International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Breast cancer remains one of the most prevalent malignancies among women globally, with triple-negative breast cancer (TNBC) representing one of its most aggressive and therapeutically challenging subtypes (1). TNBC accounts for approximately 15–20% of breast cancer cases and is defined by the lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) (2,3). This subtype is more commonly observed in younger women, particularly those under 40 years of age, and is frequently associated with early recurrence and high metastatic potential (3,4).
Patients with TNBC often face significant physical and psychological burdens during treatment, including fatigue, hepatic dysfunction, anxiety, and depressive symptoms. Supportive strategies, such as the administration of melatonin and silymarin, have shown beneficial effects in reducing chemotherapy-related fatigue and toxicity (5,6). Psychological support with agents like crocin has also been effective in mitigating distress associated with treatment (7). In parallel, biological markers such as cytokeratin 18 (CK18) are under investigation as indicators of therapeutic response, helping to personalize treatment regimens (8).
Eribulin mesylate (Halaven®), a synthetic analog of the natural product halichondrin B, functions as a microtubule dynamics inhibitor and has been approved for use in patients with advanced breast cancer (9,10). Pivotal trials such as EMBRACE (11) and Study 301 (12) have demonstrated its clinical benefit in metastatic breast cancer, particularly among patients previously treated with anthracycline and taxane-based regimens.
Notably, its effectiveness may be improved when used in combination with other agents. Its efficacy may improve when combined with synergistic agents.
Cisplatin, a platinum-based compound, induces cytotoxicity primarily through DNA crosslinking and inhibition of DNA repair, resulting in tumor cell apoptosis. Although it is an established agent in TNBC therapy, its use is often constrained by its dose-limiting toxicities (13,14). Nevertheless, platinum-based regimens continue to hold relevance in TNBC treatment, with recent clinical guidelines recommending carboplatin-taxane combinations as a neoadjuvant option for HER2-negative and TNBC cases (15,16).
In our previous investigation (17), we demonstrated the synergistic cytotoxic potential of eribulin combined with cisplatin in TNBC models. Building upon those findings, the present study explores the mechanistic basis of this synergy, particularly its relationship with autophagy-a regulated cellular process increasingly implicated in tumor progression and therapeutic resistance. Given the therapeutic promise of targeting autophagy in TNBC, we hypothesized that dual treatment with eribulin and cisplatin could potentiate antitumor effects by modulating this pathway.
Moreover, in light of global disparities in access to advanced cancer therapies, particularly in low- and middle-income countries (LMICs), there is a pressing need to develop cost-effective treatment strategies. The use of existing, clinically approved drugs with known safety profiles, such as eribulin and cisplatin, offers a pragmatic avenue for broadening treatment access and improving outcomes in resource-constrained settings (18).
Taken together, these considerations highlight the urgent need for treatment strategies that combine mechanistic efficacy with cost-effectiveness, particularly in the management of TNBC.
All cell culture reagents, including Dulbecco's Modified Eagle Medium (DMEM), fetal bovine serum (FBS), and penicillin/streptomycin, were sourced from HyClone (GE Healthcare Life Sciences, Logan, UT, USA). Trypsin-EDTA was acquired from Gibco (Thermo Fisher Scientific, Waltham, MA, USA).
Eribulin mesylate (1 mg/vial) was generously provided by Eisai Co., Ltd. (Tokyo, Japan), and cisplatin was obtained from JW Pharmaceutical (Seoul, Korea). The half-maximal inhibitory concentration (IC50) of eribulin in MDA-MB-231 cells was determined via CCK-8 assay after 72 h of treatment and found to be 40.12 µM. Based on this, a concentration of 60 µM-approximately 1.5 times the IC50- was selected for subsequent combination experiments. This supra-physiological dosing strategy aligns with previous mechanistic synergy studies, such as Ko et al (17) in TNBC cells combining eribulin and cisplatin and Swami et al (19) demonstrating similar effects in SK-BR-3 cells, as well as the in vitro/in vivo work by Terashima et al (20) on EMT/MET modulation in TNBC treated with eribulin plus S-1.
