1
|
Qin L, Huiwen M, Wang J, Wang Y, Khan SA, Zhang Y, Qiu H, Jiang L, He L, Zhang Y, Jia S. A novel polymerase β inhibitor from phage displayed peptide library augments the anti-tumour effects of temozolomide on colorectal cancer. J Chemother 2021; 34:391-400. [PMID: 34870566 DOI: 10.1080/1120009x.2021.2009987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The therapeutic efficacy of TMZ, a common used drug for chemotherapy, is limited by the resistance from colorectal cancer cells. Base excision repair (BER) pathway has been identified as one of the reasons for drug resistance. By blocking Polβ-dependent BER (Base Excision Repair) pathway, the efficacy of TMZ treatment can be improved greatly. Several Polβ inhibitors that have been identified could not become approved drugs due to lack of potency or specificity. To find therapeutic candidates with exquisite specificity and high affinity to Polβ, phage display technology was used in the current research. We screened out a candidate Polβ inhibitor, 10 D, that can inhibit the activity of Polβand SP-BER (Short-Patch Base excision Repair) pathway. Co-treatment with 10 D enhanced the sensitivity of colorectal cancer (CRC) cells to TMZ both in vitro and in vivo. Our data suggested that the novel Polβ inhibitor we identified can improve TMZ efficacy and optimize CRC chemotherapy.
Collapse
Affiliation(s)
- Lihong Qin
- Changzhou No. 7 People's Hospital, Changzhou, China
| | - Mao Huiwen
- Shuyang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Shuyang, China
| | - Jianguo Wang
- Shuyang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Shuyang, China
| | - Yuanyaun Wang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Salman A Khan
- Shuyang Hospital of Traditional Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Shuyang, China
| | - Ying Zhang
- Qinhuai Medical District, Jinlin Hospital of Nanjing University, Nanjing, China
| | - Hong Qiu
- Qinhuai Medical District, Jinlin Hospital of Nanjing University, Nanjing, China
| | - Longwei Jiang
- Qinhuai Medical District, Jinlin Hospital of Nanjing University, Nanjing, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yan Zhang
- Qinhuai Medical District, Jinlin Hospital of Nanjing University, Nanjing, China
| | - Shaochang Jia
- Qinhuai Medical District, Jinlin Hospital of Nanjing University, Nanjing, China
| |
Collapse
|
2
|
Khasraw M, Lee A, McCowatt S, Kerestes Z, Buyse ME, Back M, Kichenadasse G, Ackland S, Wheeler H. Cilengitide with metronomic temozolomide, procarbazine, and standard radiotherapy in patients with glioblastoma and unmethylated MGMT gene promoter in ExCentric, an open-label phase II trial. J Neurooncol 2016; 128:163-171. [PMID: 26935578 DOI: 10.1007/s11060-016-2094-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
Newly diagnosed glioblastoma multiforme with unmethylated MGMT promoter has a poor prognosis, with a median survival of 12 months. This phase II study investigated the efficacy and safety of combining the selective integrin inhibitor cilengitide with a combination of metronomic temozolomide and procarbazine for these patients. Eligible patients (newly diagnosed, histologically confirmed supratentorial glioblastoma with unmethylated MGMT promoter) were entered into this multicentre study. Cilengitide (2000 mg IV twice weekly) was commenced 1 week prior to radiotherapy combined with daily temozolomide (60 mg/m(2)) and procarbazine (50 or 100 mg) and, after 4 weeks' break, followed by six adjuvant cycles of temozolomide (50-60 mg/m(2)) and procarbazine (50 or 100 mg) on days 1-20, every 28 days. Cilengitide was continued for up to 12 months or until disease progression or unacceptable toxicity. The primary endpoint for efficacy was a 12-month overall survival rate of 65 %. Twenty-nine patients completed study treatment. Sixteen patients survived for 12 months or more, an overall survival rate of 55 %. The median overall survival was 14.5 months (95 % CI 11.1-19.6) and the median progression-free survival was 7.4 months (95 % CI 6.1-8). Cilengitide combined with metronomic temozolomide and procarbazine in MGMT-promoter unmethylated glioblastoma did not improve survival compared with historical data and does not warrant further investigation.
Collapse
Affiliation(s)
- Mustafa Khasraw
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia. .,University of Sydney, Sydney, Australia. .,NHMRC Clinical Trials Centre, University of Sydney, Sydney, Australia. .,Royal North Shore Hospital, Pacific HWY, St Leonards, NSW, 2065, Australia.
