1
|
Lichtor T, Tang B, Roy EJ. Cytokine Gene Vaccine Therapy for Treatment of a Brain Tumor. Brain Sci 2023; 13:1505. [PMID: 38002466 PMCID: PMC10669932 DOI: 10.3390/brainsci13111505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
A glioma is a malignant brain tumor with a poor prognosis. Attempts at the surgical removal of the tumor are the first approach, but additional treatment strategies, including radiation therapy and systemic or local chemotherapy, are necessary. Furthermore, the treatments are often associated with significant adverse side effects. Normal and malignant cells generally have antigenic differences, and this is the rationale for clinical immunotherapeutic strategies. Cytokines such as IL-15 or IL-2, which stimulate an anti-tumor immune response, have been shown to have a particularly high potential for use in immunotherapy against various tumors. In this review, treatments with either a poxvirus, genetically engineered to secrete IL-15, or allogeneic fibroblasts, transfected with tumor DNA and engineered to secrete IL-2, are shown to be effective strategies in extending the survival of mice with malignant brain tumors upon intracerebral injection of the treatment cells. Future studies with these treatment strategies in patients with intracerebral tumors are urgently needed.
Collapse
Affiliation(s)
- Terry Lichtor
- Department of Neurological Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Bingtao Tang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (B.T.); (E.J.R.)
| | - Edward J. Roy
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (B.T.); (E.J.R.)
| |
Collapse
|
2
|
Xu Q, Zhai JC, Huo CQ, Li Y, Dong XJ, Li DF, Huang RD, Shen C, Chang YJ, Zeng XL, Meng FL, Yang F, Zhang WL, Zhang SN, Zhou YM, Zhang Z. OncoPDSS: an evidence-based clinical decision support system for oncology pharmacotherapy at the individual level. BMC Cancer 2020; 20:740. [PMID: 32770988 PMCID: PMC7414679 DOI: 10.1186/s12885-020-07221-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/27/2020] [Indexed: 12/17/2022] Open
Abstract
Background Precision oncology pharmacotherapy relies on precise patient-specific alterations that impact drug responses. Due to rapid advances in clinical tumor sequencing, an urgent need exists for a clinical support tool that automatically interprets sequencing results based on a structured knowledge base of alteration events associated with clinical implications. Results Here, we introduced the Oncology Pharmacotherapy Decision Support System (OncoPDSS), a web server that systematically annotates the effects of alterations on drug responses. The platform integrates actionable evidence from several well-known resources, distills drug indications from anti-cancer drug labels, and extracts cancer clinical trial data from the ClinicalTrials.gov database. A therapy-centric classification strategy was used to identify potentially effective and non-effective pharmacotherapies from user-uploaded alterations of multi-omics based on integrative evidence. For each potentially effective therapy, clinical trials with faculty information were listed to help patients and their health care providers find the most suitable one. Conclusions OncoPDSS can serve as both an integrative knowledge base on cancer precision medicine, as well as a clinical decision support system for cancer researchers and clinical oncologists. It receives multi-omics alterations as input and interprets them into pharmacotherapy-centered information, thus helping clinicians to make clinical pharmacotherapy decisions. The OncoPDSS web server is freely accessible at https://oncopdss.capitalbiobigdata.com.
Collapse
Affiliation(s)
- Quan Xu
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Jin-Cheng Zhai
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Cai-Qin Huo
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Yang Li
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Xue-Jiao Dong
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Dong-Fang Li
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Ru-Dan Huang
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Chuang Shen
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Yu-Jun Chang
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Xi-Ling Zeng
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Fan-Lin Meng
- School of Medicine, Tsinghua University, Beijing, 100084, P.R. China
| | - Fang Yang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, P.R. China
| | - Wan-Ling Zhang
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Sheng-Nan Zhang
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Yi-Ming Zhou
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China.,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China
| | - Zhi Zhang
- National Engineering Research Center for Beijing Biochip Technology, Changping District, Beijing, 102206, P.R. China. .,CapitalBio Corporation, Changping District, Beijing, 102206, P.R. China.
