Skip to main content
Erschienen in: Current Treatment Options in Oncology 3/2011

01.09.2011 | CNS Malignancies

Pseudoprogression: Relevance With Respect to Treatment of High-Grade Gliomas

verfasst von: James Fink, MD, Donald Born, MD, Marc C. Chamberlain, MD

Erschienen in: Current Treatment Options in Oncology | Ausgabe 3/2011

Einloggen, um Zugang zu erhalten

Opinion statement

The post-treatment imaging assessment of high-grade gliomas remains challenging notwithstanding the increased utilization of advanced MRI and PET imaging. Several post-treatment imaging entities are recognized including: late-delayed radiation injury, including radionecrosis mimicking tumor progression; early-delayed (within 6 months of temozolomide-based chemoradiation) post-treatment radiographic changes, herein referred to as pseudoprogression (the subject of this review); early post-treatment changes following local glioma therapy (i.e. biodegradable BCNU wafer implantation or stereotactic radiotherapy); and pseudoresponse, seen following treatment with angiogenic inhibition based therapy such as bevacizumab. A literature review searched specifically for “pseudoprogression” within the last 5 years (2005–2010). Approximately 24 recent papers were identified and reviewed in detail. Eight small population-based studies demonstrate 26–58% (median 49%) of glioblastoma patients treated with chemoradiotherapy manifest early disease progression at first post-radiotherapy imaging. Patients with early radiographic disease progression continued on planned therapy, and a median of 38% (range 28–66%) showed radiographic improvement or stabilization and were defined retrospectively as manifesting pseudoprogression. In conclusion, pseudoprogression is a frequent early post-treatment imaging change that at present is not easily differentiated from tumor progression by anatomic or physiologic brain imaging. Consequently, an operational definition of pseudoprogression has been adopted by the Response Assessment in Neuro-Oncology Working Group wherein either the index (i.e. target) lesion stabilizes or diminishes in size on continued post-radiation (temozolomide) therapy as determined by follow-up radiologic imaging.
Literatur
1.
Zurück zum Zitat Hoffman WF, Levin VA, Wilson CB. Evaluation of malignant glioma patients during the postirradiation period. J Neurosurg. 1979;50(5):624–8.PubMedCrossRef Hoffman WF, Levin VA, Wilson CB. Evaluation of malignant glioma patients during the postirradiation period. J Neurosurg. 1979;50(5):624–8.PubMedCrossRef
2.
Zurück zum Zitat Levin VA et al. Criteria for evaluating patients undergoing chemotherapy for malignant brain tumors. J Neurosurg. 1977;47(3):329–35.PubMedCrossRef Levin VA et al. Criteria for evaluating patients undergoing chemotherapy for malignant brain tumors. J Neurosurg. 1977;47(3):329–35.PubMedCrossRef
3.
Zurück zum Zitat Cascino TLK, Kimmel DW, Dinapoli RP, et al. Report of four cases with a resolving syndrome which otherwise simulates recurrent brain tumor. Neurology. 1988;38(Supplement 1):306. Cascino TLK, Kimmel DW, Dinapoli RP, et al. Report of four cases with a resolving syndrome which otherwise simulates recurrent brain tumor. Neurology. 1988;38(Supplement 1):306.
4.
Zurück zum Zitat Macdonald DR et al. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8(7):1277–80.PubMed Macdonald DR et al. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8(7):1277–80.PubMed
5.
