1
|
Dekamin S, Ghasemi M, Dehpour AR, Ghazi-Khansari M, Shafaroodi H. Protective Effects of Glatiramer Acetate Against Paclitaxel-Induced Peripheral Neuropathy in Rats: A Role for Inflammatory Cytokines and Oxidative Stress. Neurochem Res 2024; 49:1049-1060. [PMID: 38252396 DOI: 10.1007/s11064-023-04088-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/04/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major challenge for cancer patients who undergo chemotherapy with paclitaxel. Therefore, finding effective therapies for CIPN is crucial. Glatiramer acetate is used to treat multiple sclerosis that exerts neuroprotective properties in various studies. We hypothesized that glatiramer acetate could also improve the paclitaxel-induced peripheral neuropathy. We used a rat model of paclitaxel (2 mg/kg/every other day for 7 doses)-induced peripheral neuropathy. Rats were treated with either different doses of glatiramer acetate (1, 2, 4 mg/kg/day) or its vehicle for 14 days in separate groups. The mechanical and thermal sensitivity of the rats by using the Von Frey test and the Hot Plate test, respectively, were assessed during the study. The levels of oxidative stress (malondialdehyde and superoxide dismutase), inflammatory markers (TNF-α, IL-10, NF-kB), and nerve damage (H&E and S100B staining) in the sciatic nerves of the rats were also measured at the end of study. Glatiramer acetate (2 and 4 mg/kg) exerted beneficial effects on thermal and mechanical allodynia tests. It also modulated the inflammatory response by reducing TNF-α and NF-κB levels, enhancing IL-10 production, and improving the oxidative stress status by lowering malondialdehyde and increasing superoxide dismutase activity in the sciatic nerve of the rats. Furthermore, glatiramer acetate enhanced nerve conduction velocity in all treatment groups. Histological analysis revealed that glatiramer acetate (2 and 4 mg/kg) prevented paclitaxel-induced damage to the nerve structure. These results suggest that glatiramer acetate can alleviate the peripheral neuropathy induced by paclitaxel.
Collapse
Affiliation(s)
- Sajad Dekamin
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Ghasemi
- Department of Neurology, Lahey Hospital and Medical Center, Burlington, MA, 01803, USA
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Ghazi-Khansari
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Shafaroodi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Tarasiuk O, Cavaletti G, Meregalli C. Clinical and preclinical features of eribulin-related peripheral neuropathy. Exp Neurol 2021; 348:113925. [PMID: 34801586 DOI: 10.1016/j.expneurol.2021.113925] [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: 09/02/2021] [Revised: 11/03/2021] [Accepted: 11/10/2021] [Indexed: 11/04/2022]
Abstract
Different microtubule-targeting agents (MTAs) possess distinct modes of action and their clinical use in cancer treatment is often limited by chemotherapy-induced peripheral neurotoxicity (CIPN). Eribulin is a member of the halichondrin class of antineoplastic drugs, which is correlated with a high antimitotic activity against metastatic breast cancer and liposarcoma. Current clinical evidence suggests that eribulin treatment, unlike some of the other MTAs, is associated with a relatively low incidence of severe peripheral neuropathy. This suggests that different MTAs possess unique mechanisms of neuropathologic induction. Animal models reliably reproduced eribulin-related neuropathy providing newer insights in CIPN pathogenesis, and they are highly suitable for in vivo functional, symptomatic and morphological characterizations of eribulin-related CIPN. The purpose of this review is to discuss the most recent literature on eribulin with a focus on both clinical and preclinical data, to explain the molecular events responsible for its favorable neurotoxic profile.
Collapse
Affiliation(s)
- Olga Tarasiuk
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| | - Guido Cavaletti
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy.
| | - Cristina Meregalli
- School of Medicine and Surgery, Experimental Neurology Unit and Milan Center for Neuroscience, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
3
|
Yuan P, Xu B. Clinical Utility of Eribulin Mesylate in the Treatment of Breast Cancer: A Chinese Perspective. BREAST CANCER-TARGETS AND THERAPY 2021; 13:135-150. [PMID: 33658845 PMCID: PMC7917473 DOI: 10.2147/bctt.s231298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/12/2021] [Indexed: 12/23/2022]
Abstract
Eribulin mesylate, a synthetic derivative of the anti-mitotic agent halichondrin B, has a unique tubulin-based mechanism of action that is distinct from other anti-microtubule agents including taxanes and vinca alkaloids. Consistent with this unique activity, eribulin has shown clinical efficacy in patients with metastatic breast cancer (MBC) that progressed following prior taxane and anthracycline therapy. The evidence presented in this review indicates that eribulin represents a treatment option for patients with HER2-negative metastatic breast cancer. Improved survival outcomes and better tolerability compared with vinorelbine supported the first approval of eribulin in China in 2019; eribulin was approved for women with locally advanced/metastatic HER2-negative breast cancer after treatment failure with at least two chemotherapy regimens, including an anthracycline and a taxane. Eribulin has also shown promising efficacy in patients with HER2-positive advanced breast cancer when used in combination with trastuzumab or pertuzumab, and subgroup analyses from the Phase III clinical trials support the continued evaluation of eribulin in patients with triple-negative disease. The unique non-mitotic effects of eribulin, including vascular remodeling, coupled with its clinical efficacy and safety profile, may permit the broader use of this agent in patients with MBC.
