1
|
Eroglu Z, Synold T, Badie B, Liu A, Chowdhury A, Kilpatrick J, Blanchard S, Portnow J. An intracerebral microdialysis study to determine the neuropharmacokinetics of eribulin in patients with metastatic or primary brain tumors. Cancer Chemother Pharmacol 2024; 94:807-813. [PMID: 39422741 PMCID: PMC11573798 DOI: 10.1007/s00280-024-04711-2] [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: 04/30/2024] [Accepted: 08/16/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Eribulin is an inhibitor of microtubule dynamics. It is not as highly protein bound as the taxanes and is less vulnerable to extrusion by P-glycoprotein in the blood-brain barrier (BBB). These features predict that eribulin could play an active role in managing brain tumors. Indeed, the small amount of published clinical data indicates eribulin may have some efficacy against breast cancer brain metastases. To better understand the potential of eribulin for treating brain tumors, we performed an intracerebral microdialysis study to determine the neuropharmacokinetics of eribulin in cancer patients undergoing tumor resection. METHODS After tumor removal, two microdialysis catheters were inserted into peritumoral brain tissue. Approximately 24 h after surgery, a single dose of eribulin 1.4 mg/m2 was administered intravenously. Dialysate samples were collected continuously for 72 h, with plasma samples collected in parallel. Eribulin concentrations were analyzed by tandem mass spectrometry. RESULTS Dialysate samples from 12 intracerebral microdialysis catheters placed in 7 study participants were included in the analysis. A statistically significant difference was observed between eribulin concentrations in brain tissue where BBB was disrupted versus intact, with a difference in mean maximum concentrations on log2 scale of 3.37 (std err = 0.59, p-value = 0.005). Nonetheless, overall brain to plasma ratios of eribulin only ranged from 0.13 to 1.99%. CONCLUSION Although we could detect higher concentrations of eribulin in brain tissue where BBB was disrupted, intracerebral eribulin levels were not sufficient to predict eribulin would have consistent clinically meaningful activity against tumors in the brain. CLINICALTRIALS GOV IDENTIFIER NCT02338037 (January 9, 2015).
Collapse
Affiliation(s)
- Zeynep Eroglu
- City of Hope Comprehensive Cancer Center, Department of Medical Oncology and Therapeutics Research, Duarte, CA, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Timothy Synold
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Behnam Badie
- Department of Surgery, Division of Neurosurgery, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - An Liu
- Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Arnab Chowdhury
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Julie Kilpatrick
- Departmant of Clinical Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Suzette Blanchard
- Department of Computational and Quantitative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Jana Portnow
- City of Hope Comprehensive Cancer Center, Department of Medical Oncology and Therapeutics Research, Duarte, CA, USA.
| |
Collapse
|
2
|
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
|
3
|
Hsu CY, Yanagi T, Maeda T, Nishihara H, Miyamoto K, Kitamura S, Tokuchi K, Ujiie H. Eribulin inhibits growth of cutaneous squamous cell carcinoma cell lines and a novel patient-derived xenograft. Sci Rep 2023; 13:8650. [PMID: 37244956 DOI: 10.1038/s41598-023-35811-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023] Open
Abstract
Advanced cutaneous squamous cell carcinoma (cSCC) is treated with chemotherapy and/or radiotherapy, but these typically fail to achieve satisfactory clinical outcomes. There have been no preclinical studies to evaluate the effectiveness of eribulin against cSCC. Here, we examine the effects of eribulin using cSCC cell lines and a novel cSCC patient-derived xenograft (PDX) model. In the cSCC cell lines (A431 and DJM-1 cells), eribulin was found to inhibit tumor cell proliferation in vitro as assessed by cell ATP levels. DNA content analysis by fluorescence-activated cell sorting (FACS) showed that eribulin induced G2/M cell cycle arrest and apoptosis. In xenograft models of cSCC cell lines, the administration of eribulin suppressed tumor growth in vivo. We also developed a cSCC patient-derived xenograft (PDX) which reproduces the histological and genetic characteristics of a primary tumor. Pathogenic mutations in TP53 and ARID2 were detected in the patient's metastatic tumor and in the PDX tumor. The cSCC-PDX responded well to the administration of eribulin and cisplatin. In conclusion, the present study shows the promising antineoplastic effects of eribulin in cSCC. Also, we established a novel cSCC-PDX model that preserves the patient's tumor. This PDX could assist researchers who are exploring innovative therapies for cSCC.