| |
Collapse
|
3
|
Munegowda MA, Fisher C, Molehuis D, Foltz W, Roufaiel M, Bassan J, Nitz M, Mandel A, Lilge L. Efficacy of ruthenium coordination complex-based Rutherrin in a preclinical rat glioblastoma model. Neurooncol Adv 2019; 1:vdz006. [PMID: 32642649 PMCID: PMC7212850 DOI: 10.1093/noajnl/vdz006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Background Glioblastoma is an aggressive brain cancer in adults with a grave prognosis, aggressive radio and chemotherapy provide only a 15 months median survival. Methods We evaluated the tolerability and efficacy of the Ruthenium-based photosensitizer TLD-1433 with apo-Transferrin (Rutherrin) in the rat glioma 2 (RG-2) model. The specific tumor uptake ratio and photodynamic therapy (PDT) threshold of the rat glioblastoma and normal brain were determined, survival and CD8+T-cell infiltration post-therapy were analyzed. Results were compared with those obtained for 5-aminolevulinic acid (ALA)-induced Protoporphyrin IX (PpIX)-mediated photodynamic therapy in the same animal model. As both photosensitizers have different photophysical properties, the number of absorbed photons required to achieve an equal cell kill was determined for in vitro and in vivo studies. Results A significantly lower absorbed energy was sufficient to achieve LD50 with Rutherrin versus PpIX-mediated PDT. Rutherrin provides a higher specific uptake ratio (SUR) >20 in tumors versus normal brain, whereas the SUR for ALA-induced PpIX was 10.6. To evaluate the short-term tissue response in vivo, enhanced T2-weighted magnetic resonance imaging (MRI) provided the spatial extent of edema, post PpIX-PDT at twice the cross-section versus Rutherrin-PDT suggesting reduced nonspecific damage, typically associated with a secondary wave of neuronal damage. Following a single therapy, a significant survival increase was observed in rats bearing glioma for PDT mediated by Rutherrin versus PpIX for the selected treatment conditions. Rutherrin-PDT also demonstrated an increased CD8+T-cell infiltration in the tumors. Conclusion Rutherrin-PDT was well tolerated providing a safe and effective treatment of RG-2 glioma.
Collapse
Affiliation(s)
| | - Carl Fisher
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Daniel Molehuis
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Warren Foltz
- Techna Institute, University Health Network, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Mark Roufaiel
- Theralase Technologies Inc., Toronto, Ontario, Canada
| | - Jay Bassan
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Arkady Mandel
- Theralase Technologies Inc., Toronto, Ontario, Canada
| | - Lothar Lilge
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Deshpande A, Layek RK. Fault detection and therapeutic intervention in gene regulatory networks using SAT solvers. Biosystems 2019; 179:55-62. [PMID: 30831179 DOI: 10.1016/j.biosystems.2019.02.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 11/04/2018] [Accepted: 02/28/2019] [Indexed: 11/19/2022]
Abstract
Random somatic mutations disrupt homeostasis of the cell resulting in various undesirable phenotypes including proliferation. One of the most important questions in systems medicine research is the therapeutic intervention design, which requires the knowledge of these mutations. A single or multiple mutations can occur in the diseases like cancer. These mutations have been successfully modeled as stuck-at faults in the Boolean network model of the underlying regulatory system. Identification of these fault types for multiple stuck-at faults is a non-trivial problem and requires some system theoretic introspection. This manuscript addresses the dual problem of the fault identification and the therapeutic intervention. Both the problems are mapped to the Boolean satisfiability (SAT) problem. The underlying problems are solved using a fast SAT solver. The synthetic and biological examples elucidate the effectiveness of the mapping.
Collapse
|
5
|
Ghosh D, Nandi S, Bhattacharjee S. Combination therapy to checkmate Glioblastoma: clinical challenges and advances. Clin Transl Med 2018; 7:33. [PMID: 30327965 PMCID: PMC6191404 DOI: 10.1186/s40169-018-0211-8] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/01/2018] [Indexed: 12/11/2022] Open
Abstract
Combination therapy is increasingly becoming the cornerstone of current day antitumor therapy. Glioblastoma multiforme is an aggressive brain tumor with a dismal median survival post diagnosis and a high rate of disease recurrence. The poor prognosis can be attributed to unique treatment limitations, which include the infiltrative nature of tumor cells, failure of anti-glioma drugs to cross the blood-brain barrier, tumor heterogeneity and the highly metastatic and angiogenic nature of the tumor making cells resistant to chemotherapy. Combination therapy approach is being developed against glioblastoma with new innovative combination drug regimens being tested in preclinical and clinical trials. In this review, we discuss the pathophysiology of glioblastoma, diagnostic markers, therapeutic targeting strategies, current treatment limitations, novel combination therapies in the context of current treatment options and the ongoing clinical trials for glioblastoma therapy.