Zurück zum Zitat Graeb DA, Steinbok P, Robertson WD. Transient early computed tomographic changes mimicking tumor progression after brain tumor irradiation. Radiology. 1982;144(4):813–7.PubMed Graeb DA, Steinbok P, Robertson WD. Transient early computed tomographic changes mimicking tumor progression after brain tumor irradiation. Radiology. 1982;144(4):813–7.PubMed
6.
Zurück zum Zitat de Wit MC et al. Immediate post-radiotherapy changes in malignant glioma can mimic tumor progression. Neurology. 2004;63(3):535–7.PubMed de Wit MC et al. Immediate post-radiotherapy changes in malignant glioma can mimic tumor progression. Neurology. 2004;63(3):535–7.PubMed
7.
Zurück zum Zitat Stupp R et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.PubMedCrossRef Stupp R et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–96.PubMedCrossRef
8.
Zurück zum Zitat Chamberlain MC et al. Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol. 2007;82(1):81–3.PubMedCrossRef Chamberlain MC et al. Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol. 2007;82(1):81–3.PubMedCrossRef
9.
Zurück zum Zitat Taal W et al. Incidence of early pseudo-progression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomide. Cancer. 2008;113(2):405–10.PubMedCrossRef Taal W et al. Incidence of early pseudo-progression in a cohort of malignant glioma patients treated with chemoirradiation with temozolomide. Cancer. 2008;113(2):405–10.PubMedCrossRef
10.
Zurück zum Zitat Brandes AA et al. MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol. 2008;26(13):2192–7.PubMedCrossRef Brandes AA et al. MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol. 2008;26(13):2192–7.PubMedCrossRef
11.
Zurück zum Zitat Sanghera P et al. Pseudoprogression following chemoradiotherapy for glioblastoma multiforme. Can J Neurol Sci. 2010;37(1):36–42.PubMed Sanghera P et al. Pseudoprogression following chemoradiotherapy for glioblastoma multiforme. Can J Neurol Sci. 2010;37(1):36–42.PubMed
12.
Zurück zum Zitat Roldan GB et al. Population-based study of pseudoprogression after chemoradiotherapy in GBM. Can J Neurol Sci. 2009;36(5):617–22.PubMed Roldan GB et al. Population-based study of pseudoprogression after chemoradiotherapy in GBM. Can J Neurol Sci. 2009;36(5):617–22.PubMed
13.••
Zurück zum Zitat Wen PY, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28(11):1963–72.PubMedCrossRef Wen PY, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28(11):1963–72.PubMedCrossRef
14.
Zurück zum Zitat Zeng QS et al. Multivoxel 3D proton MR spectroscopy in the distinction of recurrent glioma from radiation injury. J Neurooncol. 2007;84(1):63–9.PubMedCrossRef Zeng QS et al. Multivoxel 3D proton MR spectroscopy in the distinction of recurrent glioma from radiation injury. J Neurooncol. 2007;84(1):63–9.PubMedCrossRef
15.•
Zurück zum Zitat Hu LS, et al. Relative cerebral blood volume values to differentiate high-grade glioma recurrence from posttreatment radiation effect: direct correlation between image-guided tissue histopathology and localized dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging measurements. AJNR Am J Neuroradiol. 2009;30(3):552–8.PubMedCrossRef Hu LS, et al. Relative cerebral blood volume values to differentiate high-grade glioma recurrence from posttreatment radiation effect: direct correlation between image-guided tissue histopathology and localized dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging measurements. AJNR Am J Neuroradiol. 2009;30(3):552–8.PubMedCrossRef
16.
