1
|
Mariano NC, Marotti JD, Chen Y, Karakyriakou B, Salgado R, Christensen BC, Miller TW, Kettenbach AN. Quantitative proteomics analysis of triple-negative breast cancers. NPJ Precis Oncol 2025; 9:117. [PMID: 40269124 PMCID: PMC12019170 DOI: 10.1038/s41698-025-00907-8] [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: 09/11/2024] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15% of all Breast Cancer (BC) cases with poorer prognosis and clinical outcomes compared to other BC subtypes due to greater tumor heterogeneity and few therapeutically targetable oncogenic drivers. To reveal actionable pathways for anti-cancer treatment, we use a proteomic approach to quantitatively compare the abundances of 6306 proteins across 55 formalin-fixed and paraffin-embedded (FFPE) TNBC tumors. We identified four major TNBC clusters by unsupervised clustering analysis of protein abundances. Analyses of clinicopathological characteristics revealed associations between the proteomic profiles and clinical phenotypes exhibited by each subtype. We validate the findings by inferring immune and stromal cell type composition from genome-wide DNA methylation profiles. Finally, quantitative proteomics on TNBC cell lines was conducted to identify in vitro models for each subtype. Collectively, our data provide subtype-specific insights into molecular drivers, clinicopathological phenotypes, tumor microenvironment (TME) compositions, and potential pharmacologic vulnerabilities for further investigations.
Collapse
Affiliation(s)
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | | | | | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Brock C Christensen
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Epidemiology, Lebanon, NH, USA
- Department of Community and Family Medicine, Lebanon, NH, USA
| | - Todd W Miller
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Hanover, NH, USA.
- Dartmouth Cancer Center, Lebanon, NH, USA.
| |
Collapse
|
2
|
Grimaudo MS, D’Orazio F, Renne SL, D’Incalci M, Maki RG, Colombo P, Balzarini L, Laffi A, Santoro A, Bertuzzi AF. Assessment of the Mechanisms of Action of Eribulin in Patients with Advanced Liposarcoma Through the Evaluation of Radiological, Functional, and Tissue Responses: A Prospective Monocentric Study (Malibu Study). Cancers (Basel) 2025; 17:976. [PMID: 40149309 PMCID: PMC11940360 DOI: 10.3390/cancers17060976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Liposarcoma (LPS) is one of the most frequent histotypes of soft tissue sarcoma (STS). Eribulin is a cytotoxic agent that has improved overall survival in patients with advanced LPS. Additionally, preclinical and clinical evidence suggests its influence on vascularization and cellular differentiation. Based on these data, we developed this study to investigate non-mitotic effects of eribulin in patients with advanced LPS. Methods: In this prospective monocentric observational study, we included patients with advanced LPS eligible to receive eribulin. An assessment with dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and a biopsy were planned before treatment and after four cycles of eribulin. DCE-MRI scans were elaborated to obtain perfusion and permeability maps. Results: From September 2019 to January 2024, 11 patients were enrolled. Among them, 8/11 (73%) had successful pre- and post-treatment assessment. At the time of the analysis, 8/11 (73%) patients had disease progression and 4 (36%) had died, median progression-free survival (mPFS) was 3.3 months, and median overall survival (mOS) was 8.7 months. Among the evaluable patients, DCE-MRI perfusion decreased after eribulin treatment in patients with disease control (partial response or stable disease), while perfusion values increased in patients with progressive disease (PD). No significant change in permeability was found. Post-treatment histological changes were seen nearly in all patients, with decreased cellularity the most common change (50%), followed by vascularization modifications (20%). Conclusions: Eribulin appears to exhibit non-mitotic activity involving both vascularization and cell differentiation in LPS patients. Further studies are needed to better define these effects.
Collapse
Affiliation(s)
- Maria Susanna Grimaudo
- Medical Oncology and Hematology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (A.L.); (A.S.); (A.F.B.)
| | - Federico D’Orazio
- Radiology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (F.D.); (L.B.)
| | - Salvatore Lorenzo Renne
- Pathology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (S.L.R.); (P.C.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy;
| | - Maurizio D’Incalci
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy;
- Laboratory of Cancer Pharmacology, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy
| | - Robert G. Maki
- Memorial Sloan-Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA;
- School of Medical Sciences, Weill-Cornell Medical College, 1300 York Ave, New York, NY 10065, USA
| | - Piergiuseppe Colombo
- Pathology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (S.L.R.); (P.C.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy;
| | - Luca Balzarini
- Radiology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (F.D.); (L.B.)
| | - Alice Laffi
- Medical Oncology and Hematology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (A.L.); (A.S.); (A.F.B.)
| | - Armando Santoro
- Medical Oncology and Hematology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (A.L.); (A.S.); (A.F.B.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, MI, Italy;
| | - Alexia Francesca Bertuzzi
- Medical Oncology and Hematology Department, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089 Rozzano, MI, Italy; (A.L.); (A.S.); (A.F.B.)
| |
Collapse
|
3
|
Wu X, Yuan F, Guo L, Gao D, Zheng W, Chen C, Zheng H, Liu J. Intraductal chemotherapy for triple-negative breast cancer: a pathway to minimally invasive clinical treatment. BMC Cancer 2025; 25:285. [PMID: 39966717 PMCID: PMC11837698 DOI: 10.1186/s12885-025-13693-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is traditionally treated with systemic chemotherapy, often resulting in significant off-target toxicity. In this study, we assess the efficacy of intraductal chemotherapeutic delivery, aimed at reducing systemic side effects. Using an in situ TNBC model, created by intraductal injection of 4T1-luc cells, we identified day 3 post-tumor implantation as an optimal early intervention point. Echocardiographic analysis confirmed that intraductal administration of eribulin (ERI) or doxorubicin (DOX) did not cause cardiac dysfunction or apoptosis. Our results demonstrate that intraductal delivery of ERI and DOX significantly enhances anti-tumor and anti-metastatic effects. Mechanistically, ERI followed by DOX increased intratumoral perfusion, improved drug concentration, reversed epithelial-mesenchymal transition, and inhibited tumor cell invasion and metastasis. Additionally, this approach triggered immunogenic cell death and activated a systemic anti-tumor immune response. These findings underscore the potential of intraductal chemotherapy as a safe, highly effective approach, offering a preclinical foundation for minimally invasive TNBC therapies.
Collapse
Affiliation(s)
- Xinhong Wu
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China
| | - Feng Yuan
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China
| | - Liantao Guo
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Dongcheng Gao
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou City, Henan Province, China
| | - Weijie Zheng
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuchang District, Wuhan, 430060, People's Republic of China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuchang District, Wuhan, 430060, People's Republic of China.
| | - Hongmei Zheng
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China.
| | - Jianhua Liu
- Breast cancer center, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, National key clinical specialty discipline construction program, Hubei Provincial Clinical Research Center for Breast Cancer, Wuhan Clinical Research Center for Breast Cancer, No.116 Zhuo Daoquan South Road, 430079, Wuhan, Hubei,, China.
| |
Collapse
|
4
|
Rafiyan M, Tootoonchi E, Golpour M, Davoodvandi A, Reiter RJ, Asemi R, Sharifi M, Rasooli Manesh SM, Asemi Z. Melatonin for gastric cancer treatment: where do we stand? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1265-1282. [PMID: 39287677 DOI: 10.1007/s00210-024-03451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Gastric cancer (GC) is the third leading reason of death in men and the fourth in women. Studies have documented an inhibitory function of melatonin on the proliferation, progression and invasion of GC cells. MicroRNAs (miRNAs) are small, non-coding RNAs that play an important function in regulation of biological processes and gene expression of the cells. Some studies reported that melatonin can suppress the progression of GC by regulating the exosomal miRNAs. Thus, melatonin represents a promising potential therapeutic agent for subjects with GC. Herein, we evaluate the existing data of both in vivo and in vitro studies to clarify the molecular processes involved in the therapeutic effects of melatonin in GC. The data emphasize the critical function of melatonin in several signaling ways by which it may inhibit cancer cell proliferation, decrease chemo-resistance, induce apoptosis as well as limit invasion, angiogenesis, and metastasis. This review provides a resource that identifies some of the mechanisms by which melatonin controls GC enlargement. In light of the findings, melatonin should be considered a novel and testable therapeutic mediator for GC treatment.
Collapse
Affiliation(s)
- Mahdi Rafiyan
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Elham Tootoonchi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mahdieh Golpour
- Student Research Committee, Mazandarn University of Medical Sciences, Sari, Mazandaran, Iran
| | - Amirhossein Davoodvandi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
Zhang M, Huang J, Zheng X, Huang P, Yang X. Cost-effectiveness analysis of eribulin versus dacarbazine in patients with advanced liposarcoma. Sci Rep 2025; 15:2084. [PMID: 39814865 PMCID: PMC11735621 DOI: 10.1038/s41598-024-84247-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
A subgroup analysis of a randomized study demonstrated that patients with advanced or metastatic liposarcoma treated with eribulin had longer overall survival and progression-free survival compared to those treated with dacarbazine, suggesting eribulin as a therapeutic option for advanced liposarcoma. Therefore, this study aims to evaluate the cost-effectiveness of eribulin versus dacarbazine in the treatment of advanced liposarcoma. We established a 10-year Markov model to compare the cost-effectiveness of eribulin and dacarbazine regimens. Clinical data were sourced from a subgroup analysis of a multicenter, randomized, open-label phase 3 trials. Quality-adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs) were computed. The total cost of the dacarbazine scheme was $10,895.558, with a QALY of 0.533. In contrast, the total cost of the eribulin scheme was $16,961.891, with a QALY of 0.698. The ICER between the eribulin and dacarbazine schemes was $36,736.467, which is below the willingness-to-pay (WTP) threshold in China ($37,877.469). From the perspective of the Chinese healthcare system, eribulin is cost-effective compared to dacarbazine at the WTP threshold.
Collapse
Affiliation(s)
- Miaomiao Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Rd, Gongsu District, Hangzhou, 310014, Zhejiang, China
| | - Jinlong Huang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Rd, Gongsu District, Hangzhou, 310014, Zhejiang, China
| | - Xiaochun Zheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Rd, Gongsu District, Hangzhou, 310014, Zhejiang, China
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Rd, Gongsu District, Hangzhou, 310014, Zhejiang, China.
| | - Xiuli Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China.
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 158 Shangtang Rd, Gongsu District, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
6
|
Chan A, Gill J, Chih H, Wright SCE, Vasilevski N, Eichhorn PJA. Influence of Epithelial-Mesenchymal Transition on Risk of Relapse and Outcome to Eribulin or Cyclin-Dependent Kinase Inhibitors in Metastatic Breast Cancer. JCO Precis Oncol 2024; 8:e2400274. [PMID: 39642326 PMCID: PMC11634087 DOI: 10.1200/po.24.00274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 12/08/2024] Open
Abstract
PURPOSE The presence of epithelial-mesenchymal transition (EMT) in breast cancer (BC) cells has been linked to worse prognosis and may influence response to systemic treatment. We explored the effect of EMT in tumor samples of patients with metastatic BC on disease-free interval and overall survival in those patients receiving eribulin or cyclin-dependent kinase 4/6 inhibitors (CDK4/6i). MATERIALS AND METHODS Key inclusion criteria included available archived primary BC tissue and, where available, matched metastatic biopsy. Patients received eribulin and/or a CDK4/6i in the metastatic setting. Specimens were assessed for biomarkers by immunohistochemistry (CDH1, AE1/3, VIM, CDH2, ZEB1, pSMAD2, and SMAD4) and gene expression by droplet digital polymerase chain reaction (CDH1, CDH2, SNAI1 & 2, TWIST1, VIM, PTEN, and ZEB1 & 2). RESULTS Between 2002 and 2020, 127 patients were included (95 early-stage disease at diagnosis with metastatic relapse, 32 de novo metastatic disease). In metastatic samples, presence of ZEB1 overexpression was associated with shorter time to recurrence (48.1 months shorter; P = .003), with pSMAD2 overexpression suggesting clinical significance of 52.0 months shorter; P = .01. High gene expression levels for SNAIL1, TWIST1, and PTEN in the primary BC were associated with significantly longer survival in patients who received eribulin (P < .05); high VIM was associated with a clinically relevant trend toward shorter survival after a CDK4/6i (P = .013). CONCLUSION We demonstrate in our exploratory study that biomarkers involved in the process of EMT could have a prognostic impact in a cohort of patients with BC uniformly treated and with long-term follow-up. Genes known to be involved in EMT were associated with improved eribulin efficacy, while suggesting a poorer outcome with CDK4/6i.
Collapse
Affiliation(s)
- Arlene Chan
- Breast Cancer Research Centre-WA and Curtin University, Perth, Australia
- Curtin Medical School, Curtin University, Bentley, Australia
| | - Jespal Gill
- Anatomical Pathology, Western Diagnostics, Jandakot and PathWest, Murdoch, Australia
| | - HuiJun Chih
- School of Population Health, Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Sarah Christine Elisabeth Wright
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Natali Vasilevski
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Pieter Johan Adam Eichhorn
- Curtin Medical School, Curtin University, Bentley, Australia
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| |
Collapse
|
7
|
Nicolas E, Kosmider B, Cukierman E, Borghaei H, Golemis EA, Borriello L. Cancer treatments as paradoxical catalysts of tumor awakening in the lung. Cancer Metastasis Rev 2024; 43:1165-1183. [PMID: 38963567 PMCID: PMC11554904 DOI: 10.1007/s10555-024-10196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
Much of the fatality of tumors is linked to the growth of metastases, which can emerge months to years after apparently successful treatment of primary tumors. Metastases arise from disseminated tumor cells (DTCs), which disperse through the body in a dormant state to seed distant sites. While some DTCs lodge in pre-metastatic niches (PMNs) and rapidly develop into metastases, other DTCs settle in distinct microenvironments that maintain them in a dormant state. Subsequent awakening, induced by changes in the microenvironment of the DTC, causes outgrowth of metastases. Hence, there has been extensive investigation of the factors causing survival and subsequent awakening of DTCs, with the goal of disrupting these processes to decrease cancer lethality. We here provide a detailed overview of recent developments in understanding of the factors controlling dormancy and awakening in the lung, a common site of metastasis for many solid tumors. These factors include dynamic interactions between DTCs and diverse epithelial, mesenchymal, and immune cell populations resident in the lung. Paradoxically, among key triggers for metastatic outgrowth, lung tissue remodeling arising from damage induced by the treatment of primary tumors play a significant role. In addition, growing evidence emphasizes roles for inflammation and aging in opposing the factors that maintain dormancy. Finally, we discuss strategies being developed or employed to reduce the risk of metastatic recurrence.
Collapse
Affiliation(s)
- Emmanuelle Nicolas
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Beata Kosmider
- Center for Inflammation and Lung Research, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
- Department of Microbiology, Immunology, and Inflammation, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Hossein Borghaei
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA
| | - Lucia Borriello
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19111, USA.
