1
|
Vaz-Salgado MA, Villamayor M, Albarrán V, Alía V, Sotoca P, Chamorro J, Rosero D, Barrill AM, Martín M, Fernandez E, Gutierrez JA, Rojas-Medina LM, Ley L. Recurrent Glioblastoma: A Review of the Treatment Options. Cancers (Basel) 2023; 15:4279. [PMID: 37686553 PMCID: PMC10487236 DOI: 10.3390/cancers15174279] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Glioblastoma is a disease with a poor prognosis. Multiple efforts have been made to improve the long-term outcome, but the 5-year survival rate is still 5-10%. Recurrence of the disease is the usual way of progression. In this situation, there is no standard treatment. Different treatment options can be considered. Among them would be reoperation or reirradiation. There are different studies that have assessed the impact on survival and the selection of patients who may benefit most from these strategies. Chemotherapy treatments have also been considered in several studies, mainly with alkylating agents, with data mostly from phase II studies. On the other hand, multiple studies have been carried out with target-directed treatments. Bevacizumab, a monoclonal antibody with anti-angiogenic activity, has demonstrated activity in several studies, and the FDA has approved it for this indication. Several other TKI drugs have been evaluated in this setting, but no clear benefit has been demonstrated. Immunotherapy treatments have been shown to be effective in other types of tumors, and several studies have evaluated their efficacy in this disease, both immune checkpoint inhibitors, oncolytic viruses, and vaccines. This paper reviews data from different studies that have evaluated the efficacy of different forms of relapsed glioblastoma.
Collapse
Affiliation(s)
- Maria Angeles Vaz-Salgado
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - María Villamayor
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Víctor Albarrán
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Víctor Alía
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Pilar Sotoca
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Jesús Chamorro
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Diana Rosero
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Ana M. Barrill
- Medical Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.V.); (V.A.); (V.A.); (P.S.); (J.C.); (D.R.); (A.M.B.)
| | - Mercedes Martín
- Radiotherapy Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.M.); (E.F.)
| | - Eva Fernandez
- Radiotherapy Oncology Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (M.M.); (E.F.)
| | - José Antonio Gutierrez
- Neurosurgery Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (J.A.G.); (L.M.R.-M.); (L.L.)
| | - Luis Mariano Rojas-Medina
- Neurosurgery Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (J.A.G.); (L.M.R.-M.); (L.L.)
| | - Luis Ley
- Neurosurgery Department, Ramon y Cajal University Hospital, 28034 Madrid, Spain; (J.A.G.); (L.M.R.-M.); (L.L.)
| |
Collapse
|
2
|
Pawlowski KD, Duffy JT, Babak MV, Balyasnikova IV. Modeling glioblastoma complexity with organoids for personalized treatments. Trends Mol Med 2023; 29:282-296. [PMID: 36805210 PMCID: PMC11101135 DOI: 10.1016/j.molmed.2023.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/23/2022] [Accepted: 01/12/2023] [Indexed: 02/17/2023]
Abstract
Glioblastoma (GBM) remains a fatal diagnosis despite the current standard of care of maximal surgical resection, radiation, and temozolomide (TMZ) therapy. One aspect that impedes drug development is the lack of an appropriate model representative of the complexity of patient tumors. Brain organoids derived from cell culture techniques provide a robust, easily manipulatable, and high-throughput model for GBM. In this review, we highlight recent progress in developing GBM organoids (GBOs) with a focus on generating the GBM microenvironment (i.e., stem cells, vasculature, and immune cells) recapitulating human disease. Finally, we also discuss the use of organoids as a screening tool in drug development for GBM.
Collapse
Affiliation(s)
- Kristen D Pawlowski
- Rush Medical College, Rush University Medical Center, Chicago, IL 60612, USA; Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Joseph T Duffy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong, SAR 999077, People's Republic of China.
