1
|
Sloan L, Sen R, Liu C, Doucet M, Blosser L, Katulis L, Kamson DO, Grossman S, Holdhoff M, Redmond KJ, Quon H, Lim M, Eberhart C, Pardoll DM, Hu C, Ganguly S, Kleinberg LR. Radiation immunodynamics in patients with glioblastoma receiving chemoradiation. Front Immunol 2024; 15:1438044. [PMID: 39346903 PMCID: PMC11427284 DOI: 10.3389/fimmu.2024.1438044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/12/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction This is a prospective, rigorous inquiry into the systemic immune effects of standard adjuvant chemoradiotherapy, for WHO grade 4, glioblastoma. The purpose is to identify peripheral immunologic effects never yet reported in key immune populations, including myeloid-derived suppressor cells, which are critical to the immune suppressive environment of glioblastoma. We hypothesize that harmful immune-supportive white blood cells, myeloid derived suppressor cells, expand in response to conventionally fractionated radiotherapy with concurrent temozolomide, essentially promoting systemic immunity similar what is seen in chronic diseases like diabetes and heart disease. Methods 16 patients were enrolled in a single-institution, observational, immune surveillance study where peripheral blood was collected and interrogated by flow cytometry and RNAseq. Tumor tissue from baseline assessment was analyzed with spatial proteomics to link peripheral blood findings to baseline tissue characteristics. Results We identified an increase in myeloid-derived suppressor cells during the final week of a six-week treatment of chemoradiotherapy in peripheral blood of patients that were not alive at two years after diagnosis compared to those who were living. This was also associated with a decrease in CD8+ T lymphocytes that produced IFNγ, the potent anti-tumor cytokine. Discussion These data suggest that, as in chronic inflammatory disease, systemic immunity is impaired following delivery of adjuvant chemoradiotherapy. Finally, baseline investigation of myeloid cells within tumor tissue did not differ between survival groups, indicating immune surveillance of peripheral blood during adjuvant therapy may be a critical missing link to educate our understanding of the immune effects of standard of care therapy for glioblastoma.
Collapse
Affiliation(s)
- Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Rupashree Sen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chunnan Liu
- Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michele Doucet
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lee Blosser
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lisa Katulis
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David O. Kamson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Brain Cancer Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Stuart Grossman
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Brain Cancer Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Matthias Holdhoff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Brain Cancer Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kristin J. Redmond
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Harry Quon
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Charles Eberhart
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Drew M. Pardoll
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chen Hu
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD, United States
| | - Sudipto Ganguly
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Brain Cancer Research Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Beltran-Bless AA, Alshamsan B, Jia J, Lo V, Climans S, Nicholas G, Ng TL. Perception of pneumocystis jirovecii pneumonia (PJP) prophylaxis in glioma patients receiving concurrent temozolomide and radiation- a patient and physician survey. J Neurooncol 2024; 169:625-632. [PMID: 39105955 DOI: 10.1007/s11060-024-04764-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/29/2024] [Indexed: 08/07/2024]
Abstract
PURPOSE Pneumocystis jirovecii pneumonia (PJP) prophylaxis is required by provincial and national drug monographs during glioma treatment using temozolomide (TMZ) concurrently with radiation (TMZ-RT). However, real-world data suggest the potential benefits of PJP prophylaxis may not outweigh its potential harms in this population. METHODS We conducted a single-center patient survey and a national physician survey to explore the role of PJP prophylaxis amongst glioma patients undergoing TMZ-RT. RESULTS 23% (31/133) of physicians and 60% (44/73) of patients completed a survey. The median patient age was 42 (range 20-77); 85% (34/40) had completed adjuvant TMZ. Although only 2.4% (1/41) of patients received PJP prophylaxis, only one person (without PJP prophylaxis) was hospitalized for pneumonia. When presented with hypothetical PJP risks, 13.2% (5/38) of patients were concerned about PJP infection, while 26% (10/38) were concerned about potential side effects from prophylactic antibiotics. Most physicians (77%, 17/22) perceived the evidence for PJP prophylaxis as weak; 58% (11/19) did not routinely prescribe prophylaxis, and 73% (16/22) felt that PJP prophylaxis should be limited to patients with additional risk factors. Over 95% of physicians estimated that the incidence of PJP was < 1% in their last 5 years of practice regardless of PJP prophylaxis. For 73% (16/22) of physicians, to prescribe PJP prophylaxis, the risk of PJP infection needed to be 3-8%. CONCLUSION The current recommendation to routinely prescribe PJP prophylaxis in patients receiving TMZ-RT in the absence of other risk factors warrants reconsideration.
Collapse
Affiliation(s)
- Ana-Alicia Beltran-Bless
- Division of Medical Oncology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | - Bader Alshamsan
- Department of Medicine, College of Medicine, Qassim University, Qassim, 52571, Saudi Arabia
| | - Jason Jia
- Division of Medical Oncology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | - Victor Lo
- Division of Medical Oncology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | - Seth Climans
- Department of Oncology, Western University, London, ON, Canada
| | - Garth Nicholas
- Division of Medical Oncology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
- Ottawa Hospital Research Institute, Ottawa, ON, K1Y 4E9, Canada
| | - Terry L Ng
- Division of Medical Oncology, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada.
- Ottawa Hospital Research Institute, Ottawa, ON, K1Y 4E9, Canada.
- , 501 Smyth Road, Ottawa, Ext, ON, K1H 8L6, 613-737-7700, 70170, Canada.
| |
Collapse
|
3
|
Liu Y, Liu J, Wu X, Jiang E. Risk Factors for Central Nervous System Infections After Craniotomy. J Multidiscip Healthc 2024; 17:3637-3648. [PMID: 39100899 PMCID: PMC11296514 DOI: 10.2147/jmdh.s476125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 08/06/2024] Open
Abstract
The central nervous system (CNS) is less prone to infection owing to protection from the brain-blood barrier. However, craniotomy destroys this protection and increases the risk of infection in the brain of patients who have undergone craniotomy. CNS infection after craniotomy significantly increases the patient's mortality rate and disability. Controlling the occurrence of intracranial infection is very important for post-craniotomy patients. CNS infection after craniotomy is caused by several factors such as preoperative, intraoperative, and post-operative factors. Craniotomy may lead to postsurgical intracranial infection, which is mainly associated with surgery duration, infratentorial (posterior fossa) surgery, cerebrospinal fluid leakage, drainage tube placement, unregulated use of antibiotics, glucocorticoid use, age, diabetes, and other systemic infections. Understanding the risk factors of CNS infection after craniotomy can benefit reducing the incidence of intracranial infectious diseases. This will also provide the necessary guidance and evidence in clinical practice for planning to control intracranial infection in patients with craniotomy.
Collapse
Affiliation(s)
- Yufeng Liu
- Department of Cardiovascular Medicine, Luoyang Central Hospital affiliated to Zhengzhou University, Luoyang, Henan, 471000, People’s Republic of China
| | - Jie Liu
- Department of Cardiovascular Medicine, Luoyang Central Hospital affiliated to Zhengzhou University, Luoyang, Henan, 471000, People’s Republic of China
| | - Xiaoyan Wu
- Department of Cardiovascular Medicine, Luoyang Central Hospital affiliated to Zhengzhou University, Luoyang, Henan, 471000, People’s Republic of China
| | - Enshe Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Henan University, Kaifeng, Henan, 475004, People’s Republic of China
- Institute of Nursing and Health, Henan University, Kaifeng, Henan, 475004, People’s Republic of China
| |
Collapse
|
4
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
5
|
Lim-Fat MJ, Iorgulescu JB, Rahman R, Bhave V, Muzikansky A, Woodward E, Whorral S, Allen M, Touat M, Li X, Xy G, Patel J, Gerstner ER, Kalpathy-Cramer J, Youssef G, Chukwueke U, McFaline-Figueroa JR, Nayak L, Lee EQ, Reardon DA, Beroukhim R, Huang RY, Bi WL, Ligon KL, Wen PY. Clinical and Genomic Predictors of Adverse Events in Newly Diagnosed Glioblastoma. Clin Cancer Res 2024; 30:1327-1337. [PMID: 38252427 DOI: 10.1158/1078-0432.ccr-23-3018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
PURPOSE Adverse clinical events cause significant morbidity in patients with GBM (GBM). We examined whether genomic alterations were associated with AE (AE) in patients with GBM. EXPERIMENTAL DESIGN We identified adults with histologically confirmed IDH-wild-type GBM with targeted next-generation sequencing (OncoPanel) at Dana Farber Cancer Institute from 2013 to 2019. Seizure at presentation, lymphopenia, thromboembolic events, pseudoprogression, and early progression (within 6 months of diagnosis) were identified as AE. The biologic function of genetic variants was categorized as loss-of-function (LoF), no change in function, or gain-of-function (GoF) using a somatic tumor mutation knowledge base (OncoKB) and consensus protein function predictions. Associations between functional genomic alterations and AE were examined using univariate logistic regressions and multivariable regressions adjusted for additional clinical predictors. RESULTS Our study included 470 patients diagnosed with GBM who met the study criteria. We focused on 105 genes that had sequencing data available for ≥ 90% of the patients and were altered in ≥10% of the cohort. Following false-discovery rate (FDR) correction and multivariable adjustment, the TP53, RB1, IGF1R, and DIS3 LoF alterations were associated with lower odds of seizures, while EGFR, SMARCA4, GNA11, BRD4, and TCF3 GoF and SETD2 LoF alterations were associated with higher odds of seizures. For all other AE of interest, no significant associations were found with genomic alterations following FDR correction. CONCLUSIONS Genomic biomarkers based on functional variant analysis of a routine clinical panel may help identify AE in GBM, particularly seizures. Identifying these risk factors could improve the management of patients through better supportive care and consideration of prophylactic therapies.
Collapse
Affiliation(s)
- Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - J Bryan Iorgulescu
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rifaquat Rahman
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Varun Bhave
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alona Muzikansky
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, Massachusetts
| | - Eleanor Woodward
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sydney Whorral
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marie Allen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mehdi Touat
- Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | | | | | - Jay Patel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth R Gerstner
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Jayashree Kalpathy-Cramer
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Gilbert Youssef
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ugonma Chukwueke
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - J Ricardo McFaline-Figueroa
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lakshmi Nayak
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rameen Beroukhim
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Raymond Y Huang
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Wenya Linda Bi
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keith L Ligon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Saeed AM, Bentzen SM, Ahmad H, Pham L, Woodworth GF, Mishra MV. Systematic review and pooled analysis of the impact of treatment-induced lymphopenia on survival of glioblastoma patients. Radiat Oncol 2024; 19:36. [PMID: 38481255 PMCID: PMC10938829 DOI: 10.1186/s13014-023-02393-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/17/2023] [Indexed: 03/17/2024] Open
Abstract
PURPOSE/OBJECTIVE(S) Treatment related lymphopenia is a known toxicity for glioblastoma (GBM) patients and several single-institution studies have linked lymphopenia with poor survival outcomes. We performed a systematic review and pooled analysis to evaluate the association between lymphopenia and overall survival (OS) for GBM patients undergoing chemotherapy and radiation therapy (RT). MATERIALS/METHODS Following PRISMA guidelines, a systematic literature review of the MEDLINE database and abstracts from ASTRO, ASCO, and SNO annual meetings was conducted. A pooled analysis was performed using inverse variance-weighted random effects to generate a pooled estimate of the hazard ratio of association between lymphopenia and OS. RESULTS Ten of 104 identified studies met inclusion criteria, representing 1,718 patients. The lymphopenia cutoff value varied (400-1100 cells/uL) and as well as the timing of its onset. Studies were grouped as time-point (i.e., lymphopenia at approximately 2-months post-RT) or time-range (any lymphopenia occurrence from treatment-start to approximately 2-months post-RT. The mean overall pooled incidence of lymphopenia for all studies was 31.8%, and 11.8% vs. 39.9% for time-point vs. time-range studies, respectively. Lymphopenia was associated with increased risk of death, with a pooled HR of 1.78 (95% CI 1.46-2.17, P < 0.00001) for the time-point studies, and a pooled HR of 1.38 (95% CI 1.24-1.55, P < 0.00001) for the time-point studies. There was no significant heterogeneity between studies. CONCLUSION These results strengthen observations from previous individual single-institution studies and better defines the magnitude of the association between lymphopenia with OS in GBM patients, highlighting lymphopenia as a poor prognostic factor.
Collapse
Affiliation(s)
- Ali M Saeed
- Department of Radiation Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
- Maryland Proton Treatment Center, Baltimore, MD, USA
| | - Søren M Bentzen
- Department of Radiation Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
- Department of Epidemiology and Public Health, Division of Biostatistics and Bioinformatics, University of Maryland School of Medicine, Baltimore, USA
| | - Haroon Ahmad
- Department of Medical Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
| | - Lily Pham
- Department of Medical Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark V Mishra
- Department of Radiation Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, USA.