Collapse
Affiliation(s)
- Debarati Ghosh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Saikat Nandi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| | | |
Collapse
|
6
|
Morisse MC, Jouannet S, Dominguez-Villar M, Sanson M, Idbaih A. Interactions between tumor-associated macrophages and tumor cells in glioblastoma: unraveling promising targeted therapies. Expert Rev Neurother 2018; 18:729-737. [PMID: 30099909 DOI: 10.1080/14737175.2018.1510321] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the deadliest primary malignant central nervous system (CNS) tumor with a median overall survival of 15 months despite a very intensive therapeutic regimen including maximal safe surgery, radiotherapy, and chemotherapy. Therefore, GBM treatment still raises major biological and therapeutic challenges. Areas covered: One of the hallmarks of the GBM is its tumor microenvironment including tumor-associated macrophages (TAM). TAM, accounting for approximately 30% of the GBM bulk cell population, may explain, at least in part, the immunosuppressive features of GBMs. The TAM are active and highly plastic immune cells and include two major ontogenetically different cell populations: (i) microglia and, (ii) monocytes-derived macrophages (MDM). TAM recruited to the tumor bulk can be reprogramed by GBM cells resulting in an ineffective anti-tumor response. Interestingly, interactions between TAM and GBM cells promote tumor oncogenesis (i.e. tumor cells proliferation and migration/invasion). This review aims to explore TAM targeting in GBM as a promising therapeutic option in the near future. Expert Commentary: A better understanding of TAM-GBM interactions and dynamics will certainly uncover new anti-GBM therapeutic avenues.
Collapse
Affiliation(s)
- Mony Chenda Morisse
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France.,b Department of Medical Oncology , CHU Sud , Amiens , France
| | - Stéphanie Jouannet
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France
| | | | - Marc Sanson
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France
| | - Ahmed Idbaih
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France
| |
Collapse
|
7
|
Jan CI, Tsai WC, Harn HJ, Shyu WC, Liu MC, Lu HM, Chiu SC, Cho DY. Predictors of Response to Autologous Dendritic Cell Therapy in Glioblastoma Multiforme. Front Immunol 2018; 9:727. [PMID: 29910795 PMCID: PMC5992384 DOI: 10.3389/fimmu.2018.00727] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/23/2018] [Indexed: 12/13/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common and lethal primary malignant glioma in adults. Dendritic cell (DC) vaccines have demonstrated promising results in GBM clinical trials. However, some patients do not respond well to DC therapy, with survival rates similar to those of conventional therapy. We retrospectively analyzed clinical and laboratory data to evaluate the factors affecting vaccine treatment. Methods Forty-seven patients with de novo GBM were enrolled at China Medical University Hospital between 2005 and 2010 and divided into two subgroups. One subgroup of 27 patients received postsurgical adjuvant immunotherapy with autologous dendritic cell/tumor antigen vaccine (ADCTA) in conjunction with conventional treatment of concomitant chemoradiotherapy (CCRT) with temozolomide. The other 20 patients received only postsurgical conventional treatment without immunotherapy. Immunohistochemistry for CD45, CD4, CD8, programed death ligand 1 (PD-L1), and programed death 1 (PD-1) was performed on sections of surgical tumor specimens and peripheral blood mononuclear cells (PBMCs). Pearson's correlation, Cox proportional hazard model, and Kaplan-Meier analyses were performed to examine the correlations between the prognostic factors and survival rates. Results Younger age (<57 years), gross total resection, and CCRT and PD-1+ lymphocyte counts were significant prognostic factors of overall survival (OS) and progression-free survival (PFS) in the ADCTA group. Sex, CD45+ lymphocyte count, CD4+ or CD8+ lymphocyte count, tumor PD-L1 expression, isocitrate dehydrogenase 1 mutation, and O6 methylguanine-DNA methyltransferase promoter methylation status were not significant factors in both groups. In the ADCTA group, patients with tumor-infiltrating lymphocytes (TILs) with a lower PD-1+/CD8+ ratio (≤0.21) had longer OS and PFS (median OS 60.97 months, P < 0.001 and PFS 11.2 months, P < 0.008) compared to those with higher PD-1+/CD8+ ratio (>0.21) (median OS 20.07 months, P < 0.001 and PFS 4.43 months, P < 0.008). Similar results were observed in patients' PBMCs; lymphocyte counts with lower PD-1+/CD8+ ratio (≤0.197) had longer OS and PFS. There was a significant correlation of PD-1+/CD8+ ratio between TILs and PBMCs (Pearson's correlation R2 = 0.6002, P < 0.001). By contrast, CD4-, CD8-, but PD-1+, CD45+ tumor-infiltrating lymphocytes have no impact on OS and PFS (P = 0.073 and P = 0.249, respectively). Conclusion For patients receiving DC vaccine adjuvant therapy, better outcomes are predicted in patients with younger age, with TILs or PBMCs with lower PD-1+/CD8+ ratio, with gross tumor resection, and receiving CCRT.