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
3
|
Norden AD, Korytowsky B, You M, Kim Le T, Dastani H, Bobiak S, Singh P. A Real-World Claims Analysis of Costs and Patterns of Care in Treated Patients with Glioblastoma Multiforme in the United States. J Manag Care Spec Pharm 2019; 25:428-436. [PMID: 30917077 PMCID: PMC10398322 DOI: 10.18553/jmcp.2019.25.4.428] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Patients with glioblastoma multiforme (GBM) have a poor prognosis and high likelihood of recurrence. Routine care for incident cases in the United States involves surgical resection, followed by radiation therapy (RT) with concurrent and adjuvant temozolomide. Real-world data reporting the treatments and health care burden associated with GBM are limited. OBJECTIVE To assess patterns of care, health care resource utilization (HCRU), and costs associated with treatment of GBM in the United States. METHODS This study is a retrospective claims database analysis. Adult patients with a GBM diagnosis (index date) between January 1, 2010, and June 30, 2016, who had undergone brain surgery within 90 days of the index date, had received temozolomide and/or RT up to 90 days after index date, and had at least 6 months of continuous enrollment before the index date, were identified. Patients were excluded if they had (a) another primary cancer within 6 months pre-index, (b) secondary brain metastases, or (c) received temozolomide and/or RT pre-index. Baseline characteristics, treatments, HCRU, and costs were reported. First-line therapy began upon first receipt of RT and/or temozolomide after index date; second-line therapy began when a new drug was added > 28 days after initiation of first-line therapy or when there was a treatment gap > 90 days. Treatment regimens, duration of treatment (corrected group prognosis method), HCRU, and costs were reported descriptively in the 0- to 6-month and 7- to 12-month periods following initiation of first-line and second-line therapy. RESULTS Baseline characteristics were comparable between patients receiving temozolomide and/or RT. Patients receiving RT without chemotherapy tended to be older, be retired, and have more baseline comorbidities. Of the 4,071 patients receiving first-line therapy for GBM, most (73.0%) received temozolomide + RT; 24.4% received RT; and 2.5% received temozolomide monotherapy. Of those receiving first-line therapy, 1,283 (31.5%) patients subsequently received second-line therapy: 39.4% received bevacizumab monotherapy; 28.9% received bevacizumab combination therapy (temozolomide, 45.2% of patients; irinotecan, 24.3%; and temozolomide + lomustine, 15.4%); 15.5% received temozolomide monotherapy; and 13.7% received other systemic cancer therapies. The proportion of patients with hospitalizations increased from 2.9% (4-6 months pre-index) to 20.8% in the 3 months before the index date (likely due to diagnostic procedures) and 28.1% in the first 6 months after index (likely due to surgery) and then decreased to 13.3% in the 7- to 12-month period after index. Mean total per-patient costs at 6 and 12 months were $117,325 and $162,550 (first line) and $126,128 and $243,833 (second line). Costs in all time periods were largely driven by costs of RT/systemic cancer therapy. CONCLUSIONS Most patients with newly diagnosed GBM received treatment according to recommendations. However, relatively few patients received second-line therapy, and the HCRU burden and costs associated with both lines of therapy were substantial. Novel therapies for GBM are required to improve treatment options and outcomes in these patients. DISCLOSURES This study was funded by Bristol-Myers Squibb (Princeton Pike, NJ). Neither honoraria nor payments were provided for authorship. Norden received consultancy fees relating to this study from Bristol-Myers Squibb. Dastani, Korytowsky, Le, Singh, and You are employees of Bristol-Myers Squibb. Dastani and Korytowsky are shareholders of Bristol-Myers Squibb. Bobiak was an employee of Bristol-Myers Squibb at the time of this study. Preliminary data from this study were previously presented at the International Society for Pharmacoeconomics and Outcomes Research 22nd Annual International Meeting in Boston, MA, May 20-24, 2017.
Collapse
Affiliation(s)
- Andrew D Norden
- 1 Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts
| | | | - Min You
- 2 Bristol-Myers Squibb, Princeton, New Jersey
| | - T Kim Le
- 2 Bristol-Myers Squibb, Princeton, New Jersey
| | | | | | | |
Collapse
|
4
|
Multicenter, single arm, phase II trial on the efficacy of ortataxel in recurrent glioblastoma. J Neurooncol 2019; 142:455-462. [PMID: 30726533 DOI: 10.1007/s11060-019-03116-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/31/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE Glioblastoma (GBM) is the most aggressive and frequent subtype of all malignant gliomas. At the time of recurrence, therapeutic options are lacking. Ortataxel, a second-generation taxane was reported to be effective in pre-clinical and phase I clinical studies. The aim of this study was to evaluate a potential therapeutic activity of ortataxel in patients with GBM recurring after surgery and first line treatment. METHODS In this phase II study, according to a two stage design, adult patients with histologically confirmed GBM in recurrence after surgery or biopsy, standard radiotherapy and chemotherapy with temozolomide were considered eligible. Patients included were treated with ortataxel 75 mg/m2 i.v. every 3 weeks until disease progression. The primary objective of the study was to evaluate the activity of ortataxel in terms of progression free survival (PFS) at 6 months after the enrollment. PFS, overall survival at 9 months after the enrollment, objective response rate, compliance and safety were evaluated as secondary endpoints. RESULTS Between Nov 26, 2013 and Dec 12, 2015, 40 patients were recruited across six centres. The number of patients alive and free from progression at 6 months after the enrollment, observed in the first stage was four (11.4%), out of 35 patients included in the analysis, below the minimum number of events (7 out of 33) required to continue the study with the second stage The most important toxicities were neutropenia and hepatotoxicity that occurred in 13.2% of patients and leukopenia that occurred in 15.8% of patients. CONCLUSION Overall ortataxel treatment fail to demonstrate a significant activity in recurrent GBM patients. However in a limited number of patients the drug produced a benefit that lasted for a long time. TRIAL REGISTRATION This study is registered with ClinicalTrials.gov, number NCT01989884.