- Maryland Proton Treatment Center, Baltimore, MD, USA.
| |
Collapse
|
7
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Naumenko VA, Chekhonin VP. The need for paradigm shift: prognostic significance and implications of standard therapy-related systemic immunosuppression in glioblastoma for immunotherapy and oncolytic virotherapy. Front Immunol 2024; 15:1326757. [PMID: 38390330 PMCID: PMC10881776 DOI: 10.3389/fimmu.2024.1326757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Despite significant advances in our knowledge regarding the genetics and molecular biology of gliomas over the past two decades and hundreds of clinical trials, no effective therapeutic approach has been identified for adult patients with newly diagnosed glioblastoma, and overall survival remains dismal. Great hopes are now placed on combination immunotherapy. In clinical trials, immunotherapeutics are generally tested after standard therapy (radiation, temozolomide, and steroid dexamethasone) or concurrently with temozolomide and/or steroids. Only a minor subset of patients with progressive/recurrent glioblastoma have benefited from immunotherapies. In this review, we comprehensively discuss standard therapy-related systemic immunosuppression and lymphopenia, their prognostic significance, and the implications for immunotherapy/oncolytic virotherapy. The effectiveness of immunotherapy and oncolytic virotherapy (viro-immunotherapy) critically depends on the activity of the host immune cells. The absolute counts, ratios, and functional states of different circulating and tumor-infiltrating immune cell subsets determine the net immune fitness of patients with cancer and may have various effects on tumor progression, therapeutic response, and survival outcomes. Although different immunosuppressive mechanisms operate in patients with glioblastoma/gliomas at presentation, the immunological competence of patients may be significantly compromised by standard therapy, exacerbating tumor-related systemic immunosuppression. Standard therapy affects diverse immune cell subsets, including dendritic, CD4+, CD8+, natural killer (NK), NKT, macrophage, neutrophil, and myeloid-derived suppressor cell (MDSC). Systemic immunosuppression and lymphopenia limit the immune system's ability to target glioblastoma. Changes in the standard therapy are required to increase the success of immunotherapies. Steroid use, high neutrophil-to-lymphocyte ratio (NLR), and low post-treatment total lymphocyte count (TLC) are significant prognostic factors for shorter survival in patients with glioblastoma in retrospective studies; however, these clinically relevant variables are rarely reported and correlated with response and survival in immunotherapy studies (e.g., immune checkpoint inhibitors, vaccines, and oncolytic viruses). Our analysis should help in the development of a more rational clinical trial design and decision-making regarding the treatment to potentially improve the efficacy of immunotherapy or oncolytic virotherapy.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Victor A. Naumenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, The Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N.I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
8
|
Das A, Sylvia J, Krishnan G, Panda PK, Subramanyam P, Kumar R, Adhithyan R, Patil S, Sharma D, Jalali R. Impact of intensity-modulated proton therapy in reducing radiation-induced lymphopenia in glioma patients. Neurooncol Adv 2024; 6:vdae088. [PMID: 39045310 PMCID: PMC11263926 DOI: 10.1093/noajnl/vdae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Abstract
Background Current standard management in adult grades 2-4 gliomas includes maximal safe resection followed by adjuvant radiotherapy (RT) and chemotherapy. Radiation-induced lymphopenia (RIL) has been shown to possibly affect treatment outcomes adversely. Proton beam therapy (PBT) may reduce the volume of the normal brain receiving moderate radiation doses, and consequently RIL. Our aim was to evaluate the incidence and severity of RIL during proton beam therapy (PBT). Methods We identified patients with grades 2-4 glioma treated with PBT at our center between January 2019 and December 2021. We evaluated the incidence and severity of RIL from weekly complete blood count (CBC) data collected during PBT and compared it to the patients who were treated with photon-based RT (XRT) at our center during the same time. Results The incidence of any degree of lymphopenia (48% in PBT, vs. 81.2% in XRT, P value = .001) and severe lymphopenia (8% in PBT, vs. 24.6% in XRT, P value = .093) were both significantly lesser in patients who received PBT. Severe RIL in patients receiving PBT was seen in only CNS WHO Gr-4 tumors. Mean whole brain V20GyE and V25GyE inversely correlated to nadir ALC and were both significantly lower with PBT. Patients with lymphopenia during PBT showed a trend toward poorer progression-free survival (P = .053) compared to those with maintained lymphocyte counts. Conclusions Proton therapy seems to have a superior sparing of normal brain to moderate dose radiation than photon-based RT and reduces the incidence of lymphopenia. Glioma patients with lymphopenia possibly have worse outcomes than the ones with maintained lymphocyte counts.
Collapse
Affiliation(s)
- Anindita Das
- Department of Radiation Oncology, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| | - Jacinthlyn Sylvia
- Department of Radiation Oncology, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| | - Ganapathy Krishnan
- Department of Medical Physics, Apollo Proton Cancer Centre, Chennai, India
| | - Pankaj Kumar Panda
- Department of Clinical Research, Apollo Proton Cancer Centre, Chennai, India
| | - Preethi Subramanyam
- Department of Radiation Oncology, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| | - Roopesh Kumar
- Department of Neurosurgery, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| | - Rajendran Adhithyan
- Department of Diagnostic & Intervention Radiology, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| | - Sushama Patil
- Department of Pathology, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| | - Dayananda Sharma
- Department of Medical Physics, Apollo Proton Cancer Centre, Chennai, India
| | - Rakesh Jalali
- Department of Radiation Oncology, Neuro Oncology Cancer Management Team, Apollo Proton Cancer Centre, Chennai, India
| |
Collapse
|
9
|
Kleinberg L, Ye X, Supko J, Stevens GHJ, Shu HK, Mikkelsen T, Lieberman F, Lesser GJ, Lee E, Grossman SA. A multi-site phase I trial of Veliparib with standard radiation and temozolomide in patients with newly diagnosed glioblastoma multiforme (GBM). J Neurooncol 2023; 165:499-507. [PMID: 38015376 DOI: 10.1007/s11060-023-04514-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/16/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE A multi-site Phase I trial was conducted to determine the safety, maximum tolerated dose, and pharmacokinetics (PK) of Veliparib, a Poly (ADP-ribose) polymerase [PARP] enzyme inhibitor, when administered with temozolomide (TMZ) alone and then with temozolomide and radiation (RT) in patients with newly diagnosed glioblastoma. METHODS Given the potential for myelosuppression when a PARP inhibitor is combined with chemotherapy, the first 6 patients accrued were given Veliparib 10 mg bid and TMZ 75 mg/m2/d daily for six weeks. If this was well tolerated, the same doses of Veliparib and TMZ would be tested along with standard radiation with plans to dose escalate the Veliparib in subsequent patient cohorts. Once a maximal tolerated dose was determined, a 78 patient phase II study was planned. Peripheral blood pharmacokinetics were assessed. RESULTS Twenty-four patients were enrolled. In the first 6 patients who received 6 weeks of TMZ with Veliparib only one dose limiting toxicity (DLT) occurred. The next 12 patients received 6 weeks of RT + TMZ + veliparib and 4/12 (33%) had dose limiting hematologic toxicities. As a result, Veliparib was reduced by 50% to 10 mg BID every other week, but again 3/3 patients had dose limiting hematologic toxicities. The trial was then terminated. The mean clearance (± SD) CL/F of Veliparib for the initial dose (27.0 ± 9.0 L/h, n = 16) and at steady-state for 10 mg BID (23.5 ± 10.4 L/h, n = 18) were similar. Accumulation for BID dosing was 56% (± 33%). CONCLUSIONS Although Veliparib 10 mg BID administered with TMZ 75 mg/m2 for six weeks was well tolerated, when this regimen was combined with standard partial brain irradiation it was severely myelosuppressive even when the dose was reduced by 50%. This study again highlights the potential of localized cranial radiotherapy to significantly increase hematologic toxicity of marginally myelosuppressive systemic therapies.
Collapse
Affiliation(s)
- Lawrence Kleinberg
- Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cyberknife, Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, 401 North Broadway, Suite 1440, Baltimore, MD, 21231, USA.
| | - Xiaobu Ye
- Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jeff Supko
- Medicine, Harvard medical School, Boston, MA, USA
| | | | - Hui-Kuo Shu
- Radiation Oncology, Emory University, Atlanta, Georgia
| | - Tom Mikkelsen
- Jeffries Precision Medicine Center, Henry Ford Health, Detroit, MI, USA
| | - Frank Lieberman
- Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Glenn J Lesser
- Department of Internal Medicine, Section on Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Emerson Lee
- Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stuart A Grossman
- Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Greenlund L, Shanley R, Mulford K, Neil EC, Lawrence J, Arnold S, Olin M, Pluhar GE, Venteicher AS, Chen CC, Ferreira C, Reynolds M, Cho LC, Wilke C, Shoo BA, Yuan J, Dusenbery K, Kleinberg LR, Terezakis SA, Sloan L. Comparison of peripheral leukocyte parameters in patients receiving conventionally and hypofractionated radiotherapy schemes for the treatment of newly diagnosed glioblastoma. Front Immunol 2023; 14:1284118. [PMID: 38022656 PMCID: PMC10644882 DOI: 10.3389/fimmu.2023.1284118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Treatment for glioblastomas, aggressive and nearly uniformly fatal brain tumors, provide limited long-term success. Immunosuppression by myeloid cells in both the tumor microenvironment and systemic circulation are believed to contribute to this treatment resistance. Standard multi-modality therapy includes conventionally fractionated radiotherapy over 6 weeks; however, hypofractionated radiotherapy over 3 weeks or less may be appropriate for older patients or populations with poor performance status. Lymphocyte concentration changes have been reported in patients with glioblastoma; however, monocytes are likely a key cell type contributing to immunosuppression in glioblastoma. Peripheral monocyte concentration changes in patients receiving commonly employed radiation fractionation schemes are unknown. Methods To determine the effect of conventionally fractionated and hypofractionated radiotherapy on complete blood cell leukocyte parameters, retrospective longitudinal concentrations were compared prior to, during, and following standard chemoradiation treatment. Results This study is the first to report increased monocyte concentrations and decreased lymphocyte concentrations in patients treated with conventionally fractionated radiotherapy compared to hypofractionated radiotherapy. Discussion Understanding the impact of fractionation on peripheral blood leukocytes is important to inform selection of dose fractionation schemes for patients receiving radiotherapy.
Collapse
Affiliation(s)
- Lindsey Greenlund
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Ryan Shanley
- Clinical and Translational Science Institute, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kellen Mulford
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Elizabeth C. Neil
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Jessica Lawrence
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Susan Arnold
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Michael Olin
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, United States
| | - G. Elizabeth Pluhar
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Andrew S. Venteicher
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Clark C. Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Clara Ferreira
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Margaret Reynolds
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - L. Chinsoo Cho
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Christopher Wilke
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - B. Aika Shoo
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
| | - Jianling Yuan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Kathryn Dusenbery
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Stephanie A. Terezakis
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
11
|
Kleinberg L, Ye X, Supko J, Stevens GHJ, Shu HK, Mikkelsen T, Lieberman F, Lesser G, Lee E, Grossman S. A Multi-Site Phase I Trial of Veliparib with Standard Radiation and Temozolomide in Patients with Newly Diagnosed Glioblastoma Multiforme (GBM). RESEARCH SQUARE 2023:rs.3.rs-3466927. [PMID: 37961385 PMCID: PMC10635324 DOI: 10.21203/rs.3.rs-3466927/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Purpose A multi-site Phase I trial was conducted to determine the safety, maximum tolerated dose, and pharmacokinetics (PK) of Veliparib, a Poly (ADP-ribose) polymerase [PARP] enzyme inhibitor, when administered with temozolomide (TMZ) alone and then with temozolomide and radiation (RT) in patients with newly diagnosed glioblastoma. Methods Given the potential for myelosuppression when a PARP inhibitor is combined with chemotherapy, the first 6 patients accrued were given Veliparib 10 mg bid and TMZ 75 mg/m2/d daily for six weeks. If this was well tolerated, the same doses of Veliparib and TMZ would be tested along with standard radiation with plans to dose escalate the Veliparib in subsequent patient cohorts. Once a maximal tolerated dose was determined, a 78 patient phase II study was planned. Peripheral blood pharmacokinetics were assessed. Results Twenty-four patients were enrolled. In the first 6 patients who received 6 weeks of TMZ with Veliparib only one dose limiting toxicity (DLT) occurred. The next 12 patients received 6 weeks of RT + TMZ + veliparib and 4/12 (33%) had dose limiting hematologic toxicities. As a result, Veliparib was reduced by 50% to 10 mg BID every other week, but again 3/3 patients had dose limiting hematologic toxicities. The trial was then terminated. The mean clearance (± SD) CL/F of Veliparib for the initial dose (27.0 ± 9.0 L/h, n = 16) and at steady-state for 10 mg BID (23.5 ± 10.4 L/h, n = 18) were similar. Accumulation for BID dosing was 56% (± 33%). Conclusions Although Veliparib 10 mg BID administered with TMZ 75 mg/m2 for six weeks was well tolerated, when this regimen was combined with standard partial brain irradiation it was severely myelosuppressive even when the dose was reduced by 50%. This study again highlights the potential of localized cranial radiotherapy to significantly increase hematologic toxicity of marginally myelosuppressive systemic therapies.
