17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Phase 1 Study of Molibresib (GSK525762), a Bromodomain and Extra-Terminal Domain Protein Inhibitor, in NUT Carcinoma and Other Solid Tumors

      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

          Background

          Bromodomain and extra-terminal domain proteins are promising epigenetic anticancer drug targets. This first-in-human study evaluated the safety, recommended phase II dose, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the bromodomain and extra-terminal domain inhibitor molibresib (GSK525762) in patients with nuclear protein in testis (NUT) carcinoma (NC) and other solid tumors.

          Methods

          This was a phase I and II, open-label, dose-escalation study. Molibresib was administered orally once daily. Single-patient dose escalation (from 2 mg/d) was conducted until the first instance of grade 2 or higher drug-related toxicity, followed by a 3 + 3 design. Pharmacokinetic parameters were obtained during weeks 1 and 3. Circulating monocyte chemoattractant protein-1 levels were measured as a pharmacodynamic biomarker.

          Results

          Sixty-five patients received molibresib. During dose escalation, 11% experienced dose-limiting toxicities, including six instances of grade 4 thrombocytopenia, all with molibresib 60–100 mg. The most frequent treatment-related adverse events of any grade were thrombocytopenia (51%) and gastrointestinal events, including nausea, vomiting, diarrhea, decreased appetite, and dysgeusia (22%–42%), anemia (22%), and fatigue (20%). Molibresib demonstrated an acceptable safety profile up to 100 mg; 80 mg once daily was selected as the recommended phase II dose. Following single and repeat dosing, molibresib showed rapid absorption and elimination (maximum plasma concentration: 2 hours; t 1/2: 3–7 hours). Dose-dependent reductions in circulating monocyte chemoattractant protein-1 levels were observed. Among 19 patients with NC, four achieved either confirmed or unconfirmed partial response, eight had stable disease as best response, and four were progression-free for more than 6 months.

          Conclusions

          Once-daily molibresib was tolerated at doses demonstrating target engagement. Preliminary data indicate proof-of-concept in NC.

          Related collections

          Most cited references23

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

          Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study.

          The first-in-class small molecule inhibitor OTX015 (MK-8628) specifically binds to bromodomain motifs BRD2, BRD3, and BRD4 of bromodomain and extraterminal (BET) proteins, inhibiting them from binding to acetylated histones, which occurs preferentially at super-enhancer regions that control oncogene expression. OTX015 is active in haematological preclinical entities including leukaemia, lymphoma, and myeloma. We aimed to establish the recommended dose of OTX015 in patients with haematological malignancies. We report the results from a cohort of patients with lymphoma or multiple myeloma (non-leukaemia cohort).
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            BET inhibitors: a novel epigenetic approach

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

              BRD-NUT oncoproteins: a family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells.

              An unusual group of carcinomas, here termed nuclear protein in testis (NUT) midline carcinomas (NMC), are characterized by translocations that involve NUT, a novel gene on chromosome 15. In about 2/3rds of cases, NUT is fused to BRD4 on chromosome 19. Using a candidate gene approach, we identified two NMCs harboring novel rearrangements that result in the fusion of NUT to BRD3 on chromosome 9. The BRD3-NUT fusion gene encodes a protein composed of two tandem chromatin-binding bromodomains, an extra-terminal domain, a bipartite nuclear localization sequence, and almost the entirety of NUT that is highly homologous to BRD4-NUT. The function of NUT is unknown, but here we show that NUT contains nuclear localization and export sequences that promote nuclear-cytoplasmic shuttling via a leptomycin-sensitive pathway. In contrast, BRD3-NUT and BRD4-NUT are strictly nuclear, implying that the BRD moiety retains NUT in the nucleus via interactions with chromatin. Consistent with this idea, FRAP studies show that BRD4, BRD4-NUT and BRD3-NUT have significantly slower rates of lateral nuclear diffusion than that of NUT. To investigate the functional role of BRD-NUT fusion proteins in NMCs, we investigated the effects of siRNA-induced BRD3-NUT and BRD4-NUT withdrawal. Silencing of these proteins in NMC cell lines resulted in squamous differentiation and cell cycle arrest. Together, these data suggest that BRD-NUT fusion proteins contribute to carcinogenesis by associating with chromatin and interfering with epithelial differentiation.
                Bookmark

                Author and article information

                Journal
                JNCI Cancer Spectr
                JNCI Cancer Spectr
                jncics
                JNCI Cancer Spectrum
                Oxford University Press
                2515-5091
                April 2020
                06 November 2019
                06 November 2019
                : 4
                : 2
                : pkz093
                Affiliations
                [1 ] University of Texas MD Anderson Cancer Center , Houston, TX
                [2 ] Johns Hopkins University School of Medicine , Baltimore, MD
                [3 ] Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School , Boston, MA
                [4 ] Medical Oncology, Institute Bergonié , Bordeaux, France
                [5 ] Medical Oncology Department, Vall d’Hebron University Hospital , Barcelona, Spain
                [6 ] Medical Oncology, Léon Bérard Cancer Center , Lyon, France
                [7 ] Medical Oncology, START Madrid-FJD, Fundación Jiménez Díaz Hospital , Madrid, Spain
                [8 ] The Institute of Cancer Research and Royal Marsden Hospital , London, UK
                [9 ] GSK, Research Triangle Park , NC
                [10 ] GSK, Collegeville , PA
                [11 ] UCB Celltech , Slough, UK
                [12 ] Division of Medical Oncology, Ottawa Hospital Cancer Centre , Ottawa, ON, Canada
                [13 ] Rubius Therapeutics, Cambridge , MA
                [14 ] Abramson Cancer Center at University of Pennsylvania , Philadelphia, PA
                [15 ] Department of Medical Oncology, Dana-Farber Cancer Institute , Boston, MA
                [16 ] Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School , Boston, MA
                Author notes
                [*]

                Peter J. O’Dwyer and Geoffrey I. Shapiro contributed equally to this work and are co-senior authors.

                Correspondence to: Geoffrey I. Shapiro, MD, PhD, Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Mayer 446, Boston, MA 02215 (e-mail: geoffrey_shapiro@ 123456dfci.harvard.edu ).
                Author information
                http://orcid.org/0000-0002-1467-5557
                http://orcid.org/0000-0001-5703-8315
                http://orcid.org/0000-0002-7929-0877
                http://orcid.org/0000-0002-3331-4095
                Article
                pkz093
                10.1093/jncics/pkz093
                7165800
                32328561
                659e0858-e5cc-4e0e-9c4c-f8765f3e16e2
                © The Author(s) 2019. Published by Oxford University Press.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

                History
                : 2 May 2019
                : 21 August 2019
                : 31 October 2019
                Page count
                Pages: 9
                Funding
                Funded by: GlaxoSmithKline (GSK);
                Funded by: National Institutes of Health (NIH) Cancer Center;
                Award ID: P30 CA016672
                Award ID: P30 CA006516
                Funded by: NIH, DOI 10.13039/100000002;
                Award ID: R01 CA124633
                Categories
                Article

                Comments

                Comment on this article