as a percentage, MI is determined visually and reflects total mitotic cells in 10 high-power fields. Our objective was to integrate KI and MI into a novel metric; the cell cycle traverse rate (CCTR). Given the lack of prognostic and predictive biomarkers in TNBC, we sought to assess the potential of CCTR as a risk-stratification tool for chemotherapy-treated TNBC patients from two independent cohorts: the Nottingham group (n = 124) and the Norway group (n = 71). METHODS We evaluated the ability of CCTR to predict survival after adjuvant chemotherapy for TNBC patients (n = 195) in two independent cohorts. Using immunohistochemistry and RNA sequencing, we determined the differences in immunohistochemical biomarkers, gene ontologies, molecular pathways and immune cell fractions based on CCTR. RESULTS TNBC shows a significantly lower median CCTR compared to luminal A (p < 0.01), luminal B (p < 0.01), and HER2+ samples (p < 0.01). CCTR outperformed both KI and MI in effectively risk-stratifying TNBC patients suggesting that combining KI and MI into a single metric, namely CCTR, could serve as a superior prognostic marker for Breast Cancer Specific Survival (BCSS) (p = 0.041). CCTR-high group exhibited enriched expression of various oncogenic signatures, including angiogenesis, epithelial-to-mesenchymal transition (EMT), Hedgehog signaling, hypoxia, Notch signaling, PI3K-AKT-mTOR signaling, TGFβ signaling, p53 signaling, and TNFα signaling via NFκB. These findings suggest the potential involvement of these pathways in the aggressiveness and clinical outcomes of TNBC patients. CONCLUSIONS Collectively, these findings suggest that CCTR offers superior predictive information compared to KI and MI alone with respect to long-term outcomes from adjuvant chemotherapy in patients with TNBC that may guide treatment decision making.
Collapse
Affiliation(s)
- Shristi Bhattarai
- Department of Molecular and Cellular Biology, Kennesaw State University, Kennesaw, GA, USA
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Manali Rupji
- Biostatistics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hsueh-Ping Chao
- Department of Oncology and Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA
| | - Qi Xu
- Department of Oncology and Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA
| | - Geetanjali Saini
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Mohammed A Aleskandarany
- Division of Biomedical and Forensic Sciences, School of Human Science, University of Derby, Derby, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Ian O Ellis
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Emiel A Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Kristin Jonsdottir
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Emad Rakha
- Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| | - Jeanne Kowalski
- Department of Oncology and Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA.
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, USA.
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
10
|
Karnatak M, Yadav P, Rathi K, Shukla M, Dugam P, Suthakaran S, Rawat V, Hassam M, Pandey A, Maurya RA, Sen D, Debnath S, Das A, Mukhija A, Verma VP. New hydrazide derivatives of N-amino-11-azaartemisinin as promising epidermal growth factor receptor inhibitors for therapeutic development in triple-negative breast cancer. Arch Pharm (Weinheim) 2024; 357:e2400466. [PMID: 39267485 DOI: 10.1002/ardp.202400466] [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: 06/08/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
Triple-negative breast cancer (TNBC) treatments, such as DNA-damaging agents like carboplatin, pose considerable human toxicity and may contribute to cancer relapse. Artemisinin derivatives offer a less toxic alternative; however, their specific role in TNBC management remains to be established. To address this gap, computational models were employed to design and evaluate artemisinin-based prototypes as potential TNBC therapeutics, aiming to provide safer and more effective treatment options for this aggressive cancer subtype. Among the series of hydrazide derivatives of azaartemisinin (10a-l) reported herein, compound 10j emerged as the most promising, exhibiting notable cytotoxicity with IC50 values of 1.74 and 1.64 µM against MDA-MB-231 and MDA-MB-468 cells, respectively. The clinically useful drug doxorubicin provided IC50 values of 0.29 and 0.29 µM against MDA-MB-231 and MDA-MB-468 cells, while artemisinin provided IC50 values of 107.30 and 116.60 µM, respectively. Furthermore, putative interactions between the synthesized compounds and the epidermal growth factor receptor (EGFR) were identified using molecular docking studies, suggesting a possible mechanism for their anticancer effect. Additionally, to determine the thermodynamic parameters of the interactions between artemisinin, azaartemisinin, and biomolecules, isothermal titration calorimetry experiments were performed. The binding constant value on the order of 104 indicates a comparatively stronger binding affinity of azaartemisinin with human serum albumin (HSA) compared to artemisinin with HSA. These findings support the potential of azaartemisinin derivatives as promising EGFR inhibitors for therapeutic development in TNBC, offering a new avenue for less toxic and more effective cancer treatments.
Collapse
Affiliation(s)
- Manvika Karnatak
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
| | - Priyanka Yadav
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
| | - Komal Rathi
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
| | - Monika Shukla
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
| | - Prachi Dugam
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, India
| | - Shruthi Suthakaran
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Varun Rawat
- Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Mohammad Hassam
- Chemveda Life Sciences Pvt. Ltd., Hyderabad, Telangana, India
| | - Aditi Pandey
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
| | - Ram Awatar Maurya
- Applied Organic Chemistry Group, Chemical Science and Technology Division, CSIR-North East Institute of Science & Technology, Jorhat, Assam, India
| | - Debanjan Sen
- BCDA College of Pharmacy & Technology, Hridaypur, Barasat, Kolkata, West Bengal, India
| | - Sudhan Debnath
- Department of Chemistry, Netaji Subhash Mahavidyalaya, Udaipur, Tripura, India
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Achal Mukhija
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
| | - Ved Prakash Verma
- Department of Chemistry, Banasthali University, Banasthali Newai, Rajasthan, India
- Department of Education in Science and Mathematics (DESM), Regional Institute of Education, Bhubaneshwar, Odisha, India
| |
Collapse
|
11
|
Li P, Zheng S, Leung HM, Liu LS, Chang TJH, Maryam A, Wang F, Chin YR, Lo PK. TNA-Mediated Antisense Strategy to Knockdown Akt Genes for Triple-Negative Breast Cancer Therapy. SMALL METHODS 2024; 8:e2400291. [PMID: 38779741 PMCID: PMC11579567 DOI: 10.1002/smtd.202400291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/11/2024] [Indexed: 05/25/2024]
Abstract
Triple-negative breast cancer (TNBC) remains a significant challenge in terms of treatment, with limited efficacy of chemotherapy due to side effects and acquired drug resistance. In this study, a threose nucleic acid (TNA)-mediated antisense approach is employed to target therapeutic Akt genes for TNBC therapy. Specifically, two new TNA strands (anti-Akt2 and anti-Akt3) are designed and synthesized that specifically target Akt2 and Akt3 mRNAs. These TNAs exhibit exceptional enzymatic resistance, high specificity, enhance binding affinity with their target RNA molecules, and improve cellular uptake efficiency compared to natural nucleic acids. In both 2D and 3D TNBC cell models, the TNAs effectively inhibit the expression of their target mRNA and protein, surpassing the effects of scrambled TNAs. Moreover, when administered to TNBC-bearing animals in combination with lipid nanoparticles, the targeted anti-Akt TNAs lead to reduced tumor sizes and decreased target protein expression compared to control groups. Silencing the corresponding Akt genes also promotes apoptotic responses in TNBC and suppresses tumor cell proliferation in vivo. This study introduces a novel approach to TNBC therapy utilizing TNA polymers as antisense materials. Compared to conventional miRNA- and siRNA-based treatments, the TNA system holds promise as a cost-effective and scalable platform for TNBC treatment, owing to its remarkable enzymatic resistance, inexpensive synthetic reagents, and simple production procedures. It is anticipated that this TNA-based polymeric system, which targets anti-apoptotic proteins involved in breast tumor development and progression, can represent a significant advancement in the clinical development of effective antisense materials for TNBC, a cancer type that lacks effective targeted therapy.
Collapse
Affiliation(s)
- Pan Li
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Shixue Zheng
- Tung Biomedical Sciences CentreDepartment of Biomedical SciencesCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Hoi Man Leung
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Ling Sum Liu
- Department of ChemistryMolecular Sciences Research HubImperial College LondonWhite City CampusWood LaneLondonW12 0BZU.K.
| | - Tristan Juin Han Chang
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Alishba Maryam
- Tung Biomedical Sciences CentreDepartment of Biomedical SciencesCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Fei Wang
- The Tenth Affiliated HospitalSouthern Medical University (Dongguan People's Hospital)Dongguan523059P. R. China
| | - Y. Rebecca Chin
- Tung Biomedical Sciences CentreDepartment of Biomedical SciencesCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
| | - Pik Kwan Lo
- Department of Chemistry and State Key Laboratory of Marine PollutionCity University of Hong KongTat Chee AvenueKowloonHong Kong SARP. R. China
- Key Laboratory of Biochip TechnologyBiotechand Health CareShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| |
Collapse
|
12
|
Chen JW, Gong RH, Teng C, Lin YS, Shen LS, Lin Z, Chen S, Chen GQ. Identification of a PANoptosis-related prognostic model in triple-negative breast cancer, from risk assessment, immunotherapy, to personalized treatment. Heliyon 2024; 10:e38732. [PMID: 39430460 PMCID: PMC11489348 DOI: 10.1016/j.heliyon.2024.e38732] [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: 05/26/2024] [Revised: 09/28/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
Background Triple-negative breast cancer is a breast cancer subtype characterized by its challenging prognosis, and establishing prognostic models aids its clinical treatment. PANoptosis, a recently identified type of programmed cell death, influences tumor growth and patient outcomes. Nonetheless, the precise impact of PANoptosis-related genes on the prognosis of triple-negative breast cancer has yet to be determined. Methods Clinical information for the triple-negative breast cancer samples was collected from the Gene Expression Omnibus and The Cancer Genome Atlas databases, while 19 PANoptosis-related genes were sourced from previous studies. We first categorized PANoptosis-related subtypes and determined the differentially expressed genes between them. Subsequently, we developed and validated a PANoptosis-associated predictive model using LASSO and Cox multivariate regression analyses. Statistical evaluations were conducted using R software, and the mRNA expression levels of the genes were quantified using real-time PCR. Results Using consensus clustering analysis, we divided triple-negative breast cancer patients into two clusters based on PANoptosis-related genes and identified 1054 differentially expressed genes between these clusters. Prognostic-related genes were subsequently selected to re-cluster patients, validating their predictive ability. A prognostic model was then constructed based on four genes: BTN2A2, CACNA1H, PIGR, and S100B. The expression and enriched cell types of these genes were examined and the expression levels were validated in vitro. Furthermore, the model was validated, and a nomogram was created to enhance personalized risk assessment. The risk score, proven to be an independent prognostic indicator for triple-negative breast cancer, showed a positive correlation with both age and disease stage. Immune infiltration and drug sensitivity analyses suggested appropriate therapies for different risk groups. Mutation profiles and pathway enrichment were analyzed, providing insights into potential therapeutic targets. Conclusion A PANoptosis-related prognostic model was successfully developed for triple-negative breast cancer, offering a novel approach for predicting patient prognosis and guiding treatment strategies.