Collapse
Affiliation(s)
- Che-Yuan Hsu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Teruki Yanagi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan.
| | - Takuya Maeda
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Kodai Miyamoto
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Shinya Kitamura
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Keiko Tokuchi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, N15 W7, Kita-Ku, Sapporo, 060-8638, Japan
| |
Collapse
|
4
|
Mano Y. A separate assay of released and liposomal encapsulated eribulin in dog plasma by liquid chromatography with tandem mass spectrometry for its application to a pharmacokinetic study. J Sep Sci 2022; 45:1636-1645. [PMID: 35235250 DOI: 10.1002/jssc.202200048] [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: 01/18/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 11/08/2022]
Abstract
Eribulin has been used as a drug for the treatment of metastatic breast cancer and liposomal formulation of eribulin (E7389-LF) is under development to achieve a wider therapeutic index. It is important to separately determine released and encapsulated drugs in a systemic circulation for liposomal drugs. In this study, a separate assay method was developed for the determination of released and total (encapsulated+released) eribulin concentrations in dog plasma by liquid chromatography with tandem mass spectrometry. The released eribulin in dog plasma was separated by ultrafiltration of plasma samples. Obtained plasma ultrafiltrate and untreated plasma samples recognized as released and total eribulin, respectively, were subjected to protein precipitation for extraction of eribulin. Eribulin was quantifiable from 0.1 ng/mL. Accuracy and precision of eribulin in both matrices were within ±15% and 15%, respectively, indicating a robust assay. Released and total eribulin concentrations in plasma were determined after intravenous administration of E7389-LF to dogs, resulting in minimal released eribulin in plasma. In conclusion, a robust method for released and total eribulin levels in dog plasma was developed and was successfully applied to a pharmacokinetic study in dogs to characterize the pharmacokinetic profiles. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yuji Mano
- Global Drug Metabolism and Pharmacokinetics, Eisai Co., Ltd., Tokodai 5-1-3, Tsukuba-shi, Ibaraki 300-2635, Japan
| |
Collapse
|
5
|
Slimano F, Djerada Z, Guerin J, Bellouch MI, Brassart-Pasco S, Dukic S. Intratumoral distribution of YSNSG cyclopeptide in a mouse melanoma model using microdialysis. Eur J Pharm Sci 2019; 143:105201. [PMID: 31866565 DOI: 10.1016/j.ejps.2019.105201] [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: 08/27/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 10/25/2022]
Abstract
The YSNSG peptide is a synthetic cyclopeptide targeting αvβ3 integrin with antitumor activity. Previous study has determined main pharmacokinetic parameters in plasma and in tissue in healthy animals using microdialysis. First we aim to assess the impact of a 20 mg/kg dosage instead of 10 mg/kg in tumor growth inhibition. Secondly we aim to investigate the YSNSG peptide distribution in two different tumor regions in animals with melanoma. C57BL/6 mice were exposed at Days 8, 10 and 12 after melanoma cells implantation (B16F1) to different dosage of YSNSG peptide or control, respectively (n = 10 per group). Data analysis was performed at D16, 20 and 24 with a Nonlinear Mixed-Effects (NLME) approach. For pharmacokinetic study n = 8 mice (same disease condition) received YSNSG peptide by intravenous after insertion of two microdialysis probes in central peripheral region of tumor, respectively. Plasma and tissue samples were collected during 2 h. A non-compartmental analysis was performed to determine main pharmacokinetic parameters. There was a significant tumor growth inhibition in mice receiving 20 mg/kg vs Control (p < 0.02). Main plasma parameters were half-life elimination 25.8 ± 8.2 min, volume of distribution 11.9 ± 0.4 mL, clearance 19.8 ± 9.4 mL/h and area under the curve 1,173.6 µg.min/mL. Penetration rate of the YSNSG peptide from plasma to tumor tissue were 3.3 ± 2.1% and 3.4 ± 2.7% in central and peripheral, respectively. Contrary to subcutaneous distribution in healthy animals the distribution of the YSNSG peptide into tumoral tissue is low but seems non-heterogeneous between central and peripheral tumor region.
