1
|
Riyas Mohamed FR, Yaqinuddin A. Epigenetic reprogramming and antitumor immune responses in gliomas: a systematic review. Med Oncol 2025; 42:213. [PMID: 40380049 DOI: 10.1007/s12032-025-02760-y] [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: 02/28/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
Gliomas, particularly glioblastoma, are among the most aggressive and treatment-resistant brain tumors. Their immunosuppressive tumor microenvironment (TME) and intrinsic molecular heterogeneity hinder effective therapeutic responses. Epigenetic dysregulation in gliomas significantly impacts tumor progression and immune evasion, presenting an opportunity for therapeutic intervention. This systematic review evaluates the role of epigenetic reprogramming in modulating antitumor immune responses in gliomas and explores its potential to enhance treatment outcomes. A comprehensive literature search across major databases, adhering to PRISMA guidelines, identified preclinical and clinical studies examining the effects of epigenetic therapies on glioma-associated immune modulation. Inclusion criteria focused on studies involving DNA methylation inhibitors, histone deacetylase inhibitors, chromatin remodelers, and non-coding RNA-based therapies. Key outcomes included immune activation, tumor progression, survival, and TME modulation. Among 22 included studies, epigenetic therapies demonstrated substantial efficacy in reprogramming the glioma immune landscape. DNA methylation inhibitors such as decitabine enhanced antigen presentation and immune recognition, while histone deacetylase inhibitors improved T-cell-mediated cytotoxicity. Non-coding RNA-targeted interventions disrupted immune suppression and facilitated immune cell infiltration. These strategies showed synergistic potential with immune checkpoint inhibitors, leading to tumor growth inhibition and improved survival in preclinical models. Epigenetic therapies hold promise in overcoming glioma-induced immune resistance by modulating immune escape mechanisms and reprogramming the TME. Their integration with existing treatment modalities, including immunotherapy, represents a transformative avenue for glioma management. Further clinical validation is warranted to optimize their therapeutic potential and safety.
Collapse
|
2
|
Lin H, Liu C, Hu A, Zhang D, Yang H, Mao Y. Understanding the immunosuppressive microenvironment of glioma: mechanistic insights and clinical perspectives. J Hematol Oncol 2024; 17:31. [PMID: 38720342 PMCID: PMC11077829 DOI: 10.1186/s13045-024-01544-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma (GBM), the predominant and primary malignant intracranial tumor, poses a formidable challenge due to its immunosuppressive microenvironment, thereby confounding conventional therapeutic interventions. Despite the established treatment regimen comprising surgical intervention, radiotherapy, temozolomide administration, and the exploration of emerging modalities such as immunotherapy and integration of medicine and engineering technology therapy, the efficacy of these approaches remains constrained, resulting in suboptimal prognostic outcomes. In recent years, intensive scrutiny of the inhibitory and immunosuppressive milieu within GBM has underscored the significance of cellular constituents of the GBM microenvironment and their interactions with malignant cells and neurons. Novel immune and targeted therapy strategies have emerged, offering promising avenues for advancing GBM treatment. One pivotal mechanism orchestrating immunosuppression in GBM involves the aggregation of myeloid-derived suppressor cells (MDSCs), glioma-associated macrophage/microglia (GAM), and regulatory T cells (Tregs). Among these, MDSCs, though constituting a minority (4-8%) of CD45+ cells in GBM, play a central component in fostering immune evasion and propelling tumor progression, angiogenesis, invasion, and metastasis. MDSCs deploy intricate immunosuppressive mechanisms that adapt to the dynamic tumor microenvironment (TME). Understanding the interplay between GBM and MDSCs provides a compelling basis for therapeutic interventions. This review seeks to elucidate the immune regulatory mechanisms inherent in the GBM microenvironment, explore existing therapeutic targets, and consolidate recent insights into MDSC induction and their contribution to GBM immunosuppression. Additionally, the review comprehensively surveys ongoing clinical trials and potential treatment strategies, envisioning a future where targeting MDSCs could reshape the immune landscape of GBM. Through the synergistic integration of immunotherapy with other therapeutic modalities, this approach can establish a multidisciplinary, multi-target paradigm, ultimately improving the prognosis and quality of life in patients with GBM.
Collapse
Affiliation(s)
- Hao Lin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Chaxian Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Ankang Hu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- Institute for Translational Brain Research, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China.
- National Center for Neurological Disorders, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai Clinical Medical Center of Neurosurgery, Neurosurgical Institute of Fudan University, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
3
|
Lee SH, Kim SH, Nam TM, Jang JH, Kim KH, Lee YS, Kim MS, Kim MS, Jin SY, Lee M, Lee SH, Kim YZ. Epigenetic Regulation of the Expression of T Cell Stimulatory and Inhibitory Factors by Histone H3 Lysine Modification Enzymes and Its Prognostic Roles in Glioblastoma. J Korean Med Sci 2023; 38:e258. [PMID: 37605497 PMCID: PMC10442499 DOI: 10.3346/jkms.2023.38.e258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 06/11/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND This study aimed to identify the specific T cell co-stimulatory and co-inhibitory factors that play prognostic roles in patients with glioblastoma. Additionally, the unique histone H3 modification enzymes that regulate the expression levels of these specific co-stimulatory and co-inhibitory factors were investigated. METHODS The medical records of 84 patients newly diagnosed with glioblastoma at our institution from January 2006 to December 2020 were retrospectively reviewed. Immunohistochemical (IHC) staining for T cell co-stimulatory factors (CD27, CD28, CD137, OX40, and ICOS), T cell co-inhibitory factors (CTLA4, PD1, PD-L1, TIM3, and CD200R), and histone H3 lysine modification enzymes (MLL4, RIZ, EZH1, NSD2, KDM5c, JMJD1a, UTX, and JMJD5) was performed on archived paraffin-embedded tissues obtained by biopsy or resection. Quantitative real time-polymerase chain reaction (qRT-PCR) was performed for specific factors, which demonstrated causal relationships, in order to validate the findings of the IHC examinations. RESULTS The mean follow-up duration was 27.5 months (range, 4.1-43.5 months). During this period, 76 patients (90.5%) died, and the mean OS was 19.4 months (95% confidence interval, 16.3-20.9 months). Linear positive correlations were observed between the expression levels of CD28 and JMJD1a (R2 linear = 0.982) and those of CD137 and UTX (R2 linear = 1.528). Alternatively, significant negative correlations were observed between the expression levels of CTLA4 and RIZ (R2 linear = -1.746) and those of PD-L1 and EZH1 (R2 linear = -2.118); these relationships were confirmed by qRT-PCR. In the multivariate analysis, increased expression levels of CD28 (P = 0.042), and CD137 (P = 0.009), and decreased expression levels of CTLA4 (P = 0.003), PD-L1 (P = 0.020), and EZH1 (P = 0.040) were significantly associated with longer survival. CONCLUSION These findings suggest that the expression of certain T cell co-stimulatory factors, such as CD28 and CD 137, and co-inhibitory factors, such as CTLA4 and PD-L1 are associated with prognosis of glioblastoma patients.