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA, 19140, USA.
| |
Collapse
|
8
|
Hassouneh Z, Noel ODV, Ji N, Kim ME, Svatek J, Svatek RS, Risinger AL, Mukherjee N. Low-Dose Eribulin Promotes NK Cell-Mediated Therapeutic Efficacy in Bladder Cancer. Cancers (Basel) 2024; 16:3875. [PMID: 39594830 PMCID: PMC11592921 DOI: 10.3390/cancers16223875] [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: 10/06/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Despite its immunogenic nature, bladder cancer (BCa) responds sub-optimally to FDA-approved immunotherapy. BACKGROUND/OBJECTIVES We have previously shown that natural killer (NK) cells are major contributors to overall patient survival in BCa. In our efforts to identify clinically approved agents that enhance NK cell activation, we identified eribulin, a microtubule destabilizer primarily used in breast cancer. Ongoing clinical trials are investigating the potential integration of eribulin into the standard of care in BCa; however, the mechanistic rationale for these trials remains unclear. METHODS Here, we explore the effects of low-dose eribulin on direct NK cell activation in vitro, including on primary patient samples, and in vivo utilizing multiple murine models. Flow cytometry and RNA sequencing were employed to identify the mechanism of NK cell activation by eribulin, which was associated with increased migration and cytotoxicity of NK cells against BCa cells. RESULTS We found that localized eribulin instillation significantly reduces bladder tumor burden and improves survival in primary BCa in an NK cell-dependent manner. Importantly, eribulin promoted the shift of patient-derived intratumoral NK cells towards an anti-tumor CD49a+ CD103+ NK subset (ieILC1-like) while diminishing the dysfunctional NR4A2-expressing CD49a- NK subset. Moreover, it decreased the overall expression of exhaustion markers on NK cells, a pattern replicated in our murine models. CONCLUSIONS These findings are paradigm-shifting given that chemotherapy is traditionally considered immunosuppressive. Our study reveals the novel effect of low-dose eribulin chemotherapy in inhibiting bladder tumor growth by enhancing anti-tumor NK cell immunity, challenging previous assumptions and opening new therapeutic approaches to improve antitumor immunity.
Collapse
Affiliation(s)
- Zaineb Hassouneh
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA;
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
- Mays Cancer Center, San Antonio, TX 78229, USA;
| | - Onika D. V. Noel
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
| | - Niannian Ji
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
| | - Michelle E. Kim
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
- Long School of Medicine, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Jordan Svatek
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
| | - Robert S. Svatek
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
| | - April L. Risinger
- Mays Cancer Center, San Antonio, TX 78229, USA;
- Department of Pharmacology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA
| | - Neelam Mukherjee
- Department of Urology, University of Texas Health San Antonio (UTHSA), San Antonio, TX 78229, USA; (O.D.V.N.); (N.J.); (M.E.K.); (J.S.); (R.S.S.)
| |
Collapse
|
9
|
Mangaonkar S, Nath S, Chatterji BP. Microtubule dynamics in cancer metastasis: Harnessing the underappreciated potential for therapeutic interventions. Pharmacol Ther 2024; 263:108726. [PMID: 39349106 DOI: 10.1016/j.pharmthera.2024.108726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/01/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Microtubules, dynamic cytoskeletal structures crucial for cellular processes, have surfaced as promising targets for cancer therapy owing to their pivotal role in cancer progression and metastasis. This review comprehensively explores the multifaceted landscape of microtubule-targeting drugs and their potential to inihibit cancer metastasis. Although the role of Actin cytoskeleton is well known in controlling metastasis, only recently Microtubules are emerging as a potential controller of metastasis. We delve into the processes at the core of antimetastatic impacts of microtubule-targeting agents, both through direct modulation of microtubules and via alternative pathways. Drawing from in vitro and in vivo studies, we analyze the cytotoxic and antimetastatic doses of various compounds, shedding light on their therapeutic potential. Furthermore, we discuss the emerging class of microtubule targeting drugs, and their role in metastasis inhibition, such as microtubules acetylation inhibitory drugs, particularly histone deacetylase inihibitors and antibody-drug conjugates. Histone deacetylase (HDAC) strengthens the microtubule cytoskeleton through acetylation. Recently, HDAC inhibitors have been discovered to have antimetastatic properties. Here, the role of HDAC inhibitors in stopping metastasis is discussed with respect to microtubule cytoskeleton. Surprisingly, novel antibody conjugates of microtubule-targeting agents, which are in clinical trials, were found to be antimetastatic. This review discusses these antibody conjugates in detail. Additionally, we elucidate the intricate crosstalk between microtubules and other cytoskeletal proteins, unveiling novel therapeutic strategies for metastasis suppression. By providing a wide-ranging overview of the complex interplay between microtubules and cancer metastasis, this review contributes to the comprehension of cancer's biological mechanisms and the development of innovative therapeutic interventions to mitigate metastatic progression.
Collapse
Affiliation(s)
- Snehal Mangaonkar
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India
| | - Sangeeta Nath
- Manipal Institute of Regenerative Medicine, Bangalore, Manipal Academy of Higher Education, Manipal, India.
| | - Biswa Prasun Chatterji
- Faculty of Science, Assam Downtown University, Guwahati, India; Global Visiitng Professor, Asian University for Women, Chittagong, Bangladesh.
| |
Collapse
|
10
|
Dissanayake DS, Nagahawatta DP, Lee JS, Jeon YJ. Immunomodulatory Effects of Halichondrin Isolated from Marine Sponges and Its Synthetic Analogs in Oncological Applications. Mar Drugs 2024; 22:426. [PMID: 39330307 PMCID: PMC11432918 DOI: 10.3390/md22090426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024] Open
Abstract
Marine natural products comprise unique chemical structures and vast varieties of biological activities. This review aims to summarize halichondrin, a marine natural product, and its synthetic analogs along with its therapeutic properties and mechanisms. Halichondrin and its analogs, derived from marine sponges, exhibit potent antineoplastic properties, making them promising candidates for cancer therapeutics. These compounds, characterized by their complex molecular structures, have demonstrated significant efficacy in inhibiting microtubule dynamics, leading to cell cycle arrest and apoptosis in various cancer cell lines. Several types of halichondrins such as halichondrins B, C, norhalichondrin B, and homohalichondrin B have been discovered with similar anticancer and antitumor characteristics. Since naturally available halichondrins show hurdles in synthesis, recent advancements in synthetic methodologies have enabled the development of several halichondrin analogs, such as E7389 (eribulin), which have shown improved therapeutic indices. Eribulin has shown excellent immunomodulatory properties by several mechanisms such as reprogramming tumor microenvironments, facilitating the infiltration and activation of immune cells, and inhibiting microtubule dynamics. Despite promising results, challenges remain in the synthesis and clinical application of these compounds. This review explores the mechanisms underlying the immunomodulatory activity of halichondrin and its analogs in cancer therapy, along with their clinical applications and potential for future drug development.
Collapse
Affiliation(s)
- Dinusha Shiromala Dissanayake
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea; (D.S.D.); (D.P.N.)
| | - Dineth Pramuditha Nagahawatta
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea; (D.S.D.); (D.P.N.)
| | - Jung-Suck Lee
- Department of Seafood Science and Technology, Institute of Marine Industry, Gyeongsang National University, Tongyeong 53064, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Republic of Korea; (D.S.D.); (D.P.N.)
| |
Collapse
|
11
|
Zeng S, Chen L, Tian J, Liu Z, Liu X, Tang H, Wu H, Liu C. Integrative analysis of pan-cancer single-cell data reveals a tumor ecosystem subtype predicting immunotherapy response. NPJ Precis Oncol 2024; 8:205. [PMID: 39277681 PMCID: PMC11401940 DOI: 10.1038/s41698-024-00703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024] Open
Abstract
Tumor ecosystem shapes cancer biology and potentially influence the response to immunotherapy, but there is a lack of direct clinical evidence. In this study, we utilized EcoTyper and publicly available scRNA-Seq cohorts from ICI-treated patients. We found a ecosystem subtype (ecotype) was linked to improved responses to immunotherapy. Then, a novel immunotherapy-responsive ecotype signature (IRE.Sig) was established and validated through the analysis of pan-cancer data. Utilizing IRE.Sig, machine learning models successfully predicted ICI responses in both validation and testing cohorts, achieving area under the curve (AUC) values of 0.72 and 0.71, respectively. Furthermore, using 5 CRISPR screening cohorts, we identified several potential drugs that may augment the efficacy of ICI. We also elucidated the candidate cellular biomarkers of response to the combined treatment of pembrolizumab plus eribulin in breast cancer. This signature has the potential to serve as a valuable tool for patients in selecting appropriate immunotherapy treatments.
Collapse
Affiliation(s)
- Shengjie Zeng
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Liuxun Chen
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinyu Tian
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengxin Liu
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xudong Liu
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haibin Tang
- Department of Urology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Wu
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Chuan Liu
- Department of Urology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
12
|
Tanriverdi G, Kaleci B, Yavuz F, Sahin H, Purelku M, Yazici Z, Kokturk S. The effects of the combination of temozolomide and Eribulin on T98G human glioblastoma cell line: an ultrastructural study. Ultrastruct Pathol 2024; 48:323-337. [PMID: 38916264 DOI: 10.1080/01913123.2024.2371821] [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: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
Glioblastoma tumors are the most aggressive primary brain tumors that develop resistance to temozolomide (TMZ). Eribulin (ERB) exhibits a unique mechanism of action by inhibiting microtubule dynamics during the G2/M cell cycle phase. We utilized the T98G human glioma cell line to investigate the effects of ERB and TMZ, both individually and in combination. The experimental groups were established as follows: control, E5 (5 nM ERB), T0.75 (0.75 mM TMZ), T1 (1.0 mM TMZ), and combination groups (E5+T0.75 and E5+T1). All groups showed a significant decrease in cell proliferation. Apoptotic markers revealed a time-dependent increase in annexin-V expression, across all treatment groups at the 48-hour time point. Caspase-3, exhibited an increase in the combination treatment groups at the 48-hour mark. Transmission electron microscopy (TEM) revealed normal ultrastructural features in the glioma cells of the control group. However, treatments induced ultrastructural changes within the spheroid glioblastoma model, particularly in the combination groups. These changes included a dose-dependent increase in autophagic vacuoles and apoptotic morphology of the cells. In conclusion, the similarity in the mechanism of action between ERB and TMZ suggests the potential for synergistic effects when combined. Our results highlight that this combination induced severe damage and autophagy in glioma spheroids after 48 hours.
Collapse
Affiliation(s)
- Gamze Tanriverdi
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Belisa Kaleci
- Ministry of Health and Social Protection, University Dental Clinic, Tirane, Albania
| | - Furkan Yavuz
- Radiation Oncology Department, School of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Hakan Sahin
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Merjem Purelku
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Zeliha Yazici
- Medical Pharmacology, Medicine, Istanbul Arel University, İstanbul, Türkiye
| | - Sibel Kokturk
- Department of Histology and Embryology, Medicine Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
13
|
Li S, Zhang H, Yu H, Wu Y, Yan L, Guan X, Dong B, Zhao M, Tian X, Hao C, Wu J. Combination of eribulin and anlotinib exerts synergistic cytotoxicity in retroperitoneal liposarcoma by inducing endoplasmic reticulum stress. Cell Death Discov 2024; 10:355. [PMID: 39117615 PMCID: PMC11310505 DOI: 10.1038/s41420-024-02103-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Primary retroperitoneal liposarcoma (RLPS) is a rare heterogeneous tumor occurring within retroperitoneal space, and its overall survival has not improved much in the past few decades. Based on a small-sample clinical practice at our center, patients with RLPS can greatly benefit from anlotinib and eribulin combination. In this study, we investigated the combinational effect of anlotinib and eribulin on RLPS. In vitro experiments revealed that a low dose of anlotinib significantly enhances the cytotoxic effects of eribulin, leading to a remarkable suppression of RLPS cell proliferation, viability, colony formation, migration, and cell-cycle progression compared to individual drug treatments. At the organoid level, the combination treatment causes the spheroids in Matrigel to disintegrate earlier than the single-drug group. In vivo, RLPS patient-derived xenograft (PDX) models demonstrated that the combination of these two drugs can obviously exert a safe and effective anti-tumor effect. Through transcriptome analysis, we uncovered and validated that the synergistic effect mainly is induced by the endoplasmic reticulum stress (ERS) pathway both in vitro and in vivo. Further analyses indicate that anlotinib plus eribulin treatment results in micro-vessel density and PD-L1 expression alterations, suggesting a potential impact on the tumor microenvironment. This study extensively explored the combination regimen at multiple levels and its underlying molecular mechanism in RLPS, thus providing a foundation for translational medicine research.
Collapse
Affiliation(s)
- Shuquan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongtao Zhang
- Guowen (Changchun) International Hospital, Changchun, Jilin Province, China
| | - Hao Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yifan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Liang Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoya Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiuyun Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jianhui Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
14
|
Khan MS, Wong GL, Zhuang C, Najjar MK, Lo HW. Crosstalk between breast cancer-derived microRNAs and brain microenvironmental cells in breast cancer brain metastasis. Front Oncol 2024; 14:1436942. [PMID: 39175471 PMCID: PMC11338853 DOI: 10.3389/fonc.2024.1436942] [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: 05/22/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024] Open
Abstract
Breast cancer is the most frequent malignancy in women, constituting 15.2% of all new cancers diagnosed in the United States. Distant breast cancer metastasis accounts for the majority of breast cancer-related deaths; brain metastasis is the third most common site for metastatic breast cancer but is associated with worst prognosis of approximately eight months of survival. Current treatment options for breast cancer brain metastasis (BCBM) are limited and ineffective. To help identify new and effective therapies for BCBM, it is important to investigate the mechanisms by which breast cancer cells metastasize to the brain and thrive in the brain microenvironment. To this end, studies have reported that primary breast tumor cells can prime brain microenvironmental cells, including, astrocytes and microglia, to promote the formation of BCBM through the release of extracellular vesicle-microRNAs (miRNAs). Breast tumor-derived miRNAs can also promote breast cancer cell invasion through the blood-brain barrier by disrupting the integrity of the brain microvascular endothelial cells. In this review, we summarize current literature on breast cancer-derived BCBM-promoting miRNAs, cover their roles in the complex steps of BCBM particularly their interactions with microenvironmental cells within the brain metastatic niche, and finally discuss their therapeutic applications in the management of BCBM.