Collapse
|
12
|
Khadka S, Druffner SR, Duncan BC, Busada JT. Glucocorticoid regulation of cancer development and progression. Front Endocrinol (Lausanne) 2023; 14:1161768. [PMID: 37143725 PMCID: PMC10151568 DOI: 10.3389/fendo.2023.1161768] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023] Open
Abstract
Glucocorticoids are steroid hormones that regulate a host of cellular and physiological functions. However, they are arguably best known for their potent anti-inflammatory properties. Chronic inflammation is well-known to promote the development and progression of numerous types of cancer, and emerging evidence suggests that glucocorticoid regulation of inflammation affects cancer development. However, the timing, intensity, and duration of glucocorticoid signaling have important but often contradictory effects on cancer development. Moreover, glucocorticoids are widely used in parallel with radiation and chemotherapy to control pain, dyspnea, and swelling, but their use may compromise anti-tumor immunity. This review will explore the effects of glucocorticoids on cancer development and progression with particular focus on pro and anti-tumor immunity.
Collapse
|
13
|
Venkatesulu B, Giridhar P, Pujari L, Chou B, Lee JH, Block AM, Upadhyay R, Welsh JS, Harkenrider MM, Krishnan S, Verma V, En Hsieh C, Pradhan S, Small W, Solanki AA. Lymphocyte sparing normal tissue effects in the clinic (LymphoTEC): A systematic review of dose constraint considerations to mitigate radiation-related lymphopenia in the era of immunotherapy. Radiother Oncol 2022; 177:81-94. [PMID: 36334694 DOI: 10.1016/j.radonc.2022.10.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Radiation-related lymphopenia has been associated with suboptimal tumor control rates leading to inferior survival outcomes. To date, no standardized dose constraints are available to limit radiation dose to resident and circulating lymphocyte populations. We undertook this systemic review of the literature to provide a synopsis of the dosimetric predictors of radiation-related lymphopenia in solid malignancies. METHODOLOGY A systematic literature review of PubMed (National Institutes of Health), Cochrane Central (Cochrane collaboration), and Google Scholar was conducted with the following keywords: "radiation", "lymphopenia", "cancer", "dosimetric predictors" with an inclusion deadline of May 31, 2022. Studies that met prespecified inclusion criteria were designated either Good, Fair, or Poor Quality based on the Newcastle-Ottawa quality assessment. The dosimetric parameters derived from Good Quality studies were tabulated as LymphoTEC dose constraints. Dosimetric parameters derived from Fair and Poor-quality studies were grouped as optional. RESULTS An initial systematic search of the literature yielded 1,632 articles. After screening, a total of 48 studies met inclusion criteria and were divided into the following categories: central nervous system (CNS, 6), thoracic (11), gastrointestinal (26), gynecologic (2), head and neck, breast, and genitourinary (one each) cancers. Lung mean dose, heart mean dose, brain V25, spleen mean dose, estimated dose to immune cells, and bone marrow V10 were among the strongest predictors for severe lymphopenia related to radiotherapy. CONCLUSION Optimizing the delivery of radiation therapy to limit dose to lymphocyte-rich structures may curb the negative oncologic impact of lymphocyte depletion. The dose constraints described herein may be considered for prospective validation and future use in clinical trials to limit risk of radiation-related lymphopenia and possibly improve cancer-associated outcomes.
Collapse
Affiliation(s)
- BhanuPrasad Venkatesulu
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA; Edward Hines Veteran affairs hospital, Chicago, IL, USA.
| | | | - Lincoln Pujari
- Department of Radiation Oncology, Tata memorial center, Varanasi, India
| | - Brian Chou
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA; Edward Hines Veteran affairs hospital, Chicago, IL, USA
| | - Jae Han Lee
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Alec M Block
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA; Edward Hines Veteran affairs hospital, Chicago, IL, USA
| | - Rituraj Upadhyay
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - James S Welsh
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA; Edward Hines Veteran affairs hospital, Chicago, IL, USA
| | - Matthew M Harkenrider
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Vivek Verma
- Department of Radiation Oncology, MD Anderson cancer center, Houston, Texas, USA
| | - Cheng En Hsieh
- Department of Radiation Oncology, Institute for Radiological Research, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan City, Taiwan; Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston and The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Satyajit Pradhan
- Department of Radiation Oncology, Tata memorial center, Varanasi, India
| | - William Small
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA
| | - Abhishek A Solanki
- Loyola University Chicago, Stritch School of Medicine, Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Maywood, IL 60153, USA; Edward Hines Veteran affairs hospital, Chicago, IL, USA
| |
Collapse
|
14
|
Alsayed AR, Al-Dulaimi A, Alkhatib M, Al Maqbali M, Al-Najjar MAA, Al-Rshaidat MMD. A comprehensive clinical guide for Pneumocystis jirovecii pneumonia: a missing therapeutic target in HIV-uninfected patients. Expert Rev Respir Med 2022; 16:1167-1190. [PMID: 36440485 DOI: 10.1080/17476348.2022.2152332] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Pneumocystis jirovecii is an opportunistic, human-specific fungus that causes Pneumocystis pneumonia (PCP). PCP symptoms are nonspecific. A patient with P. jirovecii and another lung infection faces a diagnostic challenge. It may be difficult to determine which of these agents is responsible for the clinical symptoms, preventing effective treatment. Diagnostic and treatment efforts have been made more difficult by the rising frequency with which coronavirus 2019 (COVID-19) and PCP co-occur. AREAS COVERED Herein, we provide a comprehensive review of clinical and pharmacological recommendations along with a literature review of PCP in immunocompromised patients focusing on HIV-uninfected patients. EXPERT OPINION PCP may be masked by identifying co-existing pathogens that are not necessarily responsible for the observed infection. Patients with severe form COVID-19 should be examined for underlying immunodeficiency, and co-infections must be considered as co-infection with P. jirovecii may worsen COVID-19's severity and fatality. PCP should be investigated in patients with PCP risk factors who come with pneumonia and suggestive radiographic symptoms but have not previously received PCP prophylaxis. PCP prophylaxis should be explored in individuals with various conditions that impair the immune system, depending on their PCP risk.
Collapse
Affiliation(s)
- Ahmad R Alsayed
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Abdullah Al-Dulaimi
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, Applied Science Private University, Amman, Jordan
| | - Mohammad Alkhatib
- Department of Experimental Medicine, University of Rome "Tor Vergata", Roma, Italy
| | - Mohammed Al Maqbali
- Department of Nursing Midwifery and Health, Northumbria University, Newcastle-Upon-Tyne, UK
| | - Mohammad A A Al-Najjar
- Department of Pharmaceutical Sciences and Pharmaceutics, Applied Science Private University, Amman, Kingdom of Jordan
| | - Mamoon M D Al-Rshaidat
- Laboratory for Molecular and Microbial Ecology (LaMME), Department of Biological Sciences, School of Sciences, The University of Jordan, Amman, Jordan
| |
Collapse
|
15
|
Wiencke JK, Molinaro AM, Warrier G, Rice T, Clarke J, Taylor JW, Wrensch M, Hansen H, McCoy L, Tang E, Tamaki SJ, Tamaki CM, Nissen E, Bracci P, Salas LA, Koestler DC, Christensen BC, Zhang Z, Kelsey KT. DNA methylation as a pharmacodynamic marker of glucocorticoid response and glioma survival. Nat Commun 2022; 13:5505. [PMID: 36127421 PMCID: PMC9486797 DOI: 10.1038/s41467-022-33215-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/08/2022] [Indexed: 12/15/2022] Open
Abstract
Assessing individual responses to glucocorticoid drug therapies that compromise immune status and affect survival outcomes in neuro-oncology is a great challenge. Here we introduce a blood-based neutrophil dexamethasone methylation index (NDMI) that provides a measure of the epigenetic response of subjects to dexamethasone. This marker outperforms conventional approaches based on leukocyte composition as a marker of glucocorticoid response. The NDMI is associated with low CD4 T cells and the accumulation of monocytic myeloid-derived suppressor cells and also serves as prognostic factor in glioma survival. In a non-glioma population, the NDMI increases with a history of prednisone use. Therefore, it may also be informative in other conditions where glucocorticoids are employed. We conclude that DNA methylation remodeling within the peripheral immune compartment is a rich source of clinically relevant markers of glucocorticoid response.
Collapse
Affiliation(s)
- J K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA.
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Helen Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Emily Tang
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Stan J Tamaki
- Parnassus Flow Cytometry CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Courtney M Tamaki
- Parnassus Flow Cytometry CoLab, University of California San Francisco, San Francisco, CA, USA
| | - Emily Nissen
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Paige Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Ze Zhang
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Karl T Kelsey
- Department of Epidemiology, Brown University, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
16
|
Sansone G, Vivori N, Vivori C, Di Stefano AL, Picca A. Basic premises: searching for new targets and strategies in diffuse gliomas. Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00507-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
17
|
Liljedahl E, Konradsson E, Gustafsson E, Jonsson KF, Olofsson JK, Ceberg C, Redebrandt HN. Long-term anti-tumor effects following both conventional radiotherapy and FLASH in fully immunocompetent animals with glioblastoma. Sci Rep 2022; 12:12285. [PMID: 35853933 PMCID: PMC9296533 DOI: 10.1038/s41598-022-16612-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy can induce an immunological response. One limiting factor is side effects on normal tissue. Using FLASH radiotherapy, side effects could possibly be reduced. The efficacy of FLASH in relation to conventional radiotherapy (CONV-RT) has not been extensively explored in fully immunocompetent animals. Fully immunocompetent Fischer 344 rats were inoculated with NS1 glioblastoma cells subcutaneously or intracranially. Radiotherapy was delivered with FLASH or CONV-RT at 8 Gy × 2 (subcutaneous tumors) and 12.5 Gy × 2 (intracranial tumors). Cured animals were re-challenged in order to explore long-term anti-tumor immunity. Serum analytes and gene expression were explored. The majority of animals with subcutaneous tumors were cured when treated with FLASH or CONV-RT at 8 Gy × 2. Cured animals could reject tumor re-challenge. TIMP-1 in serum was reduced in animals treated with FLASH 8 Gy × 2 compared to control animals. Animals with intracranial tumors survived longer when treated with FLASH or CONV-RT at 12.5 Gy × 2, but cure was not reached. CONV-RT and FLASH were equally effective in fully immunocompetent animals with glioblastoma. Radiotherapy was highly efficient in the subcutaneous setting, leading to cure and long-term immunity in the majority of the animals.
