1
|
Narang K, Kataria T, Bisht SS, Gupta D, Banerjee S, Mayank M, Shishak S, Kaliyaperumal V, Tamilselvan S, Kamaraj D, Abraham S. Contemporary Long-term Survival Outcomes and Prognostic Factors in Adult grade 4 Astrocytoma: An Institutional Analysis. Clin Oncol (R Coll Radiol) 2025; 40:103788. [PMID: 40048926 DOI: 10.1016/j.clon.2025.103788] [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: 07/07/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 03/29/2025]
Abstract
AIMS Astrocytoma grade 4 without isocitrate dehydrogenase (IDH)-based characterisation has been called glioblastoma (GBM) in historical cohorts. There have been significant advancements in diagnostic radiology and pathology, and in the technical aspects of surgery, radiation therapy, and temozolomide (TMZ) used for treatment of this disease. We analysed the outcomes of 267 adult astrocytoma grade 4/GBM patients, consecutively treated between December 2010 and November 2018 using modern techniques at our institute. MATERIALS AND METHODS All patients underwent surgical resection, histopathology review, and O6-methylguanine-DNA methyltransferase (MGMT) methylation testing, volumetric modulated arc therapy (VMAT)-based radiation therapy using institute-specific target-delineation guidelines and image guidance, and TMZ according to Stupp protocol. Serial multiparametric magnetic resonance imaging-based follow-up ensured early detection of disease progression. Appropriate salvage therapy was determined based on clinicopathological attributes. Kaplan-Meier survival plots, log-rank test, and Cox regression analysis were performed on the prospectively recorded dataset to estimate survival and the factors affecting it. RESULTS At a median follow-up of 72 months, the median progression-free survival (PFS), 1-year PFS, and 2-year PFS were 10 months, 37.8%, and 17.5%, respectively. MGMT-methylation, a radiation dose ≥54 Gy, and ≥4 adjuvant TMZ cycles were associated with favourable PFS. Median overall survival (OS), 2-year OS and 5-year OS were 24 months, 48%, and 18%, respectively. MGMT-methylation and 1-year disease control were associated with favourable OS. Salvage treatment could be offered to 69.2% patients, with use of all the three treatment modalities in 12.4%. Salvage reirradiation could be used in 30.8% patients. Haematological toxicity ≥grade 2 was evident in 6% patients during concurrent radiation-TMZ phase and in 9% patients in adjuvant TMZ phase. Postradiation neurocognitive deficits were noted in 20.1% patients, with onset at a median duration of 10 months. CONCLUSION Modern diagnostic and therapeutic techniques affected a near-doubling of survival and acceptable late toxicity, as compared to historical data.
Collapse
Affiliation(s)
- K Narang
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India.
| | - T Kataria
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S S Bisht
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - D Gupta
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Banerjee
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - M Mayank
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Shishak
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - V Kaliyaperumal
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Tamilselvan
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - D Kamaraj
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| | - S Abraham
- Division of Radiation Oncology, Medanta Cancer Institute, Medanta The Medicity, Gurugram, India
| |
Collapse
|
2
|
Kirti A, Simnani FZ, Jena S, Lenka SS, Kalalpitiya C, Naser SS, Singh D, Choudhury A, Sahu RN, Yadav A, Sinha A, Nandi A, Panda PK, Kaushik NK, Suar M, Verma SK. Nanoparticle-mediated metronomic chemotherapy in cancer: A paradigm of precision and persistence. Cancer Lett 2024; 594:216990. [PMID: 38801886 DOI: 10.1016/j.canlet.2024.216990] [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: 09/04/2023] [Revised: 03/05/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Current methods of cancer therapy have demonstrated enormous potential in tumor inhibition. However, a high dosage regimen of chemotherapy results in various complications which affect the normal body cells. Tumor cells also develop resistance against the prescribed drugs in the whole treatment regimen increasing the risk of cancer relapse. Metronomic chemotherapy is a modern treatment method that involves administering drugs at low doses continuously, allowing the drug sufficient time to take its effect. This method ensures that the toxicity of the drugs is to a minimum in comparison to conventional chemotherapy. Nanoparticles have shown efficacy in delivering drugs to the tumor cells in various cancer therapies. Combining nanoparticles with metronomic chemotherapy can yield better treatment results. This combination stimulates the immune system, improving cancer cells recognition by immune cells. Evidence from clinical and pre-clinical trials supports the use of metronomic delivery for drug-loaded nanoparticles. This review focuses on the functionalization of nanoparticles for improved drug delivery and inhibition of tumor growth. It emphasizes the mechanisms of metronomic chemotherapy and its conjunction with nanotechnology. Additionally, it explores tumor progression and the current methods of chemotherapy. The challenges associated with nano-based metronomic chemotherapy are outlined, paving the way for prospects in this dynamic field.
Collapse
Affiliation(s)
- Apoorv Kirti
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | | | - Snehasmita Jena
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Sudakshya S Lenka
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | | | | | - Dibyangshee Singh
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Anmol Choudhury
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Rudra Narayan Sahu
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Anu Yadav
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Adrija Sinha
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Aditya Nandi
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India; Instituto de Investigaciones en Materiales, UNAM, 04510, CDMX, Mexico
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20, Uppsala, Sweden
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, Seoul, 01897, Republic of Korea.
| | - Mrutyunjay Suar
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India.
| | - Suresh K Verma
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, 751024, India.
| |
Collapse
|
3
|
Liao Y, Bai X, Cao Y, Zhang M. Effect of low-dose bevacizumab on health-related quality of life in patients with recurrent high-grade glioma: A retrospective clinical study. J Clin Neurosci 2024; 120:196-203. [PMID: 38277995 DOI: 10.1016/j.jocn.2024.01.018] [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: 09/07/2023] [Revised: 12/23/2023] [Accepted: 01/14/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND We retrospectively analyzed the effects of low-dose bevacizumab (BEV) combined with temozolomide (TMZ) on health-related quality of life (HRQL) in patients with recurrent high-grade glioma (rHGG). METHODS A total of 129 patients with rHGG were included in this study. Patients were divided into a combination group and TMZ group based on the treatment they received. The Quality of Life Questionnaire Core 30 (QLQ-C30) and EORTC Brain Cancer Module (QLQ-BN20) were used to evaluate HRQL in all patients before and after treatment. Categorical variables were compared using the chi-squared test. The data for all continuous variables were first tested for a normal distribution. If the data conformed to a normal distribution, a T test was used for comparison. If the data did not conform to a normal distribution, the rank-sum test was used. RESULTS There were differences in PFS and PFS-6 between the BEV + TMZ and TMZ groups (P<0.05). However, there was no difference in the OS between the two groups (P>0.05). The BEV + TMZ group performed better than the TMZ group in both the QLQ-C30 and QLQ-BN20. In addition, the KPS score was higher in the BEV + TMZ group than in the TMZ group. Steroid doses given were lower in the BEV + TMZ group than in the TMZ group (P < 0.05). CONCLUSIONS Low-dose BEV + TMZ can relieve the clinical symptoms of rHGG patients, reduce their steroid dose, improve HRQL, and prolong PFS, but does not bear any benefit on OS.
Collapse
Affiliation(s)
- Yonghong Liao
- Neurosurgery of The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xuexue Bai
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yiyao Cao
- Neurosurgery of The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Maoying Zhang
- Neurosurgery of The First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
4
|
Yoon WS, Chang JH, Kim JH, Kim YJ, Jung TY, Yoo H, Kim SH, Ko YC, Nam DH, Kim TM, Kim SH, Park SH, Lee YS, Yim HW, Hong YK, Yang SH. Efficacy and safety of metformin plus low-dose temozolomide in patients with recurrent or refractory glioblastoma: a randomized, prospective, multicenter, double-blind, controlled, phase 2 trial (KNOG-1501 study). Discov Oncol 2023; 14:90. [PMID: 37278858 DOI: 10.1007/s12672-023-00678-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/28/2023] [Indexed: 06/07/2023] Open
Abstract
PURPOSE Glioblastoma (GBM) has a poor prognosis after standard treatment. Recently, metformin has been shown to have an antitumor effect on glioma cells. We performed the first randomized prospective phase II clinical trial to investigate the clinical efficacy and safety of metformin in patients with recurrent or refractory GBM treated with low-dose temozolomide. METHODS Included patients were randomly assigned to a control group [placebo plus low-dose temozolomide (50 mg/m2, daily)] or an experimental group [metformin (1000 mg, 1500 mg, and 2000 mg per day during the 1st, 2nd, and 3rd week until disease progression, respectively) plus low-dose temozolomide]. The primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), disease control rate, overall response rate, health-related quality of life, and safety. RESULTS Among the 92 patients screened, 81 were randomly assigned to the control group (43 patients) or the experimental group (38 patients). Although the control group showed a longer median PFS, the difference between the two groups was not statistically significant (2.66 versus 2.3 months, p = 0.679). The median OS was 17.22 months (95% CI 12.19-21.68 months) in the experimental group and 7.69 months (95% CI 5.16-22.67 months) in the control group, showing no significant difference by the log-rank test (HR: 0.78; 95% CI 0.39-1.58; p = 0.473). The overall response rate and disease control rate were 9.3% and 46.5% in the control group and 5.3% and 47.4% in the experimental group, respectively. CONCLUSIONS Although the metformin plus temozolomide regimen was well tolerated, it did not confer a clinical benefit in patients with recurrent or refractory GBM. Trial registration NCT03243851, registered August 4, 2017.
Collapse
Affiliation(s)
- Wan-Soo Yoon
- Department of Neurosurgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jeong Hoon Kim
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Tae-Young Jung
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Heon Yoo
- Department of Neuro-Oncology Clinic, Center for Specific Organs Cancer, National Cancer Center Hospital, National Cancer Center, Goyang, Korea
| | - Se-Hyuk Kim
- Department of Neurosurgery, Ajou University Hospital, Ajou University School of Medicine, Suwon, Korea
| | - Young-Cho Ko
- Department of Neurosurgery, Konkuk University Medical Center, Seoul, Korea
| | - Do-Hyun Nam
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Hae Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Youn Soo Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyeon Woo Yim
- Department of Preventive Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Kil Hong
- Department of Neurosurgery, Hallym University Sacred Heart Hospital, The Hallym University Medical Center, 22, Gwanpyeong-ro 170 beon-gil, Dong-gu, Anyang-si, Gyeongggi-do, 14068, Korea.
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, 93 Jungbudaero, Paldal-gu, Suwon, Seoul, 16247, Korea.
| |
Collapse
|
5
|
Jenkins S, Zhang W, Steinberg SM, Nousome D, Houston N, Wu X, Armstrong TS, Burton E, Smart DD, Shah R, Peer CJ, Mozarsky B, Arisa O, Figg WD, Mendoza TR, Vera E, Brastianos P, Carter S, Gilbert MR, Anders CK, Connolly RM, Tweed C, Smith KL, Khan I, Lipkowitz S, Steeg PS, Zimmer AS. Phase I Study and Cell-Free DNA Analysis of T-DM1 and Metronomic Temozolomide for Secondary Prevention of HER2-Positive Breast Cancer Brain Metastases. Clin Cancer Res 2023; 29:1450-1459. [PMID: 36705597 PMCID: PMC10153633 DOI: 10.1158/1078-0432.ccr-22-0855] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 11/22/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023]
Abstract
PURPOSE Preclinical data showed that prophylactic, low-dose temozolomide (TMZ) significantly prevented breast cancer brain metastasis. We present results of a phase I trial combining T-DM1 with TMZ for the prevention of additional brain metastases after previous occurrence and local treatment in patients with HER2+ breast cancer. PATIENTS AND METHODS Eligible patients had HER2+ breast cancer with brain metastases and were within 12 weeks of whole brain radiation therapy (WBRT), stereotactic radiosurgery, and/or surgery. Standard doses of T-DM1 were administered intravenously every 21 days (3.6 mg/kg) and TMZ was given orally daily in a 3+3 phase I dose escalation design at 30, 40, or 50 mg/m2, continuously. DLT period was one 21-day cycle. Primary endpoint was safety and recommended phase II dose. Symptom questionnaires, brain MRI, and systemic CT scans were performed every 6 weeks. Cell-free DNA sequencing was performed on patients' plasma and CSF. RESULTS Twelve women enrolled, nine (75%) with prior SRS therapy and three (25%) with prior WBRT. Grade 3 or 4 AEs included thrombocytopenia (1/12), neutropenia (1/12), lymphopenia (6/12), and decreased CD4 (6/12), requiring pentamidine for Pneumocystis jirovecii pneumonia prophylaxis. No DLT was observed. Four patients on the highest TMZ dose underwent dose reductions. At trial entry, 6 of 12 patients had tumor mutations in CSF, indicating ongoing metastatic colonization despite a clear MRI. Median follow-up on study was 9.6 m (2.8-33.9); only 2 patients developed new parenchymal brain metastases. Tumor mutations varied with patient outcome. CONCLUSIONS Metronomic TMZ in combination with standard dose T-DM1 shows low-grade toxicity and potential activity in secondary prevention of HER2+ brain metastases.
Collapse
Affiliation(s)
- Sarah Jenkins
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | - Wei Zhang
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | - Seth M. Steinberg
- Biostatistics and Data Management Section; Center for Cancer Research, NCI, NIH
| | - Darryl Nousome
- Center for Cancer Research Collaborative Bioinformatics Resource, NCI, NIH
| | - Nicole Houston
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | - Xiaolin Wu
- Cancer Research Technology Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Dee Dee Smart
- Radiation Oncology Branch, Center for Cancer Research, NCI NIH
| | - Ritu Shah
- Neuro-Radiology, Clinical Center Cancer Research, NIH
| | - Cody J. Peer
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | - Brett Mozarsky
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | - Oluwatobi Arisa
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, NCI NIH
| | | | | | - Priscilla Brastianos
- Massachusetts General Hospital, Harvard Cancer Center, Harvard University, Boston, MA
| | - Scott Carter
- Division of Medical Sciences, Harvard University, Boston, MA
| | | | | | | | - Carol Tweed
- University of Maryland Oncology, Baltimore MD
| | - Karen L. Smith
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - Imran Khan
- Women’s Malignancies Branch; Center for Cancer Research, NCI, NIH
| | | | | | | |
Collapse
|
6
|
Prajapati HP, Ansari A. Updates in the Management of Recurrent Glioblastoma Multiforme. J Neurol Surg A Cent Eur Neurosurg 2023; 84:174-187. [PMID: 35772723 DOI: 10.1055/s-0042-1749351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND Glioblastoma is the most aggressive and diffusely infiltrative primary brain tumor. Recurrence is almost universal even after all primary standard treatments. This article aims to review the literature and update the standard treatment strategies for patients with recurrent glioblastoma. METHODS A systematic search was performed with the phrase "recurrent glioblastoma and management" as a search term in PubMed central, Medline, and Embase databases to identify all the articles published on the subject till December 2020. The review included peer-reviewed original articles, clinical trials, review articles, and keywords in title and abstract. RESULTS Out of 513 articles searched, 73 were included in this review after screening for eligibility. On analyzing the data, most of the studies report a median overall survival (OS) of 5.9 to 11.4 months after re-surgery and 4.7 to 7.6 months without re-surgery. Re-irradiation with stereotactic radiosurgery (SRS) and fractionated stereotactic radiotherapy (FSRT) result in a median OS of 10.2 months (range: 7.0-12 months) and 9.8 months (ranged: 7.5-11.0 months), respectively. Radiation necrosis was found in 16.6% (range: 0-24.4%) after SRS. Chemotherapeutic agents like nitrosourea (carmustine), bevacizumab, and temozolomide (TMZ) rechallenge result in a median OS in the range of 5.1 to 7.5, 6.5 to 9.2, and 5.1-13.0 months and six months progression free survival (PFS-6) in the range of 13 to 17.5%, 25 to 42.6%, and 23 to 58.3%, respectively. Use of epithelial growth factor receptor (EGFR) inhibitors results in a median OS in the range of 2.0 to 3.0 months and PFS-6 in 13%. CONCLUSION Although recurrent glioblastoma remains a fatal disease with universal mortality, the literature suggests that a subset of patients may benefit from maximal treatment efforts.
Collapse
Affiliation(s)
- Hanuman Prasad Prajapati
- Department of Neurosurgery, Uttar Pradesh University of Medical Sciences, Etawah, Uttar Pradesh, India
| | - Ahmad Ansari
- Department of Neurosurgery, Uttar Pradesh University of Medical Sciences, Safai, Uttar Pradesh, India
| |
Collapse
|
7
|
Metronomic Temozolomide in Heavily Pretreated Patients With Recurrent Isocitrate Dehydrogenase Wild-type Glioblastoma: A Large Real-Life Mono-Institutional Study. Clin Oncol (R Coll Radiol) 2023; 35:e319-e327. [PMID: 36858930 DOI: 10.1016/j.clon.2023.01.012] [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: 08/08/2022] [Revised: 12/08/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023]
Abstract
AIMS Glioblastoma (GBM) is the most common primary malignant brain tumour in adults and frequently relapses. The aim of this study was to assess the efficacy and safety of metronomic temozolomide (TMZ) in the recurrent GBM population. MATERIALS AND METHODS All patients treated at our centre between September 2013 and March 2021 were retrospectively reviewed. The main inclusion criteria were first-line therapy with the Stupp protocol, relapse after the first or subsequent line of therapy, treatment with a metronomic TMZ schedule (50 mg/m2 continuously) and histological diagnosis of isocitrate dehydrogenase wild-type GBM according to World Health Organization 2016 classification. RESULTS In total, 120 patients were enrolled. The median follow-up was 15.6 months, the median age was 59 years, Eastern Cooperative Oncology Group performance status (ECOG-PS) was 0-2 in 107 patients (89%). O6-methylguanine-DNA-methyltransferase (MGMT) was methylated in 66 of 105 (62%) evaluable patients. The median number of prior lines of treatment was 2 (range 1-7). Three (2%) patients showed a partial response; 48 (40%) had stable disease; 69 (57%) had progressive disease. The median overall survival from the start of metronomic TMZ was 5.4 months (95% confidence interval 4.3-6.4), whereas the median progression-free survival (PFS) was 2.6 months (95% confidence interval 2.3-2.8). At univariate analysis, MGMT methylated and unmethylated patients had a median PFS of 2.9 and 2.1 months (P = 0.001) and a median overall survival of 5.6 and 4.4 months (P = 0.03), respectively. At multivariate analysis, the absence of MGMT methylation (hazard ratio = 2.3, 95% confidence interval 1.3-3.9, P = 0.004) and ECOG-PS ≤ 2 (hazard ratio = 0.5, 95% confidence interval 0.3-0.9, P = 0.017) remained significantly associated with PFS, whereas ECOG-PS ≤ 2 (hazard ratio = 0.4, 95% confidence interval 0.3-07, P = 0.001) was the only factor associated with overall survival. The most common grade 3-4 toxicities were haematological (lymphopenia 10%, thrombocytopenia 3%). CONCLUSIONS Rechallenge with metronomic TMZ is a well-tolerated option for recurrent GBM, even in pretreated patients. Patients with methylated MGMT disease and good ECOG-PS seem to benefit the most from this treatment.
Collapse
|
8
|
Trifănescu OG, Trifănescu RA, Mitrică R, Mitrea D, Ciornei A, Georgescu M, Butnariu I, Galeș LN, Șerbănescu L, Anghel RM, Păun MA. Upstaging and Downstaging in Gliomas-Clinical Implications for the Fifth Edition of the World Health Organization Classification of Tumors of the Central Nervous System. Diagnostics (Basel) 2023; 13:diagnostics13020197. [PMID: 36673007 PMCID: PMC9858599 DOI: 10.3390/diagnostics13020197] [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: 12/03/2022] [Revised: 12/28/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
In 2021, the 5th edition of the WHO Classification of Tumors of the Central Nervous System (WHO-CNS5) was published as the sixth volume of the international standard for brain and spinal cord tumor classification. The most remarkable practical change in the current classification involves grading gliomas according to molecular characterization. IDH mutant (10%) and IDH wild-type tumors (90%) are two different entities that possess unique biological features and various clinical outcomes regarding treatment response and overall survival. This article presents two comparative cases that highlight the clinical importance of these new classification standards. The first clinical case aimed to provide a comprehensive argument for determining the IDH status in tumors initially appearing as low-grade astrocytoma upon histologic examination, thus underlining the importance of the WHO-CNS5. The second case showed the implications of the histologic overdiagnosis of glioblastoma using the previous classification system with a treatment span of 7 years that proceeded through full-dose re-irradiation up to metronomic therapy. The new WHO-CNS5 classification significantly impacted complex neurooncological cases, thus changing the initial approach to a more precise therapeutic management.
Collapse
Affiliation(s)
- Oana Gabriela Trifănescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Raluca Alexandra Trifănescu
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- “C. I. Parhon” Bucharest Institute of Endocrinology, 011863 Bucharest, Romania
| | - Radu Mitrică
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: (R.M.); (D.M.); Tel.: +40-741964311 (R.M.); +40-723226233 (D.M.)
| | - Dan Mitrea
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Neuroaxis Neurology Clinic, 011302 Bucharest, Romania
- Correspondence: (R.M.); (D.M.); Tel.: +40-741964311 (R.M.); +40-723226233 (D.M.)
| | - Ana Ciornei
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai Georgescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Ioana Butnariu
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 041914 Bucharest, Romania
| | - Laurenția Nicoleta Galeș
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Medical Oncology II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Luiza Șerbănescu
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Maricela Anghel
- Department of Oncology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Mihai-Andrei Păun
- Radiotherapy II, “Prof. Dr. Al. Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
9
|
Abstract
Survival for patients with aggressive pituitary tumours (APT) and pituitary carcinomas (PC) has significantly improved following the increasing use of temozolomide (TMZ) since the first reports of response in 2006. TMZ was established as first line chemotherapy for APT/PC in the 2018 ESE guidelines on the management of APT/PC. There is no controversy over its use as salvage therapy however there is increasing interest in exploring TMZ use earlier in the treatment algorithm for APT/PC. Overall response rates as reported in systematic reviews are around 40% but stable disease in another 25% illustrates the clinical effectiveness of TMZ. Response is higher among functional compared to non-functional tumours. Where maximal radiation thresholds have not been reached in a patient, combination radiotherapy and TMZ appears more effective. Whether combination TMZ and capecitabine (CAPTEM) offers increased benefit remains uncertain particularly given added toxicity. O6-methyl guanine DNA methyl transferase (MGMT) status is important in determining response to treatment, although examination via immunohistochemistry versus PCR-based promoter-methylation analysis remains somewhat controversial. Optimal duration of TMZ treatment has still not been determined although longer treatment courses have been associated with increased progression-free survival. Treatment options following disease progression after TMZ remain unclear but include a second course of TMZ, immunotherapy and targeted oncological agents such as bevacizumab and lapatinib as well as peptide receptor radionuclide treatment (PRRT). An experienced pituitary multidisciplinary team is essential to all management decisions in patients with APT/PC.
Collapse
Affiliation(s)
- Ann McCormack
- Department of Endocrinology, St Vincent's Hospital, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
10
|
Barciszewska AM, Belter A, Gawrońska I, Giel-Pietraszuk M, Naskręt-Barciszewska MZ. Cross-reactivity between histone demethylase inhibitor valproic acid and DNA methylation in glioblastoma cell lines. Front Oncol 2022; 12:1033035. [PMID: 36465345 PMCID: PMC9709419 DOI: 10.3389/fonc.2022.1033035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/24/2022] [Indexed: 08/22/2023] Open
Abstract
Currently, valproic acid (VPA) is known as an inhibitor of histone deacetylase (epigenetic drug) and is used for the clinical treatment of epileptic events in the course of glioblastoma multiforme (GBM). Which improves the clinical outcome of those patients. We analyzed the level of 5-methylcytosine, a DNA epigenetic modulator, and 8-oxodeoxyguanosine, an cellular oxidative damage marker, affected with VPA administration, alone and in combination with temozolomide (TMZ), of glioma (T98G, U118, U138), other cancer (HeLa), and normal (HaCaT) cell lines. We observed the VPA dose-dependent changes in the total DNA methylation in neoplastic cell lines and the lack of such an effect in a normal cell line. VPA at high concentrations (250-500 μM) induced hypermethylation of DNA in a short time frame. However, the exposition of GBM cells to the combination of VPA and TMZ resulted in DNA hypomethylation. At the same time, we observed an increase of genomic 8-oxo-dG, which as a hydroxyl radical reaction product with guanosine residue in DNA suggests a red-ox imbalance in the cancer cells and radical damage of DNA. Our data show that VPA as an HDAC inhibitor does not induce changes only in histone acetylation, but also changes in the state of DNA modification. It shows cross-reactivity between chromatin remodeling due to histone acetylation and DNA methylation. Finally, total DNA cytosine methylation and guanosine oxidation changes in glioma cell lines under VPA treatment suggest a new epigenetic mechanism of that drug action.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
- Department of Neurosurgery and Neurotraumatology, Heliodor Swiecicki Clinical Hospital, Poznan, Poland
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Iwona Gawrońska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | | | |
Collapse
|
11
|
Śledzińska P, Bebyn M, Furtak J, Koper A, Koper K. Current and promising treatment strategies in glioma. Rev Neurosci 2022:revneuro-2022-0060. [PMID: 36062548 DOI: 10.1515/revneuro-2022-0060] [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] [Received: 05/18/2022] [Accepted: 07/30/2022] [Indexed: 12/14/2022]
Abstract
Gliomas are the most common primary central nervous system tumors; despite recent advances in diagnosis and treatment, glioma patients generally have a poor prognosis. Hence there is a clear need for improved therapeutic options. In recent years, significant effort has been made to investigate immunotherapy and precision oncology approaches. The review covers well-established strategies such as surgery, temozolomide, PCV, and mTOR inhibitors. Furthermore, it summarizes promising therapies: tumor treating fields, immune therapies, tyrosine kinases inhibitors, IDH(Isocitrate dehydrogenase)-targeted approaches, and others. While there are many promising treatment strategies, none fundamentally changed the management of glioma patients. However, we are still awaiting the outcome of ongoing trials, which have the potential to revolutionize the treatment of glioma.
Collapse
Affiliation(s)
- Paulina Śledzińska
- Molecular Oncology and Genetics Department, Innovative Medical Forum, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Marek Bebyn
- Molecular Oncology and Genetics Department, Innovative Medical Forum, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Jacek Furtak
- Department of Neurosurgery, 10th Military Research Hospital and Polyclinic, 85-681 Bydgoszcz, Poland.,Department of Neurooncology and Radiosurgery, The F. Lukaszczyk Oncology Center, 85-796 Bydgoszcz, Poland
| | - Agnieszka Koper
- Department of Oncology, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, 85-067 Bydgoszcz, Poland.,Department of Oncology, Franciszek Lukaszczyk Oncology Centre, 85-796 Bydgoszcz, Poland
| | - Krzysztof Koper
- Department of Oncology, Franciszek Lukaszczyk Oncology Centre, 85-796 Bydgoszcz, Poland.,Department of Clinical Oncology, and Nursing, Departament of Oncological Surgery, Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum, 85-067 Bydgoszcz, Poland
| |
Collapse
|
12
|
Pharmacokinetics of metronomic temozolomide in cerebrospinal fluid of children with malignant central nervous system tumors. Cancer Chemother Pharmacol 2022; 89:617-627. [PMID: 35355137 PMCID: PMC9054874 DOI: 10.1007/s00280-022-04424-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/13/2022] [Indexed: 11/24/2022]
Abstract
Purpose Although temozolomide is widely used in the treatment of childhood central nervous system (CNS) tumors, information on its pharmacokinetic profile in the brain or cerebrospinal fluid (CSF) is sparse. This study aimed at investigating whether measurable and clinically relevant concentrations of temozolomide are reached and maintained in CSF for continuous oral administration in pediatric patients. A population pharmacokinetic model was developed to quantify CSF penetration of temozolomide. Methods Eleven pediatric CNS tumor patients (aged 4–14 years) treated with oral temozolomide using a metronomic schedule (24–77 mg/m2/day) were included. Temozolomide concentrations in 28 plasma samples and 64 CSF samples were analyzed by high-performance liquid chromatography. Population pharmacokinetic modeling and simulations were performed using non-linear mixed effects modeling (NONMEM 7.4.2). Results Median temozolomide concentrations in plasma and CSF were 0.96 (range 0.24–5.99) µg/ml and 0.37 (0.06–1.76) µg/ml, respectively. A two-compartment model (central/plasma [1], CSF [2]) with first-order absorption, first-order elimination, and a transit compartment between CSF and plasma adequately described the data. Population mean estimates for clearance (CL) and the volume of distribution in the central compartment (Vc) were 3.29 L/h (95% confidence interval (CI) 2.58–3.95) and 10.5 L (8.17–14.32), respectively. Based on simulations, we found a median area under the concentration vs. time curve ratio (AUCCSF / AUCplasma ratio) of 37%. Conclusion Metronomic oral temozolomide penetrates into the CSF in pediatric patients, with even higher concentration levels compared to adults. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04424-4.
Collapse
|
13
|
Ortiz R, Perazzoli G, Cabeza L, Jiménez-Luna C, Luque R, Prados J, Melguizo C. Temozolomide: An Updated Overview of Resistance Mechanisms, Nanotechnology Advances and Clinical Applications. Curr Neuropharmacol 2021; 19:513-537. [PMID: 32589560 PMCID: PMC8206461 DOI: 10.2174/1570159x18666200626204005] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/22/2022] Open
Abstract
Temozolomide (TMZ), an oral alkylating prodrug which delivers a methyl group to purine bases of DNA (O6-guanine; N7-guanine and N3-adenine), is frequently used together with radiotherapy as part of the first-line treatment of high-grade gliomas. The main advantages are its high oral bioavailability (almost 100% although the concentration found in the cerebrospinal fluid was approximately 20% of the plasma concentration of TMZ), its lipophilic properties, and small size that confer the ability to cross the blood-brain barrier. Furthermore, this agent has demonstrated activity not only in brain tumors but also in a variety of solid tumors. However, conventional therapy using surgery, radiation, and TMZ in glioblastoma results in a median patient survival of 14.6 months. Treatment failure has been associated with tumor drug resistance. This phenomenon has been linked to the expression of O6-methylguanine-DNA methyltransferase, but the mismatch repair system and the presence of cancer stem-like cells in tumors have also been related to TMZ resistance. The understanding of these mechanisms is essential for the development of new therapeutic strategies in the clinical use of TMZ, including the use of nanomaterial delivery systems and the association with other chemotherapy agents. The aim of this review is to summarize the resistance mechanisms of TMZ and the current advances to improve its clinical use.
Collapse
Affiliation(s)
- Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| | | | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| | - Cristina Jiménez-Luna
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges 1066, Switzerland
| | - Raquel Luque
- Medical Oncology Service, Virgen de las Nieves Hospital, Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Biomedical Research Centre (CIBM), University of Granada, Spain
| |
Collapse
|
14
|
Fedele P, Sanna V, Fancellu A, Marino A, Calvani N, Cinieri S. De-escalating cancer treatments during COVID 19 pandemic: Is metronomic chemotherapy a reasonable option? Crit Rev Oncol Hematol 2021; 157:103148. [PMID: 33254036 PMCID: PMC7672334 DOI: 10.1016/j.critrevonc.2020.103148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 02/08/2023] Open
Abstract
COVID 19 pandemic represents an emergency for public health services and containment measures to reduce the risk of infection have been promptly activated worldwide. The healthcare systems reorganization has had a major impact on the management of cancer patients who are considered at high risk of infection. Recommendations and guidelines on how to manage cancer patients during COVID 19 pandemic have been published. Oral administration of chemotherapy is recommended to limit the access of cancer patients to hospital facilities and in some cases to guarantee the continuum of care. Low-dose metronomic administration of chemotherapy with different drugs and schedules has emerged in the last years as a possible alternative to conventional chemotherapy, due to its promising tumor control rates and excellent safety profiles. Moreover, given that many metronomic schedules use the oral route administration, it could represent a therapeutic strategy to ensure continuum of cancer care during COVID 19 pandemic. In this review we have selected all the clinical studies that have used the metronomic strategy, especially with oral drugs, in order to identify the subgroups of cancer patients who can benefit most from a metronomic approach even during COVID 19 pandemic.
Collapse
Affiliation(s)
- Palma Fedele
- Medical Oncology, Dario Camberlingo Hospital, Francavilla Fontana (Br), Italy.
| | - Valeria Sanna
- Medical Oncology, Hospital of Sassari, Sassari, Italy
| | - Alessandro Fancellu
- Department of Medical, Surgical and Experimental Sciences, Unit of General Surgery, University of Sassari, Sassari, Italy
| | - Antonella Marino
- Medical Oncology & Breast Unit, Antonio Perrino Hospital, Brindisi, Italy
| | - Nicola Calvani
- Medical Oncology & Breast Unit, Antonio Perrino Hospital, Brindisi, Italy
| | - Saverio Cinieri
- Medical Oncology & Breast Unit, Antonio Perrino Hospital, Brindisi, Italy
| |
Collapse
|
15
|
Neth BJ, Ruff MW, Uhm JH, Johnson DR, Divekar RD, Maddox DE. Temozolomide desensitization followed by metronomic dosing in patients with hypersensitivity. Cancer Chemother Pharmacol 2020; 86:375-382. [PMID: 32778906 DOI: 10.1007/s00280-020-04123-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/03/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE Temozolomide is the most effective chemotherapy for malignant glioma. Hypersensitivity requiring interruption of therapy may significantly impact patient survival. We have successfully employed temozolomide desensitization followed by metronomic dosing of temozolomide. Our purpose was to report patient characteristics and outcomes in patients with glioma (Grade 2-4) and temozolomide hypersensitivity managed by desensitization and metronomic dosing. METHODS We performed an observational study of 15 patients at Mayo Clinic (Rochester) with a diagnosis of glioma who underwent temozolomide desensitization with subsequent metronomic dosing from May 2012 to January 2017. We calculated overall and progression-free survival using the Kaplan-Meier method, and log-rank analyses to assess for differences in survival by WHO Grade or treatment initiation. RESULTS Median age at time of desensitization was 49.3 years (26.8-64.7 years). Median follow-up after desensitization was 35.5 months. One patient (6.7%) was unable to resume temozolomide due to recurrent allergy. The median time from first desensitization to discontinuation of metronomic temozolomide was 4.2 months (0-15.2 months). Median OS and PFS for the whole sample were 181.7 months and 44.9 months. For Grade 4, OS was 100% at 1 year, 40% at 3 years, 20% at 5 years; and PFS was 60% at 1 year, 40% at 3 years, and 20% at 5 years. CONCLUSION Our results suggest that rapid-desensitization followed by metronomic temozolomide should be considered in patients with glioma who experience hypersensitivity. This strategy provides comparable outcomes to therapy with standard protocols, with the majority of patients able to tolerate temozolomide after desensitization with favorable disease control.
Collapse
Affiliation(s)
- Bryan J Neth
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| | - Michael W Ruff
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Joon H Uhm
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Derek R Johnson
- Department of Neurology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Rohit D Divekar
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Daniel E Maddox
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Minniti G, Paolini S, Rea MLJ, Isidori A, Scaringi C, Russo I, Osti MF, Cavallo L, Esposito V. Stereotactic reirradiation with temozolomide in patients with recurrent aggressive pituitary tumors and pituitary carcinomas. J Neurooncol 2020; 149:123-130. [PMID: 32632895 DOI: 10.1007/s11060-020-03579-5] [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: 05/10/2020] [Accepted: 06/29/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To evaluate the efficacy of a second course of fractionated stereotactic radiotherapy (re-SRT) and temozolomide (TMZ) as salvage treatment option in patients with aggressive pituitary tumors (APTs) and pituitary carcinomas (PCs). PATIENTS AND METHODS Twenty-one patients with recurrent or progressive APTs (n = 17) and PCs (n = 4) who received combined TMZ and re-SRT, 36 Gy/18fractions or 37.5 Gy/15fractions, were retrospectively evaluated. TMZ was given at a dose of 75 mg/m2 given concurrently to re-SRT, and then 150-200 mg/m2/day for 5 days every 4 weeks or 50 mg/m2 daily for 12 months. Local control (LC) and overall survival (OS) were calculated from the time of re-SRT by Kaplan-Meier method. RESULTS With a median follow-up of 27 months (range 12-58 months), 2-year and 4-year LC rates were 73% and 65%, respectively; 2-year and 4-year survival rates were 82% and 66%, respectively. A complete response was achieved in 2 and partial response in 11 patients. Six patients recurred with a median time to progression of 14 months. O(6)-Methylguanine-DNA methyltransferase (MGMT) status and tumor volume emerged as prognostic factors. Grade 3 radiation-related toxicities occurred in 3 (14%) patients. Grade 2 or 3 hematologic toxicities during chemotherapy occurred in 8 (38%) patients. CONCLUSION Re-SRT and TMZ is a safe treatment offering high LC in patients with progressive APTs and PCs. The potential advantages of combined chemoradiation as up-front or salvage treatment need to be explored in prospective trials.
Collapse
Affiliation(s)
- Giuseppe Minniti
- Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, 53100, Siena, Italy. .,IRCCS Neuromed, Pozzilli, IS, Italy.
| | | | - Marie Lise Jaffrain Rea
- Biotechnological and Applied Clinical Sciences Department, University of L'Aquila, L'Aquila, Italy
| | - Andrea Isidori
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Scaringi
- UPMC Hillman Cancer Center, San Pietro Hospital FBF, Rome, Italy
| | - Ivana Russo
- UPMC Hillman Cancer Center, Villa Maria, Mirabella, AV, Italy
| | | | - Luigi Cavallo
- Division of Neurosurgery, Università degli Studi di Napoli "Federico II", Naples, Italy
| | | |
Collapse
|
17
|
Karachi A, Yang C, Dastmalchi F, Sayour EJ, Huang J, Azari H, Long Y, Flores C, Mitchell DA, Rahman M. Modulation of temozolomide dose differentially affects T-cell response to immune checkpoint inhibition. Neuro Oncol 2020; 21:730-741. [PMID: 30668768 DOI: 10.1093/neuonc/noz015] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The changes induced in host immunity and the tumor microenvironment by chemotherapy have been shown to impact immunotherapy response in both a positive and a negative fashion. Temozolomide is the most common chemotherapy used to treat glioblastoma (GBM) and has been shown to have variable effects on immune response to immunotherapy. Therefore, we aimed to determine the immune modulatory effects of temozolomide that would impact response to immune checkpoint inhibition in the treatment of experimental GBM. METHODS Immune function and antitumor efficacy of immune checkpoint inhibition were tested after treatment with metronomic dose (MD) temozolomide (25 mg/kg × 10 days) or standard dose (SD) temozolomide (50 mg/kg × 5 days) in the GL261 and KR158 murine glioma models. RESULTS SD temozolomide treatment resulted in an upregulation of markers of T-cell exhaustion such as LAG-3 and TIM-3 in lymphocytes which was not seen with MD temozolomide. When temozolomide treatment was combined with programmed cell death 1 (PD-1) antibody therapy, the MD temozolomide/PD-1 antibody group demonstrated a decrease in exhaustion markers in tumor infiltrating lymphocytes that was not observed in the SD temozolomide/PD-1 antibody group. Also, the survival advantage of PD-1 antibody therapy in a murine syngeneic intracranial glioma model was abrogated by adding SD temozolomide to treatment. However, when MD temozolomide was added to PD-1 inhibition, it preserved the survival benefit that was seen by PD-1 antibody therapy alone. CONCLUSION The peripheral and intratumoral immune microenvironments are distinctively affected by dose modulation of temozolomide.
Collapse
Affiliation(s)
- Aida Karachi
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Changlin Yang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Farhad Dastmalchi
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Elias J Sayour
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Jianping Huang
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Hassan Azari
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Yu Long
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Catherine Flores
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Duane A Mitchell
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| | - Maryam Rahman
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, UF Brain Tumor Immunotherapy Program, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, Florida
| |
Collapse
|
18
|
Revealing the epigenetic effect of temozolomide on glioblastoma cell lines in therapeutic conditions. PLoS One 2020; 15:e0229534. [PMID: 32101575 PMCID: PMC7043761 DOI: 10.1371/journal.pone.0229534] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Temozolomide (TMZ) is a drug of choice in glioblastoma treatment. Its therapeutic applications expand also beyond high grade gliomas. However, a significant number of recurrences and resistance to the drug is observed. The key factor in each chemotherapy is to achieve the therapeutic doses of a drug at the pathologic site. Nonetheless, the rate of temozolomide penetration from blood to cerebrospinal fluid is only 20–30%, and even smaller into brain intestinum. That makes a challenge for the therapeutic regimens to obtain effective drug concentrations with minimal toxicity and minor side effects. The aim of our research was to explore a novel epigenetic mechanism of temozolomide action in therapeutic conditions. We analyzed the epigenetic effects of TMZ influence on different glioblastoma cell lines in therapeutically achieved TMZ concentrations through total changes of the level of 5-methylcytosine in DNA, the main epigenetic marker. That was done with classical approach of radioactive nucleotide post-labelling and separation on thin-layer chromatography. In the range of therapeutically achieved temozolomide concentrations we observed total DNA hypomethylation. The significant hypermethylating effect was visible after reaching TMZ concentrations of 10–50 μM (depending on the cell line). Longer exposure time promoted DNA hypomethylation. The demethylated state of the glioblastoma cell lines was overcome by repeated TMZ applications, where dose-dependent increase in DNA 5-methylcytosine contents was observed. Those effects were not seen in non-cancerous cell line. The increase of DNA methylation resulting in global gene silencing and consecutive down regulation of gene expression after TMZ treatment may explain better glioblastoma patients’ survival.
Collapse
|
19
|
Faustino AC, Viani GA, Hamamura AC. Patterns of recurrence and outcomes of glioblastoma multiforme treated with chemoradiation and adjuvant temozolomide. Clinics (Sao Paulo) 2020; 75:e1553. [PMID: 32935821 PMCID: PMC7470430 DOI: 10.6061/clinics/2020/e1553] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 02/12/2020] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES To assess the patterns of failure and prognostic factors in Brazilian patients with glioblastoma multiforme (GBM) treated with radiotherapy (RT) and concurrent and adjuvant temozolomide (TMZ). METHODS Patients with diagnosed GBM post-resection received postoperative RT. TMZ was administered concurrently at 75 mg/m2/day for 28 consecutive days and adjuvant therapy at 150-200 mg/m2/day for 5 days every 28 days. Radiographic failure was defined as any new T1-enhancing lesion or biopsy-confirmed progressive enhancement inside of the radiation field. When possible, patients with recurrence were salvaged with metronomic TMZ, either in combination with a local treatment or alone (surgery or re-irradiation). Several prognostic factors were evaluated for overall survival (OS). Univariate and multivariate analyses were performed to identify significant factors. A p-value <0.05 was considered significant. RESULTS This study included 50 patients. The median follow-up time was 21 months. The median RT dose was 60 Gy and all patients received concomitant TMZ. During follow-up, 41 (83.6%) failures were observed, including 34 (83%) in-field, 4 (9.7%) marginal, and 3 (7.3%) distant failures. Metronomic TMZ was used as salvage treatment in 22 (44%) cases and in combination with local treatment in 12 (24%) cases. The median OS and progression-free survival times for the entire cohort were 17 and 9 months, respectively. In univariate analysis, the following factors were significant for better OS: maximal surgical resection (p=0.03), Karnofsky Performance Score (KPS)>70 at diagnosis (p=0.01), metronomic TMZ treatment (p=0.038), recursive partitioning analysis class III (p=0.03), and time to failure >9 months (p=0.0001). In multivariate analysis, the following factors remained significant for better OS: metronomic TMZ (p=0.01) and time to failure >9 months (p=0.0001). CONCLUSION The median OS of Brazilian patients with GBM treated with RT and TMZ was satisfactory. Although TMZ therapy has become the standard of care for patients with newly diagnosed GBM, the recurrence rate is extremely high. Metronomic TMZ as salvage treatment improved survival in these patients.
Collapse
|
20
|
Wick W, Krendyukov A, Junge K, Höger T, Fricke H. Longitudinal analysis of quality of life following treatment with Asunercept plus reirradiation versus reirradiation in progressive glioblastoma patients. J Neurooncol 2019; 145:531-540. [PMID: 31679112 PMCID: PMC6881251 DOI: 10.1007/s11060-019-03320-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/19/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Glioblastoma is an aggressive malignant cancer of the central nervous system, with disease progression associated with deterioration of neurocognitive function and quality of life (QoL). As such, maintenance of QoL is an important treatment goal. This analysis presents time to deterioration (TtD) of QoL in patients with recurrent glioblastoma receiving Asunercept plus reirradiation (rRT) or rRT alone. METHODS Data from patients with a baseline and ≥ 1 post-baseline QoL assessment were included in this analysis. TtD was defined as the time from randomisation to the first deterioration in the EORTC QLQ-C15, PAL EORTC QLQ-BN20 and Medical Research Council (MRC)-Neurological status. Deterioration was defined as a decrease of ≥ 10 points from baseline in the QLQ-C15 PAL overall QoL and functioning scales, an increase of ≥ 10 points from baseline in the QLQ-C15 PAL fatigue scale and the QLQ-BN20 total sum of score, and a rating of "Worse" in the MRC-Neurological status. Patients without a deterioration were censored at the last QoL assessment. Kaplan-Meier estimates were used to describe TtD and treatment groups (Asunercept + rRT or rRT alone) were compared using the log-rank test. RESULTS Treatment with Asunercept + rRT was associated with significant improvement of TtD compared with rRT alone for QLQ-CL15 PAL overall QoL and physical functioning, and MRC Neurological Status (p ≤ 0.05). In the Asunercept + rRT group, QoL was maintained beyond progresison of disease (PoD). CONCLUSION Treatment with Asunercept plus rRT significantly prolongs TtD and maintains QoL versus rRT alone in recurrent glioblastoma patients.
Collapse
Affiliation(s)
- Wolfgang Wick
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
21
|
Karachi A, Dastmalchi F, Mitchell DA, Rahman M. Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro Oncol 2019; 20:1566-1572. [PMID: 29733389 DOI: 10.1093/neuonc/noy072] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Temozolomide is the most widely used chemotherapy for patients with glioblastoma (GBM) despite the fact that approximately half of treated patients have temozolomide resistance and all patients eventually fail therapy. Due to the limited efficacy of existing therapies, immunotherapy is being widely investigated for patients with GBM. However, initial immunotherapy trials in GBM patients have had disappointing results as monotherapy. Therefore, combinatorial treatment strategies are being investigated. Temozolomide has several effects on the immune system that are dependent on mode of delivery and the dosing strategy, which may have unpredicted effects on immunotherapy. Here we summarize the immune modulating role of temozolomide alone and in combination with immunotherapies such as dendritic cell vaccines, T-cell therapy, and immune checkpoint inhibitors for patients with GBM.
Collapse
Affiliation(s)
- Aida Karachi
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| | - Farhad Dastmalchi
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| | - Duane A Mitchell
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| | - Maryam Rahman
- Lillian S. Wells Department of Neurosurgery, UF Brain Tumor Immunotherapy Program, University of Florida, Gainesville, Florida
| |
Collapse
|
22
|
Simsek C, Esin E, Yalcin S. Metronomic Chemotherapy: A Systematic Review of the Literature and Clinical Experience. JOURNAL OF ONCOLOGY 2019; 2019:5483791. [PMID: 31015835 PMCID: PMC6446118 DOI: 10.1155/2019/5483791] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/24/2018] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
Metronomic chemotherapy, continuous and dose-dense administration of chemotherapeutic drugs with lowered doses, is being evaluated for substituting, augmenting, or appending conventional maximum tolerated dose regimens, with preclinical and clinical studies for the past few decades. To date, the principle mechanisms of its action include impeding tumoral angiogenesis and modulation of hosts' immune system, affecting directly tumor cells, their progenitors, and neighboring stromal cells. Its better toxicity profile, lower cost, and easier use are main advantages over conventional therapies. The evidence of metronomic chemotherapy for personalized medicine is growing, starting with unfit elderly patients and also for palliative treatment. The literature reviewed in this article mainly demonstrates that metronomic chemotherapy is advantageous for selected patients and for certain types of malignancies, which make it a promising therapeutic approach for filling in the gaps. More clinical studies are needed to establish a solidified role for metronomic chemotherapy with other treatment models in modern cancer management.
Collapse
Affiliation(s)
- Cem Simsek
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | - Ece Esin
- Department of Medical Oncology, A.Y. Ankara Training Hospital, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Temozolomide is a first-line treatment for newly diagnosed glioblastoma. In this review, we will examine the use of temozolomide in other contexts for treating gliomas, including recurrent glioblastoma, glioblastoma in the elderly, diffuse low- and high-grade gliomas, non-diffuse gliomas, diffuse intrinsic pontine glioma (DIPG), ependymoma, pilocytic astrocytoma, and pleomorphic xanthoastrocytoma. RECENT FINDINGS Temozolomide improved survival in older patients with glioblastoma, anaplastic gliomas regardless of 1p/19q deletion status, and progressive ependymomas. Temozolomide afforded less toxicity and comparable efficacy to radiation in high-risk low-grade gliomas and to platinum-based chemotherapy in pediatric high-grade gliomas. The success of temozolomide in promoting survival has expanded beyond glioblastoma to benefit patients with non-glioblastoma tumors. Identifying practical biomarkers for predicting temozolomide susceptibility, and establishing complementary agents for chemosensitizing tumors to temozolomide, will be key next steps for future success.
Collapse
Affiliation(s)
- Jason Chua
- Department of Neurology, University of Michigan, 1500 E. Medical Center Dr., 1914 Taubman Center, Ann Arbor, MI, 48109, USA
| | - Elizabeth Nafziger
- Department of Neurology, University of Michigan, 1500 E. Medical Center Dr., 1914 Taubman Center, Ann Arbor, MI, 48109, USA
| | - Denise Leung
- Department of Neurology, University of Michigan, 1500 E. Medical Center Dr., 1914 Taubman Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
In vitro effect of chlorambucil on human glioma cell lines (SF767 and U87-MG), and human microvascular endothelial cell (HMVEC) and endothelial progenitor cells (ECFCs), in the context of plasma chlorambucil concentrations in tumor-bearing dogs. PLoS One 2018; 13:e0203517. [PMID: 30192852 PMCID: PMC6128565 DOI: 10.1371/journal.pone.0203517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
The objective of this study was to investigate a possible mechanism of action of metronomic chlorambucil on glioma by studying the in vitro cytotoxicity and anti-angiogenic effects on glioma and endothelial cells, respectively. The in vitro LD50 and IC50 of chlorambucil were determined using human SF767 and U87-MG glioma cell lines, human microvascular endothelial cells (HMVECs) and human endothelial colony forming cells (ECFCs). Results were analyzed in the context of chlorambucil concentrations measured in the plasma of tumor-bearing dogs receiving 4 mg m-2 metronomic chlorambucil. The LD50 and IC50 of chlorambucil were 270 μM and 114 μM for SF767, and 390 μM and 96 μM for U87-MG, respectively. The IC50 of chlorambucil was 0.53 μM and 145 μM for the HMVECs and ECFCs, respectively. In pharmacokinetic studies, the mean plasma Cmax of chlorambucil was 0.06 μM. Results suggest that metronomic chlorambucil in dogs does not achieve plasma concentrations high enough to cause direct cytotoxic or growth inhibitory effects on either glioma or endothelial cells.
Collapse
|
25
|
Kim SH, Yoo H, Chang JH, Kim CY, Chung DS, Kim SH, Park SH, Lee YS, Yang SH. Procarbazine and CCNU Chemotherapy for Recurrent Glioblastoma with MGMT Promoter Methylation. J Korean Med Sci 2018; 33:e167. [PMID: 29892208 PMCID: PMC5990446 DOI: 10.3346/jkms.2018.33.e167] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 04/13/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND While procarbazine, CCNU (lomustine), and vincristine (PCV) has been an alternative chemotherapy option for malignant gliomas, it is worth investigating whether the combination of only procarbazine and CCNU is comparable because vincristine adds toxicity with uncertain benefit. The purpose of this study was to evaluate the feasibility of procarbazine and CCNU chemotherapy for recurrent glioblastoma multiforme (GBM) with O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation. METHODS Eight patients with recurrent GBM following concurrent chemoradiotherapy and temozolomide (TMZ) adjuvant therapy were enrolled in this trial; they received no other chemotherapeutic agents or target therapy. They received CCNU (75 mg/m2) on day 1 and procarbazine (60 mg/m2) through days 11 and 24 every 4 weeks. The median cycle of CCNU and procarbazine was 3.5 (range: 2-6). RESULTS One patient achieved stable disease. The median progression-free survival (PFS) with procarbazine and CCNU chemotherapy was eight weeks (range: 5-73), and the PFS rates were 25% and 12.5% at 16 and 30 weeks, respectively. The median overall survival (OS) from the initial diagnosis to death was 40 months, and the median OS from the administration of procarbazine and CCNU chemotherapy to death was 9.7 months (95% confidence interval: 6.7-12.7). Serious adverse events were found at six visits, and two cases were considered to be grade 3 toxicities. CONCLUSION The efficacy of procarbazine and CCNU chemotherapy is not satisfactory. This study suggests the need to develop other treatment strategies for recurrent and TMZ-refractory GBM. Trial registry at ClinicalTrials.gov, NCT017337346.
Collapse
Affiliation(s)
- Se-Hyuk Kim
- Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea
| | - Heon Yoo
- Neuro-Oncology Clinic, National Cancer Center, Goyang, Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Chae-Yong Kim
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Sup Chung
- Department of Neurosurgery, The Catholic University of Korea, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Sung-Hae Park
- Department of Pathology, Seoul National University Hospital, Seoul, Korea
| | - Youn Soo Lee
- Department of Hospital Pathology, The Catholic University of Korea, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Ho Yang
- Department of Neurosurgery, The Catholic University of Korea, St. Vincent's Hospital, Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Suwon, Korea
| |
Collapse
|
26
|
Houy N, Le Grand F. Administration of temozolomide: Comparison of conventional and metronomic chemotherapy regimens. J Theor Biol 2018. [PMID: 29526662 DOI: 10.1016/j.jtbi.2018.02.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE We compare the Maximum Tolerated Dose (MTD) and Metronomic Chemotherapy (MC) protocols for temozolomide administration. We develop an innovative methodology for characterizing optimal chemotherapy regimens. METHODS We use a PK/PD model based on Faivre et al. (2013) for the pharmacokinetics of temozolomide, as well as the pharmacodynamics of its efficacy. For toxicity, which is measured by the nadir of the normalized absolute neutrophil count, we formalize the myelosuppression effect of temozolomide with the physiological model of Panetta et al. (2003b). We introduce a multi-criteria tool for comparing protocols along their efficacy and toxicity dimensions. RESULTS We show that the toxicity of the MC regimen proposed by Faivre et al. (2013) can greatly be reduced without affecting its efficacy, while the standard MTD protocol efficacy cannot be improved without impairing its toxicity. We also show that for any acceptable toxicity level, the optimal protocol remains closely related to standard MTD. CONCLUSIONS Overall, our new method enables a rich comparison between protocols along multiple dimensions. We can rank protocols for temozolomide administration. It is a first step toward building optimal individual protocols.
Collapse
Affiliation(s)
- Nicolas Houy
- University of Lyon, Lyon F-69007, France; CNRS, GATE Lyon Saint-Etienne F-69130, France.
| | - François Le Grand
- Emlyon Business School, Écully F-69130, France; ETH Zurich, Zurich CH-8092, Switzerland.
| |
Collapse
|
27
|
Imber BS, Kanungo I, Braunstein S, Barani IJ, Fogh SE, Nakamura JL, Berger MS, Chang EF, Molinaro AM, Cabrera JR, McDermott MW, Sneed PK, Aghi MK. Indications and Efficacy of Gamma Knife Stereotactic Radiosurgery for Recurrent Glioblastoma: 2 Decades of Institutional Experience. Neurosurgery 2017; 80:129-139. [PMID: 27428784 DOI: 10.1227/neu.0000000000001344] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 05/23/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The role of stereotactic radiosurgery (SRS) for recurrent glioblastoma and the radionecrosis risk in this setting remain unclear. OBJECTIVE To perform a large retrospective study to help inform proper indications, efficacy, and anticipated complications of SRS for recurrent glioblastoma. METHODS We retrospectively analyzed patients who underwent Gamma Knife SRS between 1991 and 2013. We used the partitioning deletion/substitution/addition algorithm to identify potential predictor covariate cut points and Kaplan-Meier and proportional hazards modeling to identify factors associated with post-SRS and postdiagnosis survival. RESULTS One hundred seventy-four glioblastoma patients (median age, 54.1 years) underwent SRS a median of 8.7 months after initial diagnosis. Seventy-five percent had 1 treatment target (range, 1-6), and median target volume and prescriptions were 7.0 cm 3 (range, 0.3-39.0 cm 3 ) and 16.0 Gy (range, 10-22 Gy), respectively. Median overall survival was 10.6 months after SRS and 19.1 months after diagnosis. Kaplan-Meier and multivariable modeling revealed that younger age at SRS, higher prescription dose, and longer interval between original surgery and SRS are significantly associated with improved post-SRS survival. Forty-six patients (26%) underwent salvage craniotomy after SRS, with 63% showing radionecrosis or mixed tumor/necrosis vs 35% showing purely recurrent tumor. The necrosis/mixed group had lower mean isodose prescription compared with the tumor group (16.2 vs 17.8 Gy; P = .003) and larger mean treatment volume (10.0 vs 5.4 cm 3 ; P = .009). CONCLUSION Gamma Knife may benefit a subset of focally recurrent patients, particularly those who are younger with smaller recurrences. Higher prescriptions are associated with improved post-SRS survival and do not seem to have greater risk of symptomatic treatment effect.
Collapse
Affiliation(s)
- Brandon S Imber
- University of California, San Francisco School of Medicine, San Francisco, California
| | | | - Steve Braunstein
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Igor J Barani
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Shannon E Fogh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Jean L Nakamura
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | | | | | | | | | | | - Penny K Sneed
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | | |
Collapse
|
28
|
Zhou Z, Howard TA, Villano JL. Long-term daily temozolomide with dose-dependent efficacy in MGMT promotor methylation negative recurrent high-grade astrocytoma. Cancer Chemother Pharmacol 2017; 80:1043-1046. [PMID: 28791452 DOI: 10.1007/s00280-017-3415-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/03/2017] [Indexed: 10/19/2022]
Abstract
Temozolomide (TMZ) for malignant gliomas is traditionally dosed in 5 out of a 28-day cycle, however alternative regimens exist, including dose-dense. Continuous daily dosing is available, but the acceptable dose and duration of therapy is unknown. We document a 40-year-old male with recurrent anaplastic astrocytoma, IDH mutant and MGMT promotor methylation negative, who has well-tolerated continuous daily TMZ for 20 months at 100 mg per day for nearly the length of this period. A trial at 80 mg per day demonstrated disease progression with response upon return to 100 mg per day. Prior to the daily TMZ, the patient underwent three surgical resections, radiation therapy with concurrent TMZ according to the EORTC-NCIC protocol, and subsequently bevacizumab in combination with use of the Optune device. Long-term survival of patients with recurrent malignant gliomas is uncommon, and currently no standard treatment strategies exist for these patients. We present this case to demonstrate the tolerability and dose dependency of prolonged daily TMZ dosing as a therapeutic option for recurrent anaplastic astrocytomas.
Collapse
Affiliation(s)
- Zhengqiu Zhou
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Tracy A Howard
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - John L Villano
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA. .,Departments of Medicine, Neurosurgery, and Neurology, University of Kentucky, 800 Rose St., CC446, Lexington, KY, 40536-0093, USA.
| |
Collapse
|
29
|
Altwairgi AK, Raja S, Manzoor M, Aldandan S, Alsaeed E, Balbaid A, Alhussain H, Orz Y, Lary A, Alsharm AA. Management and treatment recommendations for World Health Organization Grade III and IV gliomas. Int J Health Sci (Qassim) 2017; 11:54-62. [PMID: 28936153 PMCID: PMC5604271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The treatment recommendations provided in this manuscript are intended to serve as a knowledge base for clinicians and health personals involved in treating patients with high-grade malignant glioma. In newly diagnosed patients, complete resection or biopsy is required for histological characterization of the tumor, which in turn is essential to decide the treatment strategy. In patients with good or borderline performance score, radiotherapy (RT), and chemotherapy are the preferred management. In patients with poor performance score, RT with best possible supportive care is the mainstay of the management. All patients have to undergo brain magnetic resonance imaging procedure quarterly or half-yearly for 5 years and then on an annual basis. In patients with recurrent malignant glioma, wherever possible re-resection or re-irradiation or chemotherapy can be considered along with supportive and palliative care. High-grade malignant glioma should be managed in a multidisciplinary center with the best of the possible care that is available based on the evidence as discussed in this manuscript.
Collapse
Affiliation(s)
- Abdullah K. Altwairgi
- Department of Medical Oncology, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Shanker Raja
- Department of Medical Imaging, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Mohammed Manzoor
- Department of Medical Imaging, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Sadeq Aldandan
- Department of Pathology and Clinical Laboratory Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Eyad Alsaeed
- Department Radiation Oncology, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
- Department of Oncology, King Saud University, Riyadh, Saudi Arabia
| | - Ali Balbaid
- Department Radiation Oncology, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Hussain Alhussain
- Department Radiation Oncology, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Yassir Orz
- Department of Neurosurgery, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ahmed Lary
- Department of Neurosurgery, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Abdullah A. Alsharm
- Department of Medical Oncology, Comprehensive Cancer Center, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Hovey EJ, Field KM, Rosenthal MA, Barnes EH, Cher L, Nowak AK, Wheeler H, Sawkins K, Livingstone A, Phal P, Goh C, Simes J. Continuing or ceasing bevacizumab beyond progression in recurrent glioblastoma: an exploratory randomized phase II trial. Neurooncol Pract 2017; 4:171-181. [PMID: 31386014 DOI: 10.1093/nop/npw025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background In patients with recurrent glioblastoma, the benefit of bevacizumab beyond progression remains uncertain. We prospectively evaluated continuing or ceasing bevacizumab in patients who progressed while on bevacizumab. Methods CABARET, a phase II study, initially randomized patients to bevacizumab with or without carboplatin (Part 1). At progression, eligible patients underwent a second randomization to continue or cease bevacizumab (Part 2). They could also receive additional chemotherapy regimens (carboplatin, temozolomide, or etoposide) or supportive care. Results Of 120 patients treated in Part 1, 48 (80% of the anticipated 60-patient sample size) continued to Part 2. Despite randomization, there were some imbalances in patient characteristics. The best response was stable disease in 7 (30%) patients who continued bevacizumab and 2 (8%) patients who stopped receiving bevacizumab. There were no radiological responses. Median progression-free survival was 1.8 vs 2.0 months (bevacizumab vs no bevacizumab; hazard ratio [HR], 1.08; 95% CI, .59-1.96; P = .81). Median overall survival was 3.4 vs 3.0 months (HR, .84; 95% CI, .47-1.50; P = .56 and HR .70; 95% CI .38-1.29; P = .25 after adjustment for baseline factors). Quality-of-life scores did not significantly differ between arms. While the maximum daily steroid dose was lower in the continuation arm, the difference was not statistically significant. Conclusions Patients who continued bevacizumab beyond disease progression did not have clear survival improvements, although the study was not powered to detect other than very large differences. While these data provide the only randomized evidence related to continuing bevacizumab beyond progression in recurrent glioblastoma, the small sample size precludes definitive conclusions and suggests this remains an open question.
Collapse
Affiliation(s)
- Elizabeth J Hovey
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Kathryn M Field
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Mark A Rosenthal
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Elizabeth H Barnes
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Lawrence Cher
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Anna K Nowak
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Helen Wheeler
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Kate Sawkins
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Ann Livingstone
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Pramit Phal
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - Christine Goh
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | - John Simes
- Prince of Wales Hospital, Barker Street, Randwick, Sydney, NSW 2031, Australia (E.J.H.); University of New South Wales, Sydney, NSW 2052, Australia (E.J.H.); Royal Melbourne Hospital, Grattan Street, Parkville 3050, Melbourne Victoria, Australia (K.M.F., M.A.R., P.P., C.G.); Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Grattan Street Parkville 3052, Victoria, Australia (K.M.F., M.A.R.); National Health and Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Sydney, NSW 2006, Australia (E.H.B., K.S., A.L., J.S.); Austin Hospital, 145 Studley Road, Heidelberg, Melbourne, Victoria 3084, Australia (L.C.); Sir Charles Gairdner Hospital, Nedlands, Perth 6009, Western Australia (A.K.N.); Royal North Shore Hospital, St Leonards, Sydney, NSW 2065, Australia (H.W.)
| | | |
Collapse
|
31
|
Harper A, Blackwood L. Toxicity of metronomic cyclophosphamide chemotherapy in a UK population of cancer-bearing dogs: a retrospective study. J Small Anim Pract 2017; 58:227-230. [DOI: 10.1111/jsap.12635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/20/2016] [Accepted: 11/30/2016] [Indexed: 11/30/2022]
Affiliation(s)
- A. Harper
- Institute of Veterinary Sciences; University of Liverpool; Liverpool Wirral CH64 7TE UK
| | - L. Blackwood
- Institute of Veterinary Sciences; University of Liverpool; Liverpool Wirral CH64 7TE UK
| |
Collapse
|
32
|
Duma CM, Kim BS, Chen PV, Plunkett ME, Mackintosh R, Mathews MS, Casserly RM, Mendez GA, Furman DJ, Smith G, Oh N, Caraway CA, Sanathara AR, Dillman RO, Riley AS, Weiland D, Stemler L, Cannell R, Abrams DA, Smith A, Owen CM, Eisenberg B, Brant-Zawadzki M. Upfront boost Gamma Knife “leading-edge” radiosurgery to FLAIR MRI–defined tumor migration pathways in 174 patients with glioblastoma multiforme: a 15-year assessment of a novel therapy. J Neurosurg 2016; 125:40-49. [DOI: 10.3171/2016.7.gks161460] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVEGlioblastoma multiforme (GBM) is composed of cells that migrate through the brain along predictable white matter pathways. Targeting white matter pathways adjacent to, and leading away from, the original contrast-enhancing tumor site (termed leading-edge radiosurgery [LERS]) with single-fraction stereotactic radiosurgery as a boost to standard therapy could limit the spread of glioma cells and improve clinical outcomes.METHODSBetween December 2000 and May 2016, after an initial diagnosis of GBM and prior to or during standard radiation therapy and carmustine or temozolomide chemotherapy, 174 patients treated with radiosurgery to the leading edge (LE) of tumor cell migration were reviewed. The LE was defined as a region outside the contrast-enhancing tumor nidus, defined by FLAIR MRI. The median age of patients was 59 years (range 22–87 years). Patients underwent LERS a median of 18 days from original diagnosis. The median target volume of 48.5 cm3 (range 2.5–220.0 cm3) of LE tissue was targeted using a median dose of 8 Gy (range 6–14 Gy) at the 50% isodose line.RESULTSThe median overall survival was 23 months (mean 43 months) from diagnosis. The 2-, 3-, 5-, 7-, and 10-year actual overall survival rates after LERS were 39%, 26%, 16%, 10%, and 4%, respectively. Nine percent of patients developed treatment-related imaging-documented changes due to LERS. Nineteen percent of patients were hospitalized for management of edema, 22% for resection of a tumor cyst or new tumor bulk, and 2% for shunting to treat hydrocephalus throughout the course of their disease. Of the patients still alive, Karnofsky Performance Scale scores remained stable in 90% of patients and decreased by 1–3 grades in 10% due to symptomatic treatment-related imaging changes.CONCLUSIONSLERS is a safe and effective upfront adjunctive therapy for patients with newly diagnosed GBM. Limitations of this study include a single-center experience and single-institution determination of the LE tumor target. Use of a leading-edge calculation algorithm will be described to achieve a consistent approach to defining the LE target for general use. A multicenter trial will further elucidate its value in the treatment of GBM.
Collapse
Affiliation(s)
| | - Brian S. Kim
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Peter V. Chen
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Marianne E. Plunkett
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Ralph Mackintosh
- 2Cancer Center, and
- 3Department of Radiation Oncology, Hoag Memorial Hospital Presbyterian, Newport Beach
| | - Marlon S. Mathews
- 4Department of Neurosurgery, University of California, Irvine, Orange; and
| | | | | | | | | | - Nathan Oh
- 1Neurosciences Institute,
- 5Department of Neurosurgery, Loma Linda University Health, Loma Linda, California
| | | | | | | | | | | | | | | | | | - Alexa Smith
- 4Department of Neurosurgery, University of California, Irvine, Orange; and
| | | | | | | |
Collapse
|
33
|
Wong ET, Timmons J, Callahan A, O'Loughlin L, Giarusso B, Alsop DC. Phase I study of low-dose metronomic temozolomide for recurrent malignant gliomas. BMC Cancer 2016; 16:914. [PMID: 27876012 PMCID: PMC5120517 DOI: 10.1186/s12885-016-2945-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 11/09/2016] [Indexed: 02/08/2023] Open
Abstract
Background The treatment goal for recurrent malignant gliomas centers on disease stabilization while minimizing therapy-related side effects. Metronomic dosing of cytotoxic chemotherapy has emerged as a promising option to achieve this objective. Methods This phase I study was performed using metronomic temozolomide (mTMZ) at 25 or 50 mg/m2/day continuously in 42-day cycles. Correlative studies were incorporated using arterial spin labeling MRI to assess tumor blood flow, analysis of matrix metalloproteinase-2 (MMP-2) and MMP-9 activities in the cerebrospinal fluid (CSF) as surrogates for tumor angiogenesis and invasion, as well as determination of CSF soluble interleukin-2 receptor alpha (sIL-2Rα) levels as a marker of immune modulation. Results Nine subjects were enrolled and toxicity consisted of primarily grade 1 or 2 hematological and gastrointestinal side effects; only one patient had a grade 3 elevated liver enzyme level that was reversible. Tumor blood flow was variable across subjects and time, with two experiencing a transient increase before a decrease to below baseline level while one exhibited a gradual drop in blood flow over time. MMP-2 activity correlated with overall survival but not with progression free survival, while MMP-9 activity did not correlate with either outcome parameters. Baseline CSF sIL-2Rα level was inversely correlated with time from initial diagnosis to first progression, suggesting that subjects with higher sIL-2Rα may have more aggressive disease. But they lived longer when treated with mTMZ, probably due to drug-related changes in T-cell constituency. Conclusions mTMZ possesses efficacy against recurrent malignant gliomas by altering blood flow, slowing invasion and modulating antitumor immune function.
Collapse
Affiliation(s)
- Eric T Wong
- Brain Tumor Center & Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA.
| | - Joshua Timmons
- Brain Tumor Center & Neuro-Oncology Unit, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Amy Callahan
- MRI Research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Lauren O'Loughlin
- MRI Research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - Bridget Giarusso
- MRI Research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| | - David C Alsop
- MRI Research, Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, Massachusetts, 02215, USA
| |
Collapse
|
34
|
Hepatotoxicity by combination treatment of temozolomide, artesunate and Chinese herbs in a glioblastoma multiforme patient: case report review of the literature. Arch Toxicol 2016; 91:1833-1846. [DOI: 10.1007/s00204-016-1810-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/04/2016] [Indexed: 10/21/2022]
|
35
|
Sun C, Yu Y, Wang L, Wu B, Xia L, Feng F, Ling Z, Wang S. Additive antiangiogenesis effect of ginsenoside Rg3 with low-dose metronomic temozolomide on rat glioma cells both in vivo and in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:32. [PMID: 26872471 PMCID: PMC4752767 DOI: 10.1186/s13046-015-0274-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/17/2015] [Indexed: 11/23/2022]
Abstract
Background Glioblastoma is the most common and deadly primary brain tumor in adults. Low-dose,metronomic (LDM) temozolomide (TMZ) displays improved efficacy in the treatment of glioblastoma by targeting angiogenesis, but has a limited effect on recurrence. The antiangiogenesis drug ginsenoside Rg3 (RG3) is the main active ingredient of ginseng, a popular herbal medicine. Methods Using an in vitro and a rat model of an orthotopic glioma allograft, this study was to determine whether RG3 enhanced the antiangiogenesis activity of LDM TMZ in the treatment of glioblastoma. Results Our results showed that combined use of TMZ with RG3 displayed additive inhibition on proliferation of both human umbilical vein endothelial cells (HUVEC) and rat C6 glioma cells in vitro. They additively arrested cell cycle, increased apoptosis, and decreased VEGF-A and BCL-2 expression in HUVEC. Antiangiogenesis effect was also evaluated in the rat model of orthotopic glioma allograft, based upon markers including relative cerebral blood volume (rCBV) by magnetic resonance imaging (MRI), VEGF levels and microvessel density (MVD)/CD34 staining. LDM TMZ alone was potent in suppressing angiogenesis and tumor growth, whereas RG3 alone only had modest antiangiogenesis effects. Combined treatment significantly and additively suppressed angiogenesis, without additive inhibitory effects on allografted tumor growth. Conclusions These data provide evidence showing the efficacy of LDM TMZ on glioma treatment. The combined additive antiangiogenesis effect suggests that RG3 has the potential to further increase the efficacy of LDM TMZ in the treatment of glioblastoma. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0274-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caixing Sun
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Yang Yu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Lizhen Wang
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Bin Wu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Liang Xia
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Fang Feng
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Zhiqiang Ling
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, Zhejiang, 310022, China.
| | - Shihua Wang
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, NC, 27157, USA.
| |
Collapse
|
36
|
Therapeutic options in recurrent glioblastoma--An update. Crit Rev Oncol Hematol 2016; 99:389-408. [PMID: 26830009 DOI: 10.1016/j.critrevonc.2016.01.018] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/01/2016] [Accepted: 01/19/2016] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Standards of care are not yet defined in recurrent glioblastoma. METHODS We reviewed the literature on clinical trials for recurrent glioblastoma available in PubMed and American Society of Clinical Oncology (ASCO) abstracts until June 2015. RESULTS Evidence is limited due to the paucity of randomized controlled studies. Second surgery or re-irradiation are options for selected patients. Alkylating chemotherapy such as nitrosoureas or temozolomide and the vascular endothelial growth factor (VEGF) antibody, bevacizumab, exhibit comparable single agent activity. Phase III data exploring the benefit of combining bevacizumab and lomustine are emerging. Novel approaches in the fields of targeted therapy, immunotherapy, and tumor metabolism are coming forward. Several biomarkers are being explored, but, except for O(6)-methylguanine DNA methyltransferase (MGMT) promoter methylation, none has assumed a role in clinical practice. CONCLUSION Proper patient selection, development of predictive biomarkers and randomized controlled studies are required to develop evidence-based concepts for recurrent glioblastoma.
Collapse
|
37
|
Joo JD, Kim H, Kim YH, Han JH, Kim CY. Validation of the Effectiveness and Safety of Temozolomide during and after Radiotherapy for Newly Diagnosed Glioblastomas: 10-year Experience of a Single Institution. J Korean Med Sci 2015; 30:1597-603. [PMID: 26539003 PMCID: PMC4630475 DOI: 10.3346/jkms.2015.30.11.1597] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/14/2015] [Indexed: 11/20/2022] Open
Abstract
This study was performed to validate the effectiveness and safety of concurrent chemoradiotherapy and adjuvant therapy with temozolomide for newly diagnosed glioblastoma multiforme as a standard treatment protocol. Between 2004 and 2011, patients newly diagnosed with glioblastoma who were treated with temozolomide during concurrent chemoradiotherapy and adjuvant chemotherapy were included from a single institution and analyzed retrospectively. The primary endpoint was overall survival, and the secondary endpoints were progression-free survival, response, and safety. A total of 71 patients were enrolled in this study. The response rate was 41% (29/71), and the tumor control rate was 80% (57/71). In the 67 patients who completed the concurrent chemoradiotherapy with temozolomide, the median overall survival was 19 months and the 1- and 2-yr overall survival rates were 78.3% and 41.7%, respectively. The median progression free survival was 9 months, and the 1- and 2-yr progression free survival rates were 33.8% and 14.3%, respectively. The mean duration of survival after progression of disease in salvage treatment group was 11.9 (1.3-53.2) months. Concurrent chemoradiotherapy with temozolomide resulted in grade 3 or 4 hematologic toxic effects in 2.8% of the patients. The current protocol of temozolomide during and after radiation therapy is both effective and safe and is still appropriate as the standard protocol for treatment of glioblastoma. An active salvage treatment might be required for a better prognosis.
Collapse
Affiliation(s)
- Jin-Deok Joo
- Department of Neurosurgery, Seoul National University College of Medicine, Seongnam, Korea
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hansol Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seongnam, Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seongnam, Korea
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jung Ho Han
- Department of Neurosurgery, Seoul National University College of Medicine, Seongnam, Korea
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Chae-Yong Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seongnam, Korea
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
38
|
Abstract
Current first-line treatment regimens combine surgical resection and chemoradiation for Glioblastoma that provides a slight increase in overall survival. Age on its own should not be used as an exclusion criterion of glioblastoma multiforme (GBM) treatment, but performance should be factored heavily into the decision-making process for treatment planning. Despite aggressive initial treatment, most patients develop recurrent diseases which can be treated with re-resection, systemic treatment with targeted agents or cytotoxic chemotherapy, reirradiation, or radiosurgery. Research into novel therapies is investigating alternative temozolomide regimens, convection-enhanced delivery, immunotherapy, gene therapy, antiangiogenic agents, poly ADP ribose polymerase inhibitors, or cancer stem cell signaling pathways. Given the aggressive and resilient nature of GBM, continued efforts to better understand GBM pathophysiology are required to discover novel targets for future therapy.
Collapse
Affiliation(s)
- Sanjoy Roy
- Department of Radiotherapy, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Debarshi Lahiri
- Department of Radiotherapy, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Tapas Maji
- Department of Radiotherapy, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Jaydip Biswas
- Department of Radiotherapy, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| |
Collapse
|
39
|
Wei W, Chen X, Ma X, Wang D, Guo Z. The efficacy and safety of various dose-dense regimens of temozolomide for recurrent high-grade glioma: a systematic review with meta-analysis. J Neurooncol 2015; 125:339-49. [PMID: 26334318 DOI: 10.1007/s11060-015-1920-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 08/29/2015] [Indexed: 12/30/2022]
Abstract
The goal of this meta-analysis was to identify the temozolomide (TMZ) regimen with optimal efficacy and tolerance for treatment of recurrent high-grade glioma (HGG). The PubMed and EMBASE databases were searched from the earliest records to February 2015, which identified 33 studies with 1760 participants that met the inclusion criteria. The standard schedule and three most common dose-dense regimens of TMZ therapy for recurrent HGG were included in this meta-analysis. The schedule of 7 days on/7 days off for the treatment of grade IV gliomas was significantly superior to the standard regimen with respect to progression-free survival at 6 months (34.8 %; 95 % confidence interval (CI) 27.0-43.4 %) and 12 months (15.5 %; 95 % CI 10.7-21.8 %). For grade III gliomas, this regimen conveyed a significantly greater overall survival (OS) rate at 12 months (79.0 %; 95 % CI 56.2-91.7 %), as compared to the standard schedule. Also, the 21 days on/7 days off regimen had significantly longer OS rates at 6 months (73.6 %; 95 % CI 63.4-81.8 %) and 12 months (40.6 %; 95 % CI 32.6-48.6 %) than the standard regimen for grade IV gliomas. In addition, the standard schedule showed a significantly higher clinical benefit rate than the 7 days on/7 days off and 21 days on/7 days off regimens. However, the grade 3-4 toxicity rate of lymphopenia of the standard schedule was 76.5 % (95 % CI 45.5-92.7 %), which was the highest among the four regimens. Recurrent HGG patients receiving personalized treatment should be closely followed up, especially those with concurrent hematological diseases.
Collapse
Affiliation(s)
- Wei Wei
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Xin Chen
- Department of Clinical Epidemiology, and Center of Evidence Based Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ximeng Ma
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Dawei Wang
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Zongze Guo
- Department of Neurosurgery, The First Hospital of China Medical University, No. 155 Nanjing Bei Street, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
40
|
Ringel F, Pape H, Sabel M, Krex D, Bock HC, Misch M, Weyerbrock A, Westermaier T, Senft C, Schucht P, Meyer B, Simon M. Clinical benefit from resection of recurrent glioblastomas: results of a multicenter study including 503 patients with recurrent glioblastomas undergoing surgical resection. Neuro Oncol 2015; 18:96-104. [PMID: 26243790 DOI: 10.1093/neuonc/nov145] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND While standards for the treatment of newly diagnosed glioblastomas exist, therapeutic regimens for tumor recurrence remain mostly individualized. The role of a surgical resection of recurrent glioblastomas remains largely unclear at present. This study aimed to assess the effect of repeated resection of recurrent glioblastomas on patient survival. METHODS In a multicenter retrospective-design study, patients with primary glioblastomas undergoing repeat resections for recurrent tumors were evaluated for factors affecting survival. Age, Karnofsky performance status (KPS), extent of resection (EOR), tumor location, and complications were assessed. RESULTS Five hundred and three patients (initially diagnosed between 2006 and 2010) undergoing resections for recurrent glioblastoma at 20 institutions were included in the study. The patients' median overall survival after initial diagnosis was 25.0 months and 11.9 months after first re-resection. The following parameters were found to influence survival significantly after first re-resection: preoperative and postoperative KPS, EOR of first re-resection, and chemotherapy after first re-resection. The rate of permanent new deficits after first re-resection was 8%. CONCLUSION The present study supports the view that surgical resections of recurrent glioblastomas may help to prolong patient survival at an acceptable complication rate.
Collapse
Affiliation(s)
- Florian Ringel
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Haiko Pape
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Michael Sabel
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Dietmar Krex
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Hans Christoph Bock
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Martin Misch
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Astrid Weyerbrock
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Thomas Westermaier
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Christian Senft
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Philippe Schucht
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Bernhard Meyer
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | - Matthias Simon
- Department of Neurosurgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany (F.R., H.P., B.M.); Department of Neurosurgery, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany (M.S.); Department of Neurosurgery, Universitätsklinikum Carl Gustav Carus, Dresden, Germany (D.K.); Department of Neurosurgery, Universitätsmedizin Göttingen, , Göttingen, Germany (H.C.B.); Department of Neurosurgery, Charité Universitätsmedizin, Berlin, Germany (M.M.); Department of Neurosurgery, Universitätsklinikum Freiburg, Freiburg, Germany (A.W.); Department of Neurosurgery, Universitätsklinikum Würzburg, Würzburg, Germany (T.W.); Department of Neurosurgery, Goethe Universität Frankfurt, Frankfurt, Germany (C.S.); Department of Neurosurgery, Inselspital, Universitätsklinikum Bern, Bern, Switzerland (P.S.); Department of Neurosurgery, Universitätskliniken Bonn, Rheinische Friedrich Wilhelms Universität, Bonn, Germany (M.S.)
| | | |
Collapse
|
41
|
Metronomic chemotherapy with daily low-dose temozolomide and celecoxib in elderly patients with newly diagnosed glioblastoma multiforme: a retrospective analysis. J Neurooncol 2015; 124:265-73. [DOI: 10.1007/s11060-015-1834-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 05/30/2015] [Indexed: 10/23/2022]
|
42
|
A phase II study of feasibility and toxicity of bevacizumab in combination with temozolomide in patients with recurrent glioblastoma. Clin Transl Oncol 2015; 17:743-50. [PMID: 26033428 DOI: 10.1007/s12094-015-1304-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/16/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE The aim of this prospective and multicentric phase II study was to evaluate the efficacy and safety of temozolomide (TMZ) and bevacizumab (BV) in patients (pts) with recurrent glioblastoma (GB), previously treated with chemoradiotherapy and at least three cycles of adjuvant TMZ. PATIENTS AND METHODS Patients with GB at first relapse received BV 10 mg/kg day every 2 weeks and TMZ 150 mg/m(2) days 1-7 and 15-21, every 28 days. Patients underwent brain magnetic resonance imaging every 8 weeks. RESULTS Thirty-two evaluable pts were recruited in 8 sites. Fourteen pts (44%) had gross total resection. O(6)-methylguanine-DNA methyltransferase (MGMT) promoter was methylated in 12 pts, unmethylated in 6 pts, and missing in 14 pts. The estimated 6-month progression free survival (PFS) rate was 21.9% (95% CI 9.3-40.0%). The median PFS and overall survival (OS) were 4.2 months (95% CI 3.6-5.4 months) and 7.3 months (95% CI 5.8-8.8 months), respectively. No significant association with MGMT status was found in terms of OS or PFS. Six of 32 pts (19%; 95% CI 7.2-36.4) were long-term survivors, with a median PFS and OS (50% events) of 9.5 months (95% CI 7.9-23.6) and 15.4 (95% CI 8.9-NA), respectively: no differences in baseline characteristics were identified in comparison with total population. No unexpected toxicities or treatment-related deaths were observed. CONCLUSIONS This regimen showed to be feasible and well tolerated in pts with recurrent GB pretreated with TMZ. Further investigation is warranted to identify subpopulations that are more likely to benefit from addition of BV to GB therapy.
Collapse
|
43
|
Hashimoto N, Tsuboi A, Kagawa N, Chiba Y, Izumoto S, Kinoshita M, Kijima N, Oka Y, Morimoto S, Nakajima H, Morita S, Sakamoto J, Nishida S, Hosen N, Oji Y, Arita N, Yoshimine T, Sugiyama H. Wilms tumor 1 peptide vaccination combined with temozolomide against newly diagnosed glioblastoma: safety and impact on immunological response. Cancer Immunol Immunother 2015; 64:707-16. [PMID: 25772149 PMCID: PMC11028974 DOI: 10.1007/s00262-015-1674-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 02/25/2015] [Indexed: 11/28/2022]
Abstract
To investigate the safety of combined Wilms tumor 1 peptide vaccination and temozolomide treatment of glioblastoma, a phase I clinical trial was designed. Seven patients with histological diagnosis of glioblastoma underwent concurrent radiotherapy and temozolomide therapy. Patients first received Wilms tumor 1 peptide vaccination 1 week after the end of combined concurrent radio/temozolomide therapy, and administration was continued once per week for 7 weeks. Temozolomide maintenance was started and performed for up to 24 cycles, and the observation period for safety encompassed 6 weeks from the first administration of maintenance temozolomide. All patients showed good tolerability during the observation period. Skin disorders, such as grade 1/2 injection-site reactions, were observed in all seven patients. Although grade 3 lymphocytopenia potentially due to concurrent radio/temozolomide therapy was observed in five patients (71.4 %), no other grade 3/4 hematological or neurological toxicities were observed. No autoimmune reactions were observed. All patients are still alive, and six are on Wilms tumor 1 peptide vaccination without progression, yielding a progression-free survival from histological diagnosis of 5.2-49.1 months. Wilms tumor 1 peptide vaccination was stopped in one patient after 12 injections by the patient's request. The safety profile of the combined Wilms tumor 1 peptide vaccination and temozolomide therapy approach for treating glioblastoma was confirmed.
Collapse
Affiliation(s)
- Naoya Hashimoto
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Incorporation of biomarkers in phase II studies of recurrent glioblastoma. Tumour Biol 2014; 36:153-62. [PMID: 25534238 DOI: 10.1007/s13277-014-2960-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/05/2014] [Indexed: 01/15/2023] Open
Abstract
The survival trends for glioblastoma (GBM) patients have remained largely static, reflecting a lack of improvement in the therapeutic options for patients. Less than 5 % of newly diagnosed GBM survives more than 5 years. Tumor relapse is nearly universal and the majority of patients do not respond to further systemic therapy. The results from phase II studies conducted with recurrent GBM patients have not translated to successful confirmatory studies and thus we have reached a significant roadblock in the development of new treatments for patients with recurrent GBM. The development of new, active, and potentially targeted drugs for the treatment of recurrent GBM represents a major unmet need. The incorporation of diagnostic/companion biomarker combinations into the phase II studies and appropriate stratification of the patients is lagging significantly behind other larger cancer groups such as breast, non-small cell lung cancer, and melanoma. We herein carried out a systematic review of the phase II clinical studies conducted in patients with recurrent GBM (2010-2013 inclusive) to assess the degree of biomarker incorporation within the clinical trial design.
Collapse
|
46
|
Abstract
Glioblastoma (GBM) has proven to be incurable despite recent progress on its standard of care using temozolomide (TMZ) as the main trunk of initial therapy for newly diagnosed GBM. One of the main reasons accounting for the dismal prognosis is attributed to lack of active therapeutic regimens at recurrence. Since TMZ is the most active cytotoxic agent against GBM, and the standard dosing of TMZ has shown favorable safety profile in clinical trials, re-challenge with TMZ in increased dose density schedules for recurrent tumors that have evaded from prior standard TMZ therapy appears to be a rational approach and has been intensively exploited. A number of phase II clinical trials using different alternating scheduling of dose-dense TMZ (ddTMZ) have shown superior efficacy over the standard TMZ or historical controls with other alkylating agents including nitrosoureas and procarbazine. One ddTMZ schedule, consisting of a 21-days on/7-days off regimen was applied to newly-diagnosed GBM as the adjuvant monotherapy after completion of combined radiation and TMZ and failed to demonstrate survival benefit in a large phase III trial (RTOG 0525). Thus its role in TMZ-pretreated, recurrent GBM should be carefully pursuit in randomized trials, e.g., planned JCOG 1308 trial comparing a 7-days on/7-days off ddTMZ regimen used upfront at the first relapse followed by bevacizumab on progression versus bevacizumab alone, investigating whether insertion of ddTMZ prior to bevacizumab could bestow better outcome in the recurrent setting. In this article, mode of action, past trials, and future directions of ddTMZ therapy are discussed.
Collapse
|
47
|
Jung TY, Lee JY, Kim DS, Park HJ, Kim CY, Ra YS, Lee MJ, Kim SH, Baek HJ, Kim IH, Park KD, Kim SK. Pediatric supratentorial high-grade glioma: multicenter retrospective observational study of the Korean Society for Pediatric Neuro-Oncology. J Neurooncol 2014; 121:413-9. [PMID: 25366367 DOI: 10.1007/s11060-014-1653-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/26/2014] [Indexed: 01/05/2023]
Abstract
We analyzed the prognostic factors of Korean pediatric patients with supratentorial high-grade glioma (HGG). Between 1997 and 2011, 62 patients with 34 glioblastomas and 28 anaplastic gliomas were surgically operated at nine institutions. The male-to-female ratio was 33 to 29 and the median age was 12 years (range 1-18). The prognostic significance of tumor location, extent of removal, pathologic grade, treatment method, and pattern of recurrence was analyzed. The median progression-free survival (PFS) and overall survival (OS) were 9.3 (± 0.8) and 17.8 (± 1.9) months, respectively. Glioblastoma and anaplastic glioma showed OSs of 15.9 (± 1.3) and 19.6 (± 2.4) months, respectively. Based on the univariate analysis, gross total removal (GTR) and initial combined chemoradiotherapy improved PFS (p = 0.012 and p = 0.003) and OS (p = 0.030 and p = 0.013), respectively. Cerebrospinal fluid (CSF) dissemination showed poor OS (p = 0.001). Based on the multivariate analysis, GTR and initial combined chemoradiotherapy resulted in an improved PFS [(hazard ratio 0.360; 95 % CI 0.177-0.733; p = 0.005) and (hazard ratio 0.458; 95 % CI 0.230-0.911; p = 0.026), respectively]. GTR, initial combined chemoradiotherapy, and no CSF seeding resulted in an improved OS [(hazard ratio 0.417; 95 % CI 0.201-0.861; p = 0.018), (hazard ratio 0.406; 95 % CI 0.206-0.800; p = 0.009), and (hazard ratio 0.288; 95 % CI 0.148-0.563; p = 0.000), respectively]. No significant difference in PFS and OS was observed between glioblastoma and anaplastic glioma. CSF dissemination was observed in 22 patients (35.5 %) during total follow-up. Pediatric anaplastic glioma showed poor survival, similarly to glioblastoma. GTR and initial combined chemoradiotherapy were associated with improved survival.
Collapse
Affiliation(s)
- Tae-Young Jung
- Department of Neurosurgery, Chonnam National University Hwasun Hospital, Gwangju, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Schuessler A, Smith C, Beagley L, Boyle GM, Rehan S, Matthews K, Jones L, Crough T, Dasari V, Klein K, Smalley A, Alexander H, Walker DG, Khanna R. Autologous T-cell therapy for cytomegalovirus as a consolidative treatment for recurrent glioblastoma. Cancer Res 2014; 74:3466-76. [PMID: 24795429 DOI: 10.1158/0008-5472.can-14-0296] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive human brain malignancies. Even with optimal treatment, median survival is less than 6 months for patients with recurrent GBM. Immune-based therapies have the potential to improve patient outcome by supplementing standard treatment. Expression of human cytomegalovirus (CMV) antigens in GBM tissues provides the unique opportunity to target viral antigens for GBM therapy. Here, we report findings of a formal clinical assessment of safety and potential clinical efficacy of autologous CMV-specific T-cell therapy as a consolidative treatment for recurrent GBM. From a total of 19 patients with recurrent GBM, CMV-specific T cells were successfully expanded from 13 patients (68.4%), 11 of whom received up to four T-cell infusions. Combination therapy based on T-cell infusion and chemotherapy was well tolerated, and we detected only minor adverse events. The overall survival of these patients since first recurrence ranged from 133 to 2,428 days, with a median overall survival of 403 days. Most importantly, 4 of 10 patients that completed the treatment remained progression free during the study period. Furthermore, molecular profiling of CMV-specific T-cell therapy from these patients revealed distinct gene expression signatures, which correlated with their clinical response. Our study suggests that a combination therapy with autologous CMV-specific T cells and chemotherapy is a safe novel treatment option and may offer clinical benefit for patients with recurrent GBM.
Collapse
Affiliation(s)
- Andrea Schuessler
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Corey Smith
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Leone Beagley
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | | | - Sweera Rehan
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Katherine Matthews
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Linda Jones
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Tania Crough
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Vijayendra Dasari
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | | | - Amy Smalley
- Newro Foundation, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Hamish Alexander
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory; Newro Foundation, The Wesley Hospital, Brisbane, Queensland, Australia
| | - David G Walker
- Newro Foundation, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory;
| |
Collapse
|
49
|
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]
|
50
|
Han SJ, Rolston JD, Molinaro AM, Clarke JL, Prados MD, Chang SM, Berger MS, DeSilva A, Butowski NA. Phase II trial of 7 days on/7 days off temozolmide for recurrent high-grade glioma. Neuro Oncol 2014; 16:1255-62. [PMID: 24670608 DOI: 10.1093/neuonc/nou044] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND A phase II trial was performed to evaluate the efficacy of a dose-dense, 7 days on/7 days off schedule of temozolomide for patients with recurrent high-grade gliomas (HGG). METHODS Sixty patients with recurrent HGG received temozolomide at 150 mg/m(2)/day on days 1-7 and days 15-21 during each 4-week cycle. The primary endpoint was 6-month progression-free survival (PFS-6), with a secondary endpoint of overall survival (OS). A further exploratory objective included the investigation of whether methylation status of the O(6)-methylguanine-DNA methyltransferase (MGMT) promoter within tumor tissue predicted outcomes. RESULTS Among patients with glioblastoma (n = 40), PFS-6 was 10% (95% CI, 3%-24%) with median OS of 21.6 weeks (95% CI, 16.9-30.6 weeks). PFS-6 for grade III glioma patients (n = 20) was 50% (95% CI, 27%-73%), and median OS was 100.6 weeks (95% CI, 67 weeks to not reached). There were trends towards longer PFS and OS with MGMT promoter methylation (log-rank test; P = .06 for PFS; P = .07 for OS). Additionally, bevacizumab-naïve glioblastoma patients had significantly longer PFS and OS (median PFS was 8.07 weeks [95% CI, 8 weeks to not reached] vs 7.57 weeks [95% CI, 7.29-8.29 weeks], log-rank test, P < .001; median OS was 62 weeks [26.1 weeks to not reached] vs 18.2 weeks [13.9-27.3 weeks], log-rank test, P < .001). CONCLUSIONS The dose-dense temozolomide regimen was well tolerated, although it has no significant activity in this population. Clinical trials.gov identified. NCT00619112 (available at http://clinicaltrials.gov/ct2/show/NCT00619112).
Collapse
Affiliation(s)
- Seunggu J Han
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - John D Rolston
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Jennifer L Clarke
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Michael D Prados
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Ashley DeSilva
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, California (S.J.H., J.D.R., A.M.M., J.L.C., M.D.P., S.M.C., M.S.B., A.D., N.A.B.); Department of Epidemiology and Biostatistics, University of California, San Francisco, California (A.M.M.)
| |
Collapse
|