1
|
Krishna AP, John S, Shinde PL, Mishra R. Proteo-transcriptomics meta-analysis identifies SUMO2 as a promising target in glioblastoma multiforme therapeutics. Cancer Cell Int 2021; 21:575. [PMID: 34715855 PMCID: PMC8555349 DOI: 10.1186/s12935-021-02279-y] [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: 07/13/2021] [Accepted: 10/18/2021] [Indexed: 11/10/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a deadly brain tumour with minimal survival rates due to the ever-expanding heterogeneity, chemo and radioresistance. Kinases are known to crucially drive GBM pathology; however, a rationale therapeutic combination that can simultaneously inhibit multiple kinases has not yet emerged successfully. Results Here, we analyzed the GBM patient data from several publicly available repositories and deduced hub GBM kinases, most of which were identified to be SUMOylated by SUMO2/3 isoforms. Not only the hub kinases but a significant proportion of GBM upregulated genes involved in proliferation, metastasis, invasion, epithelial-mesenchymal transition, stemness, DNA repair, stromal and macrophages maintenance were also identified to be the targets of SUMO2 isoform. Correlatively, high expression of SUMO2 isoform was found to be significantly associated with poor patient survival. Conclusions Although many natural products and drugs are evidenced to target general SUMOylation, however, our meta-analysis strongly calls for the need to design SUMO2/3 or even better SUMO2 specific inhibitors and also explore the SUMO2 transcription inhibitors for universally potential, physiologically non-toxic anti-GBM drug therapy. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02279-y. The major highlights of this study are as follows:Key upregulated hub kinases and coding genes in GBM are found to be targets of SUMO2 conjugation. SUMO2 is significantly expressed in adult primary and recurrent GBMs as well as in pediatric GBM tumours. Orthotropic xenografts from adult and pediatric GBMs confirm high expression of SUMO2 in GBM tumour samples. SUMO2 is significantly associated with patient survival plot and pan-cancer cell fitness. Rationale design of SUMO2 inhibitors or search for its transcriptional inhibitors is urgently required through industry-academia collaboration for an anti-GBM and potentially pan-cancer therapeutics.
Collapse
Affiliation(s)
- Aswani P Krishna
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Sebastian John
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Puja Laxmanrao Shinde
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India
| | - Rashmi Mishra
- Brain and Cerebro-Vascular Mechanobiology Research, Laboratory of Translational Mechanobiology, Department of Neurobiology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, 695014, Kerala, India.
| |
Collapse
|
2
|
Manju CA, Jeena K, Ramachandran R, Manohar M, Ambily AM, Sajesh KM, Gowd GS, Menon K, Pavithran K, Pillai A, Nair SV, Koyakutty M. Intracranially injectable multi-siRNA nanomedicine for the inhibition of glioma stem cells. Neurooncol Adv 2021; 3:vdab104. [PMID: 34604750 PMCID: PMC8482790 DOI: 10.1093/noajnl/vdab104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Nanoparticle siRNA-conjugates are promising clinical therapeutics as indicated by recent US-FDA approval. In glioma stem cells (GSC), multiple stemness associated genes were found aberrant. We report intracranially injectable, multi-gene-targeted siRNA nanoparticle gel (NPG) for the combinatorial silencing of 3 aberrant genes, thus inhibiting the tumorogenic potential of GSCs. Methods NPG loaded with siRNAs targeted against FAK, NOTCH-1, and SOX-2 were prepared by the self-assembly of siRNAs with protamine-hyaluronic acid combination. Electron microscopy, DLS, and agarose gel electrophoresis were used for the physicochemical characterization. Cell transfection and gene-silencing efficiency were studied using human mesenchymal stem cells and rat C6 glioma-derived GSCs. Neurosphere inhibition was tested in vitro using GSCs derived from C6 cell line and glioma patient samples. Patient-derived xenograft model and orthotopic rat glioma model were used to test the effect of NPG on in vivo tumorigenicity. Results The siRNA nanoparticles with an average size ~ 250 nm and ~ 95% loading efficiency showed cellular uptake in ~95.5% GSCs. Simultaneous gene silencing of FAK, NOTCH-1, and SOX-2 led to the inhibition of neurosphere formation by GSCs, whereas normal stem cells remained unaffected and retained neuronal differentiation capability. GBM PDX models manifested significant impairment in the tumorigenic potential of NPG treated GSCs. Intracranial injection of NPG inhibited tumor growth in orthotopic rat brain tumor model. Conclusion Intracranially injectable n-siRNA NPG targeted to multiple stem-cell signaling impairs glioma initiation capabilities of GSCs and inhibited tumor growth in vivo.
Collapse
Affiliation(s)
- Cheripelil Abraham Manju
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Kottarapat Jeena
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Ranjith Ramachandran
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesh Manohar
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Anna Mathew Ambily
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | | | | | - Krishnakumar Menon
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Keechilat Pavithran
- Department of Oncology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Ashok Pillai
- Department of Neurosurgery, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Shantikumar V Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Manzoor Koyakutty
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
3
|
Dual Specificity Kinase DYRK3 Promotes Aggressiveness of Glioblastoma by Altering Mitochondrial Morphology and Function. Int J Mol Sci 2021; 22:ijms22062982. [PMID: 33804169 PMCID: PMC8000785 DOI: 10.3390/ijms22062982] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a malignant primary brain tumor with poor patient prognosis. Although the standard treatment of GBM is surgery followed by chemotherapy and radiotherapy, often a small portion of surviving tumor cells acquire therapeutic resistance and become more aggressive. Recently, altered kinase expression and activity have been shown to determine metabolic flux in tumor cells and metabolic reprogramming has emerged as a tumor progression regulatory mechanism. Here we investigated novel kinase-mediated metabolic alterations that lead to acquired GBM radioresistance and malignancy. We utilized transcriptomic analyses within a radioresistant GBM orthotopic xenograft mouse model that overexpresses the dual specificity tyrosine-phosphorylation-regulated kinase 3 (DYRK3). We find that within GBM cells, radiation exposure induces DYRK3 expression and DYRK3 regulates mammalian target of rapamycin complex 1 (mTORC1) activity through phosphorylation of proline-rich AKT1 substrate 1 (PRAS40). We also find that DYRK3 knockdown inhibits dynamin-related protein 1 (DRP1)-mediated mitochondrial fission, leading to increased oxidative phosphorylation (OXPHOS) and reduced glycolysis. Importantly, enforced DYRK3 downregulation following irradiation significantly impaired GBM cell migration and invasion. Collectively, we suggest DYRK3 suppression may be a novel strategy for preventing GBM malignancy through regulating mitochondrial metabolism.