Collapse
Affiliation(s)
- Jia-Wen Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Rui-Hong Gong
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| | - Chi Teng
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Yu-Shan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
| | - Li-Sha Shen
- Chongqing Academy of Chinese Materia Medica, Chongqing, 400065, China
| | - Zesi Lin
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Sibao Chen
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| | - Guo-Qing Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China
| |
Collapse
|
13
|
Hou Y, Hao H, Yuan Y, Zhang J, Liu Z, Nie Y, Zhang S, Yuan S, Yang M. Nifuratel Induces Triple-Negative Breast Cancer Cell G2/M Phase Block and Apoptosis by Regulating GADD45A. Pharmaceuticals (Basel) 2024; 17:1269. [PMID: 39458909 PMCID: PMC11510481 DOI: 10.3390/ph17101269] [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: 08/01/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
(1) Background: Nifuratel (NF113), derived from nitrofuran, has a specific anti-tumor effect. However, the potential mechanisms of NF113 in triple-negative breast cancer remain unknown. (2) Methods: In the study, CCK8 assay and colony formation assays were used to evaluate the inhibition effect of NF113 on cell proliferation. Apoptosis and cell cycle distribution were tested by flow cytometry. The mechanism of NF113's anti-tumor effect was predicted by transcriptome sequencing and verified by using PCR and Western blot experiments. Breast cancer organoids constructed from the patient-derived tumor xenograft model and the MDA-MB-468 xenograft mouse model were established to evaluate the effect of NF113. (3) Results: Our study showed that NF113 had an anti-tumor effect on triple-negative breast cancer both in vitro and in vivo. NF113 also induced apoptosis and G2/M phase arrest in triple-negative breast cancer cells. Our experimental data further verified that NF113 reduced GADD5A mRNA and protein expression, which were significantly upregulated in breast cancer, with downstream CDC25C and AKT phosphorylation changes. (4) Conclusions: Our data provided compelling evidence that NF113 inhibited breast cancer growth via upregulating GADD45A. Conclusion: NF113 was able to exert inhibitory effects on the proliferation of triple-negative breast cancer in vivo and in vitro, which may induce G2/M phase arrest via the GADD45A/CyclinB/CDK1 pathway and apoptosis via GADD45A/JNK/P38.
Collapse
Affiliation(s)
- Yuhang Hou
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Hongyun Hao
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Yan Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Jing Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Zhengrui Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Yimin Nie
- Key Laboratory of Straw Comprehensive Utilization and Black Soil Conservation, Ministry of Education, College of Life Sciences, Jilin Agricultural University, Changchun 130118, China
| | - Shichang Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Shengtao Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| | - Mei Yang
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210000, China (Y.Y.); (J.Z.); (Z.L.)
| |
Collapse
|
14
|
Kepuladze S, Burkadze G, Kokhreidze I. Epithelial-Mesenchymal Transition Indexes in Triple-Negative Breast Cancer Progression and Metastases. Cureus 2024; 16:e68761. [PMID: 39371729 PMCID: PMC11456157 DOI: 10.7759/cureus.68761] [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] [Accepted: 09/06/2024] [Indexed: 10/08/2024] Open
Abstract
Background Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer characterized by the lack of expression of estrogen and progesterone receptors and the absence of HER2 protein overexpression or gene amplification. How TNBC becomes so aggressive at the molecular level is not yet fully understood. The epithelial-mesenchymal transition (EMT) has been increasingly recognized as playing a pivotal role in cancer progression and metastasis. This study aimed to elucidate the connection between TNBC progression with EMT-related markers, including vimentin, beta-catenin, and E-cadherin. Methodology Rigorous immunohistochemical analysis was employed to assess the expression of vimentin, beta-catenin, and E-cadherin in primary tumors, tumor buds, and lymph node metastases (LNMs) from 137 cases with an invasive ductal carcinoma triple-negative phenotype diagnosed between 2018 and 2024. The EMT index, which was especially important in our work, is the sum of vimentin and beta-catenin expression divided by that of E-cadherin. Estimated Pearson correlation, multiple linear regression, and Kruskal-Wallis tests were used to determine the relationships of the EMT index with tumor buds and tumor-infiltrating lymphocytes (TILs). Results Vimentin highly correlated within separate regions of interest with Pearson correlation ranging from 0.90 to 0.92 (p < 0.001). Strong negative correlations between E-cadherin and vimentin (r = -0.81 to - 0.89, p < 0.001) showed its role in preserving the epithelial phenotype. The presence of tumor buds, aggregates, or clusters of cancer cells shed from the primary tumor mass invading the connective tissue showed very strong associations with the EMT index (r = 0.91, p < 0.001). Its presence is suggestive of aggressive disease and may identify a high-risk subpopulation that may benefit from more active surveillance or adjuvant treatment. Similarly, TILs correlated inversely with the EMT index (r = -0.90, p < 0.001). The most significant predictor of the EMT index, i.e., vimentin, had a model R-squared value of 1.000 in the regression analysis. Conclusions This study brings to light the importance of EMT-related markers in TNBC progression, with special emphasis on tumor buds as possible prognostic indicators for aggressive disease. The negative correlation of TILs with the EMT index indicates that an effective immune response could antagonize EMT-mediated tumor progression. These results suggest that EMT-based treatments in TNBC should be designed from a multimarker perspective by including interactions among several markers to optimize predictions and therapeutics. The results hold the potential to set future research directions and actionable outcomes that could influence clinical utility in the battle against TNBC.
Collapse
Affiliation(s)
- Shota Kepuladze
- Pathology and Oncology, Tbilisi State Medical University, Tbilisi, GEO
| | - George Burkadze
- Molecular Pathology, Tbilisi State Medical University, Tbilisi, GEO
| | | |
Collapse
|
15
|
Banerjee R, Maitra I, Bhattacharya T, Banerjee M, Ramanathan G, Rayala SK, Venkatraman G, Rajeswari D. Next-generation biomarkers for prognostic and potential therapeutic enhancement in Triple negative breast cancer. Crit Rev Oncol Hematol 2024; 201:104417. [PMID: 38901639 DOI: 10.1016/j.critrevonc.2024.104417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024] Open
Abstract
Triple-negative breast carcinoma (TNBC) is one of the most challenging subtypes of breast carcinoma and it has very limited therapeutic options as it is highly aggressive. The prognostic biomarkers are crucial for early diagnosis of the tumor, it also helps in anticipating the trajectory of the illness and optimizing the therapy options. Several therapeutic biomarkers are being used. Among them, the next-generation biomarkers that include Circulating tumor (ct) DNA, glycogen, lipid, and exosome biomarkers provide intriguing opportunities for enhancing the prognosis of TNBC. Lipid and glycogen biomarkers serve as essential details on the development of the tumor along with the efficacy of the treatment, as it exhibits metabolic alteration linked to TNBC. Several types of biomarkers have predictive abilities in TNBC. Elevated levels are associated with worse outcomes. ctDNA being a noninvasive biomarker reveals the genetic composition of the tumor, as well as helps to monitor the progression of the disease. Traditional therapies are ineffective in TNBC due to a lack of receptors, targeted drug delivery provides a tailored approach to overcome drug resistance and site-specific action by minimizing the side effects in TNBC treatment. This enhances therapeutic outcomes against the aggressive nature of breast cancer. This paper includes all the recent biomarkers which has been researched so far in TNBC and the state of art for TNBC which is explored.
Collapse
Affiliation(s)
- Risav Banerjee
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Indrajit Maitra
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Trisha Bhattacharya
- Department of Biotechnology, Indian Academy Degree College, Autonomous, Hennur cross, Kalyan Nagar, Bengaluru, Karnataka 560043, India
| | - Manosi Banerjee
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Gnanasambandan Ramanathan
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology, Madras, Tamil Nadu 600036, India
| | - Ganesh Venkatraman
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Devi Rajeswari
- Department of Biomedical Genetics, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
16
|
Park SH, Lee J, Jung SY, Kang YH, Kim J. A higher consumption of green and white-colored vegetables and fruits is associated with lowered breast cancer risk among Korean women. Nutr Res 2024; 129:38-54. [PMID: 39213830 DOI: 10.1016/j.nutres.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer (BrCa) remains a significant health concern globally, influenced by both nonmodifiable and modifiable risk factors. Limited studies have explored the role of color-specific vegetables and fruits, which are rich in specific phytonutrients, on BrCa risk. We hypothesized that consuming color-specific vegetables and fruits may decrease BrCa risk in Korean women. This case-control study examined the relationship between the intake of different-colored vegetables and fruits and the risk of BrCa, considering menopausal, hormone receptor status, tumor subtypes. We matched 395 patients and 395 controls by age and recruited from the National Cancer Center in Korea. Dietary data was collected via food frequency questionnaire, categorizing by colors: green, orange/yellow, red/purple, and white. Odds ratio (OR) and 95% confidence intervals (CIs) were calculated by logistic regression models, with subgroup analyses for menopausal, hormone receptor status, and tumor subtypes. Results shown BrCa patients consumed less vegetables and fruits than control group. Higher consumption of green, other orange/yellow, and white vegetables and fruits was negatively associated with BrCa risk [OR (95% CIs) of Q4 vs Q1 = 0.59 (0.36-0.94); 0.55 (0.33-0.89); and 0.60 (0.37-0.96), respectively]. Particularly, a greater intake of dark green leafy vegetables was significantly associated with reduced BrCa risk (OR of Q4 vs Q1 = 0.55, 95% CI = 0.34-0.89). Subgroup analysis consistently demonstrated inverse associations between higher intake of green-color vegetables and fruits and BrCa risk. Our findings suggest that a diet rich in green and white-color vegetables and fruits may lower BrCa risk among Korean women.
Collapse
Affiliation(s)
- Sin-Hye Park
- Department of Food Science and Nutrition, Korean Institute of Nutrition, Hallym University, Chuncheon, Gangwon State, 24252, Republic of Korea; Department of Cancer Artificial Intelligence Digital Healthcare, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Gyeonggi-do, 10408, Republic of Korea
| | - Jeonghee Lee
- Department of Cancer Artificial Intelligence Digital Healthcare, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Gyeonggi-do, 10408, Republic of Korea
| | - So-Youn Jung
- Center for Breast Cancer, Research Institute and Hospital, National Cancer Center, Goyang, Gyeonggi-do, 10408, Republic of Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition, Korean Institute of Nutrition, Hallym University, Chuncheon, Gangwon State, 24252, Republic of Korea
| | - Jeongseon Kim
- Department of Cancer Artificial Intelligence Digital Healthcare, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, Gyeonggi-do, 10408, Republic of Korea.
| |
Collapse
|
17
|
Ahn J, Lee JW, Nam SM, Kim DK, Cho SK, Choi HK. Integrative multi-omics analysis reveals ortho-topolin riboside exhibits anticancer activity by regulating metabolic pathways in radio-resistant triple negative breast cancer cells. Chem Biol Interact 2024; 398:111089. [PMID: 38823535 DOI: 10.1016/j.cbi.2024.111089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/07/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Radio-resistant triple negative breast cancer (TNBC) is resistant to conventional drugs and radiation therapy. ortho-topolin riboside (oTR) has been evaluated for its anticancer activity in several types of cancer cells. However, its anti-proliferative activity in radio-resistant TNBC cells has not yet been reported. Therefore, we investigated the anti-proliferative activity of oTR in radio-resistant TNBC cells, and performed metabolome, lipidome, transcriptome, and proteome profiling to reveal the mechanisms of the anticancer activity of oTR. oTR showed cytotoxicity against radio-resistant TNBC cells with an inhibitory concentration (IC50) value of 7.78 μM. Significantly decreased (p value < 0.05) basal and compensatory glycolysis were observed in the oTR-treated group than untreated group. Mitochondrial spare respiratory capacity, which is relevant to cell fitness and flexibility, was significantly decreased (p value < 0.05) in the oTR-treated group. The major metabolic pathways significantly altered by oTR according to metabolome, transcriptome, and proteome profiles were the glycerolipid/glycerophospholipid pathway (log2(FC) of MGLL = -0.13, log2(FC) of acylglycerol lipase = -1.35, log2(FC) of glycerol = -0.81), glycolysis (log2(FC) of EGLN1 = 0.16, log2(FC) of EGLN1 = 0.62, log2(FC) of glucose = -0.76, log2(FC) of lactate = -0.81), and kynurenine pathway (log2(FC) of KYNU = 0.29, log2(FC) of kynureninase = 0.55, log2(FC) of alanine = 0.72). Additionally, proline metabolism (log2(FC) of PYCR1 = -0.17, log2(FC) of proline = -0.73) was significantly altered in the metabolomic and transcriptomic profiles. The MAPK signaling pathway (log2(FC) of CCN1 = -0.15, log2(FC) of CCN family member 1 = -1.02) and Rap 1 signaling pathway (log2(FC) of PARD6B = -0.28, log2(FC) of PAR6B = -3.13) were also significantly altered in transcriptomic and proteomic profiles. The findings of this study revealed that oTR has anticancer activity in radio-resistant TNBC cells by affecting various metabolic pathways, suggesting the potential of oTR as a novel anticancer agent for radio-resistant TNBC patients.
