1
|
Fasano M, Pirozzi M, De Falco V, Miceli CC, Farese S, Zotta A, Famiglietti V, Vitale P, Di Giovanni I, Brancati C, Carfora V, Solari D, Somma T, Cavallo LM, Cappabianca P, Conson M, Pacelli R, Ciardiello F, Addeo R. Temozolomide based treatment in glioblastoma: 6 vs. 12 months. Oncol Lett 2024; 28:418. [PMID: 39006948 PMCID: PMC11240269 DOI: 10.3892/ol.2024.14551] [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: 09/12/2023] [Accepted: 11/28/2023] [Indexed: 07/16/2024] Open
Abstract
The Stupp regimen remains the standard treatment for newly diagnosed glioblastomas, although the prognosis remains poor. Several temozolomide alternative schedules have been studied, with extended adjuvant treatment (>6 cycles of temozolomide) frequently used, although different trials have indicated contrasting results. Survival data of 87 patients who received 6 ('6C' group) or 12 ('12C' group) cycles of temozolomide were collected between 2012 and 2022. A total of 45 patients were included in the 6C group and 42 patients were included in the 12C group. Data on isocitrate dehydrogenase mutation and methylguanine-DNA-methyltransferase (MGMT) promoter methylation status were also collected. The 12C group exhibited statistically significantly improved overall survival [OS; 22.8 vs. 17.5 months; hazard ratio (HR), 0.47; 95% CI, 0.30-0.73; P=0.001] and progression-free survival (15.3 vs. 9 months; HR, 0.39; 95% CI, 0.25-0.62; P=0.001). However, in the subgroup analysis according to MGMT status, OS in the 12C group was significantly superior to OS in the 6C group only in the MGMT unmethylated tumors. The present data suggested that extended adjuvant temozolomide appeared to be more effective than the conventional six cycles.
Collapse
Affiliation(s)
- Morena Fasano
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Mario Pirozzi
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Vincenzo De Falco
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Chiara Carmen Miceli
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Stefano Farese
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Alessia Zotta
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Vincenzo Famiglietti
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Pasquale Vitale
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Ilaria Di Giovanni
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Christian Brancati
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| | - Vincenzo Carfora
- Radiation Oncology Unit, Department of Radiation Oncology, 'San Pio' Hospital, I-82100 Benevento, Italy
| | - Domenico Solari
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Luigi Maria Cavallo
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Paolo Cappabianca
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Manuel Conson
- Department of Advanced Biomedical Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, University of Naples Federico II, I-80131 Naples, Italy
| | - Fortunato Ciardiello
- Medical Oncology Unit, Department of Precision Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Raffaele Addeo
- Oncology Unit, 'San Giovanni di Dio' Hospital, ASL Napoli 2 Nord, I-80020 Frattamaggiore, Italy
| |
Collapse
|
2
|
Anvari K, Seilanian Toussi M, Saghafi M, Javadinia SA, Saghafi H, Welsh JS. Extended dosing (12 cycles) vs conventional dosing (6 cycles) of adjuvant temozolomide in adults with newly diagnosed high-grade gliomas: a randomized, single-blind, two-arm, parallel-group controlled trial. Front Oncol 2024; 14:1357789. [PMID: 38774410 PMCID: PMC11106464 DOI: 10.3389/fonc.2024.1357789] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/17/2024] [Indexed: 05/24/2024] Open
Abstract
Purpose Maximum safe surgical resection followed by adjuvant chemoradiation and temozolomide chemotherapy is the current standard of care in the management of newly diagnosed high grade glioma. However, there are controversies about the optimal number of adjuvant temozolomide cycles. This study aimed to compare the survival benefits of 12 cycles against 6 cycles of adjuvant temozolomide adults with newly diagnosed high grade gliomas. Methods Adult patients with newly diagnosed high grade gliomas, and a Karnofsky performance status>60%, were randomized to receive either 6 cycles or 12 cycles of adjuvant temozolomide. Patients were followed-up for assessment of overall survival (OS) and progression-free survival (PFS) by brain MRI every 3 months within the first year after treatment and then every six months. Results A total of 100 patients (6 cycles, 50; 12 cycles, 50) were entered. The rate of treatment completion in 6 cycles and 12 cycles groups were 91.3% and 55.1%, respectively. With a median follow-up of 26 months, the 12-, 24-, 36-, and 48-month OS rates in 6 cycles and 12 cycles groups were 81.3% vs 78.8%, 58.3% vs 49.8%, 47.6% vs 34.1%, and 47.6% vs 31.5%, respectively (p-value=.19). Median OS of 6 cycles and 12 cycles groups were 35 months (95% confidence interval (CI), 11.0 to 58.9) and 23 months (95%CI, 16.9 to 29.0). The 12-, 24-, 36-, and 48- month PFS rates in 6 cycles and 12 cycles groups were 70.8% vs 56.9%, 39.5% and 32.7%, 27.1% vs 28.8%, and 21.1% vs 28.8%, respectively (p=.88). The Median PFS of 6 cycles and 12 cycles groups was 18 months (95% CI, 14.8 to 21.1) and 16 (95% CI, 11.0 to 20.9) months. Conclusion Patients with newly diagnosed high grade gliomas treated with adjuvant temozolomide after maximum safe surgical resection and adjuvant chemoradiation do not benefit from extended adjuvant temozolomide beyond 6 cycles. Trial registration Prospectively registered with the Iranian Registry of Clinical Trials: IRCT20160706028815N3. Date registered: 18/03/14.
Collapse
Affiliation(s)
- Kazem Anvari
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Seilanian Toussi
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | | | - Seyed Alireza Javadinia
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Hamidreza Saghafi
- Faculty of Medicine, Tehran Medical Branch of Islamic Azad University, Tehran, Iran
| | - James S. Welsh
- Department of Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Edward Hines Jr., VA Hospital, Maywood, IL, United States
| |
Collapse
|
3
|
Gately L, Mesía C, Sepúlveda JM, Del Barco S, Pineda E, Gironés R, Fuster J, Hong W, Dumas M, Gill S, Navarro LM, Herrero A, Dowling A, de Las Peñas R, Vaz MA, Alonso M, Lwin Z, Harrup R, Peralta S, Long A, Perez-Segura P, Ahern E, Garate CO, Wong M, Campbell R, Cuff K, Jennens R, Gallego O, Underhill C, Martinez-Garcia M, Covela M, Cooper A, Brown S, Rosenthal M, Torres J, Collins IM, Gibbs P, Balana C. A combined analysis of two prospective randomised studies exploring the impact of extended post-radiation temozolomide on survival outcomes in newly diagnosed glioblastoma. J Neurooncol 2024; 166:407-415. [PMID: 38153582 DOI: 10.1007/s11060-023-04513-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 12/29/2023]
Abstract
PURPOSE The optimal duration of post-radiation temozolomide in newly diagnosed glioblastoma remains unclear, with no published phase III randomised trials. Standard-of-care stipulates 6 months. However, in routine care, it is often extended to 12 months, despite lacking robust supporting data. METHODS GEINO14-01 (Spain) and EX-TEM (Australia) studies enrolled glioblastoma patients without progression at the end of 6 months post-radiation temozolomide. Participants were randomised 1:1 to six additional months of temozolomide or observation. Primary endpoint was 6-month progression free survival from date of randomisation (6mPFS). Secondary endpoints included overall survival (OS) and toxicity. 204 patients were required to detect an improvement in 6mPFS from 50 to 60% (80% power). Neither study recruited sufficient patients. We performed a combined analysis of individual patient data. RESULTS 205 patients were recruited: 159 in GEINO14-01 (2014-2018) and 46 in EX-TEM (2019-2022). Median follow-up was 20.0 and 14.5 months. Baseline characteristics were balanced. There was no significant improvement in 6mPFS (57.2% vs 64.0%, OR0.75, p = 0.4), nor across any subgroups, including MGMT methylated; PFS (HR0.92, p = 0.59, median 7.8 vs 9.7 months); or OS (HR1.03, p = 0.87, median 20.1 vs 19.4 months). During treatment extension, 64% experienced any grade adverse event, mainly fatigue and gastrointestinal (both 54%). Only a minority required treatment changes: 4.5% dose delay, 7.5% dose reduction, 1.5% temozolomide discontinuation. CONCLUSION For glioblastoma patients, extending post-radiation temozolomide from 6 to 12 months is well tolerated but does not improve 6mPFS. We could not identify any subset that benefitted from extended treatment. Six months should remain standard-of-care.
Collapse
Affiliation(s)
- L Gately
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia.
| | - C Mesía
- Medical Oncology Service, Institut Català d'Oncologia, Hospitalet de Llobregat, Barcelona, Spain
| | - J M Sepúlveda
- Medical Oncology Service, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - S Del Barco
- Medical Oncology Service, Institut Català d'Oncologia Girona, Girona, Spain
| | - E Pineda
- Medical Oncology Service, Hospital Clinic de Barcelona, Barcelona, Spain
| | - R Gironés
- Medical Oncology Service, Hospital Universitario La Fe, Valencia, Spain
| | - J Fuster
- Medical Oncology Service, Hospital Son Espases, Palma de Mallorca, Spain
| | - W Hong
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - M Dumas
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - S Gill
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
| | - L M Navarro
- Medical Oncology Service, Hospital de Salamanca, Salamanca, Spain
| | - A Herrero
- Medical Oncology Service, Hospital Miguel Servet, Zaragoza, Spain
| | - A Dowling
- Department of Medical Oncology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - R de Las Peñas
- Medical Oncology Service, Hospital Provincial de Castellón, Castellón, Spain
| | - M A Vaz
- Medical Oncology Service, Hospital Ramón y Cajal, Madrid, Spain
| | - M Alonso
- Medical Oncology Service, Hospital Virgen del Rocio, Seville, Spain
| | - Z Lwin
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - R Harrup
- Department of Medical Oncology, Royal Hobart Hospital, Hobart, TAS, Australia
| | - S Peralta
- Medical Oncology Service, Hospital Sant Joan de Reus, Reus, Spain
| | - A Long
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - P Perez-Segura
- Medical Oncology Service, Hospital Clinico San Carlos, Madrid, Spain
| | - E Ahern
- Department of Medical Oncology, Monash Health, Melbourne, VIC, Australia
| | - C O Garate
- Medical Oncology Service, Hospital Universitario Fundación Alcorcón, Alcorcón, Spain
| | - M Wong
- Department of Medical Oncology, Westmead Hospital, Westmead, NSW, Australia
| | - R Campbell
- Department of Medical Oncology, Bendigo Health, Bendigo, VIC, Australia
| | - K Cuff
- Department of Medical Oncology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - R Jennens
- Department of Medical Oncology, Epworth Health, Richmond, VIC, Australia
| | - O Gallego
- Medical Oncology Service, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - C Underhill
- Department of Medical Oncology, Border Medical Oncology, East Albury, NSW, Australia
| | | | - M Covela
- Medical Oncology Service, Hospital Lucus Augusti, Lugo, Spain
| | - A Cooper
- Department of Medical Oncology, Liverpool Hospital, Liverpool, NSW, Australia
| | - S Brown
- Department of Medical Oncology, Ballarat Health Services, Ballarat, VIC, Australia
| | - M Rosenthal
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - J Torres
- Department of Medical Oncology, Goulburn Valley Health, Shepparton, VIC, Australia
| | - I M Collins
- Department of Medical Oncology, South West Regional Cancer Centre, Geelong, VIC, Australia
| | - P Gibbs
- Personalised Oncology Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - C Balana
- Medical Oncology Service, Institut Català d'Oncologia, Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), Institut Investigació Germans Trias i Pujol (IGTP), Badalona, Spain
| |
Collapse
|
4
|
Sidibe I, Tensaouti F, Gilhodes J, Cabarrou B, Filleron T, Desmoulin F, Ken S, Noël G, Truc G, Sunyach MP, Charissoux M, Magné N, Lotterie JA, Roques M, Péran P, Cohen-Jonathan Moyal E, Laprie A. Pseudoprogression in GBM versus true progression in patients with glioblastoma: A multiapproach analysis. Radiother Oncol 2023; 181:109486. [PMID: 36706959 DOI: 10.1016/j.radonc.2023.109486] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE To investigate the feasibility of using a multiapproach analysis combining clinical data, diffusion- and perfusion-weighted imaging, and 3D magnetic resonance spectroscopic imaging to distinguish true tumor progression (TP) from pseudoprogression (PSP) in patients with glioblastoma. MATERIALS AND METHODS Progression was suspected within 6 months of radiotherapy in 46 of the 180 patients included in the Phase-III SpectroGlio trial (NCT01507506). Choline/creatine (Cho/Cr), choline/N-acetyl aspartate (Cho/NAA) and lactate/N-acetyl aspartate (Lac/NAA) ratios were extracted. Apparent diffusion coefficient (ADC) and cerebral blood volume (CBV) maps were calculated. ADC, relative CBV values and tumor volume (TV) were collected at relapse. Differences between TP and PSP were evaluated using Mann-Whitney tests, and p values were adjusted with Bonferroni correction. RESULTS Patients with suspected progression underwent a new MRI scan 1 month after the first one. Of these, 28 were classified as PSP, and 18 as TP. After a median follow-up of 41 months, median overall survival was higher in PSP than in TP (25.2 vs 20.3 months; p = 0.0092). Lac/NAA and Cho/Cr ratios were higher in TP than in PSP (1.2 vs 0.5; p = 0.006; and 3 vs 2.2; p = 0.021). After multivariate regression analysis, TV was the most significant predictor of TP vs PSP, and the only one retained in the model (p = 0.028). CONCLUSION Three spectroscopic ratios could be used to differentiate PSP from TP. TV at relapse was the most predictive factor in the multivariate analysis, and overall survival was higher in PSP than in TP.
