1
|
Liu Y, Chen X, Evan T, Esapa B, Chenoweth A, Cheung A, Karagiannis SN. Folate receptor alpha for cancer therapy: an antibody and antibody-drug conjugate target coming of age. MAbs 2025; 17:2470309. [PMID: 40045156 PMCID: PMC11901361 DOI: 10.1080/19420862.2025.2470309] [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: 12/31/2024] [Revised: 02/15/2025] [Accepted: 02/17/2025] [Indexed: 03/14/2025] Open
Abstract
Folate receptor alpha (FRα) has long been the focus of therapeutics development in oncology across several solid tumors, notably ovarian, lung, and subsets of breast cancers. Its multiple roles in cellular metabolism and carcinogenesis and tumor-specific overexpression relative to normal tissues render FRα an attractive target for biological therapies. Here we review the biological significance, expression distribution, and characteristics of FRα as a highly promising and now established therapy target. We discuss the ongoing development of FRα-targeting antibodies and antibody-drug conjugates (ADCs), the first of which has been approved for the treatment of ovarian cancer, providing the impetus for heightened research and therapy development. Novel insights into the tumor microenvironment, advances in antibody engineering to enhance immune-mediated effects, the emergence of ADCs, and several studies of anti-FRα agents combined with chemotherapy, targeted and immune therapy are offering new perspectives and treatment possibilities. Hence, we highlight key translational research and discuss several preclinical studies and clinical trials of interest, with an emphasis on agents and therapy combinations with potential to change future clinical practice.
Collapse
Affiliation(s)
- Yi Liu
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
| | - Xinyi Chen
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
| | - Theodore Evan
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
| | - Benjamina Esapa
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
| | - Alicia Chenoweth
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, Innovation Hub, Guy’s Hospital, London, UK
| | - Anthony Cheung
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, Innovation Hub, Guy’s Hospital, London, UK
| | - Sophia N Karagiannis
- St. John’s Institute of Dermatology, School of Basic & Medical Biosciences & KHP Centre for Translational Medicine, King’s College London, Guy’s Hospital, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King’s College London, Guy’s Cancer Centre, Innovation Hub, Guy’s Hospital, London, UK
| |
Collapse
|
2
|
Passildas J, Paillard MJ, Uwer L, Molnar I, Dohollou N, Petit T, Hajjaji N, Boudin L, Lorgis V, Jacquin JP, Abrial C, Mouret-Reynier MA. Eribulin efficacy in long responder patients with metastatic breast cancer: A multicentric observational study. Cancer Epidemiol 2025; 96:102800. [PMID: 40090228 DOI: 10.1016/j.canep.2025.102800] [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: 11/20/2024] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Eribulin can represent a therapeutic alternative for patients with advanced breast cancer who have received at least one or two lines of anthracyclines-based chemotherapy and taxane therapy. In this observational study, we focused on long-responder patients, i.e. with an objective response or stability ≥ 6 months under eribulin to better characterize them. METHODS Metastatic breast cancer (MBC) patients treated by eribulin in 2nd, 3rd or 4th line between September 2011 and June-2018 were included. The following parameters were assessed: primary tumor and metastasis characteristics, type of response and duration, disease progression, treatment received, toxicities, progression free survival (PFS), overall survival (OS), and prognostic factors of OS and PFS. Special attention was paid to patients with hepatic disease (HD). RESULTS Among the 98 patients included, an analysis was conducted on 84 patients (median age 62). Median duration of response was 25.6 weeks (95 IC 22-27.7) with a median number of infusions of 6. Response was similar, irrespective of ERI line number. HD was observed in 70.2 % of patients. Median PFS was 9 months (95 %CI 8-10). Subgroup analysis showed similar PFS, irrespective of HD (p = 0.21) and treatment line (p = 0.46). Median OS was 24 months. (95 % IC 20-31). The main prognostic factors of OS were duration of response (p < 0.001) and, progesterone receptor positiveness was associated to PFS (p = 0.006). CONCLUSION This multicentric, retrospective study highlights eribulin as a potential second-line therapy for MBC with a median response duration of 25 weeks after 6 infusions. The safety and efficacy profiles align with previous studies, supporting its role as a viable treatment option. Notably, the response and PFS were independent of hepatic metastasis, suggesting benefit across various MBC subtypes, including those with liver involvement. However, the retrospective design warrants cautious interpretation, and further prospective studies are needed to confirm these findings and optimize eribulin's use, potentially through molecular profiling for personalized treatment strategies.
Collapse
Affiliation(s)
- J Passildas
- Division de Recherche Clinique, Délégation Recherche Clinique & Innovation, Centre Jean Perrin, Centre de Lutte contre le Cancer, 58 rue Montalembert, F-63000, Clermont-Ferrand, France; Centre d'Investigation Clinique, UMR501, Clermont-Ferrand 63011, France; Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand F-63000, France.
| | - M J Paillard
- Medical Oncology, CHRU Besancon - Hopital Jean Minjoz, Besançon, France.
| | - L Uwer
- Medical Oncology, Institut de Cancérologie de Lorraine - Alexis Vautrin, Vandoeuvre Les Nancy, France.
| | - I Molnar
- Division de Recherche Clinique, Délégation Recherche Clinique & Innovation, Centre Jean Perrin, Centre de Lutte contre le Cancer, 58 rue Montalembert, F-63000, Clermont-Ferrand, France; Centre d'Investigation Clinique, UMR501, Clermont-Ferrand 63011, France; Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand F-63000, France.
| | - N Dohollou
- Oncology, Polyclinique Bordeaux Nord Aquitaine, Bordeaux, France.
| | - T Petit
- Medical Oncology, Centre Paul Strauss Centre de Lutte contre le Cancer, Strasbourg, France.
| | - N Hajjaji
- Medical oncology, Centre Oscar Lambret, Lille, France.
| | - L Boudin
- Medical Oncology, Hôpital d'Instruction des Armées (HIA) Ste Anne, Toulon, France.
| | - V Lorgis
- Oncologue médical, Institut Cancérologie de Bourgogne, Dijon, France.
| | - J P Jacquin
- Medical Oncology, Institut de Cancérologie Lucien Neuwirth, Saint-Étienne, France.
| | - C Abrial
- Division de Recherche Clinique, Délégation Recherche Clinique & Innovation, Centre Jean Perrin, Centre de Lutte contre le Cancer, 58 rue Montalembert, F-63000, Clermont-Ferrand, France; Centre d'Investigation Clinique, UMR501, Clermont-Ferrand 63011, France; Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont-Ferrand F-63000, France.
| | - M A Mouret-Reynier
- Medical Oncology, Jean Perrin, Centre de Lutte contre le Cancer, 58 rue Montalembert, Clermont-Ferrand F-63000, France.
| |
Collapse
|
3
|
Chen YC, Takada M, Nagornyuk A, Yu M, Yamada H, Nagashima T, Ohtsuka M, DeLuca JG, Markus SM, Takaku M, Suzuki A. Inhibition of p38-MK2 pathway enhances the efficacy of microtubule inhibitors in breast cancer cells. eLife 2025; 13:RP104859. [PMID: 40439108 PMCID: PMC12122001 DOI: 10.7554/elife.104859] [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] [Indexed: 06/02/2025] Open
Abstract
Microtubule-targeting agents (MTAs) are widely used as first- and second-line chemotherapies for various cancers. However, current MTAs exhibit positive responses only in subsets of patients and are often accompanied by side effects due to their impact on normal cells. This underscores an urgent need to develop novel therapeutic strategies that enhance MTA efficacy while minimizing toxicity to normal tissues. Here, we demonstrate that inhibition of the p38 MAPK-MK2 signaling pathway sensitizes cancer cells to MTA treatment. We utilize CMPD1, a dual-target inhibitor, to concurrently suppress the p38-MK2 pathway and microtubule dynamicity. In addition to its established role as an MK2 inhibitor, we find that CMPD1 rapidly induces microtubule depolymerization, preferentially at the microtubule plus end, leading to the inhibition of tumor growth and cancer cell invasion in both in vitro and in vivo models. Notably, 10 nM CMPD1 is sufficient to induce irreversible mitotic defects in cancer cells, but not in non-transformed normal cells, highlighting its high specificity to cancer cells. We further validate that a specific p38-MK2 inhibitor significantly potentiates the efficacy of subclinical concentrations of MTA. In summary, our findings suggest that the p38-MK2 pathway presents a promising therapeutic target in combination with MTAs in cancer treatment.
Collapse
Affiliation(s)
- Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
| | - Mamoru Takada
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Aerica Nagornyuk
- Department of Biomedical Science, University of North Dakota School of Medicine and Health ScienceGrand ForksUnited States
| | - Muhan Yu
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Hideyuki Yamada
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Takeshi Nagashima
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State UniversityFort CollinsUnited States
| | - Steven M Markus
- Department of Biochemistry and Molecular Biology, Colorado State UniversityFort CollinsUnited States
| | - Motoki Takaku
- Department of Biomedical Science, University of North Dakota School of Medicine and Health ScienceGrand ForksUnited States
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-MadisonMadisonUnited States
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-MadisonMadisonUnited States
- Carbone Comprehensive Cancer Center, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
4
|
Agarwala KL, Kubara K, Seletsky BM, Sagane K, Littlefield BA. Eribulin's exclusive binding to microtubule plus ends results from discrimination between GTP and GDP forms of β-tubulin. Arch Biochem Biophys 2025:110482. [PMID: 40449645 DOI: 10.1016/j.abb.2025.110482] [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: 02/21/2025] [Revised: 05/12/2025] [Accepted: 05/28/2025] [Indexed: 06/03/2025]
Abstract
The clinically approved anticancer agent eribulin (Halaven®) exerts cytotoxic antimitotic effects by binding to high affinity sites on exposed plus ends of growing microtubules (MT). Despite X-ray crystallographic mapping of eribulin's binding pocket within β-tubulin's vinca domain, the biophysical basis for eribulin's MT plus end binding exclusivity remains unknown. We performed surface plasmon resonance (SPR) studies of tubulin binding to biotinylated eribulin probes to ask if eribulin discriminates between GTP and GDP forms of β-tubulin, which characterize growing MT plus ends and MT sides, respectively. Tubulin binding to biotin-eribulin proceeded via a single state binding reaction, while binding to biotin-vinblastine occurred via a two-state reaction incorporating a conformational change. Biochemical approaches confirmed tubulin conformational changes induced by vinblastine but not eribulin. SPR competition studies using free eribulin and vinblastine confirmed tubulin binding specificity to cognate biotinylated probes, showing that eribulin binding within the β-tubulin vinca domain is physically and functionally distinct from vinblastine. SPR studies using tubulin containing only GTP or GDP forms of β-tubulin showed that while biotin-eribulin has only slightly higher overall affinity for GTP-tubulin, dissociation from GTP-tubulin was ∼7-fold slower than from GDP-tubulin, establishing that eribulin discriminates between GTP and GDP forms of β-tubulin. In contrast, vinblastine fails to discriminate between these two tubulin forms, consistent with its known binding to both MT ends and sides. Overall, our results establish, for the first time, the biophysical basis for eribulin's MT plus end binding exclusivity as resulting from the ability to discriminate between GTP and GDP forms of β-tubulin.
Collapse
Affiliation(s)
- Kishan Lal Agarwala
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Kenji Kubara
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Boris M Seletsky
- Eisai Inc., 35 Cambridgepark Drive, Cambridge, Massachusetts 02140, USA
| | - Koji Sagane
- Tsukuba Research Laboratories, Eisai Co., Ltd., 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | | |
Collapse
|
5
|
Dickerson JC, Moen MT, Nielsen P, Riaz F, Tran E, Caswell-Jin JL, Suen W, Goldhaber-Fiebert JD, Alarid-Escudero F. Cost and Cost-Effectiveness of Treating Human Epidermal Growth Factor Receptor 2-Low Metastatic Breast Cancer. J Clin Oncol 2025:JCO2401960. [PMID: 40397834 DOI: 10.1200/jco-24-01960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 04/01/2025] [Accepted: 04/21/2025] [Indexed: 05/23/2025] Open
Abstract
PURPOSE Creating value-aligned treatment pathways in breast cancer requires understanding the cost and cost-effectiveness of new therapies. To address uncertainty in the optimal treatment sequence, we developed a decision model to assess the cost-effectiveness of various treatment sequences for patients with human epidermal growth factor receptor 2 (HER2)-low metastatic breast cancer who are eligible for trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG) under current US Food and Drug Administration labeling. METHODS We derived disease progression and therapy data from the Destiny-Breast04 trial and sourced cost and quality-of-life data from the published literature. Our simulation modeled 57-year-old women with HER2-low, endocrine refractory, and triple-negative metastatic breast cancer eligible for third-line treatment. We evaluated four sequences: chemotherapy (chemo) → chemo, T-DXd → chemo, chemo → T-DXd, and T-DXd → SG. Outcomes included quality-adjusted life years (QALYs), total lifetime costs (2020 US dollars [USD], 3% annual discount), and incremental cost-effectiveness ratios. Sequences that cost <$150,000 USD to gain an additional QALY were considered cost effective. RESULTS Chemo → chemo has the lowest cost at $176,000 (USD) per patient and yields 0.82 QALYs. T-DXd → chemo costs $282,000 (USD) and yields 1.08 QALYs, with an incremental cost-effectiveness ratio of $408,000 (USD) per QALY gained. T-DXd → SG costs $304,000 (USD) and yields 1.09 QALYs, with an incremental cost-effectiveness ratio of $2,200,000 (USD) per QALY gained. Drug cost drives the cost differences between each strategy. For T-DXd → chemo to be cost effective at the $150,000 (USD) per QALY threshold, we estimate that a 41% price reduction for T-DXd is needed. CONCLUSION At its current price, T-DXd is not cost effective for HER2-low metastatic breast cancer. Price reductions can make this drug cost effective. Optimal value-based sequencing in this patient population uses a single antibody-drug conjugate rather than back-to-back conjugates.