Collapse
Affiliation(s)
- Munazza S. Khan
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Grace L. Wong
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Chuling Zhuang
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mariana K. Najjar
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Hui-Wen Lo
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
15
|
Allsopp RC, Guo Q, Page K, Bhagani S, Kasim A, Badman P, Kenny L, Stebbing J, Shaw JA. Circulating tumour DNA dynamics during alternating chemotherapy and hormonal therapy in metastatic breast cancer: the ALERT study. Breast Cancer Res Treat 2024; 206:377-385. [PMID: 38581534 PMCID: PMC11182849 DOI: 10.1007/s10549-024-07316-8] [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: 03/01/2024] [Accepted: 03/22/2024] [Indexed: 04/08/2024]
Abstract
PURPOSE Although changes in circulating tumour DNA (ctDNA) in breast cancer are well described, the kinetics of their fluctuations has not been described over short timescales. We investigated ctDNA dynamics during alternating cycles of chemotherapy and hormonal treatment in pre-treated patients with oestrogen receptor-positive metastatic breast cancer. METHODS Patients received alternating, 9-week cycles of eribulin and aromatase inhibitors (AIs). The clinical primary endpoint, progression-free survival (PFS), was monitored at 3, 6 and 9 months; secondary endpoints, clinical benefit rate (CBR), safety and tolerability profiles, were also assessed. Importantly, ctDNA fluctuations were monitored using the Oncomine™ Breast cfDNA assay to test whether biomarkers may change rapidly between chemotherapy and aromatase inhibitor (AI) treatment in the setting of advanced breast cancer, potentially reflecting disease dynamics. RESULTS The median PFS was 202 days (95% CI: 135-undefined) and 235 days (95% CI: 235-undefined) at 6 and 9 months, respectively, with a 50% CBR at both 6 and 9 months. Dynamic changes in ctDNA were observed in short timescales between chemotherapy and AI treatment and support the clinical benefit (CB) seen in individual patients and, critically, appear informative of acquired resistance in real time. CONCLUSION Changes in ctDNA can occur rapidly and reflect changes in patients' clinical tumour responses (NCT02681523).
Collapse
Affiliation(s)
- Rebecca C Allsopp
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester Royal Infirmary, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Qi Guo
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester Royal Infirmary, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Karen Page
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester Royal Infirmary, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Shradha Bhagani
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester Royal Infirmary, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX, UK
| | - Anna Kasim
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
| | - Philip Badman
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
| | - Laura Kenny
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
- Department of Life Sciences, Anglia Ruskin University, East Road, Cambridge, CB1 1PT, UK.
| | - Jacqueline A Shaw
- Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Leicester Royal Infirmary, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX, UK
| |
Collapse
|
16
|
Wang Y, Xia B, Cao L, Yang J, Feng C, Jiang F, Li C, Gu L, Yang Y, Tian J, Cheng X, Furuuchi K, Fulmer J, Verdi A, Rybinski K, Soto A, Albone E, Uenaka T, Gong L, Liu T, Qin Q, Wei Z, Zhou Y. Preclinical studies of BB-1701, a HER2-targeting eribulin-containing ADC with potent bystander effect and ICD activity. Antib Ther 2024; 7:221-232. [PMID: 39036069 PMCID: PMC11259758 DOI: 10.1093/abt/tbae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Several HER2-targeting antibody-drug conjugates (ADC) have gained market approval for the treatment of HER2-expressing metastasis. Promising responses have been reported with the new generation of ADCs in patients who do not respond well to other HER2-targeting therapeutics. However, these ADCs still face challenges of resistance and/or severe adverse effects associated with their particular payload toxins. Eribulin, a therapeutic agent for the treatment of metastatic breast cancer and liposarcoma, is a new choice of ADC payload with a distinct mechanism of action and safety profile. METHODS We've generated a novel HER2-tageting eribulin-containing ADC, BB-1701. The potency of BB-1701 was tested in vitro and in vivo against cancer cells where HER2-expressing levels vary in a large range. Bystander killing effect and toxin-induced immunogenic cell death (ICD) of BB-1701 were also tested. RESULTS In comparison with HER2-targeting ADCs with DM1 and Dxd payload, eribulin-containing ADC demonstrated higher in vitro cytotoxicity in HER2-low cancer cell lines. BB-1701 also effectively suppressed tumors in models resistant to DM1 or Dxd containing ADCs. Mode of action studies showed that BB-1701 had a significant bystander effect on HER2-null cells adjacent to HER2-high cells. In addition, BB-1701 treatment induced ICD. Repeated doses of BB-1701 in nonhuman primates showed favorable pharmacokinetics and safety profiles at the intended clinical dosage, route of administration, and schedule. CONCLUSIONS The preclinical data support the test of BB-1701 in patients with various HER2-expressing cancers, including those resistant to other HER2-targeting ADCs. A phase I clinical trial of BB-1701 (NCT04257110) in patients is currently underway.
Collapse
Affiliation(s)
- Yang Wang
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Bing Xia
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Lixia Cao
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Jianfeng Yang
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Cui Feng
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Fangdun Jiang
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Chen Li
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Lixia Gu
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Yifan Yang
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Jing Tian
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Xin Cheng
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Keiji Furuuchi
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - James Fulmer
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Arielle Verdi
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Katherine Rybinski
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Allis Soto
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Earl Albone
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Toshimitsu Uenaka
- Epochal Precision Anti-Cancer Therapeutics (EPAT), Cell Lineage and Differentiation (CLD) Domain, Eisai Inc., Exton, PA 19341, United States
| | - Likun Gong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tingting Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qiuping Qin
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ziping Wei
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| | - Yuhong Zhou
- Bliss Biopharmaceutical (Hangzhou) Co., Ltd, Hexiang Technology Center, Hangzhou 310018, China
| |
Collapse
|
17
|
Oey O, Wijaya W, Redfern A. Eribulin in breast cancer: Current insights and therapeutic perspectives. World J Exp Med 2024; 14:92558. [PMID: 38948420 PMCID: PMC11212747 DOI: 10.5493/wjem.v14.i2.92558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/21/2024] [Accepted: 03/20/2024] [Indexed: 06/19/2024] Open
Abstract
Eribulin is a non-taxane synthetic analogue approved in many countries as third-line treatment for the treatment of patients with metastatic breast cancer. In addition to its mitotic property, eribulin has non-mitotic properties including but not limited to, its ability to induce phenotypic reversal of epithelial to mesenchymal transition, vascular remodelling, reduction in immunosuppressive tumour microenvironment. Since approval, there has been a surge in studies investigating the application of eribulin as an earlier-line treatment and also in combination with other agents such as immunotherapy and targeted therapy across all breast cancer sub-types, including hormone receptor positive, HER2 positive and triple negative breast cancer, many demonstrating promising activity. This review will focus on the application of eribulin in the treatment of metastatic breast cancer across all subtypes including its role as an earlier-line agent, its toxicity profile, and potential future directions.
Collapse
Affiliation(s)
- Oliver Oey
- Faculty of Medicine, University of Western Australia, Nedlands 6009, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands 6009, WA, Australia
| | - Wynne Wijaya
- Department of Oncology, University of Oxford, Oxford OX3 7DQ, United Kingdom
- Department of Internal Medicine, Universitas Gadjah Mada, Sleman 55281, Indonesia
| | - Andrew Redfern
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch 6150, WA, Australia
| |
Collapse
|
18
|
Guo Z, Luo J, Mashl RJ, Hoog J, Maiti P, Fettig N, Davies SR, Aft R, Held JM, Govindan R, Ding L, Li S, von Morze C, Wulf GM, Shoghi KI, Ma CX. Evaluation of Copanlisib in Combination with Eribulin in Triple-negative Breast Cancer Patient-derived Xenograft Models. CANCER RESEARCH COMMUNICATIONS 2024; 4:1430-1440. [PMID: 38717161 PMCID: PMC11152037 DOI: 10.1158/2767-9764.crc-24-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
The PI3K pathway regulates essential cellular functions and promotes chemotherapy resistance. Activation of PI3K pathway signaling is commonly observed in triple-negative breast cancer (TNBC). However previous studies that combined PI3K pathway inhibitors with taxane regimens have yielded inconsistent results. We therefore set out to examine whether the combination of copanlisib, a clinical grade pan-PI3K inhibitor, and eribulin, an antimitotic chemotherapy approved for taxane-resistant metastatic breast cancer, improves the antitumor effect in TNBC. A panel of eight TNBC patient-derived xenograft (PDX) models was tested for tumor growth response to copanlisib and eribulin, alone or in combination. Treatment-induced signaling changes were examined by reverse phase protein array, immunohistochemistry (IHC) and 18F-fluorodeoxyglucose PET (18F-FDG PET). Compared with each drug alone, the combination of eribulin and copanlisib led to enhanced tumor growth inhibition, which was observed in both eribulin-sensitive and -resistant TNBC PDX models, regardless of PI3K pathway alterations or PTEN status. Copanlisib reduced PI3K signaling and enhanced eribulin-induced mitotic arrest. The combination enhanced induction of apoptosis compared with each drug alone. Interestingly, eribulin upregulated PI3K pathway signaling in PDX tumors, as demonstrated by increased tracer uptake by 18F-FDG PET scan and AKT phosphorylation by IHC. These changes were inhibited by the addition of copanlisib. These data support further clinical development for the combination of copanlisib and eribulin and led to a phase I/II trial of copanlisib and eribulin in patients with metastatic TNBC. SIGNIFICANCE In this research, we demonstrated that the pan-PI3K inhibitor copanlisib enhanced the cytotoxicity of eribulin in a panel of TNBC PDX models. The improved tumor growth inhibition was irrespective of PI3K pathway alteration and was corroborated by the enhanced mitotic arrest and apoptotic induction observed in PDX tumors after combination therapy compared with each drug alone. These data provide the preclinical rationale for the clinical testing in TNBC.
Collapse
Affiliation(s)
- Zhanfang Guo
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jingqin Luo
- Division of Public Health Science, Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, Missouri
| | - R. Jay Mashl
- Department of Medicine, McDonnell Genome Institute, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Jeremy Hoog
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Piyush Maiti
- Mallinckrodt Institute of Radiology, St. Louis, Missouri
| | - Nikki Fettig
- Mallinckrodt Institute of Radiology, St. Louis, Missouri
| | - Sherri R. Davies
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Rebecca Aft
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Jason M. Held
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ramaswamy Govindan
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Li Ding
- Department of Medicine, McDonnell Genome Institute, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Shunqiang Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | - Gerburg M. Wulf
- Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Cynthia X. Ma
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
19
|
Azumi M, Kusama K, Yoshie M, Nakano S, Tsuru A, Kato T, Tamura K. Involvement of ferroptosis in eribulin-induced cytotoxicity in ovarian clear cell carcinoma. Eur J Pharmacol 2024; 971:176544. [PMID: 38552939 DOI: 10.1016/j.ejphar.2024.176544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
Ovarian clear cell carcinoma (OCCC) is a unique clinicopathological subtype of epithelial ovarian cancer that is resistant to standard chemotherapy. Eribulin, a microtubule dynamics inhibitor of halichondrin class, has unique effects in the cancer microenvironment such as induction of epithelization and reduction in metastatic potential in breast cancer cells; however, nothing is known about the effect of eribulin and the detailed mechanisms in OCCC. This study aimed to investigate the involvement of ferroptosis and its mechanism in the antitumor activity of eribulin in OCCC cells and a mouse xenograft model. We found that eribulin-induced cell death was reduced by ferroptosis inhibitors; deferoxamine, an iron chelator and ferrostatin-1, a lipid peroxidation inhibitor. Eribulin increased the levels of intracellular iron, reactive oxygen species (ROS), and lipid peroxides, and increased the mitochondrial membrane potential. Eribulin downregulated the expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH), and superoxide dismutase (SOD) activity. The combination of eribulin and ML210, a glutathione peroxidase 4-inhibiting ferroptosis inducer, had a synergistic effect on ferroptosis. Taken together, our findings show firstly that eribulin triggers ferroptosis in OCCC and this effect occurs via the suppression of the Nrf2-HO-1 signaling pathway, SOD activity and the promotion of lipid peroxidation. These findings suggest that eribulin-induced ferroptosis is associated with its anti-tumor effect and also could be a potential therapeutic target in OCCC.
Collapse
Affiliation(s)
- Mana Azumi
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Mikihiro Yoshie
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Saya Nakano
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Atsuya Tsuru
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan
| | - Tomoyasu Kato
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan; Department of Gynecologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazuhiro Tamura
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo, 192-0392, Japan.
| |
Collapse
|
20
|
Xerez MC, da Silva Barros CC, de Souto Medeiros MR, Mafra RP, de Lucena HF, da Silveira ÉJD, de Lisboa Lopes Costa A. CLIC4 Function in the Epithelial-Mesenchymal Transition of Epithelial Odontogenic Lesions. Head Neck Pathol 2024; 18:40. [PMID: 38727794 PMCID: PMC11087429 DOI: 10.1007/s12105-024-01646-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/04/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Odontogenic lesions constitute a heterogeneous group of lesions. CLIC4 protein regulates different cellular processes, including epithelial-mesenchymal transition and fibroblast-myofibroblast transdifferentiation. This study analyzed CLIC4, E-cadherin, Vimentin, and α-SMA immunoexpression in epithelial odontogenic lesions that exhibit different biological behavior. METHODS It analyzed the immunoexpression of CLIC4, E-cadherin, and Vimentin in the epithelial cells, as well as CLIC4 and α-SMA in the mesenchymal cells, of ameloblastoma (AM) (n = 16), odontogenic keratocyst (OKC) (n = 20), and adenomatoid odontogenic tumor (AOT) (n = 8). Immunoexpressions were categorized as score 0 (0% positive cells), 1 (< 25%), 2 (≥ 25% - < 50%), 3 (≥ 50% - < 75%), or 4 (≥ 75%). RESULTS Cytoplasmic CLIC4 immunoexpression was higher in AM and AOT (p < 0.001) epithelial cells. Nuclear-cytoplasmic CLIC4 was higher in OKC's epithelial lining (p < 0.001). Membrane (p = 0.012) and membrane-cytoplasmic (p < 0.001) E-cadherin immunoexpression were higher in OKC, while cytoplasmic E-cadherin expression was higher in AM and AOT (p < 0.001). Vimentin immunoexpression was higher in AM and AOT (p < 0.001). Stromal CLIC4 was higher in AM and OKC (p = 0.008). Similarly, α-SMA immunoexpression was higher in AM and OKC (p = 0.037). Correlations in these proteins' immunoexpression were observed in AM and OKC (p < 0.05). CONCLUSIONS CLIC4 seems to regulate the epithelial-mesenchymal transition, modifying E-cadherin and Vimentin expression. In mesenchymal cells, CLIC4 may play a role in fibroblast-myofibroblast transdifferentiation. CLIC4 may be associated with epithelial odontogenic lesions with aggressive biological behavior.
