1
|
Pachocki CJ, Hol EM. Current perspectives on diffuse midline glioma and a different role for the immune microenvironment compared to glioblastoma. J Neuroinflammation 2022; 19:276. [PMCID: PMC9675250 DOI: 10.1186/s12974-022-02630-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 10/25/2022] [Indexed: 11/21/2022] Open
Abstract
Diffuse midline glioma (DMG), formerly called diffuse intrinsic pontine glioma (DIPG), is a high-grade malignant pediatric brain tumor with a near-zero survival rate. To date, only radiation therapy provides marginal survival benefit; however, the median survival time remains less than a year. Historically, the infiltrative nature and sensitive location of the tumor rendered surgical removal and biopsies difficult and subsequently resulted in limited knowledge of the disease, as only post-mortem tissue was available. Therefore, clinical decision-making was based upon experience with the more frequent and histologically similar adult glioblastoma (GBM). Recent advances in tissue acquisition and molecular profiling revealed that DMG and GBM are distinct disease entities, with separate tissue characteristics and genetic profiles. DMG is characterized by heterogeneous tumor tissue often paired with an intact blood–brain barrier, possibly explaining its resistance to chemotherapy. Additional profiling shed a light on the origin of the disease and the influence of several mutations such as a highly recurring K27M mutation in histone H3 on its tumorigenesis. Furthermore, early evidence suggests that DMG has a unique immune microenvironment, characterized by low levels of immune cell infiltration, inflammation, and immunosuppression that may impact disease development and outcome. Within the tumor microenvironment of GBM, tumor-associated microglia/macrophages (TAMs) play a large role in tumor development. Interestingly, TAMs in DMG display distinct features and have low immune activation in comparison to other pediatric gliomas. Although TAMs have been investigated substantially in GBM over the last years, this has not been the case for DMG due to the lack of tissue for research. Bit by bit, studies are exploring the TAM–glioma crosstalk to identify what factors within the DMG microenvironment play a role in the recruitment and polarization of TAMs. Although more research into the immune microenvironment is warranted, there is evidence that targeting or stimulating TAMs and their factors provide a potential treatment option for DMG. In this review, we provide insight into the current status of DMG research, assess the knowledge of the immune microenvironment in DMG and GBM, and present recent findings and therapeutic opportunities surrounding the TAM–glioma crosstalk.
Collapse
Affiliation(s)
- Casper J. Pachocki
- grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M. Hol
- grid.5477.10000000120346234Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Pediatric brain tumors as a developmental disease. Curr Opin Oncol 2021; 33:608-614. [PMID: 34431811 DOI: 10.1097/cco.0000000000000782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Brain tumors are the most frequent solid cancer in the pediatric population. Owing to the rarity of environmental clues about their origin, it is tempting to consider these neoplasms as developmental processes gone awry. Our review will explore the heuristic power of this hypothesis and the influence of these findings on the clinical management. RECENT FINDING A more accurate description of cancer predisposition syndrome has shown their frequent association with developmental abnormalities. Several genes involved in pediatric brain tumor oncogenesis are involved in developmental processes. Modeling of several pediatric brain tumor in cerebral organoids, mimicking embryonal stage of brain development, indicates that early events during brain development create the conditions necessary for their oncogenesis. SUMMARY The onset of multiple brain tumor types early in life suggests a functional relationship between brain development and oncogenesis. A growing body of evidence seems to support the hypothesis that some of the main developmental steps in the brain can be highjacked by the tumors during their initiation. Collaborations between neuroscientists and oncologists should provide room for improvement in the knowledge for these neoplasms.
