1
|
Wang W, Ou Z, Huang X, Wang J, Li Q, Wen M, Zheng L. Microbiota and glioma: a new perspective from association to clinical translation. Gut Microbes 2024; 16:2394166. [PMID: 39185670 DOI: 10.1080/19490976.2024.2394166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Gliomas pose a significant challenge in oncology due to their malignant nature, aggressive growth, frequent recurrence, and complications posed by the blood-brain barrier. Emerging research has revealed the critical role of gut microbiota in influencing health and disease, indicating its possible impact on glioma pathogenesis and treatment responsiveness. This review focused on existing evidence and hypotheses on the relationship between microbiota and glioma from progression to invasion. By discussing possible mechanisms through which microbiota may affect glioma biology, this paper offers new avenues for targeted therapies and precision medicine in oncology.
Collapse
Affiliation(s)
- Wenhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihao Ou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixin Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianbei Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minghui Wen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Tyagi A, Wu SY, Watabe K. Metabolism in the progression and metastasis of brain tumors. Cancer Lett 2022; 539:215713. [PMID: 35513201 PMCID: PMC9999298 DOI: 10.1016/j.canlet.2022.215713] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 01/30/2023]
Abstract
Malignant brain tumors and metastases pose significant health problems and cause substantial morbidity and mortality in children and adults. Based on epidemiological evidence, gliomas comprise 30% and 80% of primary brain tumors and malignant tumors, respectively. Brain metastases affect 15-30% of cancer patients, particularly primary tumors of the lung, breast, colon, and kidney, and melanoma. Despite advancements in multimodal molecular targeted therapy and immunotherapy that do not ensure long-term treatment, malignant brain tumors and metastases contribute significantly to cancer related mortality. Recent studies have shown that metastatic cancer cells possess distinct metabolic traits to adapt and survive in new environment that differs significantly from the primary site in both nutrient composition and availability. As metabolic regulation lies at the intersection of many research areas, concerted efforts to understand the metabolic mechanism(s) driving malignant brain tumors and metastases may reveal novel therapeutic targets to prevent or reduce metastasis and predict biomarkers for the treatment of this aggressive disease. This review focuses on various aspects of metabolic signaling, interface between metabolic regulators and cellular processes, and implications of their dysregulation in the context of brain tumors and metastases.
Collapse
Affiliation(s)
- Abhishek Tyagi
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
3
|
Ruiz-Garcia H, Middlebrooks EH, Trifiletti DM, Chaichana KL, Quinones-Hinojosa A, Sheehan JP. The Extent of Resection in Gliomas-Evidence-Based Recommendations on Methodological Aspects of Research Design. World Neurosurg 2022; 161:382-395.e3. [PMID: 35505558 DOI: 10.1016/j.wneu.2021.08.140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Modern neurosurgery has established maximal safe resection as a cornerstone in the management of diffuse gliomas. Evaluation of the extent of resection (EOR), and its association with certain outcomes or interventions, heavily depends on an adequate methodology to draw strong conclusions. We aim to identify weaknesses and limitations that may threaten the internal validity and generalizability of studies involving the EOR in patients with glioma and to suggest methodological recommendations that may help mitigate these threats. METHODS A systematic search was performed by querying PubMed, Web of Science, and Scopus since inception to April 30, 2021 using PICOS/PRISMA guidelines. Articles were then screened to identify high-impact studies evaluating the EOR in patients diagnosed with diffuse gliomas in accordance with predefined criteria. We identify common weakness and limitations during the evaluation of the EOR in the selected studies and then delineate potential methodological recommendations for future endeavors dealing with the EOR. RESULTS We identified 31 high-impact studies and found several research design issues including inconsistencies regarding EOR terminology, measurement, data collection, analysis, and reporting. Although some of these issues were related to now outdated reporting standards, many were still present in recent publications and deserve attention in contemporary and future research. CONCLUSIONS There is a current need to focus more attention to the methodological aspects of glioma research. Methodological inconsistencies may introduce weaknesses into the internal validity of the studies and hamper comparative analysis of cohorts from different institutions. We hope our recommendations will eventually help develop stronger methodological designs in future research endeavors.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, Florida, USA; Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida, USA; Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Jacksonville, Florida, USA