Zurück zum Zitat Hein PA et al. Diffusion-weighted imaging in the follow-up of treated high-grade gliomas: tumor recurrence versus radiation injury. AJNR Am J Neuroradiol. 2004;25(2):201–9.PubMed Hein PA et al. Diffusion-weighted imaging in the follow-up of treated high-grade gliomas: tumor recurrence versus radiation injury. AJNR Am J Neuroradiol. 2004;25(2):201–9.PubMed
17.
Zurück zum Zitat Kashimura H et al. Diffusion tensor imaging for differentiation of recurrent brain tumor and radiation necrosis after radiotherapy–three case reports. Clin Neurol Neurosurg. 2007;109(1):106–10.PubMedCrossRef Kashimura H et al. Diffusion tensor imaging for differentiation of recurrent brain tumor and radiation necrosis after radiotherapy–three case reports. Clin Neurol Neurosurg. 2007;109(1):106–10.PubMedCrossRef
18.
Zurück zum Zitat Schlemmer HP et al. Proton MR spectroscopic evaluation of suspicious brain lesions after stereotactic radiotherapy. AJNR Am J Neuroradiol. 2001;22(7):1316–24.PubMed Schlemmer HP et al. Proton MR spectroscopic evaluation of suspicious brain lesions after stereotactic radiotherapy. AJNR Am J Neuroradiol. 2001;22(7):1316–24.PubMed
19.
Zurück zum Zitat Plotkin M et al. 123I-IMT SPECT and 1 H MR-spectroscopy at 3.0 T in the differential diagnosis of recurrent or residual gliomas: a comparative study. J Neurooncol. 2004;70(1):49–58.PubMedCrossRef Plotkin M et al. 123I-IMT SPECT and 1 H MR-spectroscopy at 3.0 T in the differential diagnosis of recurrent or residual gliomas: a comparative study. J Neurooncol. 2004;70(1):49–58.PubMedCrossRef
20.
Zurück zum Zitat Sugahara T et al. Posttherapeutic intraaxial brain tumor: the value of perfusion-sensitive contrast-enhanced MR imaging for differentiating tumor recurrence from nonneoplastic contrast-enhancing tissue. AJNR Am J Neuroradiol. 2000;21(5):901–9.PubMed Sugahara T et al. Posttherapeutic intraaxial brain tumor: the value of perfusion-sensitive contrast-enhanced MR imaging for differentiating tumor recurrence from nonneoplastic contrast-enhancing tissue. AJNR Am J Neuroradiol. 2000;21(5):901–9.PubMed
21.
Zurück zum Zitat Galban CJ et al. The parametric response map is an imaging biomarker for early cancer treatment outcome. Nat Med. 2009;15(5):572–6.PubMedCrossRef Galban CJ et al. The parametric response map is an imaging biomarker for early cancer treatment outcome. Nat Med. 2009;15(5):572–6.PubMedCrossRef
22.•
Zurück zum Zitat Tsien C., et al. Parametric response map as an imaging biomarker to distinguish progression from pseudoprogression in high-grade glioma. J Clin Oncol. 2010;28(13):2293–9.PubMedCrossRef Tsien C., et al. Parametric response map as an imaging biomarker to distinguish progression from pseudoprogression in high-grade glioma. J Clin Oncol. 2010;28(13):2293–9.PubMedCrossRef
23.
Zurück zum Zitat Ricci PE et al. Differentiating recurrent tumor from radiation necrosis: time for re-evaluation of positron emission tomography? AJNR Am J Neuroradiol. 1998;19(3):407–13.PubMed Ricci PE et al. Differentiating recurrent tumor from radiation necrosis: time for re-evaluation of positron emission tomography? AJNR Am J Neuroradiol. 1998;19(3):407–13.PubMed
24.
Zurück zum Zitat Spence AM et al. 18 F-FDG PET of gliomas at delayed intervals: improved distinction between tumor and normal gray matter. J Nucl Med. 2004;45(10):1653–9.PubMed Spence AM et al. 18 F-FDG PET of gliomas at delayed intervals: improved distinction between tumor and normal gray matter. J Nucl Med. 2004;45(10):1653–9.PubMed
25.
