1
|
Van Tine BA, Ingham MA, Attia S, Meyer CF, Baird JD, Brooks-Asplund E, D'Silva D, Kong R, Mwatha A, O'Keefe K, Weetall M, Spiegel R, Schwartz GK. Phase Ib Study of Unesbulin (PTC596) Plus Dacarbazine for the Treatment of Locally Recurrent, Unresectable or Metastatic, Relapsed or Refractory Leiomyosarcoma. J Clin Oncol 2024; 42:2404-2414. [PMID: 38684039 PMCID: PMC11227303 DOI: 10.1200/jco.23.01684] [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: 08/05/2023] [Revised: 12/13/2023] [Accepted: 02/28/2024] [Indexed: 05/02/2024] Open
Abstract
PURPOSE This multicenter, single-arm, open-label, phase Ib study was designed to determine the recommended phase II dose (RP2D) and to evaluate the safety and preliminary efficacy of unesbulin plus dacarbazine (DTIC) in patients with advanced leiomyosarcoma (LMS). PATIENTS AND METHODS Adult subjects with locally advanced, unresectable or metastatic, relapsed or refractory LMS were treated with escalating doses of unesbulin orally twice per week in combination with DTIC 1,000 mg/m2 intravenously (IV) once every 21 days. The time-to-event continual reassessment method was used to determine the RP2D on the basis of dose-limiting toxicities (DLTs) assessed during the first two 21-day treatment cycles. All explored doses of unesbulin (200 mg up to 400 mg) were in combination with DTIC. An expansion cohort was enrolled to evaluate the safety and efficacy of unesbulin at the RP2D. RESULTS Unesbulin 300 mg administered orally twice per week in combination with DTIC 1,000 mg/m2 IV once every 21 days was identified as the RP2D. On the basis of data from 27 subjects who were deemed DLT-evaluable, toxicity was higher in the unesbulin 400 mg group, with three of four subjects (75%) experiencing DLTs versus one of four subjects (25%) in the 200 mg group and three of 19 subjects (15.8%) in the 300 mg group. The most commonly reported DLTs and treatment-related grade 3 and 4 adverse events were thrombocytopenia and neutropenia. At the RP2D, seven subjects who were efficacy evaluable achieved partial response for an objective response rate of 24.1%. CONCLUSION Unesbulin 300 mg twice per week plus DTIC 1,000 mg/m2 once every 21 days was identified as the RP2D, demonstrating a favorable benefit-risk profile in a heavily pretreated population of adults with advanced LMS.
Collapse
|
2
|
Weisbrod LJ, Thiraviyam A, Vengoji R, Shonka N, Jain M, Ho W, Batra SK, Salehi A. Diffuse intrinsic pontine glioma (DIPG): A review of current and emerging treatment strategies. Cancer Lett 2024; 590:216876. [PMID: 38609002 PMCID: PMC11231989 DOI: 10.1016/j.canlet.2024.216876] [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: 01/22/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a childhood malignancy of the brainstem with a dismal prognosis. Despite recent advances in its understanding at the molecular level, the prognosis of DIPG has remained unchanged. This article aims to review the current understanding of the genetic pathophysiology of DIPG and to highlight promising therapeutic targets. Various DIPG treatment strategies have been investigated in pre-clinical studies, several of which have shown promise and have been subsequently translated into ongoing clinical trials. Ultimately, a multifaceted therapeutic approach that targets cell-intrinsic alterations, the micro-environment, and augments the immune system will likely be necessary to eradicate DIPG.
Collapse
Affiliation(s)
- Luke J Weisbrod
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Anand Thiraviyam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Winson Ho
- Department of Neurosurgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA
| | - Afshin Salehi
- Department of Neurosurgery, University of Nebraska Medical Center, Omaha, NE, 68198-5870, USA; Division of Pediatric Neurosurgery, Children's Nebraska, Omaha, NE, 68114, USA.