| | - Erik H Middlebrooks
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, Florida, USA; Department of Radiology, Mayo Clinic, Jacksonville, Florida, USA
| | - Daniel M Trifiletti
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, Florida, USA; Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | - Jason P Sheehan
- Department of Neurological Surgery, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
4
|
Meng W, Palmer JD, Siedow M, Haque SJ, Chakravarti A. Overcoming Radiation Resistance in Gliomas by Targeting Metabolism and DNA Repair Pathways. Int J Mol Sci 2022; 23:ijms23042246. [PMID: 35216362 PMCID: PMC8880405 DOI: 10.3390/ijms23042246] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 02/06/2023] Open
Abstract
Gliomas represent a wide spectrum of brain tumors characterized by their high invasiveness, resistance to chemoradiotherapy, and both intratumoral and intertumoral heterogeneity. Recent advances in transomics studies revealed that enormous abnormalities exist in different biological layers of glioma cells, which include genetic/epigenetic alterations, RNA expressions, protein expression/modifications, and metabolic pathways, which provide opportunities for development of novel targeted therapeutic agents for gliomas. Metabolic reprogramming is one of the hallmarks of cancer cells, as well as one of the oldest fields in cancer biology research. Altered cancer cell metabolism not only provides energy and metabolites to support tumor growth, but also mediates the resistance of tumor cells to antitumor therapies. The interactions between cancer metabolism and DNA repair pathways, and the enhancement of radiotherapy sensitivity and assessment of radiation response by modulation of glioma metabolism are discussed herein.
Collapse
|
5
|
Abstract
ABSTRACT High-grade gliomas are among the deadliest of all cancers despite standard treatments, and new therapeutic strategies are needed to improve patient outcome. Targeting the altered metabolic state of tumors with traditional chemotherapeutic agents has a history of success, and our increased understanding of cellular metabolism in the past 2 decades has reinvigorated the concept of novel metabolic therapies in brain tumors. Here we highlight metabolic alterations in advanced gliomas and their translation into clinical trials using both novel agents and already established drugs repurposed for cancer treatment in an effort to improve outcome for these deadly diseases.
Collapse
Affiliation(s)
- Andrew J. Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Daniel R. Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109; University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
6
|
Yin G, Yang Z. Fractional design: An alternative paradigm for late-onset toxicities in oncology dose-finding studies. Contemp Clin Trials Commun 2020; 19:100650. [PMID: 32875142 PMCID: PMC7451759 DOI: 10.1016/j.conctc.2020.100650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/05/2020] [Accepted: 08/16/2020] [Indexed: 11/17/2022] Open
Abstract
Late-onset (LO) toxicities often arise in the new era of phase I oncology dose-finding trials with targeted agents or immunotherapies. The current LO toxicities modelling is often formulated in a weighted likelihood framework, where the time-to-event continual reassessment method (TITE-CRM) is commonly used. The TITE-CRM uses the patient exposure time as a weight for the censored observation, while there is large uncertainty on which weight function to be used. As an alternative, the fractional scheme formulates an efficient and robust paradigm to address LO toxicity issues in dose finding. We review the fractional continual reassessment method (fCRM) and compare its operating characteristics with those of the TITE-CRM as well as other competitive designs via extensive simulation studies based on both the fixed and randomly generated scenarios. The fCRM is shown to possess desirable operating characteristics in identifying the maximum tolerated dose (MTD) and deliver competitive performances in comparison with other designs. It provides an alternative efficient and robust paradigm for interpreting and addressing LO toxicities in the new era of phase I dose-finding trials in precision oncology. A real trial example is used to illustrate the practical use of the fCRM design.