Collapse
Affiliation(s)
- Emma Liljedahl
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Elise Konradsson
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Emma Gustafsson
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Karolina Förnvik Jonsson
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jill K Olofsson
- Department for Geosciences and Natural Resource Management, University of Copenhagen, Copenhagen, Denmark
| | - Crister Ceberg
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Henrietta Nittby Redebrandt
- The Rausing Laboratory, Division of Neurosurgery, Department of Clinical Sciences, Lund University, Lund, Sweden. .,Department of Neurosurgery, Skåne University Hospital, Rausing Laboratory, Lund University, BMC D10, 221 84, Lund, Sweden.
| |
Collapse
|
18
|
Topical vancomycin reduces surgical site infections in patients subjected to craniotomy for primary brain tumor resection: A comprehensive cancer center experience. Clin Neurol Neurosurg 2022; 215:107206. [DOI: 10.1016/j.clineuro.2022.107206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 03/07/2022] [Indexed: 11/23/2022]
|
19
|
Molinaro AM, Wiencke JK, Warrier G, Koestler DC, Chunduru P, Lee JY, Hansen HM, Lee S, Anguiano J, Rice T, Bracci PM, McCoy L, Salas LA, Christensen BC, Wrensch M, Kelsey KT, Taylor JW, Clarke JL. Interactions of Age and Blood Immune Factors and Noninvasive Prediction of Glioma Survival. J Natl Cancer Inst 2022; 114:446-457. [PMID: 34597382 PMCID: PMC8902347 DOI: 10.1093/jnci/djab195] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/30/2021] [Accepted: 09/23/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Tumor-based classification of human glioma portends patient prognosis, but considerable unexplained survival variability remains. Host factors (eg, age) also strongly influence survival times, partly reflecting a compromised immune system. How blood epigenetic measures of immune characteristics and age augment molecular classifications in glioma survival has not been investigated. We assess the prognostic impact of immune cell fractions and epigenetic age in archived blood across glioma molecular subtypes for the first time. METHODS We evaluated immune cell fractions and epigenetic age in archived blood from the University of California San Francisco Adult Glioma Study, which included a training set of 197 patients with IDH-wild type, 1p19q intact, TERT wild type (IDH/1p19q/TERT-WT) glioma, an evaluation set of 350 patients with other subtypes of glioma, and 454 patients without glioma. RESULTS IDH/1p19q/TERT-WT patients had lower lymphocyte fractions (CD4+ T, CD8+ T, natural killer, and B cells) and higher neutrophil fractions than people without glioma. Recursive partitioning analysis delineated 4 statistically significantly different survival groups for patients with IDH/1p19q/TERT-WT based on an interaction between chronological age and 2 blood immune factors, CD4+ T cells, and neutrophils. Median overall survival ranged from 0.76 years (95% confidence interval = 0.55-0.99) for the worst survival group (n = 28) to 9.72 years (95% confidence interval = 6.18 to not available) for the best (n = 33). The recursive partitioning analysis also statistically significantly delineated 4 risk groups in patients with other glioma subtypes. CONCLUSIONS The delineation of different survival groups in the training and evaluation sets based on an interaction between chronological age and blood immune characteristics suggests that common host immune factors among different glioma types may affect survival. The ability of DNA methylation-based markers of immune status to capture diverse, clinically relevant information may facilitate noninvasive, personalized patient evaluation in the neuro-oncology clinic.
Collapse
Affiliation(s)
- Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gayathri Warrier
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Devin C Koestler
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Pranathi Chunduru
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ji Yoon Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Helen M Hansen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Sean Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Joaquin Anguiano
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Terri Rice
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Paige M Bracci
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Lucie McCoy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Brock C Christensen
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Departments of Molecular and Systems Biology and Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Karl T Kelsey
- Departments of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
20
|
Pulmonary toxicity of craniospinal irradiation using helical tomotherapy. Sci Rep 2022; 12:3221. [PMID: 35217707 PMCID: PMC8881492 DOI: 10.1038/s41598-022-07224-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/11/2022] [Indexed: 11/28/2022] Open
Abstract
Craniospinal irradiation using helical tomotherapy (HT-CSI) has advantages in aspects of homogeneous dose distribution. Physicians, however, still have concerns of pulmonary toxicity due to HT-CSI’s relatively large, low-dose irradiated volume from continuous and 360° rotation delivery. In this study, we investigated the pulmonary toxicity of HT-CSI. We retrospectively reviewed 105 patients who received HT-CSI between January 2014 and December 2019. Grade 2 + pulmonary toxicities were evaluated. Intensive systemic treatment was defined as systemic treatment administration before, during, and after HT-CSI. VX Gy was defined as % volume receiving ≥ X Gy. Thirteen patients (12.4%) presented with grade 2 + pulmonary toxicities after HT-CSI. Of these patients, only one experienced grade 2 radiation pneumonitis combined with pembrolizumab-induced pneumonitis. Conversely, pneumonia was observed in 12 patients. Intensive systemic treatment (p = 0.004), immunosuppressive drugs (p = 0.031), and bilateral lung V5 Gy ≥ 65% (p = 0.031) were identified as independent risk factors for pneumonia. The risk factor for pneumonia in pediatric patients were immunosuppressive drugs (p = 0.035) and bilateral lung V5 Gy ≥ 65% (p = 0.047). HT-CSI can be a safe treatment modality with tolerable pulmonary toxicities. Intensive systemic treatment, immunosuppressive drugs, and bilateral lung V5 Gy ≥ 65% were significantly associated with pneumonia. In these patients, close follow-up should be considered for proper management of pneumonia.
Collapse
|
21
|
Picca A, Guyon D, Santonocito OS, Baldini C, Idbaih A, Carpentier A, Naccarato AG, Caccese M, Lombardi G, Di Stefano AL. Innovating Strategies and Tailored Approaches in Neuro-Oncology. Cancers (Basel) 2022; 14:1124. [PMID: 35267432 PMCID: PMC8909701 DOI: 10.3390/cancers14051124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 01/25/2023] Open
Abstract
Diffuse gliomas, the most frequent and aggressive primary central nervous system neoplasms, currently lack effective curative treatments, particularly for cases lacking the favorable prognostic marker IDH mutation. Nonetheless, advances in molecular biology allowed to identify several druggable alterations in a subset of IDH wild-type gliomas, such as NTRK and FGFR-TACC fusions, and BRAF hotspot mutations. Multi-tyrosine kinase inhibitors, such as regorafenib, also showed efficacy in the setting of recurrent glioblastoma. IDH inhibitors are currently in the advanced phase of clinical evaluation for patients with IDH-mutant gliomas. Several immunotherapeutic approaches, such as tumor vaccines or checkpoint inhibitors, failed to improve patients' outcomes. Even so, they may be still beneficial in a subset of them. New methods, such as using pulsed ultrasound to disrupt the blood-brain barrier, gene therapy, and oncolytic virotherapy, are well tolerated and may be included in the therapeutic armamentarium soon.
Collapse
Affiliation(s)
- Alberto Picca
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, AP-HP, Hôpital Universitaire La Pitié Salpêtrière, DMU Neurosciences, 75013 Paris, France; (A.P.); (A.I.)
| | - David Guyon
- Department of Medical Oncology, Gustave Roussy University Hospital, 94800 Villejuif, France;
| | - Orazio Santo Santonocito
- Division of Neurosurgery, Spedali Riuniti di Livorno—USL Toscana Nord-Ovest, 57124 Livorno, Italy;
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy University Hospital, 94800 Villejuif, France;
| | - Ahmed Idbaih
- Institut du Cerveau-Paris Brain Institute-ICM, Sorbonne Université, Inserm, CNRS, AP-HP, Hôpital Universitaire La Pitié Salpêtrière, DMU Neurosciences, 75013 Paris, France; (A.P.); (A.I.)
| | - Alexandre Carpentier
- Service de Neurochirurgie, Hôpital Universitaire La Pitié Salpêtrière, 75013 Paris, France;
| | - Antonio Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, Division of Pathology, University of Pisa, 56100 Pisa, Italy;
- Anatomia Patologica 1, Department of Laboratory Medicine, Pisa University Hospital, 56126 Pisa, Italy
| | - Mario Caccese
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.); (G.L.)
| | - Giuseppe Lombardi
- Department of Oncology, Oncology 1, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy; (M.C.); (G.L.)
| | - Anna Luisa Di Stefano
- Division of Neurosurgery, Spedali Riuniti di Livorno—USL Toscana Nord-Ovest, 57124 Livorno, Italy;
- Department of Neurology, Foch Hospital, 92150 Suresnes, France
| |
Collapse
|
22
|
Abravan A, Salem A, Price G, Faivre-Finn C, van Herk M. Effect of systemic inflammation biomarkers on overall survival after lung cancer radiotherapy: a single-center large-cohort study. Acta Oncol 2022; 61:163-171. [PMID: 34979860 DOI: 10.1080/0284186x.2021.2022201] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Recent studies suggest that immune-related cells can be recruited for anti-tumor functions as well as tumor progression and the interplay between systemic inflammation and local immune response may play a major role in the development and progression of various cancers including lung cancer. Inflammatory markers, such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII) can be used as surrogate biomarkers of host immune status. In this work, associations between neutrophils, lymphocytes, platelets, NLR, PLR, SII and overall survival (OS) are investigated in two cohorts of non-small cell lung cancer (NSCLC) patients treated with fractionated radiotherapy (RT) and stereotactic body radiation therapy (SBRT) and a cohort of small cell lung cancer (SCLC) patients treated with fractionated RT. MATERIAL AND METHODS Data from 2513 lung cancer patients were retrospectively analyzed. Baseline NLR, PLR, and SII (NLR × platelet count) were calculated from full blood test prior to RT initiation. Cox proportional hazards regression analyses were used to evaluate the association between systemic inflammation markers and known clinical factors with OS. RESULTS The two-year OS was 42%, 63%, and 62% in the NSCLC fractionated RT, SBRT, and SCLC cohort. NLR (per 1 unit: hazard ratio [HR]: 1.04, p < 0.05) and SII (per 100 × 109/L: HR: 1.01, p < 0.05) remained the strongest independent factors of OS in multivariable Cox analyses, correcting for clinical factors in early-stage and locally advanced NSCLC and SCLC patients treated with RT. DISCUSSION This single-center large-cohort study suggests that baseline NLR and SII are independent prognostic biomarkers associated with OS in locally advanced and early-stage NSCLC patients treated with either curative-intent fractionated RT or SBRT and SCLC patients treated with curative-intent fractionated RT. External validation is warranted to evaluate the utility of these biomarkers for patients' stratification and adapting new treatment approaches.
Collapse
Affiliation(s)
- A Abravan
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK
| | - A Salem
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK
| | - G Price
- Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK
| | - C Faivre-Finn
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK
| | - M van Herk
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
23
|
Huang Q, Wang D, Yao G, Wang H. Impact of General Factors on Glioma Immunotherapy. J Clin Neurol 2022; 18:3-13. [PMID: 35021271 PMCID: PMC8762502 DOI: 10.3988/jcn.2022.18.1.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 11/17/2022] Open
Abstract
Glioma remains the most common malignant tumor in the brain and is also the most difficult to treat. Immunotherapy achieving long-lasting tumor remission in multiple cancer types has received considerable attention due to its potential to improve the treatment outcomes of patients with glioma. However, clinical trials have not yet demonstrated major improvements in prognoses, which might be attributable to the extrinsic components and intrinsic mechanisms involved in the tumor microenvironment and immune system. It is particularly noteworthy that there is emerging evidence that current routine treatment modalities and the physical and psychological characteristics of patients have different impacts on the efficacy of glioma immunotherapy. This article addresses how these factors interact with the host immune system and tumor microenvironment, and highlights their potential roles in glioma immunotherapy, with the ultimate goal of developing better immunotherapy-based personalized medicine strategies.
Collapse
Affiliation(s)
- Qilin Huang
- Department of Neurosurgery, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Dongmei Wang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Guojie Yao
- Department of Neurosurgery, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China.
| | - Hongxiang Wang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
24
|
Zhang Y, Chen S, Chen H, Chen S, Li Z, Feng E, Li W. Prognostic Value and Risk Factors of Treatment-Related Lymphopenia in Malignant Glioma Patients Treated With Chemoradiotherapy: A Systematic Review and Meta-Analysis. Front Neurol 2022; 12:726561. [PMID: 35058869 PMCID: PMC8764122 DOI: 10.3389/fneur.2021.726561] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
Background: Immunotherapy has shown promising therapeutic efficacy in various cancers but not gliomas. Circulating lymphocytes play critical roles in cancer control and responses to immune checkpoint inhibitors. Treatment-related lymphopenia has been associated with poor survival in patients with various tumors. This meta-analysis evaluated the risk and impact of lymphopenia in patients with glioma. Methods: The PubMed, Embase, Web of Science, and Cochrane Library databases were comprehensively searched. Eligible studies were included if they reported the incidence and risk factors of lymphopenia and the impact of lymphopenia on survival. Stata 16.0 was used for this meta-analysis. Results: A total of 21 studies were included in the final systematic review and 20 were included in the quantitative analysis. The overall incidence of grade III/IV lymphopenia was 31.6% [95% confidence interval (CI), 22.3-40.8%]. Pooled results based on pathology of glioma revealed that the incidence in astrocytoma and astrocytoma oligodendroglioma patients was 20.2% (95% CI:5.9-34.4%), and the incidence in glioblastoma patients was 27.6% (95% CI:16.2-38.9%). Lymphopenia was associated with poor overall survival (hazard ratio, 1.99; 95% CI, 1.74-2.27; P< 0.001) compared to no lymphopenia. Brain receiving radiation dose of 20 or 25 Gy, female sex, older age, lower baseline lymphocyte count, and dexamethasone dose > 2 mg instead of baseline use were risk factors for lymphopenia. Conclusions: Treatment-related lymphopenia was associated with decreased survival in patients with glioma. Optimization of chemoradiation regimens, particularly in patients with concurrent risk factors, can reduce lymphopenia and potentially improve survival in the era of immunotherapy.