Collapse
Affiliation(s)
- James C Dickerson
- Department of Medicine (Hematology and Oncology), Stanford University, Stanford, CA
- Department of Health Policy, Stanford University, Stanford, CA
| | - Marcus T Moen
- Department of Management Science and Engineering, Stanford University, Stanford, CA
| | - Perry Nielsen
- Department of Health Policy, Stanford University, Stanford, CA
| | - Fauzia Riaz
- Department of Medicine (Hematology and Oncology), Stanford University, Stanford, CA
| | - Edward Tran
- Department of Management Science and Engineering, Stanford University, Stanford, CA
- Department of Computer Science, Stanford University, Stanford, CA
| | | | - Wesley Suen
- Department of Health Policy, Stanford University, Stanford, CA
- Department of Biology, Stanford University, Stanford, CA
| | | | | |
Collapse
|
6
|
Sanomachi T, Shimoi T, Kojima Y, Takahashi H, Arakaki M, Kawachi A, Okuma HS, Hoshino M, Ito M, Saito A, Kita S, Maejima A, Nishikawa T, Sudo K, Mori T, Fujiwara Y, Yonemori K. Efficacy of Eribulin Monotherapy in Patients With Unresectable and/or Metastatic Adenoid Cystic Carcinoma Receiving Platinum-Based Therapy. Head Neck 2025. [PMID: 40390268 DOI: 10.1002/hed.28190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 04/18/2025] [Accepted: 05/05/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Adenoid cystic carcinoma (ACC) is the second most common and treatment-resistant salivary gland carcinoma (SGC). Although platinum-based therapies are effective, the efficacy of eribulin, a microtubule inhibitor with an 18% response rate in a Phase II trial involving recurrent/metastatic SGC, in patients with advanced ACC after platinum therapy remains unclear. METHODS We retrospectively analyzed 19 patients with unresectable/metastatic ACC treated with platinum-based therapy between December 2015 and January 2023 at the National Cancer Center Hospital, Japan. Ten patients received eribulin. RESULTS The median age was 61 years. 84% achieved disease control with the first-line or later platinum-based therapy (median progression-free survival [mPFS]: 7.6 months). Second-line or later eribulin monotherapy yielded a 50% disease control rate, with mPFS of 3.7 months and median overall survival of 41.7 months. The adverse events, predominantly neutropenia, were manageable. CONCLUSION Eribulin shows promising effects of unresectable/metastatic ACC, offering tolerable adverse events.
Collapse
Affiliation(s)
- Tomomi Sanomachi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Motoko Arakaki
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Asuka Kawachi
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Mai Hoshino
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Munehiro Ito
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ayumi Saito
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shosuke Kita
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Aiko Maejima
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Taisuke Mori
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
7
|
Natori K, Igeta M, Morimoto T, Nagahashi M, Akashi-Tanaka S, Daimon T, Miyoshi Y. Development and internal validation of a predictive model of overall and progression-free survival in eribulin-treated patients with breast cancer based on baseline peripheral blood parameters. Breast Cancer 2025; 32:500-511. [PMID: 39979692 PMCID: PMC11993485 DOI: 10.1007/s12282-025-01678-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/01/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Immune and inflammatory blood parameters have been reported as biomarkers for treatment efficacy. This study aimed to establish a predictive model that includes blood parameters for patients with metastatic breast cancer treated with eribulin. METHODS A total of 297 patients were enrolled, and their baseline neutrophil-to-lymphocyte ratio, absolute lymphocyte count (ALC), platelet-to-lymphocyte ratio (PLR), prognostic nutritional index (PNI), lymphocyte-to-monocyte ratio (LMR), lactate dehydrogenase (LDH), C-reactive protein (CRP), and clinical data were retrospectively collected. RESULTS We constructed nomograms to predict overall survival (OS) and progression-free survival (PFS) using blood parameters, including clinical factors. For OS, menopausal status, hormone receptor status, HER2 status, de novo or recurrent, metastatic site, treatment line, ALC, PLR, PNI, LMR, LDH, and CRP were selected to predict the model. We used menopausal status, hormone receptor status, HER2 status, treatment line, PLR, LMR, LDH, and CRP to predict PFS. Both the OS and PFS of patients according to the risk scores were significantly different (p < 0.001). The optimism-corrected C-indices of the nomograms for OS and PFS were 0.680 and 0.622, respectively. The mean time-dependent area under the receiver operating curve values for OS at 1, 2, and 3 years were 0.752, 0.761, and 0.784, respectively, and for PFS at 3, 6, and 12 months were 0.660, 0.661, and 0.650, respectively. CONCLUSION Nomograms incorporating peripheral blood parameters may improve the accuracy of predicting OS and PFS in patients treated with eribulin. Our prediction model may help decision-making for breast cancer patients who are considering eribulin treatment.
Collapse
Affiliation(s)
- Keiko Natori
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
- Department of Breast Surgery, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Masataka Igeta
- Department of Biostatistics, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Takashi Morimoto
- Department of Breast Surgery, Yao Municipal Hospital, 1-3-1 Ryuge-Cho, Yao, Osaka, 581-0069, Japan
| | - Masayuki Nagahashi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Sadako Akashi-Tanaka
- Department of Breast Surgery, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Takashi Daimon
- Department of Biostatistics, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yasuo Miyoshi
- Department of Surgery, Division of Breast and Endocrine Surgery, School of Medicine, Hyogo Medical University, 1-1 Mukogawa-Cho, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
8
|
Risinger AL. Beyond mitotic arrest: the diverse effects of microtubule-targeting drugs on tumor vasculature. EMBO Mol Med 2025; 17:866-868. [PMID: 40140726 DOI: 10.1038/s44321-025-00223-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Affiliation(s)
- April L Risinger
- The University of Texas Health Science Center at San Antonio, Department of Pharmacology, San Antonio, Texas, 78229, USA.
| |
Collapse
|
9
|
Banwait R, Ko H, Michalek J, Liu Q, Lathrop K, Bowhay-Carnes E, Fotopoulos G, Sarantopoulos J, Elledge R, Taverna J, Karnad A, Siziopikou KP, Kaklamani V. Pilot phase II study of the combination of lenvatinib (L) and eribulin (E) in advanced solid tumors. Int J Cancer 2025. [PMID: 40232157 DOI: 10.1002/ijc.35446] [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: 09/17/2024] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
This pilot phase II study evaluated the combination of lenvatinib, a multi-kinase inhibitor, and eribulin, a microtubule inhibitor, in patients with advanced solid tumors, including breast carcinoma, lung carcinoma, and sarcoma. Tumor angiogenesis and resistance mechanisms to anti-angiogenic therapies were primary motivations for combining these agents. The trial enrolled 29 patients, heavily pretreated with at least three prior lines of chemotherapy, and aimed to assess the efficacy and safety of the combination therapy. Overall response rate (ORR) was 24% with the highest responses observed in breast cancer (29%) and lung cancer (33%) patients. Median progression-free survival (PFS) was 7.4 months (95% CI 4.5, NA), and overall survival (OS) was 8.2 months (95% CI 6.4 to 14.9). A significant improvement in both OS and PFS was found in vimentin-negative patients, suggesting that vimentin expression may be a predictor of treatment response. The most common treatment-related adverse events (TRAEs) were oral mucositis, fatigue, neutropenia, and nausea. Grade ≥3 TRAEs included neutropenia (34.5%), febrile neutropenia (17.2%), and hypertension (13.8%), with one fatal case of sepsis reported. While the study demonstrated the potential of lenvatinib and eribulin in advanced solid tumors, particularly breast and lung cancers, it also highlighted the need for further investigation into biomarkers like vimentin to predict therapeutic outcomes. The combination therapy was manageable in terms of safety and toxicity, with a predictable safety profile. These findings suggest that lenvatinib and eribulin represent a promising treatment strategy for advanced, heavily pretreated solid tumors, warranting further exploration in larger clinical studies.
Collapse
Affiliation(s)
- Ranjit Banwait
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Heidi Ko
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Joel Michalek
- Biostatistics Division, Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas, USA
| | - Qianqian Liu
- Biostatistics Division, Department of Population Health Sciences, UT Health San Antonio, San Antonio, Texas, USA
| | - Kate Lathrop
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Elizabeth Bowhay-Carnes
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Georgios Fotopoulos
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - John Sarantopoulos
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Richard Elledge
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Josephine Taverna
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Anand Karnad
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| | - Kalliopi P Siziopikou
- Department of Medicine, Division of Pathology, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Virginia Kaklamani
- Department of Medicine, Division of Hematology Oncology, UT Health San Antonio, Mays Cancer Center, San Antonio, Texas, USA
| |
Collapse
|
10
|
Yamashita T, Saji S, Takano T, Naito Y, Tsuneizumi M, Yoshimura A, Takahashi M, Tsurutani J, Iwatani T, Kitada M, Tada H, Mori N, Higuchi T, Iwasa T, Araki K, Koizumi K, Hasegawa H, Uchida Y, Morita S, Masuda N. Trastuzumab-Pertuzumab Plus Eribulin or Taxane as First-Line Chemotherapy for Human Epidermal Growth Factor 2-Positive Locally Advanced/Metastatic Breast Cancer: The Randomized Noninferiority Phase III EMERALD Trial. J Clin Oncol 2025; 43:1302-1313. [PMID: 39787453 PMCID: PMC11974627 DOI: 10.1200/jco-24-01888] [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: 08/28/2024] [Revised: 11/07/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
PURPOSE Trastuzumab-pertuzumab (HP) plus taxane is a current standard first-line therapy for recurrent or metastatic human epidermal growth factor 2 (HER2)+ breast cancer (BC). We investigated noninferiority of eribulin to a taxane when combined with dual HER2 blockade as first-line systemic treatment for locally advanced/metastatic HER2+ BC. METHODS In the phase III EMERALD trial (target sample size, 480; ClinicalTrials.gov identifier: NCT03264547/UMIN000027938), patients were randomly assigned (1:1) to receive eribulin 1.4 mg/m2 once daily on days 1 and 8 (eribulin group) or a taxane (docetaxel 75 mg/m2 once on day 1 or paclitaxel 80 mg/m2 once daily on days 1, 8, and 15; taxane group) intravenously in a 21-day cycle, each with HP on day 1. The primary end point was progression-free survival (PFS; intention-to-treat population). Secondary end points included objective response rate, overall survival (OS), patient-reported quality of life (QoL), and safety. Noninferiority was tested using the stratified Cox proportional hazards model to estimate hazard ratios (HRs) for PFS events, with a noninferiority HR margin of 1.33. RESULTS Between August 2017 and June 2021, 446 patients (median age, 56.0 years) were enrolled. The median PFS was 14.0 and 12.9 months in the eribulin group (n = 224) and taxane group (n = 222 [docetaxel/paclitaxel, n = 186/36]), respectively (HR, 0.95 [95% CI, 0.76 to 1.19]), which confirmed the noninferiority of the study regimen. The median OS was 65.3 months in the taxane group but has not been reached in the eribulin group. Median time to QoL deterioration was numerically longer with eribulin than with taxane. Adverse event (AE) rates were similar, despite the longer duration of eribulin use. Infusion reaction, skin-related AEs, diarrhea, and edema were more common with taxane, whereas neutropenia was more common with eribulin. CONCLUSION The results suggested that eribulin + HP is an option for first-line treatment of locally advanced/metastatic HER2+ BC.
Collapse
Affiliation(s)
- Toshinari Yamashita
- Department of Breast Surgery and Oncology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | - Toshimi Takano
- Department of Breast Medical Oncology, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Chiba, Japan
| | - Michiko Tsuneizumi
- Department of Breast Surgery, Shizuoka General Hospital, Shizuoka, Japan
| | - Akiyo Yoshimura
- Department of Breast Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| | - Masato Takahashi
- Department of Breast Surgery, Hokkaido University Hospital, Hokkaido, Japan
| | - Junji Tsurutani
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Tsuguo Iwatani
- Breast and Endocrine Surgery, Okayama University Hospital, Okayama, Japan
| | - Masahiro Kitada
- Department of Breast Disease Center, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Hiroshi Tada
- Department of Surgery, Division of Breast and Endocrine Surgery, Tohoku University Hospital, Miyagi, Japan
| | - Natsuko Mori
- Department of Breast Surgery, Seirei Hamamatsu General Hospital, Shizuoka, Japan
| | - Toru Higuchi
- Department of Breast Unit, Japanese Red Cross Saitama Hospital, Saitama, Japan
| | - Tsutomu Iwasa
- Department of Medical Oncology, Kindai University Hospital, Osaka, Japan
| | - Kazuhiro Araki
- Department of Breast Medical Oncology, Gunma Prefectural Cancer Center, Gunma, Japan
| | - Kei Koizumi
- Department of Surgery 1, Division of Breast Surgery, Hamamatsu University School of Medicine, Shizuoka, Japan
| | | | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norikazu Masuda
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
O'Meara TA, Tarantino P. Lessons From Taxanes Versus Eribulin for First-Line Metastatic Human Epidermal Growth Factor Receptor 2-Positive Breast Cancer in the EMERALD Study. J Clin Oncol 2025; 43:1275-1278. [PMID: 39841942 DOI: 10.1200/jco-24-02717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/24/2025] Open
Affiliation(s)
- Tess A O'Meara
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Zhang T, Ouyang Z, Zhang Y, Sun H, Kong L, Xu Q, Qu J, Sun Y. Marine Natural Products in Inflammation-Related Diseases: Opportunities and Challenges. Med Res Rev 2025. [PMID: 40202793 DOI: 10.1002/med.22109] [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: 01/21/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
In recent decades, the potentiality of marine natural products (MNPs) in the medical field has been increasingly recognized. Natural compounds derived from marine microorganisms, algae, and invertebrates have shown significant promise for treating inflammation-related diseases. In this review, we cover the three primary sources of MNPs and their diverse and unique chemical structures and bioactivities. This review aims to summarize the progress of MNPs in combating inflammation-related diseases. Moreover, we cover the functions and mechanisms of MNPs in diseases, highlighting their functions in regulating inflammatory signaling pathways, cellular stress responses, and gut microbiota, among others. Meanwhile, we focus on key technologies and scientific methods to address the current limitations and challenges in MNPs.