Collapse
|
3
|
Price G, Bouras A, Hambardzumyan D, Hadjipanayis CG. Current knowledge on the immune microenvironment and emerging immunotherapies in diffuse midline glioma. EBioMedicine 2021; 69:103453. [PMID: 34157482 PMCID: PMC8220552 DOI: 10.1016/j.ebiom.2021.103453] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/29/2022] Open
Abstract
Diffuse midline glioma (DMG) is an incurable malignancy with the highest mortality rate among pediatric brain tumors. While radiotherapy and chemotherapy are the most common treatments, these modalities have limited promise. Due to their diffuse nature in critical areas of the brain, the prognosis of DMG remains dismal. DMGs are characterized by unique phenotypic heterogeneity and histological features. Mutations of H3K27M, TP53, and ACVR1 drive DMG tumorigenesis. Histological artifacts include pseudopalisading necrosis and vascular endothelial proliferation. Mouse models that recapitulate human DMG have been used to study key driver mutations and the tumor microenvironment. DMG consists of a largely immunologically cold tumor microenvironment that lacks immune cell infiltration, immunosuppressive factors, and immune surveillance. While tumor-associated macrophages are the most abundant immune cell population, there is reduced T lymphocyte infiltration. Immunotherapies can stimulate the immune system to find, attack, and eliminate cancer cells. However, it is critical to understand the immune microenvironment of DMG before designing immunotherapies since differences in the microenvironment influence treatment efficacy. To this end, our review aims to overview the immune microenvironment of DMG, discuss emerging insights about the immune landscape that drives disease pathophysiology, and present recent findings and new opportunities for therapeutic discovery.
Collapse
Affiliation(s)
- Gabrielle Price
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai,10 Union Square East, 5th Floor, Suite 5E, New York, NY 10003, USA; Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Alexandros Bouras
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai,10 Union Square East, 5th Floor, Suite 5E, New York, NY 10003, USA; Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dolores Hambardzumyan
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai,10 Union Square East, 5th Floor, Suite 5E, New York, NY 10003, USA; Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Constantinos G Hadjipanayis
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai,10 Union Square East, 5th Floor, Suite 5E, New York, NY 10003, USA; Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Karki A, Berlow NE, Kim JA, Hulleman E, Liu Q, Michalek JE, Keller C. Receptor-driven invasion profiles in diffuse intrinsic pontine glioma. Neurooncol Adv 2021; 3:vdab039. [PMID: 34013206 PMCID: PMC8117434 DOI: 10.1093/noajnl/vdab039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Diffuse intrinsic pontine glioma (DIPG) is a devastating pediatric cancer with unmet clinical need. DIPG is invasive in nature, where tumor cells interweave into the fiber nerve tracts of the pons making the tumor unresectable. Accordingly, novel approaches in combating the disease are of utmost importance and receptor-driven cell invasion in the context of DIPG is under-researched area. Here, we investigated the impact on cell invasion mediated by PLEXINB1, PLEXINB2, platelet growth factor receptor (PDGFR)α, PDGFRβ, epithelial growth factor receptor (EGFR), activin receptor 1 (ACVR1), chemokine receptor 4 (CXCR4), and NOTCH1. Methods We used previously published RNA-sequencing data to measure gene expression of selected receptors in DIPG tumor tissue versus matched normal tissue controls (n = 18). We assessed protein expression of the corresponding genes using DIPG cell culture models. Then, we performed cell viability and cell invasion assays of DIPG cells stimulated with chemoattractants/ligands. Results RNA-sequencing data showed increased gene expression of receptor genes such as PLEXINB2, PDGFRα, EGFR, ACVR1, CXCR4, and NOTCH1 in DIPG tumors compared to the control tissues. Representative DIPG cell lines demonstrated correspondingly increased protein expression levels of these genes. Cell viability assays showed minimal effects of growth factors/chemokines on tumor cell growth in most instances. Recombinant SEMA4C, SEM4D, PDGF-AA, PDGF-BB, ACVA, CXCL12, and DLL4 ligand stimulation altered invasion in DIPG cells. Conclusions We show that no single growth factor-ligand pair universally induces DIPG cell invasion. However, our results reveal a potential to create a composite of cytokines or anti-cytokines to modulate DIPG cell invasion.