Collapse
Affiliation(s)
- Mariana Carvalho Xerez
- Oral Pathology and Medicine, Postgraduate Program in Dental Sciences, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil.
| | - Caio César da Silva Barros
- Oral Pathology and Medicine, Postgraduate Program in Dental Sciences, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Maurília Raquel de Souto Medeiros
- Oral Pathology and Medicine, Postgraduate Program in Dental Sciences, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Rodrigo Porpino Mafra
- Oral Pathology and Medicine, Postgraduate Program in Dental Sciences, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Hévio Freitas de Lucena
- Department of Dentistry, Federal University of Rio Grande do Norte, Av. Salgado Filho, 1787, Lagoa Nova, Natal, CEP: 59056-000, RN, Brazil
| | - Éricka Janine Dantas da Silveira
- Oral Pathology and Medicine, Postgraduate Program in Dental Sciences, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
- Department of Dentistry, Federal University of Rio Grande do Norte, Av. Salgado Filho, 1787, Lagoa Nova, Natal, CEP: 59056-000, RN, Brazil
| | - Antonio de Lisboa Lopes Costa
- Oral Pathology and Medicine, Postgraduate Program in Dental Sciences, Department of Dentistry, Federal University of Rio Grande do Norte, Natal, RN, Brazil
- Department of Dentistry, Federal University of Rio Grande do Norte, Av. Salgado Filho, 1787, Lagoa Nova, Natal, CEP: 59056-000, RN, Brazil
| |
Collapse
|
21
|
Nicolò E, Gianni C, Pontolillo L, Serafini MS, Munoz-Arcos LS, Andreopoulou E, Curigliano G, Reduzzi C, Cristofanilli M. Circulating tumor cells et al.: towards a comprehensive liquid biopsy approach in breast cancer. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 5:10. [PMID: 38751670 PMCID: PMC11093063 DOI: 10.21037/tbcr-23-55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/21/2024] [Indexed: 05/18/2024]
Abstract
Liquid biopsy has emerged as a crucial tool in managing breast cancer (BC) patients, offering a minimally invasive approach to detect circulating tumor biomarkers. Until recently, the majority of the studies in BC focused on evaluating a single liquid biopsy analyte, primarily circulating tumor DNA and circulating tumor cells (CTCs). Despite the proven prognostic and predictive value of CTCs, their low abundance when detected using enrichment methods, especially in the early stages, poses a significant challenge. It is becoming evident that combining diverse circulating biomarkers, each representing different facets of tumor biology, has the potential to enhance the management of patients with BC. This article emphasizes the importance of considering these biomarkers as complementary/synergistic rather than competitive, recognizing their ability to contribute to a comprehensive disease profile. The review provides an overview of the clinical significance of simultaneously analyzing CTCs and other biomarkers, including cell-free circulating DNA, extracellular vesicles, non-canonical CTCs, cell-free RNAs, and non-malignant cells. Such a comprehensive liquid biopsy approach holds promise not only in BC but also in other cancer types, offering opportunities for early detection, prognostication, and therapy monitoring. However, addressing associated challenges, such as refining detection methods and establishing standardized protocols, is crucial for realizing the full potential of liquid biopsy in transforming our understanding and approach to BC. As the field evolves, collaborative efforts will be instrumental in unlocking the revolutionary impact of liquid biopsy in BC research and management.
Collapse
Affiliation(s)
- Eleonora Nicolò
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - Caterina Gianni
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Letizia Pontolillo
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
- Medical Oncology Department, Catholic University of Sacred Heart, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Mara Serena Serafini
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Laura Sofia Munoz-Arcos
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Eleni Andreopoulou
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Giuseppe Curigliano
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
- Division of Early Drug Development, European Institute of Oncology IRCCS, Milan, Italy
| | - Carolina Reduzzi
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
22
|
Bagheri M, Mohamed GA, Mohamed Saleem MA, Ognjenovic NB, Lu H, Kolling FW, Wilkins OM, Das S, LaCroix IS, Nagaraj SH, Muller KE, Gerber SA, Miller TW, Pattabiraman DR. Pharmacological induction of chromatin remodeling drives chemosensitization in triple-negative breast cancer. Cell Rep Med 2024; 5:101504. [PMID: 38593809 PMCID: PMC11031425 DOI: 10.1016/j.xcrm.2024.101504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/11/2023] [Accepted: 03/19/2024] [Indexed: 04/11/2024]
Abstract
Targeted therapies have improved outcomes for certain cancer subtypes, but cytotoxic chemotherapy remains a mainstay for triple-negative breast cancer (TNBC). The epithelial-to-mesenchymal transition (EMT) is a developmental program co-opted by cancer cells that promotes metastasis and chemoresistance. There are no therapeutic strategies specifically targeting mesenchymal-like cancer cells. We report that the US Food and Drug Administration (FDA)-approved chemotherapeutic eribulin induces ZEB1-SWI/SNF-directed chromatin remodeling to reverse EMT that curtails the metastatic propensity of TNBC preclinical models. Eribulin induces mesenchymal-to-epithelial transition (MET) in primary TNBC in patients, but conventional chemotherapy does not. In the treatment-naive setting, but not after acquired resistance to other agents, eribulin sensitizes TNBC cells to subsequent treatment with other chemotherapeutics. These findings provide an epigenetic mechanism of action of eribulin, supporting its use early in the disease process for MET induction to prevent metastatic progression and chemoresistance. These findings warrant prospective clinical evaluation of the chemosensitizing effects of eribulin in the treatment-naive setting.
Collapse
Affiliation(s)
- Meisam Bagheri
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Gadisti Aisha Mohamed
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | - Nevena B Ognjenovic
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Hanxu Lu
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Fred W Kolling
- Center for Quantitative Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Owen M Wilkins
- Center for Quantitative Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | | | - Ian S LaCroix
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Shivashankar H Nagaraj
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; Translational Research Institute, Brisbane, QLD 4102, Australia
| | - Kristen E Muller
- Department of Pathology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Scott A Gerber
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Todd W Miller
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA; Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Diwakar R Pattabiraman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| |
Collapse
|
23
|
Ma Y, Shi X, Zhao K, Hu S, Shi Y, Jiang Y, Liu Y, Lu L, Chang Y, Zhou F, Dai Y, Wu Z, Li S, Qian Z, Xu X, Li C, Shen B, Zhou G, Chen C, Wang X, Feng J. Optimal response to tislelizumab plus chemotherapy in metastatic triple-negative breast cancer: a case report and literature review. Front Oncol 2024; 14:1328844. [PMID: 38606104 PMCID: PMC11007123 DOI: 10.3389/fonc.2024.1328844] [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: 10/30/2023] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
Metastatic triple-negative breast cancer (mTNBC) has the worst prognosis among breast cancer subtypes. Immune checkpoint inhibitors (ICIs) plus chemotherapy have promising survival benefits. Herein, we report a 51-year-old woman whose metastatic lesions were diagnosed as triple-negative subtype and who received tislelizumab plus eribulin treatment and achieved excellent efficacy. To our knowledge, this study is the first attempt to present tislelizumab in combination with eribulin for mTNBC treatment. New treatments resulting in prolonged survival and durable clinical responses would benefit mTNBC patients. Then, we summarize the possible influencing factors of the interaction between tislelizumab and eribulin.
Collapse
Affiliation(s)
- Yuxin Ma
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Xinhong Shi
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Kun Zhao
- Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Shuyi Hu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yue Shi
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yingying Jiang
- Department of Oncology, Province Geriatric Hospital, Nanjing, Jiangsu, China
| | - Yiling Liu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Lin Lu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yuting Chang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Fei Zhou
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Yingying Dai
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Zipeng Wu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Shiyi Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Zhiying Qian
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Xia Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Chenchen Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Bo Shen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
- Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Guoren Zhou
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Cheng Chen
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Xiaohua Wang
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
- Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| | - Jifeng Feng
- Department of Medical Oncology, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
- Huaian Hospital of Huaian City, Huaian Cancer Hospital, Huaian, Jiangsu, China
| |
Collapse
|
24
|
Giulietti M, Piva F, Cecati M, Maggio S, Guescini M, Saladino T, Scortichini L, Crocetti S, Caramanti M, Battelli N, Romagnoli E. Effects of Eribulin on the RNA Content of Extracellular Vesicles Released by Metastatic Breast Cancer Cells. Cells 2024; 13:479. [PMID: 38534323 PMCID: PMC10969587 DOI: 10.3390/cells13060479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Extracellular vesicles (EVs) are small lipid particles secreted by almost all human cells into the extracellular space. They perform the essential function of cell-to-cell communication, and their role in promoting breast cancer progression has been well demonstrated. It is known that EVs released by triple-negative and highly aggressive MDA-MB-231 breast cancer cells treated with paclitaxel, a microtubule-targeting agent (MTA), promoted chemoresistance in EV-recipient cells. Here, we studied the RNA content of EVs produced by the same MDA-MB-231 breast cancer cells treated with another MTA, eribulin mesylate. In particular, we analyzed the expression of different RNA species, including mRNAs, lncRNAs, miRNAs, snoRNAs, piRNAs and tRNA fragments by RNA-seq. Then, we performed differential expression analysis, weighted gene co-expression network analysis (WGCNA), functional enrichment analysis, and miRNA-target identification. Our findings demonstrate the possible involvement of EVs from eribulin-treated cells in the spread of chemoresistance, prompting the design of strategies that selectively target tumor EVs.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Monia Cecati
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Serena Maggio
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Tiziana Saladino
- Oncology Unit AST3, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Laura Scortichini
- Oncology Unit AST3, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Sonia Crocetti
- Oncology Unit AST3, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Miriam Caramanti
- Oncology Unit AST3, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Nicola Battelli
- Oncology Unit AST3, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| | - Emanuela Romagnoli
- Oncology Unit AST3, Macerata Hospital, Via Santa Lucia 2, 62100 Macerata, Italy
| |
Collapse
|
25
|
Su MX, Lin HW, Nguyen HTH, Lin TC, Chen CJ, Wang HC, Wu CT, Wu YC, He GY, Liu LC, Huang CH. Monitoring trends in the absolute lymphocyte count and the neutrophil-to-lymphocyte ratio in patients with breast cancer receiving eribulin. BMC Cancer 2024; 24:195. [PMID: 38347468 PMCID: PMC10860250 DOI: 10.1186/s12885-024-11923-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Studies have shown that the absolute lymphocyte count (ALC) and the neutrophil-to-lymphocyte ratio (NLR) are related to the outcomes in patients with breast cancer receiving specific chemotherapies. However, the reports have focussed on the initial blood test and there is a lack of evidence or data to support that dynamic changes of ALC or NLR are associated with the patients' survival outcomes. METHODS We retrospectively reviewed electronic medical records from patients with breast cancer treated with eribulin from 2015 to 2019 at our institution. Blood test data were available prior to starting eribulin (baseline), and at 1, 3 and 6 months after initiating eribulin. We classified the patients into ALC and NLR high and low groups using the following cut-offs: 1000/µl for ALC and 3 for NLR. We defined ALC and NLR trends as increasing or decreasing compared with the initial data. We assessed the associations between the ALC and NLR with progression-free survival and overall survival. RESULTS There were 136 patients with breast cancer treated with eribulin. Of these patients, 60 had complete blood tests and follow-up data. Neither a high ALC nor a low baseline NLR was associated with the survival outcome. One month after initiating eribulin treatment, a high ALC and a low NLR were significantly associated with longer progression-free survival (p = 0.044 for each). Three months after initiating eribulin, a high ALC was significantly associated with better overall survival (p = 0.006). A high NLR at 3 or 6 months after initiating eribulin was associated with worse overall survival (p = 0.017 and p = 0.001, respectively). The ALC and NLR trends across times were not associated with survivals. CONCLUSION We showed that 1, 3 and 6 months after initiating eribulin, a high ALC and a low NLR may be related to the patients' survival outcomes. The ALC and NLR trends were not associated with survival. Accordingly, we believe patients who maintain a high ALC and a low NLR may have better clinical outcomes after initiating eribulin.
Collapse
Affiliation(s)
- Meng-Xia Su
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan
| | - Hsiang-Wen Lin
- School of Pharmacy and Graduate Institute, China Medical University, No. 100, Sec. 1, Jingmao Rd., Taichung, 406040, Taiwan
- Department of Pharmacy, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung, 404327, Taiwan
- Department of Pharmacy System, Outcomes and Policy, College of Pharmacy, University of Illinois at Chicago, Chicago, 833 S. Wood St., Chicago, 60612, Illinois, United States of America
| | - Hanh T H Nguyen
- School of Pharmacy and Graduate Institute, China Medical University, No. 100, Sec. 1, Jingmao Rd., Taichung, 406040, Taiwan
- Department of Clinical Pharmacy, Hanoi University of Pharmacy, No. 144, Xuan Thuy, Cau giay, Hanoi, Vietnam
| | - Tien-Chao Lin
- School of Pharmacy and Graduate Institute, China Medical University, No. 100, Sec. 1, Jingmao Rd., Taichung, 406040, Taiwan
- Department of Pharmacy, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung, 404327, Taiwan
| | - Chih-Jung Chen
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan
| | - Hwei-Chung Wang
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan
| | - Chen-Teng Wu
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan
| | - Yao-Chung Wu
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan
| | - Geng-Yan He
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan
| | - Liang-Chih Liu
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan.
- College of Medicine, China Medical University, No. 100, Sec. 1, Jingmao Rd., Taichung, 406040, Taiwan.
| | - Chih-Hao Huang
- Surgical Department, China Medical University Hospital, No. 2, Yude Rd. North Dist, Taichung, 404327, Taiwan.
- College of Medicine, China Medical University, No. 100, Sec. 1, Jingmao Rd., Taichung, 406040, Taiwan.
| |
Collapse
|
26
|
Shimizu T, Oba T, Oshi M, Ito KI. Eribulin promotes proliferation of CD8 + T cells and potentiates T cell-mediated anti-tumor activity against triple-negative breast cancer cells. Breast Cancer Res Treat 2024; 203:57-71. [PMID: 37733186 DOI: 10.1007/s10549-023-07111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023]
Abstract
PURPOSE Chemotherapeutic agents exert immunomodulatory effects on triple-negative breast cancer (TNBC) cells and immune cells. Eribulin favorably affects the immunological status of patients with breast cancer. However, the effects of eribulin on the immune cells remain unexplored. The aim of this study was to investigate the effects of eribulin on immune cells. METHODS Peripheral blood mononuclear cells (PBMCs) from healthy donors and mouse splenocytes were stimulated with anti-CD3 and anti-CD28 antibodies. The effects of eribulin and paclitaxel on cell proliferation and differentiation status were analyzed using flow cytometry. RNA sequencing was performed to assess alterations in gene expression in CD8+ T cells following eribulin and paclitaxel treatment. Using TNBC cell lines (MDA-MB-231, Hs578T, and MDA-MB-157), the anti-tumor activity of CD3/CD28-stimulated T cells combined with eribulin or paclitaxel was evaluated. RESULTS Eribulin did not affect CD3/CD28-stimulated PBMCs proliferation. However, eribulin significantly decreased the CD4/CD8 ratio in T cells, indicating that eribulin facilitates CD8+ T cell proliferation. Furthermore, eribulin significantly increased the frequency of less differentiated CD45RA+, CCR7+, and TCF1+ subsets of CD8+ T cells. RNA sequencing revealed that eribulin enhanced the expression of gene sets related to cell proliferation and immune responses. Moreover, eribulin augmented the anti-tumor effects of CD3/CD28-stimulated T cells against TNBC cells. These results were not observed in experiments using paclitaxel. CONCLUSIONS Eribulin promoted CD8+ T cell proliferation, repressed effector T cell differentiation, and harnessed T cell-mediated anti-tumor effects. These mechanisms may be one of the cues that eribulin can improve the immunological status of tumor-bearing hosts.