Collapse
Affiliation(s)
- Ingrid Sidibe
- Radiation Oncology Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France; Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France
| | - Fatima Tensaouti
- Radiation Oncology Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France; Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France
| | - Julia Gilhodes
- Biostatistics Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France
| | - Bastien Cabarrou
- Biostatistics Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France
| | - Thomas Filleron
- Biostatistics Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France
| | - Soleakhena Ken
- Radiation Oncology Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France; Radiation Oncology Department, Toulouse Center for Cancer Research & INSERM, Toulouse, France
| | - Georges Noël
- Radiation Oncology Department, ICANS, Strasbourg, France
| | - Gilles Truc
- Radiation Oncology Department, Georges-François Leclerc Center, Dijon, France
| | | | | | - Nicolas Magné
- Radiation Oncology Department, Lucien Neuwirth Loire Cancer Institute, Saint-Priest-en-Jarez, France
| | - Jean-Albert Lotterie
- Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France
| | - Margaux Roques
- Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France
| | - Patrice Péran
- Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France
| | - Elizabeth Cohen-Jonathan Moyal
- Radiation Oncology Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France; Radiation Oncology Department, Toulouse Center for Cancer Research & INSERM, Toulouse, France
| | - Anne Laprie
- Radiation Oncology Department, Claudius Regaud Institute/Toulouse University Cancer Institute - Oncopôle, Toulouse, France; Toulouse NeuroImaging Center (ToNIC), University of Toulouse Paul Sabatier & INSERM, Toulouse, France.
| |
Collapse
|
5
|
Gupta T, Selvarajan JMP, Kannan S, Menon N, Dasgupta A, Chatterjee A. Updated systematic review and meta-analysis of extended adjuvant temozolomide in patients with newly diagnosed glioblastoma. Neurooncol Adv 2023; 5:vdad086. [PMID: 37638346 PMCID: PMC10457033 DOI: 10.1093/noajnl/vdad086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023] Open
Abstract
Background Contemporary standard-of-care for newly diagnosed glioblastoma (GBM) is maximal safe resection followed by postoperative focal conformal radiotherapy (RT) plus concurrent temozolomide (TMZ) and 6-cycles of adjuvant TMZ (Stupp regimen). However, many patients continue to receive extended adjuvant TMZ (beyond 6-cycles) without solid scientific evidence. This review pools data from nonrandomized studies and randomized controlled trials (RCTs) comparing extended adjuvant TMZ (>6-cycles) to standard adjuvant TMZ (6-cycles) in patients with newly diagnosed GBM for updated evidence-synthesis. Methods This systematic review and meta-analysis was carried out in accordance with the Cochrane methodology including quality assessment of primary studies. Primary outcome of interest was comparative efficacy defined as progression-free survival (PFS) and overall survival (OS). Hazard ratios (HRs) for PFS and OS with corresponding 95% confidence interval (CIs) were extracted/computed from individual primary studies and pooled using random-effects model. Any p-value <0.05 was considered statistically significant. Results Evidence-synthesis was based on pooling of data from 2578 patients enrolled in 16 nonrandomized comparative studies and 5 RCTs. Overall, extended adjuvant TMZ was associated with statistically significant reduction in the risk of progression (HR = 0.72, 95%CI: 0.60-0.87; p = 0.007) and death (HR = 0.71, 95%CI: 0.57-0.90; p = 0.004) compared to standard adjuvant TMZ. However, on subgroup analysis, survival benefit of extended adjuvant TMZ was limited to data synthesized from retrospective nonrandomized comparative studies with no statistically significant difference in outcomes seen after pooling of data from RCTs only. Conclusion Apparent survival benefit of extended adjuvant TMZ in newly diagnosed GBM is largely driven by nonrandomized comparative studies with high inherent potential for multiple biases.
Collapse
Affiliation(s)
- Tejpal Gupta
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | | | - Sadhana Kannan
- Department of Clinical Research Secretariat, ACTREC/TMH, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Nandini Menon
- Department of Medical Oncology, ACTREC/TMH, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Archya Dasgupta
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
6
|
Nomogram Model for Predicting the Prognosis of High-Grade Glioma in Adults Receiving Standard Treatment: A Retrospective Cohort Study. J Clin Med 2022; 12:jcm12010196. [PMID: 36614997 PMCID: PMC9821755 DOI: 10.3390/jcm12010196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES To identify the critical factors associated with the progression-free survival (PFS) and overall survival (OS) of high-grade glioma (HGG) in adults who have received standard treatment and establish a novel graphical nomogram and an online dynamic nomogram. PATIENTS AND METHODS This is a retrospective study of adult HGG patients receiving standard treatment (surgery, postoperative radiotherapy, and temozolomide (TMZ) chemotherapy) at Huashan Hospital, Fudan University between January 2017 and December 2019. We used uni- and multi-variable COX models to identify the significant prognostic factors for PFS and OS. Based on the significant predictors, graphical and online nomograms were established. RESULTS A total of 246 patients were enrolled in the study based on the inclusion criteria. The average PFS and OS were 22.99 ± 11.43 and 30.51 ± 13.73 months, respectively. According to the multi-variable COX model, age, extent of resection (EOR), and IDH mutation were associated with PFS and OS, while edema index (EI) was relevant to PFS. In addition, patients with IDH and TERT promoter co-mutations had longer PFSs and OSs, and no apparent survival benefit was found in the long-cycle TMZ adjuvant chemotherapy compared with the standard Stupp protocol. Based on these critical factors, a graphical nomogram and online nomogram were developed for predicting PFS and OS, respectively. The calibration curve showed favorable consistency between the predicted and actual survival rates. C-index and time-dependent AUC showed good discrimination abilities. CONCLUSIONS We identified the significant predictors for the PFS and OS of HGG adults receiving standard treatment and established user-friendly nomogram models to assist neurosurgeons in optimizing clinical management and treatment strategies.
Collapse
|
7
|
Wang J, Huang Y, Zhao F, Chen J, He L, Liu Z, Pei Y, Wei Z, Li R, Ai P, Peng X. Standard or extended STUPP? Optimal duration of temozolomide for patients with high-grade gliomas: a retrospective analysis. J Neurooncol 2022; 160:433-443. [DOI: 10.1007/s11060-022-04162-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2022]
|
8
|
Gupta T, Talukdar R, Kannan S, Dasgupta A, Chatterjee A, Patil V. Efficacy and safety of extended adjuvant temozolomide compared to standard adjuvant temozolomide in glioblastoma: Updated systematic review and meta-analysis. Neurooncol Pract 2022; 9:354-363. [PMID: 36134016 PMCID: PMC9476976 DOI: 10.1093/nop/npac036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background This study was designed to compare outcomes of extended adjuvant temozolomide (TMZ) vs standard adjuvant TMZ following radiotherapy (RT) plus concurrent TMZ in newly diagnosed glioblastoma. Methods This systematic review and meta-analysis was carried out in accordance with Cochrane methodology. Only prospective clinical trials randomly assigning adults with newly diagnosed glioblastoma after concurrent RT/TMZ to 6 cycles of adjuvant TMZ (control arm) or extended (>6 cycles) adjuvant TMZ (experimental arm) were eligible. Primary outcome of interest was overall survival, while progression-free survival and toxicity were secondary endpoints. Hazard ratio (HR) for progression and death with corresponding 95% confidence interval (CI) were computed for individual primary study and pooled using random-effects model. Toxicity was defined as proportion of patients with ≥grade 3 hematologic toxicity and expressed as risk ratio (RR) with 95% CI. Any P-value <.05 was considered statistically significant. Results Systematic literature review identified five randomized controlled trials comparing standard (6 cycles) vs extended (>6 cycles) adjuvant TMZ in newly diagnosed glioblastoma. Outcome data could be extracted from 358 patients from four primary studies. Extended adjuvant TMZ was not associated with statistically significant reduction in the risk of progression (HR = 0.82, 95% CI: 0.61-1.10; P = .18) or death (HR = 0.87, 95% CI:0.60-1.27; P = .48) compared to standard adjuvant TMZ. Grade ≥3 hematologic toxicity though somewhat higher with extended adjuvant TMZ, was not significantly different between the two arms (RR = 2.01, 95% CI: 0.83-4.87; P = .12). Conclusions There is low-certainty evidence that extended adjuvant TMZ is not associated with significant survival benefit or increased hematologic toxicity in unselected patients with newly diagnosed glioblastoma compared to standard adjuvant TMZ.
Collapse
Affiliation(s)
- Tejpal Gupta
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi
Bhabha National Institute (HBNI), Mumbai,
India
| | - Riddhijyoti Talukdar
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi
Bhabha National Institute (HBNI), Mumbai,
India
| | - Sadhana Kannan
- Department of Clinical Research Secretariat, ACTREC/TMH, Tata Memorial
Centre, Homi Bhabha National Institute (HBNI),
Mumbai, India
| | - Archya Dasgupta
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi
Bhabha National Institute (HBNI), Mumbai,
India
| | - Abhishek Chatterjee
- Department of Radiation Oncology, ACTREC/TMH, Tata Memorial Centre, Homi
Bhabha National Institute (HBNI), Mumbai,
India
| | - Vijay Patil
- Department of Medical Oncology, ACTREC/TMH, Tata Memorial Centre, Homi
Bhabha National Institute (HBNI), Mumbai,
India
| |
Collapse
|
9
|
Ohno M, Miyakita Y, Takahashi M, Yanagisawa S, Tamura Y, Narita Y. Continuing maintenance temozolomide therapy beyond 12 cycles confers no clinical benefit over discontinuation at 12 cycles in patients with IDH1/2-wildtype glioblastoma. Jpn J Clin Oncol 2022; 52:1134-1142. [PMID: 35858227 DOI: 10.1093/jjco/hyac114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/30/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE The optimal duration of maintenance temozolomide therapy is controversial. We aimed to examine the clinical benefits of continuing temozolomide therapy beyond 12 cycles in patients with glioblastoma. METHODS We included 41 patients with isocitrate dehydrogenase 1/2-wildtype glioblastoma, who received 12 or more cycles of temozolomide therapy between June 2006 and December 2019. We evaluated the outcome between 16 patients who continued temozolomide therapy beyond 12 cycles up to 24 cycles (≥13 cycles group) and 25 patients wherein temozolomide therapy was discontinued at 12 cycles (12 cycles group). RESULTS The median progression-free survival and survival time after completing 12 cycles (residual progression-free survival and residual overall survival) did not differ between the 12 cycles group and ≥13 cycles group (residual progression-free survival: 11.3 vs. 9.2 months, P = 0.61, residual overall survival: 25.7 vs. 30.2 months, P = 0.76). Multivariate analysis including temozolomide therapy beyond 12 cycles, age at 12 cycles, Karnofsky performance status at 12 cycles, residual tumor at 12 cycles, maintenance therapy regimen and O-6-methylguanine deoxyribonucleic acid methyltransferase promoter methylation status revealed that extended temozolomide therapy beyond 12 cycles was not correlated with residual progression-free survival and residual overall survival (P = 0.80 and P = 0.41, respectively) but Karnofsky performance status at 12 cycles ≥80 was significantly associated with increased residual overall survival (P = 0.0012). CONCLUSIONS Continuing temozolomide beyond 12 cycles confers no clinical benefit over the discontinuation of temozolomide at 12 cycles. Karnofsky performance status at 12 cycles ≥80 may serve as a novel predictive factor for long-term survival.
Collapse
Affiliation(s)
- Makoto Ohno
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Yasuji Miyakita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Masamichi Takahashi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Shunsuke Yanagisawa
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Yukie Tamura
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| |
Collapse
|
10
|
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
|
11
|
Prather KY, O’Neal CM, Westrup AM, Tullos HJ, Hughes KL, Conner AK, Glenn CA, Battiste JD. A systematic review of amino acid PET in assessing treatment response to temozolomide in glioma. Neurooncol Adv 2022; 4:vdac008. [PMID: 35300149 PMCID: PMC8923003 DOI: 10.1093/noajnl/vdac008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The response assessment in neuro-oncology (RANO) criteria have been the gold standard for monitoring treatment response in glioblastoma (GBM) and differentiating tumor progression from pseudoprogression. While the RANO criteria have played a key role in detecting early tumor progression, their ability to identify pseudoprogression is limited by post-treatment damage to the blood-brain barrier (BBB), which often leads to contrast enhancement on MRI and correlates poorly to tumor status. Amino acid positron emission tomography (AA PET) is a rapidly growing imaging modality in neuro-oncology. While contrast-enhanced MRI relies on leaky vascularity or a compromised BBB for delivery of contrast agents, amino acid tracers can cross the BBB, making AA PET particularly well-suited for monitoring treatment response and diagnosing pseudoprogression. The authors performed a systematic review of PubMed, MEDLINE, and Embase through December 2021 with the search terms “temozolomide” OR “Temodar,” “glioma” OR “glioblastoma,” “PET,” and “amino acid.” There were 19 studies meeting inclusion criteria. Thirteen studies utilized [18F]FET, five utilized [11C]MET, and one utilized both. All studies used static AA PET parameters to evaluate TMZ treatment in glioma patients, with nine using dynamic tracer parameters in addition. Throughout these studies, AA PET demonstrated utility in TMZ treatment monitoring and predicting patient survival.
Collapse
Affiliation(s)
- Kiana Y Prather
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Christen M O’Neal
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Alison M Westrup
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hurtis J Tullos
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kendall L Hughes
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andrew K Conner
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Chad A Glenn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - James D Battiste
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
12
|
Liu J, Li C, Wang Y, Ji P, Guo S, Zhai Y, Wang N, Lou M, Xu M, Chao M, Jiao Y, Zhao W, Feng F, Qu Y, Ge S, Wang L. Prognostic and Predictive Factors in Elderly Patients With Glioblastoma: A Single-Center Retrospective Study. Front Aging Neurosci 2022; 13:777962. [PMID: 35173600 PMCID: PMC8841486 DOI: 10.3389/fnagi.2021.777962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant intracranial tumor and the median age at diagnosis is 65 years. However, elderly patients are usually excluded from clinical studies and age is considered as an independent negative prognostic factor for patients with GBM. Therefore, the best treatment method for GBM in elderly patients has remained controversial. Elderly GBM patients (≥ 60 years old) treated between January 2015 and December 2019 were enrolled in this study. Medical records were reviewed retrospectively, and clinicopathological characteristics, treatments, and outcomes were analyzed. A total of 68 patients were included, with a median age of 65.5 years (range: 60–79). The median preoperative Karnofsky performance scale (KPS) score was 90 (range 40–100) and median postoperative KPS score was 80 (range 0–90). Univariate analysis results showed that age, gender, comorbidities, preoperative KPS < 90 and MGMT promoter methylation were not significantly associated with PFS and OS. On the other hand, total resection, postoperative KPS ≥ 80, Ki67 > 25%, and Stupp-protocol treatment were significantly associated with prolonged PFS and OS. Moreover, multivariate analysis found that postoperative KPS ≥ 80, total resection, and Stupp-protocol treatment were prognostic factors for PFS and OS. The findings of this study have suggested that, on the premise of protecting function as much as possible, the more aggressive treatment regimens may prolong survival for elderly patients with GBM. However, further studies, particularly prospective randomized clinical trials, should be conducted to provide more definitive data on the appropriate management of elderly patients, especially for patients with MGMT promoter methylation.