| | - Adrian Lee
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - Sally McCowatt
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - Zoltan Kerestes
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - Marc E Buyse
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium
| | - Michael Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| | - Ganessan Kichenadasse
- International Drug Development Institute (IDDI), Louvain-la-Neuve, Belgium.,Flinders Medical Centre and Flinders University, Adelaide, Australia
| | | | - Helen Wheeler
- Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia.,University of Sydney, Sydney, Australia
| |
Collapse
|
3
|
Abstract
Background:Since temozolomide (TMZ) entry into routine practice in the first-line management of glial tumors, post-TMZ recurrences present a growing challenge. Without standard chemotherapy for TMZ failure, care in such palliative settings requires consideration not only of efficacy but of toxicity and convenience.Methods:At our institution, a combination regimen has been used: oral alkylating agents procarbazine (PCB) (100-150 mg/m2/day) and TMZ (150-200 mg/m2/day) administered on days 1-5 of a 28-day cycle. This treatment has been initiated upon radiological and/or clinical disease progression, and continued until evidence of further progression or toxicity. We retrospectively reviewed our experence with this regimen.Results:Since November 2004, 17 patients (median age 53) were treated for histologically confirmed glioma (glioblastoma multiforme (GBM), N=12; Grade 3 glioma, N=3; Grade 2 glioma, N=2) after a median of 2 recurrences. TMZ was previously given either as adjuvant therapy (post-chemoradiotherapy maintenance in 8 of 13 cases) or as salvage monotherapy (4 cases). Of 16 evaluable cases, 14 (13 high grade tumors) showed O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Two patients achieved partial response and one had complete response by RECIST criteria. Disease progressed after a median of 4 cycles (range 1 to 11+), with an actuarial progression-free survival of 42% after 6 cycles. Grade 3/4 toxicity was rare, and no dose reductions were needed. One patient discontinued treatment due to procarbazine hypersensitivity.Conclusion:Combination PCB-TMZ is well-tolerated, with modest activity in TMZ-exposed glioma.
Collapse
|
4
|
Abstract
Malignant gliomas are the most prevalent type of primary brain tumor in adults. Despite progress in brain tumor therapy, the prognosis of malignant glioma patients remains dismal. The median survival of patients with glioblastoma multiforme, the most common grade of malignant glioma, is 10-12 months. Conventional therapy of surgery, radiation and chemotherapy is largely palliative. Essentially, tumor recurrence is inevitable. Salvage treatments upon recurrence are palliative at best and rarely provide significant survival benefit. Therapies targeting the underlying molecular pathogenesis of brain tumors are urgently required. Common genetic abnormalities in malignant glioma specimens are associated with aberrant activation or suppression of cellular signal transduction pathways and resistance to radiation and chemotherapy. Several low molecular weight signal transduction inhibitors have been examined in preclinical and clinical malignant glioma trials. The efficacy of these agents as monotherapies has been modest, at best; however, small subsets of patients who harbor specific genetic changes in their tumors may display favorable clinical responses to defined small molecule inhibitors. Multitargeted kinase inhibitors or combinations of agents targeting different mitogenic pathways may overcome the resistance of tumors to single-agent targeted therapies. Well designed studies of small molecule kinase inhibitors will include assessment of safety, drug delivery, target inhibition and correlative biomarkers to define mechanisms of response or resistance to these agents. Predictive biomarkers will enrich for patients most likely to respond in future clinical trials. Additional clinical studies will combine novel targeted therapies with radiation, chemotherapies and immunotherapies.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- The Preston Robert Tisch Brain Tumor Center Division of Neurosurgery/Neuro-Oncology, Duke University Medical Center, DUMC 3624, Durham, NC 27710, USA.
| | | |
Collapse
|
5
|
Archavlis E, Tselis N, Birn G, Ulrich P, Baltas D, Zamboglou N. Survival analysis of HDR brachytherapy versus reoperation versus temozolomide alone: a retrospective cohort analysis of recurrent glioblastoma multiforme. BMJ Open 2013; 3:bmjopen-2012-002262. [PMID: 23503605 PMCID: PMC3612781 DOI: 10.1136/bmjopen-2012-002262] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVES Tumour recurrence of glioblastoma multiforme (GBM) after initial treatment with surgical resection, radiotherapy and chemotherapy is an inevitable phenomenon. This retrospective cohort study compared the efficacy of interstitial high dose rate brachytherapy (HDR-BRT), re-resection and sole dose dense temozolomide chemotherapy (ddTMZ) in the treatment of recurrent glioblastoma after initial surgery and radiochemotherapy. DESIGN Retropective cohort study. SETTING Primary level of care with two participating centres. The geographical location was central Germany. PARTICIPANTS From January 2005 to December 2010, a total of 111 patients developed recurrent GBM after initial surgery and radiotherapy with concomitant temozolomide. The inclusion criteria were as follows: (1) histology-proven diagnosis of primary GBM (WHO grade 4), (2) primary treatment with resection and radiochemotherapy, and (3) tumour recurrence/progression. INTERVENTIONS This study compared retrospectively the efficacy of interstitial HDR-BRT, re-resection and ddTMZ alone in the treatment of recurrent glioblastoma. PRIMARY AND SECONDARY OUTCOME MEASURES Median survival, progression free survival and complication rate. RESULTS Median survival after salvage therapy of the recurrence was 37, 30 and 26 weeks, respectively. The HDR-BRT group did significantly better than both the reoperation (p<0.05) and the ddTMZ groups (p<0.05). Moderate to severe complications in the HDR-BRT, reoperation and sole chemotherapy groups occurred in 5/50 (10%), 4/36 (11%) and 9/25 (36%) cases, respectively. CONCLUSIONS CT-guided interstitial HDR-BRT attained higher survival benefits in the management of recurrent glioblastoma after initial surgery and radiotherapy with concurrent temozolomide in comparison with the other treatment modalities. The low risk of complications of the HDR-BRT and the fact that it can be delivered percutaneously in local anaesthesia render it a promissing treatment option for selected patients which should be further evaluated.
