1
|
Tamura T, Nagai S, Masuda K, Imaeda K, Sugihara E, Yamasaki J, Kawaida M, Otsuki Y, Suina K, Nobusue H, Akahane T, Chiyoda T, Kisu I, Kobayashi Y, Banno K, Sakurada K, Okita H, Yamaguchi R, Ahmed AA, Yamagami W, Saya H, Aoki D, Nagano O. mTOR-mediated p62/SQSTM1 stabilization confers a robust survival mechanism for ovarian cancer. Cancer Lett 2025; 616:217565. [PMID: 39971122 DOI: 10.1016/j.canlet.2025.217565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
Over 50 % of patients with high-grade serous carcinoma (HGSC) are homologous recombination proficient, making them refractory to platinum-based drugs and poly (ADP-ribose) polymerase (PARP) inhibitors. These patients often develop progressive resistance within 6 months after primary treatment and tend to die early, thus new therapies are urgently needed. In this study, we comprehensively investigated this tumor type by leveraging a combination of machine learning analysis of a large published dataset and newly developed genetically engineered HGSC organoid models from murine fallopian tubes. Aberrant activation of RAS/PI3K signaling was a signature of poor prognosis in BRCA1/2 wild-type ovarian cancer, and mTOR-induced elevated p62 expression was a robust marker of chemotherapy-induced mTOR-p62-NRF2 signal activation. mTOR inhibition with everolimus decreased p62 and enhanced sensitivity to conventional chemotherapy, indicating that p62 serves as an important biomarker for therapeutic intervention. Combination therapy with conventional chemotherapy and mTOR inhibitors is a promising therapeutic strategy for refractory HGSC, with p62 as a biomarker.
Collapse
Affiliation(s)
- Tomohiro Tamura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan; Department of Extended Intelligence for Medicine, The Ishii-Ishibashi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Shimpei Nagai
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan.
| | - Keiyo Imaeda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Eiji Sugihara
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Juntaro Yamasaki
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Miho Kawaida
- Division of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
| | - Yuji Otsuki
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Kentaro Suina
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA; Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroyuki Nobusue
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Tomoko Akahane
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Iori Kisu
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Sakurada
- Department of Extended Intelligence for Medicine, The Ishii-Ishibashi Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Hajime Okita
- Division of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan; Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ahmed Ashour Ahmed
- Ovarian Cancer Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, OX3 9DU, UK; Department of Gynecological Oncology, Churchill Hospital, Oxford University Hospitals, Oxford, OX3 7LE, UK; Oxford NIHR Biomedical Research Centre, Oxford, OX4 2PG, UK
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Nagano
- Division of Gene Regulation, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| |
Collapse
|
2
|
Bhatt J, Ghigo A, Hirsch E. PI3K/Akt in IPF: untangling fibrosis and charting therapies. Front Immunol 2025; 16:1549277. [PMID: 40248697 PMCID: PMC12004373 DOI: 10.3389/fimmu.2025.1549277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronic, progressive lung disease characterized by abnormal epithelial repair, persistent inflammation, and excessive extracellular matrix deposition, leading to irreversible scarring and respiratory failure. Central to its pathogenesis is the dysregulation of the PI3K/Akt signaling pathway, which drives fibroblast activation, epithelial-mesenchymal transition, apoptosis resistance, and cellular senescence. Senescent cells contribute to fibrosis through the secretion of pro-inflammatory and profibrotic factors in the senescence-associated secretory phenotype (SASP). Current antifibrotic therapies, Nintedanib and Pirfenidone, only slow disease progression and are limited by side effects, highlighting the need for novel treatments. This review focuses on the role of PI3K/Akt signaling in IPF pathogenesis, its intersection with inflammation and fibrosis, and emerging therapeutic approaches targeting molecules along this pathway.
Collapse
Affiliation(s)
- Janki Bhatt
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| |
Collapse
|
3
|
Middleton G, Robbins HL, Fletcher P, Savage J, Mehmi M, Summers Y, Greystoke A, Steele N, Popat S, Jain P, Spicer J, Cave J, Shaw P, Gilligan D, Power D, Fennell D, Bajracharya M, McBride DJ, Maheswari U, Frankell AM, Swanton C, Beggs AD, Billingham L. A phase II trial of mTORC1/2 inhibition in STK11 deficient non small cell lung cancer. NPJ Precis Oncol 2025; 9:67. [PMID: 40069402 PMCID: PMC11897347 DOI: 10.1038/s41698-025-00838-4] [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: 07/30/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025] Open
Abstract
There are no current stratified medicine options for STK11-deficient NSCLC. STK11 loss mediates mTORC activation, GLUT1 up-regulation and increased glycolysis. This metabolic reprogramming might represent a therapeutic vulnerability targetable with mTORC1/2 inhibition. In arm B2 of the National Lung Matrix Trial 54 patients with NSCLC received vistusertib, of which 49 were STK11-deficient (30 with KRAS mutation (B2D), 19 without (B2S)). Objective response (OR) and durable clinical benefit (DCB) rates with 95% credible intervals (CrI) were estimated from posterior probability distributions generated using Bayesian beta-binomial conjugate analysis. In B2D, 2 per-protocol patients obtained OR (estimated true OR rate (95%CrI) 9.8% (2.4-24.3). Estimates of true DCB rate (95%CrI): B2D 24.4% (11.1-42.3), B2S 14.6% (3.6-34.7). Overall, vistusertib cannot be recommended in this context. Longitudinal ctDNA analysis demonstrates enrichment of SMARCA4 mutations post-treatment. In vitro studies show adaptive resistance to mTORC1/2 inhibition via AKT reactivation. (NCT02664935, ISRCTN38344105, EudraCT 2014-000814-73, 10 June 2015).
Collapse
Affiliation(s)
- Gary Middleton
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK.
