Skip to main content
Top
Published in: BMC Cancer 1/2024

Open Access 01-12-2024 | Glioma | Research

IDH1 mutation is detectable in plasma cell-free DNA and is associated with survival outcome in glioma patients

Authors: Stefania Crucitta, Francesco Pasqualetti, Alessandra Gonnelli, Martina Ruglioni, Giovanna Irene Luculli, Martina Cantarella, Valerio Ortenzi, Cristian Scatena, Fabiola Paiar, Antonio Giuseppe Naccarato, Romano Danesi, Marzia Del Re

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

Background

Circulating cell-free DNA (cfDNA, liquid biopsy) is a powerful tool to detect molecular alterations. However, depending on tumor characteristics, biology and anatomic localization, cfDNA detection and analysis may be challenging. Gliomas are enclosed into an anatomic sanctuary, which obstacles the release of cfDNA into the peripheral blood. Therefore, the advantages of using liquid biopsy for brain tumors is still to be confirmed. The present study evaluates the ability of liquid biopsy to detect IDH1 mutations and its correlation with survival and clinical characteristics of glioma patients.

Methods

Blood samples obtained from glioma patients were collected after surgery prior to the adjuvant therapy. cfDNA was extracted from plasma and IDH1 p.R132H mutation analysis was performed on a digital droplet PCR. χ2-test and Cohen k were used to assess the correlation between plasma and tissue IDH1 status, while Kaplan Meier curve and Cox regression analysis were applied to survival analysis. Statistical calculations were performed by MedCalc and GraphPad Prism software.

Results

A total of 67 samples were collected. A concordance between IDH1 status in tissue and in plasma was found (p = 0.0024), and the presence of the IDH1 mutation both in tissue (138.8 months vs 24.4, p < 0.0001) and cfDNA (116.3 months vs 35.8, p = 0.016) was associated with longer median OS. A significant association between IDH1 mutation both in tissue and cfDNA, age, tumor grade and OS was demonstrated by univariate Cox regression analysis. No statistically significant association between IDH1 mutation and tumor grade was found (p = 0.10).

Conclusions

The present study demonstrates that liquid biopsy may be used in brain tumors to detect IDH1 mutation which represents an important prognostic biomarker in patients with different types of gliomas, being associated to OS.
Literature
1.
go back to reference Donaldson J, Park BH. Circulating tumor DNA: measurement and clinical utility. Annu Rev Med. 2018;69:223–34.PubMedCrossRef Donaldson J, Park BH. Circulating tumor DNA: measurement and clinical utility. Annu Rev Med. 2018;69:223–34.PubMedCrossRef
2.
go back to reference Ossandon MR, Agrawal L, Bernhard EJ, Conley BA, Dey SM, Divi RL, et al. Circulating tumor DNA assays in clinical Cancer research. J Natl Cancer Inst. 2018;110(9):929–34.PubMedPubMedCentralCrossRef Ossandon MR, Agrawal L, Bernhard EJ, Conley BA, Dey SM, Divi RL, et al. Circulating tumor DNA assays in clinical Cancer research. J Natl Cancer Inst. 2018;110(9):929–34.PubMedPubMedCentralCrossRef
4.
go back to reference Mader S, Pantel K. Liquid biopsy: current status and future perspectives. Oncol Res Treat. 2017;40(7–8):404–8.PubMedCrossRef Mader S, Pantel K. Liquid biopsy: current status and future perspectives. Oncol Res Treat. 2017;40(7–8):404–8.PubMedCrossRef
5.
6.
go back to reference Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10(8):472–84.PubMedCrossRef Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10(8):472–84.PubMedCrossRef
7.
go back to reference Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol. 2017;14(9):531–48.PubMedCrossRef Siravegna G, Marsoni S, Siena S, Bardelli A. Integrating liquid biopsies into the management of cancer. Nat Rev Clin Oncol. 2017;14(9):531–48.PubMedCrossRef
8.
go back to reference Shohdy KS, West HJ. Circulating tumor DNA testing-liquid biopsy of a Cancer. JAMA Oncol. 2020;6(5):792.PubMedCrossRef Shohdy KS, West HJ. Circulating tumor DNA testing-liquid biopsy of a Cancer. JAMA Oncol. 2020;6(5):792.PubMedCrossRef
9.
go back to reference Pascual J, Attard G, Bidard FC, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO precision medicine working group. Ann Oncol. 2022;33(8):750–68.PubMedCrossRef Pascual J, Attard G, Bidard FC, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: a report from the ESMO precision medicine working group. Ann Oncol. 2022;33(8):750–68.PubMedCrossRef
10.
go back to reference Michela B. Liquid biopsy: a family of possible diagnostic tools. Diagnostics (Basel). 2021;11(8). Michela B. Liquid biopsy: a family of possible diagnostic tools. Diagnostics (Basel). 2021;11(8).