Collapse
Affiliation(s)
- Peng Yuan
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Binghe Xu
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| |
Collapse
|
4
|
First-line bevacizumab and eribulin combination therapy for HER2-negative metastatic breast cancer: Efficacy and safety in the GINECO phase II ESMERALDA study. Breast 2020; 54:256-263. [PMID: 33188992 PMCID: PMC7672314 DOI: 10.1016/j.breast.2020.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 09/20/2020] [Accepted: 09/25/2020] [Indexed: 01/17/2023] Open
Abstract
Purpose Combining bevacizumab with paclitaxel significantly improves progression-free survival (PFS) versus paclitaxel alone in HER2-negative metastatic breast cancer (MBC). Eribulin is active and tolerable in pretreated MBC. To assess whether eribulin may offer a more tolerable yet effective combination partner for bevacizumab, we evaluated a bevacizumab/eribulin combination regimen as first-line therapy for MBC. Methods In this single-arm phase II study, patients with histologically confirmed HER2-negative MBC and no prior chemotherapy for MBC received eribulin 1.23 mg/m2 on days 1 and 8 every 3 weeks for ≥6 cycles plus bevacizumab 15 mg/kg on day 1 every 3 weeks until disease progression. The primary endpoint was non-progression rate at 1 year. Secondary endpoints included objective response rate (ORR), PFS, and safety. Results The median age of the 61 treated female patients was 59 years, 16% had triple-negative MBC, 30% had ≥3 metastatic sites, and 71% had received prior (neo)adjuvant chemotherapy. Patients received a median of six eribulin and nine bevacizumab cycles. The non-progression rate at 1 year was 32% (95% confidence interval [CI]: 20–43%), ORR was 47% (95% CI: 34–60%), and median PFS was 8.3 months (95% CI: 7.0–9.6 months). The only grade ≥3 clinical adverse events in >5% of patients were hypertension (39%), neutropenia (26%), thrombosis (10%), and paresthesia/dysesthesia (7%). Conclusion First-line eribulin/bevacizumab combination therapy showed interesting activity in MBC with an acceptable safety profile, including a particularly low incidence of high-grade neuropathy. A single-arm study evaluated first-line bevacizumab–eribulin for HER2-negative MBC. The primary endpoint was non-progression rate at 1 year. The 1-year non-progression rate was 32% (95% CI 20–43%); median PFS was 8.3 months. Grade ≥3 clinical AEs in >10% comprised hypertension (39%) and neutropenia (26%). Eribulin–bevacizumab showed interesting activity and acceptable safety in MBC.
Collapse
|
5
|
Islam B, Lustberg M, Staff NP, Kolb N, Alberti P, Argyriou AA. Vinca alkaloids, thalidomide and eribulin-induced peripheral neurotoxicity: From pathogenesis to treatment. J Peripher Nerv Syst 2019; 24 Suppl 2:S63-S73. [PMID: 31647152 DOI: 10.1111/jns.12334] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/16/2019] [Indexed: 02/06/2023]
Abstract
Vinca alkaloids, thalidomide, and eribulin are widely used to treat patients with childhood acute lymphoblastic leukemia (ALL), adults affected by multiple myeloma and locally invasive or metastatic breast cancer, respectively. However, soon after their introduction into clinical practice, chemotherapy-induced peripheral neurotoxicity (CIPN) emerged as their main non-hematological and among dose-limiting adverse events. It is generally perceived that vinca alkaloids and the antiangiogenic agent thalidomide are more neurotoxic, compared to eribulin. The exposure to these chemotherapeutic agents is associated with an axonal, length-dependent, sensory polyneuropathy of mild to moderate severity, whereas it is considered that the peripheral nerve damage, unless severe, usually resolves soon after treatment discontinuation. Advanced age, high initial and prolonged dosing, coadministration of other neurotoxic chemotherapeutic agents and pre-existing neuropathy are the common risk factors. Pharmacogenetic biomarkers might be used to define patients at increased susceptibility of CIPN. Currently, there is no established therapy for CIPN prevention or treatment; symptomatic treatment for neuropathic pain and dose reduction or withdrawal in severe cases is considered, at the cost of reduced cancer therapeutic efficacy. This review critically examines the pathogenesis, epidemiology, risk factors (both clinical and pharmacogenetic), clinical phenotype and management of CIPN as a result of exposure to vinca alkaloids, thalidomide and its analogue lenalidomide as also eribulin.