PD98059 (a MAPK inhibitor) and crystal violet dye were procured from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Primary antibodies against LC3-I/II (cat. no. 4108), phospho-ERK1/2 (Thr202/Tyr204; cat. no. 9101), and β-actin (cat. no. 4967) were purchased from Cell Signaling Technology. Additional antibodies against ERK (SC-94) and p62 (sc-48389) were acquired from Santa Cruz Biotechnology (Dallas, TX, USA). Secondary antibodies conjugated with horseradish peroxidase (anti-mouse: cat. no. 7076; anti-rabbit: cat. no. 7074) were also from Cell Signaling Technology. Chemiluminescence reagents (Super Signal® West Pico) and DAPI stain were provided by Thermo Fisher. CCK-8 kits were sourced from Dojindo Molecular Technologies (Japan), and autophagy assays (ab139484) were performed using kits from Abcam (Cambridge, MA, USA). Annexin V-FITC apoptosis detection kits were obtained from Koma Biotech (Seoul, Korea).
MDA-MB-231, a human breast cancer cell line, was obtained from the Korean Cell Line Bank. Cells were grown in DMEM supplemented with 10% FBS, antibiotics (100 U/ml penicillin and 100 µg/ml streptomycin), sodium pyruvate (1 mM), sodium bicarbonate (1.5 g/l), and glucose (4.5 g/l). Cultures were maintained in a humidified 5% CO2 incubator at 37°C.
To evaluate cytotoxicity, cells were plated at 5,000 cells per well in 96-well plates and allowed to adhere overnight. Treatments with test compounds were applied for 72 h. After incubation, CCK-8 reagent (100 µl) was added, and absorbance at 450 nm was measured after a 2-h incubation using a microplate reader.
Colony formation assays were conducted to evaluate long-term proliferative capacity. MDA-MB-231 cells were seeded at low density in 6-well plates and treated with eribulin, cisplatin, or their combination for 14 days. Colonies were fixed with methanol and stained with 0.5% crystal violet.
Colonies were defined as discrete clusters containing ≥50 cells. Due to the dispersed and small-sized colonies formed by MDA-MB-231 cells even after 14 days, manual quantification under a microscope was technically unfeasible. Thus, colony numbers and areas were quantified using ImageJ software (NIH, USA) following standard image analysis steps: conversion to 8-bit grayscale, threshold adjustment, watershed segmentation, and automated particle counting via the ‘Analyze Particles’ function.
Cells at approximately 80% confluency were treated and harvested. Cell lysis was performed using buffer containing Tris, NaCl, EDTA, Triton X-100, NP-40, PI, DTT, and PMSF. Lysates were centrifuged, and protein concentrations were measured using a BCA protein assay kit. Protein (20 µg/lane) was resolved by SDS-PAGE and transferred onto PVDF membranes. Blots were blocked with 5% milk in PBST and incubated with primary antibodies overnight. HRP-conjugated secondary antibodies were applied, and bands were visualized with enhanced chemiluminescence. Band intensities were quantified and normalized to the respective loading controls.
Apoptosis was quantified using an Annexin V-FITC/PI kit as per the manufacturer's instructions. Cells treated for 72 h were collected, stained with Annexin V-FITC and PI, and analyzed via flow cytometry. Histogram analysis was done using Kaluza software.
Autophagy flux was examined using Abcam's fluorescent dye-based assay (ab139484), which selectively labels autophagic compartments. Cells were cultured in 8-well chamber slides and treated with the indicated compounds (100 µM) for the specified time, fixed with paraformaldehyde, and stained with autophagy-specific dyes according to the manufacturer's instructions. Fluorescent signals were observed using a confocal microscope and quantified with ImageJ.
To assess drug synergy, the CI was computed using the Chou-Talalay method with CompuSyn software (version 1.0, Combosyn Inc., Paramus, NJ, USA). CI values were interpreted as follows: CI <1 (synergy), CI=1 (additivity), and CI >1 (antagonism).