11.
go back to reference Urabe F, Kosaka N, Ito K, Kimura T, Egawa S, Ochiya T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am J Phys Cell Physiol. 2020;318(1):C29–39.CrossRef Urabe F, Kosaka N, Ito K, Kimura T, Egawa S, Ochiya T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am J Phys Cell Physiol. 2020;318(1):C29–39.CrossRef
12.
go back to reference Vasconcelos MH, Caires HR, Abols A, Xavier CPR, Line A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist Updat. 2019;47:100647.PubMedCrossRef Vasconcelos MH, Caires HR, Abols A, Xavier CPR, Line A. Extracellular vesicles as a novel source of biomarkers in liquid biopsies for monitoring cancer progression and drug resistance. Drug Resist Updat. 2019;47:100647.PubMedCrossRef
13.
go back to reference Taverna S, Giallombardo M, Gil-Bazo I, Carreca AP, Castiglia M, Chacartegui J, et al. Exosomes isolation and characterization in serum is feasible in non-small cell lung cancer patients: critical analysis of evidence and potential role in clinical practice. Oncotarget. 2016;7(19):28748–60.PubMedPubMedCentralCrossRef Taverna S, Giallombardo M, Gil-Bazo I, Carreca AP, Castiglia M, Chacartegui J, et al. Exosomes isolation and characterization in serum is feasible in non-small cell lung cancer patients: critical analysis of evidence and potential role in clinical practice. Oncotarget. 2016;7(19):28748–60.PubMedPubMedCentralCrossRef
14.
go back to reference Rolfo C, Mack P, Scagliotti GV, Aggarwal C, Arcila ME, Barlesi F, et al. Liquid biopsy for advanced NSCLC: a consensus statement from the International Association for the Study of Lung Cancer. J Thorac Oncol. 2021;16(10):1647–62.PubMedCrossRef Rolfo C, Mack P, Scagliotti GV, Aggarwal C, Arcila ME, Barlesi F, et al. Liquid biopsy for advanced NSCLC: a consensus statement from the International Association for the Study of Lung Cancer. J Thorac Oncol. 2021;16(10):1647–62.PubMedCrossRef
15.
go back to reference Passiglia F, Pilotto S, Facchinetti F, Bertolaccini L, Del Re M, Ferrara R, et al. Treatment of advanced non-small-cell lung cancer: the 2019 AIOM (Italian Association of Medical Oncology) clinical practice guidelines. Crit Rev Oncol Hematol. 2020;146:102858.PubMedCrossRef Passiglia F, Pilotto S, Facchinetti F, Bertolaccini L, Del Re M, Ferrara R, et al. Treatment of advanced non-small-cell lung cancer: the 2019 AIOM (Italian Association of Medical Oncology) clinical practice guidelines. Crit Rev Oncol Hematol. 2020;146:102858.PubMedCrossRef
16.
go back to reference Heidrich I, Ackar L, Mossahebi Mohammadi P, Pantel K. Liquid biopsies: potential and challenges. Int J Cancer. 2021;148(3):528–45.PubMedCrossRef Heidrich I, Ackar L, Mossahebi Mohammadi P, Pantel K. Liquid biopsies: potential and challenges. Int J Cancer. 2021;148(3):528–45.PubMedCrossRef
17.
go back to reference Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26–41.PubMedCrossRef Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26–41.PubMedCrossRef
18.
go back to reference Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, et al. Liquid biopsy for advanced non-Small cell lung Cancer (NSCLC): a statement paper from the IASLC. J Thorac Oncol. 2018;13(9):1248–68.PubMedCrossRef Rolfo C, Mack PC, Scagliotti GV, Baas P, Barlesi F, Bivona TG, et al. Liquid biopsy for advanced non-Small cell lung Cancer (NSCLC): a statement paper from the IASLC. J Thorac Oncol. 2018;13(9):1248–68.PubMedCrossRef
19.
