201
|
Bayik D, Lee J, Lathia JD. All Glioblastoma Are Not Equal: Distinct Spatial Immune Profiles Between De Novo and Recurrent Tumors. Cancer Immunol Res 2022; 10:787. [PMID: 35670711 DOI: 10.1158/2326-6066.cir-22-0400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 11/16/2022]
Abstract
Lack of insight into the immunosuppressive microenvironment of glioblastoma (GBM) hinders successful application of immunotherapy. In this issue, Alanio and colleagues identify a distinct T-cell localization profile between recurrent and de novo GBM highlighting the importance of spatial heterogeneity and providing new avenues to explore to improve GBM outcome. See related article by Alanio et al., (3).
Collapse
Affiliation(s)
- Defne Bayik
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Juyeun Lee
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Justin D Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
202
|
Gatto L, Franceschi E, Tosoni A, Nunno VD, Bartolini S, Brandes AA. Hypermutation as a potential predictive biomarker of immunotherapy efficacy in high-grade gliomas: a broken dream? Immunotherapy 2022; 14:799-813. [PMID: 35670093 DOI: 10.2217/imt-2021-0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A high tumor mutational burden and mismatch repair deficiency are observed in 'hypermutated' high-grade gliomas (HGGs); however, the molecular characterization of this distinct subtype and whether it predicts the response to immune checkpoint inhibitors (ICIs) are largely unknown. Pembrolizumab is a valid therapeutic option for the treatment of hypermutated cancers of diverse origin, but only a few clinical trials have explored the activity of ICIs in hypermutated HGGs. HGGs appear to differ from other cancers, likely due to the prevalence of subclonal versus clonal neoantigens, which are unable to elicit an immune response with ICIs. The main aim of this review is to summarize the current knowledge on hypermutation in HGGs, focusing on the broken promises of tumor mutational burden and mismatch repair deficiency as potential biomarkers of response to ICIs.
Collapse
Affiliation(s)
- Lidia Gatto
- Department of Oncology, AUSL Bologna, Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Alba Ariela Brandes
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
203
|
Revisiting the definition of glioma recurrence based on a phylogenetic investigation of primary and re-emerging tumor samples: a case report. Brain Tumor Pathol 2022; 39:218-224. [DOI: 10.1007/s10014-022-00438-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/16/2022] [Indexed: 11/27/2022]
|
204
|
Anticancer and Anti-Metastatic Role of Thymoquinone: Regulation of Oncogenic Signaling Cascades by Thymoquinone. Int J Mol Sci 2022; 23:ijms23116311. [PMID: 35682990 PMCID: PMC9181073 DOI: 10.3390/ijms23116311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/29/2022] [Accepted: 05/30/2022] [Indexed: 12/04/2022] Open
Abstract
Cancer is a life-threatening and multifaceted disease. Pioneering research works in the past three decades have mechanistically disentangled intertwined signaling networks which play contributory roles in carcinogenesis and metastasis. Phenomenal strides have been made in leveraging our scientific knowledge altogether to a new level of maturity. Rapidly accumulating wealth of information has underlined a myriad of transduction cascades which can be pharmaceutically exploited for cancer prevention/inhibition. Natural products serve as a treasure trove and compel interdisciplinary researchers to study the cancer chemopreventive roles of wide-ranging natural products in cell culture and preclinical studies. Experimental research related to thymoquinone has gradually gained momentum because of the extra-ordinary cancer chemopreventive multifunctionalities of thymoquinone. In this mini-review, we provide an overview of different cell signaling cascades reported to be regulated by thymoquinone for cancer chemoprevention. Essentially, thymoquinone efficacy has also been notably studied in animal models, which advocates for a rationale-based transition of thymoquinone from the pre-clinical pipeline to clinical trials.
Collapse
|
205
|
Challenges in glioblastoma immunotherapy: mechanisms of resistance and therapeutic approaches to overcome them. Br J Cancer 2022; 127:976-987. [DOI: 10.1038/s41416-022-01864-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/23/2022] [Accepted: 05/12/2022] [Indexed: 11/08/2022] Open
|
206
|
Sun F, Grenert JP, Tan L, Van Ziffle J, Joseph NM, Mulvey CK, Bergsland E. Checkpoint Inhibitor Immunotherapy to Treat Temozolomide-Associated Hypermutation in Advanced Atypical Carcinoid Tumor of the Lung. JCO Precis Oncol 2022; 6:e2200009. [PMID: 35737914 PMCID: PMC9249272 DOI: 10.1200/po.22.00009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/06/2022] [Accepted: 05/04/2022] [Indexed: 01/23/2023] Open
Affiliation(s)
- Fangdi Sun
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - James P. Grenert
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Lisa Tan
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Jessica Van Ziffle
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Nancy M. Joseph
- Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Claire K. Mulvey
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Emily Bergsland
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
207
|
Hu C, Wang K, Damon C, Fu Y, Ma T, Kratz L, Lal B, Ying M, Xia S, Cahill DP, Jackson CM, Lim M, Laterra J, Li Y. ATRX loss promotes immunosuppressive mechanisms in IDH1 mutant glioma. Neuro Oncol 2022; 24:888-900. [PMID: 34951647 PMCID: PMC9159463 DOI: 10.1093/neuonc/noab292] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND ATRX inactivation occurs with IDH1R132H and p53 mutations in over 80% of Grades II/III astrocytomas. It is believed that ATRX loss contributes to oncogenesis by dysregulating epigenetic and telomere mechanisms but effects on anti-glioma immunity have not been explored. This paper examines how ATRX loss contributes to the malignant and immunosuppressive phenotypes of IDH1R132H/p53mut glioma cells and xenografts. METHODS Isogenic astrocytoma cells (+/-IDH1R132H/+/-ATRXloss) were established in p53mut astrocytoma cell lines using lentivirus encoding doxycycline-inducible IDH1R132H, ATRX shRNA, or Lenti-CRISPR/Cas9 ATRX. Effects of IDH1R132H+/-ATRXloss on cell migration, growth, DNA repair, and tumorigenicity were evaluated by clonal growth, transwell and scratch assays, MTT, immunofluorence and immunoblotting assays, and xenograft growth. Effects on the expression and function of modulators of the immune microenvironment were quantified by qRT-PCR, immunoblot, T-cell function, macrophage polarization, and flow cytometry assays. Pharmacologic inhibitors were used to examine epigenetic drivers of the immunosuppressive transcriptome of IDH1R132H/p53mut/ATRXloss cells. RESULTS Adding ATRX loss to the IDH1R132H/p53mut background promoted astrocytoma cell aggressiveness, induced expression of BET proteins BRD3/4 and an immune-suppressive transcriptome consisting of up-regulated immune checkpoints (e.g., PD-L1, PD-L2) and altered cytokine/chemokine profiles (e.g., IL33, CXCL8, CSF2, IL6, CXCL9). ATRX loss enhanced the capacity of IDH1R132H/p53mut cells to induce T-cell apoptosis, tumorigenic/anti-inflammatory macrophage polarization and Treg infiltration. The transcriptional and biological immune-suppressive responses to ATRX loss were enhanced by temozolomide and radiation and abrogated by pharmacologic BET inhibition. CONCLUSIONS ATRX loss activates a BRD-dependent immune-suppressive transcriptome and immune escape mechanism in IDH1R132H/p53mut astrocytoma cells.
Collapse
Affiliation(s)
- Chengchen Hu
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Kimberly Wang
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Ceylan Damon
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Yi Fu
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Tengjiao Ma
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
| | - Lisa Kratz
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bachchu Lal
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mingyao Ying
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shuli Xia
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher M Jackson
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Lim
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John Laterra
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yunqing Li
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
208
|
Zhang J, Siller-Farfán JA. Current and future perspectives of chimeric antigen receptors against glioblastoma. IMMUNOTHERAPY ADVANCES 2022; 2:ltac014. [PMID: 36284838 PMCID: PMC9585667 DOI: 10.1093/immadv/ltac014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/27/2022] [Indexed: 11/14/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant form of cancer in the central nervous system; even with treatment, it has a 5-year survival rate of 7.2%. The adoptive cell transfer (ACT) of T cells expressing chimeric antigen receptors (CARs) has shown a remarkable success against hematological malignancies, namely leukemia and multiple myeloma. However, CAR T cell therapy against solid tumors, and more specifically GBM, is still riddled with challenges preventing its widespread adoption. Here, we first establish the obstacles in ACT against GBM, including on-target/off-tumor toxicity, antigen modulation, tumor heterogeneity, and the immunosuppressive tumor microenvironment. We then present recent preclinical and clinical studies targeting well-characterized GBM antigens, which include the interleukin-13 receptor α2 and the epidermal growth factor receptor. Afterward, we turn our attention to alternative targets in GBM, including less-explored antigens such as B7-H3 (CD276), carbonic anhydrase IX, and the GD2 ganglioside. We also discuss additional target ligands, namely CD70, and natural killer group 2 member D ligands. Finally, we present the possibilities afforded by novel CAR architectures. In particular, we examine the use of armored CARs to improve the survival and proliferation of CAR T cells. We conclude by discussing the advantages of tandem and synNotch CARs when targeting multiple GBM antigens.
Collapse
Affiliation(s)
- Josephine Zhang
- Department of Biology, Johns Hopkins University, 3400 N Charles St , Baltimore 21218, United States
- St Anne’s College, University of Oxford, Woodstock Rd , Oxford OX2 6HS, United Kingdom
| | - Jesús A Siller-Farfán
- Sir William Dunn School of Pathology, University of Oxford, S Parks Rd , Oxford OX1 3DP, United Kingdom
| |
Collapse
|
209
|
Goodman AL, Velázquez Vega JE, Glenn C, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of neuropathology in the management of progressive glioblastoma in adults. J Neurooncol 2022; 158:179-224. [PMID: 35648306 DOI: 10.1007/s11060-022-04005-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022]
Abstract
TARGET POPULATION These recommendations apply to adult patients with progressive or recurrent glioblastoma (GBM). QUESTION For adult patients with progressive glioblastoma does testing for Isocitrate Dehydrogenase (IDH) 1 or 2 mutations provide new additional management or prognostic information beyond that derived from the tumor at initial presentation? RECOMMENDATION Level III: Repeat IDH mutation testing is not necessary if the tumor is histologically similar to the primary tumor and the patient's clinical course is as expected. QUESTION For adult patients with progressive glioblastoma does repeat testing for MGMT promoter methylation provide new or additional management or prognostic information beyond that derived from the tumor at initial presentation and what methods of detection are optimal? RECOMMENDATION Level III: Repeat MGMT promoter methylation is not recommended. QUESTION For adult patients with progressive glioblastoma does EGFR amplification or mutation testing provide management or prognostic information beyond that provided by histologic analysis and if performed on previous tissue samples, does it need to be repeated? RECOMMENDATION Level III: In cases that are difficult to classify as glioblastoma on histologic features EGFR amplification testing may help in classification. If a previous EGFR amplification was detected, repeat testing is not necessary. Repeat EGFR amplification or mutational testing may be recommended in patients in which target therapy is being considered. QUESTION For adult patients with progressive glioblastoma does large panel or whole genome sequencing provide management or prognostic information beyond that derived from histologic analysis? RECOMMENDATION Level III: Primary or repeat large panel or whole genome sequencing may be considered in patients who are eligible or interested in molecularly guided therapy or clinical trials. QUESTION For adult patients with progressive glioblastoma should immune checkpoint biomarker testing be performed to provide management and prognostic information beyond that obtained from histologic analysis? RECOMMENDATION Level III: The current evidence does not support making PD-L1 or mismatch repair (MMR) enzyme activity a component of standard testing. QUESTION For adult patients with progressive glioblastoma are there meaningful biomarkers for bevacizumab responsiveness and does their assessment provide additional information for tumor management and prognosis beyond that learned by standard histologic analysis? RECOMMENDATION Level III: No established Bevacizumab biomarkers are currently available based upon the inclusion criteria of this guideline.