Collapse
Affiliation(s)
- Tadafumi Shimizu
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-861, Japan
| | - Takaaki Oba
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-861, Japan.
| | - Masanori Oshi
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ken-Ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-861, Japan
| |
Collapse
|
27
|
Nagaoka Y, Oshiro K, Yoshino Y, Matsunaga T, Endo S, Ikari A. Activation of the TGF-β1/EMT signaling pathway by claudin-1 overexpression reduces doxorubicin sensitivity in small cell lung cancer SBC-3 cells. Arch Biochem Biophys 2024; 751:109824. [PMID: 37984759 DOI: 10.1016/j.abb.2023.109824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Small-cell lung cancer (SCLC), which accounts for about 15 % of all lung cancers, progresses more rapidly than other histologic types and is rarely detected at an operable early stage. Therefore, chemotherapy, radiation therapy, or their combination are the primary treatments for this type of lung cancer. However, the tendency to acquire resistance to anticancer drugs is a severe problem. Recently, we found that an intercellular adhesion molecule, claudin (CLDN) 1, known to be involved in the migration and invasion of lung cancer cells, is involved in the acquisition of anticancer drug resistance. In the present study, we investigated the effect of CLDN1 on the anticancer-drug sensitivity of SCLC SBC-3 cells. Since epithelial-mesenchymal transition (EMT), which is involved in cancer cell migration and invasion, is well known for its involvement in anticancer-drug sensitivity via inhibition of apoptosis, we also examined EMT involvement in decreased anticancer-drug sensitivity by CLDN1. Sensitivity to doxorubicin (DOX) in SBC-3 cells was significantly decreased by CLDN1 overexpression. CLDN1 overexpression resulted in increased TGF-β1 levels, enhanced EMT induction, and increased migratory potency of SBC-3 cells. The decreased sensitivity of SBC-3 cells to anticancer drugs upon TGF-β1 treatment suggested that activation of the TGF-β1/EMT signaling pathway by CLDN1 causes the decreased sensitivity to anticancer drugs and increased migratory potency. Furthermore, treatments with antiallergic agents tranilast and zoledronic acid, known EMT inhibitors, significantly mitigated the decreased sensitivity of CLDN1-overexpressing SBC-3 cells to DOX. These results suggest that EMT inhibitors might effectively overcome reduced sensitivity to anticancer drugs in CLDN1-overexpressing SCLC cells.
Collapse
Affiliation(s)
- Yuri Nagaoka
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kotone Oshiro
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu Pharmaceutical University, Gifu 501-1196, Japan.
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| |
Collapse
|
28
|
Tochinai R, Nagashima Y, Sekizawa SI, Kuwahara M. Anti-tumor and cardiotoxic effects of microtubule polymerization inhibitors: The mechanisms and management strategies. J Appl Toxicol 2024; 44:96-106. [PMID: 37496236 DOI: 10.1002/jat.4521] [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: 06/21/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023]
Abstract
Microtubule polymerization inhibitors (MPIs) have long been used as anticancer agents because they inhibit mitosis. Microtubules are thought to play an important role in the migration of tumor cells and the formation of tumor blood vessels, and new MPIs are being developed. Many clinical trials of novel MPIs have been conducted in humans, while some clinical studies in dogs have also been reported. More attempts to apply MPIs not only in humans but also in the veterinary field are expected to be made in the future. Meanwhile, MPIs have a risk of cardiotoxicity. In this paper, we review findings on the pharmacological effects and cardiotoxicity of MPIs, as well as the mechanisms of their cardiotoxicity. Cardiotoxicity of MPIs involves not only the direct effects of MPIs on cardiomyocytes but also their effects on vascular function. For example, hypertension induced by impaired vascular function also contributes to the exacerbation of myocardial damage, and blood pressure control may be useful in reducing cardiotoxicity. By combined administration of MPIs and other anticancer agents, MPI efficacy may be enhanced, thereby potentially allowing to keep MPI dosage low. Measurement of myocardial injury markers in blood and echocardiography may be useful for monitoring cardiotoxicity. In particular, two-dimensional speckle tracking may have high sensitivity for the early detection of MPI-induced cardiac dysfunction. The exploration of the potential of new MPIs while understanding their toxicity and how to deal with them will lead to the further development of cancer chemotherapy.
Collapse
Affiliation(s)
- Ryota Tochinai
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshiyasu Nagashima
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichi Sekizawa
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masayoshi Kuwahara
- Department of Veterinary Pathophysiology and Animal Health, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
Wada A, Hirohashi Y, Kutomi G, Murata K, Iwabuchi S, Mizue Y, Murai A, Kyuno D, Shima H, Minowa T, Sasaki K, Kubo T, Kanaseki T, Tsukahara T, Nakatsugawa M, Hashimoto S, Osanai M, Torigoe T, Takemasa I. Eribulin is an immune potentiator in breast cancer that upregulates human leukocyte antigen class I expression via the induction of NOD-like receptor family CARD domain-containing 5. Cancer Sci 2023; 114:4511-4520. [PMID: 37991442 PMCID: PMC10728009 DOI: 10.1111/cas.15986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/10/2023] [Accepted: 09/18/2023] [Indexed: 11/23/2023] Open
Abstract
Eribulin inhibits microtubule polymerization and improves the overall survival of patients with recurrent metastatic breast cancer. A subgroup analysis revealed a low neutrophil to lymphocyte ratio (NLR) (<3) to be a prognostic factor of eribulin treatment. We thus hypothesized that eribulin might be related to the immune response for breast cancer cells and we analyzed the effects of eribulin on the immune system. Immunohistochemical staining revealed that human leukocyte antigen (HLA) class I expression was increased in clinical samples after eribulin treatment. In vitro assays revealed that eribulin treatment increased HLA class I expression in breast cancer line cells. RNA-sequencing demonstrated that eribulin treatment increased the expression of the NOD-like family CARD domain-containing 5 (NLRC5), a master regulator of HLA class I expression. Eribulin treatment increased the NY-ESO-1-specific T-cell receptor (TCR) transduced T (TCR-T) cell response for New York oesophageal squamous cell carcinoma 1 (NY-ESO-1) overexpressed breast cancer cells. The eribulin and TCR-T combined therapy model revealed that eribulin and immunotherapy using TCR-T cells has a synergistic effect. In summary, eribulin increases the expression of HLA class 1 via HLA class 1 transactivatior NLRC5 and eribulin combination with immunotherapy can be effective for the treatment of breast cancer.
Collapse
Affiliation(s)
- Asaka Wada
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Yoshihiko Hirohashi
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Goro Kutomi
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Kenji Murata
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Sadahiro Iwabuchi
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Yuka Mizue
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Aiko Murai
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Daisuke Kyuno
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Hiroaki Shima
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Tomoyuki Minowa
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Kenta Sasaki
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Terufumi Kubo
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Takayuki Kanaseki
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Tomohide Tsukahara
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Munehide Nakatsugawa
- Department of PathologyTokyo Medical University Hachioji Medical CenterTokyoJapan
| | - Shinichi Hashimoto
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Makoto Osanai
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Toshihiko Torigoe
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Ichiro Takemasa
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| |
Collapse
|
30
|
Ho GY, Vandenberg CJ, Lim R, Christie EL, Garsed DW, Lieschke E, Nesic K, Kondrashova O, Ratnayake G, Radke M, Penington JS, Carmagnac A, Heong V, Kyran EL, Zhang F, Traficante N, Huang R, Dobrovic A, Swisher EM, McNally O, Kee D, Wakefield MJ, Papenfuss AT, Bowtell DDL, Barker HE, Scott CL. The microtubule inhibitor eribulin demonstrates efficacy in platinum-resistant and refractory high-grade serous ovarian cancer patient-derived xenograft models. Ther Adv Med Oncol 2023; 15:17588359231208674. [PMID: 38028140 PMCID: PMC10666702 DOI: 10.1177/17588359231208674] [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: 07/01/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Despite initial response to platinum-based chemotherapy and PARP inhibitor therapy (PARPi), nearly all recurrent high-grade serous ovarian cancer (HGSC) will acquire lethal drug resistance; indeed, ~15% of individuals have de novo platinum-refractory disease. Objectives To determine the potential of anti-microtubule agent (AMA) therapy (paclitaxel, vinorelbine and eribulin) in platinum-resistant or refractory (PRR) HGSC by assessing response in patient-derived xenograft (PDX) models of HGSC. Design and methods Of 13 PRR HGSC PDX, six were primary PRR, derived from chemotherapy-naïve samples (one was BRCA2 mutant) and seven were from samples obtained following chemotherapy treatment in the clinic (five were mutant for either BRCA1 or BRCA2 (BRCA1/2), four with prior PARPi exposure), recapitulating the population of individuals with aggressive treatment-resistant HGSC in the clinic. Molecular analyses and in vivo treatment studies were undertaken. Results Seven out of thirteen PRR PDX (54%) were sensitive to treatment with the AMA, eribulin (time to progressive disease (PD) ⩾100 days from the start of treatment) and 11 out of 13 PDX (85%) derived significant benefit from eribulin [time to harvest (TTH) for each PDX with p < 0.002]. In 5 out of 10 platinum-refractory HGSC PDX (50%) and one out of three platinum-resistant PDX (33%), eribulin was more efficacious than was cisplatin, with longer time to PD and significantly extended TTH (each PDX p < 0.02). Furthermore, four of these models were extremely sensitive to all three AMA tested, maintaining response until the end of the experiment (120d post-treatment start). Despite harbouring secondary BRCA2 mutations, two BRCA2-mutant PDX models derived from heavily pre-treated individuals were sensitive to AMA. PRR HGSC PDX models showing greater sensitivity to AMA had high proliferative indices and oncogene expression. Two PDX models, both with prior chemotherapy and/or PARPi exposure, were refractory to all AMA, one of which harboured the SLC25A40-ABCB1 fusion, known to upregulate drug efflux via MDR1. Conclusion The efficacy observed for eribulin in PRR HGSC PDX was similar to that observed for paclitaxel, which transformed ovarian cancer clinical practice. Eribulin is therefore worthy of further consideration in clinical trials, particularly in ovarian carcinoma with early failure of carboplatin/paclitaxel chemotherapy.
Collapse
Affiliation(s)
- Gwo Yaw Ho
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- The Royal Women’s Hospital, Parkville, VIC, Australia
- School of Clinical Sciences, Monash University, Clayton Road, Clayton, VIC 3168, Australia
| | - Cassandra J. Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Ratana Lim
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Elizabeth L. Christie
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Dale W. Garsed
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth Lieschke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Ksenija Nesic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Olga Kondrashova
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | | | - Marc Radke
- University of Washington, Seattle, WA, USA
| | - Jocelyn S. Penington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Amandine Carmagnac
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Valerie Heong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Elizabeth L. Kyran
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Fan Zhang
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - Nadia Traficante
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | | | | | - Alexander Dobrovic
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | | | - Orla McNally
- The Royal Women’s Hospital, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| | - Damien Kee
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Medical Oncology, Austin Hospital, Heidelberg, VIC, Australia
| | - Matthew J. Wakefield
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| | - Anthony T. Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - David D. L. Bowtell
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Holly E. Barker
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- The Royal Women’s Hospital, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
31
|
Tian Z, Rao Q, He Z, Zhao W, Chen L, Liu J, Wang Y. Effect of 1 H-NMR serum lipoproteins on immunotherapy response in advanced triple-negative breast cancer patients. Cancer Sci 2023; 114:3924-3934. [PMID: 37640025 PMCID: PMC10551590 DOI: 10.1111/cas.15937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023] Open
Abstract
We previously reported the results of a phase II trial of anti-PD-1 antibody plus anti-vascular endothelial growth factor receptor 2 inhibitors and eribulin in heavily pretreated advanced triple-negative breast cancer with a favorable objective response rate (ORR) of 37.0% (NCT04303741). Here we report updated survival outcomes and serum metabolite changes of the study. Proton nuclear magnetic resonance spectroscopy was used to detect metabolite dynamics and explore biomarkers for response. We found that treatment-sensitive patients had higher very low-density lipoprotein-related metabolite expression at baseline. A lipid proteomics model consisting of six metabolites predicted ORR and progression-free survival at 6 months with area under the receiver operating characteristic curves of 0.88 and 0.87, respectively. Serum asparagine and sarcosine concentrations were significantly higher after treatment in treatment-resistant patients. In conclusion, we constructed a model consisting of six metabolites to identify patients who benefit more from the triplet treatment, and asparagine and sarcosine may be associated with treatment resistance.
Collapse
Affiliation(s)
- Zhenluan Tian
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Qunxian Rao
- Department of Gynaecological Oncology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhanghai He
- Department of Pathology, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Wei Zhao
- Tianjin Key Laboratory of Clinical MultiomicsTianjinChina
| | - Liangyu Chen
- Tianjin Key Laboratory of Clinical MultiomicsTianjinChina
| | - Jieqiong Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ying Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Breast Tumor Center, Sun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
32
|
Xu Y, Hou YY, Wu Z, Fang ZX, Wu HT, Liu J. Comprehensive analysis of cell-extracellular matrix protein Ras suppressor-1 in function and prognosis of gastrointestinal cancers. World J Methodol 2023; 13:223-237. [PMID: 37771863 PMCID: PMC10523239 DOI: 10.5662/wjm.v13.i4.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/06/2023] [Accepted: 06/19/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Ras suppressor 1 (RSU1), a highly conserved protein, plays an important role in actin cytoskeleton remodeling and cell-extracellular matrix adhesion. Aberration of RSU1 activity can cause changes in cell adhesion and migration, thereby enhancing tumor proliferation and metastasis. However, the correlation between RSU1 and gastrointestinal cancers (GICs), as well as its prognostic role related to tumor-infiltrating immune cells (TIICs) remains unclear. AIM To shows RSU1 plays a potential promoting role in facilitating tumor immune escape in GIC. METHODS Differential expression of RSU1 in different tumors and their corresponding normal tissues was evaluated by exploring the Gene Expression Profiling Interactive Analysis (GEPIA) dataset. The correlation between RSU1 expression and prognosis of GIC cancer patients was evaluated by Kaplan-Meier plotter. Then, RSU1-correlated genes were screened and functionally characterized via enrichment analysis. The correlation between RSU1 and TIICs was further characterized using the Tumor Immune Estimation Resource (TIMER). In addition, the correlation between RSU1 and immune cell surface molecules was also analyzed by TIMER. RESULTS High RSU1 expression was associated with poor overall survival of gastric cancer patients, exhibiting a hazard ratio (HR) = 1.36, first progression HR = 1.53, and post progression survival HR = 1.6. Specifically, high RSU1 Levels were associated with prognosis of gastric cancer in females, T4 and N3 stages, and Her-2-negative subtypes. Regarding immune-infiltrating cells, RSU1 expression level was positively correlated with infiltration of CD4+ T cells, macrophages, neutrophils, and dendritic cells (DCs) in colorectal adenocarcinoma and stomach adenocarcinoma. RSU1 expression was also predicted to be strongly correlated with immune marker sets in M2 macrophage, DCs and T cell exhaustion in GICs. CONCLUSION In gastrointestinal cancers, RSU1 is increased in tumor tissues, and predicts poor survival of patients. Increased RSU1 may be involved in promoting macrophage polarization, DC infiltration, and T cell exhaustion, inducing tumor immune escape and the development of tumors in GICs. We suggest that RSU1 is a promising prognostic biomarker reflecting immune infiltration level of GICs, as well as a potential therapeutic target for precision treatment through improving the immune response.