| |
Collapse
|
3
|
Maroni G, Krishnan I, Alfieri R, Maymi VA, Pandell N, Csizmadia E, Zhang J, Weetall M, Branstrom A, Braccini G, Cabrera San Millán E, Storti B, Bizzarri R, Kocher O, Daniela Sanchez Bassères DS, Welner RS, Magli MC, Merelli I, Clohessy JG, Ali A, Tenen DG, Levantini E. Tumor Microenvironment Landscapes Supporting EGFR-mutant NSCLC Are Modulated at the Single-cell Interaction Level by Unesbulin Treatment. CANCER RESEARCH COMMUNICATIONS 2024; 4:919-937. [PMID: 38546390 PMCID: PMC10964845 DOI: 10.1158/2767-9764.crc-23-0161] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 01/26/2024] [Accepted: 03/12/2024] [Indexed: 11/03/2024]
Abstract
Lung cancer is the leading cause of cancer deaths. Lethal pulmonary adenocarcinomas (ADC) present with frequent mutations in the EGFR. Genetically engineered murine models of lung cancer expedited comprehension of the molecular mechanisms driving tumorigenesis and drug response. Here, we systematically analyzed the evolution of tumor heterogeneity in the context of dynamic interactions occurring with the intermingled tumor microenvironment (TME) by high-resolution transcriptomics. Our effort identified vulnerable tumor-specific epithelial cells, as well as their cross-talk with niche components (endothelial cells, fibroblasts, and tumor-infiltrating immune cells), whose symbiotic interface shapes tumor aggressiveness and is almost completely abolished by treatment with Unesbulin, a tubulin binding agent that reduces B cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) activity. Simultaneous magnetic resonance imaging (MRI) analysis demonstrated decreased tumor growth, setting the stage for future investigations into the potential of novel therapeutic strategies for EGFR-mutant ADCs. SIGNIFICANCE Targeting the TME is an attractive strategy for treatment of solid tumors. Here we revealed how EGFR-mutant landscapes are affected at the single-cell resolution level during Unesbulin treatment. This novel drug, by targeting cancer cells and their interactions with crucial TME components, could be envisioned for future therapeutic advancements.
Collapse
Affiliation(s)
- Giorgia Maroni
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Harvard Medical School, Boston, Massachusetts
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | | | - Roberta Alfieri
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - Valerie A. Maymi
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Preclinical Murine Pharmacogenetics Core, Beth Israel Deaconess Cancer Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Nicole Pandell
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Preclinical Murine Pharmacogenetics Core, Beth Israel Deaconess Cancer Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Eva Csizmadia
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Junyan Zhang
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | | | - Giulia Braccini
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | | | - Barbara Storti
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa, Italy
| | - Ranieri Bizzarri
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa, Italy
- Department of Surgical, Medical and Molecular Pathology, and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Olivier Kocher
- Harvard Medical School, Boston, Massachusetts
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Robert S. Welner
- Department of Medicine, Hemathology/Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Maria Cristina Magli
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - Ivan Merelli
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - John G. Clohessy
- Harvard Medical School, Boston, Massachusetts
- Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Preclinical Murine Pharmacogenetics Core, Beth Israel Deaconess Cancer Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Azhar Ali
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniel G. Tenen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
- Harvard Medical School, Boston, Massachusetts
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Elena Levantini
- Harvard Medical School, Boston, Massachusetts
- Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
- Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
4
|
Sun MA, Yang R, Liu H, Wang W, Song X, Hu B, Reynolds N, Roso K, Chen LH, Greer PK, Keir ST, McLendon RE, Cheng SY, Bigner DD, Ashley DM, Pirozzi CJ, He Y. Repurposing Clemastine to Target Glioblastoma Cell Stemness. Cancers (Basel) 2023; 15:4619. [PMID: 37760589 PMCID: PMC10526458 DOI: 10.3390/cancers15184619] [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: 08/13/2023] [Revised: 09/08/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Brain tumor-initiating cells (BTICs) and tumor cell plasticity promote glioblastoma (GBM) progression. Here, we demonstrate that clemastine, an over-the-counter drug for treating hay fever and allergy symptoms, effectively attenuated the stemness and suppressed the propagation of primary BTIC cultures bearing PDGFRA amplification. These effects on BTICs were accompanied by altered gene expression profiling indicative of their more differentiated states, resonating with the activity of clemastine in promoting the differentiation of normal oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes. Functional assays for pharmacological targets of clemastine revealed that the Emopamil Binding Protein (EBP), an enzyme in the cholesterol biosynthesis pathway, is essential for BTIC propagation and a target that mediates the suppressive effects of clemastine. Finally, we showed that a neural stem cell-derived mouse glioma model displaying predominantly proneural features was similarly susceptible to clemastine treatment. Collectively, these results identify pathways essential for maintaining the stemness and progenitor features of GBMs, uncover BTIC dependency on EBP, and suggest that non-oncology, low-toxicity drugs with OPC differentiation-promoting activity can be repurposed to target GBM stemness and aid in their treatment.