Collapse
Affiliation(s)
- Anju Karki
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Noah E Berlow
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Jin-Ah Kim
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| | - Esther Hulleman
- Department of Pediatric Oncology/Hematology, Cancer Center Amsterdam, VU University Medical Center, Amsterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht,The Netherlands
| | - Qianqian Liu
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Joel E Michalek
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, Beaverton, Oregon, USA
| |
Collapse
|
5
|
Sesen J, Driscoll J, Shah N, Moses-Gardner A, Luiselli G, Alexandrescu S, Zurakowski D, Baxter PA, Su JM, Pricola Fehnel K, Smith ER. Neogenin is highly expressed in diffuse intrinsic pontine glioma and influences tumor invasion. Brain Res 2021; 1762:147348. [PMID: 33571520 DOI: 10.1016/j.brainres.2021.147348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Julie Sesen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Jessica Driscoll
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Nishali Shah
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Alexander Moses-Gardner
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Gabrielle Luiselli
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Sanda Alexandrescu
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - David Zurakowski
- Department of Surgery, Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Patricia A Baxter
- Texas Children's Hospital/Baylor College of Medicine, 77030 Houston, TX, USA
| | - Jack M Su
- Texas Children's Hospital/Baylor College of Medicine, 77030 Houston, TX, USA
| | - Katie Pricola Fehnel
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Edward R Smith
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA; Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, 02115 Boston, MA, USA.
| |
Collapse
|
6
|
Development of a clinical scale for assessment of patients with diffuse intrinsic pontine glioma (DIPG) receiving experimental therapy: the PONScore. J Neurooncol 2020; 149:263-272. [PMID: 32902768 DOI: 10.1007/s11060-020-03594-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/08/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Monitoring neurological side-effects in experimental therapy for diffuse intrinsic pontine glioma (DIPG) can be challenging. We aimed to develop a neurological scale that could be used by non-specialists to quantify neurological changes during experimental treatment of DIPG. METHODS We developed the Pontine Observational Neurological Score (PONScore) to measure signs and symptoms of DIPG by adapting validated assessment scales of neurological signs and symptoms in children. We developed a prototype score, taught it to paediatric intensive care nursing staff, who used the Score to assess children receiving awake pontine infusion of chemotherapy for treatment of DIPG. We used their feedback to develop the PONScore. Points are allocated for headache, ophthalmoplegia, facial and tongue weakness, dysarthria, paraesthesia, limb weakness and dysmetria with increasing scores reflecting increasing disability. The PONScore was administered every hour during awake pontine infusion. Correlation and agreement calculations between nursing staff, as non-specialists, and a specialist rater were performed in 30 infusions in 6 children (aged 8-11). Changes in PONScore versus volume of infusion are described in a further 55 infusions in 8 children (aged 3-11). RESULTS The PONScore demonstrated excellent intra-rater reliability with an intra-class co-efficient of 0.98 (95% CI 0.97-0.99; p-value < 0.001) between a specialist and non-specialist raters with strong correlation between scores and a Spearman correlation coefficient of 0.985 (p < 0.001). PONScores increased from 3.3 to 5.7 (p-value < 0.001) during infusion reflecting accumulation of neurological signs and symptoms during infusion. CONCLUSIONS We describe a novel neurological scale that can be used by non-specialists to describe acute neurological changes in children receiving experimental therapy for DIPG. Prospective validation as part of a clinical trial is required.
Collapse
|
7
|
Felker J, Broniscer A. Improving long-term survival in diffuse intrinsic pontine glioma. Expert Rev Neurother 2020; 20:647-658. [PMID: 32543245 DOI: 10.1080/14737175.2020.1775584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Diffuse intrinsic pontine glioma (DIPG) is an almost universally fatal pediatric brain cancer. There has been no improvement in event-free survival (EFS) or overall survival (OS) despite immense effort through a multitude of clinical trials to find a cure. Recently, there has been a surge in the knowledge of DIPG biology, including the discovery of a recurrent H3F3A mutation in over 80% of these tumors. AREAS COVERED The authors review the most recent approaches to diagnosis and treatment of DIPG including chemotherapy, biologics, surgical approaches, and immunotherapy. EXPERT OPINION The authors propose four main opportunities to improve long-term survival. First, patients should be enrolled in scientifically sound clinical trials that include molecularly profiling either via stereotactic biopsy or liquid biopsy. Second, clinical trials should include more innovative endpoints other than traditional EFS and OS such as MRI/PET imaging findings combined with surrogates of activity (e.g. serial liquid biopsies) to better ascertain biologically active treatments. Third, innovative clinical trial approaches are needed to help allow for the rapid development of combination therapies to be tested. Finally, effort should be concentrated on reversing the effects of the histone mutation, as this malfunctioning development program seems to be key to DIPG relentlessness.