Collapse
Affiliation(s)
- Sang Hyuk Lee
- Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Seung Hwan Kim
- Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Taek Min Nam
- Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Ji Hwan Jang
- Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Kyu Hong Kim
- Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
- Well Aging Research Center, Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Minseok S Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
- Translational Responsive Medicine Center, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Mee-Seon Kim
- Department of Pathology, School of Dentistry, Kyungpook National University, Kyungpook National University Hospital, Daegu, Korea
| | - Sung Yup Jin
- Department of Anesthesiology and Pain Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Moonok Lee
- Department of Anesthesiology and Pain Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Sung-Hun Lee
- Cancer Research Institute, Clinomics Inc., Suwon, Korea
| | - Young Zoon Kim
- Department of Neurosurgery, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea.
| |
Collapse
|
4
|
Ali H, Harting R, de Vries R, Ali M, Wurdinger T, Best MG. Blood-Based Biomarkers for Glioma in the Context of Gliomagenesis: A Systematic Review. Front Oncol 2021; 11:665235. [PMID: 34150629 PMCID: PMC8211985 DOI: 10.3389/fonc.2021.665235] [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: 02/07/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gliomas are the most common and aggressive tumors of the central nervous system. A robust and widely used blood-based biomarker for glioma has not yet been identified. In recent years, a plethora of new research on blood-based biomarkers for glial tumors has been published. In this review, we question which molecules, including proteins, nucleic acids, circulating cells, and metabolomics, are most promising blood-based biomarkers for glioma diagnosis, prognosis, monitoring and other purposes, and align them to the seminal processes of cancer. METHODS The Pubmed and Embase databases were systematically searched. Biomarkers were categorized in the identified biomolecules and biosources. Biomarker characteristics were assessed using the area under the curve (AUC), accuracy, sensitivity and/or specificity values and the degree of statistical significance among the assessed clinical groups was reported. RESULTS 7,919 references were identified: 3,596 in PubMed and 4,323 in Embase. Following screening of titles, abstracts and availability of full-text, 262 articles were included in the final systematic review. Panels of multiple biomarkers together consistently reached AUCs >0.8 and accuracies >80% for various purposes but especially for diagnostics. The accuracy of single biomarkers, consisting of only one measurement, was far more variable, but single microRNAs and proteins are generally more promising as compared to other biomarker types. CONCLUSION Panels of microRNAs and proteins are most promising biomarkers, while single biomarkers such as GFAP, IL-10 and individual miRNAs also hold promise. It is possible that panels are more accurate once these are involved in different, complementary cancer-related molecular pathways, because not all pathways may be dysregulated in cancer patients. As biomarkers seem to be increasingly dysregulated in patients with short survival, higher tumor grades and more pathological tumor types, it can be hypothesized that more pathways are dysregulated as the degree of malignancy of the glial tumor increases. Despite, none of the biomarkers found in the literature search seem to be currently ready for clinical implementation, and most of the studies report only preliminary application of the identified biomarkers. Hence, large-scale validation of currently identified and potential novel biomarkers to show clinical utility is warranted.
Collapse
Affiliation(s)
- Hamza Ali
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Romée Harting
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, Netherlands
| | - Meedie Ali
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Thomas Wurdinger
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| | - Myron G. Best
- Department of Neurosurgery, Brain Tumor Center Amsterdam, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center and Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
5
|
Gharbavi M, Danafar H, Amani J, Sharafi A. Immuno-informatics analysis and expression of a novel multi-domain antigen as a vaccine candidate against glioblastoma. Int Immunopharmacol 2020; 91:107265. [PMID: 33360829 DOI: 10.1016/j.intimp.2020.107265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/17/2020] [Accepted: 11/27/2020] [Indexed: 12/28/2022]
Abstract
Glioblastoma multiform is the most common of primary malignant brain tumors in adults. Currently, surgical resection of the tumor mass, followed by adjuvant radiotherapy and chemotherapy are standard treatments for glioblastoma multiform but so far are not effective treatments. Thus, the development of a vaccine, as a safe and efficient strategy for prophylactic or therapeutic purposes against glioblastoma multiform is very necessary. The present study aimed to design the multi-domain vaccine for glioblastoma multiform. An in silico approach was used to select the most potent domains of proteins to induce the host's B- and T-cell immune response against glioblastoma multiform. IL-13Rα-2 (amino acid positions 27-144), TNC (amino acid positions 1900-2100), and PTPRZ-1(amino acid positions 731-884) were found to have potent inducible immune responses. So, we considered them for fusing with a linker A(EAAAK)3A to construct the multi-domain recombinant vaccine. The immuno-informatics analysis of the designed recombinant vaccine construct was performed to evaluate its efficacy. Although the designed recombinant vaccine construct did not show allergen property, its antigenicity was estimated at 0.78. The Physico-chemical properties of the recombinant vaccine construct were characterized and revealed the potency of the vaccine candidate. Then its secondary and tertiary structures, mRNA structure, molecular docking, and immune simulation were predicted using bioinformatics tools. Next, the designed recombinant vaccine construct was synthesized, and cloned into the pET28a vector and expressed in E. coli BL21. Besides, the circular dichroism spectroscopy was utilized for the investigation of the secondary structure changes of the recombinant vaccine construct. The results of the verification assessment of the recombinant vaccine construct expression indicated that in silico analysis was relatively accurate, and relatively change occurred on the protein secondary structure. In our future plan, the vaccine candidate that was confirmed by in silico tools should be validated by further in vitro and in vivo experimental studies.
Collapse
Affiliation(s)
- Mahmoud Gharbavi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Danafar
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ali Sharafi
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
6
|
Chen PY, Wu CYJ, Fang JH, Chen HC, Feng LY, Huang CY, Wei KC, Fang JY, Lin CY. Functional Change of Effector Tumor-Infiltrating CCR5 +CD38 +HLA-DR +CD8 + T Cells in Glioma Microenvironment. Front Immunol 2019; 10:2395. [PMID: 31649684 PMCID: PMC6794477 DOI: 10.3389/fimmu.2019.02395] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/24/2019] [Indexed: 11/13/2022] Open
Abstract
Human glioma facilitates an impaired anti-tumor immunity response, including defects in circulation of T lymphocytes. The level of CD8+ T-cell activation acts as an immune regulator associated with disease progression. However, little is known about the characteristics of peripheral and tumor-infiltrating CD8+ T cells in patients with glioma. In this study, we examined the level of CD8+ T-cell activation in a group of 143 patients with glioma and determined that peripheral CD3+ T cells decreased in accordance with disease severity. The patients' peripheral CD8+ T-cell populations were similar to that of healthy donors, and a small amount of CD8+ tumor-infiltrating lymphocytes was identified in glioma tissues. An increase in activated CD8+ T cells, characterized as CD38+HLA-DR+, and their association with disease progression were identified in the patients' peripheral blood and glioma, and shown to display enriched CCR5+ and TNFR2+ expression levels. Ex vivo examination of CD38+HLA-DR+CD8+ T cells indicated that this subset of cells displayed stronger secretion of IFN-γ and IL-2 before and after a 6-h stimulation with phorbol 12-myristate 13-acetate (PMA) and ionomycin (ION) relative to healthy CD38+HLA-DR+CD8+ T cells, indicating the functional feasibility of CD38+HLA-DR+CD8+ T cells. Higher CCL5 protein and mRNA levels were identified in glioma tissues, which was consistent with the immunohistochemistry results revealing both CCL5 and CD38+HLA-DR+CD8+ T cell expression. Patients' CCR5+CD38+HLA-DR+CD8+ T cells were further validated and shown to display increases in CD45RA+CCR7- and T-bet+ accompanied by substantial CD107-a, IFN-γ, and Granzyme B levels in response to glioma cells.
Collapse
Affiliation(s)
- Pin-Yuan Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.,Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jian-He Fang
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Hsiu-Chi Chen
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Li-Ying Feng
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chiung-Yin Huang
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.,School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan.,Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chun-Yen Lin
- School of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
7
|
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: 480] [Impact Index Per Article: 80.0] [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
|
8
|
Malic L, Daoud J, Geissler M, Boutin A, Lukic L, Janta M, Elmanzalawy A, Veres T. Epigenetic subtyping of white blood cells using a thermoplastic elastomer-based microfluidic emulsification device for multiplexed, methylation-specific digital droplet PCR. Analyst 2019; 144:6541-6553. [DOI: 10.1039/c9an01316d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Digital droplet PCR for epigenetic leukocyte subtyping from clinically relevant samples is implemented using a thermoplastic elastomer microfluidic droplet generator as a first step towards an economical, customizable and easily deployable system.
