Pseudoprogression in glioblastoma patients: the impact of extent of resection
详细信息    查看全文
  • 作者:Hun Ho Park ; Tae Hoon Roh ; Seok Gu Kang ; Eui Hyun Kim…
  • 关键词:Extent of resection ; Glioblastoma ; MGMT promoter status ; Pseudoprogression
  • 刊名:Journal of Neuro-Oncology
  • 出版年:2016
  • 出版时间:February 2016
  • 年:2016
  • 卷:126
  • 期:3
  • 页码:559-566
  • 全文大小:451 KB
  • 参考文献:1.Deorah S, Lynch CF, Sibenaller ZA, Ryken TC (2006) Trends in brain cancer incidence and survival in the United States: surveillance, epidemiology, and end results program, 1973 to 2001. Neurosurg Focus 20(4):E1PubMed CrossRef
    2.Surawicz TS, Davis F, Freels S, Laws ER Jr, Menck HR (1998) Brain tumor survival: results from the National Cancer Data Base. J Neurooncol 40:151–160PubMed CrossRef
    3.Stupp R, Mason WP, van den Bent MJ et al (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352:987–996PubMed CrossRef
    4.Chamberlain MC, Glantz MJ, Chalmers L, Van Horn A, Sloan AE (2007) Early necrosis following concurrent Temodar and radiotherapy in patients with glioblastoma. J Neurooncol 82:81–83PubMed CrossRef
    5.Taal W, Brandsma D, de Bruin HG et al (2007) The incidence of pseudo-progression in a cohort of malignant glioma patients treated with chemo-radiation with temozolomide. Cancer 113:405–410CrossRef
    6.Brandes AA, Franceschi E, Tosoni A et al (2008) MGMT promoter methylation status can predict the incidence and outcome of pseudoprogression after concomitant radiochemotherapy in newly diagnosed glioblastoma patients. J Clin Oncol 26:2192–2197PubMed CrossRef
    7.Peca C, Pacelli R, Elefante A et al (2009) Early clinical and neuroradiological worsening after radiotherapy and concomitant temozolomide in patients with glioblastoma: tumour progression or radionecrosis? Clin Neurol Neurosurg 111:331–334PubMed CrossRef
    8.Yaman E, Buyukberber S, Benekli M et al (2010) Radiation induced early necrosis in patients with malignant gliomas receiving temozolomide. Clin Neurol Neurosurg 112:662–667PubMed CrossRef
    9.Ruben JD, Dally M, Bailey M, Smith R, McLean CA, Fedele P (2006) Cerebral radiation necrosis: incidence, outcomes, and risk factors with emphasis on radiation parameters and chemotherapy. Int J Radiat Oncol Biol Phys 65:499–508PubMed CrossRef
    10.Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent MJ (2008) Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol 9:453–461PubMed CrossRef
    11.Floyd NS, Woo SY, Teh BS et al (2004) Hypofractionated intensity-modulated radiotherapy for primary glioblastoma multiforme. Int J Radiat Oncol Biol Phys 58:721–726PubMed CrossRef
    12.Nieder C, Andratschke N, Wiedenmann N, Busch R, Grosu AL, Molls M (2004) Radiotherapy for high-grade gliomas. Does altered fractionation improve the outcome? Strahlenther Onkol 180:401–407PubMed CrossRef
    13.Hara W, Tran P, Li G et al (2009) Cyberknife for brain metastases of malignant melanoma and renal cell carcinoma. Neurosurgery 64:A26–A32PubMed CrossRef
    14.Esteller M, Hamilton SR, Burger PC, Baylin SB, Herman JG (1999) Inactivation of the DNA repair gene O6-methylguanine-DNA methyltransferase by promoter hypermethylation is a common event in primary human neoplasia. Cancer Res 59:793–797PubMed
    15.Esteller M, Garcia-Foncillas J, Andion E et al (2000) Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med 343:1350–1354PubMed CrossRef
    16.Hegi ME, Diserens AC, Gorlia T et al (2005) MGMT gene silencing and benefit from temozolomide in glioblastoma. NEJM 352:997–1003PubMed CrossRef
    17.Chang EF, Smith JS, Chang SM et al (2008) Preoperative prognostic classification system for hemispheric low-grade gliomas in adults. J Neurosurg 109:817–824PubMed CrossRef
    18.Wen PY, Macdonald DR, Reardon DA et al (2010) Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 28:1963–1972PubMed CrossRef
    19.Ellika SK, Jain R, Patel SC et al (2007) Role of perfusion CT in glioma grading and comparison with conventional MR imaging features. Am J Neuroradiol 28:1981–1987PubMed CrossRef
    20.Kamada K, Houkin K, Abe H, Sawamura Y, Kashiwaba T (1997) Differentiation of cerebral radiation necrosis from tumor recurrence by proton magnetic resonance spectroscopy. Neurol Med-Chir 37:250–256CrossRef
    21.Chao ST, Suh JH, Raja S, Lee SY, Barnett G (2001) The sensitivity and specificity of FDG PET in distinguishing recurrent brain tumor from radionecrosis in patients treated with stereotactic radiosurgery. Int J Cancer 96:191–197PubMed CrossRef