Collapse
Affiliation(s)
- Guosheng Yin
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zhao Yang
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
7
|
Ascha MS, Wang JF, Kumthekar P, Sloan AE, Kruchko C, Barnholtz-Sloan JS. Bevacizumab for the treatment of non-small cell lung cancer patients with synchronous brain metastases. Sci Rep 2019; 9:17792. [PMID: 31780762 PMCID: PMC6882803 DOI: 10.1038/s41598-019-54513-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Bevacizumab is FDA-approved in the treatment of primary brain tumors, but its efficacy in patients with brain metastases could be better-studied. This study examines a population of non-small cell lung cancer (NSCLC) patients with synchronous brain metastases to identify predictors of the decision to use bevacizumab and survival following bevacizumab treatment. Primary cancer registry data were used to determine which NSCLC patients diagnosed in the years 2010 through 2012 had synchronous brain metastases at the time of diagnosis, and Medicare claims used to identify a population of patients treated with bevacizumab. Record of bevacizumab treatment was found for 81 and 666 patients with and without brain metastases, respectively. After adjusting for clinical and demographic characteristics, bevacizumab was associated with 0.88 times the hazard of mortality in the elderly NSCLC population (95% CI: 0.81–0.96, p: 0.003) and a corresponding hazard ratio of 0.75 in the population of elderly NSCLC patients with synchronous brain metastases (95% CI: 0.59–0.96, p: 0.020). Bevacizumab may benefit NSCLC patients with synchronous brain metastases more than it does patients without intracranial disease, possibly as a result of its multiple potential mechanisms of action simultaneously inhibiting angiogenesis and minimizing vasogenic edema.
Collapse
Affiliation(s)
- Mustafa S Ascha
- Center for Clinical Investigation, Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Priya Kumthekar
- Northwestern University Feinberg School of Medicine, Department of Neurology, Evanston, Illinois, USA
| | - Andrew E Sloan
- Department of Neurosurgery, University Hospitals Cleveland Medical Center, Seidman Cancer Center, and the Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Carol Kruchko
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
| | - Jill S Barnholtz-Sloan
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA. .,Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.
| |
Collapse
|
8
|
A Humanized Yeast Phenomic Model of Deoxycytidine Kinase to Predict Genetic Buffering of Nucleoside Analog Cytotoxicity. Genes (Basel) 2019; 10:genes10100770. [PMID: 31575041 PMCID: PMC6826991 DOI: 10.3390/genes10100770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/17/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
Knowledge about synthetic lethality can be applied to enhance the efficacy of anticancer therapies in individual patients harboring genetic alterations in their cancer that specifically render it vulnerable. We investigated the potential for high-resolution phenomic analysis in yeast to predict such genetic vulnerabilities by systematic, comprehensive, and quantitative assessment of drug–gene interaction for gemcitabine and cytarabine, substrates of deoxycytidine kinase that have similar molecular structures yet distinct antitumor efficacy. Human deoxycytidine kinase (dCK) was conditionally expressed in the Saccharomyces cerevisiae genomic library of knockout and knockdown (YKO/KD) strains, to globally and quantitatively characterize differential drug–gene interaction for gemcitabine and cytarabine. Pathway enrichment analysis revealed that autophagy, histone modification, chromatin remodeling, and apoptosis-related processes influence gemcitabine specifically, while drug–gene interaction specific to cytarabine was less enriched in gene ontology. Processes having influence over both drugs were DNA repair and integrity checkpoints and vesicle transport and fusion. Non-gene ontology (GO)-enriched genes were also informative. Yeast phenomic and cancer cell line pharmacogenomics data were integrated to identify yeast–human homologs with correlated differential gene expression and drug efficacy, thus providing a unique resource to predict whether differential gene expression observed in cancer genetic profiles are causal in tumor-specific responses to cytotoxic agents.