- Department of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Helen L Robbins
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Immunology & Immunotherapy, University of Birmingham, Birmingham, UK
| | - Peter Fletcher
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Joshua Savage
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Manita Mehmi
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | | | | | | | | | - Pooja Jain
- St James's University Hospital, Leeds, UK
| | - James Spicer
- King's College London, Guy's Hospital, London, UK
| | - Judith Cave
- Southampton University Hospitals NHS Trust, Southampton, UK
| | | | | | | | | | | | | | | | - Alexander M Frankell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Andrew D Beggs
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Department of Cancer & Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Lucinda Billingham
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| |
Collapse
|
4
|
Scott SC, Farago A, Lai WV, Zahurak M, Rudek MA, Murray J, Carducci MA, Uziel T, Takebe N, Gore SD, Rudin CM, Hann CL. A phase 1 study of the combination of BH3-mimetic, navitoclax, and mTORC1/2 inhibitor, vistusertib, in patients with advanced solid tumors. Cancer Chemother Pharmacol 2025; 95:37. [PMID: 39998620 DOI: 10.1007/s00280-025-04760-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE To determine the, safety, tolerability and recommended phase 2 dosing of the combination of navitoclax, a dual Bcl-2/xL inhibitor, and vistusertib, a TORC1/2 inhibitor. METHODS Patients with advanced solid tumors received navitoclax plus vistusertib following a 3 + 3 dose escalation design. To mitigate thrombocytopenia, a known toxicity of navitoclax, all patients received lead-in dosing of navitoclax alone at 150 mg orally daily for a minimum of 7 days. In addition to safety and tolerability, pharmacokinetics of navitoclax and vistusertib were evaluated. RESULTS 14 patients received combination treatment which was well-tolerated at dose level 1 (navitoclax 150 mg orally daily plus vistusertib 35 mg orally twice daily). The main dose-limiting toxicity, grade 3 serum aminotransferase elevation, occurred in two of five patients at dose level 2 (navitoclax 250 mg orally daily plus vistusertib 35 mg orally twice daily). Navitoclax and vistusertib exposures appeared consistent with levels reported in prior studies of each agent. No responses were observed among the 8 response evaluable patients. CONCLUSIONS A tolerable dose of navitoclax at 150 mg orally daily plus vistusertib at 35 mg orally twice daily was identified in patients with advanced solid tumors and established as the recommended phase 2 dose (RP2D). Further efficacy assessment of this combination, in a planned phase 2 expansion in patients with relapsed small cell lung cancer, was terminated due to discontinuation of vistusertib. TRIAL REGISTRATION NCT03366103 (First posted December 8, 2017).
Collapse
Affiliation(s)
- Susan C Scott
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Anna Farago
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - W Victoria Lai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marianna Zahurak
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology Biostatistics, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Michelle A Rudek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Judy Murray
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Michael A Carducci
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Tamar Uziel
- Oncology Discovery, AbbVie Inc, North Chicago, IL, USA
| | - Naoko Takebe
- IDB/CTEP/NCI, National Cancer Institute, Rockville, MD, USA
| | - Steven D Gore
- IDB/CTEP/NCI, National Cancer Institute, Rockville, MD, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christine L Hann
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Bettedi L, Zhang Y, Yang S, Lilly MA. Unveiling GATOR2 Function: Novel Insights from Drosophila Research. Cells 2024; 13:1795. [PMID: 39513902 PMCID: PMC11545208 DOI: 10.3390/cells13211795] [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: 08/09/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The multiprotein Target of Rapamycin (TOR) Complex 1 (TORC1) is a serine/threonine kinase that stimulates anabolic metabolism and suppresses catabolism. Deregulation of TORC1 is implicated in various human pathologies, including cancer, epilepsy, and neurodegenerative disorders. The Gap Activity Towards Rags (GATOR) complex contains two subcomplexes: GATOR1, which inhibits TORC1 activity; and GATOR2, which counteracts GATOR1s function. Structural and biochemical studies have elucidated how GATOR1 regulates TORC1 activity by acting as a GTPase activating protein for Rag GTPase. However, while cryogenic electron microscopy has determined that the structure of the multi-protein GATOR2 complex is conserved from yeast to humans, how GATOR2 inhibits GATOR1 remains unclear. Here, we describe recent whole-animal studies in Drosophila that have yielded novel insights into GATOR2 function, including identifying a novel role for the GATOR2 subunit WDR59, redefining the core proteins sufficient for GATOR2 activity, and defining a TORC1-independent role for GATOR2 in the regulation of the lysosomal autophagic endomembrane system. Additionally, the recent characterization of a novel methionine receptor in Drosophila that acts through the GATOR2 complex suggests an attractive model for the evolution of species-specific nutrient sensors. Research on GATOR2 function in Drosophila highlights how whole-animal genetic models can be used to dissect intracellular signaling pathways to identify tissue-specific functions and functional redundancies that may be missed in studies confined to rapidly proliferating cell lines.
Collapse
Affiliation(s)
- Lucia Bettedi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (L.B.); (S.Y.)
| | - Yingbiao Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China;
| | - Shu Yang
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (L.B.); (S.Y.)
| | - Mary A. Lilly
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA; (L.B.); (S.Y.)
| |
Collapse
|
6
|
Aleksandrova KV, Vorobev ML, Suvorova II. mTOR pathway occupies a central role in the emergence of latent cancer cells. Cell Death Dis 2024; 15:176. [PMID: 38418814 PMCID: PMC10902345 DOI: 10.1038/s41419-024-06547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/18/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
The current focus in oncology research is the translational control of cancer cells as a major mechanism of cellular plasticity. Recent evidence has prompted a reevaluation of the role of the mTOR pathway in cancer development leading to new conclusions. The mechanistic mTOR inhibition is well known to be a tool for generating quiescent stem cells and cancer cells. In response to mTOR suppression, quiescent cancer cells dynamically change their proteome, triggering alternative non-canonical translation mechanisms. The shift to selective translation may have clinical relevance, since quiescent tumor cells can acquire new phenotypical features. This review provides new insights into the patterns of mTOR functioning in quiescent cancer cells, enhancing our current understanding of the biology of latent metastasis.
Collapse
Affiliation(s)
| | - Mikhail L Vorobev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russian Federation
| | - Irina I Suvorova
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russian Federation.
| |
Collapse
|
7
|
Szalai F, Sztankovics D, Krencz I, Moldvai D, Pápay J, Sebestyén A, Khoor A. Rictor-A Mediator of Progression and Metastasis in Lung Cancer. Cancers (Basel) 2024; 16:543. [PMID: 38339294 PMCID: PMC10854599 DOI: 10.3390/cancers16030543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Lung carcinoma is one of the most common cancer types for both men and women. Despite recent breakthroughs in targeted therapy and immunotherapy, it is characterized by a high metastatic rate, which can significantly affect quality of life and prognosis. Rictor (encoded by the RICTOR gene) is known as a scaffold protein for the multiprotein complex mTORC2. Among its diverse roles in regulating essential cellular functions, mTORC2 also facilitates epithelial-mesenchymal transition and metastasis formation. Amplification of the RICTOR gene and subsequent overexpression of the Rictor protein can result in the activation of mTORC2, which promotes cell survival and migration. Based on recent studies, RICTOR amplification or Rictor overexpression can serve as a marker for mTORC2 activation, which in turn provides a promising druggable target. Although selective inhibitors of Rictor and the Rictor-mTOR association are only in a preclinical phase, they seem to be potent novel approaches to reduce tumor cell migration and metastasis formation. Here, we summarize recent advances that support an important role for Rictor and mTORC2 as potential therapeutic targets in the treatment of lung cancer. This is a traditional (narrative) review based on Pubmed and Google Scholar searches for the following keywords: Rictor, RICTOR amplification, mTORC2, Rictor complexes, lung cancer, metastasis, progression, mTOR inhibitors.