Collapse
Affiliation(s)
- Florian Slimano
- MEDyC Research Unit, UMR CNRS/URCA n°7369, SFR CAP-Santé, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France; Department of Pharmacy, CHU Reims, Avenue du General Koenig, and Faculty of Pharmacy, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France.
| | - Zoubir Djerada
- Department of Pharmacology and Toxicology, CHU Reims, Avenue du General Koenig, 51100 Reims, France; EA3801, SFR CAP-Santé, Faculty of Medicine, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France
| | - Juline Guerin
- MEDyC Research Unit, UMR CNRS/URCA n°7369, SFR CAP-Santé, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France
| | - Morad Id Bellouch
- MEDyC Research Unit, UMR CNRS/URCA n°7369, SFR CAP-Santé, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France
| | - Sylvie Brassart-Pasco
- MEDyC Research Unit, UMR CNRS/URCA n°7369, SFR CAP-Santé, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France
| | - Sylvain Dukic
- MEDyC Research Unit, UMR CNRS/URCA n°7369, SFR CAP-Santé, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France; Department of Pharmacy, CHU Reims, Avenue du General Koenig, and Faculty of Pharmacy, Reims University, 51, rue Cognacq-Jay, 51100 Reims, France
| |
Collapse
|
6
|
Takahashi M, Miki S, Fujimoto K, Fukuoka K, Matsushita Y, Maida Y, Yasukawa M, Hayashi M, Shinkyo R, Kikuchi K, Mukasa A, Nishikawa R, Tamura K, Narita Y, Hamada A, Masutomi K, Ichimura K. Eribulin penetrates brain tumor tissue and prolongs survival of mice harboring intracerebral glioblastoma xenografts. Cancer Sci 2019; 110:2247-2257. [PMID: 31099446 PMCID: PMC6609810 DOI: 10.1111/cas.14067] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/24/2019] [Accepted: 05/13/2019] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is one of the most devastating human malignancies for which a novel efficient treatment is urgently required. This pre-clinical study shows that eribulin, a specific inhibitor of telomerase reverse transcriptase (TERT)-RNA-dependent RNA polymerase, is an effective anticancer agent against glioblastoma. Eribulin inhibited the growth of 4 TERT promoter mutation-harboring glioblastoma cell lines in vitro at subnanomolar concentrations. In addition, it suppressed the growth of glioblastoma cells transplanted subcutaneously or intracerebrally into mice, and significantly prolonged the survival of mice harboring brain tumors at a clinically equivalent dose. A pharmacokinetics study showed that eribulin quickly penetrated brain tumors and remained at a high concentration even when it was washed away from plasma, kidney or liver 24 hours after intravenous injection. Moreover, a matrix-assisted laser desorption/ionization mass spectrometry imaging analysis revealed that intraperitoneally injected eribulin penetrated the brain tumor and was distributed evenly within the tumor mass at 1 hour after the injection whereas only very low levels of eribulin were detected in surrounding normal brain. Eribulin is an FDA-approved drug for refractory breast cancer and can be safely repositioned for treatment of glioblastoma patients. Thus, our results suggest that eribulin may serve as a novel therapeutic option for glioblastoma. Based on these data, an investigator-initiated registration-directed clinical trial to evaluate the safety and efficacy of eribulin in patients with recurrent GBM (UMIN000030359) has been initiated.