Collapse
Affiliation(s)
- Badrul Islam
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Maryam Lustberg
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer, Columbus, Ohio
| | - Nathan P Staff
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Noah Kolb
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Andreas A Argyriou
- Department of Neurology, "Saint Andrew's" State General Hospital of Patras, Patras, Greece
| |
Collapse
|
6
|
Cortes J, Schöffski P, Littlefield BA. Multiple modes of action of eribulin mesylate: Emerging data and clinical implications. Cancer Treat Rev 2018; 70:190-198. [DOI: 10.1016/j.ctrv.2018.08.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
|
7
|
Cheng X, Li J, Tanaka K, Majumder U, Milinichik AZ, Verdi AC, Maddage CJ, Rybinski KA, Fernando S, Fernando D, Kuc M, Furuuchi K, Fang F, Uenaka T, Grasso L, Albone EF. MORAb-202, an Antibody-Drug Conjugate Utilizing Humanized Anti-human FRα Farletuzumab and the Microtubule-targeting Agent Eribulin, has Potent Antitumor Activity. Mol Cancer Ther 2018; 17:2665-2675. [PMID: 30262588 DOI: 10.1158/1535-7163.mct-17-1215] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/26/2018] [Accepted: 09/20/2018] [Indexed: 11/16/2022]
Abstract
Microtubule-targeting agents (MTA) have been investigated for many years as payloads for antibody-drug conjugates (ADC). In many cases, these ADCs have shown limited benefits due to lack of efficacy or significant toxicity, which has spurred continued investigation into novel MTA payloads for next-generation ADCs. In this study, we have developed ADCs using the MTA eribulin, a derivative of the macrocyclic polyether natural product halichondrin B, as a payload. Eribulin ADCs demonstrated in vitro potency and specificity using various linkers and two different conjugation approaches. MORAb-202 is an investigational agent that consists of the humanized anti-human folate receptor alpha (FRA) antibody farletuzumab conjugated via reduced interchain disulfide bonds to maleimido-PEG2-valine-citrulline-p-aminobenzylcarbamyl-eribulin at a drug-to-antibody ratio of 4.0. MORAb-202 displayed preferable biophysical properties and broad potency across a number of FRA-positive tumor cell lines as well as demonstrated improved specificity in vitro compared with farletuzumab conjugated with a number of other MTA payloads, including MMAE, MMAF, and the reducible maytansine linker-payload sulfo-SPDB-DM4. A single-dose administration of MORAb-202 in FRA-positive human tumor cell line xenograft and patient-derived tumor xenograft models elicited a robust and durable antitumor response. These data support further investigation of MORAb-202 as a potential new treatment modality for FRA-positive cancers, using the novel MTA eribulin as a payload.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Biochemistry Discovery, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Jing Li
- Department of Translational Chemistry, Eisai AiM Institute, Eisai Inc., Andover, Massachusetts
| | - Keigo Tanaka
- Department of Chemistry Research, Eisai Co. Ltd., Tsukuba-Shi, Ibaraki, Japan
| | - Utpal Majumder
- Department of Discovery Chemistry, Eisai AiM Institute, Eisai Inc., Andover, Massachusetts
| | - Andrew Z Milinichik
- Department of Biochemistry Discovery, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Arielle C Verdi
- Department of Biochemistry Discovery, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Christopher J Maddage
- Department of Preclinical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Katherine A Rybinski
- Department of Preclinical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Shawn Fernando
- Department of Bioanalytical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Danielle Fernando
- Department of Bioanalytical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Megan Kuc
- Department of Bioanalytical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Keiji Furuuchi
- Department of Preclinical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Frank Fang
- Department of Translational Chemistry, Eisai AiM Institute, Eisai Inc., Andover, Massachusetts
| | - Toshimitsu Uenaka
- Department of Preclinical Development, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania
| | - Luigi Grasso
- Department of Discovery Research, Morphotek Inc., Exton, Pennsylvania
| | - Earl F Albone
- Department of Biochemistry Discovery, Oncology Biologics Laboratories, Oncology Business Group, Eisai Inc, Exton, Pennsylvania.