Collapse
|
9
|
Zhou W, Wahl DR. Metabolic Abnormalities in Glioblastoma and Metabolic Strategies to Overcome Treatment Resistance. Cancers (Basel) 2019; 11:cancers11091231. [PMID: 31450721 PMCID: PMC6770393 DOI: 10.3390/cancers11091231] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor and is nearly universally fatal. Targeted therapy and immunotherapy have had limited success in GBM, leaving surgery, alkylating chemotherapy and ionizing radiation as the standards of care. Like most cancers, GBMs rewire metabolism to fuel survival, proliferation, and invasion. Emerging evidence suggests that this metabolic reprogramming also mediates resistance to the standard-of-care therapies used to treat GBM. In this review, we discuss the noteworthy metabolic features of GBM, the key pathways that reshape tumor metabolism, and how inhibiting abnormal metabolism may be able to overcome the inherent resistance of GBM to radiation and chemotherapy.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
10
|
Elmaci İ, Bilir A, Ozpinar A, Altinoz MA. Gemcitabine, vinorelbine and cyclooxygenase inhibitors in the treatment of glioblastoma. Ultrastructural analyses in C6 glioma in vitro. Tissue Cell 2019; 59:18-32. [PMID: 31383285 DOI: 10.1016/j.tice.2019.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/20/2019] [Accepted: 05/26/2019] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To define ultrastructural features accompanying to antitumor effects of gemcitabine, vinorelbine and cyclooxygenase inhibitors in C6 glioma cells in vitro. Vinorelbine is a semisynthetic vinca alkaloid and recent studies showed its antitumor activity in pediatric optic and pontine gliomas. Vinorelbine infusion induces a severe tumor site-pain in systemic cancers, but it is unknown whether algesia and inflammation contribute to its antitumor effects. Gemcitabine is a nucleoside-chemotherapeutic which was recently shown to act as a radiosensitizer in high-grade glioma. Some studies showed synergism of anti-inflammatory cyclooxygenase-inhibitors with microtubule inhibitors and gemcitabine. DMSO is a solvent and blocks both cylooxygenase and ribonucleotide reductase, another target of gemcitabine. Rofecoxib is withdrawn from the market, yet we used it for investigational purposes, since it blocks cylooxygenase-2 1000-times more potently than cylooxygenase -1 and is also a selective inhibitor of crinophagy. METHODS Plating efficacy, 3D-spheroid S-phase analysis with BrdU labelling and transmission electron microscopical analyses were performed. RESULTS Vinorelbine induced frequent mitotic slippage/apoptosis and autophagy. Despite both DMSO and rofecoxib induced autophagy alone and in synergy, they reduced mitotic catastrophe and autophagy triggered by vinorelbine, which was also reflected by reduced inhibition of spheroid S-phase. Gemcitabine induced karyolysis and margination of coarse chromatin towards the nuclear membrane, abundant autophagy, gutta adipis formation and decrease in mitochondria, which were enhanced by DMSO and rofecoxib. CONCLUSIONS Detailed ultrastructural analysis of the effects of chemotherapeutic drugs may provide a broader insight about their actions and pave to develop better strategies in treatment of glioblastoma.
Collapse
Affiliation(s)
- İlhan Elmaci
- Department of Neurosurgery, Acibadem Hospital, Istanbul, Turkey
| | - Ayhan Bilir
- Department of Histology and Embryology, Aydin University, Istanbul, Turkey
| | - Aysel Ozpinar
- Department of Medical Biochemistry, Acibadem University, Istanbul, Turkey
| | - Meric A Altinoz
- Department of Medical Biochemistry, Acibadem University, Istanbul, Turkey; Department of Psychiatry, Maastricht University, Holland.
| |
Collapse
|
11
|
Yang CA, Huang HY, Lin CL, Chang JG. G6PD as a predictive marker for glioma risk, prognosis and chemosensitivity. J Neurooncol 2018; 139:661-670. [DOI: 10.1007/s11060-018-2911-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/19/2018] [Indexed: 12/13/2022]
|
12
|
Ferguson SD, Xiu J, Weathers SP, Zhou S, Kesari S, Weiss SE, Verhaak RG, Hohl RJ, Barger GR, Reddy SK, Heimberger AB. GBM-associated mutations and altered protein expression are more common in young patients. Oncotarget 2018; 7:69466-69478. [PMID: 27579614 PMCID: PMC5342491 DOI: 10.18632/oncotarget.11617] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
Abstract
Background Geriatric glioblastoma (GBM) patients have a poorer prognosis than younger patients, but IDH1/2 mutations (more common in younger patients) confer a favorable prognosis. We compared key GBM molecular alterations between an elderly (age ≥ 70) and younger (18 < = age < = 45) cohort to explore potential therapeutic opportunities. Results Alterations more prevalent in the young GBM cohort compared to the older cohort (P < 0.05) were: overexpression of ALK, RRM1, TUBB3 and mutation of ATRX, BRAF, IDH1, and TP53. However, PTEN mutation was significantly more frequent in older patients. Among patients with wild-type IDH1/2 status, TOPO1 expression was higher in younger patients, whereas MGMT methylation was more frequent in older patients. Within the molecularly-defined IDH wild-type GBM cohort, younger patients had significantly more mutations in PDGFRA, PTPN11, SMARCA4, BRAF and TP53. Methods GBMs from 178 elderly patients and 197 young patients were analyzed using DNA sequencing, immunohistochemistry, in situ hybridization, and MGMT-methylation assay to ascertain mutational and amplification/expressional status. Conclusions Significant molecular differences occurred in GBMs from elderly and young patients. Except for the older cohort's more frequent PTEN mutation and MGMT methylation, younger patients had a higher frequency of potential therapeutic targets.