Collapse
Affiliation(s)
- James Felker
- Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.,Pediatric Neuro-Oncology, UPMC Children's Hospital of Pittsburgh , Pittsburgh, PA, USA
| | - Alberto Broniscer
- Department of Pediatrics, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.,Pediatric Neuro-Oncology, UPMC Children's Hospital of Pittsburgh , Pittsburgh, PA, USA
| |
Collapse
|
8
|
Sekimata K, Sato T, Sakai N. ALK2: A Therapeutic Target for Fibrodysplasia Ossificans Progressiva and Diffuse Intrinsic Pontine Glioma. Chem Pharm Bull (Tokyo) 2020; 68:194-200. [PMID: 32115526 DOI: 10.1248/cpb.c19-00882] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fibrodysplasia ossificans progressiva (FOP) and diffuse intrinsic pontine glioma (DIPG) are diseases that typically manifest in childhood and are associated with severely reduced life expectancy. However, there are currently no effective therapies for these diseases, which remain incurable. Activin receptor-like kinase-2 (ALK2), encoded by the ACVR1 gene, is a bone morphogenetic protein (BMP) type-I receptor subtype that plays an important physiological role in the development of bones, muscles, brain, and other organs. Constitutively active mutants of ALK2 have been identified as causative of FOP and involved in the tumorigenesis of DIPG owing to abnormal activation of BMP signaling, and therefore have emerged as promising treatment targets. Here, we describe these two diseases, along with the link to ALK2 signal transduction, and highlight potential ALK2 inhibitors that are under development to offer new hope for patients with FOP and DIPG.
Collapse
Affiliation(s)
- Katsuhiko Sekimata
- Drug Discovery Chemistry Platform Unit, RIKEN Center for Sustainable Resource Science
| | - Tomohiro Sato
- Drug Discovery Computational Chemistry Platform Unit, RIKEN Center for Biosystems Dynamics Research
| | - Naoki Sakai
- Drug Discovery Structural Biology Platform Unit, RIKEN Biosystems Dynamics Research
| |
Collapse
|
9
|
Kluiver TA, Alieva M, van Vuurden DG, Wehrens EJ, Rios AC. Invaders Exposed: Understanding and Targeting Tumor Cell Invasion in Diffuse Intrinsic Pontine Glioma. Front Oncol 2020; 10:92. [PMID: 32117746 PMCID: PMC7020612 DOI: 10.3389/fonc.2020.00092] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 01/17/2020] [Indexed: 12/20/2022] Open
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a rare, highly aggressive pediatric brain tumor that originates in the pons. DIPG is untreatable and universally fatal, with a median life expectancy of less than a year. Resection is not an option, due to the anatomical location of the tumor, radiotherapy has limited effect and no chemotherapeutic or targeted treatment approach has proven to be successful. This poor prognosis is partly attributed to the tumor's highly infiltrative diffuse and invasive spread. Thus, targeting the invasive behavior of DIPG has the potential to be of therapeutic value. In order to target DIPG invasion successfully, detailed mechanistic knowledge on the underlying drivers is required. Here, we review both DIPG tumor cell's intrinsic molecular processes and extrinsic environmental factors contributing to DIPG invasion. Importantly, DIPG represents a heterogenous disease and through advances in whole-genome sequencing, different subtypes of disease based on underlying driver mutations are now being recognized. Recent evidence also demonstrates intra-tumor heterogeneity in terms of invasiveness and implies that highly infiltrative tumor subclones can enhance the migratory behavior of neighboring cells. This might partially be mediated by “tumor microtubes,” long membranous extensions through which tumor cells connect and communicate, as well as through the secretion of extracellular vesicles. Some of the described processes involved in invasion are already being targeted in clinical trials. However, more research into the mechanisms of DIPG invasion is urgently needed and might result in the development of an effective therapy for children suffering from this devastating disease. We discuss the implications of newly discovered invasive mechanisms for therapeutic targeting and the challenges therapy development face in light of disease in the developing brain.