Collapse
Affiliation(s)
- Chia-Ing Jan
- Division of Molecular Pathology, Department of Pathology, China Medical University and Hospital, Taichung, Taiwan.,Department of Pathology, China Medical University and Beigang Hospital, Yunlin, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wan-Chen Tsai
- Center for Cell Therapy, China Medical University Hospital, Taichung, Taiwan
| | - Horng-Jyh Harn
- The Buddhist Tzu Chi Bioinnovation Center, Buddhist Tzu Chi University, Haualien, Taiwan.,Department of Pathology, Buddhist Tzu Chi General Hospital and Buddhist Tzu Chi University Haualien, Haualien, Taiwan
| | - Woei-Cherng Shyu
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan.,Center for Neuropsychiatry, Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Chao Liu
- Center for Cell Therapy, China Medical University Hospital, Taichung, Taiwan.,Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Hsin-Man Lu
- Department of Psychology, Asia University, Taichung, Taiwan
| | - Shao-Chih Chiu
- Center for Cell Therapy, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Der-Yang Cho
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Graduate Institute of Immunology China Medical University, Taichung, Taiwan.,Department of Neurosurgery, Neuropsychiatric Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Immunotherapy has emerged as a cornerstone of modern oncology with regulatory approvals for a variety of immunotherapeutics being achieved for a spectrum of cancer indications. Nonetheless the role of these approaches for patients with glioblastoma (GBM), the most common and deadliest primary malignant brain neoplasm, remains unknown. In this review, we summarize the current status of clinical development for the major types of immunotherapeutics, including vaccines, cell-based therapies, and immune checkpoint modulators for GBM. We also highlight potential challenges confronting the development of these agents. RECENT FINDINGS Growing preclinical and clinical data is emerging regarding the potential of immunotherapy strategies for GBM. In parallel, growing data demonstrating that historical dogma classifying the brain as immunoprivileged is inaccurate but that many tumors, including GBM evoke myriad mechanisms to suppress antitumor immune responses. SUMMARY Ongoing initial trials will provide preliminary data on the role of immunotherapy for GBM patients. Subsequent clinical development steps will likely require rationally designed combinatorial regimens.
Collapse
|
9
|
Marisetty AL, Singh SK, Nguyen TN, Coarfa C, Liu B, Majumder S. REST represses miR-124 and miR-203 to regulate distinct oncogenic properties of glioblastoma stem cells. Neuro Oncol 2017; 19:514-523. [PMID: 28040710 DOI: 10.1093/neuonc/now232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Glioblastoma (GBM) is one of the most common, aggressive, and invasive human brain tumors. There are few reliable mechanism-based therapeutic approaches for GBM patients. The transcriptional repressor RE1 silencing transcriptional factor (REST) regulates the oncogenic properties of a class of GBM stem-like cells (high-REST [HR]-GSCs) in humans. However, it has been unclear whether REST represses specific targets to regulate specific oncogenic functions or represses all targets with overlapping functions in GSCs. Methods We used genome-wide, biochemical, and mouse intracranial tumorigenic assays to identify and determine functions of microRNA (miR) targets of REST in 2 independent HR-GSC lines. Results Here we show that REST represses 2 major miR gene targets in HR-GSCs: miR-203, a new target, and miR-124, a known target. Gain of function of miR-124 or miR-203 in HR-GSCs increased survival in tumor-bearing mice. Importantly, the increased survival of tumor-bearing mice caused by knockdown of REST in HR-GSCs was reversed by double knockdown of REST and either miR-203 or miR-124, indicating that these 2 miRs are critical tumor suppressors that are repressed in REST-mediated tumorigenesis. We further show that while miR-124 and the REST-miR-124 pathways regulate self-renewal, apoptosis and invasion, miR-203 and the REST-miR-203 pathways regulate only invasion. We further identify and validate potential mRNA targets of miR-203 and miR-124 in REST-mediated HR-GSC tumor invasion. Conclusions These findings indicate that REST regulates its miR gene targets with overlapping functions and suggest how REST maintains oncogenic competence in GSCs. These mechanisms could potentially be utilized to block REST-mediated GBM tumorigenesis.