Collapse
Affiliation(s)
- Sherise D Ferguson
- Departments of Neurosurgery, Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Joanne Xiu
- Caris Life Sciences, Phoenix, AZ 85040, USA
| | - Shiao-Pei Weathers
- Departments of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Shouhao Zhou
- Departments of Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Santosh Kesari
- Department of Translational Neuro-Oncology and Neurotherapeutics, Pacific Neuroscience Institute and John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA 90404, USA
| | | | - Roeland G Verhaak
- Department of Genome Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77054, USA
| | - Raymond J Hohl
- Penn State Hershey Cancer Institute, Hershey, PA 17033, USA
| | - Geoffrey R Barger
- Department of Neurology, Wayne State University, School of Medicine, Karmanos Cancer Center, Detroit, MI 48201, USA
| | | | - Amy B Heimberger
- Departments of Neurosurgery, Biostatistics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
13
|
Bastiancich C, Bastiat G, Lagarce F. Gemcitabine and glioblastoma: challenges and current perspectives. Drug Discov Today 2017; 23:416-423. [PMID: 29074439 DOI: 10.1016/j.drudis.2017.10.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 12/15/2022]
Abstract
Gemcitabine is a nucleoside analog currently used for the treatment of various solid tumors as a single agent or in combination with other chemotherapeutic drugs. Its use against highly aggressive brain tumors (glioblastoma) has been evaluated in preclinical and clinical trials leading to controversial results. Gemcitabine can inhibit DNA chain elongation, is a potent radiosensitizer and it can enhance antitumor immune activity, but it also presents some drawbacks (e.g., short half-life, side effects, chemoresistance). The aim of this review is to discuss the challenges related to the use of gemcitabine for glioblastoma and to report recent studies that suggest overcoming these obstacles opening new perspectives for its use in the field (e.g., gemcitabine derivatives and/or nanomedicines).
Collapse
Affiliation(s)
- Chiara Bastiancich
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Université Catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Guillaume Bastiat
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | - Frederic Lagarce
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Pharmacy Department, CHU Angers, Angers University Hospital, France.
| |
Collapse
|
14
|
Veldhuijzen van Zanten SEM, El-Khouly FE, Jansen MHA, Bakker DP, Sanchez Aliaga E, Haasbeek CJA, Wolf NI, Zwaan CM, Vandertop WP, van Vuurden DG, Kaspers GJL. A phase I/II study of gemcitabine during radiotherapy in children with newly diagnosed diffuse intrinsic pontine glioma. J Neurooncol 2017; 135:307-315. [PMID: 28748343 PMCID: PMC5663796 DOI: 10.1007/s11060-017-2575-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/16/2017] [Indexed: 01/17/2023]
Abstract
The purpose of this phase I/II, open-label, single-arm trial is to investigate the safety, tolerability, maximum tolerated dose and preliminary efficacy of the potential radiosensitizer gemcitabine, administered concomitantly to radiotherapy, in children with newly diagnosed diffuse intrinsic pontine glioma (DIPG). Six doses of weekly gemcitabine were administered intravenously, concomitantly to 6 weeks of hyperfractionated radiotherapy. Successive cohorts received increasing doses of 140, 175 and 200 mg/m2 gemcitabine, respectively, following a 3 + 3 dose-escalation schedule without expansion cohort. Dose-limiting toxicities (DLT) were monitored during treatment period. Clinical response was assessed using predefined case report forms and radiological response was assessed using the modified RANO criteria. Quality of life (QoL) was assessed using PedsQL questionnaires. Between June 2012 and December 2016, nine patients were enrolled. Treatment was well tolerated, and no DLTs were observed up to the maximum dose of 200 mg/m2. All patients experienced reduction of tumor-related symptoms. QoL tended to improve during treatment. PFS and MOS were 4.8 months (95% CI 4.0–5.7) and 8.7 months (95% CI 7.0–10.4). Classifying patients according to the recently developed DIPG survival prediction model, intermediate risk patients (n = 4), showed a PFS and MOS of 6.4 and 12.4 months, respectively, versus a PFS and MOS of 4.5 and 8.1 months, respectively, in high risk patient (n = 5). Gemcitabine up to 200 mg/m2/once weekly, added to radiotherapy, is safe and well tolerated in children with newly diagnosed DIPG. PFS and MOS were not significantly different from literature.