Collapse
Affiliation(s)
- Lidija Malic
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| | - Jamal Daoud
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| | - Matthias Geissler
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| | - Alex Boutin
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| | - Ljuboje Lukic
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| | - Mojra Janta
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| | | | - Teodor Veres
- Life Sciences Division
- National Research Council of Canada
- Boucherville
- Canada
| |
Collapse
|
9
|
Zhai L, Ladomersky E, Lauing KL, Wu M, Scholtens DM, Savoor R, Zhang B, Wu JD, Horbinski C, Lukas RV, Binder DC, Wainwright DA. Commentary: preclinical efficacy of immune-checkpoint monotherapy does not recapitulate corresponding biomarkers-based clinical predictions in glioblastoma by Garg et al. (2017). Oncoimmunology 2018; 8:1548242. [PMID: 30723577 DOI: 10.1080/2162402x.2018.1548242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/29/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022] Open
Abstract
Preclinical modeling and gene expression analyses have yielded distinct observations for the role of immune checkpoint, IDO1, in glioblastoma (GBM). Accordingly, our recent work differs with Garg et al. (2017) with respect to IDO1 among preclinical and bioinformatic GBM datasets. Here, we discuss the methodological differences that affected study interpretation, and potentially, future clinical decision-making for IDO1-targeting approaches against GBM.
Collapse
Affiliation(s)
- Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Meijing Wu
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Denise M Scholtens
- Department of Preventive Medicine-Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rohan Savoor
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bin Zhang
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer D Wu
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rimas V Lukas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - David C Binder
- Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Medicine-Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
10
|
Titus AJ, Gallimore RM, Salas LA, Christensen BC. Cell-type deconvolution from DNA methylation: a review of recent applications. Hum Mol Genet 2018; 26:R216-R224. [PMID: 28977446 PMCID: PMC5886462 DOI: 10.1093/hmg/ddx275] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 07/11/2017] [Indexed: 02/07/2023] Open
Abstract
Recent advances in cell-type deconvolution approaches are adding to our understanding of the biology underlying disease development and progression. DNA methylation (DNAm) can be used as a biomarker of cell types, and through deconvolution approaches, to infer underlying cell type proportions. Cell-type deconvolution algorithms have two main categories: reference-based and reference-free. Reference-based algorithms are supervised methods that determine the underlying composition of cell types within a sample by leveraging differentially methylated regions (DMRs) specific to cell type, identified from DNAm measures of purified cell populations. Reference-free algorithms are unsupervised methods for use when cell-type specific DMRs are not available, allowing scientists to estimate putative cellular proportions or control for potential confounding from cell type. Reference-based deconvolution is typically applied to blood samples and has potentiated our understanding of the relation between immune profiles and disease by allowing estimation of immune cell proportions from archival DNA. Bioinformatic analyses using DNAm to infer immune cell proportions, part of a new field known as Immunomethylomics, provides a new direction for consideration in epigenome wide association studies (EWAS).
Collapse
Affiliation(s)
- Alexander J Titus
- Program in Quantitative Biomedical Sciences.,Department of Epidemiology
| | | | | | - Brock C Christensen
- Department of Epidemiology.,Department of Molecular and Systems Biology.,Department of Community and Family Medicine, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
11
|
Garg AD, Vandenberk L, Koks C, Verschuere T, Boon L, Van Gool SW, Agostinis P. Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci Transl Med 2016; 8:328ra27. [PMID: 26936504 DOI: 10.1126/scitranslmed.aae0105] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The promise of dendritic cell (DC)-based immunotherapy has been established by two decades of translational research. Of the four malignancies most targeted with clinical DC immunotherapy, high-grade glioma (HGG) has shown the highest susceptibility. HGG-induced immunosuppression is a roadblock to immunotherapy, but may be overcome by the application of T helper 1 (T(H)1) immunity-biased, next-generation, DC immunotherapy. To this end, we combined DC immunotherapy with immunogenic cell death (ICD; a modality shown to induce T(H)1 immunity) induced by hypericin-based photodynamic therapy. In an orthotopic HGG mouse model involving prophylactic/curative setups, both biologically and clinically relevant versions of ICD-based DC vaccines provided strong anti-HGG survival benefit. We found that the ability of DC vaccines to elicit HGG rejection was significantly blunted if cancer cell-associated reactive oxygen species and emanating danger signals were blocked either singly or concomitantly, showing hierarchical effect on immunogenicity, or if DCs, DC-associated MyD88 signal, or the adaptive immune system (especially CD8(+) T cells) were depleted. In a curative setting, ICD-based DC vaccines synergized with standard-of-care chemotherapy (temozolomide) to increase survival of HGG-bearing mice by ~300%, resulting in ~50% long-term survivors. Additionally, DC vaccines also induced an immunostimulatory shift in the brain immune contexture from regulatory T cells to T(H)1/cytotoxic T lymphocyte/T(H)17 cells. Analysis of the The Cancer Genome Atlas glioblastoma cohort confirmed that increased intratumor prevalence of T(H)1/cytotoxic T lymphocyte/T(H)17 cells linked genetic signatures was associated with good patient prognosis. Therefore, pending final preclinical checks, ICD-based vaccines can be clinically translated for glioma treatment.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research and Therapy Laboratory, Department of Cellular and Molecular Medicine, Katholieke Universiteit (KU) Leuven, Leuven 3000, Belgium
| | - Lien Vandenberk
- Laboratory of Pediatric Immunology, Department of Microbiology and Immunology, KU Leuven, Leuven 3000, Belgium
| | - Carolien Koks
- Laboratory of Pediatric Immunology, Department of Microbiology and Immunology, KU Leuven, Leuven 3000, Belgium
| | - Tina Verschuere
- Department of Neurosciences, Research Group-Neuroanatomy and Neurosurgery, KU Leuven, Leuven 3000, Belgium
| | - Louis Boon
- EPIRUS Biopharmaceuticals Netherlands BV, 3584 Utrecht, Netherlands
| | - Stefaan W Van Gool
- Laboratory of Pediatric Immunology, Department of Microbiology and Immunology, KU Leuven, Leuven 3000, Belgium.
| | - Patrizia Agostinis
- Cell Death Research and Therapy Laboratory, Department of Cellular and Molecular Medicine, Katholieke Universiteit (KU) Leuven, Leuven 3000, Belgium.