Collapse
|
4
|
Sivakumar KC, Haixiao J, Naman CB, Sajeevan TP. Prospects of multitarget drug designing strategies by linking molecular docking and molecular dynamics to explore the protein-ligand recognition process. Drug Dev Res 2020; 81:685-699. [PMID: 32329098 DOI: 10.1002/ddr.21673] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/24/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
Abstract
The designing of drugs that can simultaneously affect different protein targets is one novel and promising way to treat complex diseases. Multitarget drugs act on multiple protein receptors each implicated in the same disease state, and may be considered to be more beneficial than conventional drug therapies. For example, these drugs can have improved therapeutic potency due to synergistic effects on multiple targets, as well as improved safety and resistance profiles due to the combined regulation of potential primary therapeutic targets and compensatory elements and lower dosage typically required. This review analyzes in-silico methods that facilitate multitarget drug design that facilitate the discovery and development of novel therapeutic agents. Here presented is a summary of the progress in structure-based drug discovery techniques that study the process of molecular recognition of targets and ligands, moving from static molecular docking to improved molecular dynamics approaches in multitarget drug design, and the advantages and limitations of each.
Collapse
Affiliation(s)
- Krishnankutty Chandrika Sivakumar
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India.,Bioinformatics Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Jin Haixiao
- Li Dak Sum Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China
| | - C Benjamin Naman
- Li Dak Sum Marine Biopharmaceutical Research Center, Department of Marine Pharmacy, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, China.,Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, California, USA
| | - T P Sajeevan
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Kochi, India
| |
Collapse
|
5
|
O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, Martinez-Lage M, Brem S, Maloney E, Shen A, Isaacs R, Mohan S, Plesa G, Lacey SF, Navenot JM, Zheng Z, Levine BL, Okada H, June CH, Brogdon JL, Maus MV. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med 2018; 9:9/399/eaaa0984. [PMID: 28724573 DOI: 10.1126/scitranslmed.aaa0984] [Citation(s) in RCA: 1090] [Impact Index Per Article: 181.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/09/2017] [Indexed: 12/18/2022]
Abstract
We conducted a first-in-human study of intravenous delivery of a single dose of autologous T cells redirected to the epidermal growth factor receptor variant III (EGFRvIII) mutation by a chimeric antigen receptor (CAR). We report our findings on the first 10 recurrent glioblastoma (GBM) patients treated. We found that manufacturing and infusion of CAR-modified T cell (CART)-EGFRvIII cells are feasible and safe, without evidence of off-tumor toxicity or cytokine release syndrome. One patient has had residual stable disease for over 18 months of follow-up. All patients demonstrated detectable transient expansion of CART-EGFRvIII cells in peripheral blood. Seven patients had post-CART-EGFRvIII surgical intervention, which allowed for tissue-specific analysis of CART-EGFRvIII trafficking to the tumor, phenotyping of tumor-infiltrating T cells and the tumor microenvironment in situ, and analysis of post-therapy EGFRvIII target antigen expression. Imaging findings after CART immunotherapy were complex to interpret, further reinforcing the need for pathologic sampling in infused patients. We found trafficking of CART-EGFRvIII cells to regions of active GBM, with antigen decrease in five of these seven patients. In situ evaluation of the tumor environment demonstrated increased and robust expression of inhibitory molecules and infiltration by regulatory T cells after CART-EGFRvIII infusion, compared to pre-CART-EGFRvIII infusion tumor specimens. Our initial experience with CAR T cells in recurrent GBM suggests that although intravenous infusion results in on-target activity in the brain, overcoming the adaptive changes in the local tumor microenvironment and addressing the antigen heterogeneity may improve the efficacy of EGFRvIII-directed strategies in GBM.
Collapse
Affiliation(s)
- Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - MacLean P Nasrallah
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arati Desai
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jan J Melenhorst
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Jennifer J D Morrissette
- Division of Precision and Computational Diagnostics, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria Martinez-Lage
- Division of Neuropathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven Brem
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eileen Maloney
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Angela Shen
- Novartis Oncology, East Hanover, NJ 07936, USA
| | - Randi Isaacs
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Suyash Mohan
- Division of Neuroradiology, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon F Lacey
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean-Marc Navenot
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhaohui Zheng
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA.
| |
Collapse
|
6
|
Phosphoproteomics Reveals HMGA1, a CK2 Substrate, as a Drug-Resistant Target in Non-Small Cell Lung Cancer. Sci Rep 2017; 7:44021. [PMID: 28290473 PMCID: PMC5349541 DOI: 10.1038/srep44021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/03/2017] [Indexed: 12/13/2022] Open
Abstract
Although EGFR tyrosine kinase inhibitors (TKIs) have demonstrated good efficacy in non-small-cell lung cancer (NSCLC) patients harboring EGFR mutations, most patients develop intrinsic and acquired resistance. We quantitatively profiled the phosphoproteome and proteome of drug-sensitive and drug-resistant NSCLC cells under gefitinib treatment. The construction of a dose-dependent responsive kinase-substrate network of 1548 phosphoproteins and 3834 proteins revealed CK2-centric modules as the dominant core network for the potential gefitinib resistance-associated proteins. CK2 knockdown decreased cell survival in gefitinib-resistant NSCLCs. Using motif analysis to identify the CK2 core sub-network, we verified that elevated phosphorylation level of a CK2 substrate, HMGA1 was a critical node contributing to EGFR-TKI resistance in NSCLC cell. Both HMGA1 knockdown or mutation of the CK2 phosphorylation site, S102, of HMGA1 reinforced the efficacy of gefitinib in resistant NSCLC cells through reactivation of the downstream signaling of EGFR. Our results delineate the TKI resistance-associated kinase-substrate network, suggesting a potential therapeutic strategy for overcoming TKI-induced resistance in NSCLC.