Collapse
Affiliation(s)
- Anantha L Marisetty
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Sanjay K Singh
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tran N Nguyen
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Bin Liu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sadhan Majumder
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA.,Neuro-Oncology, The Brain Tumor Center, The University of Texas M. D. Anderson Cancer Center, University of Texas, Houston, Texas, USA
| |
Collapse
|
10
|
Reconfigurable Microfluidic Magnetic Valve Arrays: Towards a Radiotherapy-Compatible Spheroid Culture Platform for the Combinatorial Screening of Cancer Therapies. SENSORS 2017; 17:s17102271. [PMID: 28976942 PMCID: PMC5677148 DOI: 10.3390/s17102271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/08/2023]
Abstract
We introduce here a microfluidic cell culture platform or spheroid culture chamber array (SCCA) that can synthesize, culture, and enable fluorescence imaging of 3D cell aggregates (typically spheroids) directly on-chip while specifying the flow of reagents in each chamber via the use of an array of passive magnetic valves. The SCCA valves demonstrated sufficient resistance to burst (above 100 mBar), including after receiving radiotherapy (RT) doses of up to 8 Gy combined with standard 37 °C incubation for up to 7 days, enabling the simultaneous synthesis of multiple spheroids from different cell lines on the same array. Our results suggest that SCCA would be an asset in drug discovery processes, seeking to identify combinatorial treatments.
Collapse
|
11
|
Zhai L, Ladomersky E, Dostal CR, Lauing K, Swoap K, Billingham LK, Gritsina G, Wu M, McCusker RH, Binder DC, Wainwright DA. Non-tumor cell IDO1 predominantly contributes to enzyme activity and response to CTLA-4/PD-L1 inhibition in mouse glioblastoma. Brain Behav Immun 2017; 62:24-29. [PMID: 28179106 PMCID: PMC5514839 DOI: 10.1016/j.bbi.2017.01.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/28/2016] [Accepted: 01/28/2017] [Indexed: 01/09/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults with a median survival of 14.6months. A contributing factor to GBM aggressiveness is the intratumoral expression of the potently immunosuppressive enzyme, indoleamine 2,3 dioxygenase 1 (IDO1). The enzymatic activity of IDO1 is associated with the conversion of tryptophan into downstream kynurenine (Kyn), which has previously been hypothesized to contribute toward the suppression of tumor immunity. Utilizing the syngeneic, immunocompetent, intracranial GL261 cell GBM model, we previously demonstrated that tumor cell, but not non-tumor cell IDO1, suppresses T cell-mediated brain tumor regression in mice. Paradoxically, we also showed that the survival advantage mediated by immune checkpoint blockade is abrogated by non-tumor cell IDO1 deficiency. Here, we have built on our past observations and confirm the maladaptive role of tumor cell IDO1 in a novel mouse GBM model. We also demonstrate that, non-tumor cells, rather than mouse GBM cells, are the dominant contributor to IDO1-mediated enzyme activity. Finally, we show the novel associations between maximally-effective immune-checkpoint blockade-mediated survival, non-tumor cell IDO1 and intra-GBM Kyn levels. These data suggest for the first time that, GBM cell-mediated immunosuppression is IDO1 enzyme independent, while the survival benefits of immune checkpoint blockade require non-tumor cell IDO1 enzyme activity. Given that current clinical inhibitors vary in their mechanism of action, in terms of targeting IDO1 enzyme activity versus enzyme-independent effects, this work suggests that choosing an appropriate IDO1 pharmacologic will maximize the effectiveness of future immune checkpoint blockade approaches.