| |
Collapse
|
12
|
Shu Y, Hu Q, Long H, Chang C, Lu Q, Xiao R. Epigenetic Variability of CD4+CD25+ Tregs Contributes to the Pathogenesis of Autoimmune Diseases. Clin Rev Allergy Immunol 2016; 52:260-272. [DOI: 10.1007/s12016-016-8590-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
13
|
The Ratio of Regulatory (FOXP3+) to Total (CD3+) T Cells Determined by Epigenetic Cell Counting and Cardiovascular Disease Risk: A Prospective Case-cohort Study in Non-diabetics. EBioMedicine 2016; 11:151-156. [PMID: 27499494 PMCID: PMC5049920 DOI: 10.1016/j.ebiom.2016.07.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/19/2016] [Accepted: 07/29/2016] [Indexed: 01/12/2023] Open
Abstract
Background Experimental and clinical evidence indicate that inflammatory processes in atherogenesis and the development of cardiovascular complications are promoted by a loss of regulatory T cell (Treg)-mediated immunological tolerance to plaque antigens. Yet, the association between alterations of systemic Treg frequency and cardiovascular disease incidence remains uncertain. Methods A nested case-cohort study was conducted within the European Prospective Investigation into Cancer and Nutrition (EPIC)-Heidelberg, comprising a random subcohort (n = 778) and primary cases of myocardial infarction (MI, n = 276) and ischemic stroke (n = 151). Pre-diagnostic FOXP3 + Treg and total CD3 + T-lymphocyte (tTL) frequencies in blood were measured by epigenetic-based, quantitative real-time PCR-assisted cell counting. Results Multivariate, Prentice-weighted Cox regression analyses revealed that lower Treg/tTL ratios were not associated with the risk of either MI (lowest vs. highest sex-specific quartile; hazard ratio: 0.72, 95% confidence interval: 0.46 to 1.13; Ptrend = 0.51) or stroke (HR: 0.90, 95% CI: 0.51 to 1.60; Ptrend = 0.78). There were no correlations of Treg/tTL ratios with C-reactive protein, HbA1c, and various lipid parameters. Conclusions Among middle-aged adults from the general population, imbalances in the relative frequency of Tregs within the total T cell compartment do not confer an increased risk of MI or stroke. We studied if peripheral immune tolerance, as reflected by regulatory (FOXP3+) to total (CD3+) T cells, relates to CVD risk. Epigenetic-based, qPCR assisted cell counting was used to quantify T cell subsets in long-term stored buffy coat samples. Lower Treg-mediated immune tolerance does not confer an increased risk of major CVD events.
Inflammation in the arterial intima plays a central role in atherosclerotic cardiovascular disease and may develop owing to autoimmune-like responses targeted against plaque antigens. While the ratio between regulatory T cells (Tregs) and effector T cells is thought to control such immune response outcomes and tolerance within the T cell compartment, we found no association with incidence of major CVD events. These findings imply that reduced systemic Treg frequencies observed in CVD patients follow rather than precede disease manifestation and that Treg variation within a physiological range may not – as previously reported - constitute a pre-disposing risk factor for CVD.
Collapse
|
14
|
Understanding the Role of the Immune System in the Development of Cancer: New Opportunities for Population-Based Research. Cancer Epidemiol Biomarkers Prev 2015; 24:1811-9. [DOI: 10.1158/1055-9965.epi-15-0681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/16/2015] [Indexed: 11/16/2022] Open
|
15
|
Barth SD, Schulze JJ, Kühn T, Raschke E, Hüsing A, Johnson T, Kaaks R, Olek S. Treg-Mediated Immune Tolerance and the Risk of Solid Cancers: Findings From EPIC-Heidelberg. J Natl Cancer Inst 2015; 107:djv224. [PMID: 26298011 DOI: 10.1093/jnci/djv224] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 07/23/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Laboratory-based, mechanistic, and prognosis studies suggest that a shift from antitumor immunity towards tumor-immune tolerance plays a major role in carcinogenesis. However, prospective epidemiological studies on the consequences of differing immune tolerance levels prior to clinical manifestation are missing. METHODS A case-cohort study embedded in EPIC-Heidelberg was conducted comprising incident cases of breast (n = 399), colorectal (n = 185), lung (n = 149), and prostate (n = 378) cancer, which occurred during 6.6 years of follow-up, and a subcohort (n = 807). Foxp3+ regulatory T-lymphocytes and CD3+ T-lymphocytes were measured by quantitative polymerase chain reaction-based DNA methylation analysis in prediagnostic leukocyte samples. Hazard ratios (HRs) for associations of cancer risk with the ratio of both parameters, the "cellular ratio of immune tolerance" (ImmunoCRIT), were estimated using Cox regression models. All statistical tests were two-sided. RESULTS ImmunoCRIT values were positively associated with the risk of lung (highest vs lowest tertile, HR = 1.98, 95% confidence interval = 1.06 to 3.69, P trend = .0263) and colorectal cancer (HR = 1.59, 95% CI = 0.99 to 2.54, P trend = .0069) after multivariable adjustment, but not with prostate cancer risk. Regarding breast cancer significant heterogeneity by estrogen receptor (ER) status was observed (P heterogeneity = .02), and the ImmunoCRIT was associated with the risk of ER-negative breast cancer (HR = 3.34, 95% CI = 1.52 to 7.35, P trend ≤ .001), but not ER-positive breast cancer. CONCLUSION The present study indicates that increased peripheral immune tolerance may be an independent risk factor for lung, colorectal, and ER-negative breast cancer, whereas its role on the development of prostate and ER-positive breast tumors remains uncertain.
Collapse
Affiliation(s)
- Sebastian Dietmar Barth
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Janika Josephine Schulze
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Tilman Kühn
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Eva Raschke
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Anika Hüsing
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Theron Johnson
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Rudolf Kaaks
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| | - Sven Olek
- Affiliations of authors: Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany (SDB, TK, AH, TJ, RK); Ivana Türbachova Labor für Epigenetik, Epiontis GmbH, Berlin, Germany (JJS, ER, SO)
| |
Collapse
|
16
|
Differential gene methylation in paired glioblastomas suggests a role of immune response pathways in tumor progression. J Neurooncol 2015. [DOI: 10.1007/s11060-015-1869-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
17
|
Wiencke JK, Bracci PM, Hsuang G, Zheng S, Hansen H, Wrensch MR, Rice T, Eliot M, Kelsey KT. A comparison of DNA methylation specific droplet digital PCR (ddPCR) and real time qPCR with flow cytometry in characterizing human T cells in peripheral blood. Epigenetics 2015; 9:1360-5. [PMID: 25437051 DOI: 10.4161/15592294.2014.967589] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Quantitating the copy number of demethylated CpG promoter sites of the CD3Z gene can be used to estimate the numbers and proportions of T cells in human blood and tissue. Quantitative methylation specific PCR (qPCR) is useful for studying T cells but requires extensive calibration and is imprecise at low copy numbers. Here we compared the performance of a new digital PCR platform (droplet digital PCR or ddPCR) to qPCR using bisulfite converted DNA from 157 blood specimens obtained from ambulatory care controls and patients with primary glioma. We compared both ddPCR and qPCR with conventional flow cytometry (FACS) evaluation of CD3 positive T cells. Repeated measures on the same blood sample revealed ddPCR to be less variable than qPCR. Both qPCR and ddPCR correlated significantly with FACS evaluation of peripheral blood CD3 counts and CD3/total leukocyte values. However, statistical measures of agreement showed that linear concordance was stronger for ddPCR than for qPCR and the absolute values were closer to FACS for ddPCR. Both qPCR and ddPCR could distinguish clinically significant differences in T cell proportions and performed similarly to FACS. Given the higher precision, greater accuracy, and technical simplicity of ddPCR, this approach appears to be a superior DNA methylation based method than conventional qPCR for the assessment of T cells.
Collapse
Affiliation(s)
- John K Wiencke
- a Department of Neurological Surgery ; University of California, San Francisco ; San Francisco , CA USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Liquid biopsies in patients with diffuse glioma. Acta Neuropathol 2015; 129:849-65. [PMID: 25720744 PMCID: PMC4436687 DOI: 10.1007/s00401-015-1399-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 12/18/2022]
Abstract
Diffuse gliomas are the most common malignant primary tumors of the central nervous system. Like other neoplasms, these gliomas release molecular information into the circulation. Tumor-derived biomarkers include proteins, nucleic acids, and tumor-derived extracellular vesicles that accumulate in plasma, serum, blood platelets, urine and/or cerebrospinal fluid. Recently, also circulating tumor cells have been identified in the blood of glioma patients. Circulating molecules, vesicles, platelets, and cells may be useful as easily accessible diagnostic, prognostic and/or predictive biomarkers to guide patient management. Thereby, this approach may help to circumvent problems related to tumor heterogeneity and sampling error at the time of diagnosis. Also, liquid biopsies may allow for serial monitoring of treatment responses and of changes in the molecular characteristics of gliomas over time. In this review, we summarize the literature on blood-based biomarkers and their potential value for improving the management of patients with a diffuse glioma. Incorporation of the study of circulating molecular biomarkers in clinical trials is essential for further assessment of the potential of liquid biopsies in this context.