Collapse
Affiliation(s)
- Jinghui Liu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Chen Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yuan Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Peigang Ji
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Shaochun Guo
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yulong Zhai
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Na Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Miao Lou
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meng Xu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Min Chao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yang Jiao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Wenjian Zhao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Fuqiang Feng
- Department of Neurosurgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
| | - Shunnan Ge
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- *Correspondence: Shunnan Ge,
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an, China
- Liang Wang,
| |
Collapse
|
13
|
Attarian F, Taghizadeh-Hesary F, Fanipakdel A, Javadinia SA, Porouhan P, PeyroShabany B, Fazilat-Panah D. A Systematic Review and Meta-Analysis on the Number of Adjuvant Temozolomide Cycles in Newly Diagnosed Glioblastoma. Front Oncol 2021; 11:779491. [PMID: 34900732 PMCID: PMC8651479 DOI: 10.3389/fonc.2021.779491] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/29/2021] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND In newly diagnosed glioblastoma, radiation with concurrent and adjuvant (six cycles) temozolomide (TMZ) is the established standard of postsurgical care. However, the benefit of extending adjuvant TMZ therapy beyond six cycles has remained unknown. METHODS We searched PubMed, Web of Science, Scopus, and Embase up to October 1, 2021. The search keywords were "glioblastoma," "adjuvant chemotherapy," and their synonyms. The data of randomized clinical trials were extracted and included in this meta-analysis if they had reported patients' median overall survival (OS) or median progression-free survival (PFS). The standard and extended chemotherapy regimens were considered as adjuvant TMZ up to six cycles and beyond six cycles (up to a total of 12 cycles), respectively. The median OS and median PFS were pooled and compared. RESULTS Four studies consisting of 882 patients (461 patients for the standard chemotherapy group and 421 patients for the extended chemotherapy group) were included in this meta-analysis. The extended TMZ regimen was associated with a nonsignificant improvement in PFS [12.0 months (95% CI 9.0 to 15.0) vs. 10.0 months (95% CI 7.0 to 12.0), P = 0.27] without corresponding improvement in OS [23.0 months (95% CI 19.0 to 27.0) and 24.0 months (95% CI 20.0 to 28.0), P = 0.73]. CONCLUSIONS In newly diagnosed glioblastoma, continuing adjuvant TMZ beyond six cycles did not shown an increase neither in PFS nor OS.
Collapse
Affiliation(s)
- Fahimeh Attarian
- Department of Public Health, School of Health, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | | | - Azar Fanipakdel
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Alireza Javadinia
- Vasei Clinical Research Development Unit, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Pejman Porouhan
- Department of Radiation Oncology, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Babak PeyroShabany
- Department of Internal Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | |
Collapse
|
14
|
Álvarez-Torres MDM, Fuster-García E, Balaña C, Puig J, García-Gómez JM. Lack of Benefit of Extending Temozolomide Treatment in Patients with High Vascular Glioblastoma with Methylated MGMT. Cancers (Basel) 2021; 13:5420. [PMID: 34771583 PMCID: PMC8582449 DOI: 10.3390/cancers13215420] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 11/25/2022] Open
Abstract
In this study, we evaluated the benefit on survival of the combination of methylation of O6-methylguanine-DNA methyltransferase (MGMT) promotor gene and moderate vascularity in glioblastoma using a retrospective dataset of 123 patients from a multicenter cohort. MRI processing and calculation of relative cerebral blood volume (rCBV), used to define moderate- and high-vascular groups, were performed with the automatic ONCOhabitats method. We assessed the previously proposed rCBV threshold (10.7) and the new calculated ones (9.1 and 9.8) to analyze the association with survival for different populations according to vascularity and MGMT methylation status. We found that patients included in the moderate-vascular group had longer survival when MGMT is methylated (significant median survival difference of 174 days, p = 0.0129*). However, we did not find significant differences depending on the MGMT methylation status for the high-vascular group (p = 0.9119). In addition, we investigated the combined correlation of MGMT methylation status and rCBV with the prognostic effect of the number of temozolomide cycles, and only significant results were found for the moderate-vascular group. In conclusion, there is a lack of benefit of extending temozolomide treatment for patients with high vascular glioblastomas, even presenting MGMT methylation. Preliminary results suggest that patients with moderate vascularity and methylated MGMT glioblastomas would benefit more from prolonged adjuvant chemotherapy.
Collapse
Affiliation(s)
- María del Mar Álvarez-Torres
- Biomedical Data Science Laboratory, Instituto Universitario de Tecnologías de la Información y Comunicaciones, Universitat Politècnica de València, 46022 Valencia, Spain; (E.F.-G.); (J.M.G.-G.)
| | - Elies Fuster-García
- Biomedical Data Science Laboratory, Instituto Universitario de Tecnologías de la Información y Comunicaciones, Universitat Politècnica de València, 46022 Valencia, Spain; (E.F.-G.); (J.M.G.-G.)
- Department of Diagnostic Physics, Oslo University Hospital, 0450 Oslo, Norway
| | - Carmen Balaña
- Institut Catala d’Oncologia (ICO), Applied Research Group in Oncology (B-ARGO Group), Institut Investigació Germans Trias i Pujol (IGTP), 08916 Badalona, Spain;
| | - Josep Puig
- Institut de Diagnostic per la Image (IDI), Hospital Dr. Josep Trueta, 17007 Girona, Spain;
| | - Juan M. García-Gómez
- Biomedical Data Science Laboratory, Instituto Universitario de Tecnologías de la Información y Comunicaciones, Universitat Politècnica de València, 46022 Valencia, Spain; (E.F.-G.); (J.M.G.-G.)
| |
Collapse
|
15
|
Wang Y, Zhang J, Li W, Jiang T, Qi S, Chen Z, Kang J, Huo L, Wang Y, Zhuge Q, Gao G, Wu Y, Feng H, Zhao G, Yang X, Zhao H, Wang Y, Yang H, Kang D, Su J, Li L, Jiang C, Li G, Qiu Y, Wang W, Wang H, Xu Z, Zhang L, Wang R. Guideline conformity to the Stupp regimen in patients with newly diagnosed glioblastoma multiforme in China. Future Oncol 2021; 17:4571-4582. [PMID: 34519220 DOI: 10.2217/fon-2021-0435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aims: To determine how consistently Chinese glioblastoma multiforme (GBM) patients were treated according to the Stupp regimen. Patients and methods: The proportion of treatments conforming to the Stupp regimen and reasons for nonconformity were evaluated in 202 newly diagnosed GBM patients. Results: Only 15.8% of GBM patients received treatments compliant with the Stupp regimen. The main deviations were temozolomide dosages >75 mg/m2 (58/120; 48.3%) and treatment durations <42 days (84/120; 70.0%) in the concomitant phase and temozolomide dosages <150 mg/m2 (89/101; 88.1%) in the maintenance phase. Median overall survival (27.09 vs 18.21 months) and progression-free survival (14.27 vs 12.10 months) were longer in patients who received Stupp regimen-compliant treatments. Conclusion: Increased conformity to the Stupp regimen is needed for GBM patients in China.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100010, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, 310009, China
| | - Wenbin Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 100070, China
| | - Taipeng Jiang
- Department of Neurosurgery, Shenzhen Second People's Hospital, 518035, China
| | - Songtao Qi
- Department of Neurosurgery, Nanfang Hospital Southern Medical University, 510515, China
| | - Zhongping Chen
- Department of Neurosurgery, Sun Yat-Sen University Cancer Center, 510060, China
| | - Jingbo Kang
- Tumor Diagnosis & Treatment Center, The Sixth Medical Center of PLA General Hospital, 100048, China
| | - Lei Huo
- Department of Neurosurgery, Xiangya Hospital Central South University, 410008, China
| | - Yunjie Wang
- Department of Neurosurgery, The First Hospital of China Medical University, 110001, China
| | - Qichuan Zhuge
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, 325000, China
| | - Guodong Gao
- Department of Neurosurgery, The Fourth Military University Tangdu Hospital, 710032, China
| | - Yuping Wu
- Department of Craniobasal Neurology, Sichuan Cancer Hospital & Institute, The Affiliated Cancer Hospital, School of Medicine, UESTC, 610041, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, The First Affiliated Hospital of Army Medical University, 400038, China
| | - Gang Zhao
- Department of Neurosurgery, The First Bethune Hospital of Jilin University, 130021, China
| | - Xiaopeng Yang
- Department of Neurosurgery, People's Hospital of Xinjiang Uygur Autonomous Region, 830001, China
| | - Hui Zhao
- Department of Radiotherapy, People's Hospital of Xinjiang Uygur Autonomous Region, 830001, China
| | - Yirong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital of Army Medical University, 400037, China
| | - Dezhi Kang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, 350005, China
| | - Jun Su
- Department of Neurosurgery, Tumor Hospital of Harbin Medical University, 150081, China
| | - Liang Li
- Department of Neurosurgery, Peking University First Hospital, 100034, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 150001, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University, 250012, China
| | - Yongming Qiu
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, China
| | - Weimin Wang
- Department of Neurosurgery, General Hospital of Southern Theatre Command, 510010, China
| | - Handong Wang
- Department of Neurosurgery, General Hospital of Eastern Theatre Command, 210002, China
| | - Zaihua Xu
- Department of Neurosurgery, General Hospital of Northern Theatre Command, 110840, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 100070, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, 100010, China
| |
Collapse
|
16
|
Haksoyler V, A Besen A, Koseci T, Olgun P, Bayram E, Topkan E. Neutrophil-to-lymphocyte ratio is prognostic in recurrent glioblastoma multiforme treated with bevacizumab plus irinotecan. Biomark Med 2021; 15:851-859. [PMID: 33983042 DOI: 10.2217/bmm-2021-0271] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022] Open
Abstract
Aim: We intended to survey the prognostic utility of pretreatment neutrophil-to-lymphocyte ratio (NLR) as a novel prognostic index in recurrent glioblastoma multiforme (R-GBMs) treated with bevacizumab plus irinotecan (BEVIRI). Patients & methods: The present retrospective investigation incorporated the R-GBMs patients who underwent BEVIRI. The pre-BEVIRI NLR was calculated for each patient by utilizing the complete blood count tests obtained on the first day of BEVIRI. Results: The data of a total of 103 patients were analyzed. The ideal cutoff was identified at 3.04 (area under the curve: 60%; sensitivity: 60.3%; specificity 60%) for the pre-BEVIRI NLR. Low-NLR group had significantly longer overall survival times than the high-NLR group (15.8 vs 9.3 months; p = 0.015). Conclusion: NLR might be utilized as a novel biomarker in the prognostic stratification of the R-GBMs treated with BEVIRI.
Collapse
Affiliation(s)
| | - Ali A Besen
- Department of Medical Oncology, Baskent University Medical Faculty, Adana, Turkey
| | - Tolga Koseci
- Department of Medical Oncology, City Hospital, Adana, Turkey
| | - Polat Olgun
- Department of Medical Oncology, Near East University, Cyprus, Turkey
| | - Ertugrul Bayram
- Department of Medical Oncology, Cukurova University, Adana, Turkey
| | - Erkan Topkan
- Department of Radiation Oncology, Baskent University Medical Faculty, Adana, Turkey
| |
Collapse
|
17
|
Asano K, Fumoto T, Matsuzaka M, Hasegawa S, Suzuki N, Akasaka K, Katayama K, Kamataki A, Kurose A, Ohkuma H. Combination chemoradiotherapy with temozolomide, vincristine, and interferon-β might improve outcomes regardless of O6-methyl-guanine-DNA-methyltransferase (MGMT) promoter methylation status in newly glioblastoma. BMC Cancer 2021; 21:867. [PMID: 34320929 PMCID: PMC8320052 DOI: 10.1186/s12885-021-08592-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/05/2021] [Indexed: 12/30/2022] Open
Abstract
Background This investigator-initiated, open-label, single-arm, single-institute study was conducted to investigate the effectiveness of induction combination chemoradiotherapy and long-term maintenance therapy with temozolomide (TMZ) plus interferon (IFN)-β for glioblastoma. Methods The initial induction combination chemoradiotherapy comprised radiotherapy plus TMZ plus vincristine plus IFN-β. Maintenance chemotherapy comprised monthly TMZ, continued for 24–50 cycles, plus weekly IFN-β continued for as long as possible. The primary endpoint was 2-year overall survival (2y-OS). The study protocol was to be considered valid if the expected 2y-OS was over 38% and the lower limit of the 95% confidence interval (CI) was no less than 31.7% compared with historical controls, using Kaplan-Meier methods. Secondary endpoints were median progression-free survival (mPFS), median OS (mOS), 5-year OS rate (5y-OS), and mPFS and mOS classified according to MGMT promoter methylation status. Results Forty-seven patients were analyzed. The 2y-OS was 40.7% (95%CI, 27.5–55.4%). The mPFS and mOS were 11.0 months and 18.0 months, respectively, and 5y-OS was 20.3% (95%CI, 10.9–34.6%). The mPFS in groups with and without MGMT promoter methylation in the tumor was 10.0 months and 11.0 months (p = 0.59), respectively, and mOS was 24.0 months and 18.0 months (p = 0.88), respectively. The frequency of grade 3/4 neutropenia was 19.1%. Conclusions The 2y-OS with induction multidrug combination chemoradiotherapy and long-term maintenance therapy comprising TMZ plus IFN-β tended to exceed that of historical controls, but the lower limit of the 95%CI was below 31.7%. Although the number of cases was small, this protocol may rule out MGMT promoter methylation status as a prognostic factor. Trial registration University Hospital Medical Information Network (number UMIN000040599).