Collapse
Affiliation(s)
- Eleftherios Archavlis
- Department of Neurosurgery, Klinikum Offenbach, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
| | - Nikolaos Tselis
- Department of Radiotherapy and Interdisciplinary Oncology, Klinikum Offenbach, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
| | - Gerhard Birn
- Department of Neurosurgery, Klinikum Darmstadt, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
| | - Peter Ulrich
- Department of Neurosurgery, Klinikum Offenbach, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
- Department of Radiotherapy and Interdisciplinary Oncology, Klinikum Offenbach, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
| | - Dimos Baltas
- Department of Medical Physics and Engineering, Klinikum Offenbach, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
| | - Nikolaos Zamboglou
- Department of Radiotherapy and Interdisciplinary Oncology, Klinikum Offenbach, Akademisches Lehrkrankenhaus der Universität Frankfurt, Offenbach, Germany
| |
Collapse
|
6
|
Bagriacik EU, Baykaner MK, Yaman M, Sivrikaya G, Durdağ E, Emmez H, Fincan GÖ, Börcek AÖ, Seçen AE, Ercan S. Establishment of a primary pleomorphic xanthoastrocytoma cell line: in vitro responsiveness to some chemotherapeutics. Neurosurgery 2012; 70:188-97. [PMID: 21629132 DOI: 10.1227/neu.0b013e3182262c5b] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Anaplastic pleomorphic xanthoastrocytoma is an aggressively growing, malignant, and eventually fatal tumor of the central nervous system. Testing chemotherapeutic drug sensitivity under in vitro conditions would be a useful strategy to determine sensitive or resistant drugs for fatal brain cancers. OBJECTIVE To establish primary cell cultures of excised tumor tissue from pleomorphic xanthoastrocytoma-bearing patients and to test their sensitivity against various anticancer chemotherapy drugs. METHODS Prepared suspensions of the excised tumor tissue from a patient who had a recurrent grade 3 pleomorphic xanthoastrocytoma was cultured in culture dishes until cells began to grow. Immunofluorescent and immunohistochemical visualizations were performed using confocal and light microscopy. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay in comparison with ³H-thymidine incorporation assay was used to test cellular toxicity of several anticancer drugs. RESULTS We established vigorously growing primary cells of the tumor. Drug sensitivity testing was conducted successfully. CONCLUSION Primary cell cultures of surgically removed tumor tissues may be useful in studies of cancer biology and chemotherapeutic drug sensitivity for recurrent malignant brain tumors, particularly for anaplastic pleomorphic xanthoastrocytoma.
Collapse
|
7
|
Armand JP, Ribrag V, Harrousseau JL, Abrey L. Reappraisal of the use of procarbazine in the treatment of lymphomas and brain tumors. Ther Clin Risk Manag 2011; 3:213-24. [PMID: 18360630 PMCID: PMC1936303 DOI: 10.2147/tcrm.2007.3.2.213] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Procarbazine HCl is a 'nonclassical' oral alkylating anticancer agent that was first synthesized in the late 1950s. It has been used in the treatment of many cancers, but its main use is in the treatment of Hodgkin's lymphoma and brain tumors and, to a lesser extent, Non-Hodgkin's lymphoma and primary central nervous system lymphoma. Procarbazine is a prodrug that undergoes metabolic transformation into active intermediates that are thought to inhibit DNA, RNA, and protein synthesis. Early use of procarbazine in combination with mechlorethamine, vincristine, and prednisone (MOPP) was effective in the treatment of advanced Hodgkin's lymphoma, but late toxic effects such as secondary cancer and infertility led to its replacement by other regimens. However, its recent reintroduction in the dose-intensified BEACOPP (bleomycin, etoposide, doxorubicin, cyclophosphamide, vincristine, procarbazine, and prednisone) regimen has yielded very promising findings. Procarbazine alone, or more commonly combined in the PCV (procarbazine, lomustine [CCNU], and vincristine) regimen, is also effective in treating gliomas comprising astrocytomas, glioblastomas, and oligodendrogliomas. The most common side effects of procarbazine are gastrointestinal disturbances, myelosuppression, and central nervous system effects. In conclusion, the use of procarbazine in combination with other drugs means that it remains a major anticancer drug in the management of Hodgkin's lymphoma and gliomas.