Collapse
|
7
|
Puduvalli VK, Chaudhary R, McClugage SG, Markert J. Beyond Alkylating Agents for Gliomas: Quo Vadimus? Am Soc Clin Oncol Educ Book 2017; 37:175-186. [PMID: 28561663 PMCID: PMC5803081 DOI: 10.1200/edbk_175003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Recent advances in therapies have yielded notable success in terms of improved survival in several cancers. However, such treatments have failed to improve outcome in patients with gliomas for whom surgery followed by radiation therapy and chemotherapy with alkylating agents remain the standard of care. Genetic and epigenetic studies have helped identify several alterations specific to gliomas. Attempts to target these altered pathways have been unsuccessful due to various factors, including tumor heterogeneity, adaptive resistance of tumor cells, and limitations of access across the blood-brain barrier. Novel therapies that circumvent such limitations have been the focus of intense study and include approaches such as immunotherapy, targeting of signaling hubs and metabolic pathways, and use of biologic agents. Immunotherapeutic approaches including tumor-targeted vaccines, immune checkpoint blockade, antibody-drug conjugates, and chimeric antigen receptor-expressing cell therapies are in various stages of clinical trials. Similarly, identification of key metabolic pathways or converging hubs of signaling pathways that are tumor specific have yielded novel targets for therapy of gliomas. In addition, the failure of conventional therapies against gliomas has led to a growing interest among patients in the use of alternative therapies, which in turn has necessitated developing evidence-based approaches to the application of such therapies in clinical studies. The development of these novel approaches bears potential for providing breakthroughs in treatment of more meaningful and improved outcomes for patients with gliomas.
Collapse
Affiliation(s)
- Vinay K Puduvalli
- From The Ohio State University Comprehensive Cancer Center, Columbus, OH; University of Cincinnati, Cincinnati, OH; University of Alabama at Birmingham, Birmingham, AL
| | - Rekha Chaudhary
- From The Ohio State University Comprehensive Cancer Center, Columbus, OH; University of Cincinnati, Cincinnati, OH; University of Alabama at Birmingham, Birmingham, AL
| | - Samuel G McClugage
- From The Ohio State University Comprehensive Cancer Center, Columbus, OH; University of Cincinnati, Cincinnati, OH; University of Alabama at Birmingham, Birmingham, AL
| | - James Markert
- From The Ohio State University Comprehensive Cancer Center, Columbus, OH; University of Cincinnati, Cincinnati, OH; University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
8
|
Jayaram S, Gupta MK, Raju R, Gautam P, Sirdeshmukh R. Multi-Omics Data Integration and Mapping of Altered Kinases to Pathways Reveal Gonadotropin Hormone Signaling in Glioblastoma. ACTA ACUST UNITED AC 2016; 20:736-746. [DOI: 10.1089/omi.2016.0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Savita Jayaram
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- School of Life Sciences, Manipal University, Manipal, India
| | - Manoj Kumar Gupta
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- School of Life Sciences, Manipal University, Manipal, India
| | - Rajesh Raju
- Computational Biology and Bioinformatics, Rajiv Gandhi Center for Biotechnology, Thiruvananthapuram, India
| | - Poonam Gautam
- National Institute of Pathology, ICMR, New Delhi, India
| | - Ravi Sirdeshmukh
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- Mazumdar Shaw Centre for Translational Research, Narayana Hrudayalaya Health City, Bangalore, India
| |
Collapse
|
9
|
Johnson LA, Scholler J, Ohkuri T, Kosaka A, Patel PR, McGettigan SE, Nace AK, Dentchev T, Thekkat P, Loew A, Boesteanu AC, Cogdill AP, Chen T, Fraietta JA, Kloss CC, Posey AD, Engels B, Singh R, Ezell T, Idamakanti N, Ramones MH, Li N, Zhou L, Plesa G, Seykora JT, Okada H, June CH, Brogdon JL, Maus MV. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci Transl Med 2015; 7:275ra22. [PMID: 25696001 PMCID: PMC4467166 DOI: 10.1126/scitranslmed.aaa4963] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Chimeric antigen receptors (CARs) are synthetic molecules designed to redirect T cells to specific antigens. CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of surface targets with limited expression in normal tissues. The variant III mutation of the epidermal growth factor receptor (EGFRvIII) results from an in-frame deletion of a portion of the extracellular domain, creating a neoepitope. We chose a vector backbone encoding a second-generation CAR based on efficacy of a murine scFv-based CAR in a xenograft model of glioblastoma. Next, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells; a low-affinity scFv was selected on the basis of its specificity for EGFRvIII over wild-type EGFR. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to specifically lyse, proliferate, and secrete cytokines in response to antigen-bearing targets. We further evaluated the specificity of the lead CAR candidate in vitro against EGFR-expressing keratinocytes and in vivo in a model of mice grafted with normal human skin. EGFRvIII-directed CAR T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFRvIII(+) glioblastoma. On the basis of these results, we have designed a phase 1 clinical study of CAR T cells transduced with humanized scFv directed to EGFRvIII in patients with either residual or recurrent glioblastoma (NCT02209376).