Collapse
Affiliation(s)
- Kenichiro Asano
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan.
| | - Toshio Fumoto
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Masashi Matsuzaka
- Clinical Research Support Center, Hirosaki University Hospital, 53 Hon-cho, Hirosaki, Aomori, 036-8563, Japan.,Department of Medical Informatics, Hirosaki University Hospital, 53 Hon-cho, Hirosaki, Aomori, 036-8563, Japan
| | - Seiko Hasegawa
- Department of Neurosurgery, Kuroishi General Hospital, 1-70 Kitami-cho, Kuroishi, Aomori, 036-0541, Japan
| | - Naoya Suzuki
- Department of Neurosurgery, Towada City Hospital, 8-14 Nishi-Jyuniban-cho, Towada, Aomori, 034-0093, Japan
| | - Kenichi Akasaka
- Department of Neurosurgery, Towada City Hospital, 8-14 Nishi-Jyuniban-cho, Towada, Aomori, 034-0093, Japan
| | - Kosuke Katayama
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| | - Akihisa Kamataki
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 53 Honcho, Hirosaki, Aomori, 036-8563, Japan
| | - Akira Kurose
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, 53 Honcho, Hirosaki, Aomori, 036-8563, Japan
| | - Hiroki Ohkuma
- Department of Neurosurgery, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori, 036-8562, Japan
| |
Collapse
|
18
|
Anselmo P, Maranzano E, Selimi A, Lupattelli M, Palumbo I, Bini V, Casale M, Trippa F, Bufi A, Arcidiacono F, Aristei C. Clinical characterization of glioblastoma patients living longer than 2 years: A retrospective analysis of two Italian institutions. Asia Pac J Clin Oncol 2021; 17:273-279. [PMID: 33078909 DOI: 10.1111/ajco.13457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/12/2020] [Indexed: 11/28/2022]
Abstract
AIM Despite the advances in surgery and radio-chemotherapy, the prognosis of glioblastoma (GBM) remains poor with about 13% of patients alive at 24 months. METHODS A total of 75 long-term survivors (LTS), defined as alive at least 24 months from diagnosis, were retrospectively analyzed. Overall survival (OS) and recurrence-free-survival (RFS) were calculated and related to patient characteristics and treatment received. RESULTS Median age and Karnofsky performance status (KPS) were 56 years and 100%, respectively. After surgery (gross tumor resection-GTR in 62, 83% patients), all LTS received concomitant temozolomide (TMZ) with radiotherapy and 70 (93%) adjuvant TMZ. Of these, 10 (13%) discontinued TMZ prior the completion of 6 cycles, 37 (49%) received 6 cycles and 23 (31%) >6 cycles. Sixty-nine (92%) patients experienced a first tumor recurrence at a median time of 21 months. Of these, 32 (46%) were submitted to a second surgery, 34 (49%) to other no-surgical treatments and 3 (5%) only supportive care. At multivariate analysis, OS was significantly improved by second surgery after first recurrence (P = 0.0032) and by cycles of adjuvant TMZ > 6 versus ≤6 (P = 0.05). More than six cycles of TMZ significantly conditioned also first RFS (P = 0.011) and second RFS (P = 0.033). CONCLUSION The large majority of LTS had <65 years, had a high KPS and received GTR. OS and RFS resulted significantly related to an extended administration of adjuvant TMZ (>6 cycles) and a second surgery in case of recurrence.
Collapse
Affiliation(s)
- Paola Anselmo
- Radiotherapy Oncology Centre, "Santa Maria" Hospital, Terni, Italy
| | | | - Adelina Selimi
- Department of Surgical and Biomedical Science, Radiotherapy Oncology Centre, University of Perugia and "Santa Maria della Misericordia" Hospital, Perugia, Italy
| | - Marco Lupattelli
- Radiotherapy Oncology Centre, "Santa Maria della Misericordia" Hospital, Perugia, Italy
| | - Isabella Palumbo
- Department of Surgical and Biomedical Science, Radiotherapy Oncology Centre, University of Perugia and "Santa Maria della Misericordia" Hospital, Perugia, Italy
| | - Vittorio Bini
- Internal Medicine, Endocrinology & Metabolism, University of Perugia and "Santa Maria della Misericordia" Hospital, Perugia, Italy
| | - Michelina Casale
- Radiotherapy Oncology Centre, "Santa Maria" Hospital, Terni, Italy
| | - Fabio Trippa
- Radiotherapy Oncology Centre, "Santa Maria" Hospital, Terni, Italy
| | - Alessandro Bufi
- Department of Surgical and Biomedical Science, Radiotherapy Oncology Centre, University of Perugia and "Santa Maria della Misericordia" Hospital, Perugia, Italy
| | | | - Cynthia Aristei
- Department of Surgical and Biomedical Science, Radiotherapy Oncology Centre, University of Perugia and "Santa Maria della Misericordia" Hospital, Perugia, Italy
| |
Collapse
|
19
|
Balana C, Vaz MA, Manuel Sepúlveda J, Mesia C, Del Barco S, Pineda E, Muñoz-Langa J, Estival A, de Las Peñas R, Fuster J, Gironés R, Navarro LM, Gil-Gil M, Alonso M, Herrero A, Peralta S, Olier C, Perez-Segura P, Covela M, Martinez-García M, Berrocal A, Gallego O, Luque R, Perez-Martín FJ, Esteve A, Munne N, Domenech M, Villa S, Sanz C, Carrato C. A phase II randomized, multicenter, open-label trial of continuing adjuvant temozolomide beyond 6 cycles in patients with glioblastoma (GEINO 14-01). Neuro Oncol 2021; 22:1851-1861. [PMID: 32328662 DOI: 10.1093/neuonc/noaa107] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Standard treatment for glioblastoma is radiation with concomitant and adjuvant temozolomide for 6 cycles, although the optimal number of cycles of adjuvant temozolomide has long been a subject of debate. We performed a phase II randomized trial investigating whether extending adjuvant temozolomide for more than 6 cycles improved outcome. METHODS Glioblastoma patients treated at 20 Spanish hospitals who had not progressed after 6 cycles of adjuvant temozolomide were centrally randomized to stop (control arm) or continue (experimental arm) temozolomide up to a total of 12 cycles at the same doses they were receiving in cycle 6. Patients were stratified by MGMT methylation and measurable disease. The primary endpoint was differences in 6-month progression-free survival (PFS). Secondary endpoints were PFS, overall survival (OS), and safety (Clinicaltrials.gov NCT02209948). RESULTS From August 2014 to November 2018, 166 patients were screened, 7 of whom were ineligible. Seventy-nine patients were included in the stop arm and 80 in the experimental arm. All patients were included in the analyses of outcomes and of safety. There were no differences in 6-month PFS (control 55.7%; experimental 61.3%), PFS, or OS between arms. MGMT methylation and absence of measurable disease were independent factors of better outcome. Patients in the experimental arm had more lymphopenia (P < 0.001), thrombocytopenia (P < 0.001), and nausea and vomiting (P = 0.001). CONCLUSIONS Continuing temozolomide after 6 adjuvant cycles is associated with greater toxicity but confers no additional benefit in 6-month PFS. KEY POINTS 1. Extending adjuvant temozolomide to 12 cycles did not improve 6-month PFS.2. Extending adjuvant temozolomide did not improve PFS or OS in any patient subset.3. Extending adjuvant temozolomide was linked to increased toxicities.
Collapse
Affiliation(s)
- Carmen Balana
- Medical Oncology Service, Institut Català d'Oncologia, Badalona, Spain.,Applied Research Group in Oncology (B-ARGO) from the Institut Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Spain
| | | | | | - Carlos Mesia
- Medical Oncology Service, Institut Català d'Oncologia, Hospitalet de Llobregat, Spain
| | - Sonia Del Barco
- Medical Oncology Service, Institut Català d'Oncologia Girona, Girona, Spain
| | - Estela Pineda
- Medical Oncology Service, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Jose Muñoz-Langa
- Medical Oncology Service, Hospital Universitario La Fe, Valencia, Spain
| | - Anna Estival
- Medical Oncology Service, Institut Català d'Oncologia, Badalona, Spain.,Applied Research Group in Oncology (B-ARGO) from the Institut Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Ramón de Las Peñas
- Medical Oncology Service, Hospital Provincial de Castellón, Castellón, Spain
| | - Jose Fuster
- Medical Oncology Service, Hospital Son Espases, Palma De Mallorca, Spain
| | - Regina Gironés
- Medical Oncology Service, Hospital Universitario La Fe, Valencia, Spain
| | | | - Miguel Gil-Gil
- Medical Oncology Service, Institut Català d'Oncologia, Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL) Hospitalet de Llobregat, Spain
| | - Miriam Alonso
- Medical Oncology Service, Hospital Virgen del Rocio, Sevilla, Spain
| | - Ana Herrero
- Medical Oncology Service, Hospital Miguel Servet, Zaragoza, Spain
| | - Sergio Peralta
- Medical Oncology Service, Hospital Sant Joan de Reus, Reus, Spain
| | - Clara Olier
- Medical Oncology Service, Fundación Alcorcón, Madrid, Spain
| | - Pedro Perez-Segura
- Medical Oncology Service, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Maria Covela
- Medical Oncology Service, Hospital Lucus Agusti, Lugo, Spain
| | | | - Alfonso Berrocal
- Medical Oncology Service, Hospital General Universitario de Valencia, Valencia, Spain
| | - Oscar Gallego
- Medical Oncology Service, Hospital de Sant Pau, Barcelona, Spain
| | - Raquel Luque
- Medical Oncology Service, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Franciso Javier Perez-Martín
- Medical Oncology Service, Institut Català d'Oncologia, Hospitalet de Llobregat, Spain.,Bellvitge Biomedical Research Institute (IDIBELL) Hospitalet de Llobregat, Spain
| | - Anna Esteve
- Applied Research Group in Oncology (B-ARGO) from the Institut Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Spain
| | - Nuria Munne
- Pathology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Marta Domenech
- Medical Oncology Service, Institut Català d'Oncologia, Badalona, Spain
| | - Salvador Villa
- Radiation Therapy Oncology Service, Institut Català d'Oncologia, Badalona, Spain
| | - Carolina Sanz
- Pathology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Cristina Carrato
- Pathology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
20
|
Liu S, Zhao Q, Shi W, Zheng Z, Liu Z, Meng L, Dong L, Jiang X. Advances in radiotherapy and comprehensive treatment of high-grade glioma: immunotherapy and tumor-treating fields. J Cancer 2021; 12:1094-1104. [PMID: 33442407 PMCID: PMC7797642 DOI: 10.7150/jca.51107] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 11/21/2020] [Indexed: 12/18/2022] Open
Abstract
High-grade gliomas (HGGs) are the most common primary malignant brain tumors. They have a high degree of malignancy and show invasive growth. The personal treatment plan for HGG is based on the patient's age, performance status, and degree of tumor invasion. The basic treatment plan for HGG involves tumor resection, radiotherapy (RT) with concomitant temozolomide (TMZ), and adjuvant TMZ chemotherapy. The basic radiation technology includes conventional RT, three-dimensional conformal RT, intensity-modulated RT, and stereotactic RT. As our understanding of tumor pathogenesis has deepened, so-called comprehensive treatment schemes have attracted attention. These combine RT with chemotherapy, molecular targeted therapy, immunotherapy, or tumor-treating fields. These emerging treatments are expected to improve the prospects of patients with HGG. In the present article, we review the recent advances in RT and comprehensive treatment for patients with newly diagnosed and recurrent HGG.
Collapse
Affiliation(s)
- Shiyu Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Qin Zhao
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Weiyan Shi
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zhuangzhuang Zheng
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Zijing Liu
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Lihua Dong
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, China.,Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun 130021, China.,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China
| |
Collapse
|
21
|
Redjal N, Nahed BV, Dietrich J, Kalkanis SN, Olson JJ. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of chemotherapeutic management and antiangiogenic treatment of newly diagnosed glioblastoma in adults. J Neurooncol 2020; 150:165-213. [PMID: 33215343 DOI: 10.1007/s11060-020-03601-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/08/2020] [Indexed: 12/01/2022]
Abstract
QUESTION What is the role of temozolomide in the management of adult patients (aged 65 and under) with newly diagnosed glioblastoma? TARGET POPULATION These recommendations apply to adult patients diagnosed with newly diagnosed glioblastoma. RECOMMENDATION Level I: Concurrent and post-irradiation Temozolomide (TMZ) in combination with radiotherapy and post-radiotherapy as described by Stupp et al. is recommended to improve both PFS and OS in adult patients with newly diagnosed GBM. There is no evidence that alterations in the dosing regimen have additional beneficial effect. QUESTION Is there benefit to adjuvant temozolomide treatment in elderly patients (> 65 years old?). TARGET POPULATION These recommendations apply to adult patients diagnosed with newly diagnosed glioblastoma. RECOMMENDATION Level III: Adjuvant TMZ treatment is suggested as a treatment option to improve PFS and OS in adult patients (over 70 years of age) with newly diagnosed GBM. QUESTION What is the role of local regional chemotherapy with BCNU biodegradable polymeric wafers in adult patients with newly diagnosed glioblastoma? TARGET POPULATION These recommendations apply to adult patients diagnosed with newly diagnosed glioblastoma. RECOMMENDATION Level III: There is insufficient evidence for the use of BCNU wafers following resection in patients with newly diagnosed glioblastoma who undergo the Stupp protocol after surgery. Further studies of higher quality are suggested to understand the role of BCNU wafer and other locoregional therapy in the setting of Stupp Protocol. QUESTION What is the role of bevacizumab in the adult patient with newly diagnosed glioblastoma? TARGET POPULATION These recommendations apply to adult patients diagnosed with newly diagnosed glioblastoma. RECOMMENDATION Level I: Bevacizumab in general is not recommended in the initial treatment of adult patients with newly diagnosed GBM. It continues to be strongly recommended that patients with newly diagnosed GBM be enrolled in properly designed clinical trials to assess the benefit of novel chemotherapeutic agents compared to standard therapy.