Collapse
Affiliation(s)
- Fatime Szalai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Andras Khoor
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
8
|
Huang X, You L, Nepovimova E, Psotka M, Malinak D, Valko M, Sivak L, Korabecny J, Heger Z, Adam V, Wu Q, Kuca K. Inhibitors of phosphoinositide 3-kinase (PI3K) and phosphoinositide 3-kinase-related protein kinase family (PIKK). J Enzyme Inhib Med Chem 2023; 38:2237209. [PMID: 37489050 PMCID: PMC10392309 DOI: 10.1080/14756366.2023.2237209] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/11/2023] [Indexed: 02/02/2024] Open
Abstract
Phosphoinositide 3-kinases (PI3K) and phosphoinositide 3-kinase-related protein kinases (PIKK) are two structurally related families of kinases that play vital roles in cell growth and DNA damage repair. Dysfunction of PIKK members and aberrant stimulation of the PI3K/AKT/mTOR signalling pathway are linked to a plethora of diseases including cancer. In recent decades, numerous inhibitors related to the PI3K/AKT/mTOR signalling have made great strides in cancer treatment, like copanlisib and sirolimus. Notably, most of the PIKK inhibitors (such as VX-970 and M3814) related to DNA damage response have also shown good efficacy in clinical trials. However, these drugs still require a suitable combination therapy to overcome drug resistance or improve antitumor activity. Based on the aforementioned facts, we summarised the efficacy of PIKK, PI3K, and AKT inhibitors in the therapy of human malignancies and the resistance mechanisms of targeted therapy, in order to provide deeper insights into cancer treatment.
Collapse
Affiliation(s)
- Xueqin Huang
- College of Life Science, Yangtze University, Jingzhou, China
| | - Li You
- College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
| | - Miroslav Psotka
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - David Malinak
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Bratislava, Slovakia
| | - Ladislav Sivak
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Jan Korabecny
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, China
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
9
|
Banerjee S, Giannone G, Clamp AR, Ennis DP, Glasspool RM, Herbertson R, Krell J, Riisnaes R, Mirza HB, Cheng Z, McDermott J, Green C, Kristeleit RS, George A, Gourley C, Lewsley LA, Rai D, Banerji U, Hinsley S, McNeish IA. Efficacy and Safety of Weekly Paclitaxel Plus Vistusertib vs Paclitaxel Alone in Patients With Platinum-Resistant Ovarian High-Grade Serous Carcinoma: The OCTOPUS Multicenter, Phase 2, Randomized Clinical Trial. JAMA Oncol 2023; 9:675-682. [PMID: 36928279 PMCID: PMC10020933 DOI: 10.1001/jamaoncol.2022.7966] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/05/2022] [Indexed: 03/18/2023]
Abstract
Importance Patients with platinum-resistant or refractory ovarian high-grade serous carcinoma (PR-HGSC) have a poor prognosis and few therapeutic options. Preclinical studies support targeting PI3K/AKT/mTOR signaling in this setting, and a phase 1 study of the dual mTORC1/mTORC2 inhibitor vistusertib with weekly paclitaxel showed activity. Objective To evaluate whether the addition of vistusertib to weekly paclitaxel improves clinical outcomes in patients with PR-HGSC. Design, Setting, and Participants This phase 2, double-blind, placebo-controlled multicenter randomized clinical trial recruited patients from UK cancer centers between January 2016 and March 2018. Patients with PR-HGSC of ovarian, fallopian tube, or primary peritoneal origin and with measurable or evaluable disease (Response Evaluation Criteria in Solid Tumors version 1.1 and/or Gynecological Cancer Intergroup cancer antigen 125 criteria) were eligible. There were no restrictions on number of lines of prior therapy. Data analysis was performed from May 2019 to January 2022. Interventions Patients were randomized (1:1) to weekly paclitaxel (80 mg/m2 days 1, 8, and 15 of a 28-day cycle) plus oral vistusertib (50 mg twice daily) or placebo. Main Outcomes and Measures The primary end point was progression-free survival in the intention-to-treat population. Secondary end points included response rate, overall survival, and quality of life. Results A total of 140 patients (median [range] age, 63 [36-86] years; 17.9% with platinum-refractory disease; 53.6% with ≥3 prior therapies) were randomized. In the paclitaxel plus vistusertib vs paclitaxel plus placebo groups, there was no difference in progression-free survival (median, 4.5 vs 4.1 months; hazard ratio [HR], 0.84; 80% CI, 0.67-1.07; 1-sided P = .18), overall survival (median, 9.7 vs 11.1 months; HR, 1.21; 80% CI, 0.91-1.60) or response rate (odds ratio, 0.86; 80% CI, 0.55-1.36). Grade 3 to 4 adverse events were 41.2% (weekly paclitaxel plus vistusertib) vs 36.7% (weekly paclitaxel plus placebo), and there was no difference in quality of life. Conclusions and Relevance In this randomized clinical trial of weekly paclitaxel and dual mTORC1/2 inhibition in patients with PR-HGSC, vistusertib did not improve clinical activity of weekly paclitaxel. Trial Registration isrctn.org Identifier: ISRCTN16426935.
Collapse
Affiliation(s)
- Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Clinical Studies, Institute of Cancer Research, London, United Kingdom
| | - Gaia Giannone
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Andrew R. Clamp
- The Christie NHS Foundation Trust and University of Manchester, Manchester, United Kingdom
| | - Darren P. Ennis
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | | | - Rebecca Herbertson
- Sussex Cancer Centre, Royal Sussex County Hospital, Brighton, United Kingdom
| | - Jonathan Krell
- Medical Oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Ruth Riisnaes
- Division of Cancer Therapeutics, Institute of Cancer Research, London, United Kingdom
| | - Hasan B. Mirza
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Zhao Cheng
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Jacqueline McDermott
- Department of Histopathology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Clare Green
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Rebecca S. Kristeleit
- Research Department of Oncology, UCL Cancer Institute, University College London, London, United Kingdom
- Now with Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Angela George
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Charlie Gourley
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Liz-Anne Lewsley
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Debbie Rai
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Udai Banerji
- Division of Cancer Therapeutics, Institute of Cancer Research, London, United Kingdom
| | - Samantha Hinsley
- CRUK Glasgow Clinical Trials Unit, Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Iain A. McNeish
- Ovarian Cancer Action Research Centre, Department of Surgery and Cancer, Imperial College London, United Kingdom
- Medical Oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
10
|
Inhibition of branched-chain alpha-keto acid dehydrogenase kinase augments the sensitivity of ovarian and breast cancer cells to paclitaxel. Br J Cancer 2023; 128:896-906. [PMID: 36526674 PMCID: PMC9977917 DOI: 10.1038/s41416-022-02095-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
CONTEXT Many cancer patients who initially respond to chemotherapy eventually develop chemoresistance, and to address this, we previously conducted a RNAi screen to identify genes contributing to resistance. One of the hits from the screen was branched-chain α-keto acid dehydrogenase kinase (BCKDK). BCKDK controls the metabolism of branched-chain amino acids (BCAAs) through phosphorylation and inactivation of the branched-chain α-keto acid dehydrogenase complex (BCKDH), thereby inhibiting catabolism of BCAAs. METHODS We measured the impact on paclitaxel sensitivity of inhibiting BCKDK in ovarian and breast cancer cell lines. RESULTS Inhibition of BCKDK using siRNA or two chemical inhibitors (BCKDKi) was synergistic with paclitaxel in both breast and ovarian cancer cells. BCKDKi reduced levels of BCAA and the addition of exogenous BCAA suppressed this synergy. BCKDKi inactivated the mTORC1-Aurora pathway, allowing cells to overcame M-phase arrest induced by paclitaxel. In some cases, cells almost completed cytokinesis, then reverted to a single cell, resulting in multinucleate cells. CONCLUSION BCKDK is an attractive target to augment the sensitivity of cancer cells to paclitaxel.