Collapse
Affiliation(s)
- Laura A Johnson
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA. Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Scholler
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Takayuki Ohkuri
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Akemi Kosaka
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Prachi R Patel
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Arben K Nace
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tzvete Dentchev
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pramod Thekkat
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Andreas Loew
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Alina C Boesteanu
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Taylor Chen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph A Fraietta
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher C Kloss
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Avery D Posey
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Boris Engels
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Reshma Singh
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Tucker Ezell
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | | | - Melissa H Ramones
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Na Li
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Li Zhou
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Gabriela Plesa
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John T Seykora
- Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hideho Okada
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Carl H June
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA. Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Marcela V Maus
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA. Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Reardon DA, Wucherpfennig KW, Freeman G, Wu CJ, Chiocca EA, Wen PY, Curry WT, Mitchell DA, Fecci PE, Sampson JH, Dranoff G. An update on vaccine therapy and other immunotherapeutic approaches for glioblastoma. Expert Rev Vaccines 2013; 12:597-615. [PMID: 23750791 DOI: 10.1586/erv.13.41] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Outcome for glioblastoma (GBM), the most common primary CNS malignancy, remains poor. The overall survival benefit recently achieved with immunotherapeutics for melanoma and prostate cancer support evaluation of immunotherapies for other challenging cancers, including GBM. Much historical dogma depicting the CNS as immunoprivileged has been replaced by data demonstrating CNS immunocompetence and active interaction with the peripheral immune system. Several glioma antigens have been identified for potential immunotherapeutic exploitation. Active immunotherapy studies for GBM, supported by preclinical data, have focused on tumor lysate and synthetic antigen vaccination strategies. Results to date confirm consistent safety, including a lack of autoimmune reactivity; however, modest efficacy and variable immunogenicity have been observed. These findings underscore the need to optimize vaccination variables and to address challenges posed by systemic and local immunosuppression inherent to GBM tumors. Additional immunotherapy strategies are also in development for GBM. Future studies may consider combinatorial immunotherapy strategies with complimentary actions.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Alexander BM, Lee EQ, Reardon DA, Wen PY. Current and future directions for Phase II trials in high-grade glioma. Expert Rev Neurother 2013; 13:369-87. [PMID: 23545053 DOI: 10.1586/ern.12.158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite surgery, radiation and chemotherapy, the prognosis for high-grade glioma (HGG) is poor. Our understanding of the molecular pathways involved in gliomagenesis and progression has increased in recent years, leading to the development of novel agents that specifically target these pathways. Results from most single-agent trials have been modest at best, however. Despite the initial success of antiangiogenesis agents in HGG, the clinical benefit is short-lived and most patients eventually progress. Several novel agents, multi-targeted agents and combination therapies are now in clinical trials for HGG and several more strategies are being pursued.
Collapse
Affiliation(s)
- Brian M Alexander
- Department of Radiation Oncology, Dana-Farber/Brigham and Women's Cancer Center, Harvard Medical School, 75 Francis Street, ASB1-L2, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
12
|
St-Coeur PD, Touaibia M, Cuperlovic-Culf M, Morin P. Leveraging metabolomics to assess the next generation of temozolomide-based therapeutic approaches for glioblastomas. GENOMICS PROTEOMICS & BIOINFORMATICS 2013; 11:199-206. [PMID: 23732626 PMCID: PMC4357826 DOI: 10.1016/j.gpb.2013.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 03/29/2013] [Accepted: 04/13/2013] [Indexed: 01/28/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common adult primary tumor of the central nervous system. The current standard of care for glioblastoma patients involves a combination of surgery, radiotherapy and chemotherapy with the alkylating agent temozolomide. Several mechanisms underlying the inherent and acquired temozolomide resistance have been identified and contribute to treatment failure. Early identification of temozolomide-resistant GBM patients and improvement of the therapeutic strategies available to treat this malignancy are of uttermost importance. This review initially looks at the molecular pathways underlying GBM formation and development with a particular emphasis placed on recent therapeutic advances made in the field. Our focus will next be directed toward the molecular mechanisms modulating temozolomide resistance in GBM patients and the strategies envisioned to circumvent this resistance. Finally, we highlight the diagnostic and prognostic value of metabolomics in cancers and assess its potential usefulness in improving the current standard of care for GBM patients.
Collapse
|
13
|
Agarwal S, Al-Keilani MS, Alqudah MAY, Sibenaller ZA, Ryken TC, Assem M. Tumor derived mutations of protein tyrosine phosphatase receptor type k affect its function and alter sensitivity to chemotherapeutics in glioma. PLoS One 2013; 8:e62852. [PMID: 23696788 PMCID: PMC3656086 DOI: 10.1371/journal.pone.0062852] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 03/26/2013] [Indexed: 11/18/2022] Open
Abstract
Poor prognosis and resistance to therapy in malignant gliomas is mainly due to the highly dispersive nature of glioma cells. This dispersive characteristic results from genetic alterations in key regulators of cell migration and diffusion. A better understanding of these regulatory signals holds promise to improve overall survival and response to therapy. Using mapping arrays to screen for genomic alterations in gliomas, we recently identified alterations of the protein tyrosine phosphatase receptor type kappa gene (PTPRK) that correlate to patient outcomes. These PTPRK alterations are very relevant to glioma biology as PTPRK can directly sense cell–cell contact and is a dephosphorylation regulator of tyrosine phosphorylation signaling, which is a major driving force behind tumor development and progression. Subsequent sequencing of the full length PTPRK transcripts revealed novel PTPRK gene deletion and missense mutations in numerous glioma biopsies. PTPRK mutations were cloned and expressed in PTPRK-null malignant glioma cells. The effect of these mutations on PTPRK anti-oncogenic function and their association with response to anti-glioma therapeutics, such as temozolomide and tyrosine kinase inhibitors, was subsequently analyzed using in vitro cell-based assays. These genetic variations altered PTPRK activity and its post-translational processing. Reconstitution of wild-type PTPRK in malignant glioma cell lines suppressed cell growth and migration by inhibiting EGFR and β-catenin signaling and improved the effect of conventional therapies for glioma. However, PTPRK mutations abrogated tumor suppressive effects of wild-type PTPRK and altered sensitivity of glioma cells to chemotherapy.