Collapse
Affiliation(s)
- Tao Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zijun Ouyang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Yueran Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Haiyan Sun
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
13
|
Marhold M, Vaz Batista M, Blancas I, Morales C, Saura-Manich C, Saavedra C, Ruíz-Borrego M, Cortez P, Slebe F, Campolier M, Santos JC, Guerrero-Martínez JA, Jiménez-Cortegana C, Rottenmanner B, Forstner H, Bartsch R, Preusser M. TUXEDO-4: phase II study of trastuzumab-deruxtecan in HER2-low breast cancer with new or progressing brain metastases. Future Oncol 2025; 21:1065-1073. [PMID: 40018758 PMCID: PMC11988270 DOI: 10.1080/14796694.2025.2470604] [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: 09/27/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
CLINICAL TRIAL REGISTRATION NCT06048718 (clinicaltrials.gov); 2023 -506,702-39-00 (EudraCT number).
Collapse
Affiliation(s)
- Maximilian Marhold
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Marta Vaz Batista
- Department of Oncology, Hospital Professor Doutor Fernando Fonseca EPE, Lisbon, Portugal
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Isabel Blancas
- Department of Oncology, Hospital Universitario Clínico San Cecilio, Granada, Spain
- Medicine Department, University of Granada, Granada, Spain
- Área de Oncología Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Cristina Morales
- Servicio de Oncología Médica, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Cristina Saura-Manich
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Barcelona, Spain
| | - Cristina Saavedra
- Departamento de Oncología Médica, Hospital Universitario Ramón y Cajal, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | | | - Patricia Cortez
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Felipe Slebe
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | - Marta Campolier
- Medica Scientia Innovation Research (MEDSIR), Barcelona, Spain
| | | | | | | | - Beate Rottenmanner
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Heidrun Forstner
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Rupert Bartsch
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
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
|
15
|
García-Saenz JÁ, Rodríguez-Lescure Á, Cruz J, Albanell J, Alba E, Llombart A. Second-Line Treatment Options for Patients with Metastatic Triple-Negative Breast Cancer: A Review of the Clinical Evidence. Target Oncol 2025; 20:191-213. [PMID: 39806129 PMCID: PMC11933194 DOI: 10.1007/s11523-024-01125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/16/2025]
Abstract
Metastatic triple-negative breast cancer has a poor prognosis and poses significant therapeutic challenges. Until recently, limited therapeutic options have been available for patients with advanced disease after failure of first-line chemotherapy. The aim of this review is to assess the current evidence supporting second-line treatment options in patients with metastatic triple-negative breast cancer. Evidence was reviewed from controlled clinical trials in which eribulin, vinorelbine, capecitabine, gemcitabine, gemcitabine plus carboplatin, fam-trastuzumab-deruxtecan, sacituzumab govitecan, olaparib, and talazoparib were used in the second-line treatment for metastatic breast cancer, either as study drugs or as comparators. The benefit of treatment was evaluated using the European Society for Medical Oncology-Magnitude of Clinical Benefit Scale. Based on the evidence review, sacituzumab govitecan was identified as the preferred second-line treatment option for patients with metastatic triple-negative breast cancer, supported by clinical evidence and consensus across international clinical guidelines. Olaparib and talazoparib are of use in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer and germline BRCA1/2 mutations. Exploratory data for fam-trastuzumab-deruxtecan suggest a survival benefit in human epidermal growth factor receptor 2-low, hormone-receptor-negative patients, but further solid evidence is required. Other chemotherapies with lower European Society for Medical Oncology-Magnitude of Clinical Benefit Scale scores may continue to be useful in highly selected patients.
Collapse
Affiliation(s)
- José Ángel García-Saenz
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Calle Profesor Martín Lagos, S/N, 28040, Madrid, Spain.
| | | | - Josefina Cruz
- Hospital Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Joan Albanell
- Hospital del Mar Research Institute, Barcelona, Spain
- Pompeu Fabra University, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Oncología, CIBERONC-ISCIII, Madrid, Spain
| | - Emilio Alba
- Medical Oncology Unit, Universitary Hospital Virgen de la Victoria, CIBERONC, IBIMA, Malaga, Spain
| | | |
Collapse
|
16
|
Lopetegui-Lia N, Varma R, Abraham J, Roesch E. Current and Novel Treatment Options in Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Metastatic Breast Cancer. JCO Oncol Pract 2025; 21:145-154. [PMID: 39167745 DOI: 10.1200/op.23.00830] [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: 12/27/2023] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Metastatic breast cancer (mBC) remains an incurable disease, and most patients will experience disease progression during their treatment course. Although endocrine therapy remains the mainstay of treatment for hormone receptor-positive/human epidermal growth factor receptor 2-negative mBC, significant progress has been and continues to be made in the treatment of this BC subtype. The discovery of molecular markers, mutations in key cellular pathways, and genomic signatures have led to the development of novel and targeted agents, such as antibody-drug conjugates, oral selective estrogen receptor downregulators, and inhibitors of the PI3K/AKT/mTOR pathway. This has resulted in significant improvements in the survival and quality of life of patients. With the increasing number of treatment options for patients, appropriate drug sequencing remains a challenge. Treatment discussions should involve patient-physician shared decision making, with consideration of genomic data, previous lines of therapy, side effect profiles, and clinical trial enrollment.
Collapse
|
17
|
Fujimi A, Nagamachi Y, Yamauchi N, Hasebe R, Onoyama N, Hayasaka N, Matsuno T, Koike K, Gotoh Y, Ihara K, Kato J, Nishisato T, Murase K, Kutomi G, Hida T, Uhara H, Takada K. Efficacy of eribulin monotherapy for bone marrow carcinomatosis of breast cancer in a patient with Werner syndrome. Geriatr Gerontol Int 2025; 25:316-318. [PMID: 39761950 DOI: 10.1111/ggi.15070] [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: 10/16/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
Various complications and potential risks of serious adverse events lessens the intensity of chemotherapy in patients with Werner syndrome. Bone marrow carcinomatosis of breast cancer was developed in a patient with Werner syndrome. Eribulin proved well tolerated and effective in improving severe thrombocytopenia, leading to platelet transfusion-free status.
Collapse
Affiliation(s)
- Akihito Fujimi
- Department of Hematology, Sapporo Kiyota Hospital, Sapporo, Japan
| | | | - Naofumi Yamauchi
- Department of Hematology, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Riku Hasebe
- Department of Hematology, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Naoki Onoyama
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Naotaka Hayasaka
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Teppei Matsuno
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Kazuhiko Koike
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Yoshiro Gotoh
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Kohji Ihara
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Junji Kato
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Takuji Nishisato
- Department of Internal Medicine, Sapporo Kiyota Hospital, Sapporo, Japan
| | - Kazuyuki Murase
- Department of Medical Oncology, Sapporo Medical University, Sapporo, Japan
| | - Goro Kutomi
- Department of Surgery, Sapporo Medical University, Sapporo, Japan
| | - Tokimasa Hida
- Department of Dermatology, Sapporo Medical University, Sapporo, Japan
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University, Sapporo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
18
|
Guchelaar NA, Mathijssen RH, de Boer M, van Bekkum ML, Heijns JB, Vriens BE, van Rosmalen MM, Kessels LW, Hamming L, Beelen KJ, Nieboer P, van den Berg SM, Hoop EOD, Bijlsma RM, Bos ME, Dutch Breast Cancer Research Group (BOOG). Trifluridine-tipiracil in previously treated patients with oestrogen receptor-positive, HER2-negative metastatic breast cancer (BOOG 2019-01 TIBET trial): a single-arm, multicentre, phase 2 trial. EClinicalMedicine 2025; 80:103065. [PMID: 40017682 PMCID: PMC11867194 DOI: 10.1016/j.eclinm.2024.103065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 03/01/2025] Open
Abstract
Background Effective later-line chemotherapy treatment options are scarce for patients with metastatic breast cancer (MBC). Trifluridine-tipiracil has shown survival benefit in heavily pre-treated patients with metastatic colorectal and in gastric cancer refractory to a fluoropyrimidine. This study aimed to investigate the efficacy of trifluridine-tipiracil in a Western population of previously treated patients with oestrogen receptor (ER+), HER2- MBC to facilitate further optimization of this treatment strategy. Methods Adult patients at least 18 years old diagnosed with hormone receptor positive, HER2- receptor negative MBC with a performance status of 0 or 1 who have been treated with capecitabine in the metastatic setting and up to two other lines of chemotherapy, including a taxane, were enrolled in this single-arm, multicentre, phase 2 study in the Netherlands. The participants received trifluridine-tipiracil 35 mg/m2 orally twice a day on days 1-5 and days 8-12 during a 28-day cycle until disease progression, unacceptable toxicity, or withdrawal of consent. The primary endpoint was the disease control rate (DCR) at 8 weeks, defined as the percentage of patients that had stable disease, partial response or complete response according to RECIST 1.1, in all patients that received at least one dose of trifluridine-tipiracil and met the key eligibility criteria defined a priori. Secondary endpoints included progression-free survival (PFS), overall survival (OS), safety, and quality of life and were performed in all patients that received at least one dose of trifluridine-tipiracil. The primary endpoint was considered met, justifying further research of this treatment regimen, if the lower boundary of the 80% confidence interval (CI) exceeded 30%. The study was registered within ClinicalTrials.gov (NCT04489173) and is closed for inclusion. Findings Fifty female patients were enrolled from September 2020 to July 2023, with a median of 3 (IQR, 2-3) previous endocrine therapy lines and 2 (IQR, 2-3) chemotherapy lines for MBC. The DCR rate at 8 weeks was 64.0% (n = 32, 95% CI: 50.1-75.9%; 80% CI: 55.0-72.1%), thereby meeting the primary endpoint of this study. At data cutoff (January 8, 2024), the median follow-up time was 18.2 months (IQR, 13.1-25.1 months). The median PFS was 5.4 months (95% CI: 2.0-7.2 months) and the median OS 14.0 months (95% CI: 8.8-17.8 months). The safety profile of trifluridine-tipiracil aligned with expected toxicities and included leukopenia (n = 36, 69%), neutropenia (n = 43, 83%), and fatigue (n = 43, 83%). The most common grade 3-4 AEs were primarily haematological disorders and included neutropenia (n = 38, 73%), leukopenia (n = 15, 29%) and anaemia (n = 6, 12%). The most common SAEs (any grade) with a possible relationship with trifluridine-tipiracil included anaemia (n = 2) and vomiting (n = 2). No treatment-related deaths occurred. Quality of life scores remained stable throughout the treatment. Interpretation Trifluridine-tipiracil demonstrated promising efficacy in heavily pre-treated patients with MBC, despite prior exposure to a fluoropyrimidine. Clinically, this suggests that trifluridine-tipiracil holds potential as a viable oral later-line treatment option with a manageable toxicity profile while maintaining quality of life. Preparations for a phase 3 trial are underway. Funding Servier, France.
Collapse
Affiliation(s)
- Niels A.D. Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ron H.J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Maaike de Boer
- Department of Internal Medicine, Division of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, the Netherlands
| | | | - Joan B. Heijns
- Department of Internal Medicine, Amphia, Breda, the Netherlands
| | | | - Mandy M. van Rosmalen
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Lonneke W. Kessels
- Department of Internal Medicine, Deventer Hospital, Deventer, the Netherlands
| | - Lisanne Hamming
- Department of Internal Medicine, Medical Center Leeuwarden, Leeuwarden, the Netherlands
| | - Karin J. Beelen
- Department of Internal Medicine, Rijnstate, Arnhem, the Netherlands
| | - Peter Nieboer
- Department of Internal Medicine, Wilhelmina Hospital Assen, Assen, the Netherlands
| | | | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rhodé M. Bijlsma
- Department of Medical Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| | - Monique E.M.M. Bos
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dutch Breast Cancer Research Group (BOOG)
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Internal Medicine, Division of Medical Oncology, GROW-School for Oncology and Developmental Biology, Maastricht UMC+, Maastricht, the Netherlands
- Department of Internal Medicine, Reinier de Graaf Hospital, Delft, the Netherlands
- Department of Internal Medicine, Amphia, Breda, the Netherlands
- Department of Internal Medicine, Catharina Hospital, Eindhoven, the Netherlands
- Department of Internal Medicine, Deventer Hospital, Deventer, the Netherlands
- Department of Internal Medicine, Medical Center Leeuwarden, Leeuwarden, the Netherlands
- Department of Internal Medicine, Rijnstate, Arnhem, the Netherlands
- Department of Internal Medicine, Wilhelmina Hospital Assen, Assen, the Netherlands
- Dutch Breast Cancer Research Group (BOOG), the Netherlands
- Department of Medical Oncology, UMC Utrecht Cancer Center, Utrecht, the Netherlands
| |
Collapse
|
19
|
Schmidt M, Hesse T, Hoffmann O, Heinrich BJ, Park-Simon TW, Grischke EM, Weide R, Müller Huesmann H, Lüdtke-Heckenkamp K, Fischer D, Zemlin C, Kögel M, Jia Y, Schmitz H, Engelbrecht C, Jackisch C. Eribulin-Induced Peripheral Neuropathy in Locally Advanced or Metastatic Breast Cancer: Final Analysis of the Prospective Cohort IRENE Study. Cancers (Basel) 2025; 17:457. [PMID: 39941823 PMCID: PMC11815746 DOI: 10.3390/cancers17030457] [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: 11/26/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Eribulin is a preferred treatment for patients with advanced breast cancer (BC) following anthracyclines and taxanes. The final analysis of the IRENE study assessed the incidence and resolution of eribulin-induced peripheral neuropathy (EIPN), along with safety and quality of life (QoL), in patients with advanced/metastatic BC. IRENE was an observational, single-arm, prospective, multicenter cohort study. Patients aged ≥18 years with locally advanced/metastatic BC that progressed after 1-3 prior chemotherapeutic regimens received eribulin and were monitored for new-onset or worsening EIPN. Secondary endpoints included time to disease progression, safety, and health-related QoL. In total, 108 (32.2%) out of 335 patients experienced EIPN; 18 (5.4%) experienced grade ≥3 EIPN. Median time to EIPN resolution (EIPN ended or returned to baseline) was 78.7 weeks (95% CI 77.1-not estimable). Median time to disease progression was 4.5 months (95% CI 3.9-5.5). Treatment-emergent adverse events (TEAEs) occurred in 322 (96.1%) patients; serious TEAEs occurred in 185 (55.2%) patients. Incidence and resolution rates of EIPN were comparable with existing evidence from previous trials. TEAEs were consistent with the established eribulin safety profile, with no new safety signals. Eribulin treatment did not appear to affect QoL, as measured by EQ-5D-3L and EQ-VAS, or patient-reported neuropathy symptoms, as measured by the PNQ.