Collapse
|
11
|
Bernal L, Pinzi L, Rastelli G. Identification of Promising Drug Candidates against Prostate Cancer through Computationally-Driven Drug Repurposing. Int J Mol Sci 2023; 24:ijms24043135. [PMID: 36834548 PMCID: PMC9964599 DOI: 10.3390/ijms24043135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Prostate cancer (PC) is one of the most common types of cancer in males. Although early stages of PC are generally associated with favorable outcomes, advanced phases of the disease present a significantly poorer prognosis. Moreover, currently available therapeutic options for the treatment of PC are still limited, being mainly focused on androgen deprivation therapies and being characterized by low efficacy in patients. As a consequence, there is a pressing need to identify alternative and more effective therapeutics. In this study, we performed large-scale 2D and 3D similarity analyses between compounds reported in the DrugBank database and ChEMBL molecules with reported anti-proliferative activity on various PC cell lines. The analyses included also the identification of biological targets of ligands with potent activity on PC cells, as well as investigations on the activity annotations and clinical data associated with the more relevant compounds emerging from the ligand-based similarity results. The results led to the prioritization of a set of drugs and/or clinically tested candidates potentially useful in drug repurposing against PC.
Collapse
Affiliation(s)
- Leonardo Bernal
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 103, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-059-2058564
| |
Collapse
|
12
|
Martins FC, Couturier DL, de Santiago I, Sauer CM, Vias M, Angelova M, Sanders D, Piskorz A, Hall J, Hosking K, Amirthanayagam A, Cosulich S, Carnevalli L, Davies B, Watkins TBK, Funingana IG, Bolton H, Haldar K, Latimer J, Baldwin P, Crawford R, Eldridge M, Basu B, Jimenez-Linan M, Mcpherson AW, McGranahan N, Litchfield K, Shah SP, McNeish I, Caldas C, Evan G, Swanton C, Brenton JD. Clonal somatic copy number altered driver events inform drug sensitivity in high-grade serous ovarian cancer. Nat Commun 2022; 13:6360. [PMID: 36289203 PMCID: PMC9606297 DOI: 10.1038/s41467-022-33870-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 10/06/2022] [Indexed: 01/12/2023] Open
Abstract
Chromosomal instability is a major challenge to patient stratification and targeted drug development for high-grade serous ovarian carcinoma (HGSOC). Here we show that somatic copy number alterations (SCNAs) in frequently amplified HGSOC cancer genes significantly correlate with gene expression and methylation status. We identify five prevalent clonal driver SCNAs (chromosomal amplifications encompassing MYC, PIK3CA, CCNE1, KRAS and TERT) from multi-regional HGSOC data and reason that their strong selection should prioritise them as key biomarkers for targeted therapies. We use primary HGSOC spheroid models to test interactions between in vitro targeted therapy and SCNAs. MYC chromosomal copy number is associated with in-vitro and clinical response to paclitaxel and in-vitro response to mTORC1/2 inhibition. Activation of the mTOR survival pathway in the context of MYC-amplified HGSOC is statistically associated with increased prevalence of SCNAs in genes from the PI3K pathway. Co-occurrence of amplifications in MYC and genes from the PI3K pathway is independently observed in squamous lung cancer and triple negative breast cancer. In this work, we show that identifying co-occurrence of clonal driver SCNA genes could be used to tailor therapeutics for precision medicine.
Collapse
Affiliation(s)
- Filipe Correia Martins
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK.
- Experimental Medicine Initiative, University of Cambridge, Cambridge, UK.
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Gynaecological Oncology, Cambridge University Hospitals, Cambridge, UK.
| | - Dominique-Laurent Couturier
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Ines de Santiago
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Maria Vias
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Mihaela Angelova
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Deborah Sanders
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Anna Piskorz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - James Hall
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Ionut G Funingana
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Helen Bolton
- Department of Gynaecological Oncology, Cambridge University Hospitals, Cambridge, UK
| | - Krishnayan Haldar
- Department of Gynaecological Oncology, Cambridge University Hospitals, Cambridge, UK
| | - John Latimer
- Department of Gynaecological Oncology, Cambridge University Hospitals, Cambridge, UK
| | - Peter Baldwin
- Department of Gynaecological Oncology, Cambridge University Hospitals, Cambridge, UK
| | - Robin Crawford
- Department of Gynaecological Oncology, Cambridge University Hospitals, Cambridge, UK
| | - Matthew Eldridge
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Bristi Basu
- Cambridge University Hospitals, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Andrew W Mcpherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Centre, NYC, USA
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kevin Litchfield
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Centre, NYC, USA
| | - Iain McNeish
- Department of Surgery and Cancer, Imperial College of London, London, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Gerard Evan
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
| | - James D Brenton
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK.
- Department of Oncology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Dual mTORC1/2 inhibitor AZD2014 diminishes myeloid-derived suppressor cells accumulation in ovarian cancer and delays tumor growth. Cancer Lett 2021; 523:72-81. [PMID: 34560229 DOI: 10.1016/j.canlet.2021.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 02/08/2023]
Abstract
Mechanistic target of rapamycin (mTOR) forms two distinct complexes, mTOR complex 1 (mTORC1) and mTORC2. Here we investigated the antitumor effect of dual mTORC1/2 inhibitor AZD2014 on epithelial ovarian cancer (EOC) and its potential effect on immunosuppressive myeloid-derived suppressor cells (MDSCs). Immunohistochemical analysis of mTORC1 and mTORC2 was performed on a human ovarian cancer tissue microarray. High mTORC2 expression level was associated with shorter survival in EOC, whereas mTORC1 was not correlate with patients' prognosis. AZD2014 suppressed mTOR signaling pathway in ovarian cancer cells, inhibited proliferation and induced G1-phase cell cycle arrest and apoptosis. In tumor-bearing mice, AZD2014 treatment limited tumor growth, reduced peritoneal ascites, and prolonged survival. AZD2014 specifically reduced MDSCs migration and accumulation in EOC peritoneal fluid but not in the spleen. Moreover, subsequent AZD2014 treatment after cisplatin chemotherapy delayed EOC recurrence. Collectively, we observed that high mTORC2 expression level in EOC indicated a poor prognosis. Remarkably, in tumor-bearing mice, AZD2014 diminished MDSC accumulation and delayed tumor growth and recurrence.