Collapse
Affiliation(s)
- Junyoung Ahn
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji Won Lee
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung Min Nam
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dae Kyeong Kim
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Somi Kim Cho
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea; Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, 63243, Republic of Korea.
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
18
|
Tsai CC, Yang YN, Wang K, Chen YCE, Chen YF, Yang JC, Li ZL, Huang HM, Pedersen JZ, Incerpi S, Lee SY, Lin HY, Whang-Peng J. Progesterone modulates cell growth via integrin αvβ3-dependent pathway in progesterone receptor-negative MDA-MB-231 cells. Heliyon 2024; 10:e34006. [PMID: 39071644 PMCID: PMC11283053 DOI: 10.1016/j.heliyon.2024.e34006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Progesterone (P4) plays a pivotal role in regulating the cancer progression of various types, including breast cancer, primarily through its interaction with the P4 receptor (PR). In PR-negative breast cancer cells, P4 appears to function in mediating cancer progression, such as cell growth. However, the mechanisms underlying the roles of P4 in PR-negative breast cancer cells remain incompletely understood. This study aimed to investigate the effects of P4 on cell proliferation, gene expression, and signal transduction in PR-negative MDA-MB-231 breast cancer cells. P4-activated genes, associated with proliferation in breast cancer cells, exhibit a stimulating effect on cell growth in PR-negative MDA-MB-231 cells, while demonstrating an inhibitory impact in PR-positive MCF-7 cells. The use of arginine-glycine-aspartate (RGD) peptide successfully blocked P4-induced extracellular signal-regulated kinase 1/2 (ERK1/2) activation, aligning with computational models of P4 binding to integrin αvβ3. Disrupting integrin αvβ3 binding with RGD peptide or anti-integrin αvβ3 antibody altered P4-induced expression of proliferative genes and modified P4-induced cell growth in breast cancer cells. In conclusion, integrin αvβ3 appears to mediate P4-induced ERK1/2 signal pathway to regulate proliferation via alteration of proliferation-related gene expression in PR-negative breast cancer cells.
Collapse
Affiliation(s)
- Chung-Che Tsai
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yung-Ning Yang
- Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung 82445, Taiwan
- School of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Kuan Wang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chun E. Chen
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Fong Chen
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Jen-Chang Yang
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Zi-Lin Li
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jens Z. Pedersen
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Sandra Incerpi
- Department of Sciences, University Roma Tre, Rome 00133, Italy
| | - Sheng-Yang Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Hung-Yun Lin
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany 12203, NY, USA
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Jaqueline Whang-Peng
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
19
|
Gottumukkala SB, Ganesan TS, Palanisamy A. Comprehensive molecular interaction map of TGFβ induced epithelial to mesenchymal transition in breast cancer. NPJ Syst Biol Appl 2024; 10:53. [PMID: 38760412 PMCID: PMC11101644 DOI: 10.1038/s41540-024-00378-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Breast cancer is one of the prevailing cancers globally, with a high mortality rate. Metastatic breast cancer (MBC) is an advanced stage of cancer, characterised by a highly nonlinear, heterogeneous process involving numerous singling pathways and regulatory interactions. Epithelial-mesenchymal transition (EMT) emerges as a key mechanism exploited by cancer cells. Transforming Growth Factor-β (TGFβ)-dependent signalling is attributed to promote EMT in advanced stages of breast cancer. A comprehensive regulatory map of TGFβ induced EMT was developed through an extensive literature survey. The network assembled comprises of 312 distinct species (proteins, genes, RNAs, complexes), and 426 reactions (state transitions, nuclear translocations, complex associations, and dissociations). The map was developed by following Systems Biology Graphical Notation (SBGN) using Cell Designer and made publicly available using MINERVA ( http://35.174.227.105:8080/minerva/?id=Metastatic_Breast_Cancer_1 ). While the complete molecular mechanism of MBC is still not known, the map captures the elaborate signalling interplay of TGFβ induced EMT-promoting MBC. Subsequently, the disease map assembled was translated into a Boolean model utilising CaSQ and analysed using Cell Collective. Simulations of these have captured the known experimental outcomes of TGFβ induced EMT in MBC. Hub regulators of the assembled map were identified, and their transcriptome-based analysis confirmed their role in cancer metastasis. Elaborate analysis of this map may help in gaining additional insights into the development and progression of metastatic breast cancer.
Collapse
Affiliation(s)
| | - Trivadi Sundaram Ganesan
- Department of Medical Oncology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Anbumathi Palanisamy
- Department of Biotechnology, National Institute of Technology Warangal, Warangal, India.
| |
Collapse
|
20
|
Wang N, Qin L, Liu Z, Cao J, Huang J, Ma L, Huang G. Discovery of a Pimaradiene that Decreases Viability of MDA-MB-468 Cells Through Inhibition of EGFR Signaling Pathway. Chem Biodivers 2024; 21:e202400288. [PMID: 38415947 DOI: 10.1002/cbdv.202400288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 02/29/2024]
Abstract
Triple-negative breast cancer (TNBC) is characterized by strong invasiveness, high relapse rates, and poor overall survival. It occurs in approximately 15-20 % of all breast cancer cases. Natural compounds are a promising option for managing breast cancer. ent-8(14),15-Pimaradiene-2β,19-diol (JXE-23), is a pimaradiene isolated from the fern Aleuritopteris albofusca. However, the effects and molecular mechanisms of JXE-23 on cancer cells are still unknown. Thus, this study was designed to determine the potential of JXE-23 for its anticancer properties in TNBC cells. JXE-23 was evaluated for its antiproliferative activity in vitro against human breast cancer cell lines, and showed selectively cytotoxic activity against MDA-MB-468, an EGFR-overexpressing TNBC cancer cell line, with an IC50 value of 1.17±0.04 μM. Moreover, mechanistic investigations indicated that JXE-23 was significantly capable of inhibiting cell proliferation and viability in MDA-MB-468 cells. In addition, JXE-23 exerted an anticancer effect against MDA-MB-468 cells via restraining cell migration in a dose-dependent mode. Moreover, after treatment with JXE-23, the protein expressions of pEGFR, pERK, pAkt and p-p70S6K were significantly reduced in MDA-MB-468 cells. The results underscored that JXE-23 could be a potential lead compound for the treatment of EGFR-overexpressing TNBC cells.
Collapse
Affiliation(s)
- Nina Wang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, P. R. China
| | - Li Qin
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, P. R. China
| | - Zi Liu
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, P. R. China
| | - Jianguo Cao
- College of Life Sciences, Shanghai Normal University, Shanghai, P. R. China
| | - Jiayi Huang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, P. R. China
| | - Liang Ma
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, P. R. China
| | - Guozheng Huang
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, P. R. China
| |
Collapse
|
21
|
Alajroush DR, Smith CB, Anderson BF, Oyeyemi IT, Beebe SJ, Holder AA. A Comparison of In Vitro Studies between Cobalt(III) and Copper(II) Complexes with Thiosemicarbazone Ligands to Treat Triple Negative Breast Cancer. Inorganica Chim Acta 2024; 562:121898. [PMID: 38282819 PMCID: PMC10810091 DOI: 10.1016/j.ica.2023.121898] [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] [Indexed: 01/30/2024]
Abstract
Metal complexes have gained significant attention as potential anti-cancer agents. The anti-cancer activity of [Co(phen)2(MeATSC)](NO3)3•1.5H2O•C2H5OH 1 (where phen = 1,10-phenanthroline and MeATSC = 9-anthraldehyde-N(4)-methylthiosemicarbazone) and [Cu(acetylethTSC)Cl]Cl•0.25C2H5OH 2 (where acetylethTSC = (E)-N-ethyl-2-[1-(thiazol-2-yl)ethylidene]hydrazinecarbothioamide) was investigated by analyzing DNA cleavage activity. The cytotoxic effect was analyzed using CCK-8 viability assay. The activities of caspase 3/7, 9, and 1, reactive oxygen species (ROS) production, cell cycle arrest, and mitochondrial function were further analyzed to study the cell death mechanisms. Complex 2 induced a significant increase in nicked DNA. The IC50 values of complex 1 were 17.59 μM and 61.26 μM in cancer and non-cancer cells, respectively. The IC50 values of complex 2 were 5.63 and 12.19 μM for cancer and non-cancer cells, respectively. Complex 1 induced an increase in ROS levels, mitochondrial dysfunction, and activated caspases 3/7, 9, and 1, which indicated the induction of intrinsic apoptotic pathway and pyroptosis. Complex 2 induced cell cycle arrest in the S phase, ROS generation, and caspase 3/7 activation. Thus, complex 1 induced cell death in the breast cancer cell line via activation of oxidative stress which induced apoptosis and pyroptosis while complex 2 induced cell cycle arrest through the induction of DNA cleavage.
Collapse
Affiliation(s)
- Duaa R. Alajroush
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| | - Chloe B. Smith
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| | - Brittney F. Anderson
- Department of Biological Sciences, University of the Virgin Islands, 2 John Brewers Bay, St. Thomas, VI 00802, U.S.A
| | - Ifeoluwa T. Oyeyemi
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
- Department of Biological Sciences, University of Medical Sciences, Ondo City, Nigeria
| | - Stephen J. Beebe
- Frank Reidy Research center for Bioelectrics, Old Dominion University, 4211 Monarch Way, Suite 300, Norfolk, VA, 23508, U.S.A
| | - Alvin A. Holder
- Department of Chemistry and Biochemistry, Old Dominion University 4501 Elkhorn Avenue, Norfolk, VA 23529, U.S.A
| |
Collapse
|
22
|
Sonowal S, Gogoi U, Buragohain K, Nath R. Endophytic fungi as a potential source of anti-cancer drug. Arch Microbiol 2024; 206:122. [PMID: 38407579 DOI: 10.1007/s00203-024-03829-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/24/2023] [Accepted: 01/01/2024] [Indexed: 02/27/2024]
Abstract
Endophytes are considered one of the major sources of bioactive compounds used in different aspects of health care including cancer treatment. When colonized, they either synthesize these bioactive compounds as a part of their secondary metabolite production or augment the host plant machinery in synthesising such bioactive compounds. Hence, the study of endophytes has drawn the attention of the scientific community in the last few decades. Among the endophytes, endophytic fungi constitute a major portion of endophytic microbiota. This review deals with a plethora of anti-cancer compounds derived from endophytic fungi, highlighting alkaloids, lignans, terpenes, polyketides, polyphenols, quinones, xanthenes, tetralones, peptides, and spirobisnaphthalenes. Further, this review emphasizes modern methodologies, particularly omics-based techniques, asymmetric dihydroxylation, and biotic elicitors, showcasing the dynamic and evolving landscape of research in this field and describing the potential of endophytic fungi as a source of anticancer drugs in the future.
Collapse
Affiliation(s)
- Sukanya Sonowal
- Microbiology Laboratory, Department of Life Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Urvashee Gogoi
- Microbiology Laboratory, Department of Life Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Kabyashree Buragohain
- Microbiology Laboratory, Department of Life Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, 786004, India
| | - Ratul Nath
- Microbiology Laboratory, Department of Life Sciences, Dibrugarh University, Dibrugarh, Assam, 786004, India.