Collapse
Affiliation(s)
- Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Shunichiro Miki
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenji Fujimoto
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Kohei Fukuoka
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuko Matsushita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshiko Maida
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Mami Yasukawa
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Mitsuhiro Hayashi
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Raku Shinkyo
- Tsukuba Research Laboratory, Eisai, Tsukuba, Japan
| | | | - Akitake Mukasa
- Department of Neurosurgery, The University of Tokyo, Tokyo, Japan
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenkichi Masutomi
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
7
|
Watanabe K, Yui Y, Sasagawa S, Suzuki K, Kanamori M, Yasuda T, Kimura T. Low-dose eribulin reduces lung metastasis of osteosarcoma in vitro and in vivo. Oncotarget 2019; 10:161-174. [PMID: 30719211 PMCID: PMC6349434 DOI: 10.18632/oncotarget.26536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/20/2018] [Indexed: 11/25/2022] Open
Abstract
Lung metastasis markedly reduces the prognosis of osteosarcoma. Moreover, there is no effective treatment for lung metastasis, and a new treatment strategy for the treatment of osteosarcoma lung metastasis is required. Therefore, in this study, we investigated the suppressive effect of the microtubule inhibitor eribulin mesylate (eribulin) on lung metastasis of osteosarcoma. At concentrations >proliferation IC50, eribulin induced cell cycle arrest and apoptosis in a metastatic osteosarcoma cell line, LM8. However, at concentrations <proliferation IC50, (low dose), eribulin changed cell morphology and decreased LM8 migration. Low eribulin concentrations also reduced directionality during migration, peripheral localization of adenomatous polyposis coli protein, and turnover of focal adhesions. In a three-dimensional collagen culture system, low eribulin concentrations inhibited tumor cell proliferation and colony formation. Higher doses of eribulin administered on a standard schedule inhibited lung metastasis and primary tumor growth in a murine osteosarcoma metastasis model. Frequent low-dose eribulin administration (0.3 mg/kg every 4 days × 4) effectively inhibited lung metastasis but had little effect on primary tumor growth. Overall, our results indicate that eribulin could reduce osteosarcoma lung metastasis.
Collapse
Affiliation(s)
- Kenta Watanabe
- Department of Orthopedic Surgery, University of Toyama, Toyama, Japan.,Research Institute, Nozaki Tokushukai Hospital, Osaka, Japan
| | - Yoshihiro Yui
- Research Institute, Nozaki Tokushukai Hospital, Osaka, Japan
| | - Satoru Sasagawa
- Research Institute, Nozaki Tokushukai Hospital, Osaka, Japan
| | - Kayo Suzuki
- Department of Orthopedic Surgery, University of Toyama, Toyama, Japan
| | | | - Taketoshi Yasuda
- Department of Orthopedic Surgery, University of Toyama, Toyama, Japan
| | - Tomoatsu Kimura
- Department of Orthopedic Surgery, University of Toyama, Toyama, Japan
| |
Collapse
|
8
|
Schafer ES, Rau RE, Berg S, Liu X, Minard CG, D’Adamo D, Scott R, Reyderman L, Martinez G, Devarajan S, Reid JM, Fox E, Weigel BJ, Blaney SM. A phase 1 study of eribulin mesylate (E7389), a novel microtubule-targeting chemotherapeutic agent, in children with refractory or recurrent solid tumors: A Children's Oncology Group Phase 1 Consortium study (ADVL1314). Pediatr Blood Cancer 2018; 65:e27066. [PMID: 29719113 PMCID: PMC6019176 DOI: 10.1002/pbc.27066] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/12/2018] [Accepted: 03/04/2018] [Indexed: 01/21/2023]
Abstract
BACKGROUND Eribulin mesylate is a novel anticancer agent that inhibits microtubule growth, without effects on shortening, and promotes nonproductive tubulin aggregate formation. We performed a phase 1 trial to determine the dose-limiting toxicities (DLTs), maximum tolerated or recommended phase 2 dose (MTD/RP2D), and pharmacokinetics (PK) of eribulin in children with refractory or recurrent solid (excluding central nervous system) tumors. METHODS Eribulin was administered intravenously on days 1 and 8 in 21-day cycles. Three dose levels (1.1, 1.4, and 1.8 mg/m2 /dose) were evaluated using the rolling six design with additional patients enrolled into a PK expansion cohort at the MTD. PK samples were obtained following the day 1, cycle 1 dose. RESULTS Twenty-three patients, ages 3-17 (median 14) years were enrolled; 20 were evaluable for toxicity. DLTs occurred in 0/6 and 1/6 subjects at the 1.1 and 1.4 mg/m2 /dose, respectively. One subject at the 1.4 mg/m2 /dose had grade 4 neutropenia and grade 3 fatigue. At