Collapse
|
5
|
Khasraw M, Lee A, McCowatt S, Kerestes Z, Buyse ME, Back M, Kichenadasse G, Ackland S, Wheeler H. Cilengitide with metronomic temozolomide, procarbazine, and standard radiotherapy in patients with glioblastoma and unmethylated MGMT gene promoter in ExCentric, an open-label phase II trial. J Neurooncol 2016; 128:163-171. [PMID: 26935578 DOI: 10.1007/s11060-016-2094-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
Newly diagnosed glioblastoma multiforme with unmethylated MGMT promoter has a poor prognosis, with a median survival of 12 months. This phase II study investigated the efficacy and safety of combining the selective integrin inhibitor cilengitide with a combination of metronomic temozolomide and procarbazine for these patients. Eligible patients (newly diagnosed, histologically confirmed supratentorial glioblastoma with unmethylated MGMT promoter) were entered into this multicentre study. Cilengitide (2000 mg IV twice weekly) was commenced 1 week prior to radiotherapy combined with daily temozolomide (60 mg/m(2)) and procarbazine (50 or 100 mg) and, after 4 weeks' break, followed by six adjuvant cycles of temozolomide (50-60 mg/m(2)) and procarbazine (50 or 100 mg) on days 1-20, every 28 days. Cilengitide was continued for up to 12 months or until disease progression or unacceptable toxicity. The primary endpoint for efficacy was a 12-month overall survival rate of 65 %. Twenty-nine patients completed study treatment. Sixteen patients survived for 12 months or more, an overall survival rate of 55 %. The median overall survival was 14.5 months (95 % CI 11.1-19.6) and the median progression-free survival was 7.4 months (95 % CI 6.1-8). Cilengitide combined with metronomic temozolomide and procarbazine in MGMT-promoter unmethylated glioblastoma did not improve survival compared with historical data and does not warrant further investigation.
Collapse
Affiliation(s)
- Mustafa Khasraw
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia. .,University of Sydney, Sydney, Australia. .,NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia. .,Royal North Shore Hospital, Pacific HWY, St Leonards, NSW, 2065, Australia.
| | - Adrian Lee
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - Sally McCowatt
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - Zoltan Kerestes
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - Marc E Buyse
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium
| | - Michael Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - Ganessan Kichenadasse
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium.,Flinders Medical Centre and Flinders University, Adelaide, Australia
| | | | - Helen Wheeler
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| |
Collapse
|
6
|
Hypofractionated stereotactic radiotherapy in combination with bevacizumab or fotemustine for patients with progressive malignant gliomas. J Neurooncol 2015; 122:559-66. [DOI: 10.1007/s11060-015-1745-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/16/2015] [Indexed: 01/16/2023]
|
7
|
Yang WB, Xing BZ, Liang H. Comprehensive Analysis of Temozolomide Treatment for Patients with Glioma. Asian Pac J Cancer Prev 2014; 15:8405-8. [DOI: 10.7314/apjcp.2014.15.19.8405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
8
|
The role of cytotoxic chemotherapy in the management of progressive glioblastoma. J Neurooncol 2014; 118:501-55. [DOI: 10.1007/s11060-013-1338-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 12/28/2013] [Indexed: 10/25/2022]
|
9
|
An overview of fotemustine in high-grade gliomas: from single agent to association with bevacizumab. BIOMED RESEARCH INTERNATIONAL 2014; 2014:698542. [PMID: 24800248 PMCID: PMC3988896 DOI: 10.1155/2014/698542] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 03/03/2014] [Indexed: 11/25/2022]
Abstract
Fotemustine is a third-generation nitrosourea showing efficacy in various types of tumors such as melanoma and glioma. We reviewed the most important studies on fotemustine treatment in glioma patients analyzing its pharmacological profile and its activity and safety. Fotemustine was used as single agent or in association with new targeted drugs such as bevacizumab; fotemustine was used both as first-line chemotherapy before temozolomide era and in refractory-temozolomide patients during temozolomide era. Finally, analyzing and comparing the activity and safety of fotemustine alone or in combination with bevacizumab versus other nitrosoureas such as lomustine, we may suggest that the combination treatment with bevacizumab and fotemustine may be active and tolerable in patients with high grade gliomas.