Collapse
Affiliation(s)
- Navid Redjal
- Department of Neurosurgery, Capital Health Institute for Neurosciences, Capital Health Institute for Neurosciences, Two Capital Way, Pennington, NJ, 08534, USA.
| | - Brian V Nahed
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Jorg Dietrich
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Steven N Kalkanis
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI, USA
| | - Jeffrey J Olson
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
22
|
Hsu PYH, Folkman F, Ghosh S, de Robles P, Leckie C, Dersch-Mills D, Coppens R, Chambers C. Actual body weight dosing of temozolomide and overall survival in patients with glioblastoma. J Oncol Pharm Pract 2020; 27:1723-1729. [PMID: 33121351 DOI: 10.1177/1078155220968613] [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: 11/15/2022]
Abstract
BACKGROUND Adult glioblastoma patients receiving standard radiation therapy and concurrent temozolomide chemotherapy have a median survival of 14.6 months. Based on the pivotal trial data by Stupp et al., temozolomide doses were calculated based on body surface area. However, no details regarding the weight used to calculate body surface area was included in the study. As a result, temozolomide doses have been variable across the province. METHODS This retrospective chart review was conducted to determine the correlation between dose of first line temozolomide with overall survival. Patients between January 1st, 2009 and December 31st, 2014 who were newly diagnosed, pathology confirmed glioblastoma treated first line with temozolomide within Alberta Health Services were included in the study. Temozolomide doses above and below determined cut points were compared through the Kaplan-Meier method, then assessed using the log-rank test. RESULTS A cut point of 97.8% of actual body weight calculated body surface area dosing was determined for concurrent phase temozolomide. At doses above this cut point, there was a statistically significant (p = 0.0158) increase of 0.3 years in median overall survival. As for toxicity concerns, there was a statistically significant increase in the proportion of temozolomide dose reductions due to toxicity in patients dosed above the cut point. CONCLUSION Temozolomide doses at full actual body weight calculated body surface area dosing during the concurrent phase is required to achieve a similar median OS as seen in the pivotal trial by Stupp et al.
Collapse
|
23
|
Pu JK, Kwong DL. Central Nervous System Neoplasms in Hong Kong: An Inscription of Local Studies. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666190126153006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
A registry of brain and central nervous system (CNS) tumor patients in Hong Kong
comprising of data from both public and private neurosurgical practices (with approximately 98%
patients of Chinese origin), suggested geographical or racial variations in disease incidence. The
data confers the finding of a comparatively lower incidence rate of meningioma and malignant
gliomas as in other parts of Southeast Asia.
:
With data suggesting epidemiological difference, the treatment response, particularly in highgrade
glioma, was studied. Patients suffering from glioblastoma (GBM) in Hong Kong received
the standard of care, which involves safe, maximal resection followed by the Stupp regime.
5-aminolevulinic acid (5-ALA)-based fluorescence-guided surgery was found to be feasible and
safe to adopt in the treatment of local WHO Grade III & IV gliomas patients. Survival benefit was
seen in a group of patients using extended adjuvant temozolomide (TMZ) treatment for newly
diagnosed GBM as compared to those treated with the standard 6 cycles. Salvage therapies with
either single agent bevacizumab or bevacizumab plus irinotecan appeared to be effective treatment
options in Hong Kong patients with recurrent malignant glioma, with a good associated 6-
month progression-free survival (PFS) rate which was comparable to previously published overseas
data in this disease type in the same overall population.
Collapse
Affiliation(s)
- Jenny K.S. Pu
- Department of Neurosurgery, Queen Mary Hospital, Hong Kong, China
| | - Dora L.W. Kwong
- Department of Clinical Oncology, Queen Mary Hospital, University of Hong Kong, Hong Kong, China
| |
Collapse
|
24
|
Garcia-Fabiani MB, Ventosa M, Comba A, Candolfi M, Nicola Candia AJ, Alghamri MS, Kadiyala P, Carney S, Faisal SM, Schwendeman A, Moon JJ, Scheetz L, Lahann J, Mauser A, Lowenstein PR, Castro MG. Immunotherapy for gliomas: shedding light on progress in preclinical and clinical development. Expert Opin Investig Drugs 2020; 29:659-684. [PMID: 32400216 DOI: 10.1080/13543784.2020.1768528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Gliomas are infiltrating brain tumors associated with high morbidity and mortality. Current standard of care includes radiation, chemotherapy, and surgical resection. Today, survival rates for malignant glioma patients remain dismal and unchanged for decades. The glioma microenvironment is highly immunosuppressive and consequently this has motivated the development of immunotherapies for counteracting this condition, enabling the immune cells within the tumor microenvironment to react against this tumor. AREAS COVERED The authors discuss immunotherapeutic strategies for glioma in phase-I/II clinical trials and illuminate their mechanisms of action, limitations, and key challenges. They also examine promising approaches under preclinical development. EXPERT OPINION In the last decade there has been an expansion in immune-mediated anti-cancer therapies. In the glioma field, sophisticated strategies have been successfully implemented in preclinical models. Unfortunately, clinical trials have not yet yielded consistent results for glioma patients. This could be attributed to our limited understanding of the complex immune cell infiltration and its interaction with the tumor cells, the selected time for treatment, the combination with other therapies and the route of administration of the agent. Applying these modalities to treat malignant glioma is challenging, but many new alternatives are emerging to by-pass these hurdles.
Collapse
Affiliation(s)
- Maria B Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Maria Ventosa
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Alejandro J Nicola Candia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Stephen Carney
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Cancer Biology Graduate Program, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Lindsay Scheetz
- Department of Pharmaceutical Sciences, University of Michigan , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - Joerg Lahann
- Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Chemical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Ava Mauser
- Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA.,Department of Chemical Engineering, University of Michigan , Ann Arbor, MI, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School , Ann Arbor, MI, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School , Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
25
|
Zhang X, Zhang W, Mao XG, Cao WD, Zhen HN, Hu SJ. Malignant Intracranial High Grade Glioma and Current Treatment Strategy. Curr Cancer Drug Targets 2020; 19:101-108. [PMID: 29848277 DOI: 10.2174/1568009618666180530090922] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 12/06/2017] [Accepted: 12/19/2017] [Indexed: 12/17/2022]
Abstract
Malignant high-grade glioma (HGG) is the most common and extremely fatal type of primary intracranial tumor. These tumors recurred within 2 to 3 cm of the primary region of tumor resection in the majority of cases. Furthermore, the blood-brain barrier significantly limited the access of many systemically administered chemotherapeutics to the tumor, pointing towards a stringent need for new therapeutic patterns. Therefore, targeting therapy using local drug delivery for HGG becomes a priority for the development of novel therapeutic strategies. The main objectives to the effective use of chemotherapy for HGG include the drug delivery to the tumor region and the infusion of chemotherapeutic agents into the vascular supply of a tumor directly, which could improve the pharmacokinetic profile by enhancing drug delivery to the neoplasm tissue. Herein, we reviewed clinical and molecular features, different methods of chemotherapy application in HGGs, especially the existing and promising targeting therapies using local drug delivery for HGG which could effectively inhibit tumor invasion, proliferation and recurrence of HGG to combat the deadly disease. Undoubtedly, novel chemical medicines targeting these HGG may represent one of the most important directions in the Neuro-oncology.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xing-Gang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Wei-Dong Cao
- Department of Neurosurgery, Navy General Hospital, PLA, Beijing, 100048, China
| | - Hai-Ning Zhen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shi-Jie Hu
- Department of Neuro-oncology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
26
|
Quan R, Zhang H, Li Z, Li X. Survival analysis of patients with glioblastoma treated by long-term administration of temozolomide. Medicine (Baltimore) 2020; 99:e18591. [PMID: 31914038 PMCID: PMC6959873 DOI: 10.1097/md.0000000000018591] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This analysis aimed to investigate whether the long-term administration of temozolomide (TMZ) claimed a survival advantage for patients with glioblastoma in China.A total of 75 patients with newly diagnosed glioblastoma at the Department of Radiation Oncology, Shenzhen People's Hospital between August 2008 and August 2016 were retrospectively evaluated during analysis. A propensity-matched analysis was performed to balance the basic characteristics of patients between compared groups. Kaplan-Meier method and Cox proportional hazards model were used to assess progression-free survival (PFS) and overall survival (OS) of patients receiving 6 adjuvant TMZ cycles compared with patients treated with more than 6 cycles.Twenty of 75 patients received more than 6 cycles of TMZ, and the other 55 patients were treated with a median of 6 cycles ranging from 1 to 6. The patients with long-term administration of TMZ had better OS (47.0 months, 95% CI 20.0-73.9 vs 20.6 months, 95% CI 17.9-23.2, P = .014) but not PFS (17.0 months, 95% CI 10.1-24.5 vs 14.2 months, 95% CI 11.8-16.6, P = .133). Balancing the clinical factors with a propensity-matched analysis also showed the significant advantage of prolonged TMZ application in terms of OS but not PFS.Prolonged administration of TMZ beyond 6 cycles did demonstrate survival benefits for patients with initially diagnosed glioblastoma.
Collapse
Affiliation(s)
- Rencui Quan
- Department of Radiation Oncology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital
| | - Huaqing Zhang
- Department of Endocrinology, Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Zihuang Li
- Department of Radiation Oncology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital
| | - Xianming Li
- Department of Radiation Oncology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital
| |
Collapse
|
27
|
Feasibility study of finalizing the extended adjuvant temozolomide based on methionine positron emission tomography (Met-PET) findings in patients with glioblastoma. Sci Rep 2019; 9:17794. [PMID: 31780768 PMCID: PMC6883069 DOI: 10.1038/s41598-019-54398-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/14/2019] [Indexed: 01/22/2023] Open
Abstract
In the management of patients with newly diagnosed glioblastoma, there is no standard duration for adjuvant temozolomide treatment. This study aimed to assess the feasibility of finalizing adjuvant temozolomide treatment on the basis of methionine uptake in methionine positron emission tomography (Met-PET). We conducted a retrospective review of glioblastoma patients who underwent more than twelve cycles of temozolomide (extended temozolomide) treatment after resection and concomitant chemoradiotherapy with no evidence of recurrence on MRI. In addition to the methionine uptake value at the completion of extended temozolomide, local and distant recurrence and progression-free survival were also analyzed. Forty-four patients completed the extended temozolomide treatment. Among these, 18 experienced some type of tumor recurrence within one year. A Tmax/Nave value of 2.0 was the optimal cut-off value indicating progression. More than 80% of the patients with low methionine uptake completed the temozolomide treatment, and subsequent basic MRI observations showed no recurrence within one year after Met-PET. Subgroups with high uptake (≥2.0), even with continuation of temozolomide treatment, showed more frequent tumor progression than patients with low uptake (<2.0) who completed the extended temozolomide treatment (p < 0.001, odds ratio 14.7, 95% CI 3.46-62.3). The tumor recurrence rate increased in stepwise manner according to methionine uptake. Finalization of the extended temozolomide treatment on the basis of low uptake value was feasible with a low recurrence rate. Compared to MRI, Met-PET shows better ability to predict tumor progression in long-term glioblastoma survivors with extended temozolomide use.
Collapse
|
28
|
Giles BM, Underwood TT, Benhadji KA, Nelson DKS, Grobeck LM, Lin B, Wang S, Fill JA, Man M, Pitts KR, Bamberg A. Analytical Characterization of an Enzyme-Linked Immunosorbent Assay for the Measurement of Transforming Growth Factor β1 in Human Plasma. J Appl Lab Med 2019; 3:200-212. [DOI: 10.1373/jalm.2017.025619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/26/2018] [Indexed: 01/28/2023]
Abstract
Abstract
Background
The transforming growth factor β (TGF-β)–signaling pathway has emerged as a promising therapeutic target for many disease states including hepatocellular carcinoma (HCC). Because of the pleiotropic effects of this pathway, patient selection and monitoring may be important. TGF-β1 is the most prevalent isoform, and an assay to measure plasma levels of TGF-β1 would provide a rational biomarker to assist with patient selection. Therefore, the objective of this study was to analytically validate a colorimetric ELISA for the quantification of TGF-β1 in human plasma.
Methods
A colorimetric sandwich ELISA for TGF-β1 was analytically validated per Clinical and Laboratory Standards Institute protocols by assessment of precision, linearity, interfering substances, and stability. A reference range for plasma TGF-β1 was established for apparently healthy individuals and potential applicability was demonstrated in HCC patients.
Results
Precision was assessed for samples ranging from 633 to 10822 pg/mL, with total variance ranging from 28.4% to 7.2%. The assay was linear across the entire measuring range, and no interference of common blood components or similar molecules was observed. For apparently healthy individuals, the average TGF-β1 level was 1985 ± 1488 pg/mL compared to 4243 ± 2003 pg/mL for HCC patients. Additionally, the TGF-β1 level in plasma samples was demonstrated to be stable across all conditions tested, including multiple freeze–thaw cycles.
Conclusions
The ELISA described in this report is suitable for the quantification of TGF-β1 in human plasma and for investigational use in an approved clinical study.