Collapse
Affiliation(s)
- Michael A. Sun
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Pathology Graduate Program, Duke University Medical Center, Durham, NC 27710, USA
| | - Rui Yang
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Heng Liu
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Pathology Graduate Program, Duke University Medical Center, Durham, NC 27710, USA
| | - Wenzhe Wang
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Xiao Song
- The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (X.S.); (B.H.); (S.-Y.C.)
| | - Bo Hu
- The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (X.S.); (B.H.); (S.-Y.C.)
| | - Nathan Reynolds
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kristen Roso
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Lee H. Chen
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Paula K. Greer
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Stephen T. Keir
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Roger E. McLendon
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; (X.S.); (B.H.); (S.-Y.C.)
| | - Darell D. Bigner
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - David M. Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher J. Pirozzi
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yiping He
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA; (M.A.S.); (R.Y.); (H.L.); (W.W.); (N.R.); (K.R.); (L.H.C.); (P.K.G.); (S.T.K.); (R.E.M.); (D.D.B.); (D.M.A.)
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
5
|
Brubaker LW, Backos DS, Nguyen VT, Reigan P, Yamamoto TM, Woodruff ER, Iwanaga R, Wempe MF, Kumar V, Persenaire C, Watson ZL, Bitler BG. Novel chromobox 2 inhibitory peptide decreases tumor progression. Expert Opin Ther Targets 2023:1-11. [PMID: 37243607 DOI: 10.1080/14728222.2023.2218614] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
BACKGROUND The Polycomb Repressor Complex 1 (PRC1) is an epigenetic regulator of differentiation and development, consisting of multiple subunits including RING1, BMI1, and Chromobox. The composition of PRC1 dictates its function and aberrant expression of specific subunits contributes to several diseases including cancer. Specifically, the reader protein Chromobox2 (CBX2) recognizes the repressive modifications including histone H3 lysine 27 tri-methylation (H3K27me3) and H3 lysine 9 dimethylation (H3K9me2). CBX2 is overexpressed in several cancers compared to the non-transformed cell counterparts, it promotes both cancer progression and chemotherapy resistance. Thus, inhibiting the reader function of CBX2 is an attractive and unique anti-cancer approach. RESEARCH DESIGN & METHODS Compared with other CBX family members, CBX2 has a unique A/T-hook DNA binding domain that is juxtaposed to the chromodomain (CD). Using a computational approach, we constructed a homology model of CBX2 encompassing the CD and A/T hook domain. We used the model as a basis for peptide design and identified blocking peptides that are predicted to directly bind the CD and A/T-hook regions of CBX2. These peptides were tested in vitro and in vivo models. CONCLUSION The CBX2 blocking peptide significantly inhibited both 2D and 3D growth of ovarian cancer cells, downregulated a CBX2 target gene, and blunted tumor growth in vivo.
Collapse
Affiliation(s)
- Lindsay W Brubaker
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Donald S Backos
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Vu T Nguyen
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Philip Reigan
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Tomomi M Yamamoto
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth R Woodruff
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ritsuko Iwanaga
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael F Wempe
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Vijay Kumar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Christianne Persenaire
- Department of Obstetrics & Gynecology, Division of Gynecologic Oncology, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zachary L Watson
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin G Bitler
- Department of Obstetrics & Gynecology, Division of Reproductive Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Comprehensive Cancer Center, Aurora, CO, USA
| |
Collapse
|
6
|
Kerrison WGJ, Thway K, Jones RL, Huang PH. The biology and treatment of leiomyosarcomas. Crit Rev Oncol Hematol 2023; 184:103955. [PMID: 36893945 DOI: 10.1016/j.critrevonc.2023.103955] [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: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Leiomyosarcoma (LMS) is a soft tissue sarcoma of smooth muscle origin that can arise in multiple anatomical sites and is broadly classified as extra-uterine LMS or uterine LMS. There is substantial interpatient heterogeneity within this histological subtype, and despite multi-modal therapy, clinical management remains challenging with poor patient prognosis and few new therapies available. Here we discuss the current treatment landscape of LMS in both the localised and advanced disease setting. We further describe the latest advances in our evolving understanding of the genetics and biology of this group of heterogeneous diseases and summarise the key studies delineating the mechanisms of acquired and intrinsic chemotherapy resistance in this histological subtype. We conclude by providing a perspective on how novel targeted agents such as PARP inhibitors may usher in a new paradigm of biomarker-driven therapies that will ultimately impact the outcomes of patients with LMS.