Collapse
Affiliation(s)
- Supreet Agarwal
- Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa, United States of America
| | | | | | | | | | | |
Collapse
|
14
|
Joshi AD, Loilome W, Siu IM, Tyler B, Gallia GL, Riggins GJ. Evaluation of tyrosine kinase inhibitor combinations for glioblastoma therapy. PLoS One 2012; 7:e44372. [PMID: 23056179 PMCID: PMC3462750 DOI: 10.1371/journal.pone.0044372] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Accepted: 08/06/2012] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common intracranial cancer but despite recent advances in therapy the overall survival remains about 20 months. Whole genome exon sequencing studies implicate mutations in the receptor tyrosine kinase pathways (RTK) for driving tumor growth in over 80% of GBMs. In spite of various RTKs being mutated or altered in the majority of GBMs, clinical studies have not been able to demonstrate efficacy of molecular targeted therapies using tyrosine kinase inhibitors in GBMs. Activation of multiple downstream signaling pathways has been implicated as a possible means by which inhibition of a single RTK has been ineffective in GBM. In this study, we sought a combination of approved drugs that would inhibit in vitro and in vivo growth of GBM oncospheres. A combination consisting of gefitinib and sunitinib acted synergistically in inhibiting growth of GBM oncospheres in vitro. Sunitinib was the only RTK inhibitor that could induce apoptosis in GBM cells. However, the in vivo efficacy testing of the gefitinib and sunitinib combination in an EGFR amplified/ PTEN wild type GBM xenograft model revealed that gefitinib alone could significantly improve survival in animals whereas sunitinib did not show any survival benefit. Subsequent testing of the same drug combination in a different syngeneic glioma model that lacked EGFR amplification but was more susceptible to sunitinib in vitro demonstrated no survival benefit when treated with gefitinib or sunitinib or the gefitinib and sunitinib combination. Although a modest survival benefit was obtained in one of two animal models with EGFR amplification due to gefitinib alone, the addition of sunitinib, to test our best in vitro combination therapy, did not translate to any additional in vivo benefit. Improved targeted therapies, with drug properties favorable to intracranial tumors, are likely required to form effective drug combinations for GBM.
Collapse
Affiliation(s)
- Avadhut D. Joshi
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Watcharin Loilome
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - I-Mei Siu
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Betty Tyler
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gary L. Gallia
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gregory J. Riggins
- Department of Neurosurgery, School of Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
15
|
Assem M, Sibenaller Z, Agarwal S, Al-Keilani MS, Alqudah MAY, Ryken TC. Enhancing diagnosis, prognosis, and therapeutic outcome prediction of gliomas using genomics. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2012; 16:113-22. [PMID: 22401657 DOI: 10.1089/omi.2011.0031] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Malignant gliomas are the most frequent type of primary brain tumors. Patients' outcome has not improved despite new therapeutics, thus underscoring the need for a better understanding of their genetics and a fresh approach to treatment. The lack of reproducibility in the classification of many gliomas presents an opportunity where genomics may be paramount for accurate diagnosis and therefore best for therapeutic decisions. The aim of this work is to identify large and focal copy number abnormalities (CNA) and loss of heterozygosity (LOH) events in a malignant glioma population. We hypothesized that these explorations will allow discovery of genetic markers that may improve diagnosis and predict outcome. DNA from glioma specimens were subjected to CNA and LOH analyses. Our studies revealed more than 4000 CNA and several LOH loci. Losses of chromosomes 1p and/or 19q, 10, 13, 14, and 22 and gains of 7, 19, and 20 were found. Several of these alterations correlated significantly with histology and grade. Further, LOH was detected at numerous chromosomes. Interestingly, several of these loci harbor genes with potential or reported tumor suppressor properties. These novel genetic signatures may lead to critical insights into diagnosis, classification, prognosis, and design of individualized therapies.
Collapse
Affiliation(s)
- Mahfoud Assem
- Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa 52242, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Sathornsumetee S, Rich JN. Molecularly targeted therapy in neuro-oncology. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:255-78. [PMID: 22230448 DOI: 10.1016/b978-0-444-52138-5.00018-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
17
|
Walbert T, Mikkelsen T. Recurrent high-grade glioma: a diagnostic and therapeutic challenge. Expert Rev Neurother 2011; 11:509-18. [PMID: 21469924 DOI: 10.1586/ern.11.37] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The management of recurrent high-grade gliomas with conventional, as well as targeted, therapies is problematic owing to several confounding issues. First, the diagnosis of recurrence using MRI is not straightforward, making the assessment of images in daily routines, as well as in clinical trials, challenging. While chemotherapies with cytotoxic agents have demonstrated initial treatment response, most tumors recur quickly. Second, targeted therapy itself is confounded by the heterogeneous expression of drug targets and nonlinear signaling effects, with functional redundancy and sidestream feedback mechanisms resulting in treatment failure; however, several active agents have been identified, most notably, bevacizumab (an antibody that sequesters VEGF), cilengitide (an inhibitor of integrin αvβ3/5 signaling) and cediranib (an oral tyrosine kinase inhibitor targeting PDGF receptor, c-Kit and all VEGF receptor subtypes). All of these agents have undergone multiple clinical trials and have demonstrated benefits and progression-free survival prolongation in recurrent disease. Given these advances, it is likely that tailored therapies for tumors harboring specific signaling defects will become more efficient and successful in the management of glioblastoma.