Collapse
Affiliation(s)
- Yongchao Zhang
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shichao Chen
- Neurosurgery Department, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hualei Chen
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Shanshan Chen
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Emergency Department, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Enshan Feng
- Neurosurgery Department, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Werlenius K, Stragliotto G, Strandeus M, Blomstrand M, Carén H, Jakola AS, Rydenhag B, Dyregaard D, Dzhandzhugazyan KN, Kirkin AF, Raida MK, Smits A, Kinhult S. A randomized phase II trial of efficacy and safety of the immunotherapy ALECSAT as an adjunct to radiotherapy and temozolomide for newly diagnosed glioblastoma. Neurooncol Adv 2021; 3:vdab156. [PMID: 34765977 PMCID: PMC8577524 DOI: 10.1093/noajnl/vdab156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background There is an urgent need for effective treatments against glioblastoma (GBM). In this trial, we investigated the efficacy and safety of an adoptive cell-based immunotherapy. Methods Patients with newly diagnosed GBM were recruited at 4 study sites in Sweden. The patients were randomized 1:2 to receive either radiotherapy (RT), 60 Gy/30 fractions, with concomitant and adjuvant temozolomide (TMZ) only, or RT and TMZ with the addition of Autologous Lymphoid Effector Cells Specific Against Tumor (ALECSAT) in an open-label phase II trial. The primary endpoint was investigator-assessed progression-free survival (PFS). The secondary endpoints were survival and safety of ALECSAT. Results Sixty-two patients were randomized to either standard of care (SOC) with RT and TMZ alone (n = 22) or SOC with ALECSAT (n = 40). Median age was 57 years (range 38–69), 95% of the patients were in good performance status (WHO 0–1). There was no significant difference between the study arms (SOC vs ALECSAT + SOC) in PFS (7.9 vs 7.8 months; hazard ratio [HR] 1.28; 95% confidence interval [CI] 0.70–2.36; P = .42) or in median overall survival (OS) (18.3 vs 19.2 months; HR 1.16, 95% CI 0.58–2.31; P = .67). The treatment groups were balanced in terms of serious adverse events (52.4% vs 52.5%), but adverse events ≥grade 3 were more common in the experimental arm (81.0% vs 92.5%). Conclusion Addition of ALECSAT immunotherapy to standard treatment with radiochemotherapy was well tolerated but did not improve PFS or OS for patients with newly diagnosed GBM.
Collapse
Affiliation(s)
- Katja Werlenius
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Malin Blomstrand
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helena Carén
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Asgeir S Jakola
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bertil Rydenhag
- Department of Neurosurgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | | | | | | | | | - Anja Smits
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Neuroscience, Neurology, Uppsala University, Uppsala, Sweden
| | - Sara Kinhult
- Department of Clinical Sciences, Lund University, Skane University Hospital, Lund, Sweden
| |
Collapse
|
26
|
Kim K, Gwak HS, Han N, Hong EK, Choi BK, Lee S, Choi S, Park JH, Seok JH, Jeon Y, Cho H, Lee SJ, Lee Y, Nam KT, Song SW. Chimeric Antigen Receptor T Cells With Modified Interleukin-13 Preferentially Recognize IL13Rα2 and Suppress Malignant Glioma: A Preclinical Study. Front Immunol 2021; 12:715000. [PMID: 34819930 PMCID: PMC8606595 DOI: 10.3389/fimmu.2021.715000] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/15/2021] [Indexed: 12/05/2022] Open
Abstract
Background Interleukin-13 receptor α 2 (IL13Rα2) is a promising tumor-directed antigen of malignant glioma (MG). Here, we examine the efficacy and safety of T cells containing a YYB-103 chimeric antigen receptor (CAR) that can preferentially bind to IL13Rα2 on MG cells. Methods IL13 was modified on the extracellular domain by substitution of amino acids with E13K, R66D, S69D, and R109K and stably transfected into human T cells using a retroviral vector. The in vitro efficacy of YYB-103 CAR T cells was tested in cell lines with differing IL13Rα1 and IL13Rα2 expression. The in vivo efficacy of intracerebroventricular (i.c.v.) and intravenous (i.v.) routes of YYB-103 CAR T-cell administration were tested in orthotopic MG mouse models. Immunohistochemical staining of MG was performed using WHO grade 3/4 surgical specimens from 53 patients. IL13Rα2 expression was quantified by H-score calculated from staining intensity and percentage of positive cells. Results Binding affinity assay of YYB-103 verified apparently nil binding to IL13Rα1, which was more selective than previously reported IL13 modification (E13Y). YYB-103 CAR T cells showed selective toxicity toward co-cultured U87MG (IL13Rα1+/IL13Rα2+) cells but not A431 (IL13Rα1+/IL13Rα2-) cells. Consistently, YYB-103 CAR T cells suppressed tumor growth in nude mice receiving orthotopic injection of U87 MG cells. Both i.c.v. and i.v. injections of YYB-103 CAR T cells reduced tumor volume and prolonged overall survival of tumor-bearing mice. The median H-score for IL13Rα2 in patient-derived MG tissue was 5 (mean, 57.5; SD, 87.2; range, 0 to 300). Conclusion This preclinical study demonstrates the efficacy of IL13Rα2-targeted YYB-103 CAR T cells against MG cells. The use of modified IL13 to construct a CAR facilitated the selective targeting of IL13Rα2-expressing MG cells while sparing IL13Rα1-expressing cells. Notably, YYB-103 CAR T cells exhibited effective blood-brain barrier crossing, suggesting compatibility with i.v. administration rather than intracranial injection. Additionally, the high H-score for IL13Rα2 in glioblastoma, especially in conjunction with the poor prognostic markers of wild-type isocitrate dehydrogenase-1 (IDH-1) and unmethylated O6-methyl guanine methyl-transferase (MGMT), could be used to determine the eligibility of patients with recurrent glioblastoma for a future clinical trial of YYB-103 CAR T cells.
Collapse
Affiliation(s)
- Kiwan Kim
- Department of Drug Development I, CellabMED Inc., Seoul, South Korea
| | - Ho-Shin Gwak
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, South Korea
| | - Nayoung Han
- Department of Pathology, Program for Immunotherapy Research, National Cancer Center, Goyang, South Korea
| | - Eun Kyung Hong
- Department of Pathology, Program for Immunotherapy Research, National Cancer Center, Goyang, South Korea
| | - Beom K. Choi
- Biomedicine Production Branch, Program for Immunotherapy Research, National Cancer Center, Goyang, South Korea
| | - Sangeun Lee
- Department of Drug Development I, CellabMED Inc., Seoul, South Korea
| | - Soyoung Choi
- Department of Drug Development I, CellabMED Inc., Seoul, South Korea
| | - Ju-Hwang Park
- Department of Process Development, CellabMED Inc., Seoul, South Korea
| | - Ji-Hye Seok
- Department of Process Development, CellabMED Inc., Seoul, South Korea
| | - Yeongha Jeon
- Department of Drug Development II, CellabMED Inc., Seoul, South Korea
| | - Hyuntae Cho
- Department of Clinical Development, CellabMED Inc., Seoul, South Korea
| | - Song-Jae Lee
- Research Institute, CellabMED Inc., Seoul, South Korea
| | - Yura Lee
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | | |
Collapse
|
27
|
Maddison K, Graves MC, Bowden NA, Fay M, Vilain RE, Faulkner S, Tooney PA. Low tumour-infiltrating lymphocyte density in primary and recurrent glioblastoma. Oncotarget 2021; 12:2177-2187. [PMID: 34676050 PMCID: PMC8522837 DOI: 10.18632/oncotarget.28069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/28/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapies targeting tumour-infiltrating lymphocytes (TILs) that express the immune checkpoint molecule programmed cell death-1 (PD-1) have shown promise in preclinical glioblastoma models but have had limited success in clinical trials. To assess when glioblastoma is most likely to benefit from immune checkpoint inhibitors we determined the density of TILs in primary and recurrent glioblastoma. Thirteen cases of matched primary and recurrent glioblastoma tissue were immunohistochemically labelled for CD3, CD8, CD4 and PD-1, and TIL density assessed. CD3+ TILs were observed in all cases, with the majority of both primary (69.2%) and recurrent (61.5%) tumours having low density of TILs present. CD8+ TILs were observed at higher densities than CD4+ TILs in both tumour groups. PD-1+ TILs were sparse and present in only 25% of primary and 50% of recurrent tumours. Quantitative analysis of TILs demonstrated significantly higher CD8+ TIL density at recurrence (p = 0.040). No difference was observed in CD3+ (p = 0.191), CD4+ (p = 0.607) and PD-1+ (p = 0.070) TIL density between primary and recurrent groups. This study shows that TILs are present at low densities in both primary and recurrent glioblastoma. Furthermore, PD-1+ TILs were frequently absent, which may provide evidence as to why anti-PD-1 immunotherapy trials have been largely unsuccessful in glioblastoma.
Collapse
Affiliation(s)
- Kelsey Maddison
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Drug Repurposing and Medicines Research, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Moira C Graves
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Drug Repurposing and Medicines Research, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nikola A Bowden
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Drug Repurposing and Medicines Research, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Michael Fay
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Drug Repurposing and Medicines Research, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,GenesisCare, Lake Macquarie Private Hospital, Gateshead, NSW, Australia
| | - Ricardo E Vilain
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Cancer Biobank, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Pathology North, Hunter New England Area Health Service, New Lambton Heights, NSW, Australia
| | - Sam Faulkner
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Paul A Tooney
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.,Centre for Drug Repurposing and Medicines Research, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
28
|
Association between treatment-related lymphopenia and survival in glioblastoma patients following postoperative chemoradiotherapy. Strahlenther Onkol 2021; 198:448-457. [PMID: 34617129 PMCID: PMC9038819 DOI: 10.1007/s00066-021-01855-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 08/17/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE Our study investigated the association between treatment-related lymphopenia and overall survival (OS) in a series of glioblastoma (GBM) patients. We also explored clinical and dosimetric predictors of lymphocytes depletion. METHODS Between 2015 and 2019, 64 patients were treated at the same institution with postoperative chemoradiotherapy. Peripheral lymphocyte count (PLC) data and dose-volume histogram parameters were collected. Radiotherapy (RT) schedule consisted in standard total dose of 60 Gy in 30 daily fractions, with concomitant and adjuvant temozolomide (TMZ). Posttreatment acute absolute lymphopenia (nadir AAL) was calculated as a PLC lower than 1.0 × 103/mm3. Acute relative lymphopenia (ARL) was expressed by the nadir-PLC/baseline-PLC ratio < 0.5. Nadir-PLC was the lowest PLC registered between the end of RT and the first month of follow-up. Survival rates were estimated with Kaplan-Meier curves. Clinical and dosimetric variables related to AAL/ARL and OS were identified by univariate and multivariate analyses. RESULTS A total of 57 patients were eligible and included in the analyses. The median PLC was significantly decreased following chemoradiotherapy (2180/mm3 vs 900/mm3). Median OS was 16 months (range 5-55 months), with no significant difference between patients who developed nadir AAL and those who did not (16 months vs 16.5 months; p = 0.304). When considering ARL vs non-ARL, median OS was 14 months vs 26 months (p = 0.013), respectively. In multivariate Cox regression only age, sex, extent of surgery, access to adjuvant chemotherapy and brain D98% were independently associated with OS. CONCLUSION Although iatrogenic immunosuppression could be associated with inferior clinical outcomes, our data show that treatment-related lymphopenia does not adversely affect GBM survival. Prospective studies are required to confirm these findings.
Collapse
|
29
|
Wang K, Tepper JE. Radiation therapy-associated toxicity: Etiology, management, and prevention. CA Cancer J Clin 2021; 71:437-454. [PMID: 34255347 DOI: 10.3322/caac.21689] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/25/2022] Open
Abstract
Radiation therapy (RT) is a curative treatment for many malignancies and provides effective palliation in patients with tumor-related symptoms. However, the biophysical effects of RT are not specific to tumor cells and may produce toxicity due to exposure of surrounding organs and tissues. In this article, the authors review the clinical context, pathophysiology, risk factors, presentation, and management of RT side effects in each human organ system. Ionizing radiation works by producing DNA damage leading to tumor death, but effects on normal tissue may result in acute and/or late toxicity. The manifestation of toxicity depends on both cellular characteristics and affected organs' anatomy and physiology. There is usually a direct relationship between the radiation dose and volume to normal tissues and the risk of toxicity, which has led to guidelines and recommended dose limits for most tissues. Side effects are multifactorial, with contributions from baseline patient characteristics and other oncologic treatments. Technological advances in recent decades have decreased RT toxicity by dramatically improving the ability to deliver RT that maximizes tumor dose and minimizes organ dose. Thus the study of RT-associated toxicity is a complex, core component of radiation oncology training that continues to evolve alongside advances in cancer management. Because RT is used in up to one-half of all patients with cancer, an understanding of its acute and late effects in different organ systems is clinically pertinent to both oncologists and nononcologists.