Collapse
Affiliation(s)
| | | | | | | | | | - Boris Lin
- Lilly Research Laboratories, Indianapolis, IN
| | | | | | - Michael Man
- Lilly Research Laboratories, Indianapolis, IN
| | | | | |
Collapse
|
29
|
Fabbro-Peray P, Zouaoui S, Darlix A, Fabbro M, Pallud J, Rigau V, Mathieu-Daude H, Bessaoud F, Bauchet F, Riondel A, Sorbets E, Charissoux M, Amelot A, Mandonnet E, Figarella-Branger D, Duffau H, Tretarre B, Taillandier L, Bauchet L. Association of patterns of care, prognostic factors, and use of radiotherapy-temozolomide therapy with survival in patients with newly diagnosed glioblastoma: a French national population-based study. J Neurooncol 2018; 142:91-101. [PMID: 30523606 PMCID: PMC6399437 DOI: 10.1007/s11060-018-03065-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/27/2018] [Indexed: 01/11/2023]
Abstract
Background Glioblastoma is the most frequent primary malignant brain tumor. In daily practice and at whole country level, oncological care management for glioblastoma patients is not completely known. Objectives To describe oncological patterns of care, prognostic factors, and survival for all patients in France with newly-diagnosed and histologically confirmed glioblastoma, and evaluate the impact of extended temozolomide use at the population level. Methods Nationwide population-based cohort study including all patients with newly-diagnosed and histologically confirmed glioblastoma in France in 2008 and followed until 2015. Results Data from 2053 glioblastoma patients were analyzed (male/female ratio 1.5, median age 64 years). Median overall survival (OS) was 11.2 [95% confidence interval (CI) 10.7–11.9] months. The first-line therapy and corresponding median survival (MS, in months) were: 13% did not receive any oncological treatment (biopsy only) (MS = 1.8, 95% CI 1.6–2.1), 27% received treatment without the combination of radiotherapy (RT)–temozolomide (MS = 5.9, 95% CI 5.5–6.6), 60% received treatment including the initiation of the concomitant phase of RT–temozolomide (MS = 16.4, 95% CI 15.2–17.4) whom 44% of patients initiated the temozolomide adjuvant phase (MS = 18.9, 95% CI 18.0–19.8). Only 22% patients received 6 cycles or more of adjuvant temozolomide (MS = 25.5, 95% CI 24.0–28.3). The multivariate analysis showed that the risk of mortality was significantly higher for the non-progressive patients who stopped at 6 cycles (standard protocol) than those who continued the treatment, hazard ratio = 1.5 (95% CI 1.2–1.9). Conclusion In non-progressive patients, prolonging the adjuvant temozolomide beyond 6 cycles may improve OS. Electronic supplementary material The online version of this article (10.1007/s11060-018-03065-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pascale Fabbro-Peray
- Department of Biostatistics, Epidemiology, Public Health, CHU Nîmes, Nîmes, France.,EA2415 Research Unit, Montpellier University, Montpellier, France
| | - Sonia Zouaoui
- Department of Neurosurgery, Hopital Gui de Chauliac, CHU Montpellier, Montpellier University Medical Center, 80 Avenue Fliche, 34295, Montpellier, France.,INSERM U1051, Montpellier, France.,Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France
| | - Amélie Darlix
- Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France.,Department of Medical Oncology, ICM, Montpellier, France
| | - Michel Fabbro
- Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France.,Department of Medical Oncology, ICM, Montpellier, France
| | - Johan Pallud
- Department of Neurosurgery, Sainte Anne Hospital, and University Paris Descartes, Paris, France
| | - Valérie Rigau
- Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France.,Department of Neuropathology, Hopital Gui de Chauliac, CHU Montpellier, Montpellier University Medical Center, Montpellier, France
| | - Hélène Mathieu-Daude
- Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France.,Department of Medical informatics, ICM, Montpellier, France
| | - Faiza Bessaoud
- Registre des Tumeurs de l'Hérault, ICM, Montpellier, France
| | - Fabienne Bauchet
- Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France
| | - Adeline Riondel
- Department of Biostatistics, Epidemiology, Public Health, CHU Nîmes, Nîmes, France.,EA2415 Research Unit, Montpellier University, Montpellier, France
| | - Elodie Sorbets
- Department of Biostatistics, Epidemiology, Public Health, CHU Nîmes, Nîmes, France.,EA2415 Research Unit, Montpellier University, Montpellier, France
| | - Marie Charissoux
- Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France.,Department of Radiation Oncology, ICM, Montpellier, France
| | - Aymeric Amelot
- Department of Neurosurgery, CHU Lariboisière, Paris, France
| | | | | | - Hugues Duffau
- Department of Neurosurgery, Hopital Gui de Chauliac, CHU Montpellier, Montpellier University Medical Center, 80 Avenue Fliche, 34295, Montpellier, France.,INSERM U1051, Montpellier, France.,Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France
| | | | | | - Luc Bauchet
- Department of Neurosurgery, Hopital Gui de Chauliac, CHU Montpellier, Montpellier University Medical Center, 80 Avenue Fliche, 34295, Montpellier, France. .,INSERM U1051, Montpellier, France. .,Groupe de Neuro-Oncologie du Languedoc Roussillon, ICM, Montpellier, France.
| |
Collapse
|
30
|
Ghosh M, Shubham S, Mandal K, Trivedi V, Chauhan R, Naseera S. Survival and prognostic factors for glioblastoma multiforme: Retrospective single-institutional study. Indian J Cancer 2018; 54:362-367. [PMID: 29199724 DOI: 10.4103/ijc.ijc_157_17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults. The standard management has been maximum surgical resection followed by adjuvant radiotherapy with concurrent chemotherapy followed by adjuvant chemotherapy. Although the survival rate of patients with GBM has improved with recent advancements in treatment, the prognosis remains generally poor. The median survival rates are in the range of 9-12 months and 2-year survival rates are in the range of 8%-12%. MATERIALS AND METHODS A single-institution retrospective review of 61 patients of GBM from 2012 to 2014. Data regarding patient factors, disease factors, and treatment factors were collected and survival has been calculated. RESULTS A total of 61 patients with GBM were analyzed. GBM is commonly seen in sixth decade of life. Male to female ratio is 2.6:1. The right side of the brain is commonly involved with right frontal lobe being the most common site. The median follow-up was 4.6 months. The median survival of our patients was 8 months. The 1-year and 2-year survival rates were 20% and 3.27%, respectively. CONCLUSIONS The overall survival and prognosis in patients with GBM remains poor despite of constant research and studies. Concurrent chemoradiotherapy followed by adjuvant chemotherapy with temozolomide should be used after maximal resection to improve the survival.
Collapse
Affiliation(s)
- M Ghosh
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, Bihar, India
| | - S Shubham
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, Bihar, India
| | - K Mandal
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, Bihar, India
| | - V Trivedi
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, Bihar, India
| | - R Chauhan
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, Bihar, India
| | - S Naseera
- Department of Radiation Oncology, Mahavir Cancer Sansthan, Patna, Bihar, India
| |
Collapse
|
31
|
Bobustuc GC, Kassam AB, Rovin RA, Jeudy S, Smith JS, Isley B, Singh M, Paranjpe A, Srivenugopal KS, Konduri SD. MGMT inhibition in ER positive breast cancer leads to CDC2, TOP2A, AURKB, CDC20, KIF20A, Cyclin A2, Cyclin B2, Cyclin D1, ERα and Survivin inhibition and enhances response to temozolomide. Oncotarget 2018; 9:29727-29742. [PMID: 30038716 PMCID: PMC6049872 DOI: 10.18632/oncotarget.25696] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
The DNA damage repair enzyme, O6-methylguanine DNA methyltransferase (MGMT) is overexpressed in breast cancer, correlating directly with estrogen receptor (ER) expression and function. In ER negative breast cancer the MGMT promoter is frequently methylated. In ER positive breast cancer MGMT is upregulated and modulates ER function. Here, we evaluate MGMT's role in control of other clinically relevant targets involved in cell cycle regulation during breast cancer oncogenesis. We show that O6-benzylguanine (BG), an MGMT inhibitor decreases CDC2, CDC20, TOP2A, AURKB, KIF20A, cyclin B2, A2, D1, ERα and survivin and induces c-PARP and p21 and sensitizes ER positive breast cancer to temozolomide (TMZ). Further, siRNA inhibition of MGMT inhibits CDC2, TOP2A, AURKB, KIF20A, Cyclin B2, A2 and survivin and induces p21. Combination of BG+TMZ decreases CDC2, CDC20, TOP2A, AURKB, KIF20A, Cyclin A2, B2, D1, ERα and survivin. Temozolomide alone inhibits MGMT expression in a dose and time dependent manner and increases p21 and cytochrome c. Temozolomide inhibits transcription of TOP2A, AURKB, KIF20A and does not have any effect on CDC2 and CDC20 and induces p21. BG+/-TMZ inhibits breast cancer growth. In our orthotopic ER positive breast cancer xenografts, BG+/-TMZ decreases ki-67, CDC2, CDC20, TOP2A, AURKB and induces p21 expression. In the same model, BG+TMZ combination inhibits breast tumor growth in vivo compared to single agent (TMZ or BG) or control. Our results show that MGMT inhibition is relevant for inhibition of multiple downstream targets involved in tumorigenesis. We also show that MGMT inhibition increases ER positive breast cancer sensitivity to alkylator based chemotherapy.
Collapse
Affiliation(s)
- George C. Bobustuc
- Aurora Research Institute, Milwaukee, WI, USA
- Aurora Neurosciences Innovation Institute, Milwaukee, WI, USA
| | - Amin B. Kassam
- Aurora Research Institute, Milwaukee, WI, USA
- Aurora Neurosciences Innovation Institute, Milwaukee, WI, USA
| | - Richard A. Rovin
- Aurora Research Institute, Milwaukee, WI, USA
- Aurora Neurosciences Innovation Institute, Milwaukee, WI, USA
| | | | | | | | - Maharaj Singh
- Aurora Research Institute, Milwaukee, WI, USA
- Aurora Neurosciences Innovation Institute, Milwaukee, WI, USA
| | - Ameya Paranjpe
- Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | | | - Santhi D. Konduri
- Aurora Research Institute, Milwaukee, WI, USA
- Aurora Neurosciences Innovation Institute, Milwaukee, WI, USA
| |
Collapse
|
32
|
Foroughi S, Wong HL, Gately L, Lee M, Simons K, Tie J, Burgess AW, Gibbs P. Re-inventing the randomized controlled trial in medical oncology: The registry-based trial. Asia Pac J Clin Oncol 2018; 14:365-373. [PMID: 29947051 DOI: 10.1111/ajco.12992] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/29/2018] [Indexed: 01/09/2023]
Abstract
Substantial progress has recently been made in optimizing the management of cancer patients, resulting in major gains in survival and quality of life. Much of this progress has resulted from the serial testing of promising treatment strategies, typically using prospective randomized controlled trials to compare outcomes achieved with the new approach versus the current standard(s) of care. However, there is an ever-expanding list of important questions that are difficult to investigate, particularly with respect to determining the optimal sequencing and combination of proven active agents. With the rapidly growing list of clinical, pathologic and molecular characteristics that promise to predict treatment benefit and/or risk for defined patient subsets, many new questions regarding how best to personalize our approach to treatment selection are emerging. These questions can be investigated in the context of registry-based randomized clinical trials. Recently, the potential of registry-based randomized clinical trials was demonstrated in cardiology, highlighting the ability to rapidly recruit large numbers of patients to a trial addressing an important clinical question, with minimal cost and high external validity. In this review, we discuss the challenges and limitations of conventional clinical trials in multidisciplinary cancer care, describe the potential advantages of registry-based randomized trials, and highlight several registry-based oncology studies that are already underway to demonstrate the feasibility of this approach.
Collapse
Affiliation(s)
- Siavash Foroughi
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Hui-Li Wong
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Lucy Gately
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Margaret Lee
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Eastern Health, Box Hill, Victoria, Australia.,Department of Medical Oncology, Western Health, St Albans, Victoria, Australia
| | - Koen Simons
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia.,Western Centre for Health, Research and Education, Western Health, St Albans, Victoria, Australia
| | - Jeanne Tie
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Western Health, St Albans, Victoria, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Antony Wilks Burgess
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia.,Structural Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Peter Gibbs
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Medical Oncology, Western Health, St Albans, Victoria, Australia
| |
Collapse
|
33
|
Wang HC, Lin YT, Lin WC, Ho RW, Lin YJ, Tsai NW, Ho JT, Lu CH. Tumor Volume Changes During and After Temozolomide Treatment for Newly Diagnosed Higher-Grade Glioma (III and IV). World Neurosurg 2018; 114:e766-e774. [DOI: 10.1016/j.wneu.2018.03.078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/09/2018] [Indexed: 11/28/2022]
|
34
|
Bhandari M, Gandhi AK, Devnani B, Kumar P, Sharma DN, Julka PK. Comparative Study of Adjuvant Temozolomide Six Cycles Versus Extended 12 Cycles in Newly Diagnosed Glioblastoma Multiforme. J Clin Diagn Res 2017; 11:XC04-XC08. [PMID: 28658891 DOI: 10.7860/jcdr/2017/27611.9945] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/17/2017] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Studies have shown promising survival with the use of Extended Temozolomide (E-TMZ) as compared to Conventional six cycles of Temozolomide (C-TMZ) in malignant gliomas; however, the reports are mostly limited to retrospective studies with significant bias. AIM This study assesses the impact of six versus 12 cycles of adjuvant Temozolomide (TMZ) on Overall Survival (OS) in newly diagnosed postoperative patients of Glioblastoma Multiforme (GBM). MATERIALS AND METHODS Between January 2012 and July 2013, 40 postoperative patients of GBM between age 18-65 years and Karnofsky Performance Score (KPS) ≥70 were included. Patients were randomized to receive radiation (60 Gray in 30 fractions over six weeks) with concomitant TMZ (75 mg/m2/day) and adjuvant therapy with either six (C-TMZ arm) or 12 cycles (E-TMZ arm) of TMZ (150-200 mg/m2 for five days, repeated four weekly). Twenty patients were treated in each arm. Toxicity was assessed using Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. OS and Progression Free Survival (PFS) were calculated from the time of diagnosis. Kaplan Meier method was used for survival analysis. A p-value of <0.05 was taken as significant and SPSS version 12.0 was used for all statistical analysis. RESULTS Median number of adjuvant TMZ cycles was six and 12 in C-TMZ and E-TMZ arm respectively. Overall, 5% and 15% patients respectively in C-TMZ and E-TMZ arm had haematological toxicity ≥ 3 in grade. Median follow up in C-TMZ and E-TMZ arm were 14.65 months and 19.85 months. Median PFS was 12.8 months and 16.8 months in C-TMZ and E-TMZ arm respectively (p=0.069). Median OS was 15.4 months vs. 23.8 months in C-TMZ and E-TMZ arm respectively (p=0.044). CONCLUSION Our study showed that E-TMZ is well tolerated and leads to a significant increase in PFS as well as OS in newly diagnosed patients of GBM. Further prospective randomized studies are needed to validate the findings of our study.