Collapse
|
14
|
Morscher RJ, Brard C, Berlanga P, Marshall LV, André N, Rubino J, Aerts I, De Carli E, Corradini N, Nebchi S, Paoletti X, Mortimer P, Lacroix L, Pierron G, Schleiermacher G, Vassal G, Geoerger B. First-in-child phase I/II study of the dual mTORC1/2 inhibitor vistusertib (AZD2014) as monotherapy and in combination with topotecan-temozolomide in children with advanced malignancies: arms E and F of the AcSé-ESMART trial. Eur J Cancer 2021; 157:268-277. [PMID: 34543871 DOI: 10.1016/j.ejca.2021.08.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/15/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
AIM Arms E and F of the AcSé-ESMART phase I/II platform trial aimed to define the recommended dose and preliminary activity of the dual mTORC1/2 inhibitor vistusertib as monotherapy and with topotecan-temozolomide in a molecularly enriched population of paediatric patients with relapsed/refractory malignancies. In addition, we evaluated genetic phosphatidylinositol 3-kinase (PI3K)/AKT/ mammalian (or mechanistic) target of rapamycin (mTOR) pathway alterations across the Molecular Profiling for Paediatric and Young Adult Cancer Treatment Stratification (MAPPYACTS) trial (NCT02613962). EXPERIMENTAL DESIGN AND RESULTS Four patients were treated in arm E and 10 in arm F with a median age of 14.3 years. Main diagnoses were glioma and sarcoma. Dose escalation was performed as per the continuous reassessment method, expansion in an Ensign design. The vistusertib single agent administered at 75 mg/m2 twice a day (BID) on 2 days/week and vistusertib 30 mg/m2 BID on 3 days/week combined with temozolomide 100 mg/m2/day and topotecan 0.50 mg/m2/day on the first 5 days of each 4-week cycle were safe. Treatment was well tolerated with the main toxicity being haematological. Pharmacokinetics indicates equivalent exposure in children compared with adults. Neither tumour response nor prolonged stabilisation was observed, including in the 12 patients whose tumours exhibited PI3K/AKT/mTOR pathway alterations. Advanced profiling across relapsed/refractory paediatric cancers of the MAPPYACTS cohort shows genetic alterations associated with this pathway in 28.0% of patients, with 10.5% carrying mutations in the core pathway genes. CONCLUSIONS Vistusertib was well tolerated in paediatric patients. Study arms were terminated because of the absence of tumour responses and insufficient target engagement of vistusertib observed in adult trials. Targeting the PI3K/AKT/mTOR pathway remains a therapeutic avenue to be explored in paediatric patients. CLINICAL TRIAL IDENTIFIER NCT2813135.
Collapse
Affiliation(s)
- Raphael J Morscher
- Gustave Roussy Cancer Campus, Department of Paediatric and Adolescent Oncology, Villejuif, France; INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Caroline Brard
- Gustave Roussy Cancer Campus, Biostatistics and Epidemiology Unit, INSERM U1018, CESP, Université Paris-Saclay, Université Paris-Sud, UVSQ, Villejuif, France
| | - Pablo Berlanga
- Gustave Roussy Cancer Campus, Department of Paediatric and Adolescent Oncology, Villejuif, France
| | - Lynley V Marshall
- Paediatric and Adolescent Oncology Drug Development Unit, The Royal Marsden Hospital & The Institute of Cancer Research, London, United Kingdom
| | - Nicolas André
- Department of Paediatric Hematology & Oncology, Hôpital de la Timone, AP-HM, Marseille, France; UMR Inserm 1068, CNRS UMR 7258, Aix Marseille Université U105, Marseille Cancer Research Center (CRCM), Marseille, France
| | - Jonathan Rubino
- Gustave Roussy Cancer Campus, Clinical Research Direction, Villejuif, France
| | - Isabelle Aerts
- SIREDO Oncology Center, Institut Curie, PSL Research University, Paris, France
| | - Emilie De Carli
- Centre Hospitalier Universitaire, Department of Paediatric Oncology, Angers, France
| | - Nadège Corradini
- Pediatric Oncology Department, Institute of Pediatric Hematology and Oncology, Centre Leon Berard, Lyon, France
| | - Souad Nebchi
- Gustave Roussy Cancer Campus, Biostatistics and Epidemiology Unit, INSERM U1018, CESP, Université Paris-Saclay, Université Paris-Sud, UVSQ, Villejuif, France
| | - Xavier Paoletti
- Gustave Roussy Cancer Campus, Biostatistics and Epidemiology Unit, INSERM U1018, CESP, Université Paris-Saclay, Université Paris-Sud, UVSQ, Villejuif, France
| | | | - Ludovic Lacroix
- Department of Medical Biology and Pathology of Translational Research and Biobank, AMMICA, Laboratory INSERM US23/CNRS UMS3655, Gustave Roussy Cancer Campus, Université Paris-Saclay, 94805 Villejuif, France
| | - Gaelle Pierron
- Unité de Génétique Somatique, Service d'oncogénétique, Institut Curie, Centre Hospitalier, Paris, France
| | - Gudrun Schleiermacher
- SIREDO Oncology Center, Institut Curie, PSL Research University, Paris, France; Laboratory of Translational Research in Paediatric Oncology - INSERM U830, Paris, France
| | - Gilles Vassal
- Gustave Roussy Cancer Campus, Clinical Research Direction, Villejuif, France
| | - Birgit Geoerger
- Gustave Roussy Cancer Campus, Department of Paediatric and Adolescent Oncology, Villejuif, France; INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
15
|
Quantitation of vistusertib by UHPLC-MS/MS in rat plasma and its application to a pharmacokinetic study. Bioanalysis 2021; 13:1333-1341. [PMID: 34470474 DOI: 10.4155/bio-2021-0152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: The present study aimed to develop a UHPLC-MS/MS method for determination of vistusertib in biological matrix, and to describe the pharmacokinetic behavior of vistusertib in SD rats. Methodology & results: After protein precipitation with acetone and acetonitrile (1:1), the chromatographic separation was achieved on an Agilent Poroshell 120 EC-C18 column and detected with a SCIEX QTRAP 4500 mass spectrometer under positive ionization mode. The developed UHPLC-MS/MS method showed an excellent linearity within the range of 1.0-3000 ng/ml with good accuracy and precision. Vistusertib showed a rapid absorption and reached the maximum concentration of 3532.2 ± 678.0 ng/ml 20-30 min after oral administration in Sprague-Dawley rats. Conclusion: The established analytical method was fast, sensitive and robust, and successfully applied to describe the pharmacokinetic behavior of vistusertib following an oral administration in rats.