Collapse
Affiliation(s)
- William G J Kerrison
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Khin Thway
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom; The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Robin L Jones
- The Royal Marsden NHS Foundation Trust, London, United Kingdom; Division of Clinical Studies, The Institute of Cancer Research, London, United Kingdom
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
7
|
Lacuna K, Bose S, Ingham M, Schwartz G. Therapeutic advances in leiomyosarcoma. Front Oncol 2023; 13:1149106. [PMID: 36969049 PMCID: PMC10031121 DOI: 10.3389/fonc.2023.1149106] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
Leiomyosarcoma is an aggressive mesenchymal malignancy and represents one of the most common subtypes of soft tissue sarcomas. It is characterized by significant disease heterogeneity with variable sites of origin and diverse genomic profiles. As a result, the treatment of advanced leiomyosarcoma is challenging. First-line therapy for metastatic and/or unresectable leiomyosarcoma includes anthracycline or gemcitabine based regimens, which provide a median progression-free survival time of about 5 months and overall survival time between 14-16 months. Effective later-line therapies are limited. Molecular profiling has enhanced our knowledge of the pathophysiology driving leiomyosarcoma, providing potential targets for treatment. In this review, we explore recent advances in our understanding of leiomyosarcoma tumor biology and implications for novel therapeutics. We describe the development of clinical trials based on such findings and discuss available published results. To date, the most promising approaches for advanced leiomyosarcoma include targeting DNA damage repair pathways and aberrant metabolism associated with oncogenesis, as well as novel chemotherapy combinations. This review highlights the recent progress made in the treatment of advanced leiomyosarcoma. Ongoing progress is contingent upon further development of clinical trials based on molecular findings, with careful consideration for clinical trial design, strong academic collaborations, and prospective correlative analyses.
Collapse
Affiliation(s)
- Kristine Lacuna
- Division of Hematology and Medical Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | | | | | | |
Collapse
|
8
|
Dougherty J, Harvey K, Liou A, Labella K, Moran D, Brosius S, De Raedt T. Identification of therapeutic sensitivities in a spheroid drug combination screen of Neurofibromatosis Type I associated High Grade Gliomas. PLoS One 2023; 18:e0277305. [PMID: 36730269 PMCID: PMC9894422 DOI: 10.1371/journal.pone.0277305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/22/2022] [Indexed: 02/03/2023] Open
Abstract
Neurofibromatosis Type 1 (NF1) patients develop an array of benign and malignant tumors, of which Malignant Peripheral Nerve Sheath Tumors (MPNST) and High Grade Gliomas (HGG) have a dismal prognosis. About 15-20% of individuals with NF1 develop brain tumors and one third of these occur outside of the optic pathway. These non-optic pathway gliomas are more likely to progress to malignancy, especially in adults. Despite their low frequency, high grade gliomas have a disproportional effect on the morbidity of NF1 patients. In vitro drug combination screens have not been performed on NF1-associated HGG, hindering our ability to develop informed clinical trials. Here we present the first in vitro drug combination screen (21 compounds alone or in combination with MEK or PI3K inhibitors) on the only human NF1 patient derived HGG cell line available and on three mouse glioma cell lines derived from the NF1-P53 genetically engineered mouse model, which sporadically develop HGG. These mouse glioma cell lines were never exposed to serum, grow as spheres and express markers that are consistent with an Oligodendrocyte Precursor Cell (OPC) lineage origin. Importantly, even though the true cell of origin for HGG remains elusive, they are thought to arise from the OPC lineage. We evaluated drug sensitivities of the three murine glioma cell lines in a 3D spheroid growth assay, which more accurately reflects drug sensitivities in vivo. Excitingly, we identified six compounds targeting HDACs, BRD4, CHEK1, BMI-1, CDK1/2/5/9, and the proteasome that potently induced cell death in our NF1-associated HGG. Moreover, several of these inhibitors work synergistically with either MEK or PI3K inhibitors. This study forms the basis for further pre-clinical evaluation of promising targets, with an eventual hope to translate these to the clinic.