Collapse
Affiliation(s)
- Tobias Walbert
- Department of Neurosurgery, Hermelin Brain Tumor Center, Henry Ford Health System, 2799 W Grand Blvd, Detroit, MI 48202, USA
| | | |
Collapse
|
18
|
Plano D, Ibáñez E, Calvo A, Palop JA, Sanmartín C. Novel library of selenocompounds as kinase modulators. Molecules 2011; 16:6349-64. [PMID: 21796074 PMCID: PMC6264252 DOI: 10.3390/molecules16086349] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 07/20/2011] [Accepted: 07/22/2011] [Indexed: 12/30/2022] Open
Abstract
Although the causes of cancer lie in mutations or epigenic changes at the genetic level, their molecular manifestation is the dysfunction of biochemical pathways at the protein level. The 518 protein kinases encoded by the human genome play a central role in various diseases, a fact that has encouraged extensive investigations on their biological function and three dimensional structures. Selenium (Se) is an important nutritional trace element involved in different physiological functions with antioxidative, antitumoral and chemopreventive properties. The mechanisms of action for selenocompounds as anticancer agents are not fully understood, but kinase modulation seems to be a possible pathway. Various organosulfur compounds have shown antitumoral and kinase inhibition effects but, in many cases, the replacement of sulfur by selenium improves the antitumoral effect of compounds. Although Se atom possesses a larger atomic volume and nucleophilic character than sulfur, Se can also formed interactions with aminoacids of the catalytic centers of proteins. So, we propose a novel chemical library that includes organoselenium compounds as kinase modulators. In this study thirteen selenocompounds have been evaluated at a concentration of 3 or 10 µM in a 24 kinase panel using a Caliper LabChip 3000 Drug Discover Platform. Several receptor (EGFR, IGFR1, FGFR1…) and non-receptor (Abl) kinases have been selected, as well as serine/threonine/lipid kinases (AurA, Akt, CDKs, MAPKs…) implicated in main cancer pathways: cell cycle regulation, signal transduction, angiogenesis regulation among them. The obtained results showed that two compounds presented inhibition values higher than 50% in at least four kinases and seven derivatives selectively inhibited one or two kinases. Furthermore, three compounds selectively activated IGF-1R kinase with values ranging from −98% to −211%. In conclusion, we propose that the replacement of sulfur by selenium seems to be a potential and useful strategy in the search of novel chemical compound libraries against cancer as kinase modulators.
Collapse
Affiliation(s)
- Daniel Plano
- Department of Organic and Pharmaceutical Chemistry, University of Navarra, Irunlarrea, 1, Pamplona E-31008, Spain
| | - Elena Ibáñez
- Department of Organic and Pharmaceutical Chemistry, University of Navarra, Irunlarrea, 1, Pamplona E-31008, Spain
| | - Alfonso Calvo
- Oncology Division, Center for Applied Medical Research, CIMA, University of Navarra, Pío XII, 53, Pamplona E-31008, Spain
| | - Juan Antonio Palop
- Department of Organic and Pharmaceutical Chemistry, University of Navarra, Irunlarrea, 1, Pamplona E-31008, Spain
| | - Carmen Sanmartín
- Department of Organic and Pharmaceutical Chemistry, University of Navarra, Irunlarrea, 1, Pamplona E-31008, Spain
- Author to whom correspondence should be addressed;
| |
Collapse
|
19
|
Dorin-Semblat D, Schmitt S, Semblat JP, Sicard A, Reininger L, Goldring D, Patterson S, Quashie N, Chakrabarti D, Meijer L, Doerig C. Plasmodium falciparum NIMA-related kinase Pfnek-1: sex specificity and assessment of essentiality for the erythrocytic asexual cycle. MICROBIOLOGY-SGM 2011; 157:2785-2794. [PMID: 21757488 PMCID: PMC3353393 DOI: 10.1099/mic.0.049023-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The Plasmodium falciparum kinome includes a family of four protein kinases (Pfnek-1 to -4) related to the NIMA (never-in-mitosis) family, members of which play important roles in mitosis and meiosis in eukaryotic cells. Only one of these, Pfnek-1, which we previously characterized at the biochemical level, is expressed in asexual parasites. The other three (Pfnek-2, -3 and -4) are expressed predominantly in gametocytes, and a role for nek-2 and nek-4 in meiosis has been documented. Here we show by reverse genetics that Pfnek-1 is required for completion of the asexual cycle in red blood cells and that its expression in gametocytes in detectable by immunofluorescence in male (but not in female) gametocytes, in contrast with Pfnek-2 and Pfnek-4. This indicates that the function of Pfnek-1 is non-redundant with those of the other members of the Pfnek family and identifies Pfnek-1 as a potential target for antimalarial chemotherapy. A medium-throughput screen of a small-molecule library provides proof of concept that recombinant Pfnek-1 can be used as a target in drug discovery.