Collapse
|
16
|
Grosso S, Marini A, Gyuraszova K, Voorde JV, Sfakianos A, Garland GD, Tenor AR, Mordue R, Chernova T, Morone N, Sereno M, Smith CP, Officer L, Farahmand P, Rooney C, Sumpton D, Das M, Teodósio A, Ficken C, Martin MG, Spriggs RV, Sun XM, Bushell M, Sansom OJ, Murphy D, MacFarlane M, Le Quesne JPC, Willis AE. The pathogenesis of mesothelioma is driven by a dysregulated translatome. Nat Commun 2021; 12:4920. [PMID: 34389715 PMCID: PMC8363647 DOI: 10.1038/s41467-021-25173-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 07/25/2021] [Indexed: 12/22/2022] Open
Abstract
Malignant mesothelioma (MpM) is an aggressive, invariably fatal tumour that is causally linked with asbestos exposure. The disease primarily results from loss of tumour suppressor gene function and there are no 'druggable' driver oncogenes associated with MpM. To identify opportunities for management of this disease we have carried out polysome profiling to define the MpM translatome. We show that in MpM there is a selective increase in the translation of mRNAs encoding proteins required for ribosome assembly and mitochondrial biogenesis. This results in an enhanced rate of mRNA translation, abnormal mitochondrial morphology and oxygen consumption, and a reprogramming of metabolic outputs. These alterations delimit the cellular capacity for protein biosynthesis, accelerate growth and drive disease progression. Importantly, we show that inhibition of mRNA translation, particularly through combined pharmacological targeting of mTORC1 and 2, reverses these changes and inhibits malignant cell growth in vitro and in ex-vivo tumour tissue from patients with end-stage disease. Critically, we show that these pharmacological interventions prolong survival in animal models of asbestos-induced mesothelioma, providing the basis for a targeted, viable therapeutic option for patients with this incurable disease.
Collapse
Affiliation(s)
- Stefano Grosso
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Alberto Marini
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Katarina Gyuraszova
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | | | | | - Gavin D Garland
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Angela Rubio Tenor
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Ryan Mordue
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Tanya Chernova
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Nobu Morone
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Marco Sereno
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Claire P Smith
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Leah Officer
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Pooyeh Farahmand
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Claire Rooney
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - David Sumpton
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Madhumita Das
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Ana Teodósio
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Catherine Ficken
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Maria Guerra Martin
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Ruth V Spriggs
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Xiao-Ming Sun
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Owen J Sansom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK
| | - Daniel Murphy
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| | - Marion MacFarlane
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK.
| | - John P C Le Quesne
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK.
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Cancer Research UK Beatson Institute, Garscube Estate, Bearsden, UK.
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK.
- Glenfield Hospital, Groby Road, University Hospitals Leicester NHS Trust Leicester, Leicester, UK.
| | - Anne E Willis
- MRC Toxicology Unit, Gleeson Building, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Evaluating Targeted Therapies in Ovarian Cancer Metabolism: Novel Role for PCSK9 and Second Generation mTOR Inhibitors. Cancers (Basel) 2021; 13:cancers13153727. [PMID: 34359627 PMCID: PMC8345177 DOI: 10.3390/cancers13153727] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Dysregulated lipid metabolism is emerging as a hallmark in several malignancies, including ovarian cancer (OC). Specifically, metastatic OC is highly dependent on lipid-rich omentum. We aimed to investigate the therapeutic value of targeting lipid metabolism in OC. For this purpose, we studied the role of PCSK9, a cholesterol-regulating enzyme, in OC cell survival and its downstream signaling. We also investigated the cytotoxic efficacy of a small library of metabolic (n = 11) and mTOR (n = 10) inhibitors using OC cell lines (n = 8) and ex vivo patient-derived cell cultures (PDCs, n = 5) to identify clinically suitable drug vulnerabilities. Targeting PCSK9 expression with siRNA or PCSK9 specific inhibitor (PF-06446846) impaired OC cell survival. In addition, overexpression of PCSK9 induced robust AKT phosphorylation along with increased expression of ERK1/2 and MEK1/2, suggesting a pro-survival role of PCSK9 in OC cells. Moreover, our drug testing revealed marked differences in cytotoxic responses to drugs targeting metabolic pathways of high-grade serous ovarian cancer (HGSOC) and low-grade serous ovarian cancer (LGSOC) PDCs. Our results show that targeting PCSK9 expression could impair OC cell survival, which warrants further investigation to address the dependency of this cancer on lipogenesis and omental metastasis. Moreover, the differences in metabolic gene expression and drug responses of OC PDCs indicate the existence of a metabolic heterogeneity within OC subtypes, which should be further explored for therapeutic improvements.
Collapse
|
18
|
Stockton JD, Tee L, Whalley C, James J, Dilworth M, Wheat R, Nieto T, Geh I, Barros-Silva JD, Beggs AD. Complete response to neoadjuvant chemoradiotherapy in rectal cancer is associated with RAS/AKT mutations and high tumour mutational burden. Radiat Oncol 2021; 16:129. [PMID: 34256782 PMCID: PMC8278688 DOI: 10.1186/s13014-021-01853-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Pathological complete response (pathCR) in rectal cancer is beneficial, as up to 75% of patients do not experience regrowth of the primary tumour, but it is poorly understood. We hypothesised that the changes seen in the pre-treatment biopsies of pathCR but not seen in residual tumour after chemoradiotherapy were the determinants of responsiveness. METHODS Two groups of patients with either complete response (pathCR group, N = 24) or no response (poor response group, N = 24) were retrieved. Pre-treatment biopsies of cancers from these patients underwent high read depth amplicon sequencing for a targeted panel, exome sequencing, methylation profiling and immunohistochemistry for DNA repair pathway proteins. RESULTS Twenty four patients who underwent pathCR and twenty-four who underwent poor response underwent molecular characterisation. Patients in the pathCR group had significantly higher tumour mutational burden and neoantigen load, frequent copy number alterations but fewer structural variants and enrichment for driver mutations in the PI3K/AKT/mTOR signalling pathway. There were no significant differences in tumour heterogeneity as measured by MATH score. Methylation analysis demonstrated enrichment for hypomethyation in the PI3K/AKT/mTOR signalling pathway. DISCUSSION The phenomenon of pathCR in rectal cancer may be related to immunovisibility caused by a high tumour mutational burden phenotype. Potential therapy resistance mechanisms involve the PI3K/AKT/mTOR signalling pathway, but tumour heterogeneity does not seem to play a role in resistance.