Collapse
Affiliation(s)
- Abigail L Goodman
- Carolinas Pathology, Atrium Health Carolinas Medical Center, Charlotte, NC, USA.
| | - José E Velázquez Vega
- Department of Pathology and Laboratory Medicine, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, USA
| | - Chad Glenn
- Department of Neurosurgery, Stephenson Cancer Center, The University of Oklahoma, Oklahoma City, OK, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
210
|
Huang S, Xiao J, Wu J, Liu J, Feng X, Yang C, Xiang D, Luo S. Tizoxanide Promotes Apoptosis in Glioblastoma by Inhibiting CDK1 Activity. Front Pharmacol 2022; 13:895573. [PMID: 35694267 PMCID: PMC9174573 DOI: 10.3389/fphar.2022.895573] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/05/2022] [Indexed: 01/28/2023] Open
Abstract
The antiparasitic drug nitazoxanide (NTZ) has received considerable attention for its potential in cancer therapy. In this study, we demonstrate that tizoxanide (TIZ), an active metabolite of NTZ, exhibits antiglioma activity in vitro and in vivo by inducing G2/M cell cycle arrest and apoptosis. In vitro, TIZ dose-dependently inhibited the proliferation of U87, U118, and A172 human glioblastoma (GBM) cells at 48 h with IC50 values of 1.10, 2.31, and 0.73 µM, respectively. Treatment with TIZ (1 and 10 µM) also dose-dependently inhibited the colony formation of these GBM cells and accumulated ROS damage in the nucleus. In silico target fishing combined with network pharmacological disease spectrum analyses of GBM revealed that cycle-dependent kinase 1 (CDK1) is the most compatible target for TIZ and molecular docking by Molecule Operating Environment (MOE) software confirmed it. Mechanistically, TIZ inhibited the phosphorylation of CDK1 at Thr161 and decreased the activity of the CDK1/cyclin B1 complex, arresting the cell cycle at the G2/M phase. TIZ may induce apoptosis via the ROS-mediated apoptotic pathway. In vivo, TIZ suppressed the growth of established subcutaneous and intracranial orthotopic xenograft models of GBM without causing obvious side effects and prolonged the survival of nude mice bearing glioma. Taken together, our results demonstrated that TIZ might be a promising chemotherapy drug in the treatment of GBM.
Collapse
Affiliation(s)
- Si Huang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
| | - Jingxian Xiao
- School of Medical Science, Hunan University of Medicine, Huaihua, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
| | - Jiayi Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xueping Feng
- Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Chengdong Yang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Daxiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
| | - Shilin Luo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- *Correspondence: Shilin Luo,
| |
Collapse
|
211
|
Lastakchi S, Olaloko MK, McConville C. A Potential New Treatment for High-Grade Glioma: A Study Assessing Repurposed Drug Combinations against Patient-Derived High-Grade Glioma Cells. Cancers (Basel) 2022; 14:2602. [PMID: 35681582 PMCID: PMC9179370 DOI: 10.3390/cancers14112602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 02/05/2023] Open
Abstract
Repurposed drugs have demonstrated in vitro success against high-grade gliomas; however, their clinical success has been limited due to the in vitro model not truly representing the clinical scenario. In this study, we used two distinct patient-derived tumour fragments (tumour core (TC) and tumour margin (TM)) to generate a heterogeneous, clinically relevant in vitro model to assess if a combination of repurposed drugs (irinotecan, pitavastatin, disulfiram, copper gluconate, captopril, celecoxib, itraconazole and ticlopidine), each targeting a different growth promoting pathway, could successfully treat high-grade gliomas. To ensure the clinical relevance of our data, TC and TM samples from 11 different patients were utilized. Our data demonstrate that, at a concentration of 100µm or lower, all drug combinations achieved lower LogIC50 values than temozolomide, with one of the combinations almost eradicating the cancer by achieving cell viabilities below 4% in five of the TM samples 6 days after treatment. Temozolomide was unable to stop tumour growth over the 14-day assay, while combination 1 stopped tumour growth, with combinations 2, 3 and 4 slowing down tumour growth at higher doses. To validate the cytotoxicity data, we used two distinct assays, end point MTT and real-time IncuCyte life analysis, to evaluate the cytotoxicity of the combinations on the TC fragment from patient 3, with the cell viabilities comparable across both assays. The local administration of combinations of repurposed drugs that target different growth promoting pathways of high-grade gliomas have the potential to be translated into the clinic as a novel treatment strategy for high-grade gliomas.
Collapse
Affiliation(s)
| | | | - Christopher McConville
- School of Pharmacy, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (S.L.); (M.K.O.)
| |
Collapse
|
212
|
Chromatin structure predicts survival in glioma patients. Sci Rep 2022; 12:8221. [PMID: 35581287 PMCID: PMC9114333 DOI: 10.1038/s41598-022-11019-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022] Open
Abstract
The pathological changes in epigenetics and gene regulation that accompany the progression of low-grade to high-grade gliomas are under-studied. The authors use a large set of paired atac-seq and RNA-seq data from surgically resected glioma specimens to infer gene regulatory relationships in glioma. Thirty-eight glioma patient samples underwent atac-seq sequencing and 16 samples underwent additional RNA-seq analysis. Using an atac-seq/RNA-seq correlation matrix, atac-seq peaks were paired with genes based on high correlation values (|r2| > 0.6). Samples clustered by IDH1 status but not by grade. Surprisingly there was a trend for IDH1 mutant samples to have more peaks. The majority of peaks are positively correlated with survival and positively correlated with gene expression. Constructing a model of the top six atac-seq peaks created a highly accurate survival prediction model (r2 = 0.68). Four of these peaks were still significant after controlling for age, grade, pathology, IDH1 status and gender. Grade II, III, and IV (primary) samples have similar transcription factors and gene modules. However, grade IV (recurrent) samples have strikingly few peaks. Patient-derived glioma cultures showed decreased peak counts following radiation indicating that this may be radiation-induced. This study supports the notion that IDH1 mutant and IDH1 wildtype gliomas have different epigenetic landscapes and that accessible chromatin sites mapped by atac-seq peaks tend to be positively correlated with expression. The data in this study leads to a new model of treatment response wherein glioma cells respond to radiation therapy by closing open regions of DNA.
Collapse
|
213
|
Pich O, Bailey C, Watkins TBK, Zaccaria S, Jamal-Hanjani M, Swanton C. The translational challenges of precision oncology. Cancer Cell 2022; 40:458-478. [PMID: 35487215 DOI: 10.1016/j.ccell.2022.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/16/2022] [Accepted: 04/05/2022] [Indexed: 12/11/2022]
Abstract
The translational challenges in the field of precision oncology are in part related to the biological complexity and diversity of this disease. Technological advances in genomics have facilitated large sequencing efforts and discoveries that have further supported this notion. In this review, we reflect on the impact of these discoveries on our understanding of several concepts: cancer initiation, cancer prevention, early detection, adjuvant therapy and minimal residual disease monitoring, cancer drug resistance, and cancer evolution in metastasis. We discuss key areas of focus for improving cancer outcomes, from biological insights to clinical application, and suggest where the development of these technologies will lead us. Finally, we discuss practical challenges to the wider adoption of molecular profiling in the clinic and the need for robust translational infrastructure.
Collapse
Affiliation(s)
- Oriol Pich
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Chris Bailey
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Thomas B K Watkins
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Simone Zaccaria
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Computational Cancer Genomics Research Group, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK; Department of Medical Oncology, University College London Hospitals, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
214
|
Effects of Long-Term Temozolomide Treatment on Glioblastoma and Astrocytoma WHO Grade 4 Stem-Like Cells. Int J Mol Sci 2022; 23:ijms23095238. [PMID: 35563629 PMCID: PMC9100657 DOI: 10.3390/ijms23095238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma leads to a fatal course within two years in more than two thirds of patients. An essential cornerstone of therapy is chemotherapy with temozolomide (TMZ). The effect of TMZ is counteracted by the cellular repair enzyme O6-methylguanine-DNA methyltransferase (MGMT). The MGMT promoter methylation, the main regulator of MGMT expression, can change from primary tumor to recurrence, and TMZ may play a significant role in this process. To identify the potential mechanisms involved, three primary stem-like cell lines (one astrocytoma with the mutation of the isocitrate dehydrogenase (IDH), CNS WHO grade 4 (HGA)), and two glioblastoma (IDH-wildtype, CNS WHO grade 4) were treated with TMZ. The MGMT promoter methylation, migration, proliferation, and TMZ-response of the tumor cells were examined at different time points. The strong effects of TMZ treatment on the MGMT methylated cells were observed. Furthermore, TMZ led to a loss of the MGMT promoter hypermethylation and induced migratory rather than proliferative behavior. Cells with the unmethylated MGMT promoter showed more aggressive behavior after treatment, while HGA cells reacted heterogenously. Our study provides further evidence to consider the potential adverse effects of TMZ chemotherapy and a rationale for investigating potential relationships between TMZ treatment and change in the MGMT promoter methylation during relapse.
Collapse
|
215
|
Gutierrez C, Vilas CK, Wu CJ, Al'Khafaji AM. Functionalized Lineage Tracing Can Enable the Development of Homogenization-Based Therapeutic Strategies in Cancer. Front Immunol 2022; 13:859032. [PMID: 35603167 PMCID: PMC9120583 DOI: 10.3389/fimmu.2022.859032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
The therapeutic landscape across many cancers has dramatically improved since the introduction of potent targeted agents and immunotherapy. Nonetheless, success of these approaches is too often challenged by the emergence of therapeutic resistance, fueled by intratumoral heterogeneity and the immense evolutionary capacity inherent to cancers. To date, therapeutic strategies have attempted to outpace the evolutionary tempo of cancer but frequently fail, resulting in lack of tumor response and/or relapse. This realization motivates the development of novel therapeutic approaches which constrain evolutionary capacity by reducing the degree of intratumoral heterogeneity prior to treatment. Systematic development of such approaches first requires the ability to comprehensively characterize heterogeneous populations over the course of a perturbation, such as cancer treatment. Within this context, recent advances in functionalized lineage tracing approaches now afford the opportunity to efficiently measure multimodal features of clones within a tumor at single cell resolution, enabling the linkage of these features to clonal fitness over the course of tumor progression and treatment. Collectively, these measurements provide insights into the dynamic and heterogeneous nature of tumors and can thus guide the design of homogenization strategies which aim to funnel heterogeneous cancer cells into known, targetable phenotypic states. We anticipate the development of homogenization therapeutic strategies to better allow for cancer eradication and improved clinical outcomes.
Collapse
Affiliation(s)
- Catherine Gutierrez
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Caroline K Vilas
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, United States
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Catherine J Wu
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | | |
Collapse
|
216
|
Feliz Morel ÁJ, Hasanovic A, Morin A, Prunier C, Magnone V, Lebrigand K, Aouad A, Cogoluegnes S, Favier J, Pasquier C, Mus-Veteau I. Persistent Properties of a Subpopulation of Cancer Cells Overexpressing the Hedgehog Receptor Patched. Pharmaceutics 2022; 14:pharmaceutics14050988. [PMID: 35631574 PMCID: PMC9146430 DOI: 10.3390/pharmaceutics14050988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/22/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the development of new therapeutic strategies, cancer remains one of the leading causes of mortality worldwide. One of the current major challenges is the resistance of cancers to chemotherapy treatments inducing metastases and relapse of the tumor. The Hedgehog receptor Patched (Ptch1) is overexpressed in many types of cancers. We showed that Ptch1 contributes to the efflux of doxorubicin and plays an important role in the resistance to chemotherapy in adrenocortical carcinoma (ACC), a rare cancer which presents strong resistance to the standard of care chemotherapy treatment. In the present study, we isolated and characterized a subpopulation of the ACC cell line H295R in which Ptch1 is overexpressed and more present at the cell surface. This cell subpopulation is more resistant to doxorubicin, grows as spheroids, and has a greater capability of clonogenicity, migration, and invasion than the parental cells. Xenograft experiments performed in mice and in ovo showed that this cell subpopulation is more tumorigenic and metastatic than the parental cells. These results suggest that this cell subpopulation has cancer stem-like or persistent cell properties which were strengthened by RNA-seq. If present in tumors from ACC patients, these cells could be responsible for therapy resistance, relapse, and metastases.
