Brain Tumor Res Treat.  2017 Apr;5(1):1-9. 10.14791/btrt.2017.5.1.1.

Bevacizumab in Recurrent Glioma: Patterns of Treatment Failure and Implications

Affiliations
  • 1Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA. yi.li@ucsf.edu
  • 2Department of Radiology, University of Chicago, Chicago, IL, USA.
  • 3Department of Neurology, University of California, San Francisco, CA, USA.

Abstract

Glioblastoma, the most common primary malignant brain tumor in adults, is highly aggressive and associated with a poor prognosis. Bevacizumab, a monoclonal antibody against the vascular endothelial growth factor receptor, has increasingly been used in the treatment of recurrent glioblastoma. It has achieved excellent rates of radiographic response, but most patients will progress after only a few months. Upon recurrence, tumors may not enhance, secondary to vascular normalization. We describe four patterns of radiographic progression commonly associated with Bevacizumab failure: 1) Distant enhancing tumor, 2) Local tumor progression without enhancement, 3) Diffuse gliomatosis-like infiltration, and 4) Local or multifocal progression, with enhancement. Some have noted an increased incidence of distant or diffuse disease upon recurrence, suggestive of a transition to a more aggressive phenotype, but a review of the literature suggests there is no conclusive evidence that Bevacizumab treatment is associated with an increased rate of distant or diffuse recurrence.

Keyword

Brain neoplasms; Bevacizumab; Vascular endothelial growth factor; Glioma; Glioblastoma; Neuroimaging

MeSH Terms

Adult
Bevacizumab*
Brain Neoplasms
Glioblastoma
Glioma*
Humans
Incidence
Neuroimaging
Phenotype
Prognosis
Receptors, Vascular Endothelial Growth Factor
Recurrence
Treatment Failure*
Vascular Endothelial Growth Factor A
Bevacizumab
Receptors, Vascular Endothelial Growth Factor
Vascular Endothelial Growth Factor A

Figure

  • Fig. 1 Pre-treatment and post-treatment axial FLAIR (A and B, respectively) demonstrate decreased vasogenic edema post-treatment (arrows). Pre- and post-treatment axial T1 post-contrast (C and D, respectively) demonstrate decreased contrast enhancement (arrowheads). These findings are consistent with treatment response. FLAIR, fluid-attenuated inversion recovery.

  • Fig. 2 Pre-treatment and post-treatment axial (A and B, respectively) FLAIR images demonstrate decreased vasogenic edema post-treatment (arrows). Pre- and post-treatment axial T1 post-contrast images through the site of the index lesion (C and D, respectively) demonstrate decreased enhancement of disease (arrowheads). Pre-and post-treatment axial T1-post contrast images through a location distant from the index lesion (E and F, respectively) demonstrate a new distant enhancing tumor post-treatment (low arrow). Pattern of treatment failure is consistent with distant enhancing tumor. FLAIR, fluid-attenuated inversion recovery.

  • Fig. 3 Pre-treatment and post-treatment axial FLAIR images (A and B, respectively) demonstrate decreased vasogenic edema post-treatment (arrows). Pre- and post-treatment axial T1 post-contrast through the level of the index lesion (C and D, respectively) demonstrate decreased enhancement at the site of disease (arrowheads). Pre-and post-treatment axial T1-post contrast images through axial levels cranial to the index lesion (E and F, and G and H, respectively) demonstrate new distant enhancing nodules in a pattern consistent with subependymal spread of disease (long arrows). Pattern of treatment failure is consistent with distant enhancing tumor. FLAIR, fluid-attenuated inversion recovery.

  • Fig. 4 Pre-treatment and post-treatment axial FLAIR (A and B, respectively) demonstrates increased mass-like FLAIR hyperintensity post-treatment (arrows). Pre- and post-treatment axial T1 post-contrast through the level of the index lesion (C and D, respectively) demonstrate decreased enhancement of the original disease (arrowheads). Pre- and post-treatment axial DWI (E and F, respectively), and pre- and post-treatment ADC maps (G and H, respectively) demonstrate increased mass-like area of reduced diffusion post-treatment (long arrows). Pattern of treatment failure is consistent with local tumor progression without enhancement. FLAIR, fluid-attenuated inversion recovery; DWI, diffusion weighted image; ADC, apparent diffusion coefficient.

  • Fig. 5 Pre- and post-treatment axial T1 post-contrast images (A and B, respectively) demonstrate decreased tumor enhancement post-treatment (arrowheads). Multiple pre-treatment (C, E, G, and I) and post-treatment (D, F, H, and J) axial FLAIR images demonstrate development of a diffuse infiltrative FLAIR hyperintensity in a gliomatosis-like pattern (arrows). Pattern of treatment failure is consistent with diffuse gliomatosis-like tumor infiltration. FLAIR, fluid-attenuated inversion recovery.

  • Fig. 6 Pre- and post-treatment axial FLAIR (A and B, respectively) demonstrates increased mass-like FLAIR hyperintensity post-treatment (arrows). Pre- and post-treatment axial DWI (C and D, respectively), and pre- and post-treatment ADC map (E and F, respectively) demonstrate an increased mass-like area of reduced diffusion (long arrows). Pre- and post-treatment axial T1 post-contrast images through the level of the index lesion (G and H, respectively) demonstrate increased enhancement. Pattern of treatment failure is consistent with local or multifocal progression with enhancement (arrowheads). FLAIR, fluid-attenuated inversion recovery; DWI, diffusion weighted image; ADC, apparent diffusion coefficient.


