5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Phase I study of vorinostat with gefitinib in BIM deletion polymorphism/epidermal growth factor receptor mutation double‐positive lung cancer

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Patients with epidermal growth factor receptor ( EGFR)‐mutated non‐small cell lung cancer (NSCLC) harboring BIM deletion polymorphism ( BIM deletion) have poor responses to EGFR TKI. Mechanistically, the BIM deletion induces preferential splicing of the non‐functional exon 3‐containing isoform over the functional exon 4‐containing isoform, impairing TKI‐induced, BIM‐dependent apoptosis. Histone deacetylase inhibitor, vorinostat, resensitizes BIM deletion‐containing NSCLC cells to EGFR‐TKI. In the present study, we determined the safety of vorinostat‐gefitinib combination and evaluated pharmacodynamic biomarkers of vorinostat activity. Patients with EGFR‐mutated NSCLC with the BIM deletion, pretreated with EGFR‐TKI and chemotherapy, were recruited. Vorinostat (200, 300, 400 mg) was given daily on days 1‐7, and gefitinib 250 mg was given daily on days 1‐14. Vorinostat doses were escalated based on a conventional 3 + 3 design. Pharmacodynamic markers were measured using PBMC collected at baseline and 4 hours after vorinostat dose on day 2 in cycle 1. No dose‐limiting toxicities (DLT) were observed in 12 patients. We determined 400 mg vorinostat as the recommended phase II dose (RP2D). Median progression‐free survival was 5.2 months (95% CI: 1.4‐15.7). Disease control rate at 6 weeks was 83.3% (10/12). Vorinostat preferentially induced BIM mRNA‐containing exon 4 over mRNA‐containing exon 3, acetylated histone H3 protein, and proapoptotic BIM EL protein in 11/11, 10/11, and 5/11 patients, respectively. These data indicate that RP2D was 400 mg vorinostat combined with gefitinib in BIM deletion/ EGFR mutation double‐positive NSCLC. BIM mRNA exon 3/exon 4 ratio in PBMC may be a useful pharmacodynamic marker for treatment.

          Abstract

          Vorinostat, in combination with gefitinib, induced acetylated histone H3 protein expression, as well as a decrease in the BIM mRNA exon 3/exon 4 ratio in PBMC from BIM deletion polymorphism/EGFR mutation double‐positive NSCLC patients. These results provide proof of concept that the combined therapy can mitigate the functional effects of BIM deletion polymorphism.

          Related collections

          Most cited references27

          • Record: found
          • Abstract: found
          • Article: not found

          Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation.

          Patients with epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma develop acquired resistance to EGFR tyrosine kinase inhibitors (TKI) after a median of 10 to 16 months. In half of these cases, a second EGFR mutation, T790M, underlies acquired resistance. We undertook this study to examine the clinical course of patients harboring the T790M mutation following progression on TKI. EGFR-mutant lung cancer patients with acquired resistance to EGFR TKIs were identified as part of a prospective rebiopsy protocol in which postprogression tumor specimens were collected for molecular analysis. Postprogression survival and characteristics of disease progression were compared in patients with and without T790M. We identified T790M in the initial rebiopsy specimens from 58 of 93 patients (62%, 95% CI: 52-72). T790M was more common in biopsies of lung/pleura tissue and lymph nodes than in more distant sites (P = 0.014). Median postprogression survival was 16 months (interquartile range = 9-29 months); patients with T790M had a significantly longer postprogression survival (P = 0.036). Patients without T790M more often progressed in a previously uninvolved organ system (P = 0.014) and exhibited a poorer performance status at time of progression (P = 0.007). Among patients with acquired resistance to EGFR TKIs, the presence of T790M defines a clinical subset with a relatively favorable prognosis and more indolent progression. Knowledge of T790M status is therefore important both for the clinical care of these patients and for the optimal design and interpretation of clinical trials in this setting. ©2010 AACR.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A common BIM deletion polymorphism mediates intrinsic resistance and inferior responses to tyrosine kinase inhibitors in cancer.

            Tyrosine kinase inhibitors (TKIs) elicit high response rates among individuals with kinase-driven malignancies, including chronic myeloid leukemia (CML) and epidermal growth factor receptor-mutated non-small-cell lung cancer (EGFR NSCLC). However, the extent and duration of these responses are heterogeneous, suggesting the existence of genetic modifiers affecting an individual's response to TKIs. Using paired-end DNA sequencing, we discovered a common intronic deletion polymorphism in the gene encoding BCL2-like 11 (BIM). BIM is a pro-apoptotic member of the B-cell CLL/lymphoma 2 (BCL2) family of proteins, and its upregulation is required for TKIs to induce apoptosis in kinase-driven cancers. The polymorphism switched BIM splicing from exon 4 to exon 3, which resulted in expression of BIM isoforms lacking the pro-apoptotic BCL2-homology domain 3 (BH3). The polymorphism was sufficient to confer intrinsic TKI resistance in CML and EGFR NSCLC cell lines, but this resistance could be overcome with BH3-mimetic drugs. Notably, individuals with CML and EGFR NSCLC harboring the polymorphism experienced significantly inferior responses to TKIs than did individuals without the polymorphism (P = 0.02 for CML and P = 0.027 for EGFR NSCLC). Our results offer an explanation for the heterogeneity of TKI responses across individuals and suggest the possibility of personalizing therapy with BH3 mimetics to overcome BIM-polymorphism-associated TKI resistance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A class of hybrid polar inducers of transformed cell differentiation inhibits histone deacetylases.