the 1.8 mg/m2 /dose, 2/5 subjects experienced dose-limiting (grade 4) neutropenia. Grade 3/4 non-DLTs included lymphopenia and hypokalemia, while low-grade toxicities included anorexia and nausea. No episodes of grade > 2 corrected QT interval prolongation or peripheral neuropathy were reported. Eribulin pharmacokinetic parameters were highly variable; the median elimination half-life was 39.6 (range 24.2-96.4) hr. A partial response was observed in one patient (Ewing sarcoma). CONCLUSIONS Eribulin was well tolerated in children with refractory or recurrent solid tumors with neutropenia identified as the primary DLT. The RP2D of eribulin is 1.4 mg/m2 /dose on days 1 and 8 of a 21-day cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Elizabeth Fox
- Children’s Hospital of Philadelphia, Philadelphia, PA
| | | | | |
Collapse
|
9
|
Miki S, Imamichi S, Fujimori H, Tomiyama A, Fujimoto K, Satomi K, Matsushita Y, Matsuzaki S, Takahashi M, Ishikawa E, Yamamoto T, Matsumura A, Mukasa A, Nishikawa R, Masutomi K, Narita Y, Masutani M, Ichimura K. Concomitant administration of radiation with eribulin improves the survival of mice harboring intracerebral glioblastoma. Cancer Sci 2018; 109:2275-2285. [PMID: 29758120 PMCID: PMC6029838 DOI: 10.1111/cas.13637] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 04/15/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma is the most common and devastating type of malignant brain tumor. We recently found that eribulin suppresses glioma growth in vitro and in vivo and that eribulin is efficiently transferred into mouse brain tumors at a high concentration. Eribulin is a non‐taxane microtubule inhibitor approved for breast cancer and liposarcoma. Cells arrested in M‐phase by chemotherapeutic agents such as microtubule inhibitors are highly sensitive to radiation‐induced DNA damage. Several recent case reports have demonstrated the clinical benefits of eribulin combined with radiation therapy for metastatic brain tumors. In this study, we investigated the efficacy of a combined eribulin and radiation treatment on human glioblastoma cells. The glioblastoma cell lines U87MG, U251MG and U118MG, and SJ28 cells, a patient‐derived sphere culture cell line, were used to determine the radiosensitizing effect of eribulin using western blotting, flow cytometry and clonogenic assay. Subcutaneous and intracerebral glioma xenografts were generated in mice to assess the efficacy of the combined treatment. The combination of eribulin and radiation enhanced DNA damage in vitro. The clonogenic assay of U87MG demonstrated the radiosensitizing effect of eribulin. The concomitant eribulin and radiation treatment significantly prolonged the survival of mice harboring intracerebral glioma xenografts compared with eribulin or radiation alone (P < .0001). In addition, maintenance administration of eribulin after the concomitant treatment further controlled brain tumor growth. Aberrant microvasculature was decreased in these tumors. Concomitant treatment with eribulin and radiation followed by maintenance administration of eribulin may serve as a novel therapeutic strategy for glioblastomas.
Collapse
Affiliation(s)
- Shunichiro Miki
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Shoji Imamichi
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroaki Fujimori
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Arata Tomiyama
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery, National Defense Medical College, Tokorozawa, Japan
| | - Kenji Fujimoto
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Neurosurgery Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kaishi Satomi
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Yuko Matsushita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Sanae Matsuzaki
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan.,Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tetsuya Yamamoto
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.,Department of Neurosurgery, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Akira Matsumura
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Akitake Mukasa
- Department of Neurosurgery Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Neurosurgery, the University of Tokyo, Tokyo, Japan
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kenkichi Masutomi
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Mitsuko Masutani
- Division of Chemotherapy and Clinical Research, National Cancer Center Research Institute, Tokyo, Japan.,Department of Frontier Life Sciences, Nagasaki University Graduate School of Medicine, Nagasaki, Japan
| | - Koichi Ichimura
- Division of Brain Tumor Translational Research, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|