Collapse
Affiliation(s)
- Álvaro Javier Feliz Morel
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
| | - Anida Hasanovic
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
| | - Aurélie Morin
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue Contre le Cancer, CEDEX 15, 75737 Paris, France; (A.M.); (J.F.)
| | - Chloé Prunier
- INOVOTION, Biopolis-5 Av. du Grand Sablon, 38700 La Tronche, France;
| | - Virginie Magnone
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
| | - Kevin Lebrigand
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
| | - Amaury Aouad
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
| | - Sarah Cogoluegnes
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
| | - Judith Favier
- Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue Contre le Cancer, CEDEX 15, 75737 Paris, France; (A.M.); (J.F.)
| | - Claude Pasquier
- Université Côte d’Azur, CNRS-UMR7271, Laboratoire d’Informatique, Signaux et Systèmes de Sophia Antipolis (I3S), 06560 Valbonne, France;
| | - Isabelle Mus-Veteau
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), 06560 Valbonne, France; (Á.J.F.M.); (A.H.); (V.M.); (K.L.); (A.A.); (S.C.)
- Correspondence:
| |
Collapse
|
217
|
Lahoz S, Archilla I, Asensio E, Hernández‐Illán E, Ferrer Q, López‐Prades S, Nadeu F, Del Rey J, Sanz‐Pamplona R, Lozano JJ, Castells A, Cuatrecasas M, Camps J. Copy-number intratumor heterogeneity increases the risk of relapse in chemotherapy-naive stage II colon cancer. J Pathol 2022; 257:68-81. [PMID: 35066875 PMCID: PMC9790656 DOI: 10.1002/path.5870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/17/2021] [Accepted: 01/13/2022] [Indexed: 12/30/2022]
Abstract
Optimal selection of high-risk patients with stage II colon cancer is crucial to ensure clinical benefit of adjuvant chemotherapy. Here, we investigated the prognostic value of genomic intratumor heterogeneity and aneuploidy for disease recurrence. We combined targeted sequencing, SNP arrays, fluorescence in situ hybridization, and immunohistochemistry on a retrospective cohort of 84 untreated stage II colon cancer patients. We assessed the clonality of copy-number alterations (CNAs) and mutations, CD8+ lymphocyte infiltration, and their association with time to recurrence. Prognostic factors were included in machine learning analysis to evaluate their ability to predict individual relapse risk. Tumors from recurrent patients displayed a greater proportion of CNAs compared with non-recurrent (mean 31.3% versus 23%, respectively; p = 0.014). Furthermore, patients with elevated tumor CNA load exhibited a higher risk of recurrence compared with those with low levels [p = 0.038; hazard ratio (HR) 2.46], which was confirmed in an independent cohort (p = 0.004; HR 3.82). Candidate chromosome-specific aberrations frequently observed in recurrent cases included gain of the chromosome arm 13q (p = 0.02; HR 2.67) and loss of heterozygosity at 17q22-q24.3 (p = 0.05; HR 2.69). CNA load positively correlated with intratumor heterogeneity (R = 0.52; p < 0.0001). Consistently, incremental subclonal CNAs were associated with an elevated risk of relapse (p = 0.028; HR 2.20), which we did not observe for subclonal single-nucleotide variants and small insertions and deletions. The clinico-genomic model rated an area under the curve of 0.83, achieving a 10% incremental gain compared with clinicopathological markers (p = 0.047). In conclusion, tumor aneuploidy and copy-number intratumor heterogeneity were predictive of poor outcome and improved discriminative performance in early-stage colon cancer. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sara Lahoz
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Ivan Archilla
- Pathology Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Elena Asensio
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Eva Hernández‐Illán
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Queralt Ferrer
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Sandra López‐Prades
- Pathology Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Ferran Nadeu
- Molecular Pathology of Lymphoid NeoplasmsInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)BarcelonaSpain
| | - Javier Del Rey
- Department of Cell Biology, Physiology and Immunology, Faculty of MedicineUniversity Autonomous of BarcelonaBellaterraSpain
| | - Rebeca Sanz‐Pamplona
- Unit of Biomarkers and SusceptibilityOncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESPl'Hospitalet de LlobregatSpain
| | - Juan José Lozano
- Bioinformatics PlatformCentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
| | - Antoni Castells
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Miriam Cuatrecasas
- Pathology Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Jordi Camps
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain,Department of Cell Biology, Physiology and Immunology, Faculty of MedicineUniversity Autonomous of BarcelonaBellaterraSpain
| |
Collapse
|
218
|
Cho CF, Farquhar CE, Fadzen CM, Scott B, Zhuang P, von Spreckelsen N, Loas A, Hartrampf N, Pentelute BL, Lawler SE. A Tumor-Homing Peptide Platform Enhances Drug Solubility, Improves Blood-Brain Barrier Permeability and Targets Glioblastoma. Cancers (Basel) 2022; 14:cancers14092207. [PMID: 35565337 PMCID: PMC9103942 DOI: 10.3390/cancers14092207] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Glioblastoma (GBM) is a fatal and incurable brain cancer, and current treatment options have demonstrated limited success. Here, we describe the use of a dg-Bcan-Targeting-Peptide (BTP-7) that has BBB-penetrating properties and targets GBM. Conjugation of BTP-7 to an insoluble anti-cancer drug, camptothecin (CPT), improves drug solubility in aqueous solution, retains drug efficacy against patient-derived GBM stem cells (GSC), enhances BBB permeability, and enables therapeutic targeting to intracranial patient-derived GBM xenograft in mice, leading to higher toxicity in GBM cells compared to normal brain tissues and prolonged animal survival. This work demonstrates a proof-of-concept for BTP-7 as a tumor-targeting peptide for therapeutic delivery to GBM. Abstract Background: Glioblastoma (GBM) is the most common and deadliest malignant primary brain tumor, contributing significant morbidity and mortality among patients. As current standard-of-care demonstrates limited success, the development of new efficacious GBM therapeutics is urgently needed. Major challenges in advancing GBM chemotherapy include poor bioavailability, lack of tumor selectivity leading to undesired side effects, poor permeability across the blood–brain barrier (BBB), and extensive intratumoral heterogeneity. Methods: We have previously identified a small, soluble peptide (BTP-7) that is able to cross the BBB and target the human GBM extracellular matrix (ECM). Here, we covalently attached BTP-7 to an insoluble anti-cancer drug, camptothecin (CPT). Results: We demonstrate that conjugation of BTP-7 to CPT improves drug solubility in aqueous solution, retains drug efficacy against patient-derived GBM stem cells (GSC), enhances BBB permeability, and enables therapeutic targeting to intracranial GBM, leading to higher toxicity in GBM cells compared to normal brain tissues, and ultimately prolongs survival in mice bearing intracranial patient-derived GBM xenograft. Conclusion: BTP-7 is a new modality that opens the door to possibilities for GBM-targeted therapeutic approaches.
Collapse
Affiliation(s)
- Choi-Fong Cho
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
- Correspondence:
| | - Charlotte E. Farquhar
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
| | - Colin M. Fadzen
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
| | - Benjamin Scott
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
| | - Pei Zhuang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
| | - Niklas von Spreckelsen
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
- Department of General Neurosurgery, Centre of Neurosurgery, Faculty of Medicine and University Hospital, University of Cologne, 50937 Cologne, Germany
| | - Andrei Loas
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
| | - Nina Hartrampf
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Bradley L. Pentelute
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA;
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; (C.E.F.); (C.M.F.); (A.L.); (N.H.)
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sean E. Lawler
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (B.S.); (P.Z.); (N.v.S.); (S.E.L.)
- Legorreta Cancer Center, Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02912, USA
| |
Collapse
|
219
|
Liu L, Wang X, Li Y, Ma C, Shi Y, Li X, Chen J. The NDUFV2 gene silencing inhibits the proliferation of two drug-resistant cancer cell lines. J Genet Eng Biotechnol 2022; 20:64. [PMID: 35471675 PMCID: PMC9043080 DOI: 10.1186/s43141-022-00343-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 04/04/2022] [Indexed: 11/20/2022]
Abstract
Background Cancer is a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. It is unlikely that there will ever be a single cure for cancer, but the development of molecular biology and cell biology has brought new options for cancer treatment. Our research group found in the preliminary experiments that AAs exhibited significant anti-tumor activity. Studies also showed that AAs exhibited varying degrees of downregulation effects on the expression of the NDUFV2 gene in the MCF-7/ADR and SMMC-7721/ADR cell lines. However, there is no relevant report on the role of this gene expression during the growth process of drug-resistant tumor cells. To address possible objections, this paper aims to investigate the effect of NDUFV2 gene silencing on the proliferation of the MCF-7/ADR and SMMC-7721/ADR cell lines. Results The interfering plasmids pPLK/GFP+Puro-NDUFV2 shRNA-3 and shRNA-2 inhibited the NDUFV2 gene and protein expression most significantly in MCF-7/ADR and SMMC-7721/ADR cells, respectively. NDUFV2 gene silencing could effectively inhibit the proliferation of both cell lines. The inhibition rates for MCF-7/ADR were 67.31%, 73.02%, and 69.76% at 24 h, 48 h, and 72 h, while that for SMMC-7721/ADR were 68.89%, 71.97%, and 74.40% at 24 h, 48 h, and 72 h, respectively. The inhibition rate of SMMC-7721/ADR cell proliferation was positively correlated with time. Conclusions Interference with the NDUFV2 gene may significantly inhibit the proliferation of MCF-7/ADR and SMMC-7721/ADR cells. This study is the pioneer to investigate that the NDUFV2 gene has been associated with the activity of inhibiting tumor cell proliferation, suggesting that the NDUFV2 gene may become a potential target for the study of tumor genesis and the development of antineoplastic drugs.
Collapse
Affiliation(s)
- Lingling Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xunan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yue Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Chengyao Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Yeye Shi
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| | - Xiang Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China.
| | - Jianwei Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu Province, China
| |
Collapse
|
220
|
Wadden J, Ravi K, John V, Babila CM, Koschmann C. Cell-Free Tumor DNA (cf-tDNA) Liquid Biopsy: Current Methods and Use in Brain Tumor Immunotherapy. Front Immunol 2022; 13:882452. [PMID: 35464472 PMCID: PMC9018987 DOI: 10.3389/fimmu.2022.882452] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/14/2022] [Indexed: 11/27/2022] Open
Abstract
Gliomas are tumors derived from mutations in glial brain cells. Gliomas cause significant morbidity and mortality and development of precision diagnostics and novel targeted immunotherapies are critically important. Radiographic imaging is the most common technique to diagnose and track response to treatment, but is an imperfect tool. Imaging does not provide molecular information, which is becoming critically important for identifying targeted immunotherapies and monitoring tumor evolution. Furthermore, immunotherapy induced inflammation can masquerade as tumor progression in images (pseudoprogression) and confound clinical decision making. More recently, circulating cell free tumor DNA (cf-tDNA) has been investigated as a promising biomarker for minimally invasive glioma diagnosis and disease monitoring. cf-tDNA is shed by gliomas into surrounding biofluids (e.g. cerebrospinal fluid and plasma) and, if precisely quantified, might provide a quantitative measure of tumor burden to help resolve pseudoprogression. cf-tDNA can also identify tumor genetic mutations to help guide targeted therapies. However, due to low concentrations of cf-tDNA, recovery and analysis remains challenging. Plasma cf-tDNA typically represents <1% of total cf-DNA due to the blood-brain barrier, limiting their usefulness in practice and motivating the development and use of highly sensitive and specific detection methods. This mini review summarizes the current and future trends of various approaches for cf-tDNA detection and analysis, including new methods that promise more rapid, lower-cost, and accessible diagnostics. We also review the most recent clinical case studies for longitudinal disease monitoring and highlight focus areas, such as novel accurate detection methodologies, as critical research priorities to enable translation to clinic.