Collapse
|
19
|
Cai N, Chang S, Li Y, Li Q, Hu J, Liang J, Song L, Kretzschmar W, Gan X, Nicod J, Rivera M, Deng H, Du B, Li K, Sang W, Gao J, Gao S, Ha B, Ho HY, Hu C, Hu J, Hu Z, Huang G, Jiang G, Jiang T, Jin W, Li G, Li K, Li Y, Li Y, Li Y, Lin YT, Liu L, Liu T, Liu Y, Liu Y, Lu Y, Lv L, Meng H, Qian P, Sang H, Shen J, Shi J, Sun J, Tao M, Wang G, Wang G, Wang J, Wang L, Wang X, Wang X, Yang H, Yang L, Yin Y, Zhang J, Zhang K, Sun N, Zhang W, Zhang X, Zhang Z, Zhong H, Breen G, Wang J, Marchini J, Chen Y, Xu Q, Xu X, Mott R, Huang GJ, Kendler K, Flint J. Molecular signatures of major depression. Curr Biol 2015; 25:1146-56. [PMID: 25913401 PMCID: PMC4425463 DOI: 10.1016/j.cub.2015.03.008] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/03/2015] [Accepted: 03/06/2015] [Indexed: 02/05/2023]
Abstract
Adversity, particularly in early life, can cause illness. Clues to the responsible mechanisms may lie with the discovery of molecular signatures of stress, some of which include alterations to an individual’s somatic genome. Here, using genome sequences from 11,670 women, we observed a highly significant association between a stress-related disease, major depression, and the amount of mtDNA (p = 9.00 × 10−42, odds ratio 1.33 [95% confidence interval [CI] = 1.29–1.37]) and telomere length (p = 2.84 × 10−14, odds ratio 0.85 [95% CI = 0.81–0.89]). While both telomere length and mtDNA amount were associated with adverse life events, conditional regression analyses showed the molecular changes were contingent on the depressed state. We tested this hypothesis with experiments in mice, demonstrating that stress causes both molecular changes, which are partly reversible and can be elicited by the administration of corticosterone. Together, these results demonstrate that changes in the amount of mtDNA and telomere length are consequences of stress and entering a depressed state. These findings identify increased amounts of mtDNA as a molecular marker of MD and have important implications for understanding how stress causes the disease. Amount of mtDNA is increased, and telomeric DNA is shortened in major depression Both changes can be induced with stress but are contingent on the depressed state Changes are tissue specific and in part due to glucocorticoid secretion Changes are in part reversible and represent switches in metabolic strategy
Collapse
Affiliation(s)
- Na Cai
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK
| | - Simon Chang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan, ROC
| | - Yihan Li
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK
| | - Qibin Li
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Jingchu Hu
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Jieqin Liang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Li Song
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Warren Kretzschmar
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK
| | - Xiangchao Gan
- Department of Comparative Developmental Genetics, Max Planck Institute for Plant Breeding Research, Carl-von-Linne-Weg 10, Cologne 50829, Germany
| | - Jerome Nicod
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK
| | - Margarita Rivera
- Centro de Investigacion Medica en Red de Salud Mental, CIBERSAM-University of Granada, Granada, Spain; MRC SGDP Centre, Institute of Psychiatry at King's College, De Crespigny Park, London SE5 8AF, UK; National Institute for Health Research, Biomedical Research Centre for Mental Health, Institute of Psychiatry at King's College, De Crespigny Park, London SE5 8AF, UK
| | - Hong Deng
- Mental Health Center of West China Hospital of Sichuan University, No. 28 South Dianxin Street, Wuhou District, Chengdu, Sichuan 610000, China
| | - Bo Du
- Hebei Mental Health Center, No. 572 Dongfeng Road, Baoding, Hebei 71000, China
| | - Keqing Li
- Hebei Mental Health Center, No. 572 Dongfeng Road, Baoding, Hebei 71000, China
| | - Wenhu Sang
- Hebei Mental Health Center, No. 572 Dongfeng Road, Baoding, Hebei 71000, China
| | - Jingfang Gao
- Zhejiang Traditional Chinese Medical Hospital, No. 54 Youdian Road, Hangzhou, Zhejiang 310000, China
| | - Shugui Gao
- Ningbo Kang Ning Hospital, No. 1 Zhuangyu Road, Zhenhai District, Ningbo, Zhejiang 315000, China
| | - Baowei Ha
- Liaocheng No. 4 Hospital, No. 47 North Huayuan Road, Liaocheng, Shandong 252000, China
| | - Hung-Yao Ho
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan, ROC
| | - Chunmei Hu
- No. 3 Hospital of Heilongjiang Province, No. 135 Jiaotong Road, Beian, Heilongjiang 164000, China
| | - Jian Hu
- Harbin Medical University, No. 23 Youzheng Street, Nangang District, Haerbin, Heilongjiang 150000, China
| | - Zhenfei Hu
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Guoping Huang
- Sichuan Mental Health Center, No. 190, East Jiannan Road, Mianyang, Sichuan 621000, China
| | - Guoqing Jiang
- Chongqing Mental Health Center, No. 102 Jinzishan, Jiangbei District, Chongqing, Chongqing 404100, China
| | - Tao Jiang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Wei Jin
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Gongying Li
- Mental Health Institute of Jining Medical College, Dai Zhuang, Bei Jiao, Jining, Shandong 272000, China
| | - Kan Li
- Mental Hospital of Jiangxi Province, No. 43 Shangfang Road, Nanchang, Jiangxi 330000, China
| | - Yi Li
- Wuhan Mental Health Center, No. 70, Youyi Road, Wuhan, Hubei 430000, China
| | - Yingrui Li
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Youhui Li
- No. 1 Hospital of Zhengzhou University, No. 1 East Jianshe Road, Zhengzhou, Henan 450000, China
| | - Yu-Ting Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan, ROC
| | - Lanfen Liu
- Shandong Mental Health Center, No. 49 East Wenhua Road, Jinan, Shandong 250000, China
| | - Tiebang Liu
- Shenzhen Key Lab for Psychological Healthcare, Kangning Hospital, No. 1080, Cuizhu Street, Luohu District, Shenzhen, Guangdong 518000, China
| | - Ying Liu
- The First Hospital of China Medical University, No. 155 North Nanjing Street, Heping District, Shenyang, Liaoning 110001, China
| | - Yuan Liu
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Yao Lu
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Luxian Lv
- Psychiatric Hospital of Henan Province, No. 388 Middle Jianshe Road, Xinxiang, Henan 453000, China
| | - Huaqing Meng
- No. 1 Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, Chongqing 400016, China
| | - Puyi Qian
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Hong Sang
- Changchun Mental Hospital, No. 4596 Beihuan Road, Changchun, Jilin 130000, China
| | - Jianhua Shen
- Tianjin Anding Hospital, No. 13 Liulin Road, Hexi District, Tianjin, Tianjin 300000, China
| | - Jianguo Shi
- Xian Mental Health Center, No. 15 Yanyin Road, New Qujiang District, Xian, Shaanxi 710000, China
| | - Jing Sun
- Brain Hospital of Nanjing Medical University, No. 264 Guangzhou Road, Nanjing, Jiangsu 210000, China
| | - Ming Tao
- Second Affiliated Hospital of Zhejiang Chinese Medical University, No. 318 Chaowang Road, Hangzhou, Zhejiang 310000, China
| | - Gang Wang
- Beijing Anding Hospital of Capital University of Medical Sciences, No. 5 Ankang Hutong, Deshengmen wai, Xicheng District, Beijing, Beijing 100000, China
| | - Guangbiao Wang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Jian Wang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Linmao Wang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Xueyi Wang
- First Hospital of Hebei Medical University, No. 89 Donggang Road, Shijiazhuang, Hebei 50000, China
| | - Xumei Wang
- ShengJing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, Liaoning 110001, China
| | - Huanming Yang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Lijun Yang
- Jilin Brain Hospital, No. 98 West Zhongyang Road, Siping, Jilin 136000, China
| | - Ye Yin
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Jinbei Zhang
- No. 3 Hospital of Sun Yat-sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, Guangdong 510000, China
| | - Kerang Zhang
- No. 1 Hospital of Shanxi Medical University, No. 85 South Jiefang Road, Taiyuan, Shanxi 30000, China
| | - Ning Sun
- No. 1 Hospital of Shanxi Medical University, No. 85 South Jiefang Road, Taiyuan, Shanxi 30000, China