    22.de Wit MC, de Bruin HG, Eijkenboom W, Sillevis Smitt PA, van den Bent MJ (2004) Immediate post-radiotherapy changes in malignant glioma can mimic tumor progression. Neurology 63:535–537PubMed CrossRef
    23.Brandsma D, van den Bent MJAN (2009) Pseudoprogression and pseudoresponse in the treatment of gliomas. Curr Opin Neurol 22:633–638PubMed CrossRef
    24.Shah AH, Snelling B, Bregy A et al (2013) Discriminating radiation necrosis from tumor progression in gliomas: a systematic review what is the best imaging modality? J Neurooncol 112:141–152PubMed CrossRef
    25.Siu A, Wind JJ, Iorgulescu JB, Chan TA, Yamada Y, Sherman JH (2012) Radiation necrosis following treatment of high grade glioma—a review of the literature and current understanding. Acta Neurochir (Wien) 154:191–201CrossRef
    26.Liu L, Markowitz S, Gerson SL (1996) Mismatch repair mutations override alkyltransferase in conferring resistance to temozolomide, but not to 1,3-bis (2-chloroethyl) nitrosourea. Cancer Res 56:5375–5379PubMed
    27.Ochs K, Kaina B (2000) Apoptosis induced by DNA damage O6-methylguanine is Bcl-2 and caspase-9/3 regulated and Fas/caspase-8 independent. Cancer Res 60:5815–5824PubMed
    28.Olson RA, Brastianos PK, Palma DA (2011) Prognostic and predictive value of epigenetic silencing of MGMT in patients with high grade gliomas: a systematic review and meta-analysis. J Neurooncol 105:325–335PubMed CrossRef
  • 作者单位:Hun Ho Park (1) (6)
    Tae Hoon Roh (1) (6)
    Seok Gu Kang (1) (6) (7)
    Eui Hyun Kim (1) (6) (7)
    Chang-Ki Hong (1)
    Se Hoon Kim (2) (6) (7)
    Sung Soo Ahn (3) (6)
    Seung Koo Lee (3) (6)
    Hye Jin Choi (4) (6)
    Jaeho Cho (5) (6)
    Sun Ho Kim (1) (6) (7)
    Kyu-Sung Lee (1) (6) (7)
    Chang-Ok Suh (5) (6) (8)
    Jong Hee Chang (1) (6) (7) (9)

    1. Departments of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
    6. Brain Tumor Center, Yonsei University College of Medicine, Seoul, Korea
    7. Brain Research Institute, Yonsei University College of Medicine, Seoul, Korea
    2. Departments of Pathology, Yonsei University College of Medicine, Seoul, Korea
    3. Departments of Radiology, Yonsei University College of Medicine, Seoul, Korea
    4. Departments of Medical Oncology, Yonsei University College of Medicine, Seoul, Korea
    5. Departments of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
    8. Department of Radiation Oncology, Yonsei University Health System, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korea
    9. Department of Neurosurgery, Yonsei University Health System, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, Korean
  • 刊物类别:Medicine
  • 刊物主题:Medicine & Public Health
    Oncology
  • 出版者:Springer Netherlands
  • ISSN:1573-7373
文摘
Pseudoprogression (psPD) is a radiation-induced toxicity that has substantial neurological consequence in glioblastoma (GBM) patients. MGMT promoter methylation has been shown to be an important prognostic factor of psPD, but the significance of extent of resection (EOR) remains unclear. We performed a retrospective analysis on newly diagnosed GBM patients with assessable MGMT promoter status who underwent the Stupp protocol. EOR was grouped into gross total resection (GTR), subtotal resection (STR), partial resection (PR) and stereotactic biopsy. Contrast enhancing lesion enlargement was classified as psPD or non-psPD. Among a total of 101 patients, GTR, STR, PR and stereotactic biopsy was performed in 57 (56.4 %), 34 (33.7 %), 9 (8.9 %) and 1 patient (1 %), respectively. Follow-up imaging at the end of Stupp protocol classified 45 patients (44.6 %) as psPD and 56 (55.4 %) as non-psPD. psPD was observed in 24 (61.5 %) of 39 patients with methylated MGMT promoter and 21 (33.9 %) of 62 patients with unmethylated MGMT promoter (p < 0.01). psPD was documented in 17 (29.8 %), 19 (55.9 %), 8 (88.9 %) and 1 (100 %) patient with GTR, STR, PR and stereotactic biopsy (p < 0.01), respectively. On multivariate analysis MGMT promoter status (OR 3.36, 95 % CI 1.36–8.34) and EOR (OR 4.12, 95 % CI 1.71–9.91) were independent predictors of psPD. A Cox proportional hazards model showed that MGMT status (HR 2.51, p < 0.01) and EOR (HR 2.99, p < 0.01) significantly influenced survival. MGMT status and EOR have a significant impact on psPD. GTR can reduce the side effects of psPD and prolong survival. Keywords Extent of resection Glioblastoma MGMT promoter status Pseudoprogression

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700