Collapse
Affiliation(s)
- Ya Xu
- Department of Radiation Oncology, Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei 516600, Guangdong Province, China
| | - Yan-Yu Hou
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Zheng Wu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Ze-Xuan Fang
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Hua-Tao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jing Liu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
33
|
Takeyama H, Manome Y. Breast Cancers Secreting Sialyl-fibronectin Are Less Likely to Cause Epithelial-mesenchymal Transition and Have Good Prognoses. CANCER DIAGNOSIS & PROGNOSIS 2023; 3:558-570. [PMID: 37671303 PMCID: PMC10475922 DOI: 10.21873/cdp.10255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023]
Abstract
Background/Aim Elevated blood fibronectin (FN) levels have been observed in various cancers; however, their significance is controversial. We measured sialyl-fibronectin (S-FN), a type of FN secreted by tumor cells in the blood, and investigated whether blood S-FN secretion is associated with cancer malignancy and recurrent metastases. Materials and Methods We constructed an enzyme-linked immunosorbent assay (ELISA) system that recognizes S-FN as an antigen and measured the amount of S-FN secreted into the blood of 89 patients with breast tumors. The relationship between S-FN secretion and prognostic predictors was analyzed. Immunostaining was performed to identify the site of S-FN secretion in the breast tissue. Results Among the 82 patients, 21 (25.6%, 21/82) and 61 (74.4%, 61/82) were blood S-FN-positive and S-FN-negative, respectively. Regarding prognostic predictors, blood S-FN-positive and S-FN-negative patients showed significant difference in locoregional recurrence (p=0.026), remote metastases (p=0.049), and histological margins (p=0.001). Locoregional recurrence was associated with positive histological margins in S-FN-positive patients. However, remote metastases were associated with N-factor and histological classification (HC) in S-FN-negative patients. Furthermore, S-FN particles were detected in the cytoplasm of breast cancer cells through immunostaining. After the onset of recurrent metastases, two S-FN-positive and six S-FN-negative patients received anticancer drug treatment; however, further progression was observed in five S-FN-negative patients. Conclusion S-FN-positive patients are less likely to develop distant metastases, have a better prognosis, and may be less resistant to therapeutic agents than S-FN-negative patients, which contain many epithelial-mesenchymal transition cells.
Collapse
Affiliation(s)
- Hiroshi Takeyama
- Department of Breast, Thyroid, Endocrine Surgery, Jikei University School of Medicine, Tokyo, Japan
| | - Yoshinobu Manome
- Department of Molecular Cell Biology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Gui X, Liang X, Li H. Effectiveness, safety, and impact on quality of life of eribulin-based therapy in heavily pretreated patients with metastatic breast cancer: A real-world analysis. Cancer Med 2023; 12:16793-16804. [PMID: 37403746 PMCID: PMC10501238 DOI: 10.1002/cam4.6301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023] Open
Abstract
INTRODUCTION Eribulin is currently recommended for the treatment of patients with metastatic breast cancer (MBC) pre-treated with taxanes and anthracyclines. The aim of the present study was to evaluate the effectiveness and safety of eribulin and its impact on health-related quality of life in heavily pre-treated patients with MBC. METHODS Data from MBC patients who had received eribulin-based therapy at Beijing Cancer Hospital between January 2020 and July 2022 were analyzed retrospectively. Progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), adverse effects (AEs) and health-related quality of life (HRQoL) were assessed. RESULTS Data from 118 patients who had received eribulin to treat MBC were included. Median PFS was 4.2 months and median OS had not been reached. The ORR was 13.6% (16/118) and DCR was 75.4% (89/118). The median PFS in patients who received eribulin in second-line (26/118), third-line (29/118), or fourth-line or later (63/118) was 4.5, 4.2, and 3.9 months, respectively. The median OS in patients who received eribulin in third- or later line (n = 92) was 14.1 months. Patients who received eribulin combination therapy had a significantly longer median PFS compared with those who received eribulin monotherapy (4.5 vs. 3.4 months, p = 0.007) and there was a trend towards a longer median OS (not reached vs. 12.1 months). The most common grade 3-4 adverse events were neutropenia (22.9%), leukocytopenia (13.6%) and asthenia/fatigue (8.5%), without significant differences in safety between eribulin monotherapy and combination therapy. Quality of life was similar in patients who received eribulin monotherapy and combination therapy, except for cognitive function and nausea and vomiting symptoms, which were better with combination therapy. CONCLUSIONS The present study suggests that eribulin-based therapy is an effective treatment option and well tolerated for heavily pre-treated patients with MBC. Eribulin combination therapy might improve PFS and HRQoL compared with eribulin monotherapy.
Collapse
Affiliation(s)
- Xinyu Gui
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Breast Oncology, Peking University Cancer Hospital & InstituteBeijing, China 100142China
| | - Xu Liang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Breast Oncology, Peking University Cancer Hospital & InstituteBeijing, China 100142China
| | - Huiping Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Breast Oncology, Peking University Cancer Hospital & InstituteBeijing, China 100142China
| |
Collapse
|
35
|
Oya K, Nakamura Y, Watanabe R, Tanaka R, Ichimura Y, Kubota N, Matsumura Y, Tahara H, Okiyama N, Fujimoto M, Nomura T, Fujisawa Y. Eribulin mesylate exerts antitumor effects via CD103. Oncoimmunology 2023; 12:2218782. [PMID: 37261089 PMCID: PMC10228394 DOI: 10.1080/2162402x.2023.2218782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 04/30/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Eribulin mesylate (ERB) is a synthetic analog of halichondrin B, inhibiting tumor cell growth by disrupting microtubule function. Recently, anticancer drugs have been shown to not only act directly on tumor cells but also to exert antitumor effects by modifying the tumor environment. Although ERB has also been speculated to modify the tumor microenvironment including the immune response to tumors, the precise mechanism remains unclear. In our study, ERB suppressed the tumor growth of MC38 colon cancer in wildtype mice, whereas ERB failed to inhibit the tumor growth in Rag1-deficient mice which lack both B and T cells. Moreover, depletion of either CD4+ or CD8+ T cells abrogated the antitumor effect of ERB, indicating that both CD4+ and CD8+ T cells play an important role in ERB-induced antitumor effects. Furthermore, ERB treatment increased the number of tumor infiltrating lymphocytes (TILs) as well as the expression of activation markers (CD38 and CD69), immune checkpoint molecules (LAG3, TIGIT and Tim3) and cytotoxic molecules (granzyme B and perforin) in TILs. ERB upregulated E-cadherin expression in MC38. CD103 is a ligand of E-cadherin and induces T-cell activation. ERB increased the proportion of CD103+ cells in both CD4+ and CD8+ TILs. The ERB-induced antitumor effect with the increased TIL number and the increased expression of activation markers, inhibitory checkpoint molecules and cytotoxic molecules in TILs was abrogated in CD103-deficient mice. Collectively, these results suggest that ERB exerts antitumor effects by upregulation of E-cadherin expression in tumor cells and subsequent activation of CD103+ TILs.
Collapse
Affiliation(s)
- Kazumasa Oya
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiyuki Nakamura
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Rei Watanabe
- The Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryota Tanaka
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuki Ichimura
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Noriko Kubota
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yutaka Matsumura
- The Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hideaki Tahara
- Project Division of Cancer Biomolecular Therapy, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Cancer Drug Discovery and Development, Osaka International Cancer Center, Osaka, Japan
| | - Naoko Okiyama
- Department of Dermatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Manabu Fujimoto
- The Department of Dermatology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Toshifumi Nomura
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasuhiro Fujisawa
- The Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
36
|
Hsu CY, Yanagi T, Maeda T, Nishihara H, Miyamoto K, Kitamura S, Tokuchi K, Ujiie H. Eribulin inhibits growth of cutaneous squamous cell carcinoma cell lines and a novel patient-derived xenograft. Sci Rep 2023; 13:8650. [PMID: 37244956 DOI: 10.1038/s41598-023-35811-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Advanced cutaneous squamous cell carcinoma (cSCC) is treated with chemotherapy and/or radiotherapy, but these typically fail to achieve satisfactory clinical outcomes. There have been no preclinical studies to evaluate the effectiveness of eribulin against cSCC. Here, we examine the effects of eribulin using cSCC cell lines and a novel cSCC patient-derived xenograft (PDX) model. In the cSCC cell lines (A431 and DJM-1 cells), eribulin was found to inhibit tumor cell proliferation in vitro as assessed by cell ATP levels. DNA content analysis by fluorescence-activated cell sorting (FACS) showed that eribulin induced G2/M cell cycle arrest and apoptosis. In xenograft models of cSCC cell lines, the administration of eribulin suppressed tumor growth in vivo. We also developed a cSCC patient-derived xenograft (PDX) which reproduces the histological and genetic characteristics of a primary tumor. Pathogenic mutations in TP53 and ARID2 were detected in the patient's metastatic tumor and in the PDX tumor. The cSCC-PDX responded well to the administration of eribulin and cisplatin. In conclusion, the present study shows the promising antineoplastic effects of eribulin in cSCC. Also, we established a novel cSCC-PDX model that preserves the patient's tumor. This PDX could assist researchers who are exploring innovative therapies for cSCC.
Collapse
Affiliation(s)
- Che-Yuan Hsu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan.
| | - Takuya Maeda
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Kodai Miyamoto
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Shinya Kitamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Keiko Tokuchi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| |
Collapse
|
37
|
Li F, Sun H, Yu Y, Che N, Han J, Cheng R, Zhao N, Guo Y, Huang C, Zhang D. RIPK1-dependent necroptosis promotes vasculogenic mimicry formation via eIF4E in triple-negative breast cancer. Cell Death Dis 2023; 14:335. [PMID: 37217473 PMCID: PMC10203343 DOI: 10.1038/s41419-023-05841-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Necroptosis is a caspase-independent form of programmed cell death. Receptor interacting protein kinase 1 (RIPK1) is a key molecule in the initiation of necroptosis and the formation of the necrotic complex. Vasculogenic mimicry (VM) provides a blood supply to tumor cells that is not dependent on endothelial cells. However, the relationship between necroptosis and VM in triple-negative breast cancer (TNBC) is not fully understood. In this study, we found that RIPK1-dependent necroptosis promoted VM formation in TNBC. Knockdown of RIPK1 significantly suppressed the number of necroptotic cells and VM formation. Moreover, RIPK1 activated the p-AKT/eIF4E signaling pathway during necroptosis in TNBC. eIF4E was blocked by knockdown of RIPK1 or AKT inhibitors. Furthermore, we found that eIF4E promoted VM formation by promoting epithelial-mesenchymal transition (EMT) and the expression and activity of MMP2. In addition to its critical role in necroptosis-mediated VM, eIF4E was essential for VM formation. Knockdown of eIF4E significantly suppressed VM formation during necroptosis. Finally, through clinical significance, the results found that eIF4E expression in TNBC was positively correlated with the mesenchymal marker vimentin, the VM marker MMP2, and the necroptosis markers MLKL and AKT. In conclusion, RIPK1-dependent necroptosis promotes VM formation in TNBC. Necroptosis promotes VM formation by activating RIPK1/p-AKT/eIF4E signaling in TNBC. eIF4E promotes EMT and MMP2 expression and activity, leading to VM formation. Our study provides a rationale for necroptosis-mediated VM and also providing a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Fan Li
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Huizhi Sun
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Yihui Yu
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Na Che
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Jiyuan Han
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Runfen Cheng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China
| | - Yuhong Guo
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Chongbiao Huang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China.
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin 300070, Tianjin, China.
| |
Collapse
|
38
|
Sakibuzzaman M, Mahmud S, Afroze T, Fathma S, Zakia UB, Afroz S, Zafar F, Hossain M, Barua A, Akter S, Chowdhury HI, Ahsan E, Eshan SH, Fariza TT. Pathology of breast cancer metastasis and a view of metastasis to the brain. Int J Neurosci 2023; 133:544-554. [PMID: 34044732 DOI: 10.1080/00207454.2021.1935929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Despite the advances in diagnosis and management of breast cancer, metastasis has been responsible for the staggering percentage of breast cancer-related death. Mortality threat can be explained mostly by the lack of proper understanding of the diversity of pathological features and underlying mechanism of breast cancer metastasis and effective targeted therapy. Breast cancer stem cells (BCSCs) are the potential source of tumor cells spread to distant organs. BCSCs targeted therapy can suppress the breast cancer progression to metastasis. Spreading of tumor cells to the bone, lung, liver, and brain occurs through a distinct non-random process; called metastasis organotropism. Recently, brain metastasis in breast cancer patients has been detected more frequently, causing a significant clinical burden. BRCA1 and BRCA2 associated breast cancers carry a remarkably higher propensity of CNS metastasis. BRCA1 and BRCA2 associated breast cancers commonly have the propensity to be the triple-negative (TN) and hormone receptors (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative molecular subtypes, respectively. Regardless of molecular subtypes, metastasis is most commonly evident at the bone. Heterogeneity is a critical pathological feature, leads to therapeutic resistance. BCSCs, biomarkers expression patterns, and mutations contribute to heterogeneity. In this paper, we discuss crucial pathological features of breast cancer metastasis, emphasizing metastasis organotropism and heterogeneity; and mechanisms of breast cancer metastasis, highlighting the pathways of metastasis to the brain. We consider that this paper reinforces future research areas and benefits the general readers, physicians, and researchers to identify potential areas to develop targeted therapies.