Collapse
Affiliation(s)
- Marcus Schmidt
- Department of Obstetrics and Gynecology, University Medical Center Mainz, 55131 Mainz, Germany
- Department of Obstetrics and Gynecology, Sana Klinikum Offenbach GmbH, 63069 Offenbach, Germany
| | - Tobias Hesse
- Department of Gynecology, Agaplesion Diakonieklinikum Rotenburg gGmbH, 27356 Rotenburg (Wuemme), Germany;
| | - Oliver Hoffmann
- Department of Gynecology and Obstetrics, Universitätsklinikum, 45147 Essen, Germany;
| | | | | | - Eva-Maria Grischke
- Department für Frauengesundheit, Universitäts-Frauenklinik Tübingen, 72076 Tübingen, Germany;
| | - Rudolf Weide
- Oncological Outpatient Department, Praxis für Hämatologie und Onkologie Koblenz, 56073 Koblenz, Germany;
| | - Harald Müller Huesmann
- Department of Hematology/Oncology, Brüderkrankenhaus St. Josef Paderborn, 33098 Paderborn, Germany;
| | - Kerstin Lüdtke-Heckenkamp
- Department of Hematology/Oncology, Niels Stensen Clinics, Franziskus Hospital, 49124 Georgsmarienhütte, Germany;
| | - Dorothea Fischer
- Department of Gynaecology and Obstetrics, Hospital Ernst von Bergmann, 14467 Potsdam, Germany;
| | - Cosima Zemlin
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, 66421 Homburg, Germany;
| | | | - Yan Jia
- Eisai Inc., Nutley, NJ 07110, USA;
| | - Helga Schmitz
- Medical Department, Eisai GmbH, 60549 Frankfurt, Germany;
| | | | | |
Collapse
|
20
|
Bardia A, Jhaveri K, Im SA, Pernas S, De Laurentiis M, Wang S, Martínez Jañez N, Borges G, Cescon DW, Hattori M, Lu YS, Hamilton E, Zhang Q, Tsurutani J, Kalinsky K, Rubini Liedke PE, Xu L, Fairhurst RM, Khan S, Denduluri N, Rugo HS, Xu B, Pistilli B. Datopotamab Deruxtecan Versus Chemotherapy in Previously Treated Inoperable/Metastatic Hormone Receptor-Positive Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer: Primary Results From TROPION-Breast01. J Clin Oncol 2025; 43:285-296. [PMID: 39265124 PMCID: PMC11771365 DOI: 10.1200/jco.24.00920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/27/2024] [Accepted: 07/30/2024] [Indexed: 09/14/2024] Open
Abstract
PURPOSE The global, phase 3, open-label, randomized TROPION-Breast01 study assessed the trophoblast cell surface antigen 2-directed antibody-drug conjugate datopotamab deruxtecan (Dato-DXd) versus investigator's choice of chemotherapy (ICC) in hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-) breast cancer. METHODS Adult patients with inoperable/metastatic HR+/HER2‒ breast cancer, who had disease progression on endocrine therapy, for whom endocrine therapy was unsuitable, and had received one to two previous lines of chemotherapy in the inoperable/metastatic setting, were randomly assigned 1:1 to Dato-DXd (6 mg/kg once every 3 weeks) or ICC (eribulin/vinorelbine/capecitabine/gemcitabine). Dual primary end points were progression-free survival (PFS) by blinded independent central review (BICR) and overall survival (OS). RESULTS Patients were randomly assigned to Dato-DXd (n = 365) or ICC (n = 367). Dato-DXd significantly reduced the risk of progression or death versus ICC (PFS by BICR hazard ratio [HR], 0.63 [95% CI, 0.52 to 0.76]; P < .0001). Consistent PFS benefit was observed across subgroups. Although OS data were not mature, a trend favoring Dato-DXd was observed (HR, 0.84 [95% CI, 0.62 to 1.14]). The rate of grade ≥3 treatment-related adverse events (TRAEs) with Dato-DXd was lower than ICC (20.8% v 44.7%). The most common TRAEs (any grade; grade ≥3) were nausea (51.1%; 1.4%) and stomatitis (50%; 6.4%) with Dato-DXd and neutropenia (grouped term, 42.5%; 30.8%) with ICC. CONCLUSION Patients receiving Dato-DXd had statistically significant and clinically meaningful improvement in PFS and a favorable and manageable safety profile, compared with ICC. Results support Dato-DXd as a novel treatment option for patients with inoperable/metastatic HR+/HER2‒ breast cancer who have received one to two previous lines of chemotherapy in this setting.
Collapse
Affiliation(s)
- Aditya Bardia
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA
- Massachusetts General Hospital Cancer Center, Boston, MA
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sonia Pernas
- Institut Català d'Oncologia-IDIBELL, L'Hospitalet, Barcelona, Spain
| | | | - Shusen Wang
- Cancer Center of Sun Yat-sen University, Guangzhou, China
| | - Noelia Martínez Jañez
- Ramón y Cajal University Hospital, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | | | | | - Yen-Shen Lu
- National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | - Qingyuan Zhang
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Junji Tsurutani
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan
| | - Kevin Kalinsky
- Winship Cancer Institute at Emory University, Atlanta, GA
| | - Pedro Emanuel Rubini Liedke
- Hospital das Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
- UPCO—Pesquisa Clinica em Oncologia, Porto Alegre, Brazil
- Oncoclinicas Porto Alegre, Porto Alegre, Brazil
| | - Lu Xu
- AstraZeneca, Gaithersburg, MD
| | | | | | | | - Hope S. Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA
| | - Binghe Xu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
21
|
Anampa JD, Flynn DL, Leary C, Oh S, Xue X, Oktay MH, Condeelis JS, Sparano JA. Phase Ib Clinical and Pharmacodynamic Study of the TIE2 Kinase Inhibitor Rebastinib with Paclitaxel or Eribulin in HER2-Negative Metastatic Breast Cancer. Clin Cancer Res 2025; 31:266-277. [PMID: 39531537 PMCID: PMC11818423 DOI: 10.1158/1078-0432.ccr-24-2464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE Breast cancer cells disseminate to distant sites via tumor microenvironment of metastasis (TMEM) doorways. The TIE2 inhibitor rebastinib blocks TMEM doorway function in the PyMT mouse model of breast cancer. We aimed to assess the safety and pharmacodynamics of rebastinib plus paclitaxel or eribulin in patients with HER2-negative metastatic breast cancer (MBC). PATIENTS AND METHODS This phase Ib trial enrolled 27 patients with MBC who received 50 mg or 100 mg of rebastinib orally twice daily in combination with weekly paclitaxel 80 mg/m2 (if ≤2 prior non-taxane regimens) or eribulin 1.4 mg/m2 on days 1 and 8 (if ≥1 prior regimen). Safety, tolerability, and pharmacodynamic parameters indicating TIE2 kinase inhibition and TMEM doorway function were evaluated. RESULTS No dose-limiting toxicities in cycle 1 or 2 were observed among the first 12 patients at either rebastinib dose level. The most common treatment-emergent adverse events were anemia (85%), fatigue (78%), anorexia (67%), leukopenia (67%), increased alanine aminotransferase (59%), hyperglycemia (56%), nausea (52%), and neutropenia (52%). Adverse events attributed to rebastinib include muscular weakness and myalgias. Intraocular pressure increased at the 100-mg rebastinib dose level, whereas angiopoietin-2 levels increased at both dose levels, providing pharmacodynamic evidence for TIE2 blockade. Circulating tumor cells decreased significantly with the combined treatment. Objective response occurred in 5/23 (22%) evaluable patients. CONCLUSIONS In patients with MBC, the recommended phase II dose of rebastinib associated with pharmacodynamic evidence of TIE2 inhibition is either 50 or 100 mg orally twice daily in combination with paclitaxel or eribulin.
Collapse
Affiliation(s)
- Jesus D. Anampa
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | | | - Cynthia Leary
- Deciphera Pharmaceuticals LLC, Waltham, Massachusetts
| | - Sun Oh
- Department of Medical Oncology, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Maja H. Oktay
- Tumor Microenvironment and Metastasis Program, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - John S. Condeelis
- Tumor Microenvironment and Metastasis Program, Montefiore Einstein Comprehensive Cancer Center, Bronx, New York
| | - Joseph A. Sparano
- Division of Hematology Oncology, Tish Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
22
|
Fan Y, Zhang Q, Yan M, Qu X, Yin Y, Sun T, Yang J, Wang Y, Wang X, Niu Z, Wang X, Sun S, Zhao W, Liu Y, Niu M, Zhao X, Xu B. Intravenous liposomal irinotecan in metastatic triple-negative breast cancer after ≥ 2 prior lines of chemotherapy: a phase Ib study. Nat Commun 2025; 16:3. [PMID: 39746964 PMCID: PMC11696226 DOI: 10.1038/s41467-024-55090-4] [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: 06/18/2024] [Accepted: 12/01/2024] [Indexed: 01/04/2025] Open
Abstract
This study (NCT04728035) aimed to explore the safety and efficacy of liposomal irinotecan (HE072) in patients with metastatic triple-negative breast cancer (mTNBC). This study consisted of two parts. In part 1, the 3 + 3 design was used to investigate three dose levels of HE072 (50, 70 and 90 mg/m2). In part 2, patients were enrolled in two cohorts (mTNBC and HER2-negative breast cancer brain metastasis [BCBM]), and received HE072 70 mg/m2 every two weeks (Q2W). The primary endpoints were maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D), and treatment emergent adverse events (TEAEs). The secondary endpoints were pharmacokinetic profiles and efficacy including objective response rate (ORR) and disease control rate (DCR) (all patients) and Central Nervous System ORR and clinical benefit rate (CBR, for patients with HER2-negative BCBM), duration of response, progression free survival (PFS), overall survival (OS). A total of 119 patients were enrolled, including 101 mTNBC and 18 HER2-negative BCBM. One dose limiting toxicity (grade 3 nausea and vomiting) occurred at 70 mg/m2, and the MTD was not reached. The most common ≥ grade 3 TEAEs related to HE072 included neutropenia (21.0%), leukopenia (18.5%), diarrhea (10.1%). Among 87 evaluable patients with mTNBC, 22 patients (25.3%) achieved overall response. The DCR was 67.8% (59/87). The median PFS and OS were 4.8 months and 14.1 months, respectively. The RP2D was 70 mg/m2 Q2W. Promising antitumor activity in heavily pre-treated patients with mTNBC was observed, which warrants further validation.
Collapse
Affiliation(s)
- Ying Fan
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingyuan Zhang
- Harbin Medical University Affiliated Cancer Hospital, Harbin, China
| | - Min Yan
- Henan Cancer Hospital, Zhengzhou, China
| | - Xiujuan Qu
- The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yongmei Yin
- Jiangsu Provincial People's Hospital, Nanjing, China
| | - Tao Sun
- Liaoning Cancer Hospital, Shenyang, China
| | - Jin Yang
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Wang
- Sun Yat Sen Memorial Hospital of Sun Yat Sen University, Guangzhou, China
| | - Xu Wang
- Tianjin Cancer Hospital, Tianjin, China
| | | | - Xinshuai Wang
- The First Affiliated Hospital of Henan University of Science and Technology, Zhengzhou, China
| | | | - Weihong Zhao
- Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yanping Liu
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd, Shijiazhuang, China
| | - Miao Niu
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd, Shijiazhuang, China
| | - Xuemin Zhao
- CSPC Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd, Shijiazhuang, 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, China.
| |
Collapse
|
23
|
Feng M, Bi P, Kang Y, Yang D, Ren S, Lu X, Xie G, Lei H, Mo D. Real world analysis of the efficacy and safety of eribulin compared to utidelone in combination with capecitabine for the treatment of metastatic breast cancer. Cancer Cell Int 2024; 24:416. [PMID: 39702163 DOI: 10.1186/s12935-024-03608-7] [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: 09/15/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND The objective of this study was to compare the efficacy and safety of subsequent chemotherapy with single eribulin or utidelone combined with a capecitabine regimen in patients with advanced breast cancer who had previously been treated with anthracyclines and paclitaxel. METHODS This work was a retrospective analysis of 85 patients from July 2018 to July 2023. Forty-two and 43 patients were treated with the eribulin regimen and the utidelone/capecitabine regimen, respectively. The endpoints included progression-free survival, overall survival, the objective remission rate and safety. Survival analyses and multifactorial analyses were performed via the Kaplan‒Meier method, log-rank test and Cox regression models. RESULTS As of 15 April 2024, the mPFS durations of the patients in the utidelone/capecitabine and eribulin treatment groups were 7.7 and 5.2 months, respectively, and the mOS durations were 22.0 and 18.2 months, respectively. Subgroup analyses revealed that in advanced first-line therapy, the mPFS durations of the utidelone/capecitabine group and the eribulin group were 11.8 and 7.0 months, respectively, and this difference was significant. In the eribulin treatment arm, the mPFS of first-line therapy was 7.0, whereas it was 3.3 months for posterior-line therapy, and this difference was significant. The most common adverse reactions were neurotoxicity, hand‒foot syndrome, hematological toxicity, gastrointestinal toxicity, and abnormalities in hepatic and renal functions. CONCLUSIONS In conclusion, either utidelone/capecitabine or eribulin chemotherapy may result in a survival benefit with a tolerable adverse effect profile and favorable safety profile in patients with metastatic breast cancer. The first-line use of eribulin resulted in better PFS and ORR than posterior-line use, and the combination of utidelone/capecitabine represents a more efficacious approach in the advanced first-line therapy of breast cancer.