Collapse
Affiliation(s)
- Joanne D. Stockton
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Louise Tee
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Celina Whalley
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Jonathan James
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Mark Dilworth
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Rachel Wheat
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Thomas Nieto
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - S-CORT Consortium
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Ian Geh
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - João D. Barros-Silva
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
| | - Andrew D. Beggs
- Surgical Research Laboratory, Institute of Cancer and Genomic Science, University of Birmingham, Vincent Drive, Birmingham, B15 2TT UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
19
|
Yu L, Wei J, Liu P. Attacking the PI3K/Akt/mTOR signaling pathway for targeted therapeutic treatment in human cancer. Semin Cancer Biol 2021; 85:69-94. [PMID: 34175443 DOI: 10.1016/j.semcancer.2021.06.019] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
Cancer is the second leading cause of human death globally. PI3K/Akt/mTOR signaling is one of the most frequently dysregulated signaling pathways observed in cancer patients that plays crucial roles in promoting tumor initiation, progression and therapy responses. This is largely due to that PI3K/Akt/mTOR signaling is indispensable for many cellular biological processes, including cell growth, metastasis, survival, metabolism, and others. As such, small molecule inhibitors targeting major kinase components of the PI3K/Akt/mTOR signaling pathway have drawn extensive attention and been developed and evaluated in preclinical models and clinical trials. Targeting a single kinase component within this signaling usually causes growth arrest rather than apoptosis associated with toxicity-induced adverse effects in patients. Combination therapies including PI3K/Akt/mTOR inhibitors show improved patient response and clinical outcome, albeit developed resistance has been reported. In this review, we focus on revealing the mechanisms leading to the hyperactivation of PI3K/Akt/mTOR signaling in cancer and summarizing efforts for developing PI3K/Akt/mTOR inhibitors as either mono-therapy or combination therapy in different cancer settings. We hope that this review will facilitate further understanding of the regulatory mechanisms governing dysregulation of PI3K/Akt/mTOR oncogenic signaling in cancer and provide insights into possible future directions for targeted therapeutic regimen for cancer treatment, by developing new agents, drug delivery systems, or combination regimen to target the PI3K/Akt/mTOR signaling pathway. This information will also provide effective patient stratification strategy to improve the patient response and clinical outcome for cancer patients with deregulated PI3K/Akt/mTOR signaling.
Collapse
Affiliation(s)
- Le Yu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
20
|
High in vitro and in vivo synergistic activity between mTORC1 and PLK1 inhibition in adenocarcinoma NSCLC. Oncotarget 2021; 12:859-872. [PMID: 33889306 PMCID: PMC8057272 DOI: 10.18632/oncotarget.27930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/15/2021] [Indexed: 01/22/2023] Open
Abstract
Significant rational is available for specific targeting of PI3K/AKT/mTOR pathway in the treatment of non-small cell lung cancer (NSCLC). However, almost all clinical trials that have evaluated Pi3K pathway-based monotherapies/combinations did not observe an improvement of patient’s outcome. The aim of our study was therefore to define combination of treatment based on the determination of predictive markers of resistance to the mTORC1 inhibitor RAD001/Everolimus. An in vivo study showed high efficacy of RAD001 in NSCLC Patient-Derived Xenografts (PDXs). When looking at biomarkers of resistance by RT-PCR study, three genes were found to be highly expressed in resistant tumors, i.e., PLK1, CXCR4, and AXL. We have then focused our study on the combination of RAD001 + Volasertib, a PLK1 inhibitor, and observed a high antitumor activity of the combination in comparison to each monotherapy; similarly, a clear synergistic effect between the two compounds was found in an in vitro study. Pharmacodynamics study demonstrated that this synergy was due to (1) tumor vascularization decrease, increase of the HIF1 protein expression and decrease of the intracellular pH, and (2) decrease of the Carbonic Anhydrase 9 (CAIX) protein that could not correct intracellular acidosis. In conclusion, all these preclinical data strongly suggest that the inhibition of mTORC1 and PLK1 proteins may be a promising therapeutic approach for NSCLC patients.
Collapse
|
21
|
Gao M, Zhou Q. [Progress in Treatment of Advanced Squamous Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:866-874. [PMID: 32798441 PMCID: PMC7583871 DOI: 10.3779/j.issn.1009-3419.2020.101.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lung cancer ranks first both in mobidity and motality in china and worldwild. Squamous cell lung carcinoma is distinguished from lung adenocarcinoma in diagnosis and treatment due to its unique clinical & pathological manifestations and gene mutation characteristics, as a result, it is also explored as a separate type in clinical researches. In addition to the clinical manifestations of elderly, central tumors, late staging at diagnosis, and multiple comorbidities, the lack of driver genes makes it nearly impossible for these patients to benefit from targeted therapy. However, the exploration of targeted therapy for lung squamous cell carcinoma has never stopped. Several new drugs including fibroblast growth factor receptor tyrosine kinase inhibitor and cyclin dependent kinase 4 and 6 inhibitors have been studied in multiple phase I studies specifically in patients with lung squamous cell carcinoma. However, with the development of immunotherapy, the characteristics such as complex gene mutation and high tumor mutation burden makes it possible for patients with squamous cell lung cancer to benefit from immunotherapy combined with chemotherapy. This review will provide an overview of the treatment progress in advanced squamous cell lung cancer including chemotherapy, targeted therapy and immunotherapy.
.
Collapse
Affiliation(s)
- Ming Gao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China.,Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| |
Collapse
|
22
|
Wang Q, Peng H, Qi X, Wu M, Zhao X. Targeted therapies in gynecological cancers: a comprehensive review of clinical evidence. Signal Transduct Target Ther 2020; 5:137. [PMID: 32728057 PMCID: PMC7391668 DOI: 10.1038/s41392-020-0199-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/12/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Advanced and recurrent gynecological cancers are associated with poor prognosis and lack of effective treatment. The developments of the molecular mechanisms on cancer progression provide insight into novel targeted therapies, which are emerging as groundbreaking and promising cancer treatment strategies. In gynecologic malignancies, potential therapeutic targeted agents include antiangiogenic agents, poly (ADP-ribose) polymerase (PARP) inhibitors, tumor-intrinsic signaling pathway inhibitors, selective estrogen receptor downregulators, and immune checkpoint inhibitors. In this article, we provide a comprehensive review of the clinical evidence of targeted agents in gynecological cancers and discuss the future implication.
Collapse
Affiliation(s)
- Qiao Wang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hongling Peng
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
23
|
Jiang Y, Wang C, Zhou S. Targeting tumor microenvironment in ovarian cancer: Premise and promise. Biochim Biophys Acta Rev Cancer 2020; 1873:188361. [PMID: 32234508 DOI: 10.1016/j.bbcan.2020.188361] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 02/05/2023]
Abstract
Ovarian cancer is the leading cause of gynecological cancer-related mortality globally. The majority of ovarian cancer patients suffer from relapse after standard of care therapies and the clinical benefits from cancer therapies are not satisfactory owing to drug resistance. Certain novel drugs targeting the components of tumor microenvironment (TME) have been approved by US Food and Drug Administration in solid cancers. As such, the passion is rekindled to exploit the role of TME in ovarian cancer progression and metastasis for discovery of novel therapeutics for this deadly disease. In the current review, we revisit the recent mechanistic insights into the contributions of TME to the development, progression, prognosis prediction and therapeutic efficacy of ovarian cancer via modulating cancer hallmarks. We also explored potentially promising predictive and prognostic biomarkers for ovarian cancer patients.