Collapse
|
10
|
Thiepold AL, Lemercier S, Franz K, Atta J, Sulzbacher A, Steinbach JP, Rieger J. Prophylactic use of pegfilgrastim in patients treated with a nitrosourea and teniposide for recurrent glioma. Pharmacotherapy 2014; 34:633-42. [PMID: 24619825 DOI: 10.1002/phar.1409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
STUDY OBJECTIVE As chemotherapy with teniposide and a nitrosourea is commonly used for the treatment of patients with recurrent glioma but can be associated with severe myelotoxicity, we sought to determine if prophylactic administration of pegfilgrastim could reduce leukopenia or infectious complications in patients receiving this chemotherapeutic regimen. DESIGN Retrospective medical record review. SETTING University-affiliated neurooncology hospital in Frankfurt, Germany. PATIENTS Sixty-four patients who received at least one cycle of a nitrosourea agent (nimustine or lomustine) and teniposide for recurrent glioma between 2008 and 2012; of these patients, 28 did not receive prophylactic pegfilgrastim (cohort A), and 36 patients received prophylactic pegfilgrastim (cohort B). MEASUREMENTS AND MAIN RESULTS Blood counts, hospitalizations due to infection or myelosuppression, use of intravenous antibiotics, and survival parameters were analyzed. Leukopenia was more frequently observed before day 30 (early nadir) versus from 30 days until the next cycle (late nadir). In cohort B, Common Terminology Criteria for Adverse Events grade 3 leukopenia in the early nadir occurred less often compared with cohort A (9% in cohort B vs 31% in cohort A). However, the frequency of grade 4 leukopenia, number of days in the hospital due to infection or myelosuppression, days on intravenous antibiotics, progression-free survival, and overall survival were similar between the cohorts. CONCLUSION Moderate, but not severe, leukopenia or related complications could be prevented by prophylactic pegfilgrastim in patients treated with a nitrosourea and teniposide for recurrent glioma. Our results, therefore, do not support routine prophylactic use of pegfilgrastim in these patients.
Collapse
Affiliation(s)
- Anna-Luisa Thiepold
- Dr. Senckenberg Institute of Neurooncology, Goethe University Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|
11
|
Gaviani P, Simonetti G, Salmaggi A, Lamperti E, Silvani A. Safety of second-line chemotherapy with non-conventional fotemustine schedule in recurrent high grade gliomas: a single institution experience. J Neurooncol 2013; 113:527-9. [PMID: 23703296 PMCID: PMC3684713 DOI: 10.1007/s11060-013-1147-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 05/01/2013] [Indexed: 11/27/2022]
Affiliation(s)
- P. Gaviani
- Neuro-Oncology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - G. Simonetti
- Neuro-Oncology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - A. Salmaggi
- Neuro-Oncology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- SC Neurologia Ospedale Manzoni Lecco, Lecco, Italy
| | - E. Lamperti
- Neuro-Oncology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - A. Silvani
- Neuro-Oncology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
12
|
Hypofractionated stereotactic radiotherapy and continuous low-dose temozolomide in patients with recurrent or progressive malignant gliomas. J Neurooncol 2012; 111:187-94. [DOI: 10.1007/s11060-012-0999-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/31/2012] [Indexed: 02/06/2023]
|
13
|
Easaw JC, Mason WP, Perry J, Laperrière N, Eisenstat DD, Del Maestro R, Bélanger K, Fulton D, Macdonald D. Canadian recommendations for the treatment of recurrent or progressive glioblastoma multiforme. ACTA ACUST UNITED AC 2012; 18:e126-36. [PMID: 21655151 DOI: 10.3747/co.v18i3.755] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recommendation 1: Multidisciplinary ApproachTo optimize treatment outcomes, the management of patients with recurrent glioblastoma should be individualized and should involve a multidisciplinary team approach, including neurosurgery, neuropathology, radiation oncology, neuro-oncology, and allied health professions.Recommendation 2: ImagingThe standard imaging modality for assessment of recurrent glioblastoma is Gd-enhanced magnetic resonance imaging (mri). Tumour recurrence should be assessed according to the criteria set out by the Response Assessment in Neuro-Oncology Working Group. The optimal timing and frequency of mri after chemoradiation and adjunctive therapy have not been established.Recommendation 3: Pseudo-progressionProgression observed by mri after chemoradiation can be pseudo-progression. Accordingly, treated patients should not be classified as having progressive disease by Gd-enhancing mri within the first 12 weeks after the end of radiotherapy unless new enhancement is observed outside the radiotherapy field or viable tumour is confirmed by pathology at the time of a required re-operation. Adjuvant temozolomide should be continued and follow-up imaging obtained.Recommendation 4: Repeat SurgerySurgery can play a role in providing symptom relief and confirming tumour recurrence, pseudo-progression, or radiation necrosis. However, before surgical intervention, it is essential to clearly define treatment goals and the expected impact on prognosis and the patient's quality of life. In the absence of level 1 evidence, the decision to re-operate should be made according to individual circumstances, in consultation with the multidisciplinary team and the patient.Recommendation 5: Re-irradiationRe-irradiation is seldom recommended, but can be considered in carefully selected cases of recurrent glioblastoma.Recommendation 6: Systemic TherapyClinical trials, when available, should be offered to all eligible patients. In the absence of a trial, systemic therapy, including temozolomide rechallenge or anti-angiogenic therapy, may be considered. Combination therapy is still experimental; optimal drug combinations and sequencing have not been established.