| | - Wei Zhang
- Daqing No. 3 Hospital of Heilongjiang Province, No. 54 Xitai Road, Ranghulu District, Daqing, Heilongjiang 163000, China
| | - Xiuqing Zhang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Zhen Zhang
- No. 4 Hospital of Jiangsu University, No. 246 Nanmen Street, Zhenjiang, Jiangsu 212000, China
| | - Hui Zhong
- Anhui Mental Health Center, No. 316 Huangshan Road, Hefei, Anhui 230000, China
| | - Gerome Breen
- MRC SGDP Centre, Institute of Psychiatry at King's College, De Crespigny Park, London SE5 8AF, UK; National Institute for Health Research, Biomedical Research Centre for Mental Health, Institute of Psychiatry at King's College, De Crespigny Park, London SE5 8AF, UK
| | - Jun Wang
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China; Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, Copenhagen 2200, Denmark; Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China; Princess Al Jawhara Center of Excellence in the Research of Hereditary Disorders, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Jonathan Marchini
- Department of Statistics, University of Oxford, Oxford, Oxfordshire OX1 3TG, UK; Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK
| | - Yiping Chen
- CTSU, University of Oxford, Richard Doll Building, Old Road Campus, Oxford, Oxfordshire OX3 7LF, UK
| | - Qi Xu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 10005, China
| | - Xun Xu
- BGI-Shenzhen, Floor 9 Complex Building, Beishan Industrial Zone, Yantian District, Shenzhen, Guangdong 518083, China
| | - Richard Mott
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK
| | - Guo-Jen Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan 33302, Taiwan, ROC
| | - Kenneth Kendler
- Dept Psychiatry MCV, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jonathan Flint
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, Oxfordshire OX3 7BN, UK; East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China.
| |
Collapse
|
20
|
Thomas AA, Fisher JL, Rahme GJ, Hampton TH, Baron U, Olek S, Schwachula T, Rhodes CH, Gui J, Tafe LJ, Tsongalis GJ, Lefferts JA, Wishart H, Kleen J, Miller M, Whipple CA, de Abreu FB, Ernstoff MS, Fadul CE. Regulatory T cells are not a strong predictor of survival for patients with glioblastoma. Neuro Oncol 2015; 17:801-9. [PMID: 25618892 DOI: 10.1093/neuonc/nou363] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/26/2014] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are potentially prognostic indicators in patients with glioblastoma. If differences in frequency of Tregs in tumor or blood account for substantial variation in patient survival, then reliably measuring Tregs may enhance treatment selection and improve outcomes. METHODS We measured Tregs and CD3+ T cells in tumors and blood from 25 patients with newly diagnosed glioblastoma. Tumor-infiltrating Tregs and CD3+ T cells, measured by quantitative DNA demethylation analysis (epigenetic qPCR) and by immunohistochemistry, and peripheral blood Treg proportions measured by flow cytometry were correlated with patient survival. Additionally, we analyzed data from The Cancer Genome Atlas (TCGA) to correlate the expression of Treg markers with patient survival and glioblastoma subtypes. RESULTS Tregs, as measured in tumor tissue and peripheral blood, did not correlate with patient survival. Although there was a correlation between tumor-infiltrating Tregs expression by epigenetic qPCR and immunohistochemistry, epigenetic qPCR was more sensitive and specific. Using data from TCGA, mRNA expression of Forkhead box protein 3 (FoxP3) and Helios and FoxP3 methylation level did not predict survival. While the classical glioblastoma subtype corresponded to lower expression of Treg markers, these markers did not predict survival in any of the glioblastoma subtypes. CONCLUSIONS Although immunosuppression is a hallmark of glioblastoma, Tregs as measured in tissue by gene expression, immunohistochemistry, or demethylation and Tregs in peripheral blood measured by flow cytometry do not predict survival of patients. Quantitative DNA demethylation analysis provides an objective, sensitive, and specific way of identifying Tregs and CD3+ T cells in glioblastoma.
Collapse
Affiliation(s)
- Alissa A Thomas
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Jan L Fisher
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Gilbert J Rahme
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Thomas H Hampton
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Udo Baron
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Sven Olek
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Tim Schwachula
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - C Harker Rhodes
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Jiang Gui
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Laura J Tafe
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Gregory J Tsongalis
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Joel A Lefferts
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Heather Wishart
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Jonathan Kleen
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Michael Miller
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Chery A Whipple
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Francine B de Abreu
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Marc S Ernstoff
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| | - Camilo E Fadul
- Department of Neurology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (A.A.T.); Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.L.F.); Department of Genetics, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Dartmouth College, Lebanon, New Hampshire (G.J.R.); Epiontis GmbH, Berlin, Germany (U.B., S.O., T.S.); Department of Pathology, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.H.R., L.J.T., G.J.T., J.A.L., F.B.d.A.); Section of Biostatistics and Epidemiology, Department of Family and Community Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.G.); Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire (T.H.H.); Department of Psychiatry, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (H.W.); Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire (J.K., M.M.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.A.W.); Melanoma Program, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio (M.S.E.); Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire (C.E.F.)
| |
Collapse
|
21
|
|
22
|
Platten M, Ochs K, Lemke D, Opitz C, Wick W. Microenvironmental clues for glioma immunotherapy. Curr Neurol Neurosci Rep 2014; 14:440. [PMID: 24604058 DOI: 10.1007/s11910-014-0440-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gliomas have been viewed for decades as inaccessible for a meaningful antitumor immune response as they grow in a sanctuary site protected from infiltrating immune cells. Moreover, the glioma microenvironment constitutes a hostile environment for an efficient antitumor immune response as glioma-derived factors such as transforming growth factor β and catabolites of the essential amino acid tryptophan paralyze T-cell function. There is growing evidence from preclinical and clinical studies that a meaningful antitumor immunity exists in glioma patients and that it can be activated by vaccination strategies. As a consequence, the concept of glioma immunotherapy appears to be experiencing a renaissance with the first phase 3 randomized immunotherapy trials entering the clinical arena. On the basis of encouraging results from other tumor entities using immunostimulatory approaches by blocking endogenous T-cell suppressive pathways mediated by cytotoxic T-lymphocyte antigen 4 or programmed cell death protein 1/programmed cell death protein 1 ligand 1 with humanized antibodies, there is now a realistic and promising option to combine active immunotherapy with agents blocking the immunosuppressive microenvironment in patients with gliomas to allow a peripheral antitumor immune response induced by vaccination to become effective. Here we review the current clinical and preclinical evidence of antimicroenvironment immunotherapeutic strategies in gliomas.