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, 786004, India.
| |
Collapse
|
23
|
Sahu P, Camarillo IG, Sundararajan R. Efficacy of metformin and electrical pulses in breast cancer MDA-MB-231 cells. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:54-73. [PMID: 38464382 PMCID: PMC10918234 DOI: 10.37349/etat.2024.00204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/30/2023] [Indexed: 03/12/2024] Open
Abstract
Aim Triple-negative breast cancer (TNBC) is a very aggressive subset of breast cancer, with limited treatment options, due to the lack of three commonly targeted receptors, which merits the need for novel treatments for TNBC. Towards this need, the use of metformin (Met), the most widely used type-2 diabetes drug worldwide, was explored as a repurposed anticancer agent. Cancer being a metabolic disease, the modulation of two crucial metabolites, glucose, and reactive oxygen species (ROS), is studied in MDA-MB-231 TNBC cells, using Met in the presence of electrical pulses (EP) to enhance the drug efficacy. Methods MDA-MB-231, human TNBC cells were treated with Met in the presence of EP, with various concentrations Met of 1 mmol/L, 2.5 mmol/L, 5 mmol/L, and 10 mmol/L. EP of 500 V/cm, 800 V/cm, and 1,000 V/cm (with a pulse width of 100 µs at 1 s intervals) were applied to TNBC and the impact of these two treatments was studied. Various assays, including cell viability, microscopic inspection, glucose, ROS, and wound healing assay, were performed to characterize the response of the cells to the combination treatment. Results Combining 1,000 V/cm with 5 mmol/L Met yielded cell viability as low as 42.6% at 24 h. The glucose level was reduced by 5.60-fold and the ROS levels were increased by 9.56-fold compared to the control, leading to apoptotic cell death. Conclusions The results indicate the enhanced anticancer effect of Met in the presence of electric pulses. The cell growth is inhibited by suppressing glucose levels and elevated ROS. This shows a synergistic interplay between electroporation, Met, glucose, and ROS metabolic alterations. The results show promises for combinational therapy in TNBC patients.
Collapse
Affiliation(s)
- Praveen Sahu
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| | - Ignacio G. Camarillo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Raji Sundararajan
- School of Engineering Technology, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
24
|
Terragno M, Vetrova A, Semenov O, Sayan AE, Kriajevska M, Tulchinsky E. Mesenchymal-epithelial transition and AXL inhibitor TP-0903 sensitise triple-negative breast cancer cells to the antimalarial compound, artesunate. Sci Rep 2024; 14:425. [PMID: 38172210 PMCID: PMC10764797 DOI: 10.1038/s41598-023-50710-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a difficult-to-treat, aggressive cancer type. TNBC is often associated with the cellular program of epithelial-mesenchymal transition (EMT) that confers drug resistance and metastasis. EMT and reverse mesenchymal-epithelial transition (MET) programs are regulated by several signaling pathways which converge on a group of transcription factors, EMT- TFs. Therapy approaches could rely on the EMT reversal to sensitise mesenchymal tumours to compounds effective against epithelial cancers. Here, we show that the antimalarial ROS-generating compound artesunate (ART) exhibits higher cytotoxicity in epithelial than mesenchymal breast cancer cell lines. Ectopic expression of EMT-TF ZEB1 in epithelial or ZEB1 depletion in mesenchymal cells, respectively, reduced or increased ART-generated ROS levels, DNA damage and apoptotic cell death. In epithelial cells, ZEB1 enhanced expression of superoxide dismutase 2 (SOD2) and glutathione peroxidase 8 (GPX8) implicated in ROS scavenging. Although SOD2 or GPX8 levels were unaffected in mesenchymal cells in response to ZEB1 depletion, stable ZEB1 knockdown enhanced total ROS. Receptor tyrosine kinase AXL maintains a mesenchymal phenotype and is overexpressed in TNBC. The clinically-relevant AXL inhibitor TP-0903 induced MET and synergised with ART to generate ROS, DNA damage and apoptosis in TNBC cells. TP-0903 reduced the expression of GPX8 and SOD2. Thus, TP-0903 and ZEB1 knockdown sensitised TNBC cells to ART, likely via different pathways. Synergistic interactions between TP-0903 and ART indicate that combination approaches involving these compounds can have therapeutic prospects for TNBC treatment.
Collapse
Affiliation(s)
- Mirko Terragno
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan.
| | - Anastassiya Vetrova
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Oleg Semenov
- Regulation of Gene Expression Laboratory, Institute of Cytology RAS, Saint Petersburg, Russia
| | - A Emre Sayan
- Cancer Sciences Division, University of Southampton, Southampton, UK
| | - Marina Kriajevska
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Eugene Tulchinsky
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan.
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
25
|
Sua LF, Osorio ÁE, Zuñiga-Restrepo V, Ibarra CD, Quintero N, Fernández-Trujillo L. Development of a Second Primary Lung Cancer Following a Primary Breast Cancer: A Case Series. J Investig Med High Impact Case Rep 2024; 12:23247096241272013. [PMID: 39390783 PMCID: PMC11468341 DOI: 10.1177/23247096241272013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 10/12/2024] Open
Abstract
Breast cancer (BC) accounts for 24.2% of all women's malignant tumors, with rising survival rates due to advancements in chemotherapy and targeted treatments. However, second primary cancers, particularly lung cancer (LC), have become more prevalent, often emerging approximately 10 years after BC treatment. This study presents a case series of 9 women diagnosed with second primary LC following BC, treated at a high-complexity hospital in Colombia between 2014 and 2019. All initial BCs were ductal carcinomas, 7 were triple negative, 1 was human epidermal growth factor receptor 2 positive, and 1 was estrogen and progesterone positive. Each patient had undergone radiation therapy, and 7 had received chemotherapy, increasing their LC risk. The second primary LCs, all adenocarcinomas, were confirmed using immunohistochemical stains for thyroid transcription factor-1 (TTF-1), Napsin A, and estrogen receptor (ER) status. The interval between treatments and LC detection ranged from 1 to 17 years, with 4 cases identified after 10 years and 3 within 1 to 3 years, underscoring the need for prolonged surveillance. Seven LCs were ipsilateral to the BC and radiation site, while 2 were contralateral, highlighting the necessity of monitoring both sides for potential LC development. This case series enhances the local epidemiological understanding, showing that prior radiotherapy for BC and histological analysis are key in characterizing second primary LC patients. The study emphasizes the critical role of accurate histological diagnosis in guiding treatment approaches for lung lesions in BC survivors.
Collapse
Affiliation(s)
- Luz F. Sua
- Fundación Valle del Lili, Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Álvaro E. Osorio
- Fundación Valle del Lili, Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | | | | | | | | |
Collapse
|
26
|
Kim S, Park JM, Park S, Jung E, Ko D, Park M, Seo J, Nam KD, Kang YK, Lee K, Farrand L, Kim YJ, Kim JY, Seo JH. Suppression of TNBC metastasis by doxazosin, a novel dual inhibitor of c-MET/EGFR. J Exp Clin Cancer Res 2023; 42:292. [PMID: 37924112 PMCID: PMC10625208 DOI: 10.1186/s13046-023-02866-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/16/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by aggressive growth and a high propensity for recurrence and metastasis. Simultaneous overexpression of c-MET and EGFR in TNBC is associated with worse clinicopathological features and unfavorable outcomes. Although the development of new c-MET inhibitors and the emergence of 3rd-generation EGFR inhibitors represent promising treatment options, the high costs involved limit the accessibility of these drugs. In the present study, we sought to investigate the therapeutic potential of doxazosin (DOXA), a generic drug for benign prostate hyperplasia, in targeting TNBC. METHODS The effect of DOXA on TNBC cell lines in vitro was evaluated in terms of cell viability, apoptosis, c-MET/EGFR signaling pathway, molecular docking studies and impact on cancer stem cell (CSC)-like properties. An in vivo metastatic model with CSCs was used to evaluate the efficacy of DOXA. RESULTS DOXA exhibits notable anti-proliferative effects on TNBC cells by inducing apoptosis via caspase activation. Molecular docking studies revealed the direct interaction of DOXA with the tyrosine kinase domains of c-MET and EGFR. Consequently, DOXA disrupts important survival pathways including AKT, MEK/ERK, and JAK/STAT3, while suppressing CSC-like characteristics including CD44high/CD24low subpopulations, aldehyde dehydrogenase 1 (ALDH1) activity and formation of mammospheres. DOXA administration was found to suppress tumor growth, intra- and peri-tumoral angiogenesis and distant metastasis in an orthotopic allograft model with CSC-enriched populations. Furthermore, no toxic effects of DOXA were observed in hepatic or renal function. CONCLUSIONS Our findings highlight the potential of DOXA as a therapeutic option for metastatic TNBC, warranting further investigation.
Collapse
Affiliation(s)
- Seongjae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Jung Min Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Dongmi Ko
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Minsu Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Juyeon Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| | - Kee Dal Nam
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Yong Koo Kang
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Kyoungmin Lee
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea
| | - Lee Farrand
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
- Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 148 Gurodong-ro, Guro-gu, Seoul, 08308, Republic of Korea.
| |
Collapse
|
27
|
Zhang Z, Zhang R, Li D. Molecular Biology Mechanisms and Emerging Therapeutics of Triple-Negative Breast Cancer. Biologics 2023; 17:113-128. [PMID: 37767463 PMCID: PMC10520847 DOI: 10.2147/btt.s426392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is conventionally characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2), accounting for approximately 15-20% of all breast cancers. Compared to other molecular phenotypes, TNBC is typically associated with high malignancy and poor prognosis. Cytotoxic agents have been the mainstay of treatment for the past few decades due to the lack of definitive targets and limited therapeutic interventions. However, recent developments have demonstrated that TNBC has peculiar molecular classifications and biomarkers, which provide the possibility of evolving treatment from basic cytotoxic chemotherapy to an expanding domain of targeted therapies. This review presents a framework for understanding the current clinical experience surrounding molecular biology mechanisms in TNBC (Figure 1). Including immunotherapy, polymerase (PARP) and PI3K/AKT pathway inhibitors, antibody-drug conjugates, and androgen receptor (AR) blockade. Additionally, the role of miRNA therapeutics targeting TNBC and potential strategies targeting cancer stem cells (CSCs) are discussed and highlighted. As more and more treatments arise on the horizon, we believe that patients with TNBC will have a new sense of hope.
Collapse
Affiliation(s)
- Zhiying Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Rui Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Donghai Li
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| |
Collapse
|
28
|
El Gazzar WB, Albakri KA, Hasan H, Badr AM, Farag AA, Saleh OM. Poly(ADP-ribose) polymerase inhibitors in the treatment landscape of triple-negative breast cancer (TNBC). J Oncol Pharm Pract 2023; 29:1467-1479. [PMID: 37559370 DOI: 10.1177/10781552231188903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
OBJECTIVE Chemotherapy is the mainstay for triple-negative breast cancer (TNBC) patients. Over the years, the use of chemotherapy for these patients has demonstrated many adversities, including toxicity and resistance, which suggested the need to develop novel alternative therapeutic options, such as poly(ADP-ribose) polymerase inhibitors (PARPi). Herein, we provide an overview on PARPi, mechanisms of action and the role of biomarkers in PARPi sensitivity trials, clinical advances in PARPi therapy for TNBC patients based on the most recent studies and findings of clinical trials, and challenges that prevent PARP inhibitors from achieving high efficacy such as resistance and overlapping toxicities with other chemotherapies. DATA SOURCES Searching for relevant articles was done using PubMed and Cochrane Library databases by using the keywords including TNBC; chemotherapy; PARPi; BRCA; homologous recombination repair (HRR). Studies had to be published in full-text in English in order to be considered. DATA SUMMARY Although PARPi have been used in the treatment of local/metastatic breast malignancies that are HER2 negative and has a germline BRCA mutation, several questions are still to be answered in order to maximize the clinical benefit of PARP inhibitors in TNBC treatment, such as questions related to the optimal use in the neoadjuvant and metastatic settings as well as the best combinations with various chemotherapies. CONCLUSIONS PARPi are emerging treatment options for patients with gBRCA1/2 mutations. Determining patients that are most likely to benefit from PARPi and identifying the optimal treatment combinations with high efficacy and fewer side effects are currently ongoing.