Collapse
Affiliation(s)
- Menal Bhandari
- Senior Resident, Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Ajeet Kumar Gandhi
- Assistant Professor, Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Bharti Devnani
- Senior Resident, Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Pavnesh Kumar
- Senior Resident, Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Daya Nand Sharma
- Professor, Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Pramod Kumar Julka
- Professor, Department of Radiation Oncology, All India Institute of Medical Sciences, New Delhi, Delhi, India
| |
Collapse
|
35
|
Skardelly M, Dangel E, Gohde J, Noell S, Behling F, Lepski G, Borchers C, Koch M, Schittenhelm J, Bisdas S, Naumann A, Paulsen F, Zips D, von Hehn U, Ritz R, Tatagiba MS, Tabatabai G. Prolonged Temozolomide Maintenance Therapy in Newly Diagnosed Glioblastoma. Oncologist 2017; 22:570-575. [PMID: 28360216 PMCID: PMC5423504 DOI: 10.1634/theoncologist.2016-0347] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/12/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The impact of prolonging temozolomide (TMZ) maintenance beyond six cycles in newly diagnosed glioblastoma (GBM) remains a topic of discussion. We investigated the effects of prolonged TMZ maintenance on progression-free survival (PFS) and overall survival (OS). PATIENTS AND METHODS In this retrospective single-center cohort study, we included patients with GBM who were treated with radiation therapy with concomitant and adjuvant TMZ. For analysis, patients were considered who either completed six TMZ maintenance cycles (group B), continued with TMZ therapy beyond six cycles (group C), or stopped TMZ maintenance therapy within the first six cycles (group A). Patients with progression during the first six TMZ maintenance cycles were excluded. RESULTS Clinical data from 107 patients were included for Kaplan-Meier analyses and 102 for Cox regressions. Median PFS times were 8.1 months (95% confidence interval [CI] 6.1-12.4) in group A, 13.7 months (95% CI 10.6-17.5) in group B, and 20.9 months (95% CI 15.2-43.5) in group C. At first progression, response rates of TMZ/lomustine rechallenge were 47% in group B and 13% in group C. Median OS times were 12.7 months (95% CI 10.3-16.8) in group A, 25.2 months (95% CI 17.7-55.5) in group B, and 28.6 months (95% CI 24.4-open) in group C. Nevertheless, multivariate Cox regression for patients in group C compared with group B that accounted for imbalances of other risk factors showed no different relative risk (RR) for OS (RR 0.77, p = .46). CONCLUSION Our data do not support a general extension of TMZ maintenance therapy beyond six cycles. The Oncologist 2017;22:570-575 IMPLICATIONS FOR PRACTICE: Radiation therapy with concomitant and adjuvant temozolomide (TMZ) maintenance therapy is still the standard of care in patients below the age of 65 years in newly diagnosed glioblastoma. However, in clinical practice, many centers continue TMZ maintenance therapy beyond six cycles. The impact of this continuation is controversial and has not yet been addressed in prospective randomized clinical trials. We compared the effect of more than six cycles of TMZ in comparison with exactly six cycles on overall survival (OS) and progression-free survival (PFS) by multivariate analysis and found a benefit in PFS but not OS. Thus, our data do not suggest prolonging TMZ maintenance therapy beyond six cycles, which should be considered in neurooncological practice.
Collapse
Affiliation(s)
- Marco Skardelly
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Elena Dangel
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Julia Gohde
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Susan Noell
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Felix Behling
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Guilherme Lepski
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christian Borchers
- Department of Vascular Neurology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Marilin Koch
- Department of Vascular Neurology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jens Schittenhelm
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Vascular Neurology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Institute of Pathology and Neuropathology, Division of Neuropathology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sotirios Bisdas
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Vascular Neurology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Neuroradiology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Aline Naumann
- Department of Institute of Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
- Center for Personalized Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | | | - Rainer Ritz
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Marcos Soares Tatagiba
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
| | - Ghazaleh Tabatabai
- Department of Neurosurgery, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Vascular Neurology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Interdisciplinary Division of Neuro-Oncology, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Center for CNS Tumors, Comprehensive Cancer Center Tübingen Stuttgart, University Hospital Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Tübingen, Germany
- Center for Personalized Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
36
|
Efficacy and safety of long-term therapy for high-grade glioma with temozolomide: A meta-analysis. Oncotarget 2017; 8:51758-51765. [PMID: 28881684 PMCID: PMC5584285 DOI: 10.18632/oncotarget.17401] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/04/2017] [Indexed: 01/23/2023] Open
Abstract
Further treatments are warranted in preventing recurrence or progression for high-grade glioma (HGG) patients having achieved stable disease with tolerable toxicity after the Stupp regimen (6 cycles of temozolomide). This meta-analysis aims to extensively evaluate the safety, feasibility, and efficacy of long-term therapy with temozolomide (>6 cycles) for these patients. We systematically searched the pubmed, Embase and Chinese Biomedical (CBM) databases using the strategy of combination of free-text words and MeSH terms. The efficacy indicators are hazard ratio (HR) for the pooled analysis of overall survival (OS) and progression free survival (PFS). The safety indicator is risk ratio (RR) for the pooled analysis of adverse effects. Six studies comprising a total number of 396 patients met all inclusion and exclusion criteria were included. No heterogeneity and publication bias were observed across each study. It was found that patients could obtain benefits from long-term administration of temozolomide both in OS (HR 2.39, 95% CI 1.82–3.14) and PFS (HR 2.12, 95% CI 1.56–2.89). In addition, the results showed that the patients receiving long-term administration of temozolomide did not experience additional toxicity over that of the Stupp regimen (6 cycles of temozolomide). It could be concluded that it's efficacious and safe for HGG patients to receive long-term therapy with temozolomide. Nevertheless, more randomized controlled trials (RCTs) should be carried out to verify this conclusion.
Collapse
|
37
|
Gramatzki D, Kickingereder P, Hentschel B, Felsberg J, Herrlinger U, Schackert G, Tonn JC, Westphal M, Sabel M, Schlegel U, Wick W, Pietsch T, Reifenberger G, Loeffler M, Bendszus M, Weller M. Limited role for extended maintenance temozolomide for newly diagnosed glioblastoma. Neurology 2017; 88:1422-1430. [PMID: 28298550 DOI: 10.1212/wnl.0000000000003809] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 01/18/2017] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To explore an association with survival of modifying the current standard of care for patients with newly diagnosed glioblastoma of surgery followed by radiotherapy plus concurrent and 6 cycles of maintenance temozolomide chemotherapy (TMZ/RT → TMZ) by extending TMZ beyond 6 cycles. METHODS The German Glioma Network cohort was screened for patients with newly diagnosed glioblastoma who received TMZ/RT → TMZ and completed ≥6 cycles of maintenance chemotherapy without progression. Associations of clinical patient characteristics, molecular markers, and residual tumor determined by magnetic resonance imaging after 6 cycles of TMZ with progression-free survival (PFS) and overall survival (OS) were analyzed with the log-rank test. Multivariate analyses using the Cox proportional hazards model were performed to assess associations of prolonged TMZ use with outcome. RESULTS Sixty-one of 142 identified patients received at least 7 maintenance TMZ cycles (median 11, range 7-20). Patients with extended maintenance TMZ treatment had better PFS (20.5 months, 95% confidence interval [CI] 17.7-23.3, vs 17.2 months, 95% CI 10.2-24.2, p = 0.035) but not OS (32.6 months, 95% CI 28.9-36.4, vs 33.2 months, 95% CI 25.3-41.0, p = 0.126). However, there was no significant association of prolonged TMZ chemotherapy with PFS (hazard ratio [HR] = 0.8, 95% CI 0.4-1.6, p = 0.559) or OS (HR = 1.6, 95% CI 0.8-3.3, p = 0.218) adjusted for age, extent of resection, Karnofsky performance score, presence of residual tumor, O6-methylguanine DNA methyltransferase (MGMT) promoter methylation status, or isocitrate dehydrogenase (IDH) mutation status. CONCLUSION These data may not support the practice of prolonging maintenance TMZ chemotherapy beyond 6 cycles. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that in patients with newly diagnosed glioblastoma, prolonged TMZ chemotherapy does not significantly increase PFS or OS.
Collapse
Affiliation(s)
- Dorothee Gramatzki
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany.
| | - Philipp Kickingereder
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Bettina Hentschel
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Jörg Felsberg
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Ulrich Herrlinger
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Gabriele Schackert
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Jörg-Christian Tonn
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Manfred Westphal
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Michael Sabel
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Uwe Schlegel
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Wick
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Torsten Pietsch
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Guido Reifenberger
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Markus Loeffler
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Martin Bendszus
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| | - Michael Weller
- From the Department of Neurology and Brain Tumor Center (D.G., M.W.), University Hospital Zurich and University of Zurich, Switzerland; Department of Neuroradiology (P.K., M.B.), University Hospital of Heidelberg; Institute for Medical Informatics, Statistics and Epidemiology (B.H., M.L.), University of Leipzig; Departments of Neuropathology (J.F., G.R.) and Neurosurgery (M.S.), Heinrich-Heine University Düsseldorf; Division of Neurooncology (U.H.), Department of Neurology, University Medical Center Bonn; Department of Neurosurgery (G.S.), University of Dresden; Department of Neurosurgery (J.-C.T.), Ludwig Maximilians University Munich; Department of Neurosurgery (M.W.), University Medical Center Hamburg-Eppendorf, Hamburg; Department of Neurology (U.S.), Knappschaftskrankenhaus Bochum-Langendreer, Ruhr-University Bochum; Clinical Cooperation Unit Neurooncology (W.W.), German Cancer Consortium, German Cancer Research Center, and Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg; Department of Neuropathology (T.P.), DGNN Brain Tumor Reference Center, University of Bonn Medical School; and German Cancer Consortium (G.R.), German Cancer Research Center Heidelberg, Partner Site Essen/Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
38
|
Kim SK, Kim TM, Lee ST, Lee SH, Heo DS, Kim IH, Kim DG, Jung HW, Choi SH, Lee SH, Park CK. The survival significance of a measurable enhancing lesion after completing standard treatment for newly diagnosed glioblastoma. J Clin Neurosci 2016; 34:145-150. [PMID: 27475318 DOI: 10.1016/j.jocn.2016.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/18/2016] [Accepted: 06/04/2016] [Indexed: 11/17/2022]
Abstract
The goal of this study was to analyze the survival outcome according to the treatment response after completing standard treatment protocol for newly diagnosed glioblastoma (GBM) and to suggest a patient who should be considered for further treatment. After approving by our Institutional Review Board, 57 patients (38 male, 19 female; median age, 52years; age range, 16-81years) with newly diagnosed GBM who completed standard treatment protocol were examined retrospectively. According to the treatment response using the RANO criteria, there were 20 patients with complete response (CR), five patients with partial response (PR), 13 patients with stable disease (SD) and 19 patients with progressive disease (PD) after the completion of standard treatment. Patients (PR+SD+PD) with a measurable enhancing lesion were categorized the MEL group (n=37). We analyzed the difference of survival outcome between CR group and MEL group. The median progression-free survival (PFS) in the CR group was significantly better than that of the MEL group (18.0months vs. 3.0months, p=0.004). The median overall survival (OS) was also significantly longer in the CR group (25.0months vs. 15.0months, p=0.005). However, there was no significant difference in the survival outcome of the CR group compared with that of the subset of MEL group patients who showed PR or SD. Poor survival outcome was found only in MEL group patients who exhibited progression. Patients with a measurable enhancing lesion showing progression after completion of standard treatment protocol are appropriate candidates for further treatment.
Collapse
Affiliation(s)
- Sung Kwon Kim
- Department of Neurosurgery, Gyeongsang National University School of Medicine, Changwon Gyeongsang National University Hospital, Changwon, South Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Se-Hoon Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Dae Seog Heo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Il Han Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong Gyu Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Hee-Won Jung
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Seung Hong Choi
- Department of Radiology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Sang Hyung Lee
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea; Department of Neurosurgery, Seoul National University Boramae Medical Center, Seoul, South Korea.
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, 103 Daehak-ro, Jongno-gu, Seoul 03080, South Korea; Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea.
| |
Collapse
|
39
|
Rivoirard R, Falk A, Chargari C, Guy JB, Mery B, Nuti C, Peoc'h M, Forest F, Garin C, Adjabi A, Hoarau D, Kawaye S, Almokhles H, Fournel P, Magné N. Long-term Results of a Survey of Prolonged Adjuvant Treatment with Temozolomide in Patients with Glioblastoma (SV3 Study). Clin Oncol (R Coll Radiol) 2015; 27:486-7. [DOI: 10.1016/j.clon.2015.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 04/14/2015] [Indexed: 10/23/2022]
|
40
|
Liu Y, Hao S, Yu L, Gao Z. Long-term temozolomide might be an optimal choice for patient with multifocal glioblastoma, especially with deep-seated structure involvement: a case report and literature review. World J Surg Oncol 2015; 13:142. [PMID: 25889578 PMCID: PMC4393596 DOI: 10.1186/s12957-015-0558-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/23/2015] [Indexed: 11/10/2022] Open
Abstract
Background Multifocal glioblastoma is an uncommon and refractory subtype of high-grade glioma since the burden of masses could not be eliminated simply by operation, and it is getting even harder to control if some deep structures, like thalamus and pineal region, are involved. Case presentation Here we report a case of a 30-year-old male with multifocal glioblastoma affected his right thalamus, left lateral ventricle, and pineal region. Clinical manifestations include operation, concurrent radiochemotherapy, and a 12-cycle adjuvant temozolomide administration. The masses of this patient nearly disappeared after 15 months from the primary diagnosis, and no severe adverse event or neurological sequel occurred. Conclusions Long-term temozolomide might be an optimal choice for patients with multifocal glioblastoma, especially with deep-seated structure involvement.
Collapse
Affiliation(s)
- Yunpeng Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China. .,China National Clinical Research Center for Neurological Diseases, No. 6 Tiantan Xili, Dongcheng District, Beijing, China.