Collapse
Affiliation(s)
- Jack Wadden
- Department of Pediatric Hematology and Oncology, Michigan Medicine, Ann Arbor, MI, United States
| | | | | | | | - Carl Koschmann
- Department of Pediatric Hematology and Oncology, Michigan Medicine, Ann Arbor, MI, United States
| |
Collapse
|
221
|
Xu PF, Li C, Xi SY, Chen FR, Wang J, Zhang ZQ, Liu Y, Li X, Chen ZP. Whole Exome Sequencing Reveals the Genetic Heterogeneity and Evolutionary History of Primary Gliomas and Matched Recurrences. Comput Struct Biotechnol J 2022; 20:2235-2246. [PMID: 35615029 PMCID: PMC9117816 DOI: 10.1016/j.csbj.2022.04.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Peng-Fei Xu
- Scientific Research Center, The 7th Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 510275, PR China
- Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, PR China
- School of Medical, Sun Yat-Sen University, Shenzhen, Guandong 510275, PR China
| | - Cong Li
- Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, PR China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, Guandong 510120, PR China
| | - Shao-Yan Xi
- Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, PR China
| | - Fu-Rong Chen
- Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, PR China
| | - Jing Wang
- Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, PR China
| | - Zhi-Qiang Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, Guandong 510120, PR China
| | - Yan Liu
- Scientific Research Center, The 7th Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 510275, PR China
| | - Xin Li
- School of Medical, Sun Yat-Sen University, Shenzhen, Guandong 510275, PR China
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, PR China
- Corresponding authors.
| | - Zhong-Ping Chen
- Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong 510060, PR China
- Corresponding authors.
| |
Collapse
|
222
|
Sethi MK, Downs M, Shao C, Hackett WE, Phillips JJ, Zaia J. In-Depth Matrisome and Glycoproteomic Analysis of Human Brain Glioblastoma Versus Control Tissue. Mol Cell Proteomics 2022; 21:100216. [PMID: 35202840 PMCID: PMC8957055 DOI: 10.1016/j.mcpro.2022.100216] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is the most common and malignant primary brain tumor. The extracellular matrix, also known as the matrisome, helps determine glioma invasion, adhesion, and growth. Little attention, however, has been paid to glycosylation of the extracellular matrix components that constitute the majority of glycosylated protein mass and presumed biological properties. To acquire a comprehensive understanding of the biological functions of the matrisome and its components, including proteoglycans (PGs) and glycosaminoglycans (GAGs), in GBM tumorigenesis, and to identify potential biomarker candidates, we studied the alterations of GAGs, including heparan sulfate (HS) and chondroitin sulfate (CS), the core proteins of PGs, and other glycosylated matrisomal proteins in GBM subtypes versus control human brain tissue samples. We scrutinized the proteomics data to acquire in-depth site-specific glycoproteomic profiles of the GBM subtypes that will assist in identifying specific glycosylation changes in GBM. We observed an increase in CS 6-O sulfation and a decrease in HS 6-O sulfation, accompanied by an increase in unsulfated CS and HS disaccharides in GBM versus control samples. Several core matrisome proteins, including PGs (decorin, biglycan, agrin, prolargin, glypican-1, and chondroitin sulfate proteoglycan 4), tenascin, fibronectin, hyaluronan link protein 1 and 2, laminins, and collagens, were differentially regulated in GBM versus controls. Interestingly, a higher degree of collagen hydroxyprolination was also observed for GBM versus controls. Further, two PGs, chondroitin sulfate proteoglycan 4 and agrin, were significantly lower, about 6-fold for isocitrate dehydrogenase-mutant, compared to the WT GBM samples. Differential regulation of O-glycopeptides for PGs, including brevican, neurocan, and versican, was observed for GBM subtypes versus controls. Moreover, an increase in levels of glycosyltransferase and glycosidase enzymes was observed for GBM when compared to control samples. We also report distinct protein, peptide, and glycopeptide features for GBM subtypes comparisons. Taken together, our study informs understanding of the alterations to key matrisomal molecules that occur during GBM development. (Data are available via ProteomeXchange with identifier PXD028931, and the peaks project file is available at Zenodo with DOI 10.5281/zenodo.5911810).
Collapse
Affiliation(s)
- Manveen K Sethi
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - Margaret Downs
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - Chun Shao
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA
| | - William E Hackett
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA; Bioinformatics Program, Boston University, Boston, Massachusetts, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, Brain Tumor Center, Helen Diller Family Cancer Research Center, University of California San Francisco, San Francisco, California, USA; Division of Neuropathology, Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Joseph Zaia
- Department of Biochemistry, Center for Biomedical Mass Spectrometry, Boston University, Boston, Massachusetts, USA; Bioinformatics Program, Boston University, Boston, Massachusetts, USA.
| |
Collapse
|
223
|
Katzendobler S, Do A, Weller J, Dorostkar MM, Albert NL, Forbrig R, Niyazi M, Egensperger R, Thon N, Tonn JC, Quach S. Diagnostic Yield and Complication Rate of Stereotactic Biopsies in Precision Medicine of Gliomas. Front Neurol 2022; 13:822362. [PMID: 35432168 PMCID: PMC9005817 DOI: 10.3389/fneur.2022.822362] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/23/2022] [Indexed: 12/27/2022] Open
Abstract
BackgroundAn integrated diagnosis consisting of histology and molecular markers is the basis of the current WHO classification system of gliomas. In patients with suspected newly diagnosed or recurrent glioma, stereotactic biopsy is an alternative in cases in which microsurgical resection is deemed to not be safely feasible or indicated. In this retrospective study, we aimed to analyze both the diagnostic yield and the safety of a standardized biopsy technique.Material and MethodsThe institutional database was screened for frame-based biopsy procedures (January 2016 until March 2021). Only patients with a suspected diagnosis of glioma based on imaging were included. All tumors were classified according to the current WHO grading system. The clinical parameters, procedural complications, histology, and molecular signature of the tissues obtained were assessed.ResultsBetween January 2016 and March 2021, 1,214 patients underwent a stereotactic biopsy: 617 (50.8%) for a newly diagnosed lesion and 597 (49.2%) for a suspected recurrence. The median age was 56.9 years (range 5 months−94.4 years). Magnetic resonance imaging (MRI)-guidance was used in 99.3% of cases and additional positron emission tomography (PET)-guidance in 34.3% of cases. In total, stereotactic serial biopsy provided an integrated diagnosis in 96.3% of all procedures. The most frequent diagnoses were isocitrate dehydrogenase (IDH) wildtype glioblastoma (n = 596; 49.2%), oligodendroglioma grade 2 (n = 109; 9%), astrocytoma grade 3 (n = 108; 8.9%), oligodendroglioma grade 3 (n = 76; 6.3%), and astrocytoma grade 2 (n = 66; 5.4%). A detailed determination was successful for IDH 1/2 mutation in 99.4% of cases, for 1p/19q codeletion in 97.4% of cases, for TERT mutation in 98.9% of cases, and for MGMT promoter methylation in 99.1% of cases. Next-generation sequencing was evaluable in 64/67 (95.5%) of cases and DNA methylome analysis in 41/44 (93.2%) of cases. Thirteen (1.1%) cases showed glial tumors that could not be further specified. Seventy-three tumors were different non-glioma entities, e.g., of infectious or inflammatory nature. Seventy-five out of 597 suspected recurrences turned out to be post-therapeutic changes only. The rate of post-procedural complications with clinical symptoms of the Common Terminology Criteria for Adverse Events (CTCAE) grade 3 or higher was 1.2% in overall patients and 2.6% in the subgroup of brainstem biopsies. There was no fatal outcome in the entire series.ConclusionImage-guided stereotactic serial biopsy enables obtaining reliable histopathological and molecular diagnoses with a very low complication rate even in tumors with critical localization. Thus, in patients not undergoing microsurgical resection, this is a valuable tool for precision medicine of patients with glioma.
Collapse
Affiliation(s)
- Sophie Katzendobler
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Anna Do
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Jonathan Weller
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Mario M. Dorostkar
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Forbrig
- Institute of Neuroradiology, University Hospital, LMU Munich, Munich, Germany
| | - Maximilian Niyazi
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Rupert Egensperger
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Niklas Thon
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
| | - Joerg Christian Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany
- *Correspondence: Stefanie Quach
| |
Collapse
|
224
|
Löhr M, Härtig W, Schulze A, Kroiß M, Sbiera S, Lapa C, Mages B, Strobel S, Hundt JE, Bohnert S, Kircher S, Janaki-Raman S, Monoranu CM. SOAT1: A Suitable Target for Therapy in High-Grade Astrocytic Glioma? Int J Mol Sci 2022; 23:ijms23073726. [PMID: 35409086 PMCID: PMC8998855 DOI: 10.3390/ijms23073726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 02/05/2023] Open
Abstract
Targeting molecular alterations as an effective treatment for isocitrate dehydrogenase-wildtype glioblastoma (GBM) patients has not yet been established. Sterol-O-Acyl Transferase 1 (SOAT1), a key enzyme in the conversion of endoplasmic reticulum cholesterol to esters for storage in lipid droplets (LD), serves as a target for the orphan drug mitotane to treat adrenocortical carcinoma. Inhibition of SOAT1 also suppresses GBM growth. Here, we refined SOAT1-expression in GBM and IDH-mutant astrocytoma, CNS WHO grade 4 (HGA), and assessed the distribution of LD in these tumors. Twenty-seven GBM and three HGA specimens were evaluated by multiple GFAP, Iba1, IDH1 R132H, and SOAT1 immunofluorescence labeling as well as Oil Red O staining. To a small extent SOAT1 was expressed by tumor cells in both tumor entities. In contrast, strong expression was observed in glioma-associated macrophages. Triple immunofluorescence labeling revealed, for the first time, evidence for SOAT1 colocalization with Iba1 and IDH1 R132H, respectively. Furthermore, a notable difference in the amount of LD between GBM and HGA was observed. Therefore, SOAT1 suppression might be a therapeutic option to target GBM and HGA growth and invasiveness. In addition, the high expression in cells related to neuroinflammation could be beneficial for a concomitant suppression of protumoral microglia/macrophages.
Collapse
Affiliation(s)
- Mario Löhr
- Department of Neurosurgery, University Hospital Wuerzburg, 97080 Wuerzburg, Germany;
| | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, 04103 Leipzig, Germany;
| | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Matthias Kroiß
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität Munich, 80336 Munich, Germany;
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Silviu Sbiera
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany;
| | - Bianca Mages
- Institute for Anatomy, University of Leipzig, 04103 Leipzig, Germany;
| | - Sabrina Strobel
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany; (S.S.); (S.K.)
| | | | - Simone Bohnert
- Institute of Forensic Medicine, University of Wuerzburg, 97080 Wuerzburg, Germany;
| | - Stefan Kircher
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany; (S.S.); (S.K.)
| | - Sudha Janaki-Raman
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080 Wuerzburg, Germany; (S.S.); (S.K.)
- Correspondence: ; Tel.: +49-931-3181184
| |
Collapse
|
225
|
Mas-Ponte D, McCullough M, Supek F. Spectrum of DNA mismatch repair failures viewed through the lens of cancer genomics and implications for therapy. Clin Sci (Lond) 2022; 136:383-404. [PMID: 35274136 PMCID: PMC8919091 DOI: 10.1042/cs20210682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
Abstract
Genome sequencing can be used to detect DNA repair failures in tumors and learn about underlying mechanisms. Here, we synthesize findings from genomic studies that examined deficiencies of the DNA mismatch repair (MMR) pathway. The impairment of MMR results in genome-wide hypermutation and in the 'microsatellite instability' (MSI) phenotype-occurrence of indel mutations at short tandem repeat (microsatellite) loci. The MSI status of tumors was traditionally assessed by molecular testing of a selected set of MS loci or by measuring MMR protein expression levels. Today, genomic data can provide a more complete picture of the consequences on genomic instability. Multiple computational studies examined somatic mutation distributions that result from failed DNA repair pathways in tumors. These include analyzing the commonly studied trinucleotide mutational spectra of single-nucleotide variants (SNVs), as well as of other features such as indels, structural variants, mutation clusters and regional mutation rate redistribution. The identified mutation patterns can be used to rigorously measure prevalence of MMR failures across cancer types, and potentially to subcategorize the MMR deficiencies. Diverse data sources, genomic and pre-genomic, from human and from experimental models, suggest there are different ways in which MMR can fail, and/or that the cell-type or genetic background may result in different types of MMR mutational patterns. The spectrum of MMR failures may direct cancer evolution, generating particular sets of driver mutations. Moreover, MMR affects outcomes of therapy by DNA damaging drugs, antimetabolites, nonsense-mediated mRNA decay (NMD) inhibitors, and immunotherapy by promoting either resistance or sensitivity, depending on the type of therapy.