Collapse
Affiliation(s)
- Mengya Feng
- Department of Breast Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, No. 318 Lucheng South Road, Chuxiong, 675000, Yunnan, China
| | - Pingping Bi
- Department of Oncology, The People's Hospital of Lincang, No.116 Nantang Street, Linxiang District, Lincang, 677000, Yunnan, China
| | - Yihua Kang
- Department II of General Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, No. 318 Lucheng South Road, Chuxiong, 675000, Yunnan, China
| | - Dechun Yang
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, No. 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, China
| | - Shengnan Ren
- Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, No. 519 Kunzhou Road, Xishan District, Kunming, 650100, Yunnan, China
| | - Xianping Lu
- Department of Breast Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, No. 318 Lucheng South Road, Chuxiong, 675000, Yunnan, China
| | - Guojian Xie
- Department of Breast Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, No. 318 Lucheng South Road, Chuxiong, 675000, Yunnan, China
| | - Hai Lei
- Department of Breast Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, No. 318 Lucheng South Road, Chuxiong, 675000, Yunnan, China.
| | - Dan Mo
- Department of Breast Surgery, The People's Hospital of Chuxiong Yi Autonomous Prefecture, No. 318 Lucheng South Road, Chuxiong, 675000, Yunnan, China.
| |
Collapse
|
24
|
Zhang J, Su J, Ni C, Lu J. Comparative efficacy and safety of eribulin versus paclitaxel in breast cancer: a systematic review and meta-analysis. Future Oncol 2024; 20:3507-3517. [PMID: 39563608 PMCID: PMC11776855 DOI: 10.1080/14796694.2024.2431479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
AIM We conducted a meta-analysis of published randomized controlled trials to compare the effectiveness and safety of eribulin versus paclitaxel for patients with breast cancer. METHODS We systematically searched multiple databases including Cochrane, PubMed, Medline, and Embase. The primary outcomes analyzed were overall survival (OS), complete response (CR), partial response (PR), stable disease (SD), and adverse events (AEs). These outcomes were evaluated using RevMan5.3 software. RESULTS A total of 5 studies were included in the analysis. Compared to paclitaxel plus other chemotherapy drugs, eribulin plus other chemotherapy drugs not only extended the overall survival of patients but also improved the disease control rate (DCR) [risk ratio (RR) 0.98, (95% confidence intervals (CI): 0.70, 1.38), p = 0.92]. Hematological system diseases [RR 1.18 (95% CI: 1.07, 1.31), p = 0.002] were the most frequently observed adverse event with eribulin, while paclitaxel was more likely to cause nervous system lesion [RR 0.66 (95% CI: 0.54, 0.80), p < 0.0001]. CONCLUSION Compared with paclitaxel plus other chemotherapy drugs, eribulin plus other chemotherapy drugs can also prolong the PFS and OS of BC patients. Our recommendation is to use eribulin plus other chemotherapy drugs to treat advanced BC and to continuously monitor and manage the drug-related adverse events.
Collapse
Affiliation(s)
- Jialin Zhang
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Jingyang Su
- Department of General internal medicine, Tongde Hospital Affiliated to Zhejiang Chinese Medical University (Tongde Hospital of Zhejiang Province), Hangzhou, China
| | - Cui Ni
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Jinhua Lu
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
25
|
Xu B, Wang S, Yan M, Sohn J, Li W, Tang J, Wang X, Wang Y, Im SA, Jiang D, Valdez T, Dasgupta A, Zhang Y, Yan Y, Komatsubara KM, Chung WP, Ma F, Dai MS. Sacituzumab govitecan in HR +HER2 - metastatic breast cancer: the randomized phase 3 EVER-132-002 trial. Nat Med 2024; 30:3709-3716. [PMID: 39354196 PMCID: PMC11645259 DOI: 10.1038/s41591-024-03269-z] [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: 04/05/2024] [Accepted: 08/23/2024] [Indexed: 10/03/2024]
Abstract
Sacituzumab govitecan (SG) significantly improved progression-free survival (PFS) and overall survival (OS) versus chemotherapy in hormone receptor-positive human epidermal growth factor receptor 2-negative (HR+HER2-) metastatic breast cancer (mBC) in the global TROPiCS-02 study. TROPiCS-02 enrolled few Asian patients. Here we report results of SG in Asian patients with HR+HER2- mBC from the EVER-132-002 study. Patients were randomized to SG (n = 166) or chemotherapy (n = 165). The primary endpoint was met: PFS was improved with SG versus chemotherapy (hazard ratio of 0.67, 95% confidence interval 0.52-0.87; P = 0.0028; median 4.3 versus 4.2 months). OS also improved with SG versus chemotherapy (hazard ratio of 0.64, 95% confidence interval 0.47-0.88; P = 0.0061; median 21.0 versus 15.3 months). The most common grade ≥3 treatment-emergent adverse events were neutropenia, leukopenia and anemia. SG demonstrated significant and clinically meaningful improvement in PFS and OS versus chemotherapy, with a manageable safety profile consistent with prior studies. SG represents a promising treatment option for Asian patients with HR+HER2- mBC (ClinicalTrials.gov identifier no. NCT04639986 ).
Collapse
Affiliation(s)
- Binghe Xu
- Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Shusen Wang
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Min Yan
- Henan Cancer Hospital/Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | | | - Wei Li
- The First Hospital of Jilin University, Changchun, China
| | - Jinhai Tang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | - Ying Wang
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | - Yilin Yan
- Gilead Sciences Inc., Foster City, CA, USA
| | | | - Wei-Pang Chung
- National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Fei Ma
- Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Chen YC, Takada M, Nagornyuk A, Muhan W, Yamada H, Nagashima T, Ohtsuka M, DeLuca JG, Markus S, Takaku M, Suzuki A. Inhibition of p38-MK2 pathway enhances the efficacy of microtubule inhibitors in breast cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621816. [PMID: 39574707 PMCID: PMC11580888 DOI: 10.1101/2024.11.04.621816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Microtubule-targeting agents (MTAs) have been successfully translated from basic research into clinical therapies and have been widely used as first- and second-line chemotherapy drugs for various cancers. However, current MTAs exhibit positive responses only in subsets of patients and are often accompanied by side effects due to their impact on normal cells. This underscores an urgent need to develop novel therapeutic strategies that enhance MTA efficacy while minimizing toxicity to normal tissues. In this study, we demonstrate that inhibition of the p38-MK2 (MAP kinase-activated protein kinase 2) pathway sensitizes cancer cells to MTA treatment. We utilize CMPD1, a dual-target inhibitor, to concurrently suppress the p38-MK2 pathway and microtubule dynamicity. In addition to established role as an MK2 inhibitor, we find that CMPD1 rapidly induces microtubule depolymerization, preferentially at the microtubule plus-end, leading to the inhibition of tumor growth and cancer cell invasion in both in vitro and in vivo models. Notably, 10 nM CMPD1 is sufficient to induce irreversible mitotic defects in cancer cells, but not in non-transformed RPE1 cells, highlighting its high specificity to cancer cells. We further validate that a specific p38-MK2 inhibitor significantly potentiates the efficacy of sub-clinical concentrations of MTA. In summary, our findings suggest that the p38-MK2 pathway presents a promising therapeutic target in combination with MTAs in cancer treatment.
Collapse
Affiliation(s)
- Yu-Chia Chen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- These authors contributed equally
| | - Mamoru Takada
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
- These authors contributed equally
| | - Aerica Nagornyuk
- Department of Biomedical Science, University of North Dakota School of Medicine and Health Science, Grand Folks, North Dakota, USA
| | - Wu Muhan
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hideyuki Yamada
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takeshi Nagashima
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masayuki Ohtsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jennifer G. DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Steven Markus
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, Colorado, USA
| | - Motoki Takaku
- Department of Biomedical Science, University of North Dakota School of Medicine and Health Science, Grand Folks, North Dakota, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
29
|
Kitadai R, Shimoi T, Yazaki S, Okuma HS, Hoshino M, Ito M, Saito A, Kita S, Kojima Y, Nishikawa T, Sudo K, Noguchi E, Fujiwara Y, Yoshida M, Yonemori K. Clinicopathological and prognostic features of HER2-null and HER2-low advanced breast cancer treated with eribulin or capecitabine. Breast Cancer 2024; 31:1037-1045. [PMID: 39141076 PMCID: PMC11489188 DOI: 10.1007/s12282-024-01617-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND HER2-low populations constitute a heterogeneous group, and the cytotoxic anticancer agent efficacy based on HER2 status remains unclear. This study evaluated the clinicopathological features and outcomes of patients with advanced breast cancer showing HER2-low expression treated with eribulin or capecitabine, two treatment options after anthracycline and taxane treatment. METHODS We retrospectively evaluated patients who were treated with eribulin or capecitabine between 2011 and 2015. HER2 status was evaluated according to the ASCO/CAP guidelines. RESULTS No significant difference was observed in overall survival (OS; eribulin: hazard ratio [HR], 0.66; 95% CI 0.40-1.10; capecitabine: HR, 0.76; 95% CI 0.45-1.30) or progression-free survival (PFS; eribulin: HR, 1.13; 95% CI 0.72-1.78; capecitabine: HR, 0.90; 95% CI 0.56-1.44) between patients receiving eribulin (HER2-null: 35, HER2-low: 44) and those receiving capecitabine (HER2-null: 41, HER2-low: 33). Subgroup analysis revealed no significant differences in OS between the two groups in the hormone-positive and -negative populations for eribulin and capecitabine. HER2-null and HER2-low patients showed objective response rates (ORRs) of 22.5% and 9.1% (p = 0.09) overall, and 32.0% and 10.5% (p = 0.03), respectively, in hormone-positive cases among eribulin-treated patients. No response was observed in hormone-negative patients. Capecitabine treatment in HER2-null and HER2-low patients had overall ORRs of 26.8% and 15.2% (p = 0.23), respectively, with 27.3% and 16.1% (p = 0.28) for hormone-positive cases; and 25.0% and 0% (p = 1.0), respectively, for hormone-negative cases. CONCLUSIONS Eribulin and capecitabine sensitivity may vary based on HER2 expression in patients with HER2-low and HER2-null breast cancer. Prognosis was similar between the HER2-low and the HER2-null groups.
Collapse
Affiliation(s)
- Rui Kitadai
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tatsunori Shimoi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - Shu Yazaki
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hitomi Sumiyoshi Okuma
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Mai Hoshino
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Munehiro Ito
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ayumi Saito
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shosuke Kita
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yuki Kojima
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tadaaki Nishikawa
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazuki Sudo
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Emi Noguchi
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Masayuki Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Kan Yonemori
- Department of Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
30
|
Teng NMY, Malfettone A, Dalby MJ, Kiu R, Seki D, Robinson T, Gion M, Bermejo B, Pérez-García JM, Prat A, Vázquez RM, Llombart-Cussac A, Curigliano G, Schmid P, Barroso-Sousa R, Mancino M, Shimizu E, Rodríguez-Morató J, Mina L, Hall LJ, Robinson SD, Cortés J. Profiling the gut and oral microbiota of hormone receptor-positive, HER2-negative metastatic breast cancer patients receiving pembrolizumab and eribulin. MICROBIOME RESEARCH REPORTS 2024; 4:4. [PMID: 40207279 PMCID: PMC11977384 DOI: 10.20517/mrr.2024.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 04/11/2025]
Abstract
Aim: Changes in host-associated microbial communities (i.e., the microbiota) may modulate responses to checkpoint blockade immunotherapy. In the KELLY phase II study (NCT03222856), we previously demonstrated that pembrolizumab [anti-programmed cell death protein 1 (PD-1)] combined with eribulin (plus microtubule-targeting chemotherapy) showed encouraging antitumor activity in patients with hormone receptor (HR)-positive/human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer (mBC) who had received prior treatments. Methods: A total of 58 fecal and 67 saliva samples were prospectively collected from a subset of 28 patients at baseline (BL), after three treatment cycles, and end of treatment. Shotgun metagenomics, 16S rRNA gene amplicon sequencing, and bioinformatics and statistical approaches were used to characterize fecal and oral microbiota profiles. Results: Treatment caused no substantial perturbations in gut or oral microbiota, suggesting minimal drug-related microbial toxicity. Bacteroides and Faecalibacterium were the dominant gut microbiota genera, while Prevotella and Streptococcus were present in both oral and gut samples, highlighting potential gut-oral microbial interactions. Additionally, clinical benefit (CB) appeared to be associated with gut-associated Bacteroides fragilis (B. fragilis) and a BL oral abundance of Streptococcus ≥ 30%. Notably, B. fragilis NCTC 9343 supernatant induced dose-dependent lactate dehydrogenase (LDH) release from the MCF-7 (HR-positive/HER2-negative) BC cell line. Conclusion: These findings suggest that specific gut and oral microbiota may modulate the effectiveness of combinatory anti-BC therapies, potentially through the action of microbial metabolites.