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China. .,China National Clinical Research Center for Neurological Diseases, No. 6 Tiantan Xili, Dongcheng District, Beijing, China.
| | - Lanbing Yu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China. .,China National Clinical Research Center for Neurological Diseases, No. 6 Tiantan Xili, Dongcheng District, Beijing, China.
| | - Zhixian Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China. .,China National Clinical Research Center for Neurological Diseases, No. 6 Tiantan Xili, Dongcheng District, Beijing, China.
| |
Collapse
|
41
|
Rhun EL, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int 2015; 6:S9-S44. [PMID: 25722939 PMCID: PMC4338495 DOI: 10.4103/2152-7806.151331] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/15/2014] [Indexed: 01/12/2023] Open
Abstract
High-grade glioma (HGG) are optimally treated with maximum safe surgery, followed by radiotherapy (RT) and/or systemic chemotherapy (CT). Recently, the treatment of newly diagnosed anaplastic glioma (AG) has changed, particularly in patients with 1p19q codeleted tumors. Results of trials currenlty ongoing are likely to determine the best standard of care for patients with noncodeleted AG tumors. Trials in AG illustrate the importance of molecular characterization, which are germane to both prognosis and treatment. In contrast, efforts to improve the current standard of care of newly diagnosed glioblastoma (GB) with, for example, the addition of bevacizumab (BEV), have been largely disappointing and furthermore molecular characterization has not changed therapy except in elderly patients. Novel approaches, such as vaccine-based immunotherapy, for newly diagnosed GB are currently being pursued in multiple clinical trials. Recurrent disease, an event inevitable in nearly all patients with HGG, continues to be a challenge. Both recurrent GB and AG are managed in similar manner and when feasible re-resection is often suggested notwithstanding limited data to suggest benefit from repeat surgery. Occassional patients may be candidates for re-irradiation but again there is a paucity of data to commend this therapy and only a minority of selected patients are eligible for this approach. Consequently systemic therapy continues to be the most often utilized treatment in recurrent HGG. Choice of therapy, however, varies and revolves around re-challenge with temozolomide (TMZ), use of a nitrosourea (most often lomustine; CCNU) or BEV, the most frequently used angiogenic inhibitor. Nevertheless, no clear standard recommendation regarding the prefered agent or combination of agents is avaliable. Prognosis after progression of a HGG remains poor, with an unmet need to improve therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, and Neurology, Department of Medical Oncology, Oscar Lambret Center, Lille, France, Inserm U-1192, Laboratoire de Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM), Lille 1 University, Villeneuve D’Ascq, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Department of Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
42
|
Refae AA, Ezzat A, Salem DA, Mahrous M. Protracted Adjuvant Temozolomide in Glioblastoma Multiforme. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/jct.2015.68082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
43
|
Gahramanov S, Varallyay C, Tyson RM, Lacy C, Fu R, Netto JP, Nasseri M, White T, Woltjer RL, Gultekin SH, Neuwelt EA. Diagnosis of pseudoprogression using MRI perfusion in patients with glioblastoma multiforme may predict improved survival. CNS Oncol 2014; 3:389-400. [PMID: 25438810 PMCID: PMC4590772 DOI: 10.2217/cns.14.42] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS This retrospective study determined the survival of glioblastoma patients with or without pseudoprogression. METHODS A total of 68 patients were included. Overall survival was compared between patients showing pseudoprogression (in most cases diagnosed using perfusion MRI with ferumoxytol) and in patients without pseudoprogession. MGMT methylation status was also analyzed in the pseudoprogression cases. RESULTS Median survival in 24 (35.3%) patients with pseudoprogression was 34.7 months (95% CI: 20.3-54.1), and 13.4 months (95% CI: 11.1-19.5) in 44 (64.7%) patients without pseudoprogression (p < 0.0001). The longest survival was a median of 54.1 months in patients with combination of pseudoprogression and (MGMT) promoter methylation. CONCLUSION Pseudoprogression is associated with better outcome, especially if concurring with MGMT promoter methylation. Patients never diagnosed with pseudoprogression had poor survival. This study emphasizes the importance of differentiating tumor progression and pseudoprogression using perfusion MRI.
Collapse
Affiliation(s)
- Seymur Gahramanov
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Department of Neurosurgery, University of New Mexico, Albuquerque, NM 87131, USA
| | - Csanad Varallyay
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Rose Marie Tyson
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Cynthia Lacy
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Rongwei Fu
- Department of Public Health & Preventative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Department of Emergency Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Joao Prola Netto
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Morad Nasseri
- Wayne State University, Department of Neurology, 4201 St Antoine, Detroit, MI 48201 USA
| | - Tricia White
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Randy L Woltjer
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Sakir Humayun Gultekin
- Department of Pathology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Edward A Neuwelt
- Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Department of Neurosurgery, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Department of Veterans Affairs Medical Center, 3710 SW US Veterans Hospital Road, Portland, OR 97239, USA
| |
Collapse
|
44
|
Lévy S, Chapet S, Mazeron JJ. [Management of gliomas]. Cancer Radiother 2014; 18:461-7. [PMID: 25201633 DOI: 10.1016/j.canrad.2014.07.147] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 07/14/2014] [Indexed: 01/28/2023]
Abstract
Gliomas are the most frequent primary brain tumors. Their care is difficult because of the proximity of organs at risk. The treatment of glioblastoma includes surgery followed by chemoradiation with the protocol of Stupp et al. The addition of bevacizumab allows an increase in progression-free survival by 4 months but it does not improve overall survival. This treatment is reserved for clinical trials. Intensity modulation radiotherapy may be useful to reduce the neurocognitive late effects in different types of gliomas. In elderly patients an accelerated radiotherapy 40 Gy in 15 fractions allows a similar survival to standard radiotherapy. O(6)-methylguanine-DNA methyltransferase (MGMT) status may help to choose between chemotherapy and radiotherapy. There is no standard for the treatment of recurrent gliomas. Re-irradiation in stereotactic conditions allows a median survival of 8 to 12.4 months. Anaplastic gliomas with 1p19q mutation have a greater sensibility to chemotherapy by procarbazine, lomustine and vincristine. Chemoradiotherapy in these patients has become the standard treatment. Many studies are underway testing targeted therapies, their place in the therapeutic management and new radiotherapy techniques.
Collapse
Affiliation(s)
- S Lévy
- Service de radiothérapie oncologique, centre Henry-Kaplan, université François-Rabelais, CHRU de Tours, 2, boulevard Tonnelé, 37000 Tours, France
| | - S Chapet
- Service de radiothérapie oncologique, centre Henry-Kaplan, université François-Rabelais, CHRU de Tours, 2, boulevard Tonnelé, 37000 Tours, France
| | - J-J Mazeron
- Service de radiothérapie oncologique, groupe hospitalier Pitié-Salpêtrière, 47-83, boulevard de l'Hôpital, 75651 Paris cedex, France; Université Paris VI, 75651 Paris cedex, France.
| |
Collapse
|
45
|
Benouaich-Amiel A, Catalaa I, Lubrano V, Cohen-Jonathan Moyal E, Uro-Coste E. Glioma di alto grado: astrocitoma anaplastico e glioblastoma. Neurologia 2014. [DOI: 10.1016/s1634-7072(14)67978-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
46
|
Ajaz M, Jefferies S, Brazil L, Watts C, Chalmers A. Current and investigational drug strategies for glioblastoma. Clin Oncol (R Coll Radiol) 2014; 26:419-30. [PMID: 24768122 DOI: 10.1016/j.clon.2014.03.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 03/27/2014] [Indexed: 11/21/2022]
Abstract
Medical treatments for glioblastoma face several challenges. Lipophilic alkylators remain the mainstay of treatment, emphasising the primacy of good blood-brain barrier penetration. Temozolomide has emerged as a major contributor to improved patient survival. The roles of procarbazine and vincristine in the procarbazine, lomustine and vincristine (PCV) schedule have attracted scrutiny and several lines of evidence now support the use of lomustine as effective single-agent therapy. Bevacizumab has had a convoluted development history, but clearly now has no major role in first-line treatment, and may even be detrimental to quality of life in this setting. In later disease, clinically meaningful benefits are achievable in some patients, but more impressively the combination of bevacizumab and lomustine shows early promise. Over the last decade, investigational strategies in glioblastoma have largely subscribed to the targeted kinase inhibitor paradigm and have mostly failed. Low prevalence dominant driver lesions such as the FGFR-TACC fusion may represent a niche role for this agent class. Immunological, metabolic and radiosensitising approaches are being pursued and offer more generalised efficacy. Finally, trial design is a crucial consideration. Progress in clinical glioblastoma research would be greatly facilitated by improved methodologies incorporating: (i) routine pharmacokinetic and pharmacodynamic assessments by preoperative dosing; and (ii) multi-stage, multi-arm protocols incorporating new therapy approaches and high-resolution biology in order to guide necessary improvements in science.
Collapse
Affiliation(s)
- M Ajaz
- Surrey Cancer Research Institute, University of Surrey, Guildford, UK.
| | - S Jefferies
- Oncology Centre, Addenbrooke's Hospital, Cambridge, UK
| | - L Brazil
- Guy's, St Thomas' and King's College Hospitals, London, UK
| | - C Watts
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - A Chalmers
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
47
|
Abstract
The influence of the microenvironment on tumour progression is becoming clearer. In this Review we address the role of an essential signalling pathway, that of transforming growth factor-β, in the regulation of components of the tumour microenvironment and how this contributes to tumour progression.
Collapse
Affiliation(s)
- Michael Pickup
- Vanderbilt University Medical Center, Vanderbilt-Ingram Comprehensive Cancer Center, Medicine and Pathology, Cancer Biology, 2220 Pierce Avenue, 691 Preston Research Building, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
48
|
Concurrent and adjuvant temozolomide-based chemoradiotherapy schedules for glioblastoma. Strahlenther Onkol 2013; 189:926-31. [DOI: 10.1007/s00066-013-0410-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/17/2013] [Indexed: 10/26/2022]
|
49
|
Balañá C, Vaz MA, Lopez D, de la Peñas R, García-Bueno JM, Molina-Garrido MJ, Sepúlveda JM, Cano JM, Bugés C, Sanz SM, Arranz JL, Perez-Segura P, Rodriguez A, Martin JM, Benavides M, Gil M. Should we continue temozolomide beyond six cycles in the adjuvant treatment of glioblastoma without an evidence of clinical benefit? A cost analysis based on prescribing patterns in Spain. Clin Transl Oncol 2013; 16:273-9. [PMID: 23793813 DOI: 10.1007/s12094-013-1068-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/05/2013] [Indexed: 01/06/2023]
Abstract
PURPOSE The standard adjuvant treatment for glioblastoma is temozolomide concomitant with radiotherapy, followed by a further six cycles of temozolomide. However, due to the lack of empirical evidence and international consensus regarding the optimal duration of temozolomide treatment, it is often extended to 12 or more cycles, even in the absence of residual disease. No clinical trial has shown clear evidence of clinical benefit of this extended treatment. We have explored the economic impact of this practice in Spain. MATERIALS AND METHODS Spanish neuro-oncologists completed a questionnaire on the clinical management of glioblastomas in their centers. Based on their responses and on available clinical and demographic data, we estimated the number of patients who receive more than six cycles of temozolomide and calculated the cost of this extended treatment. RESULTS Temozolomide treatment is continued for more than six cycles by 80.5 % of neuro-oncologists: 44.4 % only if there is residual disease; 27.8 % for 12 cycles even in the absence of residual disease; and 8.3 % until progression. Thus, 292 patients annually will continue treatment beyond six cycles in spite of a lack of clear evidence of clinical benefit. Temozolomide is covered by the National Health Insurance System, and the additional economic burden to society of this extended treatment is nearly 1.5 million euros a year. CONCLUSIONS The optimal duration of adjuvant temozolomide treatment merits investigation in a clinical trial due to the economic consequences of prolonged treatment without evidence of greater patient benefit.
Collapse
Affiliation(s)
- C Balañá
- Medical Oncology Service, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Carretera Canyet, s/n, 08916, Badalona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gupta T, Mohanty S, Moiyadi A, Jalali R. Factors predicting temozolomide induced clinically significant acute hematologic toxicity in patients with high-grade gliomas: a clinical audit. Clin Neurol Neurosurg 2013; 115:1814-9. [PMID: 23764039 DOI: 10.1016/j.clineuro.2013.05.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 04/30/2013] [Accepted: 05/15/2013] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Myelo-suppression, the dose-limiting toxicity of alkylating cytotoxic agents is generally perceived to be uncommon with temozolomide (TMZ), a novel oral second generation imidazotetrazinone prodrug, with a reported incidence of 5-10% of grade 3-4 acute hematologic toxicity. We were observing a higher incidence of clinically significant myelo-toxicity with the standard schedule of TMZ, particularly in females, prompting us to do a clinical audit in our patient population. METHODS One hundred two adults (>18 years of age) treated with TMZ either for newly diagnosed or recurrent/progressive high-grade glioma constituted the study cohort. Clinically significant acute hematologic toxicity was defined as any one or more of the following: any grade 3-4 hematologic toxicity; omission of daily TMZ dose for ≥ 3 consecutive days during concurrent phase; deferral of subsequently due TMZ cycle by ≥ 7 days during adjuvant phase; dose reduction or permanent discontinuation of TMZ; use of growth factors, platelets or packed-cell transfusions during the course of TMZ. Uni-variate and multi-variate analysis was performed to correlate incidence of acute hematologic toxicity with baseline patient, disease, and treatment characteristics. RESULTS The incidence of clinically significant neutropenia and thrombocytopenia was 7% and 12% respectively. Seven (7%) patients needed packed-cells, growth factors, and/or platelet transfusions. Grade 3-4 lymphopenia though common (32%) was self-limiting and largely asymptomatic. Two (2%) patients, both women succumbed to community acquired pneumonia during adjuvant TMZ. Multi-variate logistic regression analysis identified female gender, grade IV histology, baseline total leukocyte count <7700/mm(3) and baseline serum creatinine ≥1mg/dl as factors associated with significantly increased risk of clinically significant acute hematologic toxicity. CONCLUSION The incidence of TMZ induced clinically significant neutropenia and thrombocytopenia was low in our patient population. Severe lymphopenia though high was largely asymptomatic and self-limiting. Gender, grade, leukocyte count, and serum creatinine were significant independent predictors of severe acute myelo-toxicity.
Collapse
Affiliation(s)
- Tejpal Gupta
- Department of Radiation Oncology, Advanced Centre for Treatment Research & Education in Cancer and Tata Memorial Hospital Tata Memorial Centre, Mumbai, India.
| | | | | | | |
Collapse
|