Collapse
Affiliation(s)
| | - Fatma E El-Khouly
- Department of Pediatric Oncology - Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Pharmacology & Pharmacy, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc H A Jansen
- Department of Pediatric Oncology - Hematology, VU University Medical Center, Amsterdam, The Netherlands
| | - Dewi P Bakker
- Department of Child Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Esther Sanchez Aliaga
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Cornelis J A Haasbeek
- Department of Radiotherapy, VU University Medical Center, Amsterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, VU University Medical Center, Amsterdam, The Netherlands.,Neuroscience Amsterdam, Amsterdam, The Netherlands
| | - C Michel Zwaan
- Department of Pediatric Oncology - Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - W Peter Vandertop
- Neurosurgical Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands.,Neurosurgical Center Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - Dannis G van Vuurden
- Department of Pediatric Oncology - Hematology, VU University Medical Center, Amsterdam, The Netherlands. .,Department of Pediatrics, Division of Oncology - Hematology, VU University Medical Center, De Boelelaan 1117, Room 9D36, 1081 HV, Amsterdam, The Netherlands.
| | - Gertjan J L Kaspers
- Department of Pediatric Oncology - Hematology, VU University Medical Center, Amsterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
15
|
Wahl DR, Dresser J, Wilder-Romans K, Parsels JD, Zhao SG, Davis M, Zhao L, Kachman M, Wernisch S, Burant CF, Morgan MA, Feng FY, Speers C, Lyssiotis CA, Lawrence TS. Glioblastoma Therapy Can Be Augmented by Targeting IDH1-Mediated NADPH Biosynthesis. Cancer Res 2016; 77:960-970. [PMID: 27923831 DOI: 10.1158/0008-5472.can-16-2008] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 12/21/2022]
Abstract
NADPH is a critical reductant needed in cancer cells to fuel the biosynthesis of deoxynucleotides and antioxidants and to sustain stress-survival responses after radiation-induced DNA damage. Thus, one rational strategy to attack cancer cells is to target their heavy reliance on NADPH. Here, we report that the isocitrate dehydrogenase IDH1 is the most strongly upregulated NADPH-producing enzyme in glioblastoma (GBM). IDH1 silencing in GBM cells reduced levels of NADPH, deoxynucleotides, and glutathione and increased their sensitivity to radiation-induced senescence. Rescuing these metabolic restrictions was sufficient to reverse IDH1-mediated radiosensitization. In a murine xenograft model of human GBM, we found that IDH1 silencing significantly improved therapeutic responses to fractionated radiotherapy, when compared with either treatment alone. In summary, our work offers a mechanistic rationale for IDH1 inhibition as a metabolic strategy to improve the response of GBM to radiotherapy. Cancer Res; 77(4); 960-70. ©2016 AACR.
Collapse
Affiliation(s)
- Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
| | - Joseph Dresser
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Joshua D Parsels
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Shuang G Zhao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Mary Davis
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Maureen Kachman
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Stefanie Wernisch
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Charles F Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Meredith A Morgan
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA
| | - Corey Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Costas A Lyssiotis
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology; University of Michigan, Ann Arbor, Michigan
| | - Theodore S Lawrence
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|