Collapse
Affiliation(s)
- Nancy MY Teng
- Gut Microbes&Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Andrea Malfettone
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
| | - Matthew J Dalby
- Institute of Microbiology and Infection, University of Birmingham, Birmingham B12 2TT, UK
| | - Raymond Kiu
- Gut Microbes&Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham B12 2TT, UK
| | - David Seki
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food&Health Technical University of Munich, Freising D-80333, Germany
| | - Tim Robinson
- Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - María Gion
- Medical Onology Department, Hospital Universitario Ramón y Cajal, Madrid 28034, Spain
| | - Begoña Bermejo
- Medical Oncology, Hospital Clínico de Valencia, INCLIVA, CIBERONC, Medicine Department, Universidad de Valencia, Valencia 46010, Spain
| | - José Manuel Pérez-García
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quirón Group, Barcelona 08017, Spain
| | - Aleix Prat
- Medical Oncology Department, Hospital Clínic y Provincial de Barcelona, Barcelona 08036, Spain
| | | | - Antonio Llombart-Cussac
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
- Department of medical oncology, Hospital Arnau de Vilanova, FISABIO, Valencia 46800, Spain
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, University of Milano, Milano 20141, Italy
- Department of Oncology and Hemato-Oncology, University of Milano, Milano 20122, Italy
| | - Peter Schmid
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK
| | | | - Mario Mancino
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
| | - Eileen Shimizu
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
| | | | - Leonardo Mina
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
| | - Lindsay J Hall
- Gut Microbes&Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham B12 2TT, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL - Institute for Food&Health Technical University of Munich, Freising D-80333, Germany
- Norwich Medical School, University of East Anglia, Norwich NR4 7TJ, UK
| | - Stephen D Robinson
- Gut Microbes&Health, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Javier Cortés
- Medica Scientia Innovation Research (MEDSIR), Barcelona 08005, Spain
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quirón Group, Barcelona 08017, Spain
- Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid 28670, Spain
| |
Collapse
|
31
|
Yadav SK, Leon-Ferre RA. Current treatment paradigms for triple-negative breast cancer. Minerva Med 2024; 115:589-598. [PMID: 39016529 DOI: 10.23736/s0026-4806.24.09458-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Triple negative breast cancer (TNBC) accounts for 15% of all breast cancers and is characterized by more aggressive biology, significant disease heterogeneity, and worse clinical outcomes. In recent years, improved understanding of TNBC tumor biology and its heterogeneity have led to the identification of new molecular targets, opening avenues for novel treatment strategies. Clinical trials evaluating immunotherapy, poly-ADP ribose polymerase (PARP) inhibitors, and antibody drug conjugates have shown improvement in clinical outcomes, leading to their incorporation to the treatment options available for patients with TNBC. This review aimed to provide the internal medicine specialist and primary care provider with a comprehensive overview of the current systemic therapy approaches for TNBC and introduce clinicians to novel therapies that have recently been added to the treatment armamentarium against this disease.
Collapse
Affiliation(s)
- Sumeet K Yadav
- Department of Hospital Internal Medicine, Mayo Clinic Health System, Mankato, MN, USA -
| | | |
Collapse
|
32
|
El Kaddissi A, Vernerey D, Falcoz A, Mansi L, Bazan F, Chaigneau L, Dobi E, Goujon M, Meneveau N, Paillard MJ, Selmani Z, Viot J, Molimard C, Monnien F, Woronoff AS, Curtit E, Borg C, Meynard G. Prognostic Factors for Long-Term Eribulin Response in a Cohort of Patients With HER2-Negative Metastatic Breast Cancer. Clin Breast Cancer 2024; 24:e622-e632.e5. [PMID: 38972830 DOI: 10.1016/j.clbc.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/03/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
CONTEXT AND AIMS Eribulin is used in taxane and anthracycline refractory HER2-negative metastatic breast cancers (MBC). Patients treated in pivotal clinical trials achieved low survival rates, therefore, the identification of prognostic criteria for long progression-free survival (PFS) is still an unmet medical need. In this study, we sought to determine potential prognostic criteria for long-term eribulin response in HER2-negative MBC. METHODS Our retrospective cohort includes female patients with HER2-negative MBC treated with eribulin in Franche-Comté, France. We defined a long-term response as at least 6 months of eribulin treatment. The primary endpoint was the analysis of criteria that differ according to the progression-free survival. Secondary outcomes concerned overall survival and response rate. RESULTS From January 2011 to April 2020, 431 patients treated with eribulin were screened. Of them, 374 patients were included. Median PFS was 3.2 months (2.8-3.7). Eighty-eight patients (23.5%) had a long-term response to eribulin. Four discriminant criteria allowed to separate PFS in 2 arms (PFS < 3 months or > 6 months) with a 78% positive predictive value: histological grade, absence of meningeal metastasis, response to prior chemotherapy, and OMS status. We have developed a nomogram combining these 4 criteria. Median overall survival was 8.5 months (7.0-9.5). CONCLUSION Eribulin response in MBC can be driven by clinical and biological factors. Application of our nomogram could assist in the prescription of eribulin.
Collapse
Affiliation(s)
- Antoine El Kaddissi
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France.
| | - Dewi Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France; INSERM, Besançon 25020, France
| | - Antoine Falcoz
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France; INSERM, Besançon 25020, France
| | - Laura Mansi
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Fernando Bazan
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Loïc Chaigneau
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Erion Dobi
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Morgan Goujon
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Nathalie Meneveau
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Marie-Justine Paillard
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Zohair Selmani
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Julien Viot
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France; Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France; INSERM, Besançon 25020, France
| | - Chloé Molimard
- Department of Pathology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Franck Monnien
- Department of Pathology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Anne-Sophie Woronoff
- Cancer Registry of Doubs, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| | - Elsa Curtit
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France; INSERM, Besançon 25020, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France; INSERM, Besançon 25020, France
| | - Guillaume Meynard
- Department of Medical Oncology, University Hospital of Besançon Jean-Minjoz, Besançon 25030, France
| |
Collapse
|
33
|
Ni M, Zhou L, Lu Y, Guo D, Li X, Li L, Zhang L, Chen M, Zhang L, Xu F, Yuan Z, Wang S, Shi Y, Yang A, An X. Eribulin plus carboplatin combination for HER2-negative metastatic breast cancer: a multicenter, real-world cohort study. BMC Cancer 2024; 24:1214. [PMID: 39350055 PMCID: PMC11443731 DOI: 10.1186/s12885-024-12953-9] [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: 03/29/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Pre-clinical data suggests a potential synergistic effect of eribulin and platinum. However, clinical data on the combination for metastatic breast cancer (mBC) is lacking. We evaluated the efficacy and safety of eribulin plus carboplatin (ErCb) in patients with mBC. PATIENTS AND METHODS This multicenter, real-world cohort study included patients with pre-treated metastatic triple negative breast cancer (TNBC) or endocrine-refractory hormone receptor (HR) positive, HER2-negative mBC who received ErCb. Eribulin (1.4 mg/m2) and carboplatin (target AUC = 2) were administered intravenously on day 1 and 8 of 21-day cycle. Objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were evaluated. RESULTS From March 2022 to December 2023, a cohort of 37 patients were recruited to the study. Among them, 22 patients have TNBC and 15 have HR + HER2 - mBC. Of the 22 patients with TNBC, 8 had an initial diagnosis of the HR + HER2 - subtype. The median treatment was 6 cycles (range, 2 - 8 cycles). In the full cohort, TNBC, and HR + HER2 - subgroup, the ORR were 51.4%, 54.5% and 46.7%, the DCR were 81.1%, 81.8% and 80%, and the median PFS were 5 months, 5 months, and 5.2 months, respectively. The median OS was 12.7 months in the entire cohort and 12.8 months in TNBC subgroup. The most common grade 3/4 hematological AEs were neutropenia (37.8%), leukopenia (35.1%), febrile neutropenia (10.8%), thrombocytopenia (5.4%), and anemia (2.7%). No grade 3/4 non-hematological AEs were observed. CONCLUSION ErCb demonstrated favorable efficacy and tolerability in patients with heavily pre-treated mBC, especially TNBC. The findings of the current study warrant further investigation of the application of this combination in earlier lines of mBC treatment.
Collapse
Affiliation(s)
- Mengqian Ni
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Lijia Zhou
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Yongkui Lu
- Affiliated Cancer Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Dachuan Guo
- Department of Minimally Invasive Interventional Therapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiuyue Li
- Central Hospital of Pan-Yu, Guangzhou, 511486, China
| | - Lixia Li
- Cancer Hospital, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Lidong Zhang
- Puning People's Hospital, Jieyang, Guangdong, 515300, China
| | - Meiting Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Lulu Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Fei Xu
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Zhongyu Yuan
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Shusen Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China
| | - Yanxia Shi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China.
| | - Anli Yang
- Department of Breast Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China.
| | - Xin An
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, No. 651, Dongfeng Road East, Yuexiu District, Guangzhou, 510060, China.
| |
Collapse
|
34
|
Carrión-Madroñal IM, Díaz-Acedo R, Lora-Escobar SJ, Naranjo-Llamas E, Jaramillo-Ruiz D, Artacho-Criado S, Prado-Mel E. Sacituzumab-govitecan in metastatic triple-negative breast cancer: a multicenter effectiveness and safety study. Future Oncol 2024; 20:2565-2572. [PMID: 39263951 PMCID: PMC11534107 DOI: 10.1080/14796694.2024.2394408] [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: 02/26/2024] [Accepted: 08/16/2024] [Indexed: 09/13/2024] Open
Abstract
Aim: Sacituzumab-govitecan (Sgov) is a new antibody-drug conjugate recently approved for metastatic triple negative breast cancer (mTNBC), so there are still few data published in the real-world setting.Materials & methods: This study was to analyze the effectiveness and safety of Sgov in mTNBC of patients from the three main hospitals of a city and to compare with the pivotal ASCENT-trial. A total of 46 patients were included, all women diagnosed with mTNBC, with a median age of 52 years and Eastern Cooperative Oncology Group performance status 0-1 71.8% of patients.Results: Sgov effectiveness data seem to be slightly inferior than expected. Furthermore, it is observed that patients with an Eastern Cooperative Oncology Group of two or higher benefit significantly less from treatment with the drug. Safety profile of Sgov is acceptable.
Collapse
Affiliation(s)
| | - Rocío Díaz-Acedo
- Servicio de Farmacia, Hospital Universitario Virgen del Rocío, Seville, 41013, SPAIN
| | | | | | | | | | - Elena Prado-Mel
- Servicio de Farmacia, Hospital Universitario Virgen del Rocío, Seville, 41013, SPAIN
| |
Collapse
|
35
|
Tolaney SM, Punie K, Carey LA, Kurian AW, Ntalla I, Sjekloca N, Shah A, Rehnquist MK, Stokes M, Fraeman K, Verret W, Jhaveri K. Real-world treatment patterns and outcomes in patients with HR+/HER2- metastatic breast cancer treated with chemotherapy in the United States. ESMO Open 2024; 9:103691. [PMID: 39241499 PMCID: PMC11406087 DOI: 10.1016/j.esmoop.2024.103691] [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: 06/20/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Until recently, treatment options for patients with hormone receptor-positive/human epidermal growth factor 2-negative (HR+/HER2-) metastatic breast cancer (mBC) and resistance to endocrine therapy were limited to chemotherapy. This real-world study describes treatment patterns and outcomes in patients treated with chemotherapy in the United States before approval of antibody-drug conjugates. PATIENTS AND METHODS This retrospective, observational study included adults with HR+/HER2- mBC from the ConcertAI Patient360™ Breast Cancer dataset who initiated their first chemotherapy in the metastatic setting between January 2011 and June 2021. Treatment patterns were described; real-world overall survival, time to next treatment or death, and real-world progression-free survival were evaluated for all eligible patients and patients treated with subsequent chemotherapy. Index dates were the start date of each chemotherapy treatment. RESULTS Among 1545 eligible patients, 76% were white, 12% had Eastern Cooperative Oncology Group performance status ≥2, 38% had de novo mBC, and median age was 61 years (range, 52-69 years). Within the index period, capecitabine was used the most as the first chemotherapy agent and decreased in later treatments, while the use of eribulin increased between first and fourth chemotherapies. Median (95% confidence interval) real-world overall survival was 23.3 months (21.3-25.4 months) from start of first chemotherapy, time to next treatment or death was 6.5 months (5.9-7.1 months), and real-world progression-free survival was 6.9 months (6.4-7.6 months); median times from second, third, and fourth chemotherapies decreased with each additional chemotherapy treatment. CONCLUSIONS This real-world study demonstrates that for patients with HR+/HER2- mBC, chemotherapy provides relatively limited survival benefit which decreases with each additional chemotherapy line, and highlights the need for improved treatment options.