All experiments were independently repeated at least three times. All statistical analyses were performed using SPSS version 29.0 for Windows (IBM Corp.). One-way ANOVA was used with Tukey's Honestly Significant Difference post hoc analysis. Data are shown as the mean ± SD. P<0.05 was considered to indicate a statistically significant difference.
Cell viability was assessed using the CCK-8 assay (Fig. 1A), and the synergistic efficacy of the drug combination was further confirmed by colony formation assays (Fig. 1B and C). Table I summarizes the CI values derived for the two drugs in this cell model. Single treatment with eribulin mesylate (60 µM) decreased cell viability to 75.11±0.41% after 72 h of exposure. Cisplatin (60 µM) single treatment showed a reduction of 50.57±0.20%. The combination of these drugs significantly inhibited cell viability to 17.15±0.10% (Fig. 1A). Colony-forming assays were performed to test the ability of single cells to grow into colonies. This drug combination synergistically inhibited colony formation by MDA-MB-231 cells (Fig. 1B and C).
We observed that the ERK phosphorylation level increased more than 5.5-fold in the experimental cells compared to that in the control cells after the administration of 60 µM eribulin for 72 h (Fig. 2A and B). Moreover, 60 µM cisplatin increased ERK phosphorylation 6.0-fold (Fig. 2A and B). In addition, when cisplatin was added to eribulin, ERK activation increased by 14.8 times. When cisplatin was added to eribulin, ERK activation was elevated 14.8-fold compared to the control, reflecting a 2.7- and 2.5-fold increase relative to eribulin and cisplatin treatment alone, respectively. These findings confirmed that ERK1/2 activation increased synergistically with eribulin-cisplatin combination.
To evaluate autophagy induction, the LC3-I/II ratio and p62 expression were measured. As autophagy markers, the microtubule-associated protein LC3-I/II ratio and p62 expression were determined using western blot analysis (Fig. 3A). The LC3-I/II level increased 4.3-fold in the eribulin group compared to that in the control group. Cisplatin alone increased the expression of these parameters by 6.1-fold; the corresponding value for eribulin-cisplatin combination treatment was 13.9-fold, indicating a synergistic effect (Fig. 3B). A significant downregulation of p62 was observed following co-treatment with eribulin and cisplatin, suggesting promoted autophagic degradation (Fig. 3C). The corresponding values for eribulin alone and eribulin-cisplatin combination were 9.0 and 23.6%, respectively, indicating a significant increase in autophagic activity in the combination group compared with the eribulin-only group (Fig. 3D and E). Autophagic vacuole staining showed a slight increase in autophagic activity with either eribulin or cisplatin alone, whereas their combination resulted in a marked enhancement, indicating cumulative autophagic activation.
The apoptotic activity of the combination of eribulin and cisplatin was analyzed by flow cytometry after double staining with annexin V and propidium iodide (Fig. 4A). The apoptotic activity levels were 28.61, 64.77, and 99.26% when the cells were treated with eribulin alone, cisplatin alone, or a combination thereof, respectively. Upon co-treatment with 3-methyladenine, a known autophagy inhibitor, the apoptotic rate induced by the eribulin-cisplatin combination significantly decreased, suggesting that the observed cytotoxicity is dependent on autophagic mechanisms. When 3-methyladenine samples were treated with a combination of eribulin and cisplatin, the apoptotic cell volume was reduced to 13.68% compared with 99.26% in the eribulin-cisplatin combination group (Fig. 4A and B).
To assess the impact of ERK inhibition, cells were pretreated with PD98059 prior to exposure to eribulin and cisplatin, followed by a viability assessment using the CCK-8 assay. When PD98059 was combined with the two-drug combination, the cell viability increased significantly from 33.63 to 53.37% (Fig. 5A). Flow cytometry was used to quantify apoptotic cells following dual labeling with Annexin V and propidium iodide (Fig. 5B). Compared with the eribulin and cisplatin combination group, the apoptosis rate decreased from 92.72 to 18.01% when PD98059 was used (Fig. 5C).