Collapse
Affiliation(s)
- Kyle Wang
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, Ohio
| | - Joel E Tepper
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
30
|
The Influence of Severe Radiation-Induced Lymphopenia on Overall Survival in Solid Tumors: A Systematic Review and Meta-Analysis. Int J Radiat Oncol Biol Phys 2021; 111:936-948. [PMID: 34329738 DOI: 10.1016/j.ijrobp.2021.07.1695] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/10/2021] [Accepted: 07/19/2021] [Indexed: 11/22/2022]
Abstract
PURPOSE Emerging evidence suggests a detrimental prognostic association between radiation-induced lymphopenia (RIL) and pathologic response, progression-free survival, and overall survival (OS) in patients who undergo radiation therapy for cancer. The aim of this study was to systematically review and meta-analyze the prognostic impact of RIL on OS in patients with solid tumors. METHODS AND MATERIALS PubMed/MEDLINE and Embase were systematically searched. The analysis included intervention and prognostic studies that reported on the prognostic relationship between RIL and survival in patients with solid tumors. An overall pooled adjusted hazard ratio (aHR) was calculated using a random-effects model. Subgroup analyses for different patient-, tumor-, treatment-, and study-related characteristics were performed using meta-regression. RESULTS Pooling of 21 cohorts within 20 eligible studies demonstrated a statistically significant association between OS and grade ≥3 versus grade 0-2 RIL (n = 16; pooled aHR, 1.65; 95% confidence interval [CI], 1.43-1.90) and grade 4 RIL versus grade 0-3 (n = 5; aHR, 1.53; 95% CI, 1.24-1.90). Moderate heterogeneity among aHRs was observed, mostly attributable to overestimated aHRs in 7 studies likely subject to model-overfitting. Subgroup analysis showed significant prognostic impact of grade ≥3 RIL in 4 brain tumor (aHR, 1.63; 95% CI, 1.06-2.51), 4 lung cancer (aHR, 1.52; 95% CI, 1.01-2.29), and 3 pancreatic cancer (aHR, 1.92; 95% CI, 1.10-3.36) cohorts. CONCLUSIONS This meta-analysis demonstrates a significant detrimental prognostic association between grade ≥3 lymphopenia and OS in patients receiving radiation therapy for solid tumors. This finding appears consistent for tumors of the brain, thorax, and upper abdomen and provides an imperative to further elucidate the potential survival benefit of lymphopenia-mitigating strategies.
Collapse
|
31
|
Wielgat P, Wawrusiewicz-Kurylonek N, Czarnomysy R, Rogowski K, Bielawski K, Car H. The Paired Siglecs in Brain Tumours Therapy: The Immunomodulatory Effect of Dexamethasone and Temozolomide in Human Glioma In Vitro Model. Int J Mol Sci 2021; 22:ijms22041791. [PMID: 33670244 PMCID: PMC7916943 DOI: 10.3390/ijms22041791] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The paired sialic acid-binding immunoglobulin like lectins (Siglecs) are characterized by similar cellular distribution and ligand recognition but opposing signalling functions attributed to different intracellular sequences. Since sialic acid—Siglec axis are known to control immune homeostasis, the imbalance between activatory and inhibitory mechanisms of glycan-dependent immune control is considered to promote pathology. The role of sialylation in cancer is described, however, its importance in immune regulation in gliomas is not fully understood. The experimental and clinical observation suggest that dexamethasone (Dex) and temozolomide (TMZ), used in the glioma management, alter the immunity within the tumour microenvironment. Using glioma-microglia/monocytes transwell co-cultures, we investigated modulatory action of Dex/TMZ on paired Siglecs. Based on real-time PCR and flow cytometry, we found changes in SIGLEC genes and their products. These effects were accompanied by altered cytokine profile and immune cells phenotype switching measured by arginases expression. Additionally, the exposure to Dex or TMZ increased the binding of inhibitory Siglec-5 and Siglec-11 fusion proteins to glioma cells. Our study suggests that the therapy-induced modulation of the interplay between sialoglycans and paired Siglecs, dependently on patient’s phenotype, is of particular signification in the immune surveillance in the glioma management and may be useful in glioma patient’s therapy plan verification.
Collapse
Affiliation(s)
- Przemyslaw Wielgat
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Correspondence: ; Tel.: +48-85-7450-647
| | | | - Robert Czarnomysy
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilińskiego 1, 15-089 Bialystok, Poland; (R.C.); (K.B.)
| | - Karol Rogowski
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland;
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilińskiego 1, 15-089 Bialystok, Poland; (R.C.); (K.B.)
| | - Halina Car
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15A, 15-274 Bialystok, Poland;
- Department of Experimental Pharmacology, Medical University of Bialystok, Szpitalna 37, 15-295 Bialystok, Poland;
| |
Collapse
|
32
|
|
33
|
Abravan A, Faivre-Finn C, Kennedy J, McWilliam A, van Herk M. Radiotherapy-Related Lymphopenia Affects Overall Survival in Patients With Lung Cancer. J Thorac Oncol 2020; 15:1624-1635. [PMID: 32553694 DOI: 10.1016/j.jtho.2020.06.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Lymphopenia after radiotherapy has an adverse effect on the patient's outcome. However, the relationship between radiotherapy dose delivery and lymphopenia is not fully understood. This work used image-based data mining to identify anatomical regions where the received dose is correlated with severe lymphopenia. METHODS A total of 901 patients with lung cancer were analyzed. A Cox model was used to assess prognostic factors of overall survival (OS). Two matched groups were defined-patients with lymphopenia of grade 3 or higher and patients without lymphopenia of grade 3-based on tumor volume, baseline lymphocytes, and prescribed dose. Then, data mining was used to identify regions where dose correlates significantly with lymphopenia of grade 3 or higher. For this, dose matrices were aligned using registration of the computed tomography images to one reference patient. Mean dose distributions were obtained for the two groups, and organs of significance were detected. Dosimetric parameters from the identified organs that had the highest correlation with lymphocytes at nadir were selected. Multivariable analysis was conducted for lymphopenia of grade 3 or higher on the full lung cohort, and the model was tested on 305 patients with esophageal cancer. RESULTS Adjusted Cox regression revealed that lymphopenia of grade 3 or higher is an independent factor of OS. The anatomical regions identified were the heart, lung, and thoracic vertebrae. Dosimetric parameters for lymphopenia included thoracic vertebrae V20, mean lung dose, and mean heart dose, which were further validated in the esophageal cancer cohort. CONCLUSIONS We report that severe lymphopenia during radiotherapy is a poor prognostic factor for OS in patients with lung cancer and could be mitigated by minimizing thoracic vertebrae V20, mean lung dose, and mean heart dose to limit the irradiation of stem cells and blood pool.
Collapse
Affiliation(s)
- Azadeh Abravan
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, United Kingdom.
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Jason Kennedy
- Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Alan McWilliam
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Marcel van Herk
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom; Radiotherapy Related Research, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
34
|
Abstract
OPINION STATEMENT Corticosteroids have been essential in the management of brain tumor patients for decades, primarily for the treatment of peritumoral cerebral edema and its associated neurologic deficits. Dexamethasone is the drug of choice with standard practice being administration up to four times per day, however, because of its long biologic half-life and high potency, once or twice a day dosing is likely adequate in patients without elevated intracranial pressure. The length of corticosteroid treatment should be limited to the shortest period of time to minimize the risk of potential toxicities that can significantly affect quality of life, as well as to avoid a possible detrimental impact on survival in high-grade glioma patients and abrogation of the effect of immunotherapy. Agents such as bevacizumab should be considered in patients who are unable to wean completely off of steroids as well as those who have symptomatic edema and are on immunotherapy. Several other agents have been studied without much success. An increased understanding of the complex pathophysiology of peritumoral vasogenic edema is critically needed to discover new agents that are safer and more effective.
Collapse
|
35
|
Alcorn SR, Fiksel J, Wright JL, Elledge CR, Smith TJ, Perng P, Saleemi S, McNutt TR, DeWeese TL, Zeger S. Developing an Improved Statistical Approach for Survival Estimation in Bone Metastases Management: The Bone Metastases Ensemble Trees for Survival (BMETS) Model. Int J Radiat Oncol Biol Phys 2020; 108:554-563. [PMID: 32446952 DOI: 10.1016/j.ijrobp.2020.05.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 04/23/2020] [Accepted: 05/15/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE To determine whether a machine learning approach optimizes survival estimation for patients with symptomatic bone metastases (SBM), we developed the Bone Metastases Ensemble Trees for Survival (BMETS) to predict survival using 27 prognostic covariates. To establish its relative clinical utility, we compared BMETS with 2 simpler Cox regression models used in this setting. METHODS AND MATERIALS For 492 bone sites in 397 patients evaluated for palliative radiation therapy (RT) for SBM from January 2007 to January 2013, data for 27 clinical variables were collected. These covariates and the primary outcome of time from consultation to death were used to build BMETS using random survival forests. We then performed Cox regressions as per 2 validated models: Chow's 3-item (C-3) and Westhoff's 2-item (W-2) tools. Model performance was assessed using cross-validation procedures and measured by time-dependent area under the curve (tAUC) for all 3 models. For temporal validation, a separate data set comprised of 104 bone sites treated in 85 patients in 2018 was used to estimate tAUC from BMETS. RESULTS Median survival was 6.4 months. Variable importance was greatest for performance status, blood cell counts, recent systemic therapy type, and receipt of concurrent nonbone palliative RT. tAUC at 3, 6, and 12 months was 0.83, 0.81, and 0.81, respectively, suggesting excellent discrimination of BMETS across postconsultation time points. BMETS outperformed simpler models at each time, with respective tAUC at each time of 0.78, 0.76, and 0.74 for the C-3 model and 0.80, 0.78, and 0.77 for the W-2 model. For the temporal validation set, respective tAUC was similarly high at 0.86, 0.82, and 0.78. CONCLUSIONS For patients with SBM, BMETS improved survival predictions versus simpler traditional models. Model performance was maintained when applied to a temporal validation set. To facilitate clinical use, we developed a web platform for data entry and display of BMETS-predicted survival probabilities.
Collapse
Affiliation(s)
- Sara R Alcorn
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD.
| | - Jacob Fiksel
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Jean L Wright
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Christen R Elledge
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Thomas J Smith
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Powell Perng
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Sarah Saleemi
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Todd R McNutt
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Theodore L DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Scott Zeger
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
36
|
Herting CJ, Chen Z, Maximov V, Duffy A, Szulzewsky F, Shayakhmetov DM, Hambardzumyan D. Tumour-associated macrophage-derived interleukin-1 mediates glioblastoma-associated cerebral oedema. Brain 2020; 142:3834-3851. [PMID: 31665239 DOI: 10.1093/brain/awz331] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/12/2019] [Accepted: 09/10/2019] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most common and uncompromising primary brain tumour and is characterized by a dismal prognosis despite aggressive treatment regimens. At the cellular level, these tumours are composed of a mixture of neoplastic cells and non-neoplastic cells, including tumour-associated macrophages and endothelial cells. Cerebral oedema is a near-universal occurrence in patients afflicted with glioblastoma and it is almost exclusively managed with the corticosteroid dexamethasone despite significant drawbacks associated with its use. Here, we demonstrate that dexamethasone blocks interleukin-1 production in both bone marrow-derived and brain resident macrophage populations following stimulation with lipopolysaccharide and interferon gamma. Additionally, dexamethasone is shown to inhibit downstream effectors of interleukin-1 signalling in both macrophage populations. Co-culture of bone marrow-derived macrophages with organotypic tumour slices results in an upregulation of interleukin-1 cytokines, an effect that is absent in co-cultured microglia. Genetic ablation of interleukin-1 ligands or receptor in mice bearing RCAS/tv-a-induced platelet-derived growth factor B-overexpressing glioblastoma results in reduced oedema and partial restoration of the integrity of the blood-brain barrier, respectively; similar to results obtained with vascular endothelial growth factor neutralization. We establish that tumours from dexamethasone-treated mice exhibit reduced infiltration of cells of the myeloid and lymphoid compartments, an effect that should be considered during clinical trials for immunotherapy in glioblastoma patients. Additionally, we emphasize that caution should be used when immune profiling and single-cell RNA sequencing data are interpreted from fresh glioblastoma patient samples, as nearly all patients receive dexamethasone after diagnosis. Collectively, this evidence suggests that interleukin-1 signalling inhibition and dexamethasone treatment share therapeutic efficacies and establishes interleukin-1 signalling as an attractive and specific therapeutic target for the management of glioblastoma-associated cerebral oedema.