| |
Collapse
|
8
|
Chine VB, Au NPB, Kumar G, Ma CHE. Targeting Axon Integrity to Prevent Chemotherapy-Induced Peripheral Neuropathy. Mol Neurobiol 2018; 56:3244-3259. [DOI: 10.1007/s12035-018-1301-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
|
9
|
Cook BM, Wozniak KM, Proctor DA, Bromberg RB, Wu Y, Slusher BS, Littlefield BA, Jordan MA, Wilson L, Feinstein SC. Differential Morphological and Biochemical Recovery from Chemotherapy-Induced Peripheral Neuropathy Following Paclitaxel, Ixabepilone, or Eribulin Treatment in Mouse Sciatic Nerves. Neurotox Res 2018; 34:677-692. [PMID: 30051419 DOI: 10.1007/s12640-018-9929-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 12/29/2022]
Abstract
The reversibility of chemotherapy-induced peripheral neuropathy (CIPN), a disabling and potentially permanent side effect of microtubule-targeting agents (MTAs), is becoming an increasingly important issue as treatment outcomes improve. The molecular mechanisms regulating the variability in time to onset, severity, and time to recovery from CIPN between the common MTAs paclitaxel and eribulin are unknown. Previously (Benbow et al. in Neurotox Res 29:299-313, 2016), we found that after 2 weeks of a maximum tolerated dose (MTD) in mice, paclitaxel treatment resulted in severe reductions in axon area density, higher frequency of myelin abnormalities, and increased numbers of Schwann cell nuclei in sciatic nerves. Biochemically, eribulin induced greater microtubule-stabilizing effects than paclitaxel. Here, we extended these comparative MTD studies to assess the recovery from these short-term effects of paclitaxel, eribulin, and a third MTA, ixabepilone, over the course of 6 months. Paclitaxel induced a persistent reduction in axon area density over the entire 6-month recovery period, unlike ixabepilone- or eribulin-treated animals. The abundance of myelin abnormalities rapidly declined after cessation of all drugs but recovered most slowly after paclitaxel treatment. Paclitaxel- and ixabepilone- but not eribulin-treated animals exhibited increased Schwann cell numbers during the recovery period. Tubulin composition and biochemistry rapidly returned from MTD-induced levels of α-tubulin, acetylated α-tubulin, and end-binding protein 1 to control levels following cessation of drug treatment. Taken together, sciatic nerve axons recovered more rapidly from morphological effects in eribulin- and ixabepilone-treated animals than in paclitaxel-treated animals and drug-induced increases in protein expression levels following paclitaxel and eribulin treatment were relatively transient.
Collapse
Affiliation(s)
- B M Cook
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,Department of Biomolecular Sciences and Engineering, University of California Santa Barbara, Santa Barbara, CA, 93016, USA
| | - K M Wozniak
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - D A Proctor
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93016, USA
| | - R B Bromberg
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93016, USA
| | - Y Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - B S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - B A Littlefield
- Scientific Administration, Eisai Research Institute, Andover, MA, 01810, USA
| | - M A Jordan
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93016, USA
| | - L Wilson
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93016, USA
| | - Stuart C Feinstein
- Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, CA, 93106, USA. .,Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93016, USA.