Collapse
Affiliation(s)
- David Mas-Ponte
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
| | - Marcel McCullough
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
| | - Fran Supek
- Genome Data Science, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, Baldiri Reixac 10, Barcelona 08028, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Pg Lluís Companys, 23, Barcelona 08010, Spain
| |
Collapse
|
226
|
Pienkowski T, Kowalczyk T, Garcia-Romero N, Ayuso-Sacido A, Ciborowski M. Proteomics and metabolomics approach in adult and pediatric glioma diagnostics. Biochim Biophys Acta Rev Cancer 2022; 1877:188721. [PMID: 35304294 DOI: 10.1016/j.bbcan.2022.188721] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 12/26/2022]
Abstract
The diagnosis of glioma is mainly based on imaging methods that do not distinguish between stage and subtype prior to histopathological analysis. Patients with gliomas are generally diagnosed in the symptomatic stage of the disease. Additionally, healing scar tissue may be mistakenly identified based on magnetic resonance imaging (MRI) as a false positive tumor recurrence in postoperative patients. Current knowledge of molecular alterations underlying gliomagenesis and identification of tumoral biomarkers allow for their use as discriminators of the state of the organism. Moreover, a multiomics approach provides the greatest spectrum and the ability to track physiological changes and can serve as a minimally invasive method for diagnosing asymptomatic gliomas, preceding surgery and allowing for the initiation of prophylactic treatment. It is important to create a vast biomarker library for adults and pediatric patients due to their metabolic differences. This review focuses on the most promising proteomic, metabolomic and lipidomic glioma biomarkers, their pathways, the interactions, and correlations that can be considered characteristic of tumor grade or specific subtype.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland.
| | - Tomasz Kowalczyk
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Noemi Garcia-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain
| | - Angel Ayuso-Sacido
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain; Brain Tumor Laboratory, Fundación Vithas, Grupo Hospitales Vithas, 28043 Madrid, Spain; Faculty of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| |
Collapse
|
227
|
Whitfield BT, Huse JT. Classification of adult-type diffuse gliomas: Impact of the World Health Organization 2021 update. Brain Pathol 2022; 32:e13062. [PMID: 35289001 PMCID: PMC9245936 DOI: 10.1111/bpa.13062] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Over the last decade, developments in molecular profiling have radically altered the diagnosis, classification, and management of numerous cancer types, with primary brain tumors being no exception. Although historically brain tumors have been classified based on their morphological characteristics, recent advances have allowed refinement of tumor classification based on molecular alterations. This shift toward molecular classification of primary brain tumors is reflected in the 2021 5th edition of the WHO classification of central nervous system tumors (WHO 2021). In this review, we will discuss the most recent updates to the classification of adult‐type diffuse gliomas, a group of highly infiltrative and largely incurable CNS malignancies. It is our hope continued that refinement of molecular criteria will improve diagnosis, prognostication, and eventually treatment of these devastating tumors.
Collapse
Affiliation(s)
- Benjamin T Whitfield
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jason T Huse
- Departments of Pathology and Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
228
|
Morano F, Raimondi A, Pagani F, Lonardi S, Salvatore L, Cremolini C, Murgioni S, Randon G, Palermo F, Antonuzzo L, Pella N, Racca P, Prisciandaro M, Niger M, Corti F, Bergamo F, Zaniboni A, Ratti M, Palazzo M, Cagnazzo C, Calegari MA, Marmorino F, Capone I, Conca E, Busico A, Brich S, Tamborini E, Perrone F, Di Maio M, Milione M, Di Bartolomeo M, de Braud F, Pietrantonio F. Temozolomide Followed by Combination With Low-Dose Ipilimumab and Nivolumab in Patients With Microsatellite-Stable, O 6-Methylguanine-DNA Methyltransferase-Silenced Metastatic Colorectal Cancer: The MAYA Trial. J Clin Oncol 2022; 40:1562-1573. [PMID: 35258987 PMCID: PMC9084437 DOI: 10.1200/jco.21.02583] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This is a multicenter, single-arm phase II trial evaluating the efficacy and safety of an immune-sensitizing strategy with temozolomide priming followed by a combination of low-dose ipilimumab and nivolumab in patients with microsatellite-stable (MSS) and O6-methylguanine–DNA methyltransferase (MGMT)–silenced metastatic colorectal cancer (mCRC). MAYA shows that temozolomide priming followed by Ipi/Nivo combo induces durable benefit in MSS/MGMT-silenced mCRC.![]()
Collapse
Affiliation(s)
- Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Raimondi
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pagani
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Medical Oncology 3, Istituto Oncologico Veneto IOV-IRCSS, Padua, Italy
| | - Lisa Salvatore
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Sabina Murgioni
- Medical Oncology 1, Istituto Oncologico Veneto IOV-IRCSS, Padua, Italy
| | - Giovanni Randon
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Palermo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy.,Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nicoletta Pella
- Department of Oncology, ASUFC University Hospital of Udine, Udine, Italy
| | - Patrizia Racca
- ColoRectal Cancer Unit, Department of Oncology, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Michele Prisciandaro
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Monica Niger
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Bergamo
- Medical Oncology 1, Istituto Oncologico Veneto IOV-IRCSS, Padua, Italy
| | | | - Margherita Ratti
- Department of Medical Oncology, Azienda Socio Sanitaria Territoriale of Cremona, Cremona, Italy
| | - Michele Palazzo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Celeste Cagnazzo
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Maria Alessandra Calegari
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federica Marmorino
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Iolanda Capone
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elena Conca
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Adele Busico
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Silvia Brich
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Elena Tamborini
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Federica Perrone
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Turin, Division of Medical Oncology, Ordine Mauriziano Hospital, Turin, Italy
| | - Massimo Milione
- Department of the Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Maria Di Bartolomeo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
229
|
Blonski M, Obara T, Brzenczek C, Pouget C, Dillier C, Meyer M, Lavigne L, Forthoffer N, Broussois A, Gauchotte G, Baron MH, Rech F, Mézières S, Gaudeau Y, Verger A, Vogin G, Anxionnat R, Moureaux JM, Taillandier L. Initial PCV Chemotherapy Followed by Radiotherapy Is Associated With a Prolonged Response But Late Neurotoxicity in 20 Diffuse Low-Grade Glioma Patients. Front Oncol 2022; 12:827897. [PMID: 35311144 PMCID: PMC8931287 DOI: 10.3389/fonc.2022.827897] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundStudy RTOG 9802 in high-risk diffuse low-grade gliomas (DLGGs) showed the potential synergistic effect on survival of the procarbazine, CCNU, and vincristine (PCV) radiotherapy (RT) combination. Limited data on long-term neurocognitive impact and quality of life (QoL) have yet been reported.Patients and MethodsWe described a monocentric series of patients treated at first line by the combination of PCV immediately followed by RT between January 01, 1982 and January 01, 2017. Radiological data were collected and included volume, velocity of diametric expansion (VDE), and MRI aspects. Long-term neurocognitive and QoL were analyzed.ResultsTwenty patients fulfilled the eligibility criteria. The median response rate was 65.1% (range, 9.6%–99%) at the time of maximal VDE decrease corresponding to a median volume reduction of 79.7 cm3 (range, 3.1 to 174.2 cm3), which occurred after a median period of 7.2 years (range, 0.3–21.9) after the end of RT. An ongoing negative VDE was measured in 13/16 patients after the end of RT, with a median duration of 6.7 years (range, 9 months–21.9 years). The median follow-up since radiological diagnosis was 17.5 years (range, 4.8 to 29.5). Estimated median survival was 17.4 years (95% CI: 12; NR). After a long-term follow-up, substantial neurotoxicity was noticed with dementia in six progression-free patients (30%), leading to ventriculo-peritoneal shunt procedures in three, and premature death in five. Thirteen patients (65%) were unable to work with disability status. Successive longitudinal neurocognitive assessments for living patients showed verbal episodic memory deterioration.ConclusionsPCV-RT combination seems to have not only an oncological synergy but also a long-term neurotoxic synergy to consider before initial therapeutic decision.
Collapse
Affiliation(s)
- Marie Blonski
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
- *Correspondence: Marie Blonski,
| | - Tiphaine Obara
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Cyril Brzenczek
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Celso Pouget
- Department of Pathology, CHRU, Nancy, France
- Centre de ressources Biologiques, BB-0033-00035, CHRU, Nancy, France
| | - Céline Dillier
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
| | - Mylène Meyer
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
| | - Laura Lavigne
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
| | | | | | - Guillaume Gauchotte
- Department of Pathology, CHRU, Nancy, France
- Centre de ressources Biologiques, BB-0033-00035, CHRU, Nancy, France
| | | | - Fabien Rech
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
- Department of Neurosurgery, CHRU, Nancy, France
| | - Sophie Mézières
- Department of Mathematics, Elie Cartan Institute, Nancy, France
- INRIA Biology, Genetics and Statistics, Nancy, France
| | - Yann Gaudeau
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Antoine Verger
- Department of Nuclear Medicine and Nancyclotep Imaging Platform, CHRU, Nancy, France
- IADI, INSERM U1254, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Guillaume Vogin
- Department of Radiation Therapy, Baclesse Radiation Therapy Center, Esch/Alzette, Luxembourg
- UMR 7635 CNRS, IMoPA Biopole Lorraine University Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | | | - Jean-Marie Moureaux
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| | - Luc Taillandier
- Department of Neurology, Neurooncology Unit, CHRU, Nancy, France
- Centre de Recherche en Automatique Nancy France - UMR 7039 - BioSiS Department, Faculty of Medicine, Université de Lorraine, Vandoeuvre-lès Nancy, France
| |
Collapse
|
230
|
Nussinov R, Zhang M, Maloney R, Tsai C, Yavuz BR, Tuncbag N, Jang H. Mechanism of activation and the rewired network: New drug design concepts. Med Res Rev 2022; 42:770-799. [PMID: 34693559 PMCID: PMC8837674 DOI: 10.1002/med.21863] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/06/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022]
Abstract
Precision oncology benefits from effective early phase drug discovery decisions. Recently, drugging inactive protein conformations has shown impressive successes, raising the cardinal questions of which targets can profit and what are the principles of the active/inactive protein pharmacology. Cancer driver mutations have been established to mimic the protein activation mechanism. We suggest that the decision whether to target an inactive (or active) conformation should largely rest on the protein mechanism of activation. We next discuss the recent identification of double (multiple) same-allele driver mutations and their impact on cell proliferation and suggest that like single driver mutations, double drivers also mimic the mechanism of activation. We further suggest that the structural perturbations of double (multiple) in cis mutations may reveal new surfaces/pockets for drug design. Finally, we underscore the preeminent role of the cellular network which is deregulated in cancer. Our structure-based review and outlook updates the traditional Mechanism of Action, informs decisions, and calls attention to the intrinsic activation mechanism of the target protein and the rewired tumor-specific network, ushering innovative considerations in precision medicine.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Mingzhen Zhang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| | - Ryan Maloney
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| | - Chung‐Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| | - Bengi Ruken Yavuz
- Department of Health Informatics, Graduate School of InformaticsMiddle East Technical UniversityAnkaraTurkey
| | - Nurcan Tuncbag
- Department of Health Informatics, Graduate School of InformaticsMiddle East Technical UniversityAnkaraTurkey
- Department of Chemical and Biological Engineering, College of EngineeringKoc UniversityIstanbulTurkey
- Koc University Research Center for Translational Medicine, School of MedicineKoc UniversityIstanbulTurkey
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| |
Collapse
|
231
|
Kovachka S, Malloci G, Simsir M, Ruggerone P, Azoulay S, Mus-Veteau I. Inhibition of the drug efflux activity of Ptch1 as a promising strategy to overcome chemotherapy resistance in cancer cells. Eur J Med Chem 2022; 236:114306. [DOI: 10.1016/j.ejmech.2022.114306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022]
|
232
|
Zhong L, Yang P, Zhang C, Wang Z, Jiang T, Chen B, Shan X, Qiu X. Long-term adjuvant administration of temozolomide impacts serum ions concentration in high-grade glioma. Chin Neurosurg J 2022; 8:6. [PMID: 35216639 PMCID: PMC8876447 DOI: 10.1186/s41016-022-00271-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 01/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background Adjuvant temozolomide (TMZ) chemotherapy with standard regimen remarkably improves survival in patients with high-grade glioma (HGG). However, the influence of long-term TMZ chemotherapy on serum ions concentration is unclear. Methods One hundred and thirty-eight patients with HGG were included. Their blood samples were collected for blood biochemistry and routine test. The alteration in serum ions concentration, total protein, albumin, globin, and blood cells counts were used to identify the impact of long-term TMZ chemotherapy. Results Through the comparation of quantitative value of diverse parameters among different chemotherapy cycles, we identified that serum potassium concentration had a downward trend after TMZ administration (1st vs. 6th, p < 0.001; 1st vs. 12th, p < 0.001). Additionally, the correlation analysis showed that platelets was negatively correlated with chemotherapy cycles (r = − 0.649, p = 0.023). The hematological adverse events mainly centered on grade 1 to 2. Conclusion Long-term administration of TMZ may lead to serum ions disturbance. Besides the myelosuppression, we should pay attention to the alteration in serum ions concentration, and give patients proper symptomatic treatment when necessary. Supplementary Information The online version contains supplementary material available at 10.1186/s41016-022-00271-7.