Collapse
Affiliation(s)
| | - Vincent Ribrag
- Institut Gustave-RoussyDesmolins, Villejuif Cedex, France
| | | | - Lauren Abrey
- Memorial Sloan-Kettering Cancer CenterNew York, New York, USA
| |
Collapse
|
8
|
Rubie H, Geoerger B, Frappaz D, Schmitt A, Leblond P, Ndiaye A, Aerts I, Deley MCL, Gentet JC, Paci A, Chastagner P, Dias N, Djafari L, Pasquet M, Chatelut E, Landman-Parker J, Corradini N, Vassal G. Phase I study of topotecan in combination with temozolomide (TOTEM) in relapsed or refractory paediatric solid tumours. Eur J Cancer 2010; 46:2763-70. [PMID: 20558056 DOI: 10.1016/j.ejca.2010.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 04/08/2010] [Accepted: 05/04/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE To evaluate maximum tolerated dose and recommended dose (RD) for phase II studies of topotecan (TPT) combined with temozolomide (TMZ) (TOTEM) in children and adolescents with relapsed or refractory solid malignancies. PATIENTS AND METHODS Multicentre, phase I study with a standard '3+3' design in five dose increments. Eligible patients: aged 6 months to 21 years, diagnosis of a solid malignancy failed at least 2 previous lines of therapy. TMZ was administered orally, starting at 100 mg/m(2)/d, and TPT intravenously over 30 min, starting at 0.75 mg/m(2)/d over 5 consecutive days every 28 d. A pharmacokinetics analysis was performed on Day 1 and Day 5 of cycle 1. RESULTS Between February and October 2007, 16 patients were treated. The median age was 8.5 years (range, 3-19 years). Dose-limiting toxicity (grade 4 neutropenia and/or thrombocytopenia lasting more than 7 d) during the first cycle occurred in 2 of 3 patients at level 3 (TMZ 150 mg/m(2)/d and TPT 1.0 mg/m(2)/d) and was always manageable. Confirmed complete and partial responses were observed in 4 patients (25%), three with metastatic neuroblastoma and one with high-grade glioma. Seven patients had a stable disease. Pharmacokinetic data show a wide inter-individual variability. No significant differences were observed between plasma TMZ and TPT concentrations on Day 1 and Day 5 indicating the absence of pharmacokinetic interaction between the drugs. CONCLUSIONS The RD for the combination is TMZ 150 mg/m(2)/d and TPT 0.75 mg/m(2)/d with dose-limiting haematological toxicity. The observed activity deserves further evaluation in paediatric malignancies.
Collapse
|
9
|
Omar AI, Mason WP. Temozolomide: The evidence for its therapeutic efficacy in malignant astrocytomas. CORE EVIDENCE 2010; 4:93-111. [PMID: 20694068 PMCID: PMC2899776 DOI: 10.2147/ce.s6010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Indexed: 11/23/2022]
Abstract
Introduction: Malignant gliomas are a heterogeneous group of primary central nervous system neoplasms that represent less than 2% of all cancers yet carry a significant burden to society. They are frequently associated with considerable and progressive neurological disability and are ultimately intractable to all forms of treatment. Temozolomide (TMZ) is a new second generation DNA alkylating agent that has become part of malignant astrocytoma management paradigms because of its proven efficacy, ease of administration, and favorable toxicity profile. Aims: To review the role of TMZ in the management of malignant astrocytomas (World Health Organization grades III and IV) including newly diagnosed (n) and recurrent (r) anaplastic astrocytomas (AA) and glioblastomas. Evidence review: A series of pivotal clinical trials have established a role for TMZ in the treatment of malignant astrocytomas. A large phase II trial examining the role of TMZ in rAA showed a response rate of 35%, and a 6-month progression-free survival of 46%. This led to the accelerated approval of TMZ by the FDA and the EU for the treatment of rAA. Evidence for a role of TMZ in nAA is currently limited but research is ongoing in this area. The role of TMZ in the management of glioblastoma at the time of recurrence (rGBM) is less impressive but evidence for its activity was demonstrated in two large phase II trials that led to the approval of TMZ for this indication in Europe and Canada but not in the US. A recent large prospective randomized phase III trial showed that the addition of TMZ during and after radiation therapy (RT) in newly diagnosed (nGBM) patients prolonged median overall survival by 2.5 months; perhaps more importantly, the 2-year survival rate for patients receiving TMZ and RT was 26% compared with 10% for those receiving RT alone. Concurrent TMZ with RT followed by adjuvant TMZ has become the standard of care for nGBM patients. Based on the evidence presented in this trial, TMZ received approval from the FDA and the EU for patients with nGBM in 2005. Place in therapy: There is evidence to support the use of TMZ for the following diseases in the order of most to least convincing: nGBM, rAA, rGBM, and nAA. This order may quickly change as more trials are being designed and implemented, particularly with novel TMZ dosing schedules.
Collapse
Affiliation(s)
- Ayman I Omar
- Department of Medicine, Princess Margaret Hospital and the University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
10
|
López T, Figueras F, Manjarrez J, Bustos J, Alvarez M, Silvestre-Albero J, Rodríguez-Reinoso F, Martínez-Ferre A, Martínez E. Catalytic nanomedicine: A new field in antitumor treatment using supported platinum nanoparticles. In vitro DNA degradation and in vivo tests with C6 animal model on Wistar rats. Eur J Med Chem 2010; 45:1982-90. [PMID: 20153564 DOI: 10.1016/j.ejmech.2010.01.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2009] [Revised: 12/29/2009] [Accepted: 01/19/2010] [Indexed: 10/19/2022]
|
11
|
A Case Report of High-dose Local Chemotherapy With Carmustine Wafers in a Large-size Glioblastoma Multiforme. ACTA ACUST UNITED AC 2010. [DOI: 10.1097/wnq.0b013e3181ce6726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
12
|
Abstract
BACKGROUND Gliomas account for 42% of all primary CNS neoplasms and 77% of all malignant primary CNS neoplasms. Unfortunately the high-grade variant of gliomas, glioblastoma multiforme (GBM), is difficult to treat and generally considered incurable. Survival rates are generally poor, and neurological morbidity in the setting of disease progression is high. Fortunately, significant progress has been achieved in the past decade in our understanding of the molecular biology of this aggressive tumour histology and, as a consequence, there is renewed clinical trial activity in this area focused on improving quality of life, treatment-related morbidity and outcomes. METHODS A review of literature from June 2005 to June 2008 was conducted on multimodal treatment of malignant glioma (MG) patients, using specific search criteria in Medline, EMBASE, and BIOSIS. Abstracts from relevant US and European medical (cancer) meetings were also evaluated. RESULTS The established therapies for MG include surgery, radiotherapy (RT), and local or systemic chemotherapy. However, over the last 10 years only two chemotherapeutic agents have received regulatory approval for treatment of MG: polifeprosan 20 with carmustine (BCNU implant) and temozolomide (TMZ), an imidazotetrazine derivative of dacarbazine. More recent advances in the treatment of brain tumours have been in the development of multimodal approaches. Specific interest in the combination of BCNU implant and TMZ has arisen due to the demonstrable depletion by TMZ of the DNA repair enzyme responsible for resistance to a nitrosourea such as BCNU. Further interest in this combination stems from the observation that there is a difference in the time to peak effect for each agent. Additional emerging data suggest that multimodal therapy with maximal resection and BCNU implants, followed by adjuvant therapy with radiation and TMZ, is effective and well-tolerated in patients with initial high-grade, resectable MG. CONCLUSIONS The increasing body of efficacy data suggests that this combination of BCNU implants and TMZ within a multimodal treatment strategy including surgery and RT may provide an enhanced benefit compared with the use of either of these agents alone in select patients with high-grade glioma.
Collapse
|
13
|
Sathornsumetee S, Rich JN, Reardon DA. Diagnosis and treatment of high-grade astrocytoma. Neurol Clin 2008; 25:1111-39, x. [PMID: 17964028 DOI: 10.1016/j.ncl.2007.07.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High-grade astrocytomas include the most common adult central nervous system (CNS) tumor, glioblastoma multiforme, and anaplastic astrocytoma--a highly aggressive cancer with short median survival despite maximal multimodality therapy. Diagnosis is by clinical and radiographic findings confirmed by histopathology. Standard-of-care therapy includes surgical resection, radiotherapy, and temozolomide. Nearly all patients who have high-grade astrocytomas develop tumor recurrence or progression after this multimodality treatment. Two treatment challenges are molecular/genetic heterogeneity of tumors and limited CNS tumor delivery. It is probable that targeted therapies will be most effective in combination with one another or with cytotoxic therapies. This article discusses diagnosis and current treatment of high-grade astrocytomas.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- Division of Neurology, The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, DUMC 3624, Durham, NC 27710, USA
| | | | | |
Collapse
|
14
|
Contribution of DNA repair mechanisms to determining chemotherapy response in high-grade glioma. J Clin Neurosci 2008; 15:1-8. [DOI: 10.1016/j.jocn.2007.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 05/23/2007] [Indexed: 01/07/2023]
|
15
|
Abstract
BACKGROUND We describe a retrospective series of children with low-grade glioma who received temozolomide. PROCEDURE Eligible patients had had a diagnosis of low-grade glioma with or without histological confirmation. Temozolomide was administered at a dose of 200 mg/m(2) daily for 5 days, in a 4-week cycle. Therapy was stopped on completion of the targeted 12 cycles of chemotherapy or on evidence of tumor progression. RESULTS Thirteen eligible patients were identified, eight male and five female. Median age at diagnosis was 5.5 years (range 2.6-15.0 years) and at commencement of temozolomide treatment was 9.0 years (range 3.8-15.2 years). Nine patients had a histological diagnosis of pilocytic astrocytoma. Twelve patients had received carboplatin prior to temozolomide, including three in combination with vincristine. A total of 111 cycles of therapy have been administered. Hematological toxicity and nausea were the most common adverse effects. Median time to progression was 6.7 months (range 1.5-41.8 months). Event-free survival rate at 3 years was 57%. Twelve of 13 patients remain alive at the time of report. Eleven have stable disease (SD). CONCLUSION Temozolomide appears to be active in pediatric low-grade glioma, with the advantage of oral administration and excellent tolerability.