Collapse
Affiliation(s)
- Yuting Jiang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, PR China
| | - Chengdi Wang
- Department of Respiratory and Critical Care Medicine, West China Medical School/West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, PR China.
| |
Collapse
|
24
|
Abstract
With the discovery of rapamycin 45 years ago, studies in the mechanistic target of rapamycin (mTOR) field started 2 decades before the identification of the mTOR kinase. Over the years, studies revealed that the mTOR signaling is a master regulator of homeostasis and integrates a variety of environmental signals to regulate cell growth, proliferation, and metabolism. Deregulation of mTOR signaling, particularly hyperactivation, frequently occurs in human tumors. Recent advances in molecular profiling have identified mutations or amplification of certain genes coding proteins involved in the mTOR pathway (eg, PIK3CA, PTEN, STK11, and RICTOR) as the most common reasons contributing to mTOR hyperactivation. These genetic alterations of the mTOR pathway are frequently observed in lung neoplasms and may serve as a target for personalized therapy. mTOR inhibitor monotherapy has met limited clinical success so far; however, rational drug combinations are promising to improve efficacy and overcome acquired resistance. A better understanding of mTOR signaling may have the potential to help translation of mTOR pathway inhibitors into the clinical setting.
Collapse
|
25
|
Kaipio K, Chen P, Roering P, Huhtinen K, Mikkonen P, Östling P, Lehtinen L, Mansuri N, Korpela T, Potdar S, Hynninen J, Auranen A, Grénman S, Wennerberg K, Hautaniemi S, Carpén O. ALDH1A1-related stemness in high-grade serous ovarian cancer is a negative prognostic indicator but potentially targetable by EGFR/mTOR-PI3K/aurora kinase inhibitors. J Pathol 2019; 250:159-169. [PMID: 31595974 DOI: 10.1002/path.5356] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 09/05/2019] [Accepted: 10/03/2019] [Indexed: 12/16/2022]
Abstract
Poor chemotherapy response remains a major treatment challenge for high-grade serous ovarian cancer (HGSC). Cancer stem cells are the major contributors to relapse and treatment failure as they can survive conventional therapy. Our objectives were to characterise stemness features in primary patient-derived cell lines, correlate stemness markers with clinical outcome and test the response of our cells to both conventional and exploratory drugs. Tissue and ascites samples, treatment-naive and/or after neoadjuvant chemotherapy, were prospectively collected. Primary cancer cells, cultured under conditions favouring either adherent or spheroid growth, were tested for stemness markers; the same markers were analysed in tissue and correlated with chemotherapy response and survival. Drug sensitivity and resistance testing was performed with 306 oncology compounds. Spheroid growth condition HGSC cells showed increased stemness marker expression (including aldehyde dehydrogenase isoform I; ALDH1A1) as compared with adherent growth condition cells, and increased resistance to platinum and taxane. A set of eight stemness markers separated treatment-naive tumours into two clusters and identified a distinct subgroup of HGSC with enriched stemness features. Expression of ALDH1A1, but not most other stemness markers, was increased after neoadjuvant chemotherapy and its expression in treatment-naive tumours correlated with chemoresistance and reduced survival. In drug sensitivity and resistance testing, five compounds, including two PI3K-mTOR inhibitors, demonstrated significant activity in both cell culture conditions. Thirteen compounds, including EGFR, PI3K-mTOR and aurora kinase inhibitors, were more toxic to spheroid cells than adherent cells. Our results identify stemness markers in HGSC that are associated with a decreased response to conventional chemotherapy and reduced survival if expressed by treatment-naive tumours. EGFR, mTOR-PI3K and aurora kinase inhibitors are candidates for targeting this cell population. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Katja Kaipio
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Ping Chen
- Integrated Cardio Metabolic Centre (ICMC), Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Pia Roering
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kaisa Huhtinen
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Piia Mikkonen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Päivi Östling
- Science for Life Laboratory Department of Oncology & Pathology, Karolinska Institutet, Huddinge, Sweden.,Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Laura Lehtinen
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Naziha Mansuri
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Taina Korpela
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland, High Throughput Biomedicine Unit (HTB), University of Helsinki, Helsinki, Finland
| | - Johanna Hynninen
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Annika Auranen
- Department of Obstetrics and Gynaecology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Seija Grénman
- Department of Obstetrics and Gynaecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, High Throughput Biomedicine Unit (HTB), University of Helsinki, Helsinki, Finland.,Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Sampsa Hautaniemi
- Research Programs Unit, Genome-Scale Biology and Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli Carpén
- Research Center for Cancer, Infections and Immunity, Institute of Biomedicine, University of Turku, Turku, Finland.,Research Programs Unit, Genome-Scale Biology and Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
Katopodis P, Chudasama D, Wander G, Sales L, Kumar J, Pandhal M, Anikin V, Chatterjee J, Hall M, Karteris E. Kinase Inhibitors and Ovarian Cancer. Cancers (Basel) 2019; 11:E1357. [PMID: 31547471 PMCID: PMC6770231 DOI: 10.3390/cancers11091357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is fifth in the rankings of cancer deaths among women, and accounts for more deaths than any other gynecological malignancy. Despite some improvement in overall-(OS) and progression-free survival (PFS) following surgery and first-line chemotherapy, there is a need for development of novel and more effective therapeutic strategies. In this mini review, we provide a summary of the current landscape of the clinical use of tyrosine kinase inhibitors (TKIs) and mechanistic target of rapamycin (mTOR) inhibitors in ovarian cancer. Emerging data from phase I and II trials reveals that a combinatorial treatment that includes TKIs and chemotherapy agents seems promising in terms of PFS despite some adverse effects recorded; whereas the use of mTOR inhibitors seems less effective. There is a need for further research into the inhibition of multiple signaling pathways in ovarian cancer and progression to phase III trials for drugs that seem most promising.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
| | - Dimple Chudasama
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Gurleen Wander
- Chelsea and Westminster Hospital NHS Trust, London UB9 6JH, UK.
| | - Louise Sales
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Juhi Kumar
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Manreen Pandhal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
- Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Jayanta Chatterjee
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK.
| | - Marcia Hall
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK.
| | - Emmanouil Karteris
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
27
|
Hua H, Kong Q, Zhang H, Wang J, Luo T, Jiang Y. Targeting mTOR for cancer therapy. J Hematol Oncol 2019; 12:71. [PMID: 31277692 PMCID: PMC6612215 DOI: 10.1186/s13045-019-0754-1] [Citation(s) in RCA: 601] [Impact Index Per Article: 100.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/14/2019] [Indexed: 02/05/2023] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a protein kinase regulating cell growth, survival, metabolism, and immunity. mTOR is usually assembled into several complexes such as mTOR complex 1/2 (mTORC1/2). In cooperation with raptor, rictor, LST8, and mSin1, key components in mTORC1 or mTORC2, mTOR catalyzes the phosphorylation of multiple targets such as ribosomal protein S6 kinase β-1 (S6K1), eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), Akt, protein kinase C (PKC), and type-I insulin-like growth factor receptor (IGF-IR), thereby regulating protein synthesis, nutrients metabolism, growth factor signaling, cell growth, and migration. Activation of mTOR promotes tumor growth and metastasis. Many mTOR inhibitors have been developed to treat cancer. While some of the mTOR inhibitors have been approved to treat human cancer, more mTOR inhibitors are being evaluated in clinical trials. Here, we update recent advances in exploring mTOR signaling and the development of mTOR inhibitors for cancer therapy. In addition, we discuss the mechanisms underlying the resistance to mTOR inhibitors in cancer cells.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qingbin Kong
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Luo
- Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|