Collapse
Affiliation(s)
- Liyun Zhong
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Chuanbao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Zheng Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, 119 South 4th Ring Road West, Beijing, 100070, China
| | - Baoshi Chen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xia Shan
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China. .,Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China. .,Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, 119 South 4th Ring Road West, Beijing, 100070, China.
| | - Xiaoguang Qiu
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, 119 South 4th Ring Road West, Beijing, 100070, China.
| |
Collapse
|
233
|
Ling Z, Zhang J, Liu Q. Oncogenic Forkhead box D3 antisense RNA 1 promotes cell survival and confers temozolomide resistance in glioblastoma cells through the miR-128-3p/WEE1 G2 checkpoint kinase axis. Bioengineered 2022; 13:6012-6023. [PMID: 35191808 PMCID: PMC8974031 DOI: 10.1080/21655979.2022.2042133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Although temozolomide (TMZ) is recommended for glioblastoma (GBM) treatment, patients treated with TMZ usually develop TMZ resistance. Thus, there is an urgent need to elucidate the mechanism through which GBM cells acquire TMZ resistance. FOXD3-AS1, a recently discovered lncRNA, shows high expression in diverse cancer types. Nonetheless, its role in GBM remains unclear. This study found that FOXD3-AS1 was overexpressed in GBM cells and associated with dismal prognostic outcome in GBM patients. Functional studies revealed that depletion of FOXD3-AS1 inhibited cell growth and induced apoptosis of GBM cells. Results also showed that FOXD3-AS1 participates in the tolerance of GBM cells to TMZ. Specifically, TMZ-resistant cells exhibited higher FOXD3-AS1 expression compared to parental cells. Overexpression of FOXD3-AS1 increased TMZ tolerance in TMZ sensitive cells, whereas depletion of FOXD3-AS1 sensitized TMZ-resistant cells to TMZ treatment. Mechanistically, WEE1 was positively expressed with FOXD3-AS1. Given that both FOXD3-AS1 and WEE1 contain a binding site for miR-128-3p, FOXD3-AS1 could act as a competing endogenous RNA (ceRNA) to promote WEE1 expression by sponging miR-128-3p. Furthermore, we demonstrated that WEE1 was upregulated in TMZ-resistant GBM cells. Overexpression of WEE1 increased TMZ tolerance in TMZ sensitive cells, whereas deletion of FOXD3-AS1 promoted TMZ-resistant cells to be more sensitive to TMZ. Importantly, depletion of WEE1 could reverse TMZ resistant phenotype in FOXD3-AS1-overexpressed GBM cells. Collectively, our findings reveal a critical role of FOXD3-AS1 in the survival of GBM cells and TMZ resistance, which suggests that FOXD3-AS1 is a potential biomarker for the diagnosis and treatment of GBM.
Collapse
Affiliation(s)
- Zaisheng Ling
- Department of Ct Diagnosis, The Second Affiliated Hospital of Harbin Medical University, Harbin, P. R. China
| | - Jinpeng Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, P. R. China
| | - Qingqing Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, P. R. China
| |
Collapse
|
234
|
Baak JPA, Li H, Guo H. Clinical and Biological Interpretation of Survival Curves of Cancer Patients, Exemplified With Stage IV Non-Small Cell Lung Cancers With Long Follow-up. Front Oncol 2022; 12:837419. [PMID: 35186767 PMCID: PMC8849109 DOI: 10.3389/fonc.2022.837419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Worldwide, 18.1 million new invasive cancers and 9.9 million cancer deaths occurred in 2020. Lung cancer is the second most frequent (11.4%) and, with 1.8 million deaths, remains the leading cause of cancer mortality. About 1.7 million of lung cancers are of the non-small cell lung cancer (NSCLC) subtype, and of these, 60%-70% are in advanced stage IV at the time of diagnosis. Thus, the annual worldwide number of new NSCLC stage IV patients is about 1 million, and they have a very poor prognosis. Indeed, 25%-30% die within 3 months of diagnosis. However, the survival duration of the remaining 700,000 new patients per year surviving >3 months varies enormously. Surprisingly, little research has been done to explain these survival differences, but recently it was found that classical patient, tumour and treatment features cannot accurately distinguish short- and very long-term survivors. What then are the causes of these bewildering survival variations amongst "the same cancers"? Clonality, proliferation differences, neovascularization, intra-tumour heterogeneity, genetic inhomogeneity and other cancer hallmarks play important roles. Considering each of these, single or combined, can greatly improve our understanding. Another technique is analysis of the survival curve of a seemingly homogeneous group of cancer patients. This can give valuable information about the existence of subgroups and their biological characteristics. Different basic survival curves and what their shapes tell about the biological properties of these invasive cancers are discussed. Application of this analysis technique to the survival curve of 690 stage IV NSCLC patients with a 3.2-120.0-month survival suggests that this seemingly homogeneously group of patients probably consists of 4-8 subgroups with a very different survival. A subsequent detailed mathematical analysis shows that a model of 8 subgroups gives a very good match with the original survival curve of the whole group. In conclusion, the survival curve of a seemingly homogeneous group of cancer patients can give valuable information about the existence of subgroups and their biological characteristics. Application of this technique to 690 NSCLC Stage IV patients makes it probable that 8 different subgroups with very different survival rates exist in this group of cancers.
Collapse
Affiliation(s)
- Jan P A Baak
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway.,Medical Practice Dr. Med Jan Baak AS, Tananger, Norway
| | - Hegen Li
- Department of Medical Oncology, Longhua University Hospital, Shanghai, China
| | - Huiru Guo
- Department of Medical Oncology, Longhua University Hospital, Shanghai, China
| |
Collapse
|
235
|
Svec RL, McKee SA, Berry MR, Kelly AM, Fan TM, Hergenrother PJ. Novel Imidazotetrazine Evades Known Resistance Mechanisms and Is Effective against Temozolomide-Resistant Brain Cancer in Cell Culture. ACS Chem Biol 2022; 17:299-313. [PMID: 35119837 DOI: 10.1021/acschembio.2c00022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is the most lethal primary brain tumor. Currently, frontline treatment for primary GBM includes the DNA-methylating drug temozolomide (TMZ, of the imidazotetrazine class), while the optimal treatment for recurrent GBM remains under investigation. Despite its widespread use, a majority of GBM patients do not respond to TMZ therapy; expression of the O6-methylguanine DNA methyltransferase (MGMT) enzyme and loss of mismatch repair (MMR) function as the principal clinical modes of resistance to TMZ. Here, we describe a novel imidazotetrazine designed to evade resistance by MGMT while retaining suitable hydrolytic stability, allowing for effective prodrug activation and biodistribution. This dual-substituted compound, called CPZ, exhibits activity against cancer cells irrespective of MGMT expression and MMR status. CPZ has greater blood-brain barrier penetrance and comparable hematological toxicity relative to TMZ, while also matching its maximum tolerated dose in mice when dosed once-per-day over five days. The activity of CPZ is independent of the two principal mechanisms suppressing the effectiveness of TMZ, making it a promising new candidate for the treatment of GBM, especially those that are TMZ-resistant.
Collapse
Affiliation(s)
- Riley L. Svec
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Sydney A. McKee
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Matthew R. Berry
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Aya M. Kelly
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
236
|
Barresi V, Mafficini A, Calicchia M, Piredda ML, Musumeci A, Ghimenton C, Scarpa A. Recurrent oligodendroglioma with changed 1p/19q status. Neuropathology 2022; 42:160-166. [PMID: 35144313 PMCID: PMC9546156 DOI: 10.1111/neup.12789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/15/2021] [Accepted: 10/15/2021] [Indexed: 11/28/2022]
Abstract
We report a case of oligodendroglioma that had consistent histopathological features as well as a distinct change in 1p/19q status in the second recurrence, after temozolomide chemotherapy and radiotherapy. The first tumor recurrence had oligodendroglial morphology, IDH1 R132H and TERT promoter mutations, and 1p/19q codeletion detected by fluorescent in situ hybridization (FISH). Copy number analysis, assessed by next‐generation sequencing, confirmed 1p/19q codeletion, and disclosed loss of heterozygosity (LOH) of chromosomes 4 and 9 and chromosome 11 gain. The second recurrence featured not only oligodendroglial morphology but also the appearance of admixed multinucleated giant cells or neoplastic cells having oval nuclei and mitoses and showing microvascular proliferation; it maintained IDH1 R132H and TERT promoter mutations, acquired TP53 mutation, and showed 19q LOH, but disomic 1p, detected by FISH. Copy number analysis depicted LOH of chromosomes 3p, 13, and 19q, 1p partial deletion (1p chr1p34.2‐p11), and gain of chromosomes 2p25.3‐p24.1, 8q12.2‐q24.3, and 11q13.3‐q25. B‐allele frequency analysis of polymorphic sites disclosed copy‐neutral LOH at 1p36.33‐p34.2, supporting the initial deletion of 1p, followed by reduplication of 1p36.33‐p34.2 alone. These findings suggest that the two tumor recurrences might have originated from an initial neoplastic clone, featuring 1p/19q codeletion and IDH1 and TERT promoter mutations, and have independently acquired other copy number alterations. The reduplication of chromosome 1p might be the result of temozolomide treatment, and gave rise to false negative 1p deletion detected by FISH. The possibility of 1p copy‐neutral LOH should be considered in recurrent oligodendrogliomas with altered 1p/19q status detected by FISH.