| |
Collapse
|
10
|
Wozniak KM, Vornov JJ, Wu Y, Liu Y, Carozzi VA, Rodriguez-Menendez V, Ballarini E, Alberti P, Pozzi E, Semperboni S, Cook BM, Littlefield BA, Nomoto K, Condon K, Eckley S, DesJardins C, Wilson L, Jordan MA, Feinstein SC, Cavaletti G, Polydefkis M, Slusher BS. Peripheral Neuropathy Induced by Microtubule-Targeted Chemotherapies: Insights into Acute Injury and Long-term Recovery. Cancer Res 2017; 78:817-829. [PMID: 29191802 DOI: 10.1158/0008-5472.can-17-1467] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/30/2017] [Accepted: 11/21/2017] [Indexed: 01/01/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major cause of disability in cancer survivors. CIPN investigations in preclinical model systems have focused on either behaviors or acute changes in nerve conduction velocity (NCV) and amplitude, but greater understanding of the underlying nature of axonal injury and its long-term processes is needed as cancer patients live longer. In this study, we used multiple independent endpoints to systematically characterize CIPN recovery in mice exposed to the antitubulin cancer drugs eribulin, ixabepilone, paclitaxel, or vinorelbine at MTDs. All of the drugs ablated intraepidermal nerve fibers and produced axonopathy, with a secondary disruption in myelin structure within 2 weeks of drug administration. In addition, all of the drugs reduced sensory NCV and amplitude, with greater deficits after paclitaxel and lesser deficits after ixabepilone. These effects correlated with degeneration in dorsal root ganglia (DRG) and sciatic nerve and abundance of Schwann cells. Although most injuries were fully reversible after 3-6 months after administration of eribulin, vinorelbine, and ixabepilone, we observed delayed recovery after paclitaxel that produced a more severe, pervasive, and prolonged neurotoxicity. Compared with other agents, paclitaxel also displayed a unique prolonged exposure in sciatic nerve and DRG. The most sensitive indicator of toxicity was axonopathy and secondary myelin changes accompanied by a reduction in intraepidermal nerve fiber density. Taken together, our findings suggest that intraepidermal nerve fiber density and changes in NCV and amplitude might provide measures of axonal injury to guide clinical practice.Significance: This detailed preclinical study of the long-term effects of widely used antitubulin cancer drugs on the peripheral nervous system may help guide clinical evaluations to improve personalized care in limiting neurotoxicity in cancer survivors. Cancer Res; 78(3); 817-29. ©2017 AACR.
Collapse
Affiliation(s)
- Krystyna M Wozniak
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ying Liu
- Department of Neurology and the Cutaneous Nerve Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Valentina A Carozzi
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Virginia Rodriguez-Menendez
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Elisa Ballarini
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Eleonora Pozzi
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Sara Semperboni
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Brett M Cook
- Neurosci Research Institute, University of California, Santa Barbara, California.,Biomolecular Science and Engineering Program, University of California, Santa Barbara, California
| | | | | | | | | | | | - Leslie Wilson
- Neurosci Research Institute, University of California, Santa Barbara, California.,Biomolecular Science and Engineering Program, University of California, Santa Barbara, California.,Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California
| | - Mary A Jordan
- Neurosci Research Institute, University of California, Santa Barbara, California.,Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California
| | - Stuart C Feinstein
- Neurosci Research Institute, University of California, Santa Barbara, California.,Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California
| | - Guido Cavaletti
- Experimental Neurology Unit and PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Michael Polydefkis
- Department of Neurology and the Cutaneous Nerve Laboratory, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery and Departments of Neurology, Psychiatry, Neuroscience, Medicine and Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
11
|
Garrone O, Miraglio E, Vandone AM, Vanella P, Lingua D, Merlano MC. Eribulin in advanced breast cancer: safety, efficacy and new perspectives. Future Oncol 2017; 13:2759-2769. [PMID: 29219017 DOI: 10.2217/fon-2017-0283] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Eribulin is a synthetic analog of halichondrin B belonging to microtubule-targeted agents with a distinct mechanism of inhibition of microtubule dynamics. This molecule has multiple nonmitotic effects on tumor biology, exhibiting effects on epithelial-mesenchimal transition and tumor vasculature. We review here preclinical and clinical studies on eribulin. The mitotic and nonmitotic effects together with its favorable safety profile make eribulin a unique drug with high potential in the treatment of metastatic breast cancer. The new emphasis of eribulin mechanism of action on vascular remodeling, microenvironment modifications and reversal of epithelial-mesenchimal transition paves the way to rethinking the use of the drug in an immunological perspective.