Collapse
Affiliation(s)
- Md Sakibuzzaman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Shahriar Mahmud
- Sher-E-Bangla Medical College and Hospital, Barisal, Bangladesh
| | | | - Sawsan Fathma
- Bangladesh Medical College and Hospital, Dhaka, Bangladesh
| | | | - Sabrina Afroz
- Faridpur Medical College and Hospital, Faridpur, Bangladesh
| | - Farzina Zafar
- Shaheed Suhrawardy Medical College and Hospital, Dhaka, Bangladesh
| | - Maksuda Hossain
- Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Amit Barua
- Institute of Applied Health Sciences, Chattogram, Bangladesh
| | - Sabiha Akter
- Sher-E-Bangla Medical College and Hospital, Barisal, Bangladesh
| | | | - Eram Ahsan
- Medical College for Women and Hospital, Dhaka, Bangladesh
| | - Shayet Hossain Eshan
- Department of Internal Medicine, Amita Health Saint Joseph Hospital Chicago, Chicago, IL, USA
| | | |
Collapse
|
39
|
Bagheri M, Aisha Mohamed G, Mohamed Saleem MA, Ognjenovic NB, Lu H, Kolling FW, Wilkins OM, Das S, La Croix IS, Nagaraj SH, Muller KE, Gerber SA, Miller TW, Pattabiraman DR. Pharmacological Induction of mesenchymal-epithelial transition chemosensitizes breast cancer cells and prevents metastatic progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537586. [PMID: 37131809 PMCID: PMC10153261 DOI: 10.1101/2023.04.19.537586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The epithelial-mesenchymal transition (EMT) is a developmental program co-opted by tumor cells that aids the initiation of the metastatic cascade. Tumor cells that undergo EMT are relatively chemoresistant, and there are currently no therapeutic avenues specifically targeting cells that have acquired mesenchymal traits. We show that treatment of mesenchymal-like triple-negative breast cancer (TNBC) cells with the microtubule-destabilizing chemotherapeutic eribulin, which is FDA-approved for the treatment of advanced breast cancer, leads to a mesenchymal-epithelial transition (MET). This MET is accompanied by loss of metastatic propensity and sensitization to subsequent treatment with other FDA-approved chemotherapeutics. We uncover a novel epigenetic mechanism of action that supports eribulin pretreatment as a path to MET induction that curtails metastatic progression and the evolution of therapy resistance.
Collapse
Affiliation(s)
- Meisam Bagheri
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
| | - Gadisti Aisha Mohamed
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | | | - Nevena B. Ognjenovic
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Hanxu Lu
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Fred W. Kolling
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
| | - Owen M. Wilkins
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover NH 03755 USA
| | | | - Ian S. La Croix
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Shivashankar H. Nagaraj
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
- Translational Research Institute, Brisbane QLD 4102, Australia
| | - Kristen E. Muller
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
| | - Scott A. Gerber
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
| | - Todd W. Miller
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
| | - Diwakar R. Pattabiraman
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon NH 03756, USA
- Lead contact
| |
Collapse
|
40
|
Masuda T, Ueo H, Okumura Y, Kai Y, Ando Y, Masuguchi K, Kitagawa M, Kitagawa A, Hayashi N, Tsuruda Y, Hisamatsu Y, Suehiro S, Ohmura H, Fujiyoshi K, Tanaka F, Mimori K. Dynamic Changes in Peripheral Systemic Immunity Markers During Chemotherapy in HER2-negative Advanced Breast Cancer. Cancer Genomics Proteomics 2023; 20:182-194. [PMID: 36870689 PMCID: PMC9989675 DOI: 10.21873/cgp.20373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND/AIM The immune system has a pivotal role in modulating the response to chemotherapy in breast cancer (BC). However, the immune status during chemotherapy remains unclear. We evaluated the sequential changes in peripheral systemic immunity markers in BC patients treated with various chemotherapeutic agents. MATERIALS AND METHODS We examined the correlation between the peripheral systemic immunity markers, neutrophil-to-lymphocyte ratio (NLR), absolute lymphocyte count (ALC) and the local cytolytic activity (CYT) score obtained by quantitative reverse-transcription polymerase chain reaction of 84 preoperative BC patients. Next, we observed the sequential changes in the peripheral systemic immunity markers during treatment with four anticancer drugs: oral 5-fluorouracil derivative; S-1, epirubicin plus cyclophosphamide; paclitaxel plus the anti-vascular endothelial growth factor antibody bevacizumab, and eribulin in 172 HER2-negative advanced BC patients. Finally, we examined the correlation between the changes in the peripheral systemic immunity markers, time to treatment failure (TTF) and progression-free survival (PFS). RESULTS A negative correlation was found between ALC and NLR. ALC-low and NLR-high cases were positively associated with CYT score-low cases. The ratio of ALC-increase and NLR-decrease varies depending on the anticancer drugs used. The responder group (TTF ≥3 months) had a higher NLR-decrease ratio than the nonresponder group (TTF <3 months). Patients with a high NLR-decrease ratio showed higher PFS. CONCLUSION The change in ALC or NLR varies according to the anticancer drugs, suggesting differential immunomodulatory effects of the drugs. Furthermore, the change in NLR reflects the therapeutic efficacy of chemotherapy in advanced BC.
Collapse
Affiliation(s)
- Takaaki Masuda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | | | - Yuta Okumura
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | | | - Yuki Ando
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Ken Masuguchi
- Department of Pharmacy, Kyushu University Beppu Hospital, Oita, Japan
| | - Miwa Kitagawa
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan.,Department of Breast Surgery, Medical Corporation Kyoujinkai Komatsu Hospital, Osaka, Japan
| | - Akihiro Kitagawa
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan.,Department of Surgery, Sakai City Medical Center, Osaka, Japan
| | - Naoki Hayashi
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Yusuke Tsuruda
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Yuichi Hisamatsu
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan
| | - Shuji Suehiro
- Department of Breast Surgery, Tsurumi Hospital, Oita, Japan
| | - Hirofumi Ohmura
- Department of Internal Medicine, Kyushu University Beppu Hospital, Oita, Japan
| | | | - Fumiaki Tanaka
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan.,Tanaka Breast, Surgery, Internal Medicine Clinic, Oita, Japan
| | - Koshi Mimori
- Department of Surgery, Kyushu University Beppu Hospital, Oita, Japan;
| |
Collapse
|
41
|
Xie X, Lee J, Fuson JA, Liu H, Iwase T, Yun K, Margain C, Tripathy D, Ueno NT. Identification of Kinase Targets for Enhancing the Antitumor Activity of Eribulin in Triple-Negative Breast Cell Lines. Biomedicines 2023; 11:735. [PMID: 36979714 PMCID: PMC10045293 DOI: 10.3390/biomedicines11030735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most aggressive molecular subtype of breast cancer, and current treatments are only partially effective in disease control. More effective combination approaches are needed to improve the survival of TNBC patients. Eribulin mesylate, a non-taxane microtubule dynamics inhibitor, is approved by the U.S. Food and Drug Administration to treat metastatic breast cancer after at least two previous chemotherapeutic regimens. However, eribulin as a single agent has limited therapeutic efficacy against TNBC. METHODS High-throughput kinome library RNAi screening, Ingenuity Pathway Analysis, and STRING analysis were performed to identify target kinases for combination with eribulin. The identified combinations were validated using in vivo and ex vivo proliferation assays. RESULTS We identified 135 potential kinase targets whose inhibition enhanced the antiproliferation effect of eribulin in TNBC cells, with the PI3K/Akt/mTOR and the MAPK/JNK pathways emerging as the top candidates. Indeed, copanlisib (pan-class I PI3K inhibitor), everolimus (mTOR inhibitor), trametinib (MEK inhibitor), and JNK-IN-8 (pan-JNK inhibitor) produced strong synergistic antiproliferative effects when combined with eribulin, and the PI3K and mTOR inhibitors had the most potent effects in vitro. CONCLUSIONS Our data suggest a new strategy of combining eribulin with PI3K or mTOR inhibitors to treat TNBC.
Collapse
Affiliation(s)
- Xuemei Xie
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jangsoon Lee
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jon A. Fuson
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huey Liu
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Toshiaki Iwase
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kyuson Yun
- Research Institute at Houston Methodist, Weill Cornell Medical College, Houston, TX 77030, USA
| | | | - Debu Tripathy
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Cancer Biology and Therapeutics, University of Hawai’i Cancer Center, Honolulu, HI 96813, USA
| |
Collapse
|
42
|
Hayashi T, Kobayashi N, Ushida K, Asai N, Nakano S, Fujii K, Ando T, Utsumi T. Effect of eribulin on epithelial-mesenchymal transition plasticity in metastatic breast cancer: An exploratory, prospective study. Genes Cells 2023; 28:364-373. [PMID: 36849792 DOI: 10.1111/gtc.13019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 02/01/2023] [Accepted: 02/20/2023] [Indexed: 03/01/2023]
Abstract
Epithelial-mesenchymal transition (EMT) plays a pivotal role in cancer metastasis and treatment resistance, which worsens prognosis. In phase III trials, eribulin improved overall survival in metastatic breast cancer (MBC) patients. In preclinical studies, eribulin suppressed EMT. However, clinical data on the use of eribulin for MBC patients are limited. In this exploratory, prospective study, we examined the effect of eribulin on EMT in MBC patients. Twenty-two patients aged 44-82 years with recurrent breast cancer or MBC were treated with eribulin. Breast cancer tissue samples were obtained before treatment and on Day 15 ± 5 of the first cycle of eribulin treatment. EMT markers (E-cadherin, claudin-3, vimentin, and N-cadherin) were analyzed using western blotting. EMT changes were evaluated based on the ratio of epithelial to mesenchymal markers before and after treatment in individual tumors. E-cadherin/vimentin, claudin-3/vimentin, E-cadherin/N-cadherin, and claudin-3/N-cadherin ratios were significantly higher after treatment (p = .007, p = .005, p = .006, and p = .011, respectively). Based on E-cadherin/vimentin, 65.0% of tumors shifted to an epithelial phenotype, as compared to 66.7% based on claudin-3/vimentin, 84.6% based on E-cadherin/N-cadherin, and 71.4% based on claudin-3/N-cadherin ratios. Thus, our results showed that eribulin suppressed EMT in breast cancer tissues.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Naomi Kobayashi
- Department of Breast Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Aichi, Japan
| | - Kaori Ushida
- Department of Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Naoya Asai
- Department of Pathology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Shogo Nakano
- Division of Breast and Endocrine Surgery, Department of Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kimihito Fujii
- Division of Breast and Endocrine Surgery, Department of Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takahito Ando
- Division of Breast and Endocrine Surgery, Department of Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Toshiaki Utsumi
- Department of Surgery, Fujita Health University School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
43
|
Legátová A, Pelantová M, Rösel D, Brábek J, Škarková A. The emerging role of microtubules in invasion plasticity. Front Oncol 2023; 13:1118171. [PMID: 36860323 PMCID: PMC9969133 DOI: 10.3389/fonc.2023.1118171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
The ability of cells to switch between different invasive modes during metastasis, also known as invasion plasticity, is an important characteristic of tumor cells that makes them able to resist treatment targeted to a particular invasion mode. Due to the rapid changes in cell morphology during the transition between mesenchymal and amoeboid invasion, it is evident that this process requires remodeling of the cytoskeleton. Although the role of the actin cytoskeleton in cell invasion and plasticity is already quite well described, the contribution of microtubules is not yet fully clarified. It is not easy to infer whether destabilization of microtubules leads to higher invasiveness or the opposite since the complex microtubular network acts differently in diverse invasive modes. While mesenchymal migration typically requires microtubules at the leading edge of migrating cells to stabilize protrusions and form adhesive structures, amoeboid invasion is possible even in the absence of long, stable microtubules, albeit there are also cases of amoeboid cells where microtubules contribute to effective migration. Moreover, complex crosstalk of microtubules with other cytoskeletal networks participates in invasion regulation. Altogether, microtubules play an important role in tumor cell plasticity and can be therefore targeted to affect not only cell proliferation but also invasive properties of migrating cells.
Collapse
Affiliation(s)
- Anna Legátová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Markéta Pelantová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Jan Brábek
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Aneta Škarková
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia,*Correspondence: Aneta Škarková,
| |
Collapse
|
44
|
Ho GY, Kyran EL, Bedo J, Wakefield MJ, Ennis DP, Mirza HB, Vandenberg CJ, Lieschke E, Farrell A, Hadla A, Lim R, Dall G, Vince JE, Chua NK, Kondrashova O, Upstill-Goddard R, Bailey UM, Dowson S, Roxburgh P, Glasspool RM, Bryson G, Biankin AV, Cooke SL, Ratnayake G, McNally O, Traficante N, DeFazio A, Weroha SJ, Bowtell DD, McNeish IA, Papenfuss AT, Scott CL, Barker HE. Epithelial-to-Mesenchymal Transition Supports Ovarian Carcinosarcoma Tumorigenesis and Confers Sensitivity to Microtubule Targeting with Eribulin. Cancer Res 2022; 82:4457-4473. [PMID: 36206301 PMCID: PMC9716257 DOI: 10.1158/0008-5472.can-21-4012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 06/15/2022] [Accepted: 10/04/2022] [Indexed: 01/24/2023]
Abstract
Ovarian carcinosarcoma (OCS) is an aggressive and rare tumor type with limited treatment options. OCS is hypothesized to develop via the combination theory, with a single progenitor resulting in carcinomatous and sarcomatous components, or alternatively via the conversion theory, with the sarcomatous component developing from the carcinomatous component through epithelial-to-mesenchymal transition (EMT). In this study, we analyzed DNA variants from isolated carcinoma and sarcoma components to show that OCS from 18 women is monoclonal. RNA sequencing indicated that the carcinoma components were more mesenchymal when compared with pure epithelial ovarian carcinomas, supporting the conversion theory and suggesting that EMT is important in the formation of these tumors. Preclinical OCS models were used to test the efficacy of microtubule-targeting drugs, including eribulin, which has previously been shown to reverse EMT characteristics in breast cancers and induce differentiation in sarcomas. Vinorelbine and eribulin more effectively inhibited OCS growth than standard-of-care platinum-based chemotherapy, and treatment with eribulin reduced mesenchymal characteristics and N-MYC expression in OCS patient-derived xenografts. Eribulin treatment resulted in an accumulation of intracellular cholesterol in OCS cells, which triggered a downregulation of the mevalonate pathway and prevented further cholesterol biosynthesis. Finally, eribulin increased expression of genes related to immune activation and increased the intratumoral accumulation of CD8+ T cells, supporting exploration of immunotherapy combinations in the clinic. Together, these data indicate that EMT plays a key role in OCS tumorigenesis and support the conversion theory for OCS histogenesis. Targeting EMT using eribulin could help improve OCS patient outcomes. SIGNIFICANCE Genomic analyses and preclinical models of ovarian carcinosarcoma support the conversion theory for disease development and indicate that microtubule inhibitors could be used to suppress EMT and stimulate antitumor immunity.