Collapse
Affiliation(s)
- Valeria Barresi
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy.,ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Martina Calicchia
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Maria Liliana Piredda
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy
| | - Angelo Musumeci
- Department of Neurosciences, Unit of Neurosurgery, Hospital Trust of Verona, Verona, Italy
| | - Claudio Ghimenton
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Anatomic Pathology, University of Verona, Verona, Italy.,ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
237
|
3D microgels to quantify tumor cell properties and therapy response dynamics. Biomaterials 2022; 283:121417. [DOI: 10.1016/j.biomaterials.2022.121417] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/18/2022] [Accepted: 02/15/2022] [Indexed: 12/21/2022]
|
238
|
Brady SW, Gout AM, Zhang J. Therapeutic and prognostic insights from the analysis of cancer mutational signatures. Trends Genet 2022; 38:194-208. [PMID: 34483003 PMCID: PMC8752466 DOI: 10.1016/j.tig.2021.08.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/06/2021] [Accepted: 08/11/2021] [Indexed: 02/08/2023]
Abstract
The somatic mutations in each cancer genome are caused by multiple mutational processes, each of which leaves a characteristic imprint (or 'signature'), potentially caused by specific etiologies or exposures. Deconvolution of these signatures offers a glimpse into the evolutionary history of individual tumors. Recent work has shown that mutational signatures may also yield therapeutic and prognostic insights, including the identification of cell-intrinsic signatures as biomarkers of drug response and prognosis. For example, mutational signatures indicating homologous recombination deficiency are associated with poly(ADP)-ribose polymerase (PARP) inhibitor sensitivity, whereas APOBEC-associated signatures are associated with ataxia telangiectasia and Rad3-related kinase (ATR) inhibitor sensitivity. Furthermore, therapy-induced mutational signatures implicated in cancer progression have also been uncovered, including the identification of thiopurine-induced TP53 mutations in leukemia. In this review, we explore the various ways mutational signatures can reveal new therapeutic and prognostic insights, thus extending their traditional role in identifying disease etiology.
Collapse
Affiliation(s)
- Samuel W Brady
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Alexander M Gout
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
239
|
Richardson LG, Miller JJ, Kitagawa Y, Wakimoto H, Choi BD, Curry WT. Implications of IDH mutations on immunotherapeutic strategies for malignant glioma. Neurosurg Focus 2022; 52:E6. [DOI: 10.3171/2021.11.focus21604] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/19/2021] [Indexed: 11/06/2022]
Abstract
Immunotherapy has emerged as a promising approach for treating aggressive solid tumors, even within the CNS. Mutation in the metabolic gene isocitrate dehydrogenase 1 (IDH1) represents not only a major glioma defining biomarker but also an attractive therapeutic neoantigen. As patients with IDH-mutant glioma enter early-phase vaccine and immune checkpoint inhibitor clinical trials, there is emerging evidence that implicates the oncometabolite, 2-hydroxyglutarate (2HG), generated by the neomorphic activity of mutant IDH, as a potential barrier to current immunotherapeutic approaches. Here, the authors review the immunomodulatory and immunosuppressive roles of 2HG within the unique IDH-mutant glioma tumor immune microenvironment and discuss promising immunotherapeutic approaches currently being investigated in preclinical models.
Collapse
Affiliation(s)
- Leland G. Richardson
- Translational Brain Tumor Immunology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Julie J. Miller
- Pappas Center for Neuro-Oncology, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yosuke Kitagawa
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Bryan D. Choi
- Translational Brain Tumor Immunology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - William T. Curry
- Translational Brain Tumor Immunology Laboratory, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
240
|
Kawaguchi K, Otani R, Kikuchi M, Kushihara Y, Funata N, Yamada R, Shinoura N. Genetic Characteristics of Mismatch Repair-deficient Glioblastoma. NMC Case Rep J 2022; 8:565-571. [PMID: 35079518 PMCID: PMC8769403 DOI: 10.2176/nmccrj.cr.2020-0366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/18/2021] [Indexed: 11/20/2022] Open
Abstract
Mismatch repair (MMR) gene deficiency is rarely observed in gliomas, a constitutional defect is associated with tumorigenesis in Lynch syndrome, and an acquired defect is associated with hypermutation after temozolomide treatment. However, the meaning of MMR gene deficiency in gliomas is unclear. Two cases of MMR-deficient glioblastomas are reported, and mutational status of oncogenes was compared between primary and recurrent tumor samples in a glioblastoma patient with Lynch syndrome. Additionally, the characteristics of MMR-deficient glioblastomas were analyzed using public glioma datasets to determine the meaning of MMR deficiency in gliomas. Case 1 was a glioblastoma patient with Lynch syndrome, and treatment with pembrolizumab for the recurrent tumor was temporarily effective for a short period. Comparison of mutational changes between primary and recurrent tumor samples showed many additional mutated genes associated with multiple signaling pathways in the recurrent tumor. Tumor recurrence and chemoresistance could be associated with intratumoral heterogeneity and accelerated tumor progression due to defects of multiple signaling pathways. Case 2 was a glioblastoma patient with acquired MMR gene deficiency, and she died of rapid progression of bone marrow metastases. This rare clinical course was considered to be associated with gene expression changes and heterogeneity that resulted from MMR gene deficiency. Two cases of MMR gene-deficient glioblastomas were presented, and their genetic characteristics suggested that their clinical courses could be associated with MMR gene deficiency.
Collapse
Affiliation(s)
- Kei Kawaguchi
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Ryohei Otani
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Miyu Kikuchi
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yoshihiro Kushihara
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Nobuaki Funata
- Department of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Ryoji Yamada
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Nobusada Shinoura
- Department of Neurosurgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| |
Collapse
|
241
|
Barzegar Behrooz A, Talaie Z, Jusheghani F, Łos MJ, Klonisch T, Ghavami S. Wnt and PI3K/Akt/mTOR Survival Pathways as Therapeutic Targets in Glioblastoma. Int J Mol Sci 2022; 23:ijms23031353. [PMID: 35163279 PMCID: PMC8836096 DOI: 10.3390/ijms23031353] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a devastating type of brain tumor, and current therapeutic treatments, including surgery, chemotherapy, and radiation, are palliative at best. The design of effective and targeted chemotherapeutic strategies for the treatment of GBM require a thorough analysis of specific signaling pathways to identify those serving as drivers of GBM progression and invasion. The Wnt/β-catenin and PI3K/Akt/mTOR (PAM) signaling pathways are key regulators of important biological functions that include cell proliferation, epithelial–mesenchymal transition (EMT), metabolism, and angiogenesis. Targeting specific regulatory components of the Wnt/β-catenin and PAM pathways has the potential to disrupt critical brain tumor cell functions to achieve critical advancements in alternative GBM treatment strategies to enhance the survival rate of GBM patients. In this review, we emphasize the importance of the Wnt/β-catenin and PAM pathways for GBM invasion into brain tissue and explore their potential as therapeutic targets.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Brain Cancer Department, Asu vanda Gene Industrial Research Company, Tehran 1533666398, Iran; (A.B.B.); (Z.T.)
| | - Zahra Talaie
- Brain Cancer Department, Asu vanda Gene Industrial Research Company, Tehran 1533666398, Iran; (A.B.B.); (Z.T.)
| | - Fatemeh Jusheghani
- Department of Biotechnology, Asu vanda Gene Industrial Research Company, Tehran 1533666398, Iran;
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland;
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
- Department of Pathology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Department of Surgery, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Department of Medical Microbiology and Infectious Diseases, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine, Katowice School of Technology, 40-555 Katowice, Poland
- Correspondence:
| |
Collapse
|
242
|
Fabro F, Lamfers MLM, Leenstra S. Advancements, Challenges, and Future Directions in Tackling Glioblastoma Resistance to Small Kinase Inhibitors. Cancers (Basel) 2022; 14:600. [PMID: 35158868 PMCID: PMC8833415 DOI: 10.3390/cancers14030600] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
Despite clinical intervention, glioblastoma (GBM) remains the deadliest brain tumor in adults. Its incurability is partly related to the establishment of drug resistance, both to standard and novel treatments. In fact, even though small kinase inhibitors have changed the standard clinical practice for several solid cancers, in GBM, they did not fulfill this promise. Drug resistance is thought to arise from the heterogeneity of GBM, which leads the development of several different mechanisms. A better understanding of the evolution and characteristics of drug resistance is of utmost importance to improve the current clinical practice. Therefore, the development of clinically relevant preclinical in vitro models which allow careful dissection of these processes is crucial to gain insights that can be translated to improved therapeutic approaches. In this review, we first discuss the heterogeneity of GBM, which is reflected in the development of several resistance mechanisms. In particular, we address the potential role of drug resistance mechanisms in the failure of small kinase inhibitors in clinical trials. Finally, we discuss strategies to overcome therapy resistance, particularly focusing on the importance of developing in vitro models, and the possible approaches that could be applied to the clinic to manage drug resistance.
Collapse
Affiliation(s)
| | | | - Sieger Leenstra
- Department of Neurosurgery, Brain Tumor Center, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands; (F.F.); (M.L.M.L.)
| |
Collapse
|
243
|
Shamshiripour P, Nikoobakht M, Mansourinejad Z, Ahmadvand D, Akbarpour M. A comprehensive update to DC therapy for glioma; a systematic review and meta-analysis. Expert Rev Vaccines 2022; 21:513-531. [DOI: 10.1080/14760584.2022.2027759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Parisa Shamshiripour
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of medical imaging technology and molecular imaging, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Nikoobakht
- Department of Neurosurgery, Iran University of Medical Sciences, Tehran, Iran
| | - zahra Mansourinejad
- Department of systems biology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Davoud Ahmadvand
- Department of medical imaging technology and molecular imaging, Iran University of Medical Sciences, Tehran, Iran
| | - Mahzad Akbarpour
- Advanced Cellular Therapeutics Facility, David and Etta Jonas Center for Cellular Therapy, Hematopoietic Cellular Therapy Program, The University of Chicago Medical Center, Chicago 60637 IL, USA
- Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno-TACT), Universal Science and Education Research Network (USERN), Chicago, USA
| |
Collapse
|
244
|
Hwang H, Huang J, Khaddour K, Butt OH, Ansstas G, Chen J, Katumba RG, Kim AH, Leuthardt EC, Campian JL. Prolonged response of recurrent IDH-wild-type glioblastoma to laser interstitial thermal therapy with pembrolizumab. CNS Oncol 2022; 11:CNS81. [PMID: 35043686 PMCID: PMC8988254 DOI: 10.2217/cns-2021-0013] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Despite the improved understanding of the molecular and genetic heterogeneity of glioblastoma, there is still an unmet need for better therapeutics, as treatment approaches have remained unchanged in recent years. Research into the role of the immune microenvironment has generated enthusiasm for testing immunotherapy (specifically, immune checkpoint inhibitors). However, to date, trials of immunotherapy in glioblastoma have not demonstrated a survival advantage. Combination approaches aimed at optimally inducing response to immune checkpoint inhibitors with radiotherapy are currently being investigated. Herein, the authors describe their experience of the potential benefit and clinical outcomes of using combination pembrolizumab (an immune checkpoint inhibitor) and laser interstitial thermal therapy in a case series of patients with recurrent IDH-wild-type glioblastoma.
Collapse
Affiliation(s)
- Helen Hwang
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jiayi Huang
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Karam Khaddour
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA.,Department of Medicine, Division of Hematology & Oncology, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Omar H Butt
- Department of Neurology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - George Ansstas
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jie Chen
- Department of Pathology, Washington University School of Medicine, Saint Louis, MO 63110, USA.,Department of Pathology & Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ruth Gn Katumba
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Albert H Kim
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Eric C Leuthardt
- Department of Neurosurgery, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jian L Campian
- Department of Medicine, Division of Oncology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
245
|
Attia AM, Eltybe HA, Sedik MF, Hefni AM, Abdelgawad MI, Farrag A, Essa AA, El-Barody MM, Attia NM. The efficacy and safety of extended adjuvant temozolomide following concurrent radio-chemotherapy among Egyptian patients with newly diagnosed glioblastoma multiforme. Am J Cancer Res 2022; 12:355-370. [PMID: 35141023 PMCID: PMC8822288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023] Open
Abstract
Although concurrent radio-chemotherapy and adjuvant temozolomide (TMZ) treatment for 6 cycles has been established as a standard of care for newly diagnosed glioblastoma multiforme (GBM) patients, the recommended duration of adjuvant TMZ remains a matter of debate. Hereby, we aimed to report for the first time our experience from Upper Egypt through comparing survival and toxicity profile between two treatment modalities of adjuvant TMZ (> six cycles versus six cycles) and delineating factors of prognostic significance in Egyptian patients with newly diagnosed GBM treated by radiation therapy with concomitant and adjuvant TMZ. Between June 2016 and February 2018, the medical records of 121 patients were eligible to be retrospectively reviewed to extract the study relevant data. All patients received concurrent radio-chemotherapy, followed by TMZ for 6 cycles in 29 patients (Group 1) and for >6 cycles in 26 patients (Group 2). Patients in Group 1 had a median PFS of 15 months (95% CI: 10.215-19.785), while those in Group 2 had a median PFS of 18 months (95% CI: 16.611-19.389). After a median follow up duration of 20 months (range: 12-41), the median OS was 18 months (95% CI: 13.420-22.580) in Group 1 and 22 months (95% CI: 18.777-25.223) in Group 2. There was no statistically significant correlation between the number of chemotherapy cycles and PFS (P=0.513) or OS (P=0.867). The extent of surgical resection was the only independent prognostic factor for both PFS (P=0.015) and OS (P=0.028) by multivariate analysis. Three grade ≥3 hematologic toxicity were encountered in 3 patients. One in the six-cycle group (neutropenia), and two in the extended cycles group (one had neutropenia and the other one developed thrombocytopenia). No statistically significant difference in the toxicity profile between both groups. The results of our study suggest that extended TMZ therapy is safe and tolerable, however it did not significantly improve PFS or OS as compared to the standard six-cycle course. Larger randomized studies are required to shed more light on this issue.