Collapse
Affiliation(s)
- Cameron J Herting
- Graduate Division of Molecular and Systems Pharmacology, Emory University, Atlanta, GA, USA.,Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA
| | - Zhihong Chen
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Victor Maximov
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA
| | - Alyssa Duffy
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA
| | - Frank Szulzewsky
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Dmitry M Shayakhmetov
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA
| | - Dolores Hambardzumyan
- Aflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta and Emory University Department of Pediatrics, Atlanta, GA, USA.,Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
37
|
Kleinberg L, Sloan L, Grossman S, Lim M. Radiotherapy, Lymphopenia, and Host Immune Capacity in Glioblastoma: A Potentially Actionable Toxicity Associated With Reduced Efficacy of Radiotherapy. Neurosurgery 2020; 85:441-453. [PMID: 31232425 DOI: 10.1093/neuros/nyz198] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 02/24/2019] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is cytotoxic to tumor cells and is therefore a critical component of therapy for many malignancies, including glioblastoma (GBM). We now appreciate the value of the immunomodulatory effects of radiation that may be important to overall therapeutic success in some patients with this primary brain tumor. Although potentially beneficial immune-stimulating properties of radiotherapy treatment have been the focus of recent study, this modality is actually at the same time associated with the depletion of lymphocytes, which are crucial to the defense against neoplastic development and progression. In this review, we describe the association of systemic lymphopenia with poor tumor outcome, present evidence that radiotherapy is an important contributing cause of lymphodepletion, describe the systemic immune context of tumor and brain injury that contributes to immunosuppression, describe other contributing factors to lymphopenia including concomitant medications and treatments, and speculate about the role of the normal physiologic response to brain injury in the immunosuppressive dynamics of GBM. Radiotherapy is one significant and potentially actionable iatrogenic suppressor of immune response that may be limiting the success of therapy in GBM and other tumor types. Altered strategies for radiotherapy more permissive of a vigorous antineoplastic immune response may improve outcome for malignancy.
Collapse
Affiliation(s)
- Lawrence Kleinberg
- Department of Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Lindsey Sloan
- Department of Radiation Oncology and Radiation Molecular Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Stuart Grossman
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
38
|
Hammi A, Paganetti H, Grassberger C. 4D blood flow model for dose calculation to circulating blood and lymphocytes. Phys Med Biol 2020; 65:055008. [PMID: 32119649 PMCID: PMC8268045 DOI: 10.1088/1361-6560/ab6c41] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To better understand how radiotherapy delivery parameters affect the depletion of circulating lymphocytes in patients treated for intra-cranial tumors, we developed a computational human body blood flow model (BFM), that enables to estimate the dose to the circulating blood during the course of fractionated radiation therapy. A hemodynamic cardiovascular system based on human body reference values was developed to distribute the cardiac output to 24 different organs, described by a discrete Markov Chain. For explicit intracranial blood flow modeling, we extracted major cerebral vasculature from MRI data of a patient and complemented them with an extension network of generic vessels in the frontal and occipital lobes to guarantee even overall blood supply to the entire brain volume. An explicit Monte Carlo simulation was implemented to track the propagation of each individual blood particle (BP) through the brain and time-dependent radiation fields, accumulating dose along their trajectories. The cerebral model includes 1050 path lines and explicitly simulates more than 266 000 BP at any given time that are tracked with a time resolution of 10 ms. The entire BFM for the whole body contains 22 178 000 BP, corresponding to 4200 BP per ml of blood. We have used the model to investigate the difference between proton and photon therapy, and the effect of different dose rates and patient characteristics on the dose to the circulating blood pool. The mean dose to the blood pool is estimated to be 0.06 and 0.13 Gy after 30 fractions of proton and photon therapy, respectively, and the highest dose to 1% of blood was found to be 0.19 Gy and 0.34 Gy. The fraction of blood volume receiving any dose after the first fraction is significantly lower for proton therapy, 10.1% compared to 18.4% for the photon treatment plan. 90% of the blood pool will have received dose after the 11th fraction using photon therapy compared to the 21st fraction with proton therapy. Higher dose rates can effectively reduce the fraction of blood irradiated to low doses but increase the amount of blood receiving high doses. Patient characteristics such as blood pressure, gender and age lead to smaller effects than variations in the dose rate. We developed a 4D human BFM including recirculating to estimate the radiation dose to the circulating blood during intracranial treatment and demonstrate its application to proton- versus photon-based delivery, various dose rates and patient characteristics. The radiation dose estimation to the circulating blood provides us better insight into the origins of radiation-induced lymphopenia.
Collapse
Affiliation(s)
- Abdelkhalek Hammi
- Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States of America
| | | | | |
Collapse
|
39
|
Kelly WJ, Gilbert MR. Glucocorticoids and immune checkpoint inhibitors in glioblastoma. J Neurooncol 2020; 151:13-20. [PMID: 32108294 DOI: 10.1007/s11060-020-03439-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 02/14/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Immunotherapy, activation of the immune system to target tumor cells, represents a paradigm shift in the treatment of cancer. Immune checkpoint therapies, which target immunomodulatory molecules expressed on T-lymphocytes, have demonstrated improved survival in a variety of malignancies. However, benefit in glioblastoma, the most common and devastating malignant brain tumor, remains to be seen. With several recent clinical trials failing to show efficacy of immunotherapy, concerns have been raised regarding the impact of glucocorticoid use in this patient population that may impair the ability for immune checkpoint inhibitors to affect a response. METHODS For this article we examined the mechanism by which immune checkpoint inhibitors activate, and glucocorticoids impair, T-lymphocyte function. RESULTS In this context, we review the clinical data of immune checkpoint inhibitors in glioblastoma as well as the impact glucocorticoids have on immune checkpoint inhibitor efficacy. Finally, we highlight key questions that remain in the field, and the potential benefit of further research for central nervous system tumors. CONCLUSION More information on the extent, character and duration of glucocorticoids on patients treated with PD-(L)1 will better inform both clinical management and novel therapeutic development of immunotherapy in patients with CNS malignancies.
Collapse
Affiliation(s)
- William J Kelly
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Building 82, Room 235, 9030 Old Georgetown Road, Bethesda, MD, 20892, USA.
| |
Collapse
|
40
|
Abravan A, Eide HA, Helland Å, Malinen E. Radiotherapy-related lymphopenia in patients with advanced non-small cell lung cancer receiving palliative radiotherapy. Clin Transl Radiat Oncol 2020; 22:15-21. [PMID: 32181373 PMCID: PMC7063172 DOI: 10.1016/j.ctro.2020.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
Abstract
Risk of grade 3 lymphopenia increased with RT dose to the soft tissue and trabecular bone. High baseline CRP/Albumin was negatively associated with overall survival. Risk of lymphopenia may decrease by limiting irradiation field in palliative RT.
Background Lymphopenia during radiotherapy (RT) may have an adverse effect on treatment outcome. The aim of this study is to investigate associations between lymphopenia and RT parameters in patients with advanced lung cancer. Moreover, to investigate the prognostic role of lymphopenia, blood protein levels, and treatment and patient-related factors. Material and Methods Sixty-two advanced stage non-small cell lung cancer (NSCLC) patients were retrospectively analyzed. Blood counts were available prior to, during, and after RT (3Gyx10). For each patient, a thoracic volume of interest (VOI) –including thoracic soft tissue and trabecular bone– was obtained by applying a CT window of −500 to 1200 HU in the planning CT. Dose parameters from thoracic VOI and other regions including lungs and vertebrae were calculated. Association between risk of lymphopenia ≥ G3 (lymphocytes at nadir according to CTCAE v4.0) and therapeutic parameters was investigated using Logistic regression. Relationships between overall survival (OS) and RT dose parameters, baseline blood counts and protein levels, and clinical factors were evaluated using Log-rank and Cox models. Result Mean thoracic RT dose (odds ratio [OR] 1.67; p = 0.04), baseline lymphocytes (OR 0.65; p = 0.01), and corticosteroids use (OR 6.07; p = 0.02) were significantly associated with increased risk of lymphopenia ≥ G3 in multivariable analysis. Worse OS was associated with high mean thoracic RT dose, high CRP/Albumin, large tumor volume and corticosteroids use (p < 0.05, univariate analysis), but not with lymphopenia ≥ G3. CRP/Albumin ratio > 0.12 (hazard ratio [HR] 2.28, p = 0.03) and corticosteroid use (HR 2.52, p = 0.01) were independently associated with worse OS. Conclusion High thoracic RT dose gave a higher risk of lymphopenia ≥ G3; hence limiting dose volume to the thorax may be valuable in preventing severe lymphopenia for patients receiving palliative fractionated RT. Still, lymphopenia ≥ G3 was not associated with worse OS. however, high baseline CRP/Albumin was associated with poorer OS and may carry important information as a prognostic factor of OS in advanced NSCLC receiving palliative RT.
Collapse
Key Words
- C-reactive protein/Albumin
- CRP, C-Reactive Protein
- CRT, Chemo-radiotherapy
- CT, Computed Tomography
- CTCAE, Common Terminology Criteria for Adverse Events
- Corticosteroid
- ECOG, Eastern Cooperative Oncology Group
- GTV, Gross Tumor Volume
- HR, Hazard Ratio
- Hematologic toxicity
- Lung cancer
- Lymphopenia
- NSCLC, Non-Small Cell Lung Cancer
- OR, Odds Ratio
- OS, Overall Survival
- Overall survival
- RT, Radiotherapy
- Radiotherapy
- VOI, Volume of Interest
Collapse
Affiliation(s)
- Azadeh Abravan
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
- Corresponding author at: Department of Medical Physics, Oslo University Hospital, PO Box 4953 Nydalen, N-0424 Oslo, Norway.
| | - Hanne Astrid Eide
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Åslaug Helland
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Dutoit V, Philippin G, Widmer V, Marinari E, Vuilleumier A, Migliorini D, Schaller K, Dietrich PY. Impact of Radiochemotherapy on Immune Cell Subtypes in High-Grade Glioma Patients. Front Oncol 2020; 10:89. [PMID: 32117743 PMCID: PMC7034105 DOI: 10.3389/fonc.2020.00089] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 01/17/2020] [Indexed: 11/23/2022] Open
Abstract
Glioblastoma is a dreadful disease with very poor prognosis, median overall survival being <2 years despite standard-of-care treatment. This has led to the development of alternative strategies, among which immunotherapy is being actively tested. In particular, many clinical trials of therapeutic vaccination using peptides or tumor cells are ongoing. A major issue in implementing therapeutic vaccines in patients with high-grade glioma is that immune responses have to be elicited in the context of immunosuppressive treatments. Indeed, radiotherapy, chemotherapy, and steroids, which are part of the standard of care for patients with glioblastoma, are known to deplete leukocytes. Whether lymphopenia is beneficial or detrimental to elicitation of efficient immune responses is still debated. Here, in order to determine the impact of standard radiochemotherapy on immune cell subsets, we analyzed the phenotype and function of immune populations in 25 patients with high-grade glioma along concomitant radiochemotherapy and adjuvant chemotherapy with temozolomide. Thirteen healthy individuals were studied along the same period. We show that absolute T and B cell counts are reduced upon concomitant radiochemotherapy. Importantly, T cell counts were not restored long-term after discontinuation of treatment. In addition, the percentage of T regulatory cells among CD4 T cells was increased during the same period and was not decreased upon treatment discontinuation. Finally, we show that the ability of T cells to proliferate is transiently reduced after concomitant radiochemotherapy but is restored at the time of adjuvant TMZ cycles. Although not experimentally validated, transient reduction in proliferation associated with strong lymphopenia during radiochemotherapy may suggest that vaccine-induced T cell stimulation would be suboptimal in that period and that therapeutic vaccination should be performed outside radiochemotherapy administration. In addition, strategies aiming at depleting Treg cells should be implemented in future trials.