go back to reference Del Re M, Bordi P, Rofi E, Restante G, Valleggi S, Minari R, et al. The amount of activating EGFR mutations in circulating cell-free DNA is a marker to monitor osimertinib response. Br J Cancer. 2018;119(10):1252–8.PubMedPubMedCentralCrossRef Del Re M, Bordi P, Rofi E, Restante G, Valleggi S, Minari R, et al. The amount of activating EGFR mutations in circulating cell-free DNA is a marker to monitor osimertinib response. Br J Cancer. 2018;119(10):1252–8.PubMedPubMedCentralCrossRef
20.
go back to reference Del Re M, Petrini I, Mazzoni F, Valleggi S, Gianfilippo G, Pozzessere D, et al. Incidence of T790M in patients with NSCLC progressed to Gefitinib, Erlotinib, and Afatinib: a study on circulating cell-free DNA. Clin Lung Cancer. 2020;21(3):232–7.PubMedCrossRef Del Re M, Petrini I, Mazzoni F, Valleggi S, Gianfilippo G, Pozzessere D, et al. Incidence of T790M in patients with NSCLC progressed to Gefitinib, Erlotinib, and Afatinib: a study on circulating cell-free DNA. Clin Lung Cancer. 2020;21(3):232–7.PubMedCrossRef
21.
22.
go back to reference Groot VP, Mosier S, Javed AA, Teinor JA, Gemenetzis G, Ding D, et al. Circulating tumor DNA as a clinical test in resected pancreatic Cancer. Clin Cancer Res. 2019;25(16):4973–84.PubMedPubMedCentralCrossRef Groot VP, Mosier S, Javed AA, Teinor JA, Gemenetzis G, Ding D, et al. Circulating tumor DNA as a clinical test in resected pancreatic Cancer. Clin Cancer Res. 2019;25(16):4973–84.PubMedPubMedCentralCrossRef
23.
go back to reference Schwaederle M, Chattopadhyay R, Kato S, Fanta PT, Banks KC, Choi IS, et al. Genomic alterations in circulating tumor DNA from diverse Cancer patients identified by next-generation sequencing. Cancer Res. 2017;77(19):5419–27.PubMedPubMedCentralCrossRef Schwaederle M, Chattopadhyay R, Kato S, Fanta PT, Banks KC, Choi IS, et al. Genomic alterations in circulating tumor DNA from diverse Cancer patients identified by next-generation sequencing. Cancer Res. 2017;77(19):5419–27.PubMedPubMedCentralCrossRef
24.
go back to reference Zill OA, Banks KC, Fairclough SR, Mortimer SA, Vowles JV, Mokhtari R, et al. The landscape of actionable genomic alterations in cell-free circulating tumor DNA from 21,807 advanced Cancer patients. Clin Cancer Res. 2018;24(15):3528–38.PubMedCrossRef Zill OA, Banks KC, Fairclough SR, Mortimer SA, Vowles JV, Mokhtari R, et al. The landscape of actionable genomic alterations in cell-free circulating tumor DNA from 21,807 advanced Cancer patients. Clin Cancer Res. 2018;24(15):3528–38.PubMedCrossRef
25.
go back to reference Gracie L, Pan Y, Atenafu EG, Ward DG, Teng M, Pallan L, et al. Circulating tumour DNA (ctDNA) in metastatic melanoma, a systematic review and meta-analysis. Eur J Cancer. 2021;158:191–207.PubMedCrossRef Gracie L, Pan Y, Atenafu EG, Ward DG, Teng M, Pallan L, et al. Circulating tumour DNA (ctDNA) in metastatic melanoma, a systematic review and meta-analysis. Eur J Cancer. 2021;158:191–207.PubMedCrossRef
26.
go back to reference Marczynski GT, Laus AC, Dos Reis MB, Reis RM, Vazquez VL. Circulating tumor DNA (ctDNA) detection is associated with shorter progression-free survival in advanced melanoma patients. Sci Rep. 2020;10(1):18682.PubMedPubMedCentralCrossRef Marczynski GT, Laus AC, Dos Reis MB, Reis RM, Vazquez VL. Circulating tumor DNA (ctDNA) detection is associated with shorter progression-free survival in advanced melanoma patients. Sci Rep. 2020;10(1):18682.PubMedPubMedCentralCrossRef
27.