Zurück zum Zitat Chen W et al. Predicting treatment response of malignant gliomas to bevacizumab and irinotecan by imaging proliferation with [18 F] fluorothymidine positron emission tomography: a pilot study. J Clin Oncol. 2007;25(30):4714–21.PubMedCrossRef Chen W et al. Predicting treatment response of malignant gliomas to bevacizumab and irinotecan by imaging proliferation with [18 F] fluorothymidine positron emission tomography: a pilot study. J Clin Oncol. 2007;25(30):4714–21.PubMedCrossRef
26.
Zurück zum Zitat Muzi M et al. Kinetic analysis of 3′-deoxy-3′-18 F-fluorothymidine in patients with gliomas. J Nucl Med. 2006;47(10):1612–21.PubMed Muzi M et al. Kinetic analysis of 3′-deoxy-3′-18 F-fluorothymidine in patients with gliomas. J Nucl Med. 2006;47(10):1612–21.PubMed
27.
Zurück zum Zitat Spence AM et al. NCI-sponsored trial for the evaluation of safety and preliminary efficacy of 3′-deoxy-3′-[18 F]fluorothymidine (FLT) as a marker of proliferation in patients with recurrent gliomas: preliminary efficacy studies. Mol Imaging Biol. 2009;11(5):343–55.PubMedCrossRef Spence AM et al. NCI-sponsored trial for the evaluation of safety and preliminary efficacy of 3′-deoxy-3′-[18 F]fluorothymidine (FLT) as a marker of proliferation in patients with recurrent gliomas: preliminary efficacy studies. Mol Imaging Biol. 2009;11(5):343–55.PubMedCrossRef
28.
Zurück zum Zitat Terakawa Y et al. Diagnostic accuracy of 11 C-methionine PET for differentiation of recurrent brain tumors from radiation necrosis after radiotherapy. J Nucl Med. 2008;49(5):694–9.PubMedCrossRef Terakawa Y et al. Diagnostic accuracy of 11 C-methionine PET for differentiation of recurrent brain tumors from radiation necrosis after radiotherapy. J Nucl Med. 2008;49(5):694–9.PubMedCrossRef
29.
Zurück zum Zitat Rachinger W et al. Positron emission tomography with O-(2-[18F]fluoroethyl)-l-tyrosine versus magnetic resonance imaging in the diagnosis of recurrent gliomas. Neurosurgery. 2005;57(3):505–11. discussion 505–11.PubMedCrossRef Rachinger W et al. Positron emission tomography with O-(2-[18F]fluoroethyl)-l-tyrosine versus magnetic resonance imaging in the diagnosis of recurrent gliomas. Neurosurgery. 2005;57(3):505–11. discussion 505–11.PubMedCrossRef
30.
Zurück zum Zitat Pytel P, Lukas RV. Update on diagnostic practice: tumors of the nervous system. Arch Pathol Lab Med. 2009;133(7):1062–77.PubMed Pytel P, Lukas RV. Update on diagnostic practice: tumors of the nervous system. Arch Pathol Lab Med. 2009;133(7):1062–77.PubMed
31.
Zurück zum Zitat Fabi A et al. Pseudoprogression and MGMT status in glioblastoma patients: implications in clinical practice. Anticancer Res. 2009;29(7):2607–10.PubMed Fabi A et al. Pseudoprogression and MGMT status in glioblastoma patients: implications in clinical practice. Anticancer Res. 2009;29(7):2607–10.PubMed
32.
Zurück zum Zitat Sayyari A. A., et al., Distinguishing recurrent primary brain tumor from radiation injury: a preliminary study using a susceptibility-weighted MR imaging-guided apparent diffusion coefficient analysis strategy. AJNR Am J Neuroradiol. 2010;31(6):1049–54.PubMedCrossRef Sayyari A. A., et al., Distinguishing recurrent primary brain tumor from radiation injury: a preliminary study using a susceptibility-weighted MR imaging-guided apparent diffusion coefficient analysis strategy. AJNR Am J Neuroradiol. 2010;31(6):1049–54.PubMedCrossRef
33.
Zurück zum Zitat Miyatake S et al. Pseudoprogression in boron neutron capture therapy for malignant gliomas and meningiomas. Neuro Oncology. 2009;11(4):430–6.PubMedCrossRef Miyatake S et al. Pseudoprogression in boron neutron capture therapy for malignant gliomas and meningiomas. Neuro Oncology. 2009;11(4):430–6.PubMedCrossRef