Using the control value as the reference (100%), LC3-I/II expression was markedly increased in the eribulin plus cisplatin group (824.99%) but was reduced to 119.30% when PD98059 was additionally administered, whereas p62 expression increased from 38.70 to 72.59% with PD98059 treatment (Fig. 5D-F). Thus, ERK inhibition affected the expression of autophagy-related proteins.
An autophagy assay (ab139484, Abcam) confirmed that the levels of autophagosomes decreased when PD98059 was used in combination with eribulin and cisplatin (Fig. 5G). Quantitative analysis of fluorescence intensity showed consistent results, decreasing from 15.59 to 2.92% (P<0.05; Fig. 5H).
In this study, MDA-MB-231 cells were used to investigate the cytotoxic effect of a combination of eribulin and cisplatin, both widely utilized in TNBC treatment. After 72 h of treatment, significant cancer cell death was observed, mediated by autophagy-dependent mechanisms involving ERK pathway activation. These findings reveal a novel therapeutic vulnerability in TNBC.
Eribulin, a microtubule-targeting agent derived from the marine sponge Halichondria okadai, has demonstrated clinical efficacy in breast cancer, particularly in taxane- and anthracycline-resistant cases (21,22). The EMBRACE trial notably supported its use in metastatic breast cancer, with further analyses confirming its benefit in TNBC (11). Previous studies have shown that eribulin can synergize with various agents, including HDAC inhibitors and RAF/MEK inhibitors, primarily through ERK pathway inactivation (23,24). However, our study uniquely reveals that eribulin induces cell death via ERK pathway activation, representing, to our knowledge, the first report of this mechanism in TNBC.
Cisplatin, a platinum-based chemotherapeutic, also activates ERK and induces apoptosis across multiple carcinoma types including cervical cancer (25), hepatocellular carcinoma (26), human glioma (27), and mouse proximal tubule cancer (28). Consistent with prior reports, our study confirms that ERK activation persists in MDA-MB-231 cells following cisplatin treatment, contributing to cell death. Remarkably, the combination of eribulin and cisplatin further amplified this ERK-driven autophagic response, leading to significantly enhanced cytotoxic effects against TNBC cells.
TNBC is molecularly defined by the absence of estrogen receptor (ER), progesterone receptor (PR), and HER2 overexpression, rendering it unresponsive to endocrine or HER2-targeted therapies. This receptor-negative profile contributes to poor prognosis and compels TNBC cells to rely on alternative survival pathways such as the MAPK/ERK axis (29,30). Our finding that ERK activation mediates autophagy-dependent cell death in response to the combination therapy suggests that this pathway-typically associated with cellular adaptation and drug resistance-may instead represent a vulnerability in TNBC (31). Furthermore, high basal autophagic activity, often driven by MAPK/ERK signaling, has been implicated in TNBC progression and response to therapy, indicating that modulation of this pathway could sensitize cells to cytotoxic agents (32,33).
Autophagy, which often intersects with apoptotic mechanisms, is increasingly recognized as a modulator of cancer therapy response (34,35). Our data indicate that ERK activation is not merely associated with autophagic flux but is a functional mediator of combination-induced cell death. This positions ERK-mediated autophagy as a viable target for combination therapy in TNBC, particularly in the context of eribulin and cisplatin co-treatment (36,37).
Importantly, the clinical relevance of this dual-agent strategy extends beyond mechanistic insights. Although targeted therapies have revolutionized TNBC management in high-income countries, their high cost continues to restrict access in low- and middle-income countries (LMICs) (38). Given that both eribulin and cisplatin are already approved and relatively affordable, their combination may offer a scalable and cost-effective solution to mitigate global disparities in breast cancer treatment (11,17,18). This approach is consistent with current global oncology efforts aimed at expanding access to effective therapies in resource-limited settings.