Collapse
Affiliation(s)
- Alia M Attia
- Radiation Oncology Department, South Egypt Cancer Institute, Assiut UniversityAssiut 71515, Egypt
| | - Hanan A Eltybe
- Department of Medical Oncology and Hematological Malignancies, South Egypt Cancer Institute, Assiut UniversityAssiut 71111, Egypt
| | - Mayada F Sedik
- Department of Medical Oncology and Hematological Malignancies, South Egypt Cancer Institute, Assiut UniversityAssiut 71111, Egypt
| | - Ahmed Mubarak Hefni
- Department of Medical Oncology and Hematological Malignancies, South Egypt Cancer Institute, Assiut UniversityAssiut 71111, Egypt
| | | | - Ashraf Farrag
- Clinical Oncology Department, Assiut UniversityAssiut 71111, Egypt
| | | | - Mohamed M El-Barody
- Radiology Department, South Egypt Cancer Institute, Assiut UniversityAssiut 71111, Egypt
| | - Noha M Attia
- Radiology Department, Assiut University Hospital, Assiut UniversityAssiut 71515, Egypt
| |
Collapse
|
246
|
Craig DJ, Bailey MM, Noe OB, Williams KK, Stanbery L, Hamouda DM, Nemunaitis JJ. Subclonal landscape of cancer drives resistance to immune therapy. Cancer Treat Res Commun 2022; 30:100507. [PMID: 35007928 DOI: 10.1016/j.ctarc.2021.100507] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023]
Abstract
Tumor mutation burden (TMB) is often used as a biomarker for immunogenicity and prerequisite for immune checkpoint inhibitor (ICI) therapy. However, it is becoming increasingly evident that not all tumors with high TMB respond to ICIs as expected. It has been shown that the ability of T-cells to infiltrate the tumor microenvironment and elicit a specific immune response is dependent not only on the TMB, but also on intra-tumor heterogeneity and the fraction of low-frequency subclonal mutations that make up the tumor. High intra-tumor heterogeneity leads to inefficient recognition of tumor neoantigens by T-cells due to their diluted frequency and spatial heterogeneity. Clinical studies have shown that tumors with a high degree of intra-tumor heterogeneity respond poorly to ICI therapy, and previous cytotoxic treatment may increase the intra-tumor heterogeneity and render second-line ICI therapy less effective. This paper reviews the role of ICI therapy when following chemotherapy or radiation to determine if they may be better suited as first-line therapy in patients with high TMB, low intra-tumor heterogeneity, and high PD-1, PD-L1, or CTLA-4 expression.
Collapse
Affiliation(s)
- Daniel J Craig
- University of Toledo Medical Center, Toledo, OH, 43614, USA
| | | | - Olivia B Noe
- University of Toledo Medical Center, Toledo, OH, 43614, USA
| | | | | | | | | |
Collapse
|
247
|
Yearley AG, Iorgulescu JB, Chiocca EA, Peruzzi PP, Smith TR, Reardon DA, Mooney MA. The current state of glioma data registries. Neurooncol Adv 2022; 4:vdac099. [PMID: 36196363 PMCID: PMC9521197 DOI: 10.1093/noajnl/vdac099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Background The landscape of glioma research has evolved in the past 20 years to include numerous large, multi-institutional, database efforts compiling either clinical data on glioma patients, molecular data on glioma specimens, or a combination of both. While these strategies can provide a wealth of information for glioma research, obtaining information regarding data availability and access specifications can be challenging. Methods We reviewed the literature for ongoing clinical, molecular, and combined database efforts related to glioma research to provide researchers with a curated overview of the current state of glioma database resources. Results We identified and reviewed a total of 20 databases with data collection spanning from 1975 to 2022. Surveyed databases included both low- and high-grade gliomas, and data elements included over 100 clinical variables and 12 molecular data types. Select database strengths included large sample sizes and a wide variety of variables available, while limitations of some databases included complex data access requirements and a lack of glioma-specific variables. Conclusions This review highlights current databases and registries and their potential utility in clinical and genomic glioma research. While many high-quality resources exist, the fluid nature of glioma taxonomy makes it difficult to isolate a large cohort of patients with a pathologically confirmed diagnosis. Large, well-defined, and publicly available glioma datasets have the potential to expand the reach of glioma research and drive the field forward.
Collapse
Affiliation(s)
- Alexander G Yearley
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Julian Bryan Iorgulescu
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Ennio Antonio Chiocca
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Pier Paolo Peruzzi
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Timothy R Smith
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Michael A Mooney
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
248
|
Luo J, Junaid M, Hamid N, Duan JJ, Yang X, Pei DS. Current understanding of gliomagenesis: from model to mechanism. Int J Med Sci 2022; 19:2071-2079. [PMID: 36483593 PMCID: PMC9724244 DOI: 10.7150/ijms.77287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/03/2022] [Indexed: 11/24/2022] Open
Abstract
Glioma, a kind of central nervous system (CNS) tumor, is hard to cure and accounts for 32% of all CNS tumors. Establishing a stable glioma model is critically important to investigate the underlying molecular mechanisms involved in tumorigenesis and tumor progression. Various core signaling pathways have been identified in gliomagenesis, such as RTK/RAS/PI3K, TP53, and RB1. Traditional methods of establishing glioma animal models have included chemical induction, xenotransplantation, and genetic modifications (RCAS/t-va system, Cre-loxP, and TALENs). Recently, CRISPR/Cas9 has emerged as an efficient gene editing tool with high germline transmission and has extended the scope of stable and efficient glioma models that can be generated. Therefore, this review will highlight the documented evidence about the molecular characteristics, critical genetic markers, and signaling pathways responsible for gliomagenesis and progression. Moreover, methods of establishing glioma models using gene editing techniques and therapeutic aspects will be discussed. Finally, the prospect of applying gene editing in glioma by using CRISPR/Cas9 strategy and future research directions to establish a stable glioma model are also included in this review. In-depth knowledge of glioma signaling pathways and use of CRISPR/Cas9 can greatly assist in the development of a stable, efficient, and spontaneous glioma model, which can ultimately improve the effectiveness of therapeutic responses and cure glioma patients.
Collapse
Affiliation(s)
- Juanjuan Luo
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
| | - Muhammad Junaid
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Naima Hamid
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin-Jing Duan
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
| | - Xiaojun Yang
- Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- ✉ Corresponding authors: De-Sheng Pei, E-mail: ; Xiaojun Yang, E-mail:
| | - De-Sheng Pei
- School of Public Health and Management, Chongqing Medical University, Chongqing 400016, China
- ✉ Corresponding authors: De-Sheng Pei, E-mail: ; Xiaojun Yang, E-mail:
| |
Collapse
|
249
|
Li X, Thirumalai D. A mathematical model for phenotypic heterogeneity in breast cancer with implications for therapeutic strategies. J R Soc Interface 2022; 19:20210803. [PMID: 35078336 PMCID: PMC8790361 DOI: 10.1098/rsif.2021.0803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Inevitably, almost all cancer patients develop resistance to targeted therapy. Intratumour heterogeneity is a major cause of drug resistance. Mathematical models that explain experiments quantitatively are useful in understanding the origin of intratumour heterogeneity, which then could be used to explore scenarios for efficacious therapy. Here, we develop a mathematical model to investigate intratumour heterogeneity in breast cancer by exploiting the observation that HER2+ and HER2- cells could divide symmetrically or asymmetrically. Our predictions for the evolution of cell fractions are in quantitative agreement with single-cell experiments. Remarkably, the colony size of HER2+ cells emerging from a single HER2- cell (or vice versa), which occurs in about four cell doublings, also agrees with experimental results, without tweaking any parameter in the model. The theory explains experimental data on the responses of breast tumours under different treatment protocols. We then used the model to predict that, not only the order of two drugs, but also the treatment period for each drug and the tumour cell plasticity could be manipulated to improve the treatment efficacy. Mathematical models, when integrated with data on patients, make possible exploration of a broad range of parameters readily, which might provide insights in devising effective therapies.
Collapse
Affiliation(s)
- Xin Li
- Department of Chemistry, University of Texas, Austin, TX 78712, USA
| | - D. Thirumalai
- Department of Chemistry, University of Texas, Austin, TX 78712, USA
| |
Collapse
|
250
|
Van Gool SW, Makalowski J, Bitar M, Van de Vliet P, Schirrmacher V, Stuecker W. Synergy between TMZ and individualized multimodal immunotherapy to improve overall survival of IDH1 wild-type MGMT promoter-unmethylated GBM patients. Genes Immun 2022; 23:255-259. [PMID: 35173295 PMCID: PMC9758045 DOI: 10.1038/s41435-022-00162-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 01/07/2023]
Abstract
The prognosis of IDH1 wild-type MGMT promoter-unmethylated GBM patients remains poor. Addition of Temozolomide (TMZ) to first-line local treatment shifted the median overall survival (OS) from 11.8 to 12.6 months. We retrospectively analyzed the value of individualized multimodal immunotherapy (IMI) to improve OS in these patients. All adults meeting the criteria and treated 06/2015-06/2021 were selected. Thirty-two patients (12f, 20m) had a median age of 47 y (range 18-69) and a KPI of 70 (50-100). Extent of resection was complete (11), <complete (12) or not documented (9). Seven patients were treated with surgery/radio(chemo)therapy and subsequent IMI (Group-1); 25 patients were treated with radiochemotherapy followed by maintenance TMZ plus IMI during and after TMZ (Group-2). Age, KPI and extent of resection were not different amongst both groups. The median OS of group-1 patients was 11 m (2 y OS: 0%). Surprisingly the median OS of group-2 patients was 22 m with 2 y OS of 36% (CI95%: 16-57), which was significantly (Log-rank: p = 0.0001) different from group-1. The data suggest that addition of IMI after local therapy on its own has no relevant effect on OS in these GBM patients, similar to maintenance TMZ. However, the combination of both TMZ + IMI significantly improved OS.
Collapse
Affiliation(s)
- Stefaan W. Van Gool
- Immun-Onkologisches Zentrum Köln (IOZK), Hohenstaufenring 30-32, 50674 Köln, Germany
| | - Jennifer Makalowski
- Immun-Onkologisches Zentrum Köln (IOZK), Hohenstaufenring 30-32, 50674 Köln, Germany
| | - Michael Bitar
- Immun-Onkologisches Zentrum Köln (IOZK), Hohenstaufenring 30-32, 50674 Köln, Germany
| | - Peter Van de Vliet
- Immun-Onkologisches Zentrum Köln (IOZK), Hohenstaufenring 30-32, 50674 Köln, Germany
| | - Volker Schirrmacher
- Immun-Onkologisches Zentrum Köln (IOZK), Hohenstaufenring 30-32, 50674 Köln, Germany
| | - Wilfried Stuecker
- Immun-Onkologisches Zentrum Köln (IOZK), Hohenstaufenring 30-32, 50674 Köln, Germany
| |
Collapse
|