Collapse
Affiliation(s)
- J C Easaw
- Department of Oncology, Tom Baker Cancer Centre and the University of Calgary, Calgary, AB.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Kuhnhenn J, Kowalski T, Steenken S, Ostermann K, Schlegel U. Procarbazine, carmustine, and vincristine (PBV) for chemotherapy pre-treated patients with recurrent glioblastoma: a single-institution analysis. J Neurooncol 2012; 109:433-8. [DOI: 10.1007/s11060-012-0913-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 06/05/2012] [Indexed: 11/28/2022]
|
15
|
Carpentier A, Chauvet D, Reina V, Beccaria K, Leclerq D, McNichols RJ, Gowda A, Cornu P, Delattre JY. MR-guided laser-induced thermal therapy (LITT) for recurrent glioblastomas. Lasers Surg Med 2012; 44:361-8. [DOI: 10.1002/lsm.22025] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2012] [Indexed: 11/10/2022]
|
16
|
Balañá C, Villá S, Teixidor P. Evolution of care for patients with relapsed glioblastoma. Expert Rev Anticancer Ther 2012; 11:1719-29. [PMID: 22050021 DOI: 10.1586/era.11.152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glioblastoma is the most common and aggressive form of brain tumor in adults and has a poor prognosis and a high recurrence rate despite optimal care. The management of patients with relapsed glioblastoma is challenging, with no widely agreed standard of care. Different chemotherapy and radiotherapy combination treatment strategies provide only modest benefits. Recently, several novel agents, including bevacizumab and XL-184, have demonstrated promising results in Phase II trials with their further evaluation ongoing in Phase III randomized trials. Interpreting the data from such trials is a key challenge in glioblastoma, highlighting the need for relevant and standardized patient assessment techniques. Data from ongoing and planned trials should help to define optimal treatment strategies for this disease.
Collapse
Affiliation(s)
- Carmen Balañá
- Medical Oncology Service, Institut Català d'Oncologia, Germans Trias i Pujol, Carretera Canyet sn, 08916 Badalona, Barcelona, Spain.
| | | | | |
Collapse
|
17
|
Beauchesne P. Fotemustine: a third-generation nitrosourea for the treatment of recurrent malignant gliomas. Cancers (Basel) 2012; 4:77-87. [PMID: 24213227 PMCID: PMC3712672 DOI: 10.3390/cancers4010077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 01/17/2012] [Accepted: 01/18/2012] [Indexed: 11/16/2022] Open
Abstract
Malignant gliomas account for approximately 60% of all primary brain tumors in adults. The prognosis for patients with malignant glioma has not changed significantly in recent years. Despite debulking surgery, radiotherapy and cytotoxic chemotherapy, the median survival time is nine to 12 months, and very few, if any, patients are cured from this illness. Fotemustine is an alkylating agent characterized by the grafting of a phosphonoalanine group onto the nitrosourea radical with consequent high lipophilicity and improved diffusion through the cell membrane and the blood-brain barrier. Fotemustine has been registered for use in two indications: disseminated malignant melanoma, including cerebral metastases, and primary brain tumors. Fotemustine is currently used in Europe, particularly in France and Italy, as a salvage therapy for recurrent malignant gliomas. Myelosuppression, leucopenia and thrombocytopenia are the most frequent side effects of treatment with fotemustine. The objective response to this treatment is between 26% and 70%, and the reported median survival time is 10 months. New drug combinations containing fotemustine and angiogenesis inhibitors, such as bevacizumab, are currently under development. In this review, we describe all the combinations of fotemustine currently used in clinical practice for recurrent malignant gliomas.