Collapse
Affiliation(s)
- S M Tolaney
- Breast Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA.
| | - K Punie
- GZA Hospitals Sint-Augustinus, Wilrijk, Belgium
| | - L A Carey
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, USA
| | - A W Kurian
- Stanford University School of Medicine, Stanford, USA
| | - I Ntalla
- Gilead Sciences Europe Ltd., Stockley Park, UK
| | - N Sjekloca
- Gilead Sciences Europe Ltd., Stockley Park, UK
| | - A Shah
- Gilead Sciences, Inc., Foster City, USA
| | | | | | | | - W Verret
- Gilead Sciences, Inc., Foster City, USA
| | - K Jhaveri
- Memorial Sloan Kettering Cancer Center (MSKCC), New York, USA; Weill Cornell Medical College, New York, USA
| |
Collapse
|
36
|
Yamashita T, Sohn JH, Tokunaga E, Niikura N, Park YH, Lee KS, Chae YS, Xu B, Wang X, Im SA, Li W, Lu YS, Aguilar CO, Nishijima S, Nishiyama Y, Sugihara M, Modi S, Tsurutani J. Trastuzumab deruxtecan versus treatment of physician's choice in previously treated Asian patients with HER2-low unresectable/metastatic breast cancer: subgroup analysis of the DESTINY-Breast04 study. Breast Cancer 2024; 31:858-868. [PMID: 38884900 PMCID: PMC11341650 DOI: 10.1007/s12282-024-01600-7] [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: 03/29/2024] [Accepted: 05/25/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND In the global phase 3 DESTINY-Breast04 study (NCT03734029), the anti-human epidermal growth factor 2 (HER2) antibody-drug conjugate trastuzumab deruxtecan (T-DXd) demonstrated a statistically significant improvement in progression-free survival (PFS) and overall survival (OS), with manageable safety compared with treatment of physician's choice (TPC) in patients with HER2-low metastatic breast cancer (mBC) who had received 1-2 prior lines of chemotherapy. METHODS This subgroup analysis examined the efficacy and safety of T-DXd versus TPC in 213 patients from Asian countries and regions who were enrolled in the DESTINY-Breast04 trial and randomized to T-DXd (n = 147) or TPC (n = 66). RESULTS Median PFS with T-DXd and TPC was 10.9 and 5.3 months, respectively, in Asian patients with hormone receptor-positive mBC, and 10.9 and 4.6 months, respectively, in the overall Asian population. In both populations, median OS was not reached with T-DXd and was 19.9 months with TPC. The objective response rate was higher with T-DXd versus TPC in all Asian patients. Median treatment duration was 8.4 months with T-DXd and 3.5 months with TPC. The most common grade ≥ 3 drug-related treatment-emergent adverse events in Asian patients treated with T-DXd were neutropenia (16.3%), anemia (12.9%), and leukopenia (11.6%); the incidences of neutropenia and leukopenia were higher with TPC versus T-DXd. Adjudicated drug-related interstitial lung disease or pneumonitis with T-DXd was 14.3%; the majority of events were grade 1-2. CONCLUSIONS T-DXd demonstrated clinically meaningful survival benefits versus TPC in Asian HER2-low mBC patients, regardless of hormone receptor status, with no new safety signals. CLINICAL TRIAL REGISTRATION NUMBER ClinicalTrials.gov, NCT03734029.
Collapse
Affiliation(s)
| | - Joo Hyuk Sohn
- Yonsei University Health System, Seoul, Republic of Korea
| | | | - Naoki Niikura
- Tokai University School of Medicine Hospital, Kanagawa, Japan
| | | | - Keun Seok Lee
- National Cancer Center, Gyeonggi-do, Republic of Korea
| | - Yee Soo Chae
- Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Binghe Xu
- Cancer Hospital Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | | | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wei Li
- The First Hospital of Jilin University, Jilin, China
| | - Yen-Shen Lu
- National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | - Shanu Modi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Junji Tsurutani
- The Innovative Center of Translational Research and Clinical Science for Cancer Therapy, Showa University Hospital, Tokyo, Japan.
- Advanced Cancer Translational Research Institute, Showa University, Tokyo, Japan.
| |
Collapse
|
37
|
Kranthikumar R, Kishi Y. Application of Ni/Zr-Mediated Ketone Coupling for the Scalable Synthesis of Homohalichondrin B. Org Lett 2024; 26:7105-7109. [PMID: 39158056 DOI: 10.1021/acs.orglett.4c02297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
A practical and improved synthetic route for the scalable synthesis of homohalichondrin B has been developed. Formation of the highly stereoselective [6,6]-spiroskeleton system through a convergent Ni/Zr-mediated ketone coupling, followed by acid-promoted spiroketalization, simplified the route considerably. Higher coupling efficiency was achieved by fine-tuning the NiI/NiII (1:50) catalyst loadings with an equimolar (1:1) mixture of coupling partners.
Collapse
Affiliation(s)
- Ramagonolla Kranthikumar
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| | - Yoshito Kishi
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
38
|
Shi X, Wang X, Yao W, Shi D, Shao X, Lu Z, Chai Y, Song J, Tang W, Wang X. Mechanism insights and therapeutic intervention of tumor metastasis: latest developments and perspectives. Signal Transduct Target Ther 2024; 9:192. [PMID: 39090094 PMCID: PMC11294630 DOI: 10.1038/s41392-024-01885-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 08/04/2024] Open
Abstract
Metastasis remains a pivotal characteristic of cancer and is the primary contributor to cancer-associated mortality. Despite its significance, the mechanisms governing metastasis are not fully elucidated. Contemporary findings in the domain of cancer biology have shed light on the molecular aspects of this intricate process. Tumor cells undergoing invasion engage with other cellular entities and proteins en route to their destination. Insights into these engagements have enhanced our comprehension of the principles directing the movement and adaptability of metastatic cells. The tumor microenvironment plays a pivotal role in facilitating the invasion and proliferation of cancer cells by enabling tumor cells to navigate through stromal barriers. Such attributes are influenced by genetic and epigenetic changes occurring in the tumor cells and their surrounding milieu. A profound understanding of the metastatic process's biological mechanisms is indispensable for devising efficacious therapeutic strategies. This review delves into recent developments concerning metastasis-associated genes, important signaling pathways, tumor microenvironment, metabolic processes, peripheral immunity, and mechanical forces and cancer metastasis. In addition, we combine recent advances with a particular emphasis on the prospect of developing effective interventions including the most popular cancer immunotherapies and nanotechnology to combat metastasis. We have also identified the limitations of current research on tumor metastasis, encompassing drug resistance, restricted animal models, inadequate biomarkers and early detection methods, as well as heterogeneity among others. It is anticipated that this comprehensive review will significantly contribute to the advancement of cancer metastasis research.
Collapse
Affiliation(s)
- Xiaoli Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xinyi Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, China
| | - Dongmin Shi
- Department of Medical Oncology, Shanghai Changzheng Hospital, Shanghai, China
| | - Xihuan Shao
- The Fourth Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengqing Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Yue Chai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jinhua Song
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
- School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
39
|
Vicari HP, Gomes RDC, Lima K, Rossini NDO, Rodrigues Junior MT, de Miranda LBL, Dias MVB, Costa-Lotufo LV, Coelho F, Machado-Neto JA. Cyclopenta[b]indoles as novel antimicrotubule agents with antileukemia activity. Toxicol In Vitro 2024; 99:105856. [PMID: 38821378 DOI: 10.1016/j.tiv.2024.105856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Acute leukemias present therapeutic challenges despite advances in treatments. Microtubule inhibitors have played a pivotal role in cancer therapy, inspiring exploration into novel compounds like C2E1 from the cyclopenta[b]indole class. In the present study, we investigated C2E1's potential as a therapeutic agent for acute leukemia at molecular, cellular, and genetic levels. C2E1 demonstrated tubulin depolarization activity, significantly reducing leukemia cell viability. Its impact involved multifaceted mechanisms: inducing apoptosis, arrest of cell cycle progression, and inhibition of clonogenicity and migration in leukemia cells. At a molecular level, C2E1 triggered DNA damage, antiproliferative, and apoptosis markers and altered gene expression related to cytoskeletal regulation, disrupting essential cellular processes crucial for leukemia cell survival and proliferation. These findings highlight C2E1's promise as a potential candidate for novel anti-cancer therapies. Notably, its distinct mode of action from conventional microtubule-targeting drugs suggests the potential to bypass common resistance mechanisms encountered with existing treatments. In summary, C2E1 emerges as a compelling compound with diverse effects on leukemia cells, showcasing promising antineoplastic properties. Its ability to disrupt critical cellular functions selective to leukemia cells positions it as a candidate for future therapeutic development.
Collapse
Affiliation(s)
- Hugo Passos Vicari
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ralph da Costa Gomes
- Department of Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas, São Paulo, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fernando Coelho
- Department of Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas, São Paulo, Brazil.
| | | |
Collapse
|
40
|
Ramos A, Bizri N, Novak E, Mollen K, Khan S. The role of cGAS in epithelial dysregulation in inflammatory bowel disease and gastrointestinal malignancies. Front Pharmacol 2024; 15:1409683. [PMID: 39050748 PMCID: PMC11266671 DOI: 10.3389/fphar.2024.1409683] [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: 03/30/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024] Open
Abstract
The gastrointestinal tract is lined by an epithelial monolayer responsible for selective permeability and absorption, as well as protection against harmful luminal contents. Recognition of foreign or aberrant DNA within these epithelial cells is, in part, regulated by pattern recognition receptors such as cyclic GMP-AMP synthase (cGAS). cGAS binds double-stranded DNA from exogenous and endogenous sources, resulting in the activation of stimulator of interferon genes (STING) and a type 1 interferon response. cGAS is also implicated in non-canonical pathways involving the suppression of DNA repair and the upregulation of autophagy via interactions with PARP1 and Beclin-1, respectively. The importance of cGAS activation in the development and progression of inflammatory bowel disease and gastrointestinal cancers has been and continues to be explored. This review delves into the intricacies of the complex role of cGAS in intestinal epithelial inflammation and gastrointestinal malignancies, as well as recent therapeutic advances targeting cGAS pathways.
Collapse
Affiliation(s)
- Anna Ramos
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Nazih Bizri
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Elizabeth Novak
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Kevin Mollen
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Division of Pediatric General and Thoracic Surgery, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Sidrah Khan
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Yamada H, Takada M, Ghone D, Yu M, Nagashima T, Fujimoto H, Sakakibara J, Hasegawa Y, Takao S, Yamada A, Narui K, Ishikawa T, Suzuki A, Otsuka M. Eribulin induces micronuclei and enhances the nuclear localization of cGAS in triple-negative breast cancer cells. Sci Rep 2024; 14:14146. [PMID: 38898119 PMCID: PMC11187130 DOI: 10.1038/s41598-024-64651-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/11/2024] [Indexed: 06/21/2024] Open
Abstract
Eribulin (ERI), clinically utilized for locally advanced or metastatic breast tumors, has shown potential links to the immune system. Notably, the cGAS-STING pathway, a key component of innate immunity, has gained prominence. Yet, limited reports explore ERI's effects on the cGAS-STING pathway. Additionally, the nuclear presence of cGAS remains poorly understood. This study uniquely delves into ERI's impact on both the cytosolic cGAS-STING pathway and nuclear cGAS. ERI enhances nuclear localization of cGAS, resulting in hyper-activation of the cGAS-STING pathway in triple-negative breast cancer cells. Reduction of cGAS heightened both cell proliferation and ERI sensitivity. In clinical data using ERI in a neo-adjuvant setting, patients with low cGAS cases exhibited reduced likelihood of achieving pathological complete response after ERI treatment. These findings illuminate the potential of cGAS and IFNβ as predictive biomarkers for ERI sensitivity, providing valuable insights for personalized breast cancer treatment strategies.
Collapse
Affiliation(s)
- Hideyuki Yamada
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Mamoru Takada
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan.
| | - Dhaval Ghone
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Muhan Yu
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Takeshi Nagashima
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Hiroshi Fujimoto
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Junta Sakakibara
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| | - Yoshie Hasegawa
- Department of Breast Surgery, Hachinohe City Hospital, Hachinohe, Aomori, Japan
| | - Shintaro Takao
- Department of Breast Surgery, Konan Medical Center, Kobe, Hyogo, Japan
| | - Akimitsu Yamada
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kazutaka Narui
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Takashi Ishikawa
- Department of Breast Oncology and Surgery, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Aussie Suzuki
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Masayuki Otsuka
- Department of General Surgery, Graduate School of Medicine, Chiba University, Chiba, Chiba, Japan
| |
Collapse
|
44
|
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
|
45
|
Miglietta F, Pontolillo L, De Angelis C, Caputo R, Marino M, Bria E, Di Rienzo R, Verrazzo A, Buonerba C, Tortora G, Di Lorenzo G, Del Mastro L, Giuliano M, Montemurro F, Puglisi F, Guarneri V, De Laurentiis M, Scafuri L, Arpino G. Gender minorities in breast cancer - Clinical trials enrollment disparities: Focus on male, transgender and gender diverse patients. Breast 2024; 75:103713. [PMID: 38493590 PMCID: PMC10959718 DOI: 10.1016/j.breast.2024.103713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND The last years have seen unprecedented improvement in breast cancer (BC) survival rates. However, this entirely apply to female BC patients, since gender minorities (male, transgender/gender-diverse) are neglected in BC phase III registration clinical trials. METHODS We conducted a scoping review of phase III clinical trials of agents with a current positioning within the therapeutic algorithms of BC. RESULTS We selected 51 phase III trials. Men enrollment was allowed in 35.3% of trials. In none of the trial inclusion/exclusion criteria referred to transgender/gender-diverse people. A numerical higher rate of enrolled men was observed in the contemporary as compared to historical group. We found a statistically significant association between the drug class and the possibility of including men: 100%, 80%, 50%, 33.3%, 25%, 10% and 9.1% of trials testing ICI/PARP-i, ADCs, PI3K/AKT/mTOR-i, anti-HER2 therapy, CDK4/6-i, ET alone, and CT alone. Overall, 77409 patients were enrolled, including 112 men (0.2%). None of the trial reported transgender/gender-diverse people proportion. Studies investigating PARP-i were significantly associated with the highest rate of enrolled men (1.42%), while the lowest rates were observed for trials of CT (0.13%), ET alone (0.10%), and CDK 4/6-I (0.08%), p < 0.001. CONCLUSIONS We confirmed that gender minorities are severely underrepresented among BC registration trials. We observed a lower rate of men in trials envisaging endocrine manipulation or in less contemporary trials. This work sought to urge the scientific community to increase the awareness level towards the issue of gender minorities and to endorse more inclusive criteria in clinical trials.