Collapse
Affiliation(s)
- Michael Platten
- Department of Neurooncology, University Hospital Heidelberg and National Center for Tumor Diseases, German Cancer Consortium (DKTK) Clinical Cooperation Units, Im Neuenheimer Feld, Heidelberg, Germany,
| | | | | | | | | |
Collapse
|
23
|
Zhuo C, Li Z, Xu Y, Wang Y, Li Q, Peng J, Zheng H, Wu P, Li B, Cai S. Higher FOXP3-TSDR demethylation rates in adjacent normal tissues in patients with colon cancer were associated with worse survival. Mol Cancer 2014; 13:153. [PMID: 24938080 PMCID: PMC4074420 DOI: 10.1186/1476-4598-13-153] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 06/11/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The influence of natural regulatory T cells (nTregs) on the patients with colon cancer is unclear. Demethylated status of the Treg-specific demethylated region (TSDR) of the FOXP3 gene was reported to be a potential biomarker for the identification of nTregs. METHODS The demethylation rate of the TSDR (TSDR-DMR) was calculated by using methylation-specific quantitative polymerase chain reaction (MS-qPCR) assay. The expression of TSDR-DMR and FOXP3 mRNA was investigated in various colorectal cancer cell lines. A total of 130 colon carcinoma samples were utilized to study the DMR at tumor sites (DMRT) and adjacent normal tissue (DMRN). The correlations between DMRs and clinicopathological variables of patients with colon cancer were studied. RESULTS The TSDR-DMRs varied dramatically among nTregs (97.920 ± 0.466%) and iTregs (3.917 ± 0.750%). Significantly, DMRT (3.296 ± 0.213%) was higher than DMRN (1.605 ± 0.146%) (n = 130, p = 0.000). Higher DMRN levels were found in female patients (p = 0.001) and those with distant metastases (p = 0.017), and were also associated with worse recurrence-free survival in non-stage IV patients (low vs. high, p = 0.022). However, further Cox multivariate analysis revealed that the FOXP3-TSDR status does not have prognostic value. CONCLUSION MS-qPCR assays of FOXP3-TSDR can efficiently distinguish nTregs from non-nTregs. Abnormal recruitment of nTregs occurs in the local tumor microenvironment. Infiltration of tissue-resident nTregs may have a negative role in anti-tumor effects in patients with colon cancer; however, this role is limited and complicated.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Bin Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, No, 270 Dong-an Road, Shanghai 20032, People's Republic of China.
| | | |
Collapse
|
24
|
Langevin SM, Houseman EA, Accomando WP, Koestler DC, Christensen BC, Nelson HH, Karagas MR, Marsit CJ, Wiencke JK, Kelsey KT. Leukocyte-adjusted epigenome-wide association studies of blood from solid tumor patients. Epigenetics 2014; 9:884-95. [PMID: 24671036 DOI: 10.4161/epi.28575] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Epigenome-wide studies of DNA methylation using blood-derived DNA from cancer patients are complicated by the heterogeneity of cell types within blood and the associated cell lineage specification of DNA methylation signatures. Here, we applied a novel set of analytic approaches to assess the association between cancer case-status and DNA methylation adjusted for leukocyte variation using blood specimens from three case-control cancer studies (bladder: 223 cases, 205 controls; head and neck: 92 cases, 92 controls; and ovarian: 131 cases, 274 controls). Using previously published data on leukocyte-specific CpG loci and a recently described approach to deconvolute subject-specific blood composition, we performed an epigenome-wide analysis to examine the association between blood-based DNA methylation patterns and each of the three aforementioned solid tumor types adjusted for cellular heterogeneity in blood. After adjusting for leukocyte profile in our epigenome-wide analysis, the omnibus association between case-status and methylation was significant for all three studies (bladder cancer: P = 0.047; HNSCC: P = 0.013; ovarian cancer: P = 0.0002). Subsequent analyses revealed that CpG sites associated with cancer were enriched for transcription factor binding motifs involved with cancer-associated pathways. These results support the existence of cancer-associated DNA methylation profiles in the blood of solid tumor patients that are independent of alterations in normal leukocyte distributions. Adoption of the methods developed here will make it feasible to rigorously assess the influence of variability of normal leukocyte profiles when investigating cancer related changes in blood-based epigenome-wide association studies.
Collapse
Affiliation(s)
- Scott M Langevin
- Department of Environmental Health; University of Cincinnati College of Medicine; Cincinnati, OH USA; Department of Epidemiology; Brown University; Providence, RI USA
| | - E Andres Houseman
- Department of Biostatistics; Oregon State University College of Public Health and Human Sciences; Corvallis, OR USA
| | - William P Accomando
- Department of Pathology & Laboratory Medicine; Brown University; Providence, RI USA
| | - Devin C Koestler
- Department of Community and Family Medicine; Section of Biostatistics and Epidemiology; Dartmouth Medical School; Lebanon, NH USA
| | - Brock C Christensen
- Department of Community and Family Medicine; Section of Biostatistics and Epidemiology; Dartmouth Medical School; Lebanon, NH USA; Department of Pharmacology and Toxicology; Dartmouth Medical School; Lebanon, NH USA
| | - Heather H Nelson
- Division of Epidemiology and Community Health; University of Minnesota Masonic Cancer Center; Minneapolis, MN USA
| | - Margaret R Karagas
- Department of Community and Family Medicine; Section of Biostatistics and Epidemiology; Dartmouth Medical School; Lebanon, NH USA
| | - Carmen J Marsit
- Department of Community and Family Medicine; Section of Biostatistics and Epidemiology; Dartmouth Medical School; Lebanon, NH USA; Department of Pharmacology and Toxicology; Dartmouth Medical School; Lebanon, NH USA
| | - John K Wiencke
- Department of Neurological Surgery; University of California San Francisco; San Francisco, CA USA
| | - Karl T Kelsey
- Department of Epidemiology; Brown University; Providence, RI USA; Department of Pathology & Laboratory Medicine; Brown University; Providence, RI USA
| |
Collapse
|
25
|
Accomando WP, Wiencke JK, Houseman EA, Nelson HH, Kelsey KT. Quantitative reconstruction of leukocyte subsets using DNA methylation. Genome Biol 2014; 15:R50. [PMID: 24598480 PMCID: PMC4053693 DOI: 10.1186/gb-2014-15-3-r50] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 03/05/2014] [Indexed: 11/23/2022] Open
Abstract
Background Cell lineage-specific DNA methylation patterns distinguish normal human leukocyte subsets and can be used to detect and quantify these subsets in peripheral blood. We have developed an approach that uses DNA methylation to simultaneously quantify multiple leukocyte subsets, enabling investigation of immune modulations in virtually any blood sample including archived samples previously precluded from such analysis. Here we assess the performance characteristics and validity of this approach. Results Using Illumina Infinium HumanMethylation27 and VeraCode GoldenGate Methylation Assay microarrays, we measure DNA methylation in leukocyte subsets purified from human whole blood and identify cell lineage-specific DNA methylation signatures that distinguish human T cells, B cells, NK cells, monocytes, eosinophils, basophils and neutrophils. We employ a bioinformatics-based approach to quantify these cell types in complex mixtures, including whole blood, using DNA methylation at as few as 20 CpG loci. A reconstruction experiment confirms that the approach could accurately measure the composition of mixtures of human blood leukocyte subsets. Applying the DNA methylation-based approach to quantify the cellular components of human whole blood, we verify its accuracy by direct comparison to gold standard immune quantification methods that utilize physical, optical and proteomic characteristics of the cells. We also demonstrate that the approach is not affected by storage of blood samples, even under conditions prohibiting the use of gold standard methods. Conclusions Cell mixture distributions within peripheral blood can be assessed accurately and reliably using DNA methylation. Thus, precise immune cell differential estimates can be reconstructed using only DNA rather than whole cells.