In LMICs, financial barriers significantly limit access to novel targeted therapies (39). The monthly cost of immune checkpoint inhibitors or PARP inhibitors can exceed $5,000 USD, making them unattainable for most patients (40). In contrast, cisplatin and eribulin-being off-patent or comparatively affordable-are frequently listed in public health formularies. For instance, in South Korea, the cost of eribulin treatment under the national insurance system is approximately $1,500-$2,000 USD per cycle, substantially lower than newer biologics [HIRA (Health Insurance Review and Assessment Service, Korea's national health technology assessment body)]. These economic factors reinforce the feasibility of the eribulin-cisplatin combination, especially in regions striving to deliver cost-effective cancer care (17,18).
In summary, this is the first report demonstrating that eribulin alone can activate the ERK pathway to induce autophagy-mediated cytotoxicity in TNBC. These findings highlight ERK-mediated autophagy as a promising therapeutic target and underscore the clinical feasibility of the eribulin-cisplatin combination in both advanced and resource-limited settings.
Nevertheless, this study has limitations. As an in vitro investigation, it cannot fully recapitulate the complex tumor microenvironment. In vivo validation using xenograft models will be necessary to confirm the translational potential of the eribulin-cisplatin combination, particularly its impact on ERK-mediated autophagic cell death. However, the recent NCCN guidelines supporting platinum-based neoadjuvant regimens in TNBC provide a clinical rationale for future studies (18).
In conclusion, our findings demonstrate that ERK-driven autophagy mediates the synergistic cytotoxicity of eribulin and cisplatin in TNBC. This mechanism highlights a clinically feasible and cost-effective therapeutic strategy, warranting further in vivo validation and clinical development.
Not applicable.
This work was supported by Chungnam National University.
The data generated in the present study may be requested from the corresponding author.
HK and JL conceived and designed the study. HK, ML, EC and JL contributed to the design of the methodology and performed experiments. Data analysis and validation were conducted by HK and JL. Software tool use and visualization were performed by HK. The experiments were performed by ML, EC and JL, with resources provided by ML and EC. Data curation was performed by HK, ML and EC. HK drafted the original manuscript, and JL performed critical review and editing. Supervision and project administration were led by JL. Funding for the project was acquired by JL. HK and JL confirm the authenticity of all the raw data. All authors have read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
ORCID IDs: HK, 0000-0002-6621-8761; JL, 0000-0002-9345-724X; ML, 0000-0003-1732-3001; EC, 0000-0002-7281-3698.
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, Bilous M, Fitzgibbons P, et al: Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American society of clinical oncology/college of American pathologists clinical practice guideline update. J Clin Oncol. 31:3997–4013. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Morris GJ, Naidu S, Topham AK, Guiles F, Xu Y, McCue P, Schwartz GF, Park PK, Rosenberg AL, Brill K and Mitchell EP: Differences in breast carcinoma characteristics in newly diagnosed African-American and Caucasian patients: A single-institution compilation compared with the national cancer institute's surveillance, epidemiology, and end results database. Cancer. 110:876–884. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P and Narod SA: Triple-negative breast cancer: Clinical features and patterns of recurrence. Clin Cancer Res. 13:4429–4434. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Sedighi Pashaki A, Sheida F, Moaddab Shoar L, Hashem T, Fazilat-Panah D, Nemati Motehaver A, Ghanbari Motlagh A, Nikzad S, Bakhtiari M, Tapak L, et al: A randomized, controlled, parallel-group, trial on the long-term effects of melatonin on fatigue associated with breast cancer and its adjuvant treatments. Integr Cancer Ther. 22:153473542311686242023. View Article : Google Scholar : PubMed/NCBI | |