Collapse
Affiliation(s)
- Gwo Yaw Ho
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- The Royal Women's Hospital, Parkville, Victoria, Australia
| | - Elizabeth L. Kyran
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Cancer Research UK Cambridge Institute, Cambridge, United Kingdom
| | - Justin Bedo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- School of Computing and Information Systems, the University of Melbourne, Parkville, Victoria, Australia
| | - Matthew J. Wakefield
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
| | - Darren P. Ennis
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Hasan B. Mirza
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Cassandra J. Vandenberg
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Lieschke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew Farrell
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Anthony Hadla
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ratana Lim
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Genevieve Dall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - James E. Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ngee Kiat Chua
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Olga Kondrashova
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Rosanna Upstill-Goddard
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Ulla-Maja Bailey
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Suzanne Dowson
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Patricia Roxburgh
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Rosalind M. Glasspool
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Gareth Bryson
- Department of Pathology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Andrew V. Biankin
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | - Susanna L. Cooke
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
| | | | - Orla McNally
- The Royal Women's Hospital, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Nadia Traficante
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Anna DeFazio
- Centre for Cancer Research, The Westmead Institute for Medical Research, Sydney, Australia
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, Australia
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, Australia
| | - S. John Weroha
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - David D. Bowtell
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Iain A. McNeish
- Division of Cancer and Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, United Kingdom
- Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Anthony T. Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Clare L. Scott
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- The Royal Women's Hospital, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Sir Peter MacCallum Cancer Centre Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Holly E. Barker
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
45
|
Luong T, Cukierman E. Eribulin normalizes pancreatic cancer-associated fibroblasts by simulating selected features of TGFβ inhibition. BMC Cancer 2022; 22:1255. [PMID: 36461015 PMCID: PMC9719234 DOI: 10.1186/s12885-022-10330-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Less than 11% of pancreatic cancer patients survive 5-years post-diagnosis. The unique biology of pancreatic cancer includes a significant expansion of its desmoplastic tumor microenvironment, wherein cancer-associated fibroblasts (CAFs) and their self-produced extracellular matrix are key components. CAF functions are both tumor-supportive and tumor-suppressive, while normal fibroblastic cells are solely tumor-suppressive. Knowing that CAF-eliminating drugs are ineffective and can accelerate cancer progression, therapies that "normalize" CAF function are highly pursued. Eribulin is a well-tolerated anti-microtubule drug used to treat a plethora of neoplasias, including advanced/metastatic cancers. Importantly, eribulin can inhibit epithelial to mesenchymal transition via a mechanism akin to blocking pathways induced by transforming growth factor-beta (TGFβ). Notably, canonical TGFβ signaling also plays a pivotal role in CAF activation, which is necessary for the development and maintenance of desmoplasia. Hence, we hypothesized that eribulin could modulate, and perhaps "normalize" CAF function. METHODS To test this premise, we used a well-established in vivo-mimetic fibroblastic cell-derived extracellular matrix (CDM) system and gauged the effects of eribulin on human pancreatic CAFs and cancer cells. This pathophysiologic fibroblast/matrix functional unit was also used to query eribulin effects on CDM-regulated pancreatic cancer cell survival and invasive spread. RESULTS Demonstrated that intact CAF CDMs modestly restricted eribulin from obstructing pancreatic cancer cell growth. Nonetheless, eribulin-treated CAFs generated CDMs that limited nutrient-deprived pancreatic cancer cell survival, similar to reported tumor-suppressive CDMs generated by TGFβ-deficient CAFs. CONCLUSIONS Data from this study support the central proposed premise suggesting that eribulin could be used as a CAF/matrix-normalizing drug.
Collapse
Affiliation(s)
- Tiffany Luong
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, 19111, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment, Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, 19111, USA.
| |
Collapse
|
46
|
Narui K, Miura D, Hasegawa Y, Tachibana A, Horiguchi J, Hayashi M, Miyashita M, Kubota T, Suzuki M, Yamada K, Yamada A, Akazawa K, Kohno N, Ishikawa T. A Randomized Controlled Phase 2 Study of Neoadjuvant Eribulin Versus Paclitaxel in Women with Operable Breast Cancer: The JONIE-3 Study. Clin Breast Cancer 2022; 22:e881-e891. [PMID: 36151017 DOI: 10.1016/j.clbc.2022.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/20/2022] [Accepted: 08/17/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Neoadjuvant chemotherapy (NAC) is essential for surgical downstaging of early-stage breast cancer, but taxane administration is associated with neuropathy. We investigated whether eribulin induces less neuropathy than paclitaxel. METHODS In this multicentre, randomised study (UMIN000012817), patients diagnosed with invasive breast cancer between December 2013 and April 2016 were randomly assigned to group E (eribulin followed by fluorouracil, epirubicin, and cyclophosphamide; FEC) or group P (paclitaxel followed by FEC). The primary endpoint was incidence of grade 1 or higher peripheral neuropathy according to the Common Terminology Criteria for Adverse Events (CTCAE). Secondary endpoints were pathological complete response (pCR), clinical response, breast-conserving surgery, adverse events, disease-free survival (DFS), and patient neurotoxicity questionnaire (PNQ) analysis. RESULTS One hundred and eighteen cases were analyzed for safety and 115 were evaluated for efficacy. Peripheral sensory neuropathy was significantly lower in group E after week 6, while peripheral motor neuropathy in group E was significantly lower at weeks 9, 12, and 15. pCR in groups E and P was 20.7% and 29.8% (P = .289), respectively, and clinical response was 55.2% and 77.2% (P = .017), respectively. Three-year DFS was 89.7% in group E and 86.0% in group P (P = .561). Neutropenia was more frequent and more severe in group E. PNQ was evaluated for 4 years, and item 1 (sensory) was consistently lower in group E. CONCLUSION Neuropathy was significantly less frequent and less severe in patients who received eribulin compared with paclitaxel. Thus, eribulin could be a good alternative to paclitaxel in patients suffering severe neuropathy.
Collapse
Affiliation(s)
- Kazutaka Narui
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Japan
| | | | - Yoshie Hasegawa
- Department of Breast Surgery, Hirosaki Municipal Hospital, Aomori, Japan
| | - Akihiko Tachibana
- Department of Breast Surgery, Kanto Central Hospital of the Mutual Aid Association of Public School Teachers, Tokyo, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Chiba, Japan
| | | | | | - Tomoyuki Kubota
- Department of Breast Surgery, Kamiiida Daiichi General Hospital, Aichi, Japan
| | - Masato Suzuki
- Department of Breast Surgery and Breast Center, National Hospital Organization, Chiba Medical Center, Chiba, Japan
| | - Kimito Yamada
- Department of Breast Oncology and Surgery, Tokyo Medical University, Tokyo, Japan
| | - Akimitsu Yamada
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Japan
| | - Kohei Akazawa
- Department of Medical Informatics, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Norio Kohno
- Department of Breast Surgery, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University, Tokyo, Japan.
| |
Collapse
|
47
|
Jafri M, Kristeleit H, Misra V, Baxter M, Ahmed S, Jegannathen A, Jain A, Maskell D, Barthakur U, Edwards G, Walter HS, Walshaw R, Khan M, Borley A, Rea D. Eribulin Treatment for Patients with Metastatic Breast Cancer: The UK Experience - A Multicenter Retrospective Study. Oncology 2022; 100:666-673. [PMID: 36044833 PMCID: PMC9808648 DOI: 10.1159/000526140] [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: 12/02/2021] [Accepted: 06/20/2022] [Indexed: 01/07/2023]
Abstract
INTRODUCTION This study examined real-world data from patients who received eribulin for metastatic breast cancer (MBC) collected from 14 hospitals across the UK. METHODS Anonymized data were collected retrospectively from patients with MBC who had received eribulin. The data included the hormone-receptor status, histological diagnosis, age, prior chemotherapy, response to eribulin, progression-free survival (PFS), and overall survival (OS). RESULTS Among 577 patients analyzed, the median age was 56 years, and most patients (73%) were estrogen-receptor positive. The median OS was 288 days (95% confidence interval [CI]: 261-315), and the PFS was 117 days (95% CI: 105-129). The median OS was higher among older patients (≥65 vs. <65 years: 325 days [95% CI: 264-385] vs. 285 days [95% CI: 252-317]; p = 0.028). The median OS was also higher in patients who received eribulin after fewer prior lines of chemotherapy (≤2 vs. >2 prior: 328 days [95% CI: 264-385] vs. 264 days [95% CI: 229-298]; p = 0.042). DISCUSSION/CONCLUSION These retrospective data suggest that eribulin can be successfully used in older patients with MBC. Eribulin treatment was more effective in earlier-line settings, which, while predictable, supports consideration of eribulin as a second-line treatment option.
Collapse
Affiliation(s)
- Mariam Jafri
- Medical Oncology, University Hospitals Birmingham Foundation Trust, Birmingham, UK,*Mariam Jafri,
| | - Hartmut Kristeleit
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Vivek Misra
- Clinical Oncology, The Christie NHS Foundation Trust, Cancer Centre, Manchester, UK
| | - Mark Baxter
- Molecular and Clinical Medicine, Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Samreen Ahmed
- Medical Oncology, University Hospitals of Leicester, Leicester, UK
| | - Apurna Jegannathen
- Clinical Oncology, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, UK
| | - Ankit Jain
- Medical Oncology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK
| | - David Maskell
- Clinical Oncology, Norfolk and Norwich University Hospitals NHS Trust, Norwich, UK
| | - Urmila Barthakur
- Clinical Oncology, Taunton and Somerset Foundation Trust, Taunton, UK
| | | | - Harriet S. Walter
- Department of Oncology, University Hospitals of Leicester, Leicester, UK
| | - Richard Walshaw
- Division of Cancer Sciences, The Christie NHS Foundation Trust, Cancer Centre, Manchester, UK
| | - Madeha Khan
- Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Annabel Borley
- Clinical Oncology, Velindre University NHS Trust, Cardiff, UK
| | - Daniel Rea
- Medical Oncology, University Hospitals Birmingham Foundation Trust, Birmingham, UK,Medical Oncology, University of Birmingham Clinical Trials Unit, Birmingham, UK
| |
Collapse
|
48
|
López González A, Del Barco Berrón S, Grau I, Galan M, Castelo Fernández B, Cortés A, Sánchez Rovira P, Martinez-Bueno A, Gonzalez X, García A, Gener P, Mina L, Alcalá-López D, Sampayo M, Cortés J, Pérez-Garcia JM, Llombart-Cussac A, López-Miranda E. Challenging Endocrine Sensitivity of Hormone Receptor-Positive/HER2-Negative Advanced Breast Cancer with the Combination of Eribulin and Endocrine Therapy: The REVERT Study. Cancers (Basel) 2022; 14:cancers14235880. [PMID: 36497361 PMCID: PMC9737152 DOI: 10.3390/cancers14235880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Luminal advanced breast cancer (ABC) patients eventually progress on endocrine therapy. REVERT aimed to explore whether eribulin could restore endocrine sensitivity in a randomized, non-comparative phase II trial. METHODS Aromatase inhibitor (AI)-resistant patients with luminal ABC were randomized 1:1 to receive eribulin +/- AI. Patients were stratified by prior cyclin-dependent kinases 4/6 inhibitor (CDK4/6i) treatment. The primary endpoint was an investigator-assessed overall response rate (ORR) according to RECIST version 1.1 in the eribulin + AI arm. An interim analysis was planned with 11 evaluable patients according to a two-stage Simon design. RESULTS Twenty-two patients were enrolled (15 eribulin + AI arm; 7 eribulin arm). The trial was terminated early in March 2021, with eight (36.4%) patients still on treatment. ORR was 26.7% in the eribulin + AI arm (95% CI, 7.8-55.1%; p = 0.0541). In the eribulin arm, two (28.6%) patients had an objective response (95% CI, 3.7-71.0%). The difference between the study arms was not significant (p = 0.918). The addition of AI to eribulin also failed to show improvement in other efficacy endpoints. A significant interaction between the treatment arm and previous CDK4/6i treatment was observed for ORR (p = 0.018) and progression-free survival (p = 0.084). Overall, the toxicity profile was consistent with the known safety profile of eribulin. No treatment-related deaths were reported. CONCLUSION Eribulin + AI does not seem to improve outcomes compared with eribulin monotherapy in patients with AI-resistant luminal ABC. This chemo-endocrine approach deserves further investigation after progression to CDK4/6i-based therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Alfonso Cortés
- Hospital Universitario Ramón y Cajal, 2559 Madrid, Spain
| | | | | | - Xavier Gonzalez
- Instituto Oncológico Dr. Rosell, Hospital General de Cataluña, 08190 San Cugat del Vallés, Spain
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, 08017 Barcelona, Spain
| | - Almudena García
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
| | - Petra Gener
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
| | - Leonardo Mina
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
| | | | - Miguel Sampayo
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, 08017 Barcelona, Spain
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - José Manuel Pérez-Garcia
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, 08017 Barcelona, Spain
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
| | - Antonio Llombart-Cussac
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
- Arnau de Vilanova Hospital, Universidad Católica de Valencia, 46015 Valencia, Spain
| | - Elena López-Miranda
- Hospital Universitario Ramón y Cajal, 2559 Madrid, Spain
- Medica Scientia Innovation Research SL (MEDSIR), 08018 Barcelona, Spain
- Correspondence:
| |
Collapse
|
49
|
Kenny L, Beresford M, Brown I, Misra V, Kristeleit H. Eribulin for the treatment of advanced breast cancer: A prospective observational registry study. Eur J Cancer Care (Engl) 2022; 31:e13747. [PMID: 36336468 PMCID: PMC9787722 DOI: 10.1111/ecc.13747] [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: 01/11/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Eribulin treatment improved overall survival with predictable toxicities in phase 3 trials of patients with previously treated, locally advanced/metastatic breast cancer. This study (NCT02443428) prospectively observed eribulin-treated patients in real-world clinical practice. METHODS This observational multicentre registry study enrolled 76 patients with locally advanced/metastatic breast cancer who had ≤2 prior chemotherapeutic regimens for advanced disease. Eribulin was administered at a 1.23 mg/m2 dose (days 1 and 8 of every 21-day cycle). Adverse events (AEs) were monitored and effectiveness was assessed per local practice. RESULTS AEs occurred in 98.7% of patients; 88.2% had eribulin-related AEs. The most common AEs were fatigue (64.5%), alopecia (36.8%), nausea (35.5%) and constipation (30.3%). Serious AEs occurred in 42.1% of patients. The most common grade 3/4 AEs were neutropenia (9.2%), febrile neutropenia (9.2%), dyspnoea (5.3%) and pleural effusion (5.3%). No fatal AEs occurred. Dose reductions occurred in 31.6% of patients, 42.1% experienced dose delays and 9.2% discontinued due to worsening condition. There were complete responses in 2.6% and partial responses in 15.8% of patients. Median time to progression and overall survival were 4.0 and 8.3 months, respectively. CONCLUSION Eribulin was well tolerated in real-world clinical practice, comparable to safety and effectiveness reported in other clinical trials.
Collapse
Affiliation(s)
- Laura Kenny
- Imperial College LondonLondonUK,Imperial College Healthcare NHS TrustLondonUK
| | | | | | - Vivek Misra
- The Christie NHS Foundation TrustManchesterUK
| | | |
Collapse
|
50
|
Azumi M, Yoshie M, Takano W, Ishida A, Kusama K, Tamura K. The Impact of Eribulin on Stathmin Dynamics and Paclitaxel Sensitivity in Ovarian Cancer Cells. Biol Pharm Bull 2022; 45:1627-1635. [DOI: 10.1248/bpb.b22-00251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Mana Azumi
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences
| | - Mikihiro Yoshie
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences
| | - Wataru Takano
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences
| | - Akari Ishida
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences
| | - Kazuhiro Tamura
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|