Collapse
Affiliation(s)
- Walaa Bayoumie El Gazzar
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha City, Egypt
| | | | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Amira M Badr
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amina A Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha City, Egypt
| | | |
Collapse
|
29
|
Hedayat M, Khezri MR, Jafari R, Malekinejad H, Majidi Zolbanin N. Concomitant effects of paclitaxel and celecoxib on genes involved in apoptosis of triple-negative metastatic breast cancer cells. Med Oncol 2023; 40:263. [PMID: 37548777 DOI: 10.1007/s12032-023-02119-1] [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: 04/24/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023]
Abstract
Although triple-negative breast cancer accounts for less than one-fifth of breast cancers, it has a higher rate of metastasis and mortality. This study investigated the effects of combination treatment with paclitaxel and celecoxib on the expression of genes involved in the apoptosis of triple-negative metastatic breast cancer cells. MDA-MB-231 cells were cultured and then treated with certain concentrations of celecoxib (CLX), paclitaxel (PTX), and combination of them for 24 and 48 h. Cell viability was assessed by the MTT method. The real-time PCR method was utilized to assess the expression level of the genes involved in apoptosis. Western blotting was used for evaluating protein expression. IC50 values for CLX and PTX were 73.95 μM and 3.15 μM, respectively. The results demonstrated that PTX, CLX, and PTX + CLX significantly (p < 0.05) reduced cell viability. The comparison of combination treatment with PTX showed a significant increase in caspase 3 gene expression at both time points, in Bax gene expression after 48 h, and a remarkable decrease in Bcl-2 gene expression at both times. Western blotting results were in line with genes' expression. These findings indicate that a combination of PTX and CLX results in a significantly more reduction in cell viability of breast cancer cells. In addition, it seems CLX may be an effective agent in regulating the expression level of caspase 3, Bax, and Bcl-2 when combined with PTX.
Collapse
Affiliation(s)
- Mohaddeseh Hedayat
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hassan Malekinejad
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Sero Road, Urmia, 5715799313, Iran.
| |
Collapse
|
30
|
Passos ID, Papadimitriou D, Katsouda A, Papavasileiou GE, Galatas A, Tzitzis P, Mpakosi A, Mironidou-Tzouveleki M. In Vitro and In Vivo Effects of Docetaxel and Dasatinib in Triple-Negative Breast Cancer: A Research Study. Cureus 2023; 15:e43534. [PMID: 37719631 PMCID: PMC10500968 DOI: 10.7759/cureus.43534] [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] [Accepted: 08/15/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) comprises a heterogeneous group of tumors with a single trait in common: an evident aggressive nature with higher rates of relapse and lower overall survival in the metastatic context when compared to other subtypes of breast cancer. To date, not a single targeted therapy has been approved for the treatment of TNBC, and cytotoxic chemotherapy remains the standard treatment. In the present experimental study, we examine the effects of the chemotherapeutic docetaxel and the bcr/abl kinase inhibitor dasatinib on TNBC cell lines (in vitro) and on TNBC tumor xenograft mouse models (in vivo). Materials and methods TNBC cell lines were cultivated and treated with various concentrations of docetaxel and dasatinib (5 nM to 100 nM). Cell death and apoptosis were studied by flow cytometry. TNBC cell lines were then injected in BALB/c athymic nude mice to express the tumor in vivo. Four groups of mice were created (group A: control; group B: DOC; group C: DAS; group D: DOC + DAS) and treated, respectively, with the drugs and their combination. Tumors were obtained, maintained in a 10% formaldehyde solution, embedded in paraffin, and sent for further histological evaluation (hematoxylin-eosin staining and immune-histochemical analysis) to assess the tumor growth inhibition. Results The cytotoxic effects of docetaxel seem statistically important, with little effect on apoptosis. The effect of dasatinib in vitro and vivo is statistically important, in terms of apoptosis and tumor reduction, with little adverse effects. Conclusions TNBC is a difficult-to-treat oncologic condition, even in an experimental setting. Promising results concerning the addition of targeted therapies (dasatinib) to the conventional cytotoxic ones (docetaxel) have been shown, awaiting further evaluation.
Collapse
Affiliation(s)
- Ioannis D Passos
- Surgical Department, 219 Mobile Army Surgical Hospital, Didymoteicho, GRC
| | - Dimochristos Papadimitriou
- Laboratory of Clinical Pharmacology, Faculty of Medicine, School of Health Sciences, General Hospital of Thessaloniki "G. Gennimatas" /Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Areti Katsouda
- Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, GRC
| | | | - Apostolos Galatas
- Surgical Department, 219 Mobile Army Surgical Hospital, Didymoteicho, GRC
| | - Panagiotis Tzitzis
- 1st Department of Obstetrics & Gynaecology, Medical Faculty, Papageorgiou General Hospital/Aristotle University of Thessaloniki, Thessaloniki, GRC
| | - Alexandra Mpakosi
- Department of Microbiology, General State Hospital of Nikaia "Saint Panteleimon", Nikaia, GRC
| | - Maria Mironidou-Tzouveleki
- 1st Department of Pharmacology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, GRC
| |
Collapse
|
31
|
Bae KR, Ahn Y, Park JW, Kim SJ. Exploring Fear of Cancer Recurrence (FCR) in cancer survivors from a medical social work perspective: A qualitative study of medical social workers in South Korea. PLoS One 2023; 18:e0288059. [PMID: 37410785 DOI: 10.1371/journal.pone.0288059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/18/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Fear of Cancer Recurrence (FCR) in cancer survivors has been insufficiently addressed despite its imperativeness in cancer journey. Although several studies have investigated healthcare professionals' experience with FCR in cancer survivors, a medical social work perspective has rarely been reflected. This study aimed to explore Korean medical social workers' experience with intervening FCR in cancer survivors. METHODS Snowball sampling recruited 12 experienced medical social workers intervening with cancer survivors at tertiary or university cancer hospitals in South Korea. Individual and focus-group interviews (FGI) were conducted with the medical social workers. The interviews were recorded, transcribed, and analyzed by using an inductive qualitative content analysis. RESULTS Content analysis of the interviews extracted the following major themes regarding FCR in cancer survivors. First, when and how FCR among cancer survivors emerged at the early stage of medical social work interventions was identified. Second, how medical social workers dealt with FCR in cancer survivors was illustrated. Third, the responses of cancer survivors to medical social work interventions for FCR were assessed. Finally, the internal and external issues underlying the medical social work interventions for FCR among cancer survivors were revealed and discussed. CONCLUSION Based on the results, this study suggested the implications on dealing with FCR in cancer survivors in the realm of medial social work profession. Furthermore, it expanded the discussion about FCR in cancer survivors from cancer hospitals to community.
Collapse
Affiliation(s)
- Ka Ryeong Bae
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Yeojin Ahn
- Patient-Centered Outcomes Research Institute, Samsung Medical Center, Seoul, South Korea
| | - Joung Won Park
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
- Division of Social Work, Samsung Medical Center, Seoul, South Korea
| | - Seok-Joo Kim
- Department of Social Welfare, College of Social Sciences, Daegu University, Gyeongbuk, South Korea
| |
Collapse
|
32
|
Dhanushkumar T, Kamaraj B, Vasudevan K, Gopikrishnan M, Dasegowda KR, Rambabu M, George Priya Doss C. Structural immunoinformatics approach for rational design of a multi-epitope vaccine against triple negative breast cancer. Int J Biol Macromol 2023:125209. [PMID: 37271264 DOI: 10.1016/j.ijbiomac.2023.125209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
TNBC is a highly malignant breast cancer known for its aggressive behavior affecting young female adults. The standard treatment for TNBC includes surgery, chemotherapy, and radiotherapy, which often have significant side effects. Therefore, novel preventive methods are required to combat TNBC effectively. In this study, we utilized immunoinformatics to construct an in-silico vaccine against TNBC using the TRIM25 molecule via the reverse vaccinology method. Four vaccines were designed by generating T and B-cell epitopes linked with four different linkers. The modeled vaccine was docked and the results showed that vaccine-3 exhibited the highest affinity with the immune receptors. The molecular dynamics results revealed that the binding affinity and stability of Vaccine-3 were greater than those of Vaccine 2 complexes. This study has great potential preventive measures for TNBC, and further research is warranted to evaluate its efficacy in preclinical settings. This study presents an innovative preventive strategy for triple-negative breast cancer (TNBC) through immunoinformatics and reverse vaccinology to develop an in-silico vaccine. Leveraging these innovative techniques offers a novel avenue for combating the complex challenges associated with TNBC. This approach demonstrates considerable potential as a significant breakthrough in preventive measures for this particularly aggressive and malignant form of breast cancer.
Collapse
Affiliation(s)
- T Dhanushkumar
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Balu Kamaraj
- Department of Dental Education, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India.
| | - Mohanraj Gopikrishnan
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - K R Dasegowda
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Majji Rambabu
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India.
| |
Collapse
|
33
|
Xie X, Chauhan GB, Edupuganti R, Kogawa T, Park J, Tacam M, Tan AW, Mughees M, Vidhu F, Liu DD, Taliaferro JM, Pitner MK, Browning LS, Lee JH, Bertucci F, Shen Y, Wang J, Ueno NT, Krishnamurthy S, Hortobagyi GN, Tripathy D, Van Laere SJ, Bartholomeusz G, Dalby KN, Bartholomeusz C. Maternal Embryonic Leucine Zipper Kinase is Associated with Metastasis in Triple-negative Breast Cancer. CANCER RESEARCH COMMUNICATIONS 2023; 3:1078-1092. [PMID: 37377604 PMCID: PMC10281291 DOI: 10.1158/2767-9764.crc-22-0330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 03/21/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
Triple-negative breast cancer (TNBC) has high relapse and metastasis rates and a high proportion of cancer stem-like cells (CSC), which possess self-renewal and tumor initiation capacity. MELK (maternal embryonic leucine zipper kinase), a protein kinase of the Snf1/AMPK kinase family, is known to promote CSC maintenance and malignant transformation. However, the role of MELK in TNBC metastasis is unknown; we sought to address this in the current study. We found that MELK mRNA levels were higher in TNBC tumors [8.11 (3.79-10.95)] than in HR+HER2- tumors [6.54 (2.90-9.26)]; P < 0.001]. In univariate analysis, patients with breast cancer with high-MELK-expressing tumors had worse overall survival (P < 0.001) and distant metastasis-free survival (P < 0.01) than patients with low-MELK-expressing tumors. In a multicovariate Cox regression model, high MELK expression was associated with shorter overall survival after adjusting for other baseline risk factors. MELK knockdown using siRNA or MELK inhibition using the MELK inhibitor MELK-In-17 significantly reduced invasiveness, reversed epithelial-to-mesenchymal transition, and reduced CSC self-renewal and maintenance in TNBC cells. Nude mice injected with CRISPR MELK-knockout MDA-MB-231 cells exhibited suppression of lung metastasis and improved overall survival compared with mice injected with control cells (P < 0.05). Furthermore, MELK-In-17 suppressed 4T1 tumor growth in syngeneic BALB/c mice (P < 0.001). Our findings indicate that MELK supports metastasis by promoting epithelial-to-mesenchymal transition and the CSC phenotype in TNBC. Significance These findings indicate that MELK is a driver of aggressiveness and metastasis in TNBC.