Collapse
Affiliation(s)
- Jacquelyn Dougherty
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kyra Harvey
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Angela Liou
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Katherine Labella
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Deborah Moran
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Stephanie Brosius
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department or Neurology, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Thomas De Raedt
- Department of Pediatrics, Children’s Hospital Philadelphia, Philadelphia, Pennsylvania, United States of America
- School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
9
|
Rubanov A, Berico P, Hernando E. Epigenetic Mechanisms Underlying Melanoma Resistance to Immune and Targeted Therapies. Cancers (Basel) 2022; 14:cancers14235858. [PMID: 36497341 PMCID: PMC9738385 DOI: 10.3390/cancers14235858] [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: 11/11/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Melanoma is an aggressive skin cancer reliant on early detection for high likelihood of successful treatment. Solar UV exposure transforms melanocytes into highly mutated tumor cells that metastasize to the liver, lungs, and brain. Even upon resection of the primary tumor, almost thirty percent of patients succumb to melanoma within twenty years. Identification of key melanoma genetic drivers led to the development of pharmacological BRAFV600E and MEK inhibitors, significantly improving metastatic patient outcomes over traditional cytotoxic chemotherapy or pioneering IFN-α and IL-2 immune therapies. Checkpoint blockade inhibitors releasing the immunosuppressive effects of CTLA-4 or PD-1 proved to be even more effective and are the standard first-line treatment. Despite these major improvements, durable responses to immunotherapy and targeted therapy have been hindered by intrinsic or acquired resistance. In addition to gained or selected genetic alterations, cellular plasticity conferred by epigenetic reprogramming is emerging as a driver of therapy resistance. Epigenetic regulation of chromatin accessibility drives gene expression and establishes distinct transcriptional cell states. Here we review how aberrant chromatin, transcriptional, and epigenetic regulation contribute to therapy resistance and discuss how targeting these programs sensitizes melanoma cells to immune and targeted therapies.
Collapse
Affiliation(s)
- Andrey Rubanov
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Pietro Berico
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Eva Hernando
- Department of Pathology, NYU Grossman School of Medicine, New York, NY 10016, USA
- Interdisciplinary Melanoma Cooperative Group, Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
10
|
Yang J, Xu Y, Chen Y, Li T, Zhang X, Hu T, Xing R, Yang Y. Therapeutic perspectives for adult soft tissue sarcoma-updates from the 2022 ASCO annual meeting. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0394. [PMID: 36350007 PMCID: PMC9630518 DOI: 10.20892/j.issn.2095-3941.2022.0394] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/05/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Jilong Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, China
| | - Yu Xu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Yong Chen
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Tao Li
- Department of Bone and Soft tissue Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xiaowei Zhang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Tu Hu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Ruwei Xing
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, China
| | - Yun Yang
- Department of Bone and Soft Tissue Tumor, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology, Tianjin 300060, China
| |
Collapse
|
11
|
Doyle EJ, Morey L, Conway E. Know when to fold 'em: Polycomb complexes in oncogenic 3D genome regulation. Front Cell Dev Biol 2022; 10:986319. [PMID: 36105358 PMCID: PMC9464936 DOI: 10.3389/fcell.2022.986319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Chromatin is spatially and temporally regulated through a series of orchestrated processes resulting in the formation of 3D chromatin structures such as topologically associating domains (TADs), loops and Polycomb Bodies. These structures are closely linked to transcriptional regulation, with loss of control of these processes a frequent feature of cancer and developmental syndromes. One such oncogenic disruption of the 3D genome is through recurrent dysregulation of Polycomb Group Complex (PcG) functions either through genetic mutations, amplification or deletion of genes that encode for PcG proteins. PcG complexes are evolutionarily conserved epigenetic complexes. They are key for early development and are essential transcriptional repressors. PcG complexes include PRC1, PRC2 and PR-DUB which are responsible for the control of the histone modifications H2AK119ub1 and H3K27me3. The spatial distribution of the complexes within the nuclear environment, and their associated modifications have profound effects on the regulation of gene transcription and the 3D genome. Nevertheless, how PcG complexes regulate 3D chromatin organization is still poorly understood. Here we glean insights into the role of PcG complexes in 3D genome regulation and compaction, how these processes go awry during tumorigenesis and the therapeutic implications that result from our insights into these mechanisms.