34.
Zurück zum Zitat Perry A, Schmidt RE. Cancer therapy-associated CNS neuropathology: an update and review of the literature. Acta Neuropathol. 2006;111(3):197–212.PubMedCrossRef Perry A, Schmidt RE. Cancer therapy-associated CNS neuropathology: an update and review of the literature. Acta Neuropathol. 2006;111(3):197–212.PubMedCrossRef
35.
Zurück zum Zitat Tihan T et al. Prognostic value of detecting recurrent glioblastoma multiforme in surgical specimens from patients after radiotherapy: should pathology evaluation alter treatment decisions? Hum Pathol. 2006;37(3):272–82.PubMedCrossRef Tihan T et al. Prognostic value of detecting recurrent glioblastoma multiforme in surgical specimens from patients after radiotherapy: should pathology evaluation alter treatment decisions? Hum Pathol. 2006;37(3):272–82.PubMedCrossRef
36.
Zurück zum Zitat Cao Y et al. Use of magnetic resonance imaging to assess blood-brain/blood-glioma barrier opening during conformal radiotherapy. J Clin Oncol. 2005;23(18):4127–36.PubMedCrossRef Cao Y et al. Use of magnetic resonance imaging to assess blood-brain/blood-glioma barrier opening during conformal radiotherapy. J Clin Oncol. 2005;23(18):4127–36.PubMedCrossRef
37.
Zurück zum Zitat Lemasson B et al. Monitoring blood-brain barrier status in a rat model of glioma receiving therapy: dual injection of low-molecular-weight and macromolecular MR contrast media. Radiology. 2010;257(2):342–52.PubMedCrossRef Lemasson B et al. Monitoring blood-brain barrier status in a rat model of glioma receiving therapy: dual injection of low-molecular-weight and macromolecular MR contrast media. Radiology. 2010;257(2):342–52.PubMedCrossRef
38.•
Zurück zum Zitat Chamberlain MC. Emerging clinical principles on the use of bevacizumab for the treatment of malignant gliomas. Cancer 2010;116(17):3988–99.PubMedCrossRef Chamberlain MC. Emerging clinical principles on the use of bevacizumab for the treatment of malignant gliomas. Cancer 2010;116(17):3988–99.PubMedCrossRef
39.
40.
Zurück zum Zitat Henson JW, Ulmer S, Harris GJ. Brain tumor imaging in clinical trials. AJNR Am J Neuroradiol. 2008;29(3):419–24.PubMedCrossRef Henson JW, Ulmer S, Harris GJ. Brain tumor imaging in clinical trials. AJNR Am J Neuroradiol. 2008;29(3):419–24.PubMedCrossRef
41.
Zurück zum Zitat Gerstner ER, Batchelor TT. Imaging and response criteria in gliomas. Curr Opin Oncol. 2010;22(6):598–603.PubMedCrossRef Gerstner ER, Batchelor TT. Imaging and response criteria in gliomas. Curr Opin Oncol. 2010;22(6):598–603.PubMedCrossRef
42.••
Zurück zum Zitat Brandsma, D., van den Bent MJ. Pseudoprogression and pseudoresponse in the treatment of gliomas. Curr Opin Neurol. 2009;22(6):633–8.PubMedCrossRef Brandsma, D., van den Bent MJ. Pseudoprogression and pseudoresponse in the treatment of gliomas. Curr Opin Neurol. 2009;22(6):633–8.PubMedCrossRef
Metadaten
Titel
Pseudoprogression: Relevance With Respect to Treatment of High-Grade Gliomas
verfasst von
James Fink, MD
Donald Born, MD
Marc C. Chamberlain, MD
Publikationsdatum
01.09.2011
Verlag
Current Science Inc.
Erschienen in
Current Treatment Options in Oncology / Ausgabe 3/2011
Print ISSN: 1527-2729
Elektronische ISSN: 1534-6277
DOI
https://doi.org/10.1007/s11864-011-0157-1

Weitere Artikel der Ausgabe 3/2011

Current Treatment Options in Oncology 3/2011 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.