|
Moezian GSA, Javadinia SA, Sales SS, Fanipakdel A, Elyasi S and Karimi G: Oral silymarin formulation efficacy in management of AC-T protocol induced hepatotoxicity in breast cancer patients: A randomized, triple blind, placebo-controlled clinical trial. J Oncol Pharm Pract. 28:827–835. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Salek R, Dehghani M, Mohajeri SA, Talaei A, Fanipakdel A and Javadinia SA: Amelioration of anxiety, depression, and chemotherapy related toxicity after crocin administration during chemotherapy of breast cancer: A double blind, randomized clinical trial. Phytother Res. 35:5143–5153. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Fazilat-Panah D, Vakili Ahrari Roudi S, Keramati A, Fanipakdel A, Sadeghian MH, Homaei Shandiz F, Shahidsales S and Javadinia SA: Changes in cytokeratin 18 during neoadjuvant chemotherapy of breast cancer: A prospective study. Iran J Pathol. 15:117–126. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Swami U, Chaudhary I, Ghalib MH and Goel S: Eribulin-a review of preclinical and clinical studies. Crit Rev Oncol Hematol. 81:163–184. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Mani S and Swami U: Eribulin mesilate, a halichondrin B analogue, in the treatment of breast cancer. Drugs Today (Barc). 46:641–653. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Cortes J, O'Shaughnessy J, Loesch D, Blum JL, Vahdat LT, Petrakova K, Chollet P, Manikas A, Diéras V, Delozier T, et al: Eribulin monotherapy versus treatment of physician's choice in patients with metastatic breast cancer (EMBRACE): A phase 3 open-label randomised study. Lancet. 377:914–923. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Kaufman PA, Awada A, Twelves C, Yelle L, Perez EA, Velikova G, Olivo MS, He Y, Dutcus CE and Cortes J: Phase III open-label randomized study of eribulin mesylate versus capecitabine in patients with locally advanced or metastatic breast cancer previously treated with an anthracycline and a taxane. J Clin Oncol. 33:594–601. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Ghosh S: Cisplatin: The first metal based anticancer drug. Bioorg Chem. 88:1029252019. View Article : Google Scholar : PubMed/NCBI | |
|
Dasari S and Tchounwou PB: Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J Pharmacol. 740:364–378. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang C, Xu C, Gao X and Yao Q: Platinum-based drugs for cancer therapy and anti-tumor strategies. Theranostics. 12:2115–2132. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lin C, Cui J, Peng Z, Qian K, Wu R, Cheng Y and Yin W: Efficacy of platinum-based and non-platinum-based drugs on triple-negative breast cancer: meta-analysis. Eur J Med Res. 27:2012022. View Article : Google Scholar : PubMed/NCBI | |
|
Ko H, Lee M, Cha E, Sul J, Park J and Lee J: Eribulin mesylate improves cisplatin-induced cytotoxicity of triple-negative breast cancer by extracellular signal-regulated kinase 1/2 activation. Medicina (Kaunas). 58:4572022. | |
|
Obidiro O, Battogtokh G and Akala EO: Triple Negative breast cancer treatment options and limitations: Future outlook. Pharmaceutics. 15:17962023. View Article : Google Scholar : PubMed/NCBI | |
|
Swami U, Shah U and Goel S: Eribulin in cancer treatment. Mar Drugs. 13:5016–5058. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Terashima M, Sakai K, Togashi Y, Hayashi H, De Velasco MA, Tsurutani J and Nishio K: Synergistic antitumor effects of S-1 with eribulin in vitro and in vivo for triple-negative breast cancer cell lines. Springerplus. 3:4172014. View Article : Google Scholar : PubMed/NCBI | |
|