Collapse
Affiliation(s)
- Emma J. Doyle
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Lluis Morey
- Sylvester Comprehensive Cancer Centre, Miami, FL, United States
- Department of Human Genetics, Biomedical Research Building, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Eric Conway
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Xu J, Li L, Shi P, Cui H, Yang L. The Crucial Roles of Bmi-1 in Cancer: Implications in Pathogenesis, Metastasis, Drug Resistance, and Targeted Therapies. Int J Mol Sci 2022; 23:ijms23158231. [PMID: 35897796 PMCID: PMC9367737 DOI: 10.3390/ijms23158231] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/01/2022] Open
Abstract
B-cell-specific Moloney murine leukemia virus integration region 1 (Bmi-1, also known as RNF51 or PCGF4) is one of the important members of the PcG gene family, and is involved in regulating cell proliferation, differentiation and senescence, and maintaining the self-renewal of stem cells. Many studies in recent years have emphasized the role of Bmi-1 in the occurrence and development of tumors. In fact, Bmi-1 has multiple functions in cancer biology and is closely related to many classical molecules, including Akt, c-MYC, Pten, etc. This review summarizes the regulatory mechanisms of Bmi-1 in multiple pathways, and the interaction of Bmi-1 with noncoding RNAs. In particular, we focus on the pathological processes of Bmi-1 in cancer, and explore the clinical relevance of Bmi-1 in cancer biomarkers and prognosis, as well as its implications for chemoresistance and radioresistance. In conclusion, we summarize the role of Bmi-1 in tumor progression, reveal the pathophysiological process and molecular mechanism of Bmi-1 in tumors, and provide useful information for tumor diagnosis, treatment, and prognosis.
Collapse
Affiliation(s)
- Jie Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Lin Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
| | - Pengfei Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| | - Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China; (J.X.); (L.L.); (P.S.)
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China
- Correspondence: (H.C.); (L.Y.)
| |
Collapse
|
13
|
Recent progress on small molecules targeting epigenetic complexes. Curr Opin Chem Biol 2022; 67:102130. [DOI: 10.1016/j.cbpa.2022.102130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 12/16/2022]
|
14
|
Branstrom A, Cao L, Furia B, Trotta C, Santaguida M, Graci JD, Colacino JM, Ray B, Li W, Sheedy J, Mollin A, Yeh S, Kong R, Sheridan R, Baird JD, O'Keefe K, Spiegel R, Goodwin E, Keating S, Weetall M. Emvododstat, a Potent Dihydroorotate Dehydrogenase Inhibitor, Is Effective in Preclinical Models of Acute Myeloid Leukemia. Front Oncol 2022; 12:832816. [PMID: 35223511 PMCID: PMC8864546 DOI: 10.3389/fonc.2022.832816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Blocking the pyrimidine nucleotide de novo synthesis pathway by inhibiting dihydroorotate dehydrogenase (DHODH) results in the cell cycle arrest and/or differentiation of rapidly proliferating cells including activated lymphocytes, cancer cells, or virally infected cells. Emvododstat (PTC299) is an orally bioavailable small molecule that inhibits DHODH. We evaluated the potential for emvododstat to inhibit the progression of acute myeloid leukemia (AML) using several in vitro and in vivo models of the disease. Broad potent activity was demonstrated against multiple AML cell lines, AML blasts cultured ex vivo from patient blood samples, and AML tumor models including patient-derived xenograft models. Emvododstat induced differentiation, cytotoxicity, or both in primary AML patient blasts cultured ex vivo with 8 of 10 samples showing sensitivity. AML cells with diverse driver mutations were sensitive, suggesting the potential of emvododstat for broad therapeutic application. AML cell lines that are not sensitive to emvododstat are likely to be more reliant on the salvage pathway than on de novo synthesis of pyrimidine nucleotides. Pharmacokinetic experiments in rhesus monkeys demonstrated that emvododstat levels rose rapidly after oral administration, peaking about 2 hours post-dosing. This was associated with an increase in the levels of dihydroorotate (DHO), the substrate for DHODH, within 2 hours of dosing indicating that DHODH inhibition is rapid. DHO levels declined as drug levels declined, consistent with the reversibility of DHODH inhibition by emvododstat. These preclinical findings provide a rationale for clinical evaluation of emvododstat in an ongoing Phase 1 study of patients with relapsed/refractory acute leukemias.
Collapse
Affiliation(s)
- Arthur Branstrom
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Liangxian Cao
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Bansri Furia
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | | | | | - Jason D Graci
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Joseph M Colacino
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Balmiki Ray
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Wencheng Li
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Josephine Sheedy
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Anna Mollin
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Shirley Yeh
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Ronald Kong
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | | | - John D Baird
- Clinical, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Kylie O'Keefe
- Commercial, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Robert Spiegel
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Elizabeth Goodwin
- Scientific Writing, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Suzanne Keating
- Scientific Writing, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| | - Marla Weetall
- Research, PTC Therapeutics, Inc., South Plainfield, NJ, United States
| |
Collapse
|