              Hybrid polar compounds (HPCs) have been synthesized that induce terminal differentiation and/or apoptosis in various transformed cells. We have previously reported on the development of the second-generation HPCs suberoylanilide hydroxamic acid (SAHA) and m-carboxycinnamic acid bishydroxamide (CBHA) that are 2,000-fold more potent inducers on a molar basis than the prototype HPC hexamethylene bisacetamide (HMBA). Herein we report that CBHA and SAHA inhibit histone deacetylase 1 (HDAC1) and histone deacetylase 3 (HDAC3) activity in vitro. Treatment of cells in culture with SAHA results in a marked hyperacetylation of histone H4, but culture with HMBA does not. Murine erythroleukemia cells developed for resistance to SAHA are cross-resistant to trichostatin A, a known deacetylase inhibitor and differentiation inducer, but are not cross-resistant to HMBA. These studies show that the second-generation HPCs, unlike HMBA, are potent inhibitors of HDAC activity. In this sense, HMBA and the second-generation HPCs appear to induce differentiation by different pathways.
                Bookmark

                Author and article information

                Contributors
                syano@staff.kanazawa-u.ac.jp
                Journal
                Cancer Sci
                Cancer Sci
                10.1111/(ISSN)1349-7006
                CAS
                Cancer Science
                John Wiley and Sons Inc. (Hoboken )
                1347-9032
                1349-7006
                06 January 2020
                February 2020
                : 111
                : 2 ( doiID: 10.1111/cas.v111.2 )
                : 561-570
                Affiliations
                [ 1 ] Division of Medical Oncology Cancer Research Institute Kanazawa University Kanazawa Japan
                [ 2 ] Nano Life Science Institute Kanazawa University Kanazawa Japan
                [ 3 ] Department of Respiratory Medicine Nagoya University Graduate School of Medicine Nagoya Japan
                [ 4 ] Department of Advanced Medicine Nagoya University Hospital Nagoya Japan
                [ 5 ] Division of Integrated Oncology Institute of Biomedical Research and Innovation Kobe Japan
                [ 6 ] Department of Medical Oncology Kobe Minimally Invasive Cancer Center Kobe Japan
                [ 7 ] Division of Thoracic Oncology Shizuoka Cancer Center Shizuoka Japan
                [ 8 ] Innovative Clinical Research Center (iCREK) Kanazawa University Hospital Kanazawa Japan
                [ 9 ] Department of Data Science Center for Integrated Medical Research Hiroshima University Hospital Hiroshima Japan
                [ 10 ] Clinical Research Center Chiba University Hospital Chiba Japan
                [ 11 ] Department of Medical Oncology Takarazuka City Hospital Takarazuka Japan
                [ 12 ] National Hospital Organization Nagoya Medical Center Nagoya Japan
                [ 13 ] Cancer and Stem Cell Biology Signature Research Program Duke‐NUS Medical School Singapore
                [ 14 ] Department of Haematology Singapore General Hospital Singapore
                [ 15 ] Department of Medical Oncology National Cancer Centre Singapore Singapore
                [ 16 ] Department of Medicine Duke University Medical Center Durham North Carolina USA
                Author notes
                [*] [* ] Correspondence

                Seiji Yano, Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan.

                Email: syano@ 123456staff.kanazawa-u.ac.jp

                Author information
                https://orcid.org/0000-0003-2416-546X
                https://orcid.org/0000-0001-8544-6244
                https://orcid.org/0000-0002-6151-2988
                Article
                CAS14260
                10.1111/cas.14260
                7004511
                31782583
                7dcdd46b-7412-4fb9-b3e3-6a12ba4e64f9
                © 2019 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 29 October 2019
                : 13 November 2019
                : 25 November 2019
                Page count
                Figures: 3, Tables: 5, Pages: 10, Words: 7241
                Funding
                Funded by: Japan Agency for Medical Research and Development , open-funder-registry 10.13039/100009619;
                Award ID: 15Aak0101016h0003
                Award ID: 15Ack0106113h0002
                Award ID: 16ck0106207h0001
                Funded by: Kanazawa University Hospital
                Categories
                Original Article
                Original Articles
                Clinical Research
                Custom metadata
                2.0
                February 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.7.5 mode:remove_FC converted:06.02.2020

                Oncology & Radiotherapy
                bim deletion polymorphism,egfr‐tki,nsclc,resistance,vorinostat
                Oncology & Radiotherapy
                bim deletion polymorphism, egfr‐tki, nsclc, resistance, vorinostat

                Comments

                Comment on this article