Collapse
|
26
|
The prognostic value of Foxp3+ tumor-infiltrating lymphocytes in patients with glioblastoma. J Neurooncol 2013; 116:251-9. [PMID: 24276989 PMCID: PMC3890045 DOI: 10.1007/s11060-013-1314-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 11/10/2013] [Indexed: 12/26/2022]
Abstract
Forkhead box protein 3 (Foxp3) is known as a specific marker for regulatory T cells which contribute to immunosuppression in tumor microenvironment. However, existing studies regarding clinical significance of Foxp3+ tumor-infiltrating lymphocytes (TILs) in glioblastoma (GBM) remained discrepant. In this study, we aimed to explore whether this subtype of TILs correlated with prognosis in patients with GBM. Foxp3+ TILs as well as CD8+ ones were detected by immunohistochemistry on paraffin-embedded tumor samples from 62 patients. Staining for p53, MGMT and Ki-67 were also performed. The correlation of TIL subtypes with clinicopathologic features were analyzed. Progression-free survival (PFS) and overall survival (OS) were estimated by Kaplan–Meier method and compared using log-rank test. Independent prognostic factors for PFS and OS were determined through univariate and multivariate analysis. Significant correlation was found between Foxp3 and CD8 expression (P = 0.003), but not between TIL subtypes and clinicopathologic characteristics. Patients with higher density of Foxp3+ TILs showed relatively shorter PFS (P < 0.001) and OS (P = 0.003) whereas patients with higher density of CD8+ TILs obtained no significant differences in survival. Survival analysis based on molecular classifications further clarified these predictive values. Univariate and multivariate analysis revealed that frequency of Foxp3+ TILs was probably associated with both PFS (P = 0.002) and OS (P = 0.003). In conclusion, the results suggest that Foxp3 positive infiltrates could provide an independent predictive factor in GBM.
Collapse
|
27
|
Langevin SM, Kelsey KT. The fate is not always written in the genes: epigenomics in epidemiologic studies. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2013; 54:533-41. [PMID: 23444110 PMCID: PMC4093796 DOI: 10.1002/em.21762] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/08/2013] [Accepted: 01/10/2013] [Indexed: 05/11/2023]
Abstract
Cost-effective, high-throughput epigenomic technologies have begun to emerge, rapidly replacing the candidate gene approach to molecular epidemiology and offering a comprehensive strategy for the study of epigenetics in human subjects. Epigenome-wide association studies (EWAS) provide new opportunities for advancing our understanding of epigenetic changes associated with complex disease states. However, such analyses are complicated by the dynamic nature of DNA methylation. In contrast to genomic studies, where genotype is essentially constant across somatic cells, EWAS present a new set of challenges, largely due to differential DNA methylation across distinct cell types, particularly for studies involving heterogeneous tissue sources, and changes in the epigenetic profile that occur over time. This review describes potential applications of EWAS from the viewpoint of the molecular epidemiologist, along with special considerations and pitfalls involved in the design of such studies.
Collapse
Affiliation(s)
- Scott M. Langevin
- Department of Epidemiology, Brown University, Providence, RI
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI
| | - Karl T. Kelsey
- Department of Epidemiology, Brown University, Providence, RI
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI
- Corresponding author: 70 Ship Street, Box G-E5, Providence, RI 02912, Phone: 401-863-6420, Fax: 401-863-9008,
| |
Collapse
|
28
|
Thomas AA, Fisher JL, Ernstoff MS, Fadul CE. Vaccine-based immunotherapy for glioblastoma. CNS Oncol 2013; 2:331-49. [PMID: 25054578 PMCID: PMC6166520 DOI: 10.2217/cns.13.29] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glioblastoma remains the most lethal human brain tumor, despite the advent of multimodal treatment approaches. Because immune tolerance plays an important role in tumor progression, adding immunotherapy has become an attractive and innovative treatment approach for these aggressive tumors. Several early-phase clinical trials have demonstrated that vaccine-based immunotherapies, including dendritic cell therapy, peptide-based vaccines and vaccines containing autologous tumor lysates, are feasible and well tolerated. These trials have revealed promising trends in overall survival and progression-free survival for patients with glioblastoma, and have paved the way for ongoing randomized controlled trials.
Collapse
Affiliation(s)
- Alissa A Thomas
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Jan L Fisher
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Marc S Ernstoff
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Camilo E Fadul
- Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
29
|
Implantation of GL261 neurospheres into C57/BL6 mice: a more reliable syngeneic graft model for research on glioma-initiating cells. Int J Oncol 2013; 43:477-84. [PMID: 23708048 DOI: 10.3892/ijo.2013.1962] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Accepted: 04/05/2013] [Indexed: 11/05/2022] Open
Abstract
Recent studies have demonstrated that inflammatory cells and inflammatory mediators are indispensable components of the tumor-initiating cell (TIC) niche and regulate the malignant behavior of TICs. However, conventional animal models for glioma-initiating cell (GIC) studies are based on the implantation of GICs from human glioblastoma (GBM) into immunodeficient mice without the regulation of immune system. Whether animal models can mimic the cellular microenvironment of malignancy and evaluate the biological features of GICs accurately is unclear. Here, we detected the biological features of neurosphere-like tumor cells derived from the murine GBM cell line GL261 (GL261-NS) and from primary human GBM (PGBM-NS) in vitro, injected GL261-NS into syngeneic C57/BL6 mouse brain and injected PGBM-NS into NOD/SCID mouse brain, respectively. The tumorigenic characteristics of the two different orthotopic transplantation models were analyzed and the histological discrepancy between grafts and human primary GBM was compared. We found that GICs enriched in GL261-NS, GL261-NS and PGBM-NS exhibited increased GIC potential and enhanced chemoresistance in vitro. GL261-NS was significantly more aggressive compared to GL261 adhesive cells (GL261-AC) in vivo and the enhanced aggression was more significant in syngeneic mice compared to immunodeficient mice. The discrepancy of tumorigenicity between GL261-NS and GL261-AC in C57/BL6 mice was also larger compared to that between PGBM-NS and PGBM-AC in immunodeficient mice. Syngrafts derived from GL261-NS in C57/BL6 mice corresponded to the human GBM histologically better, compared with xenografts derived from PGBM-NS in NOD/SCID mice, which lack inflammatory cells and inflammatory mediators. We conclude that the inflammatory niche is involved in the tumorigenicity of GICs and implantation of GL261-NS into C57/BL6 mice is a more reliable syngeneic graft model for in vivo study on GICs relative to the immunodeficiency model.
Collapse
|
30
|
Abstract
The GTC Cancer Summit: Novel Approaches to Drug Discovery was divided into two parallel tracks: the 2nd Cancer Epigenetics Conference, and the Protein Kinases and Drug Design Conference. The 2nd Cancer Epigenetics Conference focused on exciting changes in drug discovery that include an unprecedented private and public collaboration on drug discovery in epigenetics through the Structural Genomics Consortium (SGC), which has led to several major breakthroughs including: the development of small-molecule inhibitors that interfere with protein interactions, especially bromodomain-containing protein acetylation readers; the indirect but successful targeting of the elusive MYC oncogene; and the identification of epigenetic drugs that are disease-specific. Also reported were the development of clinically useful DNA methylation assays; cell, peptide and protein arrays for testing antibody- and protein-binding specificity; and tools for chromatin capture and DNA modification analysis. Several groups reported on the lack of specificity of some commercial, but unnamed, antibodies used for epigenetic studies.
Collapse
|