Collapse
Affiliation(s)
- Dominique Dorin-Semblat
- INSERM U609, Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK.,INSERM-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, GHI-SV-EPFL Station 19, CH-1015 Lausanne, Switzerland
| | - Sophie Schmitt
- Cell Cycle Group, C.N.R.S., Station Biologique, 29680 Roscoff, Bretagne, France
| | - Jean-Philippe Semblat
- INSERM U609, Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK.,INSERM-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, GHI-SV-EPFL Station 19, CH-1015 Lausanne, Switzerland
| | - Audrey Sicard
- INSERM U609, Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK.,INSERM-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, GHI-SV-EPFL Station 19, CH-1015 Lausanne, Switzerland
| | - Luc Reininger
- INSERM U609, Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK.,INSERM-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, GHI-SV-EPFL Station 19, CH-1015 Lausanne, Switzerland
| | - Dean Goldring
- Biochemistry, University of Kwazulu-Natal, PB X01 Scottsville, South Africa
| | - Shelley Patterson
- Department of Molecular Biology and Microbiology, University of Central Florida, 12722 Research Parkway, Orlando, FL 32826, USA
| | - Neils Quashie
- Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Debopam Chakrabarti
- Department of Molecular Biology and Microbiology, University of Central Florida, 12722 Research Parkway, Orlando, FL 32826, USA
| | - Laurent Meijer
- Cell Cycle Group, C.N.R.S., Station Biologique, 29680 Roscoff, Bretagne, France
| | - Christian Doerig
- INSERM U609, Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK.,INSERM-EPFL Joint Laboratory, Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, GHI-SV-EPFL Station 19, CH-1015 Lausanne, Switzerland
| |
Collapse
|
20
|
Reardon DA, Perry JR, Brandes AA, Jalali R, Wick W. Advances in malignant glioma drug discovery. Expert Opin Drug Discov 2011; 6:739-53. [DOI: 10.1517/17460441.2011.584530] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
21
|
Luan X, Gao C, Zhang N, Chen Y, Sun Q, Tan C, Liu H, Jin Y, Jiang Y. Exploration of acridine scaffold as a potentially interesting scaffold for discovering novel multi-target VEGFR-2 and Src kinase inhibitors. Bioorg Med Chem 2011; 19:3312-9. [PMID: 21576023 DOI: 10.1016/j.bmc.2011.04.053] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 04/27/2011] [Accepted: 04/28/2011] [Indexed: 01/16/2023]
Abstract
VEGFR-2 and Src kinases both play important roles in cancers. In certain cancers, Src works synergistically with VEGFR-2 to promote its activation. Development of multi-target drugs against VEGFR-2 and Src is of therapeutic advantage against these cancers. By using molecular docking and SVM virtual screening methods and based on subsequent synthesis and bioassay studies, we identified 9-aminoacridine derivatives with an acridine scaffold as potentially interesting novel dual VEGFR-2 and Src inhibitors. The acridine scaffold has been historically used for deriving topoisomerase inhibitors, but has not been found in existing VEGFR-2 inhibitors and Src inhibitors. A series of 21 acridine derivatives were synthesized and evaluated for their antiproliferative activities against K562, HepG-2, and MCF-7 cells. Some of these compounds showed better activities against K562 cells in vitro than imatinib. The structure-activity relationships (SAR) of these compounds were analyzed. One of the compounds (7r) showed low μM activity against K562 and HepG-2 cancer cell-lines, and inhibited VEGFR-2 and Src at inhibition rates of 44% and 8% at 50μM, respectively, without inhibition of topoisomerase. Moreover, 10μM compound 7r could reduce the levels of activated ERK1/2 in a time dependant manner, a downstream effector of both VEGFR-2 and Src. Our study suggested that acridine scaffold is a potentially interesting scaffold for developing novel multi-target kinase inhibitors such as VEGFR-2 and Src dual inhibitors.
Collapse
Affiliation(s)
- Xudong Luan
- The Guangdong Province Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhang C, Tan C, Zu X, Zhai X, Liu F, Chu B, Ma X, Chen Y, Gong P, Jiang Y. Exploration of (S)-3-aminopyrrolidine as a potentially interesting scaffold for discovery of novel Abl and PI3K dual inhibitors. Eur J Med Chem 2011; 46:1404-14. [DOI: 10.1016/j.ejmech.2011.01.020] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 12/29/2010] [Accepted: 01/12/2011] [Indexed: 01/04/2023]
|
23
|
Mellinghoff IK, Lassman AB, Wen PY. Signal transduction inhibitors and antiangiogenic therapies for malignant glioma. Glia 2011; 59:1205-12. [PMID: 21351155 DOI: 10.1002/glia.21137] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 12/08/2010] [Indexed: 11/09/2022]
Abstract
Detailed characterization of the cancer genome in a large number of primary human glioblastomas has identified recurrent alterations that result in deregulation of signal transduction pathways and are "druggable" with a growing number of small molecule pharmaceuticals. While many of these compounds have shown clinical activity in other human cancers harboring similar genetic alterations, the clinical experience in glioblastoma has been disappointing thus far with only rare and transient radiographic responses. Our understanding of drug resistance is confounded by the uncertainty of drug delivery across the blood brain barrier and the limited knowledge to what extent the growth of these tumors depends on any particular signaling pathway. This uncertainty is, at least in part, due to shortcomings in the current approach to evaluate signal transduction inhibitors in glioma patients, including drug testing in molecularly unselected patient populations, limited documentation of drug penetration and target inhibition in tumor tissue, and use of radiographic response criteria that may not be optimal for the evaluation of these compounds. © 2011 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Ingo K Mellinghoff
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, USA.
| | | | | |
Collapse
|
24
|
Jin F, Gao C, Zhao L, Zhang H, Wang HT, Shao T, Zhang SL, Wei YJ, Jiang XB, Zhou YP, Zhao HY. Using CD133 positive U251 glioblastoma stem cells to establish nude mice model of transplanted tumor. Brain Res 2011; 1368:82-90. [DOI: 10.1016/j.brainres.2010.10.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/12/2010] [Accepted: 10/14/2010] [Indexed: 02/06/2023]
|
25
|
Abstract
IMPORTANCE OF THE FIELD Despite numerous advances made during the last decade in brain tumor therapy, the prognosis of glioblastoma has not improved and these tumors inevitably recur with no effective treatment. Thus, any new therapeutic strategy to target this most malignant tumor will be of significant benefit. RNAi is a powerful gene silencing method that might be used in combination with other agents to improve the efficacy of glioblastoma treatment. AREAS COVERED IN THIS REVIEW Recent progress and challenges of pre-clinical and clinical research of RNAi therapy for glioblastoma. The review covers literature from 2003 to 2009. WHAT THE READER WILL GAIN The principle of RNA interference therapy, three categories of RNAi triggers, different RNAi delivery system and pre-clinical and clinical studies that are currently underway to evaluate the validity of RNAi as a potential therapeutic strategy against glioblastoma are discussed. TAKE HOME MESSAGE RNA inference therapy combined with other therapeutics may offer therapeutic potential for glioblastoma multiforme. Further studies are required to develop more efficient and specific delivery systems, select suitable gene targets, optimize treatment dose and administration schedule, evaluate the efficacy of combination treatment strategies, establish a validated clinical response measure system etc.