Collapse
Affiliation(s)
- Valérie Dutoit
- Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland.,Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | - Géraldine Philippin
- Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland.,Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | - Valérie Widmer
- Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland.,Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | - Eliana Marinari
- Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland.,Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | | | - Denis Migliorini
- Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland.,Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | - Karl Schaller
- Department of Clinical Neurosciences, Division of Neurosurgery, Geneva University Hospital, Geneva, Switzerland
| | - Pierre-Yves Dietrich
- Laboratory of Tumor Immunology and Center of Oncology, Geneva University Hospital, Geneva, Switzerland.,Translational Research Center for Oncohematology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland.,Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| |
Collapse
|
42
|
Abstract
Herpes simplex virus 1 (HSV-1) can be responsible for life-threatening HSV encephalitis (HSE). The mortality rate of patients with HSE who do not receive antiviral treatment is 70%, with most survivors suffering from permanent neurological sequelae. The use of intravenous acyclovir together with improved diagnostic technologies such as PCR and magnetic resonance imaging has resulted in a reduction in the mortality rate to close to 20%. However, 70% of surviving patients still do not recover complete neurological functions. Thus, there is an urgent need to develop more effective treatments for a better clinical outcome. It is well recognized that cerebral damage resulting from HSE is caused by viral replication together with an overzealous inflammatory response. Both of these processes constitute potential targets for the development of innovative therapies against HSE. In this review, we discuss recent progress in therapy that may be used to ameliorate the outcome of patients with HSE, with a particular emphasis on immunomodulatory agents. Ideally, the administration of adjunctive immunomodulatory drugs should be initiated during the rise of the inflammatory response, and its duration should be limited in time to reduce undesired effects. This critical time frame should be optimized by the identification of reliable biomarkers of inflammation.
Collapse
|
43
|
Adhikaree J, Moreno-Vicente J, Kaur AP, Jackson AM, Patel PM. Resistance Mechanisms and Barriers to Successful Immunotherapy for Treating Glioblastoma. Cells 2020; 9:E263. [PMID: 31973059 PMCID: PMC7072315 DOI: 10.3390/cells9020263] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is inevitably refractory to surgery and chemoradiation. The hope for immunotherapy has yet to be realised in the treatment of GBM. Immune checkpoint blockade antibodies, particularly those targeting the Programme death 1 (PD-1)/PD-1 ligand (PD-L1) pathway, have improved the prognosis in a range of cancers. However, its use in combination with chemoradiation or as monotherapy has proved unsuccessful in treating GBM. This review focuses on our current knowledge of barriers to immunotherapy success in treating GBM, such as diminished pre-existing anti-tumour immunity represented by low levels of PD-L1 expression, low tumour mutational burden and a severely exhausted T-cell tumour infiltrate. Likewise, systemic T-cell immunosuppression is seen driven by tumoural factors and corticosteroid use. Furthermore, unique anatomical differences with primary intracranial tumours such as the blood-brain barrier, the type of antigen-presenting cells and lymphatic drainage contribute to differences in treatment success compared to extracranial tumours. There are, however, shared characteristics with those known in other tumours such as the immunosuppressive tumour microenvironment. We conclude with a summary of ongoing and future immune combination strategies in GBM, which are representative of the next wave in immuno-oncology therapeutics.
Collapse
Affiliation(s)
- Jason Adhikaree
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| | - Julia Moreno-Vicente
- Antibody and Vaccine Group, Centre for Cancer Immunology, University of Southampton, Southampton General Hospital, Southampton, Hants SO16 6YD, UK;
| | - Aanchal Preet Kaur
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| | - Andrew Mark Jackson
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| | - Poulam M. Patel
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| |
Collapse
|
44
|
Tea MN, Poonnoose SI, Pitson SM. Targeting the Sphingolipid System as a Therapeutic Direction for Glioblastoma. Cancers (Basel) 2020; 12:cancers12010111. [PMID: 31906280 PMCID: PMC7017054 DOI: 10.3390/cancers12010111] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/28/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most commonly diagnosed malignant brain tumor in adults. The prognosis for patients with GBM remains poor and largely unchanged over the last 30 years, due to the limitations of existing therapies. Thus, new therapeutic approaches are desperately required. Sphingolipids are highly enriched in the brain, forming the structural components of cell membranes, and are major lipid constituents of the myelin sheaths of nerve axons, as well as playing critical roles in cell signaling. Indeed, a number of sphingolipids elicit a variety of cellular responses involved in the development and progression of GBM. Here, we discuss the role of sphingolipids in the pathobiology of GBM, and how targeting sphingolipid metabolism has emerged as a promising approach for the treatment of GBM.
Collapse
Affiliation(s)
- Melinda N. Tea
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5001, Australia;
| | - Santosh I. Poonnoose
- Department of Neurosurgery, Flinders Medical Centre, Adelaide, SA 5042, Australia;
| | - Stuart M. Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, UniSA CRI Building, North Tce, Adelaide, SA 5001, Australia;
- Adelaide Medical School and School of Biological Sciences, University of Adelaide, SA 5001, Australia
- Correspondence: ; Tel.: +61-8-8302-7832; Fax: +61-8-8302-9246
| |
Collapse
|
45
|
Rifino N, Rigamonti A, Guida FM, De Nobili G, Spena G, Ferrarese C, Salmaggi A. Lack of development of Pneumocystis jirovecii Pneumonia in a cohort of 103 Italian glioblastoma patients not receiving prophylaxis during post-surgical chemoradiotherapy. J Neurol Sci 2019; 405:116431. [DOI: 10.1016/j.jns.2019.116431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/16/2019] [Accepted: 08/19/2019] [Indexed: 10/26/2022]
|
46
|
Molinaro AM, Taylor JW, Wiencke JK, Wrensch MR. Genetic and molecular epidemiology of adult diffuse glioma. Nat Rev Neurol 2019; 15:405-417. [PMID: 31227792 PMCID: PMC7286557 DOI: 10.1038/s41582-019-0220-2] [Citation(s) in RCA: 429] [Impact Index Per Article: 85.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
The WHO 2007 glioma classification system (based primarily on tumour histology) resulted in considerable interobserver variability and substantial variation in patient survival within grades. Furthermore, few risk factors for glioma were known. Discoveries over the past decade have deepened our understanding of the molecular alterations underlying glioma and have led to the identification of numerous genetic risk factors. The advances in molecular characterization of glioma have reframed our understanding of its biology and led to the development of a new classification system for glioma. The WHO 2016 classification system comprises five glioma subtypes, categorized by both tumour morphology and molecular genetic information, which led to reduced misclassification and improved consistency of outcomes within glioma subtypes. To date, 25 risk loci for glioma have been identified and several rare inherited mutations that might cause glioma in some families have been discovered. This Review focuses on the two dominant trends in glioma science: the characterization of diagnostic and prognostic tumour markers and the identification of genetic and other risk factors. An overview of the many challenges still facing glioma researchers is also included.
Collapse
Affiliation(s)
- Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret R Wrensch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
47
|
Ye LL, Fan XW, Hu CS, He XY, Wang XS, Shen CY, Xu TT, Ying HM. Dosimetry of the brain and hypothalamus predicting acute lymphopenia and the survival of glioma patients with postoperative radiotherapy. Cancer Med 2019; 8:2759-2768. [PMID: 30983159 PMCID: PMC6558490 DOI: 10.1002/cam4.2159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 01/20/2023] Open
Abstract
Background The aim of this study was to investigate dosimetric factors for predicting acute lymphopenia and the survival of glioma patients with postoperative intensity‐modulated radiotherapy (IMRT). Methods A total of 148 glioma patients were reviewed. Acute lymphopenia was defined as a peripheral lymphocyte count (PLC) lower than 1.0 × 109/L during radiotherapy with a normal level at pretreatment. PLCs with the corresponding dates and dose volume histogram parameters were collected. Univariate and multivariate Cox regression analyses were constructed to assess the significance of risk factors associated with lymphopenia and overall survival (OS). Results Sixty‐nine (46.6%) patients developed lymphopenia during radiotherapy. Multivariate analyses revealed that the risk increased with the maximal dose of the hypothalamus (HT Dmax) ≥56 Gy (58.9% vs 28.5%, P = 0.002), minimal dose of the whole brain (WB Dmin) ≥2 Gy (54.3% vs 33.9%, P = 0.006), or mean dose of the WB (WB Dmean) ≥34 Gy (56.0% vs 37.0%, P = 0.022). Patients with older age, high‐grade glioma, development of lymphopenia, high HT Dmax, WB Dmin, and WB Dmean had significantly inferior OS in the multivariate analyses. Conclusions HT Dmax, WB Dmin, and WB Dmean are promising indicators of lymphopenia and the survival of glioma patients undergoing postoperative IMRT. The necessity and feasibility of dosimetric constraints for HT and WB is warranted with further investigation.
Collapse
Affiliation(s)
- Lu-Lu Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Xing-Wen Fan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Chao-Su Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Xia-Yun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Xiao-Shen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Chun-Ying Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Ting-Ting Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| | - Hong-Mei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, P.R China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, P.R China
| |
Collapse
|
48
|
Cenciarini M, Valentino M, Belia S, Sforna L, Rosa P, Ronchetti S, D'Adamo MC, Pessia M. Dexamethasone in Glioblastoma Multiforme Therapy: Mechanisms and Controversies. Front Mol Neurosci 2019; 12:65. [PMID: 30983966 PMCID: PMC6449729 DOI: 10.3389/fnmol.2019.00065] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/26/2019] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant of the glial tumors. The world-wide estimates of new cases and deaths annually are remarkable, making GBM a crucial public health issue. Despite the combination of radical surgery, radio and chemotherapy prognosis is extremely poor (median survival is approximately 1 year). Thus, current therapeutic interventions are highly unsatisfactory. For many years, GBM-induced brain oedema and inflammation have been widely treated with dexamethasone (DEX), a synthetic glucocorticoid (GC). A number of studies have reported that DEX also inhibits GBM cell proliferation and migration. Nevertheless, recent controversial results provided by different laboratories have challenged the widely accepted dogma concerning DEX therapy for GBM. Here, we have reviewed the main clinical features and genetic and epigenetic abnormalities underlying GBM. Finally, we analyzed current notions and concerns related to DEX effects on cerebral oedema, cancer cell proliferation and migration and clinical outcome.
Collapse
Affiliation(s)
- Marta Cenciarini
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Mario Valentino
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Paolo Rosa
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome "Sapienza", Polo Pontino, Latina, Italy
| | - Simona Ronchetti
- Section of Pharmacology, Department of Medicine, University of Perugia School of Medicine, Perugia, Italy
| | - Maria Cristina D'Adamo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Mauro Pessia
- Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia School of Medicine, Perugia, Italy.,Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| |
Collapse
|
49
|
Hui CY, Rudra S, Ma S, Campian JL, Huang J. Impact of overall corticosteroid exposure during chemoradiotherapy on lymphopenia and survival of glioblastoma patients. J Neurooncol 2019; 143:129-136. [PMID: 30864102 DOI: 10.1007/s11060-019-03146-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Corticosteroids are commonly used to alleviate symptoms from cerebral vasogenic edema in glioblastoma (GBM) patients. This study evaluated the impact of overall corticosteroid exposure during chemoradiotherapy (CRT) on acute severe lymphopenia (ASL) and survival outcomes of GBM patients. METHODS GBM patients treated with CRT from 2007 to 2016 were retrospectively analyzed. Overall corticosteroid exposure was estimated as the average daily dexamethasone dose during 6 weeks of CRT. ASL was defined as grade 3 or higher lymphopenia within 3 months of starting CRT. ASL rates, overall survival (OS), and progression-free survival (PFS) were analyzed using Kaplan-Meier method. Multivariable analysis (MVA) was performed using logistic and Cox regression to identify independent predictors of ASL and survival outcomes, respectively. RESULTS Of the 319 eligible patients, the median daily dexamethasone use was 2 mg/day. The high-dose dexamethasone cohort (> 2 mg/day) had significantly higher ASL and worse OS than the low-dose dexamethasone cohort: 3-month ASL of 43.7% versus 19.8% (p < 0.003) and median OS of 12.6 months versus 17.9 months (p < 0.001), respectively. On MVA, higher dexamethasone use was independently associated with higher ASL and worse OS, but not worse PFS. A subset analysis of patients with gross-total resection found that higher dexamethasone use was significantly associated with ASL, but not OS. CONCLUSION Increased corticosteroid use among GBM patients during CRT appears to be an independent risk factor for developing subsequent ASL. Its apparent association with worse OS may be influenced by other confounding factors and would need to be validated through prospective investigations.
Collapse
Affiliation(s)
- Caressa Y Hui
- Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Soumon Rudra
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Sirui Ma
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA
| | - Jian L Campian
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jiayi Huang
- Department of Radiation Oncology, Washington University School of Medicine, 4921 Parkview Place, Campus Box 8224, St. Louis, MO, 63110, USA.
| |
Collapse
|
50
|
Kleinberg L, Lim M. Commentary: Does Stereotactic Radiosurgery Have a Role in the Management of Patients Presenting With 4 or More Brain Metastases? Neurosurgery 2019; 84:567-568. [DOI: 10.1093/neuros/nyy255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 05/15/2018] [Indexed: 11/14/2022] Open
|