go back to reference Malla M, Loree JM, Kasi PM, Parikh AR. Using circulating tumor DNA in colorectal Cancer: current and evolving practices. J Clin Oncol. 2022;40(24):2846–57.PubMedPubMedCentralCrossRef Malla M, Loree JM, Kasi PM, Parikh AR. Using circulating tumor DNA in colorectal Cancer: current and evolving practices. J Clin Oncol. 2022;40(24):2846–57.PubMedPubMedCentralCrossRef
28.
go back to reference Kang Y, Lin X, Kang D. Diagnostic value of circulating tumor DNA in molecular characterization of glioma: a meta-analysis. Medicine (Baltimore). 2020;99(33):e21196.PubMedCrossRef Kang Y, Lin X, Kang D. Diagnostic value of circulating tumor DNA in molecular characterization of glioma: a meta-analysis. Medicine (Baltimore). 2020;99(33):e21196.PubMedCrossRef
29.
go back to reference Boisselier B, Gallego Perez-Larraya J, Rossetto M, Labussiere M, Ciccarino P, Marie Y, et al. Detection of IDH1 mutation in the plasma of patients with glioma. Neurology. 2012;79(16):1693–8.PubMedCrossRef Boisselier B, Gallego Perez-Larraya J, Rossetto M, Labussiere M, Ciccarino P, Marie Y, et al. Detection of IDH1 mutation in the plasma of patients with glioma. Neurology. 2012;79(16):1693–8.PubMedCrossRef
30.
go back to reference Cabezas-Camarero S, Garcia-Barberan V, Perez-Alfayate R, Casado-Farinas I, Sloane H, Jones FS, et al. Detection of IDH1 mutations in plasma using BEAMing Technology in Patients with gliomas. Cancers (Basel). 2022;14(12). Cabezas-Camarero S, Garcia-Barberan V, Perez-Alfayate R, Casado-Farinas I, Sloane H, Jones FS, et al. Detection of IDH1 mutations in plasma using BEAMing Technology in Patients with gliomas. Cancers (Basel). 2022;14(12).
31.
go back to reference Nassiri F, Chakravarthy A, Feng S, Shen SY, Nejad R, Zuccato JA, et al. Detection and discrimination of intracranial tumors using plasma cell-free DNA methylomes. Nat Med. 2020;26(7):1044–7.PubMedPubMedCentralCrossRef Nassiri F, Chakravarthy A, Feng S, Shen SY, Nejad R, Zuccato JA, et al. Detection and discrimination of intracranial tumors using plasma cell-free DNA methylomes. Nat Med. 2020;26(7):1044–7.PubMedPubMedCentralCrossRef
32.
go back to reference Piccioni DE, Achrol AS, Kiedrowski LA, Banks KC, Boucher N, Barkhoudarian G, et al. Analysis of cell-free circulating tumor DNA in 419 patients with glioblastoma and other primary brain tumors. CNS. Oncol. 2019;8(2):CNS34. Piccioni DE, Achrol AS, Kiedrowski LA, Banks KC, Boucher N, Barkhoudarian G, et al. Analysis of cell-free circulating tumor DNA in 419 patients with glioblastoma and other primary brain tumors. CNS. Oncol. 2019;8(2):CNS34.
33.
go back to reference Yi Z, Qu C, Zeng Y, Liu Z. Liquid biopsy: early and accurate diagnosis of brain tumor. J Cancer Res Clin Oncol. 2022;148(9):2347–73.PubMedCrossRef Yi Z, Qu C, Zeng Y, Liu Z. Liquid biopsy: early and accurate diagnosis of brain tumor. J Cancer Res Clin Oncol. 2022;148(9):2347–73.PubMedCrossRef
34.
go back to reference Simonelli M, Dipasquale A, Orzan F, Lorenzi E, Persico P, Navarria P, et al. Cerebrospinal fluid tumor DNA for liquid biopsy in glioma patients' management: close to the clinic? Crit Rev Oncol Hematol. 2020;146:102879.PubMedCrossRef Simonelli M, Dipasquale A, Orzan F, Lorenzi E, Persico P, Navarria P, et al. Cerebrospinal fluid tumor DNA for liquid biopsy in glioma patients' management: close to the clinic? Crit Rev Oncol Hematol. 2020;146:102879.PubMedCrossRef
35.
go back to reference Sareen H, Garrett C, Lynch D, Powter B, Brungs D, Cooper A, et al. The role of liquid biopsies in detecting molecular tumor biomarkers in brain Cancer patients. Cancers (Basel). 2020;12(7). Sareen H, Garrett C, Lynch D, Powter B, Brungs D, Cooper A, et al. The role of liquid biopsies in detecting molecular tumor biomarkers in brain Cancer patients. Cancers (Basel). 2020;12(7).