Collapse
Affiliation(s)
- Dongsheng Guo
- Department of Neurosurgery, Huazhong University of Sciences and Technology, Tongji Medical College, Tongji Hospital, Wuhan, China.
| | | | | | | |
Collapse
|
26
|
Morphy R. Selectively nonselective kinase inhibition: striking the right balance. J Med Chem 2010; 53:1413-37. [PMID: 20166671 DOI: 10.1021/jm901132v] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Richard Morphy
- Medicinal Chemistry Department, Schering-Plough, Newhouse, Lanarkshire, ML1 5SH, UK.
| |
Collapse
|
27
|
Ma XH, Shi Z, Tan C, Jiang Y, Go ML, Low BC, Chen YZ. In-silico approaches to multi-target drug discovery : computer aided multi-target drug design, multi-target virtual screening. Pharm Res 2010; 27:739-49. [PMID: 20221898 DOI: 10.1007/s11095-010-0065-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 01/08/2010] [Indexed: 01/25/2023]
Abstract
Multi-target drugs against selective multiple targets improve therapeutic efficacy, safety and resistance profiles by collective regulations of a primary therapeutic target together with compensatory elements and resistance activities. Efforts have been made to employ in-silico methods for facilitating the search and design of selective multi-target agents. These methods have shown promising potential in facilitating drug discovery directed at selective multiple targets.
Collapse
Affiliation(s)
- Xiao Hua Ma
- Bioinformatics and Drug Design Group, Department of Pharmacy, Centre for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, Singapore, 117543, Singapore
| | | | | | | | | | | | | |
Collapse
|
28
|
Thaker NG, Zhang F, McDonald PR, Shun TY, Lazo JS, Pollack IF. Functional genomic analysis of glioblastoma multiforme through short interfering RNA screening: a paradigm for therapeutic development. Neurosurg Focus 2010; 28:E4. [PMID: 20043719 DOI: 10.3171/2009.10.focus09210] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Glioblastoma multiforme (GBM) is a high-grade brain malignancy arising from astrocytes. Despite aggressive surgical approaches, optimized radiation therapy regimens, and the application of cytotoxic chemotherapies, the median survival of patients with GBM from time of diagnosis remains less than 15 months, having changed little in decades. Approaches that target genes and biological pathways responsible for tumorigenesis or potentiate the activity of current therapeutic modalities could improve treatment efficacy. In this regard, several genomic and proteomic strategies promise to impact significantly on the drug discovery process. High-throughput genome-wide screening with short interfering RNA (siRNA) is one strategy for systematically exploring possible therapeutically relevant targets in GBM. Statistical methods and protein-protein interaction network databases can also be applied to the screening data to explore the genes and pathways that underlie the pathological basis and development of GBM. In this study, we highlight several genome-wide siRNA screens and implement these experimental concepts in the T98G GBM cell line to uncover the genes and pathways that regulate GBM cell death and survival. These studies will ultimately influence the development of a new avenue of neurosurgical therapy by placing the drug discovery process in the context of the entire biological system.
Collapse
Affiliation(s)
- Nikhil G Thaker
- University of Medicine and Dentistry of New Jersey-New Jersey Medical School, Newark, New Jersey, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
Protein phosphorylation plays key roles in the regulation of normal and cancer cells. It is a highly dynamic process. Protein kinases are the targets of several new cancer drugs and drug candidates. However, some of the main issues related to new drugs are how they function and the selection of those patients that will likely respond best to a particular treatment regime. There is an urgent need to understand and monitor kinase signalling pathways. Phosphoproteomics requires the enrichment of phosphorylated proteins or peptides from tissue or bodily fluids, and the application of technologies such as mass spectrometry (MS) to the identification and quantification of protein phosphorylation sites. As the field develops it will provide pharmacodynamic readouts of disease states and cellular drug responses in tumour samples. There have been a number of recent advances, but there are still technical hurdles and bioinformatics challenges that need to be addressed.
Collapse
Affiliation(s)
- Keith Ashman
- Biotechnology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), ES-28029 Madrid, Spain.
| | | |
Collapse
|
30
|
|
31
|
Grzmil M, Hemmings BA. Deregulated signalling networks in human brain tumours. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2009; 1804:476-83. [PMID: 19879382 DOI: 10.1016/j.bbapap.2009.10.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 10/13/2009] [Accepted: 10/21/2009] [Indexed: 01/17/2023]
Abstract
Despite the variety of modern therapies against human brain cancer, in its most aggressive form of glioblastoma multiforme (GBM) it is a still deadly disease with a median survival of approximately 1 year. Over the past 2 decades, molecular profiling of low- and high-grade malignant brain tumours has led to the identification and molecular characterisation of mechanisms leading to brain cancer development, maintenance and progression. Genetic alterations occurring during gliomagenesis lead to uncontrolled tumour growth stimulated by deregulated signal transduction pathways. The characterisation of hyperactivated signalling pathways has identified many potential molecular targets for therapeutic interference in human gliomas. Overexpressed or mutated and constitutively active kinases are attractive targets for low-molecular-weight inhibitors. Although the first attempts with mono-therapy using a single targeted kinase inhibitor were not satisfactory, recent studies based on the simultaneous targeting of several core hyperactivated pathways show great promise for the development of novel therapeutic approaches. This review focuses on genetic alterations leading to the activation of key deregulated pathways in human gliomas.
Collapse
Affiliation(s)
- Michal Grzmil
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH-4058 Basel, Switzerland.
| | | |
Collapse
|