Collapse
Affiliation(s)
- Patrick Beauchesne
- Neuro-oncology/Neurology, University Hospital of Nancy, Hôpital CENTRAL, CO N 34, 54035 Nancy cedex, France.
| |
Collapse
|
18
|
Johnson DR, Chang SM. Recent Medical Management of Glioblastoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:26-40. [DOI: 10.1007/978-1-4614-3146-6_3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Silvani A, Gaviani P, Lamperti E, Botturi A, Ferrari D, Simonetti G, Salmaggi A. Lecture: fotemustine in brain tumors. Neurol Sci 2011; 32 Suppl 2:S255-7. [DOI: 10.1007/s10072-011-0800-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
Combined chemotherapy with temozolomide and fotemustine in recurrent glioblastoma patients. J Neurooncol 2011; 104:617-8. [PMID: 21229293 DOI: 10.1007/s11060-010-0515-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 12/20/2010] [Indexed: 10/18/2022]
|
21
|
Guida M, Cramarossa A, Fistola E, Porcelli M, Giudice G, Lubello K, Colucci G. High activity of sequential low dose chemo-modulating Temozolomide in combination with Fotemustine in metastatic melanoma. A feasibility study. J Transl Med 2010; 8:115. [PMID: 21067582 PMCID: PMC2992498 DOI: 10.1186/1479-5876-8-115] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 11/10/2010] [Indexed: 11/24/2022] Open
Affiliation(s)
- Michele Guida
- Department of Medical Oncology; National Institute of Cancer, Bari, Italy.
| | | | | | | | | | | | | |
Collapse
|
22
|
Minniti G, Armosini V, Salvati M, Lanzetta G, Caporello P, Mei M, Osti MF, Maurizi RE. Fractionated stereotactic reirradiation and concurrent temozolomide in patients with recurrent glioblastoma. J Neurooncol 2010; 103:683-91. [DOI: 10.1007/s11060-010-0446-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2010] [Accepted: 10/21/2010] [Indexed: 12/20/2022]
|
23
|
Addeo R, Caraglia M, De Santi MS, Montella L, Abbruzzese A, Parlato C, Vincenzi B, Carraturo M, Faiola V, Genovese M, Cennamo G, Del Prete S. A new schedule of fotemustine in temozolomide-pretreated patients with relapsing glioblastoma. J Neurooncol 2010; 102:417-24. [PMID: 20694830 PMCID: PMC3089727 DOI: 10.1007/s11060-010-0329-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 07/21/2010] [Indexed: 02/08/2023]
Abstract
In the present study we investigated the feasibility and effectiveness of a new biweekly schedule of fotemustine (FTM) in patients with recurrent glioblastoma, after at least one previous treatment. The primary endpoint was progression-free survival at 6 months; secondary objectives were clinical response, overall survival, disease-free survival, and toxicity. Forty patients (median age 52.8 years; median Karnofsky Performance Status at progression 90) underwent second-line chemotherapy with FTM. Selected patients were previously treated with a standard radiotherapy course with concomitant temozolomide (TMZ). After tumor relapse or progression proven by magnetic resonance imaging (MRI), all patients underwent chemotherapy with FTM, given intravenously at dose of 80 mg/m2 every 2 weeks for five consecutive administrations (induction phase), and then every 3 weeks at 100 mg/m2 as maintenance. A total of 329 infusions were administered; the median number of cycles administered was 8. All patients completed the induction phase, and 29 patients received at least one maintenance infusion. Response to treatment was assessed using MacDonald criteria. One complete response [2.5%, 95% confidence interval (CI): 0–10%], 9 partial responses (22.5%, 95% CI: 15–37%), and 16 stable diseases (40%, 95% CI: 32–51%) were observed. Median time to progression was 6.7 months (95% CI: 3.9–9.1 months). Progression-free survival at 6 months was 61%. Median survival from beginning of FTM chemotherapy was 11.1 months. The schedule was generally well tolerated; the main toxicities were hematologic (grade 3 thrombocytopenia in two cases). To the best of our knowledge, this is the first report specifically dealing with the use of a biweekly induction schedule of FTM. The study demonstrates that FTM has therapeutic efficacy as single-drug second-line chemotherapy with a favorable safety profile.
Collapse
Affiliation(s)
- Raffaele Addeo
- Oncology Department, "S. Giovanni di Dio" Hospital, ASL Napoli2 Nord Via Giovanni XXIII, 80028 Frattaminore, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gallo C, Buonerba C, Di Lorenzo G, Romeo V, De Placido S, Marinelli A. Can high-dose fotemustine reverse MGMT resistance in glioblastoma multiforme? J Neurooncol 2010; 100:311-9. [PMID: 20556481 DOI: 10.1007/s11060-010-0168-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 03/23/2010] [Indexed: 12/30/2022]
Abstract
Glioblastoma multiforme (GBM), the highest grade malignant glioma, is associated with a grim prognosis-median overall survival is in the range 12-15 months, despite optimum treatment. Surgery to the maximum possible extent, external beam radiotherapy, and systemic temozolomide chemotherapy are current standard treatments for newly diagnosed GBM, with intracerebral delivery of carmustine wafers (Gliadel). Unfortunately, the effectiveness of chemotherapy can be hampered by the DNA repair enzyme O6-methylguanine methyltransferase (MGMT), which confers resistance both to temozolomide and nitrosoureas, for example fotemustine and carmustine. MGMT activity can be measured by PCR and immunohistochemistry, with the former being the current validated technique. High-dose chemotherapy can deplete MGMT levels in GBM cells and has proved feasible in various trials on temozolomide, in both newly diagnosed and recurrent GBM. We here report the unique case of a GBM patient, with high MGMT expression by immunohistochemistry, who underwent an experimental, high-dose fotemustine schedule after surgery and radiotherapy. Although treatment caused two episodes of grade 3-4 thrombocytopenia, a complete response and survival of more than three years were achieved, with a 30% increase in dose intensity compared with the standard fotemustine schedule.
Collapse
Affiliation(s)
- Chiara Gallo
- Department of Molecular and Clinical Endocrinology and Oncology, University Federico II of Naples, 80100 Via Sergio Pansini 5, Naples, Italy
| | | | | | | | | | | |
Collapse
|
25
|
Fabi A, Metro G, Vidiri A, Lanzetta G, Carosi M, Telera S, Maschio M, Russillo M, Sperduti I, Carapella CM, Cognetti F, Pace A. Low-dose fotemustine for recurrent malignant glioma: a multicenter phase II study. J Neurooncol 2010; 100:209-15. [PMID: 20352471 DOI: 10.1007/s11060-010-0163-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 03/15/2010] [Indexed: 10/19/2022]
Abstract
Fotemustine at the conventional dose of 100 mg/m(2) is an active treatment for recurrent malignant gliomas (RMGs). However, it is associated with a relevant incidence of severe myelotoxicity, which is not justified in the palliative setting of this disease. This study was conducted to address whether administration of fotemustine at 60 mg/m(2) (induction) followed by 75 mg/m(2) (maintenance) would preserve clinical activity with the advantage of improved tolerance. Forty patients with RMGs pretreated with ≤2 lines of chemotherapy were enrolled. Median age was 57 years (26-80) and median Karnofsky performance status was 80 (60-100). Thirty-one patients (77.5%) had tissue available for analysis of the O(6)-methylguanine methyltransferase (MGMT) gene promoter which was found to be methylated in 14 cases (45%). Overall, 8 partial responses (20%) and 13 disease stabilizations (32.5%) were observed for a disease-control rate of 52.5%. At 6 months, 21% of patients were free from progression. Grades 3 and 4 platelet and white blood cell toxicity occurred in ≤10% of patients, and no patients discontinued treatment because of toxicity. No significant difference was observed for disease control rate between methylated and unmethylated patients, although a trend toward improved progression-free survival was reported for methylated patients. Low-dose fotemustine has activity comparable with that of the full-dose regimen, therefore it should be preferred for its greater tolerability. The role of MGMT gene promoter methylation status in relation to sensitivity to fotemustine is still unclear and needs further evaluation in future clinical trials.
Collapse
Affiliation(s)
- Alessandra Fabi
- Division of Medical Oncology, Regina Elena Cancer Institute, Via Elio Chianesi, 53, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Addeo R, De Santi MS, Del Prete S, Caraglia M. Fotemustine and recurrent glioblastoma: possible new opportunities for an old drug. Cancer Chemother Pharmacol 2009; 64:863-6. [PMID: 19639315 DOI: 10.1007/s00280-009-1086-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Accepted: 07/14/2009] [Indexed: 02/04/2023]
|
27
|
Concurrent radiotherapy: fotemustine combination for newly diagnosed malignant glioma patients, a phase II study. Cancer Chemother Pharmacol 2009; 64:171-5. [DOI: 10.1007/s00280-009-0993-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 03/18/2009] [Indexed: 11/28/2022]
|
28
|
Treatment of recurrent malignant gliomas with fotemustine monotherapy: impact of dose and correlation with MGMT promoter methylation. BMC Cancer 2009; 9:101. [PMID: 19335893 PMCID: PMC2667532 DOI: 10.1186/1471-2407-9-101] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Accepted: 03/31/2009] [Indexed: 12/02/2022] Open
Abstract
Background In recurrent malignant gliomas (MGs), a high rate of haematological toxicity is observed with the use of fotemustine at the conventional schedule (100 mg/m2 weekly for 3 consecutive weeks followed by triweekly administration after a 5-week rest period). Also, the impact of O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status on fotemustine activity has never been explored in the clinical setting. Methods 40 patients with recurrent pretreated MG were identified as being treated with fotemustine at doses ranging from 65 mg/m2 to 100 mg/m2. Patients were classified into 3 groups according to the dose of fotemustine received, from the lowest dosage received in group A, to the highest in group C. Analysis of MGMT promoter methylation in tumor tissue was successfully performed in 19 patients. Results Overall, 20% of patients responded to treatment, for a disease control rate (DCR, responses plus stabilizations) of 47.5%. Groups A and B experienced a response rate of 40% and 26.5% respectively, while the corresponding value for group C was 10%. Out of 19 patients, MGMT promoter was found methylated in 12 cases among which a DCR of 66.5% was observed. All 7 patients with unmethylated MGMT promoter were progressive to fotemustine. Conclusion Low-dose fotemustine at 65–75 mg/m2 (induction phase) followed by 75–85 mg/m2 (maintenance phase) has an activity comparable to that of the conventional schedule. By determination of the MGMT promoter methylation status patients might be identified who are more likely to benefit from fotemustine chemotherapy.