36.
go back to reference Eibl RH, Schneemann M. Liquid biopsy and primary brain tumors. Cancers (Basel). 2021;13(21). Eibl RH, Schneemann M. Liquid biopsy and primary brain tumors. Cancers (Basel). 2021;13(21).
37.
go back to reference Ray A. Liquid biopsy in gliomas- a review. Neurol India. 2020;68(6):1295–300.PubMed Ray A. Liquid biopsy in gliomas- a review. Neurol India. 2020;68(6):1295–300.PubMed
38.
go back to reference Orzan F, De Bacco F, Lazzarini E, Crisafulli G, Gasparini A, Dipasquale A, et al. Liquid biopsy of cerebrospinal fluid enables selective profiling of glioma molecular subtypes at first clinical presentation. Clin Cancer Res. 2023;29(7):1252–66.PubMedPubMedCentralCrossRef Orzan F, De Bacco F, Lazzarini E, Crisafulli G, Gasparini A, Dipasquale A, et al. Liquid biopsy of cerebrospinal fluid enables selective profiling of glioma molecular subtypes at first clinical presentation. Clin Cancer Res. 2023;29(7):1252–66.PubMedPubMedCentralCrossRef
40.
go back to reference De Mattos-Arruda L, Mayor R, Ng CKY, Weigelt B, Martinez-Ricarte F, Torrejon D, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:8839.PubMedPubMedCentralCrossRef De Mattos-Arruda L, Mayor R, Ng CKY, Weigelt B, Martinez-Ricarte F, Torrejon D, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat Commun. 2015;6:8839.PubMedPubMedCentralCrossRef
41.
go back to reference Seoane J, De Mattos-Arruda L, Le Rhun E, Bardelli A, Weller M. Cerebrospinal fluid cell-free tumour DNA as a liquid biopsy for primary brain tumours and central nervous system metastases. Ann Oncol. 2019;30(2):211–8.PubMedCrossRef Seoane J, De Mattos-Arruda L, Le Rhun E, Bardelli A, Weller M. Cerebrospinal fluid cell-free tumour DNA as a liquid biopsy for primary brain tumours and central nervous system metastases. Ann Oncol. 2019;30(2):211–8.PubMedCrossRef
42.
go back to reference Miller AM, Shah RH, Pentsova EI, Pourmaleki M, Briggs S, Distefano N, et al. Tracking tumour evolution in glioma through liquid biopsies of cerebrospinal fluid. Nature. 2019;565(7741):654–8.PubMedPubMedCentralCrossRef Miller AM, Shah RH, Pentsova EI, Pourmaleki M, Briggs S, Distefano N, et al. Tracking tumour evolution in glioma through liquid biopsies of cerebrospinal fluid. Nature. 2019;565(7741):654–8.PubMedPubMedCentralCrossRef
43.
44.
go back to reference Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet. 2018;392(10145):432–46.PubMedCrossRef Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet. 2018;392(10145):432–46.PubMedCrossRef
45.
go back to reference Wesseling P, Capper D. WHO 2016 classification of gliomas. Neuropathol Appl Neurobiol. 2018;44(2):139–50.PubMedCrossRef Wesseling P, Capper D. WHO 2016 classification of gliomas. Neuropathol Appl Neurobiol. 2018;44(2):139–50.PubMedCrossRef
47.
go back to reference Picca A, Berzero G, Di Stefano AL, Sanson M. The clinical use of IDH1 and IDH2 mutations in gliomas. Expert Rev Mol Diagn. 2018;18(12):1041–51.PubMedCrossRef Picca A, Berzero G, Di Stefano AL, Sanson M. The clinical use of IDH1 and IDH2 mutations in gliomas. Expert Rev Mol Diagn. 2018;18(12):1041–51.PubMedCrossRef
48.
go back to reference Sonoda Y, Kumabe T, Nakamura T, Saito R, Kanamori M, Yamashita Y, et al. Analysis of IDH1 and IDH2 mutations in Japanese glioma patients. Cancer Sci. 2009;100(10):1996–8.PubMedCrossRef Sonoda Y, Kumabe T, Nakamura T, Saito R, Kanamori M, Yamashita Y, et al. Analysis of IDH1 and IDH2 mutations in Japanese glioma patients. Cancer Sci. 2009;100(10):1996–8.PubMedCrossRef
49.
go back to reference Liu Y, Lang F, Chou FJ, Zaghloul KA, Yang C. Isocitrate dehydrogenase mutations in glioma: genetics, biochemistry, and clinical indications. Biomedicines. 2020;8(9). Liu Y, Lang F, Chou FJ, Zaghloul KA, Yang C. Isocitrate dehydrogenase mutations in glioma: genetics, biochemistry, and clinical indications. Biomedicines. 2020;8(9).