Collapse
Affiliation(s)
- Xuemei Xie
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Current Institution: Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, Hawaii, USA
| | - Gaurav B. Chauhan
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ramakrishna Edupuganti
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Takahiro Kogawa
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jihyun Park
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Moises Tacam
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alex W. Tan
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohd Mughees
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fnu Vidhu
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Diane D. Liu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juliana M. Taliaferro
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Mary Kathryn Pitner
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luke S. Browning
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Ju-Hyeon Lee
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - François Bertucci
- Predictive Oncology Laboratory, Marseille Research Cancer Center, INSERM U1068, CNRS U7258, Institut Paoli-Calmettes, Aix Marseille University, 13009 Marseille, France
| | - Yu Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jian Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Current Institution: Cancer Biology Program, University of Hawai'i Cancer Center, Honolulu, Hawaii, USA
| | - Savitri Krishnamurthy
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas
- Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel N. Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven J. Van Laere
- Center for Oncological Research, Integrated Personalized and Precision Oncology Network, University of Antwerp, Antwerp, Wilrijk
- Department Oncology, KU Leuven, Leuven, Belgium
| | - Geoffrey Bartholomeusz
- Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kevin N. Dalby
- Division of Chemical Biology & Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Chandra Bartholomeusz
- Section of Translational Breast Cancer Research, Houston, Texas
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
34
|
Ncube KN, Jurgens T, Steenkamp V, Cromarty AD, van den Bout I, Cordier W. Comparative Evaluation of the Cytotoxicity of Doxorubicin in BT-20 Triple-Negative Breast Carcinoma Monolayer and Spheroid Cultures. Biomedicines 2023; 11:biomedicines11051484. [PMID: 37239157 DOI: 10.3390/biomedicines11051484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/10/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Three-dimensional cell culture models are increasingly adopted as preferred pre-clinical drug testing platforms, as they circumvent limitations associated with traditional monolayer cell cultures. However, many of these models are not fully characterized. This study aimed to characterize a BT-20 triple-negative breast carcinoma spheroid model and assess its susceptibility to doxorubicin in comparison to a monolayer model. Spheroids were developed using the liquid overlay method. Phenotypic attributes were analyzed by characterizing changes in size, gross morphology, protein content, metabolic activity, hypoxic status, and cell-cell junctions. The cytotoxic range of doxorubicin in monolayers was determined using the sulforhodamine B assay, and the comparative effect of toxic and sub-toxic concentrations was assessed in both spheroids and monolayers. Similar to the in vivo microenvironment, spheroids had a heterogeneous spatial cytoarchitecture, inherent hypoxia and strong adherens junctions. Doxorubicin induced dose-dependent cytotoxicity in monolayers (IC25: 130 nM, IC50: 320 nM and IC75: 1580 nM); however, these concentrations did not alter the spheroid size or acid phosphatase activity. Only concentrations ≥6 µM had any effect on spheroid integrity. In comparison to monolayers, the BT-20 spheroid model has decreased sensitivity to doxorubicin and could serve as a better model for susceptibility testing in triple-negative breast cancer.
Collapse
Affiliation(s)
- Keith N Ncube
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria 0007, South Africa
| | - Tamarin Jurgens
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0007, South Africa
| | - Vanessa Steenkamp
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria 0007, South Africa
| | - Allan D Cromarty
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria 0007, South Africa
| | - Iman van den Bout
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria 0007, South Africa
- Centre for Neuroendocrinology, Department of Immunology, University of Pretoria, Pretoria 0007, South Africa
| | - Werner Cordier
- Department of Pharmacology, Faculty of Health Sciences, University of Pretoria, Pretoria 0007, South Africa
| |
Collapse
|
35
|
Elmakaty I, Abdo R, Elsabagh A, Elsayed A, Malki MI. Comparative efficacy and safety of PD-1/PD-L1 inhibitors in triple negative breast cancer: a systematic review and network meta-analysis of randomized controlled trials. Cancer Cell Int 2023; 23:90. [PMID: 37170090 PMCID: PMC10173590 DOI: 10.1186/s12935-023-02941-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 05/07/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Triple-Negative Breast Cancer (TNBC) is a lethal subtype of breast cancer with limited treatment options. The purpose of this Network Meta-Analysis (NMA) is to compare the efficacy and safety of inhibitors of programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) in treating TNBC. METHODS Our search strategy was used in six databases: PubMed, Cochrane Library, Cumulative Index to Nursing and Allied Health Literature database, Embase, Scopus, and Web of Science up to November 2nd, 2022, as well as a thorough search in the most used trial registries. We included phase II and III randomized controlled trials that looked at the efficacy of PD-1/PD-L1 inhibitors in the treatment of TNBC and reported either Overall Survival (OS), Progression-Free Survival (PFS), or pathological Complete Response (pCR). The risk of bias was assessed utilizing Cochrane's risk of bias 2 tool, and the statistical analysis was performed using a frequentist contrast-based method for NMA by employing standard pairwise meta-analysis applying random effects model. RESULTS 12 trials (5324 patients) were included in our NMA including seven phase III trials. Pembrolizumab in a neoadjuvant setting achieved a pooled OS of 0.82 (95% Confidence Interval (CI) 0.65 to 1.03), a PFS of 0.82 (95% CI 0.71 to 0.94) and a pCR 2.79 (95% CI 1.07 to 7.24) compared to Atezolizumab's OS of 0.92 (95% CI 0.74 to 1.15), PFS of 0.82 (95% CI 0.69 to 0.97), and pCR of 1.94 (95% CI 0.86 to 4.37). Atezolizumab had less grade ≥ 3 adverse events (OR 1.48, 95% CI 0.90 to 2.42) than Pembrolizumab (OR 1.90, 95% CI 1.08 to 3.33) in the neoadjuvant setting. CONCLUSIONS PD-1/PD-L1 inhibitors exhibited varying efficacy in terms of OS, PFS, and pCR. They were associated with an increase in immune-related adverse effects. When used early in the course of TNBC, PD-1/PD-L1 inhibitors exert their maximum benefit. Durvalumab as a maintenance treatment instead of chemotherapy has shown promising outcomes. Future studies should focus on PD-L1 expression status and TNBC subtypes, since these factors may contribute to the design of individualized TNBC therapy regimens. Systematic review registration PROSPERO Identifier: CRD42022380712.
Collapse
Affiliation(s)
- Ibrahim Elmakaty
- College of Medicine, QU Health, Qatar University, P. O. Box: 2713, Doha, Qatar
| | - Ruba Abdo
- College of Medicine, QU Health, Qatar University, P. O. Box: 2713, Doha, Qatar
| | - Ahmed Elsabagh
- College of Medicine, QU Health, Qatar University, P. O. Box: 2713, Doha, Qatar
| | - Abdelrahman Elsayed
- College of Medicine, QU Health, Qatar University, P. O. Box: 2713, Doha, Qatar
| | - Mohammed Imad Malki
- Pathology Unit, Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
36
|
Wudtiwai B, Kodchakorn K, Shwe TH, Pothacharoen P, Phitak T, Suninthaboonrana R, Kongtawelert P. Brazilein inhibits epithelial-mesenchymal transition (EMT) and programmed death ligand 1 (PD-L1) expression in breast cancer cells. Int Immunopharmacol 2023; 118:109988. [PMID: 36933493 DOI: 10.1016/j.intimp.2023.109988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
Triple-negative breast cancer (TNBC) exhibits high levels of Epithelial-mesenchymal transition (EMT) and Programmed death ligand 1 (PD-L1) expression, which promotes immune escape and metastasis. Brazilein is a natural compound extracted from Caesalpinia sappan L., and has been demonstrated to be an anti-inflammatory anti- proliferative and apoptosis-inducer in various cancer cells. Here, we investigated the effect of brazilein on EMT and PD-L1 expression in breast cancer cells and its related molecular mechanisms using MCF-7 and MDA-MB-231 cells as a model. Since the AKT, NF-κB, and GSK3β/β-catenin signaling were reported to be important mechanisms in immune escape and metastasis, the effect of brazilein on these signaling pathways were also found out in our study. Firstly, brazilein was treated on breast cancer cells at various concentrations to study cell viability, apoptosis, and apoptosis proteins. Then, breast cancer cells were treated with non-toxic concentrations of brazilein to study its influence on EMT and expression of PD-L1 protein using MTT, flow cytometry, western blot, and wound healing analysis, respectively. We found that brazilein exerts an anti-cancer effect by reducing cell viability via induction of apoptosis, while it also downregulated EMT and PD-L1 through suppression of phosphorylation of AKT, NF-κB, and GSK3β/β-catenin. Moreover, the migration ability was diminished by inhibiting the activation of MMP-9 and MMP-2. Taken together, brazilein might delay cancer progression through inhibition of EMT, PD-L1, and metastasis suggesting it might be a potential therapeutic option in breast cancer patients having a high level of EMT and PD-L1.
Collapse
Affiliation(s)
- Benjawan Wudtiwai
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Kanchanok Kodchakorn
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Thuzar Hla Shwe
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Peraphan Pothacharoen
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Thanyaluck Phitak
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| | | | - Prachya Kongtawelert
- Thailand Excellence Center for Tissue Engineering and Stem Cells, Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
37
|
Pandurangi RS, Cseh O, Luchman HA, Ma CX, Senadheera SN, Forrest ML. Rational Drug Design of Targeted and Enzyme-Cleavable Vitamin E Analogs as a Neoadjuvant to Chemotherapy: In Vitro and In Vivo Evaluation on Reduction of the Cardiotoxicity Side Effect of Doxorubicin. ACS Pharmacol Transl Sci 2023; 6:372-386. [PMID: 36926453 PMCID: PMC10012254 DOI: 10.1021/acsptsci.2c00091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Indexed: 02/09/2023]
Abstract
Traditional drug design focuses on specific biological targets where specific receptors or biomarkers are overexpressed by cancer cells. Cancer cells circumvent the interventions by activating survival pathways and/or downregulating cell death pathways for their survival. A priori activation of apoptosis pathways of tumor (AAAPT) is a novel tumor-sensitizing technology that sensitizes tumor cells that are not responding well to the current treatments by targeting specific survival pathways involved in the desensitization of tumor cells and tries to revive them selectively in cancer cells, sparing normal cells. Several vitamin E derivatives (AMP-001, AMP-002, AMP-003, and AMP-004) were synthesized, characterized, and studied for their anti-tumorigenic properties and their synergistic potential with the standard chemotherapy doxorubicin in various cancer cells including brain cancer stem cells in vitro. Preliminary studies revealed that AAAPT drugs (a) reduced the invasive potential of brain tumor stem cells, (b) synergized with Federal Drug Application-approved doxorubicin, and (c) enhanced the therapeutic index of doxorubicin in the triple-negative breast cancer tumor rat model, preserving the ventricular function compared to cardiotoxic doxorubicin alone at therapeutic dose. The AAAPT approach has the advantage of inhibiting survival pathways and activating cell death pathways selectively in cancer cells by using targeting, linkers cleavable by tumor-specific Cathepsin B, and PEGylation technology to enhance the bioavailability. We propose AAAPT drugs as a neoadjuvant to chemotherapy and not as stand-alone therapy, which is shown to be effective in expanding the therapeutic index of doxorubicin and making it work at lower doses.
Collapse
Affiliation(s)
- Raghu S. Pandurangi
- Sci-Engi-Medco
Solutions Inc. (SEMCO), 573, Lexington Landing Pl, St. Charles, Missouri 63303, United States
| | - Orsolya Cseh
- HRIC
2A25, 3330 Hospital Drive NW, Calgary, AB T2N 4N, Canada
| | | | - Cynthia Xiuguang Ma
- Siteman
Cancer Center, Washington University School
of Medicine, St. Louis, Missouri 63110, United States
| | - Sanjeewa N. Senadheera
- Department
of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
| | - Marcus Laird Forrest
- Department
of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas 66047, United States
| |
Collapse
|
38
|
S R Dwivedi P, Shastry C. Anti-tumor potential and mode of action of karanjin against breast cancer; an in-silico approach. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
|
39
|
Ezenwafor T, Anye V, Madukwe J, Amin S, Obayemi J, Odusanya O, Soboyejo W. Nanoindentation study of the viscoelastic properties of human triple negative breast cancer tissues: Implications for mechanical biomarkers. Acta Biomater 2023; 158:374-392. [PMID: 36640950 DOI: 10.1016/j.actbio.2023.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
This paper presents the results of a combined experimental and theoretical study of the structure and viscoelastic properties of human non-tumorigenic mammary breast tissues and triple negative breast cancer (TNBC) tissues of different histological grades. A combination of immunofluorescence and confocal microscopy, and atomic force microscopy is used to study the actin cytoskeletal structures of non-tumorigenic and tumorigenic breast tissues (grade I to grade III). A combination of nanoindentation and statistical techniques is then used to measure viscoelastic properties of non-tumorigenic and human TNBC of different histological grades. A Standard Fluid Model/Anti-Zener Model II is also used to characterize the viscoelastic properties of the non-tumorigenic and tumorigenic TNBC tissues of different grades. The implications of the results are discussed for the potential application of nanoindentation and statistical deconvolution techniques to the development of mechanical biomarkers for TNBC detection/cancer diagnosis. STATEMENT OF SIGNIFICANCE: There is increasing interest in the development of mechanical biomarkers for cancer diagnosis. Here, we show that nanoindentation techniques can be used to characterize the viscoelastic properties of normal breast tissue and TNBC tissues of different histological grades. The Standard Fluid Model (Anti-Zener Model II) is used to classify the viscoelastic properties of breast tissues of different TNBC histological grades. Our results suggest that breast tissue and TNBC tissue viscoelastic properties can be used as mechanical biomarkers for the detection of TNBC at different stages.