Reference

1. Schwartzbaum JA, Fisher JL, Aldape KD, Wrensch M. Epidemiology and molecular pathology of glioma. Nat Clin Pract Neurol. 2006; 2:494–503.
Article
2. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005; 352:987–996.
Article
3. Gilbert MR, Wang M, Aldape KD, et al. Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J Clin Oncol. 2013; 31:4085–4091.
Article
4. Ostrom QT, Gittleman H, Liao P, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007-2011. Neuro Oncol. 2014; 16:Suppl 4. iv1–iv63.
Article
5. Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009; 27:4733–4740.
Article
6. Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009; 27:740–745.
Article
7. Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008; 70:779–787.
Article
8. Pope WB, Xia Q, Paton VE, et al. Patterns of progression in patients with recurrent glioblastoma treated with bevacizumab. Neurology. 2011; 76:432–437.
Article
9. Kargiotis O, Rao JS, Kyritsis AP. Mechanisms of angiogenesis in gliomas. J Neurooncol. 2006; 78:281–293.
Article
10. Rubenstein JL, Kim J, Ozawa T, et al. Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia. 2000; 2:306–314.
Article
11. de Groot JF, Fuller G, Kumar AJ, et al. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol. 2010; 12:233–242.
Article
12. Lu KV, Chang JP, Parachoniak CA, et al. VEGF inhibits tumor cell invasion and mesenchymal transition through a MET/VEGFR2 complex. Cancer Cell. 2012; 22:21–35.
Article
13. Ellis LM, Hicklin DJ. VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 2008; 8:579–591.
Article
14. Thompson EM, Frenkel EP, Neuwelt EA. The paradoxical effect of bevacizumab in the therapy of malignant gliomas. Neurology. 2011; 76:87–93.
Article
15. Pope WB, Lai A, Nghiemphu P, Mischel P, Cloughesy TF. MRI in patients with high-grade gliomas treated with bevacizumab and chemotherapy. Neurology. 2006; 66:1258–1260.
Article
16. Ali SA, McHayleh WM, Ahmad A, et al. Bevacizumab and irinotecan therapy in glioblastoma multiforme: a series of 13 cases. J Neurosurg. 2008; 109:268–272.
Article
17. Bokstein F, Shpigel S, Blumenthal DT. Treatment with bevacizumab and irinotecan for recurrent high-grade glial tumors. Cancer. 2008; 112:2267–2273.
Article
18. Kang TY, Jin T, Elinzano H, Peereboom D. Irinotecan and bevacizumab in progressive primary brain tumors, an evaluation of efficacy and safety. J Neurooncol. 2008; 89:113–118.
Article
19. Narayana A, Kelly P, Golfinos J, et al. Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: impact on local control and patient survival. J Neurosurg. 2009; 110:173–180.
Article
20. Thompson EM, Dosa E, Kraemer DF, Neuwelt EA. Treatment with bevacizumab plus carboplatin for recurrent malignant glioma. Neurosurgery. 2010; 67:87–93.
Article
21. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007; 13:1253–1259.
Article
22. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007; 25:4722–4729.
Article
23. Taal W, Oosterkamp HM, Walenkamp AM, et al. Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014; 15:943–953.
Article
24. Brandes AA, Finocchiaro G, Zagonel V, et al. AT-11. Final results from the randomized phase II trial avareg (ML25739) with bevacizumab (BEV) or fotemustine (FTM) in recurrent GBM. Neuro Oncol. 2014; 16:suppl 5. v10.
25. Wick W, Brandes AA, Gorlia T, et al. EORTC 26101 phase III trial exploring the combination of bevacizumab and lomustine in patients with first progression of a glioblastoma. J Clin Oncol. 2016; 34:suppl. abstract 2001.
Article
26. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008; 8:592–603.
Article
27. Kunkel P, Ulbricht U, Bohlen P, et al. Inhibition of glioma angiogenesis and growth in vivo by systemic treatment with a monoclonal antibody against vascular endothelial growth factor receptor-2. Cancer Res. 2001; 61:6624–6628.
28. Macdonald DR, Cascino TL, Schold SC Jr, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990; 8:1277–1280.
Article
29. Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010; 28:1963–1972.
Article
30. Claes A, Gambarota G, Hamans B, et al. Magnetic resonance imaging-based detection of glial brain tumors in mice after antiangiogenic treatment. Int J Cancer. 2008; 122:1981–1986.
Article
31. Shapiro LQ, Beal K, Goenka A, et al. Patterns of failure after concurrent bevacizumab and hypofractionated stereotactic radiation therapy for recurrent high-grade glioma. Int J Radiat Oncol Biol Phys. 2013; 85:636–642.
Article
32. Prados M, Cloughesy T, Samant M, et al. Response as a predictor of survival in patients with recurrent glioblastoma treated with bevacizumab. Neuro Oncol. 2011; 13:143–151.
Article
33. Nowosielski M, Wiestler B, Goebel G, et al. Progression types after antiangiogenic therapy are related to outcome in recurrent glioblastoma. Neurology. 2014; 82:1684–1692.
Article
34. Zuniga RM, Torcuator R, Jain R, et al. Efficacy, safety and patterns of response and recurrence in patients with recurrent high-grade gliomas treated with bevacizumab plus irinotecan. J Neurooncol. 2009; 91:329–336.
Article
Full Text Links
  • BTRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr