Carmustine implants and Temozolomide for the treatment of newly diagnosed high grade glioma

Similar documents
Scottish Medicines Consortium

University of Zurich. Temozolomide and MGMT forever? Zurich Open Repository and Archive. Weller, M. Year: 2010

Clinical Management Protocol Chemotherapy [Glioblastoma Multiforme (CNS)] Protocol for Planning and Treatment

Concomitant (without adjuvant) temozolomide and radiation to treat glioblastoma: A retrospective study

doi: /bjr/

Gl i o b l a s t o m a multiforme is the most common

Chemotherapy in Adults with Gliomas

Adjuvant treatment of high grade gliomas

Newcastle Neuro-oncology Team Audit of Outcome of Glioblastoma Multiforme Chemoradiotherapy Treatment

Cilengitide (Impetreve) for glioblastoma multiforme. February 2012

National Institute for Health and Clinical Excellence. Single Technology Appraisal (STA)

CNS Tumors: The Med Onc Perspective. Ronald J. Scheff, MD Associate Clinical Professor Weill Medical College of Cornell U.

PROCARBAZINE, lomustine, and vincristine (PCV) is

Technology appraisal guidance Published: 27 June 2007 nice.org.uk/guidance/ta121

Chemotherapy in malignant brain tumors

Temozolomide with Radiotherapy for the Treatment of Malignant Gliomas, Center Experience

Corporate Medical Policy

This study was designed to evaluate an online prognosis

Dose dense 1 week on/1 week off temozolomide in recurrent glioma: a retrospective study

Corporate Medical Policy

Title: Carmustine (BCNU) for recurrent glioblastoma multiforme: efficacy, toxicity and prognostic factors

3-D conformal radiotherapy with concomitant and adjuvant temozolomide for patients with glioblastoma multiforme and evaluation of prognostic factors

Glioblastoma: Adjuvant Treatment Abdulrazag Ajlan, MD, MSc, FRCSC, UCNS(D)

Contemporary Management of Glioblastoma

ORIGINAL PAPERS. The Impact of Surgery on the Efficacy of Adjuvant Therapy in Glioblastoma Multiforme

Temozolomide in the treatment of recurrent malignant glioma in Chinese patients!"#$%&'()*+,-./0,1234

Treatment of recurrent high-grade gliomas

Corporate Medical Policy

Epidemiology and outcome research of glioma patients in Southern Switzerland: A population based analysis

Examining large groups of cancer patients to identify ways of predicting which therapies cancers might respond to.

21/03/2017. Disclosure. Practice Changing Articles in Neuro Oncology for 2016/17. Gliomas. Objectives. Gliomas. No conflicts to declare

Appraisal of carmustine. implants and temozolomide. for newly diagnosed high. Brain and Spine Foundation

PRESURGICAL PLANNING. Strongly consider neuropsychological evaluation before functional imaging study Strongly consider functional imaging study

Corporate Medical Policy Tumor-Treatment Fields Therapy for Glioblastoma

Stephanie E. Combs Æ Marc Bischof Æ Thomas Welzel Æ Holger Hof Æ Susanne Oertel Æ Jürgen Debus Æ Daniela Schulz-Ertner

Original. Key words: Glioblastoma multiforme, temozolomide, radiation NEMROCK

Zurich Open Repository and Archive. Long-term survival of glioblastoma patients treated with radiotherapy and lomustine plus temozolomide

Zurich Open Repository and Archive. Long-term survival of glioblastoma patients treated with radiotherapy and lomustine plus temozolomide

Rapid regression of glioblastoma following carmustine wafer implantation: A case report

An international study under the guidance of the European Organization

Hypofractionated radiation therapy for glioblastoma

Low grade glioma: a journey towards a cure

Imaging for suspected glioma

First-line treatment of malignant glioma with carmustine implants followed by concomitant radiochemotherapy: a multicenter experience

Neuro-Oncology. Martin J. van den Bent. Department of Neuro-oncology/Neurology, Erasmus M.C. Cancer Institute, Rotterdam, Netherlands

Concomitant Accelerated Hyper-Fractionated Radiotherapy and Temozolomide in the Treatment of High Grade Astrocytoma

Elderly Patients with Glioblastoma Multiforme Treated with Concurrent Temozolomide and Standard- versus Abbreviated-Course Radiotherapy

Management of Glioma: The Basics Glioma Update The clinical challenge. Glioma a malignant disease of the CNS

See the corresponding editorial in this issue, pp 1 2. J Neurosurg 115:3 8, An extent of resection threshold for newly diagnosed glioblastomas

Systemic Treatment. Third International Neuro-Oncology Course. 23 May 2014

Collection of Recorded Radiotherapy Seminars

Management of malignant glioma: steady progress with multimodal approaches

THE EFFECTIVE OF BRAIN CANCER AND XAY BETWEEN THEORY AND IMPLEMENTATION. Mustafa Rashid Issa

Background. Central nervous system (CNS) tumours. High-grade glioma

Lynn S. Ashby 1*, Kris A. Smith 2 and Baldassarre Stea 3

PRINCESS MARGARET CANCER CENTRE CLINICAL PRACTICE GUIDELINES

Phase 2 study of BCNU and temozolomide for recurrent glioblastoma multiforme: North American Brain Tumor Consortium study 1

Efficacy of Treatment for Glioblastoma Multiforme in Elderly Patients (65+): A Retrospective Analysis

Glioblastoma multiforme has been described as. Bevacizumab and irinotecan therapy in glioblastoma multiforme: a series of 13 cases

Chemotherapy plus Radiotherapy versus Radiotherapy Alone for Patients with Anaplastic Oligodendroglioma: Long Term Results of RTOG 9402

Oncological Management of Brain Tumours. Anna Maria Shiarli SpR in Clinical Oncology 15 th July 2013

Incidence of Early Pseudo-progression in a Cohort of Malignant Glioma Patients Treated With Chemoirradiation With Temozolomide

Precision medicine for gliomas

Changing Paradigms An Update on the Multidisciplinary Management of Malignant Glioma

Practice of Interferon Therapy

Copyright information:

High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up

Bevacizumab in combination with temozolomide and regional radiation therapy for up-front treatment of patients with newly-diagnosed glioblastoma

BRAIN TUMOURS: SUCCESSES AND CHALLENGES ON THE OTHER SIDE OF THE BLOOD-BRAIN BARRIER

Protocol. Tumor Treatment Fields Therapy for Glioblastoma

New Approaches in Brain Tumor Treatment. Virginia Stark-Vance, M.D.

The Response to Chemo Radiation. Therapy in Unresectable Glioblastoma

Intracavitary Balloon Catheter Brain Brachytherapy for Malignant Gliomas or Metastasis to the Brain. Original Policy Date

Radiation and concomitant chemotherapy for patients with glioblastoma multiforme

Related Policies None

Interferon β and temozolomide combination therapy for temozolomide monotherapy refractory malignant gliomas

High-grade glioma (World Health Organization Grades III and IV) Salvage Therapy in Patients with Glioblastoma. Is There any Benefit?

Supratentorial Glioblastoma Multiforme: The Role of Surgical Resection Versus Biopsy Among Older Patients

Technology appraisal guidance Published: 26 April 2001 nice.org.uk/guidance/ta23

Glioblastoma (GBM) is the most common and

Bevacizumab rescue therapy extends the survival in patients with recurrent malignant glioma

Monitoring temozolomide treatment of low-grade glioma with proton magnetic resonance spectroscopy

VAL-083: Validated DNA-targeting Agent for Underserved Cancer Patients. September 2018

ORE Open Research Exeter

MP Intracavitary Balloon Catheter Brain Brachytherapy for Malignant Gliomas or Metastasis to the Brain. Related Policies None

Citation Pediatrics international (2015), 57.

Radioterapia no Tratamento dos Gliomas de Baixo Grau

University of Colorado Cancer Center Brain Disease Site Schema

Safety and efficacy of carmustine (BCNU) wafers for metastatic brain tumors

National Horizon Scanning Centre. Bevacizumab (Avastin) for glioblastoma multiforme - relapsed. August 2008

Survival of High Grade Glioma Patients Treated by Three Radiation Schedules with Chemotherapy: A Retrospective Comparative Study

NICE guideline Published: 11 July 2018 nice.org.uk/guidance/ng99

Of all neoplastic lesions and across all medical specialties, Eosinophilic meningitis triggered by implanted Gliadel wafers: case report

UNDERSTANDING MOLECULAR TESTING IN BRAIN TUMORS: HOW CLINICALLY USEFUL IS IT?

Neuro-Oncology Program

Hematologic Toxicity of Temozolomide and Radiation in Glioblastoma Patients Correlation with Clinicopathological Factors

ONC201: a new treatment option being tested clinically for recurrent glioblastoma

Treatment and outcomes for glioblastoma in elderly compared with non-elderly patients: a population-based study

Wafer migration: Monitor patients for signs of obstructive hydrocephalus (5.5). Embryo-fetal toxicity: Can cause fetal harm (5.6)

Brain Schema March 2018

Transcription:

National Institute for Health and Clinical Excellence Health Technology Appraisal Carmustine implants and Temozolomide for the treatment of newly diagnosed high grade glioma Personal statement Conventional treatment for malignant gliomas, anaplastic astrocytoma WHO grade III (AA) and glioblastoma multiforme WHO grade IV (GBM), consists of surgery followed by external beam radiotherapy. While the survival and quality of life benefit of surgery has not been tested in prospective randomised trials, the evidence for the benefit of external beam radiotherapy was demonstrated in three randomised trials (1, 2) (3). The role of adjuvant chemotherapy, particularly with nitrosoureas, has been under test since 1970 s. While individual randomised trials showed marginal or no survival benefit, a meta-analysis of 12 trials of over 3000 patients demonstrated a 6% survival benefit at 1 year and 5% at 2 years and prolongation of median survival by approximately 2 months (4). Despite many years of research, the reality of malignant glioma is that it represents an incurable malignancy with a median life expectancy in the region of 12 months and few, if any, long term survivors. The principal prognostic factors for survival are age, performance status and tumour histology. More favourable outcome is seen in younger patients with anaplastic astrocytoma and good performance status, and less favourable in older patients with glioblastoma histology and functional impairment. The two new treatments under consideration, Carmustine implants (Gliadel) and Temozolomide, involve the use of alkylating agents given as intralesional or systemic treatment. Carmustine (BCNU) impregnated polymer wafers (Gliadel) are a means of intralesional delivery of alkylating agent by insertion of wafers into the resection cavity at the time of surgical removal of malignant glioma. The efficacy of Gliadel was first examined at the time of recurrence. In patients with recurrent GBM treated with salvage surgery, the insertions of BCNU impregnated wafers compared to polymer wafers alone (controls), was shown to prolong median survival by 8 weeks (5). Only one adequately powered randomised study tested Gliadel against placebo wafers as an adjuvant treatment to surgery and radiotherapy in newly diagnosed malignant glioma. In the study of 240 patients with GBM and AA, Gliadel was shown to prolong median survival by 2.3 months with approximately 10% survival benefit at 1 year and no demonstrable survival benefit at 18 months. In the principal subgroup of 207 patients with GBM the survival difference did not reach 1

statistical significance and no survival difference was seen beyond 18 months (6). Temozolomide, an orally administered alkylating agent, was first tested at the time of recurrence. In a randomised phase II study in patients with recurrent GBM, median progression free survival was prolonged by one month when compared to Procarbazine and this did not translate into a significant survival benefit (7). Temozolomide was tested in a randomised study of 573 newly diagnosed patients with GBM as an additional treatment to surgery and radiotherapy, given as concomitant treatment with radiation and as adjuvant chemotherapy for 6 months following completion of radiotherapy. The use of chemotherapy was associated with prolongation of median survival by 2 months and an improvement in 2 year survival of 15% (8). In summary, the addition of Gliadel wafers at the time of surgery was shown to result in prolongation of median survival in a group of patients with GBM and AA. Although this is also likely to be the case for patients with GBM, the size of the study precluded the demonstration of a statistically significant survival benefit in this group. In the available peer review publication, the survival benefit is most pronounced at the median timepoint with no clear benefit seen beyond 16-18 months. Temozolomide given as concomitant and adjuvant treatment has shown survival benefit in patients with GBM, both at the median timepoint and persisting for the duration of the study with a persistent survival difference at 2 years. Nevertheless, all patients with GBM regardless of the use of Temozolomide, progress and no long term cures have been reported. Both trials show encouraging results with survival benefit in a population of patients with poor prognosis not shown in individual trials carried out in the preceding decade. Nevertheless, the published results are in line with the existing knowledge of the effectiveness of systemic nitrosoureas in the treatment of malignant gliomas. Neither of the treatments under consideration have been compared to nitrosoureas (BCNU, CCNU or ACNU) and on the basis of the available data the magnitude of benefit of the new treatments is within the range seen for systemic nitrosoureas. A commonly held belief is that the toxicity of the new approaches is less that that seen previously with nitrosoureas but sound comparative data is lacking. Both randomised studies are the first demonstration of the survival benefit of the treatments under test and contain a relatively small number of patients. Despite a possible statistical desire to confirm the results in more robust studies, it is unlikely that the desperate plight of patients with malignant glioma would make repeat trials acceptable. While both studies show a potential way ahead with prolongation of survival apparently without marked additional toxicity, they also leave a number of unanswered questions. The first question is a comparison to nitrosourea based chemotherapy. Realistically, a randomised study of Temozolomide against 2

nitrosoureas would be unlikely to accrue patients. A study comparing systemic and intralesional nitrosoureas is a marginally better prospect but in the current climate would have to allow for administration of Temozolomide and therefore only limited time for exposure to systemic nitrosoureas. Further question is the applicability of the results demonstrated largely in patients with GBM to patients with AA. Based on the apparent equivalent benefit for AA and GBM shown in the meta-analysis (4) and on the apparently superior efficacy of Temozolomide in terms of response rate in patients with recurrent AA (9) compared in separate studies to recurrent GBM (7) (10) the treatment under test may be considered effective in AA. However, existing knowledge about the toxicity of concomitant chemotherapy and radiotherapy at other sites raises a concern about the potential damaging effect of combination of concomitant chemotherapy and brain irradiation and this is of particularly relevance for the best prognostic group of patients with malignant glioma who are young with AA and have a median life expectancy measured in years. One or more trials addressing the treatments under test in patients with AA should be encouraged. A trial addressing the role of concomitant Temozolomide in patients with AA is in advanced stage of preparation by the NCRI Brain Tumour Clinical Studies Group. Another unanswered question is the potential relationship between the two treatments. On the basis of the available evidence, it is not possible to make a statement about the comparative efficacy of the two treatments under consideration unless a specific analysis of matched patients is performed. In this respect, a randomised trial comparing the individual treatments alone and in combination would be useful but in practice such a study would be difficult to organise. The reality of malignant glioma is that patients seek any treatment which may offer some benefit. If both treatments are approved, it is likely that patients who have Gliadel wafers inserted at initial surgery will be considered for additional treatment with Temozolomide, based on the unproven assumption of additive effect. Prospective collection of national and international outcome data on combined treatment would be of value. One of the determinants of the efficacy of alkylating agents is the status of the DNA repair machinery. Studies of MGMT gene methylation status, used as a measure of the activity of the repair enzyme of alkylating agent induced damage, suggest that the benefit for adjuvant chemotherapy is seen predominantly in patients with impaired repair capacity (11). Further use of Gliadel and Temozolomide, both within and outside trials, should be accompanied by tests of repair enzyme status with the future aim of tailoring treatment to its predicted efficacy. Future studies of molecular predictors of treatment effectiveness should also lead to the development of strategies to overcome potential treatment resistance. 3

References: 1. Walker MD, Alexander E, Jr., Hunt WE, MacCarty CS, Mahaley MS, Jr., Mealey J, Jr., et al. Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas. A cooperative clinical trial. J Neurosurg. 1978;49(3):333-43. 2. Kristiansen K, Hagen S, Kollevold T, Torvik A, Holme I, Nesbakken R, et al. Combined modality therapy of operated astrocytomas grade III and IV. Confirmation of the value of postoperative irradiation and lack of potentiation of bleomycin on survival time: a prospective multicenter trial of the Scandinavian Glioblastoma Study Group. Cancer. 1981;47(4):649-52. 3. Sandberg WM, Malmström P, Strömblad LG, Anderson H, Borgström S, Brun A, et al. A randomized study of chemotherapy with procarbazine, vincristine, and lomustine with and without radiation therapy for astrocytoma grades 3 and/or 4. Cancer. 1991;68:22-9. 4. Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta- analysis of individual patient data from 12 randomised trials. Lancet. 2002;359(9311):1011-8. 5. Brem H, Piantadosi S, Burger PC, Walker M, Selker R, Vick NA, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group [see comments]. Lancet. 1995;345(8956):1008-12. 6. Westphal M, Hilt DC, Bortey E, Delavault P, Olivares R, Warnke PC, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neurooncol. 2003 Apr;5(2):79-88. 7. Yung WK, Albright RE, Olson J, Fredericks R, Fink K, Prados MD, et al. A phase II study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse. Br J Cancer. 2000;83(5):588-93. 8. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005 Mar 10;352(10):987-96. 9. Yung W, Prados M, Yaya-Tur R, Rosenfeld S, Brada M, Friedman H, et al. Multicenter Phase II Trial of Temozolomide in Patients With Anaplastic Astrocytoma or Anaplastic Oligoastrocytoma at First Relapse. Journal of Clinical Oncology. 1999;17:2762. 4

10. Brada M, Viviers L, Abson C, Hines F, Britton J, Ashley S, et al. Phase II study of primary temozolomide chemotherapy in patients with WHO grade II gliomas. Ann Oncol. 2003 Dec;14(12):1715-21. 11. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005 Mar 10;352(10):997-1003. Michael Brada Professor of Clinical Oncology The Institute of Cancer Research & The Royal Marsden NHS Foundation Trust 5