Collapse
Affiliation(s)
- Theresa Ezenwafor
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria; NASENI Centre of Excellence in Nanotechnology and Advanced Materials, Km 4, Ondo Road, Akure, Ondo State, Nigeria; Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA 01609, United States; Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Gateway Park Life Sciences and Bioengineering Centre, Worcester, MA 01609, United States
| | - Vitalis Anye
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria
| | - Jonathan Madukwe
- Department of Histopathology, National Hospital Abuja, Federal Capital Territory (FCT), Nigeria
| | - Said Amin
- Department of Histopathology, National Hospital Abuja, Federal Capital Territory (FCT), Nigeria
| | - John Obayemi
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA 01609, United States; Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Gateway Park Life Sciences and Bioengineering Centre, Worcester, MA 01609, United States
| | - Olushola Odusanya
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria; Biotechnology and Genetic Engineering Advanced Laboratory, Sheda Science and Technology Complex (SHESTCO), Kwale, Federal Capital Territory, Abuja, Nigeria
| | - Winston Soboyejo
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria; Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA 01609, United States; Department of Biomedical Engineering, Worcester Polytechnic Institute, 60 Prescott Street, Gateway Park Life Sciences and Bioengineering Centre, Worcester, MA 01609, United States.
| |
Collapse
|
40
|
Michlewska S, Maly M, Wójkowska D, Karolczak K, Skiba E, Hołota M, Kubczak M, Ortega P, Watala C, Javier de la Mata F, Bryszewska M, Ionov M. Carbosilane ruthenium metallodendrimer as alternative anti-cancer drug carrier in triple negative breast cancer mouse model: A preliminary study. Int J Pharm 2023; 636:122784. [PMID: 36858135 DOI: 10.1016/j.ijpharm.2023.122784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023]
Abstract
The carbosilane metallodendrimer G1-[[NCPh(o-N)Ru(η6- p-cymene)Cl]Cl]4 (CRD13), based on an arene Ru(II) complex coordinated to imino-pyridine surface groups, has been conjugated with anti-cancer drugs. Ruthenium in the positively-charged dendrimer structure allows this nanoparticle to be considered as an anticancer drug carrier, made more efficient because ruthenium has anticancer properties. The ability of CRD13 to form complexes with Doxorubicin (DOX), 5-Fluorouracil (5-Fu), and Methotrexate (MTX) has been evaluated using zeta potential measurement, transmission electron microscopy (TEM) and computer simulation. The results show that it forms stable nanocomplexes with all those drugs, enhancing their effectiveness against MDA-MB-231 cancer cells. In vivo tests indicate that the CRD13/DOX system caused a decrease of tumor weight in mice with triple negative breast cancer. However, the tumors were most visibly reduced when naked dendrimers were injected.
Collapse
Affiliation(s)
- Sylwia Michlewska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Poland; Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland.
| | - Marek Maly
- Department of Physics, Faculty of Science, J.E. Purkyně University in Ústí nad Labem, Pasteurova 15, 400 96 Ústí nad Labem, Czech Republic
| | - Dagmara Wójkowska
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Mazowiecka st. 6/8, 92-215 Lodz, Poland
| | - Kamil Karolczak
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Mazowiecka st. 6/8, 92-215 Lodz, Poland
| | - Elżbieta Skiba
- Institute of General and Ecological Chemistry, Lodz University of Technology, Żeromskiego 116, 90-924 Łódź, Poland
| | - Marcin Hołota
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Małgorzata Kubczak
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Paula Ortega
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain; Instituto de Investigación Sanitaria Ramón y Cajal, IRYCIS, Spain
| | - Cezary Watala
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Mazowiecka st. 6/8, 92-215 Lodz, Poland
| | - F Javier de la Mata
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain; Instituto de Investigación Sanitaria Ramón y Cajal, IRYCIS, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| |
Collapse
|
41
|
Castellanos G, Valbuena DS, Pérez E, Villegas VE, Rondón-Lagos M. Chromosomal Instability as Enabling Feature and Central Hallmark of Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:189-211. [PMID: 36923397 PMCID: PMC10010144 DOI: 10.2147/bctt.s383759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/11/2022] [Indexed: 03/11/2023]
Abstract
Chromosomal instability (CIN) has become a topic of great interest in recent years, not only for its implications in cancer diagnosis and prognosis but also for its role as an enabling feature and central hallmark of cancer. CIN describes cell-to-cell variation in the number or structure of chromosomes in a tumor population. Although extensive research in recent decades has identified some associations between CIN with response to therapy, specific associations with other hallmarks of cancer have not been fully evidenced. Such associations place CIN as an enabling feature of the other hallmarks of cancer and highlight the importance of deepening its knowledge to improve the outcome in cancer. In addition, studies conducted to date have shown paradoxical findings about the implications of CIN for therapeutic response, with some studies showing associations between high CIN and better therapeutic response, and others showing the opposite: associations between high CIN and therapeutic resistance. This evidences the complex relationships between CIN with the prognosis and response to treatment in cancer. Considering the above, this review focuses on recent studies on the role of CIN in cancer, the cellular mechanisms leading to CIN, its relationship with other hallmarks of cancer, and the emerging therapeutic approaches that are being developed to target such instability, with a primary focus on breast cancer. Further understanding of the complexity of CIN and its association with other hallmarks of cancer could provide a better understanding of the cellular and molecular mechanisms involved in prognosis and response to treatment in cancer and potentially lead to new drug targets.
Collapse
Affiliation(s)
- Giovanny Castellanos
- Maestría en Ciencias Biológicas, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia.,School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Duván Sebastián Valbuena
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Erika Pérez
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| | - Victoria E Villegas
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Milena Rondón-Lagos
- School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia, Tunja, Colombia
| |
Collapse
|
42
|
Lv H, Zhu Y, Xue J, Jia X, Chen J. Targeted Drug Delivery System Based on Copper Sulfide for Synergistic Near-Infrared Photothermal Therapy/Photodynamic Therapy/Chemotherapy of Triple Negative Breast Cancer. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:15766-15775. [PMID: 36508193 DOI: 10.1021/acs.langmuir.2c02667] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Multi-modal synergistic therapy, especially the integration of near-infrared laser phototherapies and chemotherapy, is often sought after owing to its minimal invasiveness, low side effects, and improved anticancer therapeutic efficacy. Herein, CuS nanoparticles were first coated with zinc phthalocyanine derivant (Pc)-functionalized mesoporous silica (mSiO2-Pc) to achieve a drug delivery system (CuS@mSiO2-Pc) with photothermal/photodynamic therapy. Chemical drug DOX was subsequently loaded for chemotherapy, and hyaluronic acid (HA) was employed as a covering material with cancer targeting. The as-obtained CuS@mSiO2-Pc(DOX)@HA nanoparticles were nano-sized with good biocompatibility, effective DOX loading, and controllable DOX releasing. Expectedly, this multifunctional nanoplatform exhibits effective generation of reactive oxygen species and hyperthermia upon the near-infrared laser irradiation. Most importantly, the nanoparticles were targeted into 4T1 cells and showed significantly remarkable cytotoxicity under near-infrared laser irradiation, proving their synergistic therapeutic efficacy. Therefore, this targeted drug system based on CuS with synergistic photothermal therapy/photodynamic therapy/chemotherapy has great application prospects in clinical anticancer treatment for triple negative breast cancer.
Collapse
Affiliation(s)
- Huihui Lv
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116Fujian, P. R. China
| | - Yuchao Zhu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116Fujian, P. R. China
| | - Jinping Xue
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116Fujian, P. R. China
| | - Xiao Jia
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116Fujian, P. R. China
| | - Juanjuan Chen
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350116Fujian, P. R. China
| |
Collapse
|
43
|
Ortiz Valdez E, Rangel-Escareño C, Matus Santos JA, Vázquez Romo R, Guijosa A, Villarreal-Garza C, Arrieta O, Rodríguez-Bautista R, Caro-Sánchez CH, Ortega Gómez A. Characterization of triple negative breast cancer gene expression profiles in Mexican patients. Mol Clin Oncol 2022; 18:5. [PMID: 36605097 PMCID: PMC9808158 DOI: 10.3892/mco.2022.2601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/05/2022] [Indexed: 12/23/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive type of cancer that accounts for ~23% of breast tumors in Mexico. In an attempt to understand in an improved way the behavior of TNBC, throughout the years, gene expression in these tumors has been studied. Lehman et al identified 6 subtypes of gene expression in TNBC with distinct characteristics. In the present study, it was aimed to assess clinical, pathological and prognostic characteristics of TNBC in a Mexican-based cohort. A total of 55 patients diagnosed with TNBC at Mexico's National Institute of Cancer (INCan) were included. Tumor needle biopsy samples were obtained and subjected to microarray analysis. Patients were thus classified into one of the 6 TNBC molecular subtypes. The prognostic, clinical and pathological information of patients was obtained, and differences across molecular subtypes were sought. Out of the 55 included patients, the following subtypes were identified: 9 basal-like-1, 11 basal-like-2 (BSL2), 16 immunomodulatory (IM), 12 mesenchymal, 6 androgen receptor-like and 1 mesenchymal stem-like. Mean follow-up time was 47.1 months. The IM molecular subtype had the best overall survival (OS) (median OS was not reached). BSL2 had the worst OS (15 months). A complete pathologic response to neoadjuvant chemotherapy was obtained more often in the IM subtype (P=0.032). No significant associations were found between any of the clinical or pathological characteristics and the TNBC molecular subtypes. The results obtained from the present study should be considered when seeking to implement a clinical-molecular model for TNBC patient care, particularly in Hispanic-based populations, as they have been frequently underrepresented in clinical studies assessing TNBC molecular subtypes.
Collapse
Affiliation(s)
- Eric Ortiz Valdez
- Breast Tumors Department, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Claudia Rangel-Escareño
- Computational Genomics Department, Instituto Nacional de Medicina Genómica, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico
| | - Juan Antonio Matus Santos
- Breast Tumors Department, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Rafael Vázquez Romo
- Breast Tumors Department, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Alberto Guijosa
- School of Medicine, Universidad Panamericana, Benito Juárez, Mexico City 03920, Mexico
| | - Cynthia Villarreal-Garza
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnológico de Monterrey, Real San Agustín, San Pedro Garza García, Nuevo León 66278, Mexico
| | - Oscar Arrieta
- Thoracic Oncology Unit, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Rubén Rodríguez-Bautista
- Thoracic Oncology Unit, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Claudia H. Caro-Sánchez
- Pathology Department, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Alette Ortega Gómez
- Laboratory of Translational Medicine, Mexico's National Institute of Cancer, Sección XVI, Tlalpan, Mexico City 14080, Mexico,Correspondence to: Dr Alette Ortega Gómez, Laboratory of Translational Medicine, Mexico's National Institute of Cancer, 22 San Fernando Avenue, Sección XVI, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
44
|
PKR-Mediated Phosphorylation of eIF2a and CHK1 Is Associated with Doxorubicin-Mediated Apoptosis in HCC1143 Triple-Negative Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms232415872. [PMID: 36555509 PMCID: PMC9779813 DOI: 10.3390/ijms232415872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/11/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Triple-negative breast cancer is more aggressive than other types of breast cancer. Protein kinase R (PKR), which is activated by dsRNA, is known to play a role in doxorubicin-mediated apoptosis; however, its role in DNA damage-mediated apoptosis is not well understood. In this study, we investigated the roles of PKR and its downstream players in doxorubicin-treated HCC1143 triple-negative breast cancer cells. Doxorubicin treatment induces DNA damage and apoptosis. Interestingly, doxorubicin treatment induced the phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α) via PKR, whereas the inhibition of PKR with inhibitor C16 reduced eIF2α phosphorylation. Under these conditions, doxorubicin-mediated DNA fragmentation, cell death, and poly(ADP ribose) polymerase and caspase 7 levels were recovered. In addition, phosphorylation of checkpoint kinase 1 (CHK1), which is known to be involved in doxorubicin-mediated DNA damage, was increased by doxorubicin treatment, but blocked by PKR inhibition. Protein translation was downregulated by doxorubicin treatment and upregulated by blocking PKR phosphorylation. These results suggest that PKR activation induces apoptosis by increasing the phosphorylation of eIF2α and CHK1 and decreasing the global protein translation in doxorubicin-treated HCC1143 triple-negative breast cancer cells.