51.
go back to reference Khan I, Waqas M, Shamim MS. Prognostic significance of IDH 1 mutation in patients with glioblastoma multiforme. J Pak Med Assoc. 2017;67(5):816–7.PubMed Khan I, Waqas M, Shamim MS. Prognostic significance of IDH 1 mutation in patients with glioblastoma multiforme. J Pak Med Assoc. 2017;67(5):816–7.PubMed
52.
go back to reference Hartmann C, Hentschel B, Wick W, Capper D, Felsberg J, Simon M, et al. Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol. 2010;120(6):707–18.PubMedCrossRef Hartmann C, Hentschel B, Wick W, Capper D, Felsberg J, Simon M, et al. Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol. 2010;120(6):707–18.PubMedCrossRef
53.
go back to reference Xia L, Wu B, Fu Z, Feng F, Qiao E, Li Q, et al. Prognostic role of IDH mutations in gliomas: a meta-analysis of 55 observational studies. Oncotarget. 2015;6(19):17354–65.PubMedPubMedCentralCrossRef Xia L, Wu B, Fu Z, Feng F, Qiao E, Li Q, et al. Prognostic role of IDH mutations in gliomas: a meta-analysis of 55 observational studies. Oncotarget. 2015;6(19):17354–65.PubMedPubMedCentralCrossRef
54.
go back to reference Pappula AL, Rasheed S, Mirzaei G, Petreaca RC, Bouley RA. A genome-wide profiling of glioma patients with an IDH1 mutation using the catalogue of somatic mutations in Cancer database. Cancers (Basel). 2021;13(17). Pappula AL, Rasheed S, Mirzaei G, Petreaca RC, Bouley RA. A genome-wide profiling of glioma patients with an IDH1 mutation using the catalogue of somatic mutations in Cancer database. Cancers (Basel). 2021;13(17).
55.
go back to reference Stoyanov GS, Lyutfi E, Georgieva R, Georgiev R, Dzhenkov DL, Petkova L, et al. Reclassification of glioblastoma Multiforme according to the 2021 World Health Organization classification of central nervous system tumors: a single institution report and practical significance. Cureus. 2022;14(2):e21822.PubMedPubMedCentral Stoyanov GS, Lyutfi E, Georgieva R, Georgiev R, Dzhenkov DL, Petkova L, et al. Reclassification of glioblastoma Multiforme according to the 2021 World Health Organization classification of central nervous system tumors: a single institution report and practical significance. Cureus. 2022;14(2):e21822.PubMedPubMedCentral
56.
go back to reference Sledzinska P, Bebyn M, Szczerba E, Furtak J, Harat M, Olszewska N, et al. Glioma 2021 WHO classification: the superiority of NGS over IHC in routine diagnostics. Mol Diagn Ther. 2022;26(6):699–713.PubMedPubMedCentralCrossRef Sledzinska P, Bebyn M, Szczerba E, Furtak J, Harat M, Olszewska N, et al. Glioma 2021 WHO classification: the superiority of NGS over IHC in routine diagnostics. Mol Diagn Ther. 2022;26(6):699–713.PubMedPubMedCentralCrossRef
57.
go back to reference Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro-Oncology. 2021;23(8):1231–51.PubMedPubMedCentralCrossRef Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro-Oncology. 2021;23(8):1231–51.PubMedPubMedCentralCrossRef
58.
go back to reference Mellinghoff IK, van den Bent MJ, Blumenthal DT, Touat M, Peters KB, Clarke J, et al. Vorasidenib in IDH1- or IDH2-mutant low-grade glioma. N Engl J Med. 2023;389(7):589–601.PubMedCrossRef Mellinghoff IK, van den Bent MJ, Blumenthal DT, Touat M, Peters KB, Clarke J, et al. Vorasidenib in IDH1- or IDH2-mutant low-grade glioma. N Engl J Med. 2023;389(7):589–601.PubMedCrossRef
59.