Collapse
Affiliation(s)
- Ornella Garrone
- Medical Oncology A.O. S. Croce e Carle Ospedale di Insegnamento, Cuneo, Italy
| | - Emanuela Miraglio
- Medical Oncology A.O. S. Croce e Carle Ospedale di Insegnamento, Cuneo, Italy
| | - Anna Maria Vandone
- Medical Oncology A.O. S. Croce e Carle Ospedale di Insegnamento, Cuneo, Italy
| | - Paola Vanella
- Medical Oncology A.O. S. Croce e Carle Ospedale di Insegnamento, Cuneo, Italy
| | - Daniele Lingua
- Medical Oncology A.O. S. Croce e Carle Ospedale di Insegnamento, Cuneo, Italy
| | - Marco C Merlano
- Medical Oncology A.O. S. Croce e Carle Ospedale di Insegnamento, Cuneo, Italy
| |
Collapse
|
12
|
|
13
|
Park YH, Kim TY, Im YH, Lee KS, Park IH, Sohn J, Lee SH, Im SA, Kim JH, Kim SH, Lee SJ, Koh SJ, Lee KH, Choi YJ, Cho EK, Lee S, Kang SY, Seo JH, Kim SB, Jung KH. Feasibility and Efficacy of Eribulin Mesilate in Korean Patients with Metastatic Breast Cancer: Korean Multi-center Phase IV Clinical Study Results. Cancer Res Treat 2016; 49:423-429. [PMID: 27488876 PMCID: PMC5398406 DOI: 10.4143/crt.2016.191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/13/2016] [Indexed: 01/26/2023] Open
Abstract
PURPOSE Eribulin mesilate was approved for the treatment of patients with locally advanced or metastatic breast cancer (MBC), who had received at least two chemotherapeutic regimens, including anthracycline and taxane. On the other hand, the efficacy and safety information of eribulin in Korean patients is limited by the lack of clinical trials. MATERIALS AND METHODS In this multicenter, open-label, single-arm, phase IV study, locally advanced or MBC patients were enrolled between June 2013 and April 2014 from 14 centers in Korea. One point four mg/m2 dose of eribulin was administered on days 1 and 8 of every 21 days. The primary endpoint was the frequency and intensity of the treatment emergent adverse event. The secondary endpoint was the disease control rate, which included the rate of complete responses, partial responses, and stable disease. RESULTS A total of 101 patients received at least one dose of eribulin and were included in the safety set. The patients received a total of 543 treatment cycles, with a median of three cycles (range, 1 to 31 cycles). The most common adverse event was neutropenia (91.1% of patients, 48.3% of cycles). The frequent non-hematological adverse events included alopecia, decrease in appetite, fatigue/asthenia, and myalgia/arthralgia. The peripheral neuropathy of any grade occurred in 27 patients (26.7%), including grade 3 in two patients. Disease control rate was 52.7% and 51.3% of patients in the full analysis set and per-protocol set, respectively. CONCLUSION This study demonstrated the feasible safety profile and activity of eribulin in Korean patients with MBC.
Collapse
Affiliation(s)
- Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Yong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Young-Hyuck Im
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keun-Seok Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, National Cancer Center, Goyang, Korea
| | - In Hae Park
- Division of Hematology and Medical Oncology, Department of Internal Medicine, National Cancer Center, Goyang, Korea
| | - Joohyuk Sohn
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Soo-Hyeon Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Se Hyun Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Soo Jung Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Kyungpook National University Medical Center, Kyungpook National University School of Medicine, Daegu, Korea
| | - Su-Jin Koh
- Division of Medical Oncology, Department of Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Ki Hyeong Lee
- Division of Hematology-Oncology, Department of Medicine, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Yoon Ji Choi
- Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Eun Kyung Cho
- Division of Hematology and Oncology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Suee Lee
- Department of Internal Medicine, Dong-A University Medical Center, Busan, Korea
| | - Seok Yun Kang
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Ajou University School of Medicine, Suwon, Korea
| | - Jae Hong Seo
- Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Wozniak KM, Vornov JJ, Wu Y, Nomoto K, Littlefield BA, DesJardins C, Yu Y, Lai G, Reyderman L, Wong N, Slusher BS. Sustained Accumulation of Microtubule-Binding Chemotherapy Drugs in the Peripheral Nervous System: Correlations with Time Course and Neurotoxic Severity. Cancer Res 2016; 76:3332-9. [PMID: 27197173 DOI: 10.1158/0008-5472.can-15-2525] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 04/03/2016] [Indexed: 12/13/2022]
Abstract
Chemotherapy-induced peripheral neuropathy is a dose-limiting side effect of many antineoplastic agents, but the mechanisms underlying the toxicities are unclear. At their MTDs, the microtubule-binding drugs paclitaxel and ixabepilone induce more severe neuropathy in mice relative to eribulin mesylate, paralleling their toxicity profiles in clinic. We hypothesized that the severity of their neurotoxic effects might be explained by the levels at which they accumulate in the peripheral nervous system. To test this hypothesis, we compared their pharmacokinetics and distribution in peripheral nerve tissue. After administration of a single intravenous dose, each drug was rapidly cleared from plasma but all persisted in the dorsal root ganglia (DRG) and sciatic nerve (SN) for up to 72 hours. Focusing on paclitaxel and eribulin, we performed a 2-week MTD-dosing regimen, followed by a determination of drug pharmacokinetics, tissue distribution, and multiple functional measures of peripheral nerve toxicity for 4 weeks. Consistent with the acute dosing study, both drugs persisted in peripheral nervous tissues for weeks, in contrast to their rapid clearance from plasma. Notably, although eribulin exhibited greater DRG and SN penetration than paclitaxel, the neurotoxicity observed functionally was consistently more severe with paclitaxel. Overall, our results argue that sustained exposure of microtubule-binding chemotherapeutic agents in peripheral nerve tissues cannot by itself account for their associated neurotoxicity. Cancer Res; 76(11); 3332-9. ©2016 AACR.