Collapse
Affiliation(s)
- Federica Miglietta
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Oncology 2 Unit, Istituto Oncologico Veneto - IOV IRCCS, Padova, Italy
| | - Letizia Pontolillo
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Medical Oncology, Department of Traslational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Roberta Caputo
- Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Pascale, Naples, Italy
| | - Monica Marino
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Oncology 2 Unit, Istituto Oncologico Veneto - IOV IRCCS, Padova, Italy
| | - Emilio Bria
- Medical Oncology, Department of Traslational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Rossana Di Rienzo
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Annarita Verrazzo
- Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Pascale, Naples, Italy
| | - Carlo Buonerba
- Oncology Unit, Hospital "Andrea Tortora", ASL Salerno, Pagani, Italy; Associazione O.R.A. ETS - Oncology Research Assistance, Salerno, Italy
| | - Giampaolo Tortora
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Medical Oncology, Department of Traslational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Di Lorenzo
- Oncology Unit, Hospital "Andrea Tortora", ASL Salerno, Pagani, Italy; Associazione O.R.A. ETS - Oncology Research Assistance, Salerno, Italy
| | - Lucia Del Mastro
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Internal Medicine and Medical Specialities, School of Medicine, University of Genoa, Genoa, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | | | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy; Oncology 2 Unit, Istituto Oncologico Veneto - IOV IRCCS, Padova, Italy
| | - Michelino De Laurentiis
- Department of Breast and Thoracic Oncology, Division of Breast Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Pascale, Naples, Italy
| | - Luca Scafuri
- Oncology Unit, Hospital "Andrea Tortora", ASL Salerno, Pagani, Italy; Associazione O.R.A. ETS - Oncology Research Assistance, Salerno, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.
| |
Collapse
|
46
|
Grammoustianou M, Dimitrakopoulos FI, Koutras A. Current Status and Future Perspectives of Antibody-Drug Conjugates in Hormone Receptor-Positive Breast Cancer. Cancers (Basel) 2024; 16:1801. [PMID: 38791880 PMCID: PMC11120191 DOI: 10.3390/cancers16101801] [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: 02/25/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer is the most common cancer type in women. The vast majority of breast cancer patients have hormone receptor-positive (HR+) tumors. In advanced HR+ breast cancer, the combination of endocrine therapy with cyclin-dependent kinase 4/6 (CDK4/6) inhibitors is considered the standard of care in the front-line setting. Nevertheless, resistance to hormonal therapy and CDK4/6 inhibitors eventually occurs, leading to progression of the disease. Antibody-drug conjugates (ADCs) comprise a promising therapeutic choice with significant efficacy in patients with HR+ breast cancer, which is resistant to endocrine treatment. ADCs typically consist of a cytotoxic payload attached by a linker to a monoclonal antibody that targets a specific tumor-associated antigen, offering the advantage of a more selective delivery of chemotherapy to cancer cells. In this review, we focus on the ADC mechanisms of action, their toxicity profile and therapeutic uses as well as on related biomarkers and future perspectives in advanced HR+ breast cancer.
Collapse
Affiliation(s)
- Maria Grammoustianou
- Oncology Department, Sotiria General Hospital, 115 27 Athens, Greece;
- Breast Cancer Survivorship Research Group, Gustave Roussy, 94805 Villejuif, France
| | | | - Angelos Koutras
- Division of Oncology, Department of Medicine, University Hospital, Medical School, University of Patras, 265 04 Patras, Greece;
| |
Collapse
|
47
|
Chen L, Yan X, Luo T, Tian T, He P, Zhong X. Efficacy and safety of eribulin mesylate in patients with locally advanced or metastatic breast cancer previously treated with anthracycline/taxanes. Cancer Med 2024; 13:e7295. [PMID: 38785215 PMCID: PMC11117449 DOI: 10.1002/cam4.7295] [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: 10/09/2023] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND This prospective real-world study aimed to assess the efficacy and safety of eribulin in the clinical practice against advanced breast cancer (ABC) in China. PATIENTS AND METHODS In this study, eligible patients with inoperable locally advanced or metastatic breast cancer who had experienced prior neo-/adjuvant or failed the palliative treatment with anthracycline/taxanes were included. Eribulin (1.4 mg/m2) was infused intravenously on Day 1 and Day 8 every 3 weeks until disease progression or intolerable toxicity occurred. The progression-free survival (PFS), overall response rate (ORR), disease control rate (DCR), and safety of the treatment were assessed. RESULTS One hundred and thirty-four patients were enrolled. The median PFS (mPFS) was 4.3 months (95% CI: 0.3-15.4). The ORR and DCR was 32.1% and 79.1%, respectively. The mPFS of patients who received eribulin as first- or second-line treatment was significantly better than those who received eribulin as ≥3-line treatment (6.9 months [95% CI: 3.2-8.8] vs. 4.0 months [95% CI: 3.4-4.6], p = 0.006). The mPFS of patients with triple-negative, HER2-positive, and HER2(-)/HR(+) was 3.4 (95% CI: 2.7-4.1), 6.2 (95% CI: 2.3-10.1) and 5.0 months (95% CI: 4.1-5.9), respectively. HER2(+) patients had significantly longer PFS than TNBC patients (p = 0.022). Patients received combination therapy had a significantly longer mPFS than those who received eribulin monotherapy (5.0 months [95% CI 3.6-6.3] vs. 4.0 months [95% CI: 3.3-4.7] [p = 0.016]). Multivariate analysis revealed that MBC patients with a molecular typing of non-TNBC receiving eribulin as ≤2-line therapy and combination therapy had a low risk of disease progression. Neutropenia (33.58%), leukopenia (11.94%), and thrombocytopenia (4.48%) were the most common treatment-related adverse events. CONCLUSION Eribulin demonstrated effective clinical activity and a favorable tolerability profile in Chinese patients with ABC in the real-world. The efficacy and safety profile were consistent with those reported in previous randomized phase 3 trials.
Collapse
Affiliation(s)
- Lan Chen
- Department of Medical Oncology, West China HospitalSichuan UniversityChengduSichuan ProvincePeople's Republic of China
| | - Xi Yan
- Department of Medical OncologyHead and Neck Cancer Department, West China HospitalChengduSichuan ProvincePeople's Republic of China
| | - Ting Luo
- Department of Medical OncologyHead and Neck Cancer Department, West China HospitalChengduSichuan ProvincePeople's Republic of China
| | - Tinglun Tian
- Department of Medical OncologyHead and Neck Cancer Department, West China HospitalChengduSichuan ProvincePeople's Republic of China
| | - Ping He
- Department of Medical OncologyHead and Neck Cancer Department, West China HospitalChengduSichuan ProvincePeople's Republic of China
| | - Xiaorong Zhong
- Department of Medical OncologyHead and Neck Cancer Department, West China HospitalChengduSichuan ProvincePeople's Republic of China
| |
Collapse
|
48
|
Goyal RK, Zhang J, Davis KL, Sluga-O'Callaghan M, Kaufman PA. Real-world treatment patterns and clinical outcomes in patients treated with eribulin after prior phosphoinositide 3-Kinase inhibitor treatment for metastatic breast cancer. Breast Cancer Res Treat 2024; 205:201-210. [PMID: 38310616 PMCID: PMC11062963 DOI: 10.1007/s10549-023-07080-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/09/2023] [Indexed: 02/06/2024]
Abstract
PURPOSE In 2010, the US Food and Drug Administration approved eribulin for the treatment of metastatic breast cancer (MBC). Since then, the treatment landscape has evolved with many new therapy classes, a more recent one being the small molecule inhibitors of phosphoinositide 3 kinase (PI3K). We sought to characterize the treatment patterns and clinical outcomes of patients with MBC who received eribulin following prior treatment with a PI3K inhibitor. METHODS A retrospective cohort study based on medical record review included MBC patients who initiated eribulin between March 2019 and September 2020 following prior treatment with a PI3K inhibitor was conducted. Patient demographics, treatment characteristics, and clinical outcomes were analyzed descriptively. Real-world progression-free survival (rwPFS) and overall survival (OS) were estimated from the initiation of eribulin therapy using Kaplan-Meier analyses. RESULTS 82 eligible patients were included. Patients' median age at eribulin initiation was 62 years; 86.5% had hormone receptor-positive, human epidermal growth factor receptor 2-negative tumors. Eribulin was most often administered in the second or third line (82.9%) in the metastatic setting. Best overall response on eribulin was reported as complete or partial response in 72% of the patients. The median rwPFS was 18.9 months (95% confidence interval [CI], 12.4-not estimable); median OS was not reached. The estimated rwPFS and OS rates at 12 months were 63.3% (95% CI, 50.5-73.7) and 82.6% (95% CI, 72.4-89.3), respectively. CONCLUSION Our real-world study suggests that eribulin may be a potential treatment option for MBC patients who fail a prior PI3K inhibitor.
Collapse
Affiliation(s)
- Ravi K Goyal
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | | - Keith L Davis
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | | - Peter A Kaufman
- Larner College of Medicine, Division of Hematology/Oncology, University of Vermont Cancer Center, 111 Colchester Avenue, EP2, Burlington, VT, 05401, USA.
| |
Collapse
|
49
|
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
|
50
|
Bryce AH, Crawford ED, Agarwal N, Hussain MH, Beltran H, Cooperberg MR, Petrylak DP, Shore N, Spratt DE, Tagawa ST, Antonarakis ES, Aparicio AM, Armstrong AJ, Boike TP, Calais J, Carducci MA, Chapin BF, Cookson MS, Davis JW, Dorff T, Eggener SE, Feng FY, Gleave M, Higano C, Iagaru A, Morgans AK, Morris M, Murray KS, Poage W, Rettig MB, Sartor O, Scher HI, Sieber P, Small E, Srinivas S, Yu EY, Zhang T, Koo PJ. Expert Perspectives on Controversies in Metastatic Castration-Resistant Prostate Cancer Management: Narrative Review and Report of the First US Prostate Cancer Conference Part 2. JU OPEN PLUS 2024; 2:e00032. [PMID: 38774467 PMCID: PMC11107999 DOI: 10.1097/ju9.0000000000000138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/22/2024] [Indexed: 05/24/2024]
Abstract
Background Management strategies for metastatic castration-resistant prostate cancer (mCRPC) have rapidly shifted in recent years. As novel imaging and therapeutic approaches have made their way to the clinic, providers are encountering increasingly challenging clinical scenarios, with limited guidance from the current literature. Materials and Methods The US Prostate Cancer Conference (USPCC) is a multidisciplinary meeting of prostate cancer experts intended to address the many challenges of prostate cancer management. At the first annual USPCC meeting, areas of controversy and consensus were identified during a 2-day meeting that included expert presentations, full-panel discussions, and postdiscussion responses to questions developed by the USPCC cochairs and session moderators. Results This narrative review covers the USPCC expert discussion and perspectives relevant to mCRPC, including neuroendocrine/aggressive-variant prostate cancer (NEPC/AVPC). Areas of broad agreement identified among USPCC experts include the benefits of poly (ADP-ribose) polymerase (PARP) inhibitors for patients with BRCA1/2 mutations, the use of radioligand therapy in patients with prostate-specific membrane antigen (PSMA)-positive mCRPC, and the need for clinical trials that address real-world clinical questions, including the performance of novel therapies when compared with modern standard-of-care treatment. Ongoing areas of controversy and uncertainty included the appropriateness of PARP inhibitors in patients with non-BRCA1/2 mutations, the optimal definition of PSMA positivity, and systemic therapies for patients with NEPC/AVPC after progression on platinum-based therapies. Conclusions The first annual USPCC meeting identified several areas of controversy in the management of mCRPC, highlighting the urgent need for clinical trials designed to facilitate treatment selection and sequencing in this heterogeneous disease state.
Collapse
Affiliation(s)
- Alan H. Bryce
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, Arizona
| | - E. David Crawford
- Department of Urology, University of California San Diego, La Jolla, California
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Maha H. Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, Illinois
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Matthew R. Cooperberg
- Department of Urology, University of California at San Francisco, San Francisco, California
| | | | - Neal Shore
- Carolina Urologic Research Center/Genesis Care, Myrtle Beach, South Carolina
| | | | - Scott T. Tagawa
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, New York
| | | | - Ana M. Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrew J. Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina
| | | | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California
| | | | - Brian F. Chapin
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael S. Cookson
- Department of Urology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma
| | - John W. Davis
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tanya Dorff
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Scott E. Eggener
- Departments of Surgery (Urology), University of Chicago Medical Center, Chicago, Illinois
| | - Felix Y. Feng
- Departments of Radiation Oncology, Urology, and Medicine, University of California San Francisco, San Francisco, California
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Celestia Higano
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Alicia K. Morgans
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Michael Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katie S. Murray
- Department of Urology, NYU Langone Health, New York, New York
| | - Wendy Poage
- Prostate Conditions Education Council, Centennial, Colorado
| | - Matthew B. Rettig
- Department of Medicine, Division of Hematology-Oncology, VA Greater Los Angeles, Los Angeles, California
- Departments of Medicine and Urology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Howard I. Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Paul Sieber
- Keystone Urology Specialists, Lancaster, Pennsylvania
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University Medical Center, Stanford, California
| | - Evan Y. Yu
- Department of Medicine, Division of Hematology & Oncology, University of Washington and Fred Hutchinson Cancer Center, Seattle, Washington
| | - Tian Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|