Collapse
Affiliation(s)
- T A Kluiver
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute, KNAW Utrecht, Utrecht, Netherlands.,Cancer Genomics Center, Utrecht, Netherlands
| | - M Alieva
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute, KNAW Utrecht, Utrecht, Netherlands.,Cancer Genomics Center, Utrecht, Netherlands
| | - D G van Vuurden
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Ellen J Wehrens
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute, KNAW Utrecht, Utrecht, Netherlands.,Cancer Genomics Center, Utrecht, Netherlands
| | - Anne C Rios
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Cancer Research, Oncode Institute, Hubrecht Institute, KNAW Utrecht, Utrecht, Netherlands.,Cancer Genomics Center, Utrecht, Netherlands
| |
Collapse
|
10
|
A New Treatment Opportunity for DIPG and Diffuse Midline Gliomas: 5-ALA Augmented Irradiation, the 5aai Regimen. Brain Sci 2020; 10:brainsci10010051. [PMID: 31963414 PMCID: PMC7016657 DOI: 10.3390/brainsci10010051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/19/2022] Open
Abstract
Prognosis for diffuse intrinsic pontine glioma (DIPG) and generally for diffuse midline gliomas (DMG) has only marginally improved over the last ~40 years despite dozens of chemotherapy and other therapeutic trials. The prognosis remains invariably fatal. We present here the rationale for a planned study of adding 5-aminolevulinic acid (5-ALA) to the current irradiation of DIPG or DMG: the 5aai regimen. In a series of recent papers, oral 5-ALA was shown to enhance standard therapeutic ionizing irradiation. 5-ALA is currently used in glioblastoma surgery to enable demarcation of overt tumor margins by virtue of selective uptake of 5-ALA by neoplastic cells and selective conversion to protoporphyrin IX (PpIX), which fluoresces after excitation by 410 nm (blue) light. 5-ALA is also useful in treating glioblastomas by virtue of PpIX's transfer of energy to O2 molecules, producing a singlet oxygen that in turn oxidizes intracellular DNA, lipids, and proteins, resulting in selective malignant cell cytotoxicity. This is called photodynamic treatment (PDT). Shallow penetration of light required for PpIX excitation and resultant energy transfer to O2 and cytotoxicity results in the inaccessibility of central structures like the pons or thalamus to sufficient light. The recent demonstration that keV and MeV photons can also excite PpIX and generate singlet O2 allows for reconsideration of 5-ALA PDT for treating DMG and DIPG. 5-ALA has an eminently benign side effect profile in adults and children. A pilot study in DIPG/DMG of slow uptitration of 5-ALA prior to each standard irradiation session-the 5aai regimen-is warranted.
Collapse
|
11
|
Wang Y, Zhao W, Liu X, Guan G, Zhuang M. ARL3 is downregulated and acts as a prognostic biomarker in glioma. J Transl Med 2019; 17:210. [PMID: 31234870 PMCID: PMC6591946 DOI: 10.1186/s12967-019-1914-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Glioma is the most common primary malignant brain tumor in adults with a poor prognosis. ARL3 is a member of the ARF family, and plays a key role in ciliary function and lipid-modified protein trafficking. ARL3 has been reported to be involved in ciliary diseases, in which it affects kidney and photoreceptor development. However, the functional role of ARL3 in cancer remains unknown. In this study, we aimed to explore ARL3 expression and its roles in glioma prognosis. METHODS RT-PCR and immunohistochemistry were performed to examine the expression level of ARL3 in glioma samples. Data from The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA) and Repository for Molecular Brain Neoplasia Data (REMBRANDT) databases were employed to investigate ARL3 expression and its roles in glioma prognosis. A nomogram for predicting 3- or 5-year survival was established using Cox proportional hazards regression. Finally, gene ontology (GO) analysis, gene set enrichment analysis (GSEA), and gene set variation analysis (GSVA) were performed to explore the biological function. RESULTS ARL3 expression was downregulated in glioma, and associated with poor prognosis in glioma patients. The C-indexes, areas under the ROC curve and calibration plots of the nomogram indicated an effective predictive performance for glioma patients. In addition, GO and pathway analyses suggested the involvement of ARL3 in angiogenesis and immune cell infiltration in the microenvironment. CONCLUSIONS Low ARL3 expression predicted poor prognosis and contributed to antiangiogenesis and the proportion of infiltrating immune cells in the GBM microenvironment. Thus, ARL3 may be a prognostic marker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Yulin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, 515041, Guangdong, China
| | - Weijiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xin Liu
- Department of Stomatology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Minghua Zhuang
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, 515041, Guangdong, China.
| |
Collapse
|