Collapse
Affiliation(s)
- Krystyna M Wozniak
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | | | - Yanke Yu
- Eisai Inc., Andover, Massachusetts
| | | | | | | | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Neurology, Medicine Psychiatry, and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
15
|
Smith JA, Benbow SJ. Meeting Report: Inaugural Chemotherapy-Induced Peripheral Neuropathy Symposium - Santa Barbara, CA, February 2015. Cancer Res 2015; 75:3696-8. [PMID: 26081810 DOI: 10.1158/0008-5472.can-15-1145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Chemotherapy-induced peripheral neuropathy is a common, dose-limiting side effect of cancer treatment. This conference was the first of its kind to bring together a wide range of clinicians, researchers, and industry professionals to address the potential causes, preventions, and treatments for this drug toxicity. Intraepidermal nerve fiber loss, axonal degeneration, immune cell infiltration, alterations in tubulin protein expression and microtubule stability, axonal transport, and mitochondrial dysfunction were addressed as possible mechanisms. Problems with animal models of the disease were discussed, as well as the potential of patient-derived induced sensory neurons to serve as a novel in vitro model.
Collapse
Affiliation(s)
- Jennifer A Smith
- Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara
| | - Sarah J Benbow
- Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara
| |
Collapse
|
16
|
Swami U, Shah U, Goel S. Eribulin in Cancer Treatment. Mar Drugs 2015; 13:5016-58. [PMID: 26262627 PMCID: PMC4557013 DOI: 10.3390/md13085016] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/18/2015] [Accepted: 07/24/2015] [Indexed: 11/20/2022] Open
Abstract
Halichondrin B is a complex, natural, polyether macrolide derived from marine sponges. Eribulin is a structurally-simplified, synthetic, macrocyclic ketone analogue of Halichondrin B. Eribulin was approved by United States Food and Drug Administration in 2010 as a third-line therapy for metastatic breast cancer patients who have previously been treated with an anthracycline and a taxane. It has a unique microtubule dynamics inhibitory action. Phase III studies have either been completed or are currently ongoing in breast cancer, soft tissue sarcoma, and non-small cell lung cancer. Phase I and II studies in multiple cancers and various combinations are currently ongoing. This article reviews the available information on eribulin with respect to its clinical pharmacology, pharmacokinetics, pharmacodynamics, mechanism of action, metabolism, preclinical studies, and with special focus on clinical trials.
Collapse
Affiliation(s)
- Umang Swami
- University of Iowa Hospitals and Clinics, 200 Hawkins Dr, Iowa City, IA 52242, USA.
| | - Umang Shah
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, Bronx, NY 10461, USA.
| | - Sanjay Goel
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, Bronx, NY 10461, USA.
| |
Collapse
|
17
|
Scavelli C, Gallù F. Long-term efficacy and safety of a third-line treatment with eribulin plus trastuzumab in a young breast cancer patient. Future Oncol 2015; 10:2127-32. [PMID: 25471027 DOI: 10.2217/fon.14.134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Eribulin mesylate is approved for the treatment of metastatic breast cancer (MBC) patients after progression with anthracyclines and taxanes. Eribulin appears especially promising when combined with trastuzumab, according to the results of a recent Phase II trial in first-line setting. Here we report the case of a young, pretreated, HER2(-) MBC patient, who achieved a long-term clinical benefit with eribulin alone and in combination with trastuzumab after re-biopsy on liver metastases showed HER2 amplification. Although it is unique for its evolving clinical/biomolecular picture, this case adds anecdotal evidence to the efficacy and tolerability of this combination. However, Phase III trials are warranted to confirm its potential in first and subsequent lines of MBC treatment.