Collapse
|
29
|
A multi-institutional phase II study on second-line Fotemustine chemotherapy in recurrent glioblastoma. J Neurooncol 2008; 92:79-86. [PMID: 19018476 DOI: 10.1007/s11060-008-9739-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Accepted: 11/04/2008] [Indexed: 10/21/2022]
Abstract
The present study aims to assess the feasibility and the effectiveness of a second-line Fotemustine chemotherapy in patients with recurrent Glioblastoma after standard primary treatment. Between 2005 and 2007, 50 patients with relapsed malignant glioma (median age=56.8 years; median KPS=90) underwent a second-line chemotherapy with Fotemustine. Selected patients were previously treated with a standard 60 Gy Radiotherapy course and Temozolomide Chemotherapy. Patients were stratified into classes according to the prognostic Recursive Partition Analysis. Endpoints of the study were Progression Free Survival at 6 months, duration of Objective Response and Stabilization, Overall Survival and toxicity. At analysis, 36 patients were dead and 14 were alive. Median follow-up from primary diagnosis was 26.6 months. The Efficacy control of the disease was 62%. PFS was 6.1 months; PFS-6 was 52% and median overall survival from primary diagnosis was 24.5 months, with few manageable haematological toxicities. Fotemustine was safe and effective as second-line chemotherapy in recurrent glioblastoma.
Collapse
|
30
|
Second-line chemotherapy with fotemustine in temozolomide-pretreated patients with relapsing glioblastoma: a single institution experience. Anticancer Drugs 2008; 19:613-20. [PMID: 18525321 DOI: 10.1097/cad.0b013e3283005075] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To evaluate efficacy and safety of fotemustine chemotherapy in temozolomide (TMZ) pretreated adults with recurrent glioblastoma multiforme (GBM). Primary endpoint was progression-free survival at 6 months. Twenty-seven patients (median age: 56 years; median Karnofsky performance status at progression: 80) with relapsed glioblastoma multiforme underwent fotemustine as second-line chemotherapy after failure of homogeneous postoperative treatment consisting of conformal radiotherapy (60 Gy in 30 fractions) with concomitant TMZ (75 mg/m2 per day), followed by six courses of TMZ (150-200 mg/m2 for 5 days every 28 days). Patients were assigned to Radiation Therapy Oncology Group recursive partitioning analysis classes for gliomas. After MRI-proven tumor relapse or progression, all patients underwent chemotherapy with fotemustine, given intravenously 100 mg/m2 every week for 3 consecutive weeks (induction phase) and then every 3 weeks (maintenance phase). Adequate liver, renal, and bone marrow functions were required. Toxicity grading was based on the National Cancer Institute's Common Toxicity Criteria (version 2.0). Response to treatment was assessed on MacDonald criteria. According to an intention-to-treat-analysis, data on all enrolled patients were included in statistical analysis. Eight partial responses (29.6%) and five cases of stable disease (18.5%) were observed. Median time to progression was 5.7 months. Progression-free survival at 6 months was 48.15%. Median survival from the beginning of fotemustine chemotherapy was 9.1 months. Median survival from diagnosis of glioblastoma was 21.2 months. Toxicity was manageable and mainly hematological (grade 3 thrombocytopenia: three cases; grade 4 leukopenia: one case). Fotemustine has shown therapeutic efficacy as single-drug second-line chemotherapy in treatment of TMZ pretreated patients.
Collapse
|