Collapse
Affiliation(s)
- Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Carlos R. Dostal
- Neuroscience Program, The University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Kristen Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Kathleen Swoap
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Leah K. Billingham
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Galina Gritsina
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Meijing Wu
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Robert H. McCusker
- Department of Animal Sciences, The University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - David C. Binder
- Commitee on Cancer Biology, The University of Chicago, Chicago, IL 60637,Department of Pathology, The University of Chicago, Chicago, IL 60637
| | - Derek A. Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611,Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611,Department of Medicine-Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
12
|
The development of dendritic cell vaccine-based immunotherapies for glioblastoma. Semin Immunopathol 2017; 39:225-239. [PMID: 28138787 DOI: 10.1007/s00281-016-0616-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 12/17/2022]
Abstract
In this review, we focus on the biologic advantages of dendritic cell-based vaccinations as a therapeutic strategy for cancer as well as preclinical and emerging clinical data associated with such approaches for glioblastoma patients.
Collapse
|
13
|
Royer-Perron L, Idbaih A, Sanson M, Delattre JY, Hoang-Xuan K, Alentorn A. Precision medicine in glioblastoma therapy. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1241128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
14
|
Urup T, Michaelsen SR, Olsen LR, Toft A, Christensen IJ, Grunnet K, Winther O, Broholm H, Kosteljanetz M, Issazadeh-Navikas S, Poulsen HS, Lassen U. Angiotensinogen and HLA class II predict bevacizumab response in recurrent glioblastoma patients. Mol Oncol 2016; 10:1160-8. [PMID: 27262894 DOI: 10.1016/j.molonc.2016.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/01/2016] [Accepted: 05/19/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Bevacizumab combination therapy is among the most frequently used treatments in recurrent glioblastoma and patients who achieve response to bevacizumab have improved survival as well as quality of life. Accordingly, the aim of this study was to identify predictive biomarkers for bevacizumab response in recurrent glioblastoma patients. METHODS The study included a total of 82 recurrent glioblastoma patients treated with bevacizumab combination therapy whom were both response and biomarker evaluable. Gene expression of tumor tissue was analyzed by using a customized NanoString platform covering 800 genes. Candidate gene predictors associated with response were analyzed by multivariate logistic and Cox regression analysis. RESULTS Two genes were independently associated with response: Low expression of angiotensinogen (2-fold decrease in AGT; OR = 2.44; 95% CI: 1.45-4.17; P = 0.0009) and high expression of a HLA class II gene (2-fold increase in HLA-DQA1; OR = 1.22; 95% CI: 1.01-1.47; P = 0.04). These two genes were included in a model that is able predict response to bevacizumab combination therapy in clinical practice. When stratified for a validated prognostic index, the predictive model for response was significantly associated with improved overall survival. CONCLUSION Two genes (low angiotensinogen and high HLA-class II expression) were predictive for bevacizumab response and were included in a predictive model for response. This model can be used in clinical practice to identify patients who will benefit from bevacizumab combination therapy.
Collapse
Affiliation(s)
- Thomas Urup
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | - Signe Regner Michaelsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Lars Rønn Olsen
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kemitorvet, Building 208, DK-2800 Lyngby, Denmark; Bioinformatics Centre, Department of Biology and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200, Denmark
| | - Anders Toft
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ib Jarle Christensen
- Department of Gastroenterology, Hvidovre Hospital, Kettegård Allé 30, DK-2650 Hvidovre, Denmark
| | - Kirsten Grunnet
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ole Winther
- Bioinformatics Centre, Department of Biology and Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200, Denmark
| | - Helle Broholm
- Department of Neuropathology, Center of Diagnostic Investigation, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Michael Kosteljanetz
- Department of Neurosurgery, The Neurocenter, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | | | - Hans Skovgaard Poulsen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Oncology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ulrik Lassen
- Department of Radiation Biology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Oncology, The Finsen Center, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Phase I Unit, Finsencenter, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
15
|
Gilbert MR. Antiangiogenic Therapy for Glioblastoma: Complex Biology and Complicated Results. J Clin Oncol 2016; 34:1567-9. [DOI: 10.1200/jco.2016.66.5364] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|