Collapse
Affiliation(s)
- Claudio Scavelli
- Medical Oncology Unit, 'S Cuore di Gesù' Hospital, SP Alezio-Gallipoli, I-73014 Gallipoli, LE, Italy
| | | |
Collapse
|
18
|
Dybdal-Hargreaves NF, Risinger AL, Mooberry SL. Eribulin mesylate: mechanism of action of a unique microtubule-targeting agent. Clin Cancer Res 2015; 21:2445-52. [PMID: 25838395 DOI: 10.1158/1078-0432.ccr-14-3252] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 02/05/2015] [Indexed: 01/22/2023]
Abstract
Eribulin mesylate (eribulin), an analogue of the marine natural product halichondrin B, is a microtubule-depolymerizing drug that has utility in the treatment of patients with breast cancer. Clinical trial results have demonstrated that eribulin treatment provides a survival advantage to patients with metastatic or locally advanced breast cancer previously treated with an anthracycline and a taxane. Furthermore, a pooled analysis of two pivotal phase III trials has demonstrated that eribulin also improves overall survival in several patient subgroups, including in women with HER2-negative disease and triple-negative breast cancer. This review covers the preclinical research that led to the clinical testing and approval of eribulin, as well as subsequent research that was prompted by distinct and unexpected effects of eribulin in the clinic. Initial studies with halichondrin B demonstrated unique effects on tubulin binding that resulted in distinct microtubule-dependent events and antitumor actions. Consistent with the actions of the natural product, eribulin has potent microtubule-depolymerizing activities and properties that distinguish it from other microtubule-targeting agents. Here, we review new results that further differentiate the effects of eribulin from other agents on peripheral nerves, angiogenesis, vascular remodeling, and epithelial-to-mesenchymal transition. Together, these data highlight the distinct properties of eribulin and begin to delineate the mechanisms behind the increased survival benefit provided by eribulin for patients.
Collapse
Affiliation(s)
| | - April L Risinger
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas. Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Susan L Mooberry
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas. Cancer Therapy & Research Center, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
19
|
Rivera E, Cianfrocca M. Overview of neuropathy associated with taxanes for the treatment of metastatic breast cancer. Cancer Chemother Pharmacol 2015; 75:659-70. [PMID: 25596818 PMCID: PMC4365177 DOI: 10.1007/s00280-014-2607-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 10/07/2014] [Indexed: 12/26/2022]
Abstract
Taxanes are an established option in the standard treatment paradigm for patients with metastatic breast cancer (MBC). Neuropathy is a common, dose-limiting side effect of taxane therapy that is often managed by dose reductions and delays. The severity, time to onset, and improvement in neuropathy are important considerations for patient management and vary among currently approved taxanes. The rate of grade ≥3 neuropathy with taxanes has been shown to be dose and schedule dependent; however, time to improvement to grade ≤1 is typically shorter for nab-paclitaxel than for other taxanes in patients with MBC. Many tools for assessing patient-reported neuropathy exist. Because MBC is incurable and patient quality of life must be critically considered when making treatment decisions, there is a need for more prospective trials to assess patient-reported neuropathy. Validated predictors of taxane-related neuropathy may play an important role in treatment decisions in the future. This review will focus on the toxicity profile (i.e., neuropathy) of each of the taxanes used in the treatment of MBC, will provide updates on tools used for the assessment of neuropathy, and will highlight newly discovered predictors of taxane-related neuropathy.
Collapse
Affiliation(s)
- Edgardo Rivera
- Banner MD Anderson Cancer Center, 2946 E. Banner Gateway Drive, Gilbert, AZ, 85234, USA,
| | | |
Collapse
|
20
|
Doherty MK, Morris PG. Eribulin for the treatment of metastatic breast cancer: an update on its safety and efficacy. Int J Womens Health 2015; 7:47-58. [PMID: 25610001 PMCID: PMC4294649 DOI: 10.2147/ijwh.s74462] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Breast cancer remains a leading cause of cancer-related death internationally. Treatment approaches for metastatic breast cancer have evolved in recent years; however chemotherapy remains a core component for the majority of patients. Agents such as anthracyclines and taxanes have been extensively studied and form standard treatment. Eribulin mesylate is a novel synthetic microtubule-directed chemotherapy, based on a naturally-occurring compound. Through phase I studies, eribulin was found to be tolerable and activity was seen in patients with metastatic breast cancer. Phase II studies in metastatic breast cancer further demonstrated its efficacy, with responses and survival which compare favorably with other studied chemotherapy agents. The phase III EMBRACE study showed superior survival for patients treated with eribulin compared with those who received a physician's choice control. This led to its approval for use in many countries in this setting. Its toxicity profile is well established and manageable for the most part, with the commonest reported toxicities being alopecia, fatigue, neutropenia and peripheral neuropathy. A second reported phase III study comparing eribulin to capecitabine failed to show an improvement in survival in pretreated patients. This article reviews the clinical pharmacology and mechanism of action of eribulin, and summarizes the results of the major preclinical and clinical studies of eribulin in metastatic breast cancer.
Collapse
Affiliation(s)
- Mark K Doherty
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|