Kuznetsov G, Towle MJ, Cheng H, Kawamura T, TenDyke K, Liu D, Kishi Y, Yu MJ and Littlefield BA: Induction of morphological and biochemical apoptosis following prolonged mitotic blockage by halichondrin B macrocyclic ketone analog E7389. Cancer Res. 64:5760–5766. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Okouneva T, Azarenko O, Wilson L, Littlefield BA and Jordan MA: Inhibition of centromere dynamics by eribulin (E7389) during mitotic metaphase. Mol Cancer Ther. 7:2003–2011. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ono H, Sowa Y, Horinaka M, Iizumi Y, Watanabe M, Morita M, Nishimoto E, Taguchi T and Sakai T: The histone deacetylase inhibitor OBP-801 and eribulin synergistically inhibit the growth of triple-negative breast cancer cells with the suppression of survivin, Bcl-xL, and the MAPK pathway. Breast Cancer Res Treat. 171:43–52. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ono H, Horinaka M, Sukeno M, Morita M, Yasuda S, Nishimoto E, Konishi E and Sakai T: Novel RAF/MEK inhibitor CH5126766/VS-6766 has efficacy in combination with eribulin for the treatment of triple-negative breast cancer. Cancer Sci. 112:4166–4175. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Martindale JL and Holbrook NJ: Requirement for ERK activation in cisplatin-induced apoptosis. J Biol Chem. 275:39435–39443. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Guégan JP, Ezan F, Théret N, Langouët S and Baffet G: MAPK signaling in cisplatin-induced death: Predominant role of ERK1 over ERK2 in human hepatocellular carcinoma cells. Carcinogenesis. 34:38–47. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Choi BK, Choi CH, Oh HL and Kim YK: Role of ERK activation in cisplatin-induced apoptosis in A172 human glioma cells. Neurotoxicology. 25:915–924. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Arany I, Megyesi JK, Kaneto H, Price PM and Safirstein RL: Cisplatin-induced cell death is EGFR/src/ERK signaling dependent in mouse proximal tubule cells. Am J Physiol Renal Physiol. 287:F543–F549. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Bianchini G, Balko JM, Mayer IA, Sanders ME and Gianni L: Triple-negative breast cancer: Challenges and opportunities of a heterogeneous disease. Nat Rev Clin Oncol. 13:674–690. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Giltnane JM and Balko JM: Rationale for targeting the Ras/MAPK pathway in triple-negative breast cancer. Discov Med. 17:275–283. 2014.PubMed/NCBI | |
|
Sivaprasad U and Basu A: Inhibition of ERK attenuates autophagy and potentiates tumour necrosis factor-alpha-induced cell death in MCF-7 cells. J Cell Mol Med. 12:1265–1271. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Yu S, Cao Z, Cai F, Yao Y, Chang X, Wang X, Zhuang H and Hua ZC: ADT-OH exhibits anti-metastatic activity on triple-negative breast cancer by combinatorial targeting of autophagy and mitochondrial fission. Cell Death Dis. 15:4632024. View Article : Google Scholar : PubMed/NCBI | |
|
Han Y, Fan S, Qin T, Yang J, Sun Y, Lu Y, Mao J and Li L: Role of autophagy in breast cancer and breast cancer stem cells (review). Int J Oncol. 52:1057–1070. 2018.PubMed/NCBI | |
|
Eisenberg-Lerner A, Bialik S, Simon HU and Kimchi A: Life and death partners: Apoptosis, autophagy and the cross-talk between them. Cell Death Differ. 16:966–975. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Rosenfeldt MT and Ryan KM: The multiple roles of autophagy in cancer. Carcinogenesis. 32:955–963. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, He S and Ma B: Autophagy and autophagy-related proteins in cancer. Mol Cancer. 19:122020. View Article : Google Scholar : PubMed/NCBI | |
|
Gewirtz DA: The four faces of autophagy: Implications for cancer therapy. Cancer Res. 74:647–651. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
World Health Organization, . Global breast cancer initiative implementation framework: Assessing, strengthening and scaling up of services for the early detection and management of breast cancer: Executive summary. Geneva: World Health Organization; 2023 | |
|
Arnold M, Morgan E, Rumgay H, Mafra A, Singh D, Laversanne M, Vignat J, Gralow JR, Cardoso F, Siesling S and Soerjomataram I: Current and future burden of breast cancer: Global statistics for 2020 and 2040. Breast. 66:15–23. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Goldstein DA, Chen Q, Ayer T, Howard DH, Lipscomb J, El-Rayes BF and Flowers CR: First- and second-line bevacizumab in addition to chemotherapy for metastatic colorectal cancer: A United States-based cost-effectiveness analysis. J Clin Oncol. 33:1112–1118. 2015. View Article : Google Scholar : PubMed/NCBI |