Collapse
Affiliation(s)
- Seong L Khaw
- Children's Cancer Centre, Royal Children's Hospital, Melbourne, Australia.
| | | | | | | | | |
Collapse
|
16
|
Ferguson PJ, Kurowska EM, Freeman DJ, Chambers AF, Koropatnick J. In vivo inhibition of growth of human tumor lines by flavonoid fractions from cranberry extract. Nutr Cancer 2007; 56:86-94. [PMID: 17176222 DOI: 10.1207/s15327914nc5601_12] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Edible fruits and berries may serve as sources for novel anticancer agents, given that extracts of these foods have demonstrated cytotoxic activity against tumor cell lines. Semipurified, flavonoid-rich extracts of cranberry (Vaccinia macrocarpa) were shown previously to arrest proliferation of tumor cells and induce apoptosis. However, the ability of cranberry flavonoids to inhibit tumor growth in vivo has not been reported other than in a preliminary report. As model systems for testing this activity, human tumor cell lines representative of three malignancies were chosen: glioblastoma multiforme (U87), colon carcinoma (HT-29), and androgen-independent prostate carcinoma (DU145). A flavonoid-rich fraction 6 (Fr6) and a more purified proanthocyanidin (PAC)-rich fraction were isolated from cranberry presscake and whole cranberry, respectively, by column chromatography. Fr6 and PAC each significantly slowed the growth of explant tumors of U87 in vivo, and PAC inhibited growth of HT-29 and DU145 explants (P < 0.05), inducing complete regression of two DU145 tumor explants. Flow cytometric analyses of in vitro-treated U87 cells indicated that Fr6 and PAC could arrest cells in G1 phase of the cell cycle (P < 0.05) and also induce cell death within 24 to 48 h of exposure (P < 0.05). These results indicate the presence of a potential anticancer constituent in the flavonoid-containing fractions from cranberry extracts.
Collapse
Affiliation(s)
- Peter J Ferguson
- London Regional Cancer Program, Health Sciences Centre, 790 Commissioners Road East, London, Ontario, Canada.
| | | | | | | | | |
Collapse
|
17
|
Simpson L, Galanis E. Recurrent glioblastoma multiforme: advances in treatment and promising drug candidates. Expert Rev Anticancer Ther 2007; 6:1593-607. [PMID: 17134364 DOI: 10.1586/14737140.6.11.1593] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Recurrent glioblastoma multiforme is a lethal disease with currently available treatment options having a limited impact on outcome. In this article, current and novel therapeutic approaches in the treatment of recurrent glioblastoma multiforme, including chemotherapy, targeted molecular agents, virotherapy/gene therapy and immunotherapy and challenges in developing novel therapeutic agents for glioblastoma multiforme will be discussed.
Collapse
Affiliation(s)
- Lijo Simpson
- Mayo Clinic, Department of Oncology, 200 1st Str SW, Rochester, MN 55905, USA.
| | | |
Collapse
|
18
|
Abstract
Primary brain tumors represent over 100 different tumor types with widely divergent biologies and clinical outcomes, but these neoplasms frequently pose similar challenges to neuro-oncologists. Malignant gliomas are the most common type of primary intrinsic brain tumor in adults and remain extremely lethal. Current standard-of-care therapies for these cancers include surgery, radiation and palliative cytotoxics, which have significant side-effects and limited efficacy. Advances in our understanding of the molecular underpinnings of cancer have led to targeted molecular therapies that may permit improvement in therapeutic efficacy and reduced toxicity; these therapies, however, still face many challenges. Signal transduction pathways that are inappropriately regulated in brain cancers include growth factors and their receptors (e.g. epidermal growth factor receptor, vascular endothelial growth factor receptor and platelet-derived growth factor receptor), which regulate cellular interactions with the microenvironment and intracellular oncogenic pathways. Low-molecular-weight inhibitors have been developed to target many kinases and may have advantages in terms of delivery. Monoclonal antibodies may have greater specificity, but face delivery restrictions. Preferential tumor delivery of chemotherapies, conjugated toxins and radioisotopes has been achieved through convection-enhanced delivery, intratumoral implants and intra-arterial infusion. Despite these advances, few molecularly targeted therapies have demonstrated significant antineoplastic activity for a broad range of patients, possibly due to tumor and patient heterogeneity. Improved functional neuropathology and imaging may permit identification of patient subgroups for which clinical responses may be enriched. It is probable, however, that targeted therapies will be most effective in combination either with one another or with cytotoxic therapies. In this study, we review the current state of new therapies for malignant gliomas.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- Departments of Medicine, The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
19
|
Mason WP, Stupp R. Recent advances in the medical therapy of high-grade gliomas. FUTURE NEUROLOGY 2006. [DOI: 10.2217/14796708.1.6.831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Malignant glial neoplasms, including glioblastoma, are amongst the most devastating and intractable of solid tumors. Until recently the standard of care for newly diagnosed glioblastoma was surgical resection to the extent feasible followed by conventional fractionated radiotherapy. When administered for disease progression, chemotherapy had modest benefit and its use in the adjuvant setting was controversial. Temozolomide, an oral alkylating chemotherapeutic agent, has now been demonstrated to increase survival time in patients with newly diagnosed glioblastoma when used concurrently with radiotherapy and as adjuvant or maintenance treatment for six cycles thereafter. Correlative molecular studies suggested that the benefit of temozolomide is largely restricted to patients whose tumor has silenced the gene for methylguanine methyltransferase, a repair enzyme implicated in resistance to alkylator chemotherapy. Use of temozolomide chemotherapy upfront in the management of glioblastoma is now considered the standard of care. This significant advance has also stimulated development of therapeutic strategies that incorporate temozolomide, and other agents, in the initial management of most high-grade gliomas. Furthermore, our increased understanding of the molecular derangements that underlie gliomagenesis has identified a number of putative molecular targets against which novel therapeutics have been tested with encouraging preliminary results. Finally, the challenges presented by the blood–brain barrier to adequate drug delivery have stimulated the development of unique locoregional delivery techniques that are currently undergoing clinical evaluation. This review summarizes these recent advances, and speculates on how the field is likely to evolve in the near future.
Collapse
Affiliation(s)
- Warren P Mason
- Princess Margaret Hospital, 610 University Avenue, Suite 18–717, Toronto, ON M5G 2M9, Canada
| | - Roger Stupp
- University of Lausanne Hospitals, Multidisciplinary Oncology Center, 46 Rue du Bugnon, Lausanne, 1011, Switzerland
| |
Collapse
|
20
|
Morrone FB, Oliveira DL, Gamermann P, Stella J, Wofchuk S, Wink MR, Meurer L, Edelweiss MIA, Lenz G, Battastini AMO. In vivo glioblastoma growth is reduced by apyrase activity in a rat glioma model. BMC Cancer 2006; 6:226. [PMID: 16995949 PMCID: PMC1592110 DOI: 10.1186/1471-2407-6-226] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2006] [Accepted: 09/23/2006] [Indexed: 12/03/2022] Open
Abstract
Background ATP is an important signalling molecule in the peripheral and central nervous system. Both glioma growth and tumor resection induces cell death, thus liberating nucleotides to the extracellular medium. Nucleotides are hydrolyzed very slowly by gliomas when compared with astrocytes and induce neuronal cell death and glioma proliferation. The objective of the present study was to test the involvement of extracellular ATP in glioblastoma growth in a rat glioma model. Methods To deplete the extracellular ATP, the enzyme apyrase was tested on the treatment of gliomas implanted in the rats CNS. One million glioma C6 cells in 3 microliters of DMEM/FCS were injected in the right striata of male Wistar rats, 250–270 g. After 20 days, the rats were decapitated and the brain sectioning and stained with hematoxylin and eosine. We performed immunohistochemical experiments with Ki67, CD31 and VEGF. Total RNA was isolated from cultured glioma C6 cells and the cDNA was analyzed by Real Time-PCR with primers for the NTPDase family. Results C6 glioma cells effectively have a low expression of all NTPDases investigated, in comparison with normal astrocytes. The implanted glioma co-injected with apyrase had a significant reduction in the tumor size (p < 0.05) when compared with the rats injected only with gliomas or with gliomas plus inactivated apyrase. According to the pathological analysis, the malignant gliomas induced by C6 injection and co-injected with apyrase presented a significant reduction in the mitotic index and other histological characteristics that indicate a less invasive/proliferative tumor. Reduction of proliferation induced by apyrase co-injection was confirmed by counting the percentage of Ki67 positive glioma cell nuclei. According to counts with CD31, vessel density and neoformation was higher in the C6 group 20 days after implantation. Confirming this observation, rats treated with apyrase presented less VEGF staining in comparison to the control group. Conclusion These results indicate that the participation of extracellular ATP and the ecto-nucleotidases may be associated with the development of this type of brain tumor in an in vivo glioma model.
Collapse
Affiliation(s)
- Fernanda B Morrone
- Departamento de Bioquímica, ICBS, UFRGS, Porto Alegre, RS, Brazil
- Faculdade de Farmácia, PUCRS, Porto Alegre, RS, Brazil
| | - Diogo L Oliveira
- Departamento de Bioquímica, ICBS, UFRGS, Porto Alegre, RS, Brazil
| | | | - Joseli Stella
- Departamento de Bioquímica, ICBS, UFRGS, Porto Alegre, RS, Brazil
| | - Suzana Wofchuk
- Departamento de Bioquímica, ICBS, UFRGS, Porto Alegre, RS, Brazil
| | - Márcia R Wink
- Departamento de Biofísica IB, UFRGS, Porto Alegre, RS, Brazil
| | - Luise Meurer
- Departamento de Patologia, HCPA, UFRGS, Porto Alegre, RS, Brazil
| | | | - Guido Lenz
- Departamento de Biofísica IB, UFRGS, Porto Alegre, RS, Brazil
| | | |
Collapse
|
21
|
Boiardi A, Eoli M, Salmaggi A, Lamperti E, Botturi A, Broggi G, Bissola L, Finocchiaro G, Silvani A. Systemic temozolomide combined with loco-regional mitoxantrone in treating recurrent glioblastoma. J Neurooncol 2006; 75:215-20. [PMID: 16283445 DOI: 10.1007/s11060-005-3030-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Twenty-two recurrent GBM patients were enrolled for second tumor debulking with local positioning of a Rickam reservoir, in order to locally deliver chemotherapy with the aim of controlling local tumor recurrence. We designed a protocol using systemic temozolomide (150 mg/sqm days 1-5 every 28) in association with mitoxantrone, delivered through the reservoir (4 mg/day 1-5 every 28) positioned into the area of tumor exeresis. After re-operation a residual tumor mass no larger than 2 cm was identified in 18/22 patients. The patients were treated with monthly cycles of chemotherapy until evolution of the tumor, but in no case for more than 10 cycles. Responses were evaluated by MRI scans performed every 2 months and images assessed according to MacDonald's criteria. Response rate: no complete responses (CR), 5 partial responses (PR), 13 stable disease (SD) and 4 progressive disease (PD) occurred. The median progression-free survival (PFS) and survival time (ST) of the whole group of treated patients was 7 and 11 months, respectively and more than a quarter of the patients survived over 18 months. During the study, the patients' compliance was complete and no dropouts occurred. Hematological toxicity was mild and after repeated local injections only minor neurological side-effects occurred. Despite some bias in patients' selection not excluded in this pilot study, results are interesting: the PFS was as long as the survival of recurrent GBM reported in the literature.
Collapse
Affiliation(s)
- A Boiardi
- Department of Neuro-Oncology, Istituto Nazionale Neurologico "Carlo Besta", Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Payne MJ, Pratap SE, Middleton MR. Temozolomide in the treatment of solid tumours: current results and rationale for dosing/scheduling. Crit Rev Oncol Hematol 2005; 53:241-52. [PMID: 15718149 DOI: 10.1016/j.critrevonc.2004.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2004] [Indexed: 11/26/2022] Open
Abstract
This review examines the current evidence for the use of temozolomide in the treatment of solid tumours. The possible molecular and clinical advantages of temozolomide are identified and the molecular mechanism of temozolomide resistance is explored. Attempts to maximise efficacy have led to manipulation of both dosage and drug scheduling and the evidence for the various strategies is reviewed. Finally, the potential role of combination therapy is considered.
Collapse
Affiliation(s)
- M J Payne
- The Churchill Hospital, CRUK Medical Oncology Unit, Oxford Radcliffe Hospitals Trust, Headington, Oxford OX37LJ, UK
| | | | | |
Collapse
|
24
|
Abstract
The designation of a tumor as anaplastic astrocytoma (AA) reflects a distinct histologic classification of malignant glioma characterized by an abundance of pleomorphic astrocytes with evidence of mitosis. Although these tumors are malignant, they have a better prognosis and a higher likelihood of response to treatment than glioblastoma. Despite advances in brain tumor imaging, making an accurate diagnosis requires the evaluation of tumor tissue and is essential for treatment planning. Currently, most patients undergo maximal surgical debulking of tumor followed by external beam radiation, often with subsequent adjuvant chemotherapy. However, despite the use of these treatment modalities, most tumors recur within a few years and these recurrent tumors are more refractory to subsequent therapies. This review examines the diagnosis, prognosis, and treatment of AAs. Ongoing clinical research investigations are also summarized, reflecting advances in our knowledge of the molecular pathogenesis of these tumors and providing hope for significant improvements in patient outcomes.
Collapse
Affiliation(s)
- Siew Ju See
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore
| | | |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The discovery of temozolomide in the 1980s was expected to be an important advance in improving survival for patients with malignant brain tumors. Numerous clinical studies have demonstrated the activity of temozolomide against recurrent or refractory gliomas and noncentral nervous system malignancies. In the last 2 years, studies have focused on exploring strategies to optimize the efficacy of temozolomide, including evaluating different temozolomide dosing schedules and combining temozolomide with other antineoplastic agents, radiation therapy, or drug resistance-modifying agents. RECENT FINDINGS A critical review of these studies suggests that temozolomide, as currently used, has limited efficacy in treating refractory malignant infiltrative brain tumors, and survival benefit is, at best, a few weeks longer than that with procarbazine. There is enthusiasm about the activity of temozolomide in the treatment of recurrent low-grade gliomas and advanced malignant melanomas. Temozolomide has activity and a favorable safety profile in all dosing schedules tested. Nevertheless, the trials evaluating the efficacy of temozolomide suffer from being uncontrolled, with short follow-up periods. SUMMARY Despite the advantages of a favorable safety profile and oral administration, temozolomide has yet to realize its initial promise and full potential. Studies of temozolomide combined with novel drug resistance-modifying agents will likely improve disease control while minimizing toxicities, leading to improved survival benefit. Larger, randomized trials comparing temozolomide with standard therapy are needed to confirm the suggested benefit from temozolomide in malignant brain tumors. Temozolomide will continue to be attractive as an agent in the treatment of brain tumors because of its desirable features and safety.
Collapse
Affiliation(s)
- R Nagasubramanian
- Department of Pediatrics, Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Illinois 60637, USA
| | | |
Collapse
|
26
|
Abstract
Malignant gliomas are the most common type of primary brain tumor. Although therapy for patients with these tumors remains limited, there has been important progress recently. In this review, some of these advances are discussed, with an emphasis on targeted molecular therapies.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology, Dana Farber Cancer Institute, SW430D, 44 Binney Street, Boston, MA 02115, USA.
| | | |
Collapse
|