go back to reference Crucitta S, Ruglioni M, Novi C, Manganiello M, Arici R, Petrini I, et al. Comparison of digital PCR systems for the analysis of liquid biopsy samples of patients affected by lung and colorectal cancer. Clin Chim Acta. 2023;541:117239.PubMedCrossRef Crucitta S, Ruglioni M, Novi C, Manganiello M, Arici R, Petrini I, et al. Comparison of digital PCR systems for the analysis of liquid biopsy samples of patients affected by lung and colorectal cancer. Clin Chim Acta. 2023;541:117239.PubMedCrossRef
60.
go back to reference Nørøxe DS, Østrup O, Yde CW, Ahlborn LB, Nielsen FC, Michaelsen SR, et al. Cell-free DNA in newly diagnosed patients with glioblastoma - a clinical prospective feasibility study. Oncotarget. 2019;10(43):4397–406.PubMedPubMedCentralCrossRef Nørøxe DS, Østrup O, Yde CW, Ahlborn LB, Nielsen FC, Michaelsen SR, et al. Cell-free DNA in newly diagnosed patients with glioblastoma - a clinical prospective feasibility study. Oncotarget. 2019;10(43):4397–406.PubMedPubMedCentralCrossRef
61.
go back to reference Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426–37.PubMedCrossRef Schwarzenbach H, Hoon DS, Pantel K. Cell-free nucleic acids as biomarkers in cancer patients. Nat Rev Cancer. 2011;11(6):426–37.PubMedCrossRef
62.
go back to reference Husain A, Mishra S, Siddiqui MH, Husain N. Detection of IDH1 mutation in cfDNA and tissue of adult diffuse glioma with allele-specific qPCR. Asian Pac J Cancer Prev. 2023;24(3):961–8.PubMedPubMedCentralCrossRef Husain A, Mishra S, Siddiqui MH, Husain N. Detection of IDH1 mutation in cfDNA and tissue of adult diffuse glioma with allele-specific qPCR. Asian Pac J Cancer Prev. 2023;24(3):961–8.PubMedPubMedCentralCrossRef
64.
go back to reference Deng L, Xiong P, Luo Y, Bu X, Qian S, Zhong W, et al. Association between IDH1/2 mutations and brain glioma grade. Oncol Lett. 2018;16(4):5405–9.PubMedPubMedCentral Deng L, Xiong P, Luo Y, Bu X, Qian S, Zhong W, et al. Association between IDH1/2 mutations and brain glioma grade. Oncol Lett. 2018;16(4):5405–9.PubMedPubMedCentral
65.
go back to reference Yan W, Zhang W, You G, Bao Z, Wang Y, Liu Y, et al. Correlation of IDH1 mutation with clinicopathologic factors and prognosis in primary glioblastoma: a report of 118 patients from China. PLoS One. 2012;7(1):e30339.PubMedPubMedCentralCrossRef Yan W, Zhang W, You G, Bao Z, Wang Y, Liu Y, et al. Correlation of IDH1 mutation with clinicopathologic factors and prognosis in primary glioblastoma: a report of 118 patients from China. PLoS One. 2012;7(1):e30339.PubMedPubMedCentralCrossRef
66.
go back to reference Sanson M, Marie Y, Paris S, Idbaih A, Laffaire J, Ducray F, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol. 2009;27(25):4150–4.PubMedCrossRef Sanson M, Marie Y, Paris S, Idbaih A, Laffaire J, Ducray F, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol. 2009;27(25):4150–4.PubMedCrossRef
67.
go back to reference Polivka J, Polivka J Jr, Rohan V, Pesta M, Repik T, Pitule P, et al. Isocitrate dehydrogenase-1 mutations as prognostic biomarker in glioblastoma multiforme patients in West Bohemia. Biomed Res Int. 2014;2014:735659.PubMedPubMedCentralCrossRef Polivka J, Polivka J Jr, Rohan V, Pesta M, Repik T, Pitule P, et al. Isocitrate dehydrogenase-1 mutations as prognostic biomarker in glioblastoma multiforme patients in West Bohemia. Biomed Res Int. 2014;2014:735659.PubMedPubMedCentralCrossRef
68.