Collapse
|
45
|
Kesharwani P, Chadar R, Shukla R, Jain GK, Aggarwal G, Abourehab MAS, Sahebkar A. Recent advances in multifunctional dendrimer-based nanoprobes for breast cancer theranostics. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2433-2471. [PMID: 35848467 DOI: 10.1080/09205063.2022.2103627] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Breast cancer (BC) undoubtedly is one of the most common type of cancers amongst women, which causes about 5 million deaths annually. The treatments and diagnostic therapy choices currently available for Breast Cancer is very much limited . Advancements in novel nanocarrier could be a promising strategy for diagnosis and treatments of this deadly disease. Dendrimer nanoformulation could be functionalized and explored for efficient targeting of overexpressed receptors on Breast Cancer cells to achieve targeted drug delivery, for diagnostics and to overcome the resistance of the cells towards particular chemotherapeutic. Additionally, the dendrimer have shown promising potential in the improvement of therapeutic value for Breast Cancer therapy by achieving synergistic co-delivery of chemotherapeutics and genetic materials for multidirectional treatment. In this review, we have highlighted the application of dendrimer as novel multifunctional nanoplatforms for the treatment and diagnosis of Breast Cancer.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.,University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, U.P, India
| | - Gaurav K Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
46
|
Ezenwafor TC, Uzonwanne VO, Madukwe JUA, Amin SM, Anye VC, Obayemi JD, Odusanya OS, Soboyejo WO. Adhesion of LHRH/EphA2 to human Triple Negative Breast Cancer tissues. J Mech Behav Biomed Mater 2022; 136:105461. [PMID: 36195050 DOI: 10.1016/j.jmbbm.2022.105461] [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: 06/15/2022] [Revised: 08/26/2022] [Accepted: 09/08/2022] [Indexed: 11/19/2022]
Abstract
The adhesive interactions between molecular recognition units (such as specific peptides and antibodies) and antigens or other receptors on the surfaces of tumors are of great value in the design of targeted nanoparticles and drugs for the detection and treatment of specific cancers. In this paper, we present the results of a combined experimental and theoretical study of the adhesion between Luteinizing Hormone Releasing Hormone (LHRH)/Epherin type A2 (EphA2)-AFM coated tips and LHRH/EphA2 receptors that are overexpressed on the surfaces of human Triple Negative Breast Cancer (TNBC) tissues of different histological grades. Following a histochemical and immuno-histological study of human tissue extracts, the receptor overexpression, and their distributions are characterized using Immunohistochemistry (IHC), Immunofluorescence (IF), and a combination of fluorescence microscopy and confocal microscopy. The adhesion forces between LHRH or EphA2 and human TNBC breast tissues are measured using force microscopy techniques that account for the potential effects of capillary forces due to the presence of water vapor. The corresponding adhesion energies are also determined using adhesion theory. The pull off forces and adhesion energies associated with higher grades of TNBC are shown to be greater than those associated with normal/non-tumorigenic human breast tissues, which were studied as controls. The observed increase in adhesion forces and adhesion energies are also correlated with the increasing incidence of LHRH/EphA2 receptors at higher grades of TNBC. The implications of the results are discussed for the development of targeted nanostructures for the detection and treatment of TNBC.
Collapse
Affiliation(s)
- Theresa C Ezenwafor
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria; NASENI Centre of Excellence in Nanotechnology and Advanced Materials, Km 4, Ondo Road, Akure, Ondo State, Nigeria; Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA; Department of Biomedical Engineering, Worcester Polytechnic Institute, Gateway Park Life Sciences and Bioengineering Centre, 60 Prescott Street, Worcester, MA, 01609, USA
| | - Vanessa O Uzonwanne
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA; Department of Biomedical Engineering, Worcester Polytechnic Institute, Gateway Park Life Sciences and Bioengineering Centre, 60 Prescott Street, Worcester, MA, 01609, USA
| | - Jonathan U A Madukwe
- Department of Histopathology, National Hospital, Abuja, Federal Capital Territory (FCT), Nigeria
| | - Said M Amin
- Department of Histopathology, National Hospital, Abuja, Federal Capital Territory (FCT), Nigeria
| | - Vitalis C Anye
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria
| | - John D Obayemi
- Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA; Department of Biomedical Engineering, Worcester Polytechnic Institute, Gateway Park Life Sciences and Bioengineering Centre, 60 Prescott Street, Worcester, MA, 01609, USA
| | - Olushola S Odusanya
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria; Biotechnology and Genetic Engineering Advanced Laboratory, Sheda Science and Technology Complex (SHESTCO), Kwale, Abuja, Federal Capital Territory, Nigeria
| | - Winston O Soboyejo
- Department of Materials Science and Engineering, African University of Science and Technology, Km 10 Airport Road, Galadimawa, Abuja, Federal Capital Territory (FCT), Nigeria; Department of Mechanical and Materials Engineering, Worcester Polytechnic Institute (WPI), 100 Institute Road, Worcester, MA, 01609, USA; Department of Biomedical Engineering, Worcester Polytechnic Institute, Gateway Park Life Sciences and Bioengineering Centre, 60 Prescott Street, Worcester, MA, 01609, USA.
| |
Collapse
|
47
|
Parmanik A, Bose A, Ghosh B, Paul M, Itoo A, Biswas S, Arakha M. Development of triphala churna extract mediated iron oxide nanoparticles as novel treatment strategy for triple negative breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
48
|
Wang H, Su W, Lowe S, Zhou Z, Bentley R, Zhou Q, Cheng C, Guo X, Song Q, Liang Q, Li N, Liang M, Zhu Y, Sun C. Association of Apatinib and Breast Cancer: A systematic review and meta-analysis. Surg Oncol 2022; 44:101818. [DOI: 10.1016/j.suronc.2022.101818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/15/2022] [Accepted: 07/08/2022] [Indexed: 10/17/2022]
|
49
|
Xia J, Wang Q, Ju F, Luo X, Wang F, Zhou Y, Huang H, Wang H, Bao X. Chloride Intracellular Channel 1 is a Potential Biomarker for Breast Cancer. BREAST CANCER: TARGETS AND THERAPY 2022; 14:247-258. [PMID: 36081926 PMCID: PMC9447450 DOI: 10.2147/bctt.s367519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022]
Abstract
Purpose Multiple reports have demonstrated that highly expressed chloride intracellular channel 1 (CLIC1) exists in a range of malignant tumors and is involved in proliferation, invasion, and migration of cancer cells. There are few studies on CLIC1 and breast cancer (BC). The purpose of this research was to evaluate the expression level of CLIC1 in BC and its impact on prognosis of BC patients. Patients and Methods Differences in CLIC1 expression levels in 25 pairs of BC and corresponding paracancerous specimens were tested by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot (WB). Immunohistochemistry (IHC) was performed to discuss the relevance between CLIC1 expression in BC tissue chips and clinicopathological parameters of BC patients. The effect of CLIC1 expression on patient prognosis was evaluated by Kaplan–Meier survival curve and Cox regression analysis. Receiver operating characteristic (ROC) curve assessed the diagnostic performance of CLIC1 for BC. Results The experimental results of qRT-PCR and WB demonstrated that CLIC1 was highly expressed in BC tissues. IHC results showed that overexpression of CLIC1 was strictly correlated with tumor size, TNM classification, pathological grade, lymph node metastasis and Ki67. Patients with lower CLIC1 expression had longer overall survival (OS) and progression-free survival (PFS). Cox regression analysis and ROC curve confirmed that CLIC1 could independently influence the prognosis of BC patients and might have diagnostic efficiency. Conclusion Overexpressed CLIC1 is closely related to the progression of BC and the poor prognosis of the patients, suggesting that it may act as a potential biological diagnostic index for BC.
Collapse
Affiliation(s)
- Jinwen Xia
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
- Clinical Medicine, Medical College, Nantong University, Nantong, People’s Republic of China
| | - Quhui Wang
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Fei Ju
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Xiang Luo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Feng Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Youlang Zhou
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Hua Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Hua Wang
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
- Clinical Medicine, Medical College, Nantong University, Nantong, People’s Republic of China
- Correspondence: Hua Wang, Department of Breast and Thyroid Surgery, Affiliated Hospital of Nantong University, No. 20, Xisi Road, Chongchuan District, Nantong City, Jiangsu Province, 226001, People’s Republic of China, Tel +86 137 062 92250, Email
| | - Xingli Bao
- Department of Medical Equipment, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| |
Collapse
|
50
|
Pe KCS, Saetung R, Yodsurang V, Chaotham C, Suppipat K, Chanvorachote P, Tawinwung S. Triple-negative breast cancer influences a mixed M1/M2 macrophage phenotype associated with tumor aggressiveness. PLoS One 2022; 17:e0273044. [PMID: 35960749 PMCID: PMC9374254 DOI: 10.1371/journal.pone.0273044] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 08/02/2022] [Indexed: 12/05/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by excessive accumulation of tumor-infiltrating immune cells, including tumor-associated macrophages (TAMs). TAMs consist of a heterogeneous population with high plasticity and are associated with tumor aggressiveness and poor prognosis. Moreover, breast cancer cells can secrete factors that influence TAM polarization. Therefore, this study aimed to evaluate the crosstalk between cancer cells and macrophages in the context of TNBC. Cytokine-polarized M2 macrophage were used as control. Distinct from the classical M2 macrophage, TAMs generated from TNBC-conditioned media upregulated both M1- and M2-associated genes, and secreted both the anti-inflammatory cytokine interleukin IL-10 and the proinflammatory cytokine IL-6 and tumor necrosis factor- α. Theses TNBC-induced TAMs exert aggressive behavior of TNBC cells. Consistently, TCGA and MTABRIC analyses of human breast cancer revealed upregulation of M1- associated genes in TNBC comparing with non-TNBC. Among these M1-associated genes, CXCL10 and IL1B were revealed to be independent prognostic factors for disease progression. In conclusion, TNBC cells induce macrophage polarization with a mixture of M1 and M2 phenotypes. These cancer-induced TAMs further enhance tumor cell growth and aggressiveness.
Collapse
Affiliation(s)
- Kristine Cate S. Pe
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology, Chulalongkorn University, Bangkok, Thailand
| | - Rattana Saetung
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology, Chulalongkorn University, Bangkok, Thailand
| | - Varalee Yodsurang
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology, Chulalongkorn University, Bangkok, Thailand
| | - Chatchai Chaotham
- Faculty of Pharmaceutical Sciences, Department of Biochemistry and Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Koramit Suppipat
- Faculty of Medicine, Department of Research Affair, Chulalongkorn University, Bangkok, Thailand
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Pithi Chanvorachote
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology, Chulalongkorn University, Bangkok, Thailand
| | - Supannikar Tawinwung
- Faculty of Pharmaceutical Sciences, Department of Pharmacology and Physiology, Chulalongkorn University, Bangkok, Thailand
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|