69.
go back to reference Minniti G, Scaringi C, Arcella A, Lanzetta G, Di Stefano D, Scarpino S, et al. IDH1 mutation and MGMT methylation status predict survival in patients with anaplastic astrocytoma treated with temozolomide-based chemoradiotherapy. J Neuro-Oncol. 2014;118(2):377–83.CrossRef Minniti G, Scaringi C, Arcella A, Lanzetta G, Di Stefano D, Scarpino S, et al. IDH1 mutation and MGMT methylation status predict survival in patients with anaplastic astrocytoma treated with temozolomide-based chemoradiotherapy. J Neuro-Oncol. 2014;118(2):377–83.CrossRef
70.
go back to reference Muralidharan K, Yekula A, Small JL, Rosh ZS, Kang KM, Wang L, et al. Promoter mutation analysis for blood-based diagnosis and monitoring of gliomas. Clin Cancer Res. 2021;27(1):169–78.PubMedCrossRef Muralidharan K, Yekula A, Small JL, Rosh ZS, Kang KM, Wang L, et al. Promoter mutation analysis for blood-based diagnosis and monitoring of gliomas. Clin Cancer Res. 2021;27(1):169–78.PubMedCrossRef
71.
go back to reference Fontanilles M, Marguet F, Beaussire L, Magne N, Pépin LF, Alexandru C, et al. Cell-free DNA and circulating TERT promoter mutation for disease monitoring in newly-diagnosed glioblastoma. Acta Neuropathol Commun. 2020;8(1):179.PubMedPubMedCentralCrossRef Fontanilles M, Marguet F, Beaussire L, Magne N, Pépin LF, Alexandru C, et al. Cell-free DNA and circulating TERT promoter mutation for disease monitoring in newly-diagnosed glioblastoma. Acta Neuropathol Commun. 2020;8(1):179.PubMedPubMedCentralCrossRef
72.
go back to reference Husain A, Mishra S, Hadi R, Sahu A, Kumari S, Rastogi M, et al. Dynamics of cell-free DNA in predicting response in adult diffuse glioma on chemoradiotherapy. Cancer Gene Ther. 2022;268-269:55–63.CrossRef Husain A, Mishra S, Hadi R, Sahu A, Kumari S, Rastogi M, et al. Dynamics of cell-free DNA in predicting response in adult diffuse glioma on chemoradiotherapy. Cancer Gene Ther. 2022;268-269:55–63.CrossRef
73.
go back to reference Riviere-Cazaux C, Dong X, Mo W, Luo S, Wang A, Du P, et al. Biom-16. Longitudinal glioma cerebrospinal fluid cell-free dna for monitoring and treatment response assessment. Neuro-Oncology. 2023;25(Supplement_5):v7-v.CrossRef Riviere-Cazaux C, Dong X, Mo W, Luo S, Wang A, Du P, et al. Biom-16. Longitudinal glioma cerebrospinal fluid cell-free dna for monitoring and treatment response assessment. Neuro-Oncology. 2023;25(Supplement_5):v7-v.CrossRef
75.
go back to reference Saenz-Antonanzas A, Auzmendi-Iriarte J, Carrasco-Garcia E, Moreno-Cugnon L, Ruiz I, Villanua J, et al. Liquid biopsy in glioblastoma: opportunities, applications and challenges. Cancers (Basel). 2019;11(7). Saenz-Antonanzas A, Auzmendi-Iriarte J, Carrasco-Garcia E, Moreno-Cugnon L, Ruiz I, Villanua J, et al. Liquid biopsy in glioblastoma: opportunities, applications and challenges. Cancers (Basel). 2019;11(7).
76.
go back to reference Pasqualetti F, Rizzo M, Franceschi S, Lessi F, Paiar F, Buffa FM. New perspectives in liquid biopsy for glioma patients. Curr Opin Oncol. 2022;34(6):705–12.PubMedCrossRef Pasqualetti F, Rizzo M, Franceschi S, Lessi F, Paiar F, Buffa FM. New perspectives in liquid biopsy for glioma patients. Curr Opin Oncol. 2022;34(6):705–12.PubMedCrossRef
Metadata
Title
IDH1 mutation is detectable in plasma cell-free DNA and is associated with survival outcome in glioma patients
Authors
Stefania Crucitta
Francesco Pasqualetti
Alessandra Gonnelli
Martina Ruglioni
Giovanna Irene Luculli
Martina Cantarella
Valerio Ortenzi
Cristian Scatena
Fabiola Paiar
Antonio Giuseppe Naccarato
Romano Danesi
Marzia Del Re
Publication date
01-12-2024
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2024
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-11726-0

Other articles of this Issue 1/2024

BMC Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine