1
|
d'Angelo M, Castelli V, Benedetti E, Antonosante A, Catanesi M, Dominguez-Benot R, Pitari G, Ippoliti R, Cimini A. Theranostic Nanomedicine for Malignant Gliomas. Front Bioeng Biotechnol 2019; 7:325. [PMID: 31799246 PMCID: PMC6868071 DOI: 10.3389/fbioe.2019.00325] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022] Open
Abstract
Brain tumors mainly originate from glial cells and are classified as gliomas. Malignant gliomas represent an incurable disease; indeed, after surgery and chemotherapy, recurrence appears within a few months, and mortality has remained high in the last decades. This is mainly due to the heterogeneity of malignant gliomas, indicating that a single therapy is not effective for all patients. In this regard, the advent of theranostic nanomedicine, a combination of imaging and therapeutic agents, represents a strategic tool for the management of malignant brain tumors, allowing for the detection of therapies that are specific to the single patient and avoiding overdosing the non-responders. Here, recent theranostic nanomedicine approaches for glioma therapy are described.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Reyes Dominguez-Benot
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giuseppina Pitari
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Kim JS, Shin DH, Kim JS. Dual-targeting immunoliposomes using angiopep-2 and CD133 antibody for glioblastoma stem cells. J Control Release 2017; 269:245-257. [PMID: 29162480 DOI: 10.1016/j.jconrel.2017.11.026] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 01/10/2023]
Abstract
Glioblastoma stem cells (GSCs), which are identified as subpopulation of CD133+/ALDH1+, are known to show resistance to the most of chemotherapy and radiation therapy, leading to the recurrence of tumor in glioblastoma multiforme (GBM) patients. Also, delivery of temozolomide (TMZ), a mainline treatment of GBM, to the GBM site is hampered by various barriers including the blood-brain barrier (BBB). A dual-targeting immunoliposome encapsulating TMZ (Dual-LP-TMZ) was developed by using angiopep-2 (An2) and anti-CD133 monoclonal antibody (CD133 mAb) for BBB transcytosis and specific delivery to GSCs, respectively. The size, zeta potential and drug encapsulation efficiency of Dual-LP-TMZ were 203.4nm in diameter, -1.6mV and 99.2%, respectively. The in vitro cytotoxicity of Dual-LP-TMZ against U87MG GSCs was increased by 425- and 181-folds when compared with that of free TMZ and non-targeted TMZ liposome (LP-TMZ) (10.3μM vs. 4380μM and 1869μM in IC50, respectively). Apoptosis and anti-migration ability of Dual-LP-TMZ in U87MG GSCs were also significantly enhanced comparing with those of free TMZ or LP-TMZ. In vivo study clearly showed a significant reduction in tumor size after intravenous administrations of Dual-LP-TMZ to the orthotopically-implanted brain tumor mice when compared with free TMZ or LP-TMZ. Increased life span (ILS) and median survival time (MST) of tumor-bearing mice were also increased when treated with Dual-LP-TMZ (211.2% in ILS and 49.2days in MST) than with free TMZ (0% in ILS and 23.3day in MST). These data indicate that conjugation of both An2 peptide and CD133 mAb to TMZ-encapsulating liposome is very effective in delivering the TMZ to GSCs via BBB, suggesting a potential use of Dual-LP-TMZ as a therapeutic modality for GBM.
Collapse
Affiliation(s)
- Jung Seok Kim
- Research Center for Cell Fate Control (RCCFC), Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jin-Seok Kim
- Research Center for Cell Fate Control (RCCFC), Drug Information Research Institute (DIRI), College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| |
Collapse
|
3
|
Greanya ED, Taylor SCM, BscPharm FH, Barnett J, Thiessen B. Temozolomide for malignant gliomas in British Columbia: A population-based cost-effectiveness analysis. J Oncol Pharm Pract 2016. [DOI: 10.1191/1078155204jp138oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Study objectives. To evaluate the cost-effectiveness and outcomes achieved in patients with recurrent malignant glioma treated with temozolomide in British Columbia, as compared to previous lomustine use in the same patient population, and to temozolomide literature reports. Outcomes assessed included median overall survival, 6-month overall survival and 6-month progression free survival. Methods. A retrospective analysis was conducted to identify patients who received single-agent temozolomide or lomustine during successive, prespecified time periods. Data was collected on survival, disease progression, duration of therapy, cost of drug, labour and supplies, and successive or prior chemotherapy. Results. Six-month progression free survival (PFS) occurred in 52% and 42.9% of patients in the temozolomide and lomustine cohorts, respectively (P=0.44). Six-month overall survival and median overall survival (OS) were 72% and 40.86 weeks for temozolomide patients and 64.3% and 46.7 weeks for lomustine patients. These outcomes were not statistically different between the two treatment groups. Associated with these outcomes, temozolomide patients received a median of six cycles of drug treatment, with a median cost per patient of $11 660 (CAN). Alternatively, lomustine patients received a median of four cycles with a median cost per patient of $189 (CAN). In the cost-effectiveness analysis for median OS, temozolomide was not a cost-effective alternative, and for 6-month PFS, the incremental cost effectiveness ratio (ICER) of temozolomide was $1261 (CAN) for each additional percent of patients progression free at 6-months. Sensitivity analysis varying both median OS and 6-month PFS resulted in ICER’s of temozolomide ranging from $332 to $3277. Conclusions. No significant differences in outcomes were observed between patients treated with single-agent lomustine or temozolomide. Temozolomide therapy has an incremental cost increase over lomustine of $11 471 per patient. It appears when only survival outcomes and direct treatment costs are considered, lomustine is a more cost-effective treatment strategy in the specific setting of recurrent malignant glioma.
Collapse
Affiliation(s)
- ED Greanya
- University of British Columbia, BC, Canada
| | - SCM Taylor
- British Columbia Cancer Agency (BCCA) Provincial Pharmacy Program, 600 West 10th Avenue, Vancouver, BC, Canada V5Z 4E6
| | - F Hu BscPharm
- British Columbia Cancer Agency (BCCA) Provincial Pharmacy Program, 600 West 10th Avenue, Vancouver, BC, Canada V5Z 4E6
| | - J Barnett
- British Columbia Cancer Agency (BCCA) Provincial Pharmacy Program, 600 West 10th Avenue, Vancouver, BC, Canada V5Z 4E6
| | - B Thiessen
- Division of Medical Oncology, British Columbia Cancer Agency (BCCA), 600 West 10th Avenue, Vancouver, BC, V5Z 4E6 Canada
| |
Collapse
|
4
|
Gwak HS, Yee GT, Park CK, Kim JW, Hong YK, Kang SG, Kim JH, Seol HJ, Jung TY, Chang JH, Yoo H, Hwang JH, Kim SH, Park BJ, Hwang SC, Kim MS, Kim SH, Kim EY, Kim E, Kim HY, Ko YC, Yun HJ, Youn JH, Kim J, Lee B, Lee SH. Temozolomide salvage chemotherapy for recurrent anaplastic oligodendroglioma and oligo-astrocytoma. J Korean Neurosurg Soc 2013; 54:489-95. [PMID: 24527191 PMCID: PMC3921276 DOI: 10.3340/jkns.2013.54.6.489] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/22/2013] [Accepted: 09/30/2013] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To evaluate the efficacy of temozolomide (TMZ) chemotherapy for recurrent anaplastic oligodendroglioma (AO) and anaplastic oligoastrocytoma (AOA). METHODS A multi-center retrospective trial enrolled seventy-two patients with histologically proven AO/AOA who underwent TMZ chemotherapy for their recurrent tumors from 2006 to 2010. TMZ was administered orally (150 to 200 mg/m(2)/day) for 5 days per 28 days until unacceptable toxicity occurred or tumor progression was observed. RESULTS TMZ chemotherapy cycles administered was median 5.3 (range, 1-41). The objective response rate was 24% including 8 cases (11%) of complete response and another 23 patients (32%) were remained as stable disease. Severe side effects (≥grade 3) occurred only in 9 patients (13%). Progression-free survival (PFS) of all patients was a median 8.0 months (95% confidence interval, 6.0-10.0). The time to recurrence of a year or after was a favorable prognostic factor for PFS (p<0.05). Overall survival (OS) was apparently differed by the patient's histology, as AOA patients survived a median OS of 18.0 months while AO patients did not reach median OS at median follow-up of 11.5 months (range 2.7-65 months). Good performance status of Eastern Cooperative Oncology Group 0 and 1 showed prolonged OS (p<0.01). CONCLUSION For recurrent AO/AOA after surgery followed by radiation therapy, TMZ could be recommended as a salvage therapy at the estimated efficacy equal to procarbazine, lomustine, and vincristine (PCV) chemotherapy at first relapse. For patients previously treated with PCV, TMZ is a favorable therapeutic option as 2nd line salvage chemotherapy with an acceptable toxicity rate.
Collapse
Affiliation(s)
- Ho-Shin Gwak
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Gi Taek Yee
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Chul-Kee Park
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Jin Wook Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Yong-Kil Hong
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Seok-Gu Kang
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Jeong Hoon Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Ho Jun Seol
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Tae-Young Jung
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Jong Hee Chang
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Heon Yoo
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | | | - Se-Hyuk Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Bong Jin Park
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Sun-Chul Hwang
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Min Su Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Seon-Hwan Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Eun-Young Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Ealmaan Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Hae Yu Kim
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Young-Cho Ko
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Hwan Jung Yun
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Ji Hye Youn
- Registration Group, Korean Society for Neuro-Oncology, Korea
| | - Juyoung Kim
- Pharmaceutical Benefit Department, Health Insurance Review and Assessment Service, Korea
| | - Byeongil Lee
- Pharmaceutical Benefit Department, Health Insurance Review and Assessment Service, Korea
| | - Seung Hoon Lee
- Registration Group, Korean Society for Neuro-Oncology, Korea
| |
Collapse
|
5
|
Ling Y, Wei K, Zou F, Zhong S. Temozolomide loaded PLGA-based superparamagnetic nanoparticles for magnetic resonance imaging and treatment of malignant glioma. Int J Pharm 2012; 430:266-75. [PMID: 22486964 DOI: 10.1016/j.ijpharm.2012.03.047] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Revised: 02/29/2012] [Accepted: 03/25/2012] [Indexed: 12/12/2022]
Abstract
Polysorbate 80 coated temozolomide-loaded PLGA-based superparamagnetic nanoparticles (P80-TMZ/SPIO-NPs) were successfully synthesized and characterized as drug carriers and diagnosis agent for malignant brain glioma. The mean size of P80-TMZ/SPIO-NPs was 220 nm with narrow hydrodynamic particle size distribution. The superparamagnetic characteristic of P80-TMZ/SPIO-NPs was proved by vibration simple magnetometer. P80-TMZ/SPIO-NPs exhibited high drug loading and encapsulation efficiency as well as good sustained drug release performance for 15 days. MTT assay demonstrated the antiproliferative effect of P80-TMZ/SPIO-NPs for C6 glioma cells. Significant cellular uptake of P80-TMZ/SPIO-NPs was evaluated in C6 glioma cells by fluorescence microscopy, Prussian blue staining, and atomic absorption spectrophotometer (AAS) for qualitative and quantitative study, respectively. MRI scanning analyses in vitro indicated that P80-TMZ/SPIO-NPs could be used as a good MRI contrast agent. Polysorbate 80 coated temozolomide-loaded PLGA-based superparamagnetic nanoparticles could be able to promise a multifunctional theragnostic carrier of brain cancer.
Collapse
Affiliation(s)
- You Ling
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | | | | | | |
Collapse
|
6
|
Cimini A, Ippoliti R. Innovative Therapies against Human Glioblastoma Multiforme. ISRN ONCOLOGY 2011; 2011:787490. [PMID: 22091432 PMCID: PMC3195804 DOI: 10.5402/2011/787490] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/24/2011] [Accepted: 05/25/2011] [Indexed: 01/20/2023]
Abstract
Glioblastoma multiforme is the most invasive and aggressive brain tumor in humans, and despite the latest chemical and radiative therapeutic approaches, it is still scarcely sensitive to these treatments and is generally considered an incurable disease. This paper will focus on the latest approaches to the treatment of this cancer, including the new chemicals such as proautophagic drugs and kinases inhibitors, and differentiating agents. In this field, there have been opening new perspectives as the discovery of possible specific targets such as the EGFRvIII, a truncated form of the EGF receptor. Antibodies against these targets can be used as proapoptotic agents and as possible carriers for chemicals, drugs, radioisotopes, and toxins. In this paper, we review the possible mechanism of action of these therapies, with particular attention to the combined use of toxic substances (for example, immunotoxins) and antiproliferative/differentiating compounds (i.e., ATRA, PPARγ agonists). All these aspects will be discussed in the view of progress clinical trials and of possible new approaches for directed drug formulations.
Collapse
Affiliation(s)
- Annamaria Cimini
- Department of Basic and Applied Biology, University of l'Aquila, Via Vetoio No. 10, 67010 L'Aquila, Italy
| | | |
Collapse
|
7
|
Sure D, Dunn I, Norden A, Anderson WS. Intracerebral hemorrhage secondary to thrombocytopenia in a patient treated with temozolomide. Clin Neurol Neurosurg 2010; 112:741-2. [PMID: 20434832 DOI: 10.1016/j.clineuro.2010.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Revised: 03/08/2010] [Accepted: 04/05/2010] [Indexed: 11/29/2022]
Abstract
Thrombocytopenia related intracerebral hemorrhage (ICH) secondary to chemotherapy induced myelosuppression can be very difficult to manage and may result in the death of the patient. We present the case of a 26-year-old male suffering a fatal ICH in the context of treatment of a high grade glioma with temozolomide. The clinician should be aware that a dose dependent severe and possible irreversible myelotoxicity can occur with temozolomide treatment resulting in substantial morbidity.
Collapse
|
8
|
Treatment of recurrent glioblastoma: can local delivery of mitoxantrone improve survival? J Neurooncol 2008; 88:105-13. [PMID: 18283418 DOI: 10.1007/s11060-008-9540-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2007] [Accepted: 01/28/2008] [Indexed: 10/22/2022]
Abstract
In this study, the records of 276 adult patients with recurrent glioblastoma (GBM) treated at recurrence at our institution between 2004 and 2006 were reviewed for progression-free survival (PFS), overall survival (OS), and toxicity. At recurrence, all patients underwent systemic treatment with temozolomide (200 mg/sqm on days 1-5 every 28 days) until tumor progression. Patients, whose tumor was judged resectable without risk of adjunctive neurological deficit, underwent a second surgery with or without positioning of a Rickam/Ommaya reservoir. The reservoir was used for locoregional chemotherapy with mitoxantrone. Two hundred seventy-six rGBL patients (pts) were divided into three subgroups: A 161 pts treated only with temozolomide, B 50 pts re-operated-on +temozolomide, and C 65 pts re-operated on + temozolomide + locoregional CHT. For group A, the 6 month PFS and 6 month survival (ST) were 39.3 and 43%, respectively, with a median survival time (mST) of 5 months (range 4-6) and 25% of pts alive at 9 months. For group B, the 6 month PFS and 6 month survivors were 64 and 74.1%, respectively, with a mST of 8 months (range 6-10) and 25% of pts alive at 12 months. For group C, the 6 month PFS and 6 month survivors were 70.7 and 87.7%, respectively, with a mST of 11 months (range 9-13) and 25% of pts alive at 18 months (A vs. B vs. C, log-rank P < 0.001) (B vs. C, P = 0.041) (A vs. B P = 0.009). Cox proportional hazard model was used to obtain Hazard Ratio (HR) for type of treatment corrected by age and time (in months) between diagnosis and first recurrence: second tumor debulking was statistically effective for survival, reducing by 36% the risk of death (HR = 0.64; 0.46-0.89), but the most significant favorable prognostic factor for survival was the local delivery of mitoxantrone which reduced the risk of death to 50% (HR = 0.50; 0.38-0.68).
Collapse
|
9
|
Abstract
Malignant primary brain tumors, gliomas, often overexpress both platelet-derived growth factor (PDGF) ligands and receptors providing an autocrine and/or paracrine boost to tumor growth. Glioblastoma multiforme (GBM) is the most frequent glioma. Its aggressive and infiltrative growth renders it extremely difficult to treat. Median survival after diagnosis is currently only 12-14 months. The present review describes the use of retroviral tagging to identify candidate cancer-causing genes that cooperate with PDGF in brain tumor formation. Newborn mice injected intracerebrally with a Moloney murine leukemia retrovirus carrying the sis/PDGF-B oncogene and a replication competent helper virus developed brain tumors with many characteristics of human gliomas. Analysis of proviral integrations in the brain tumors identified almost 70 common insertion sites (CISs). These CISs were named brain tumor loci and harbored known but also putative novel cancer-causing genes. Microarray analysis identified differentially expressed genes in the mouse brain tumors compared to normal brain. Known tumor genes and markers of immature cells were upregulated in the tumors. Tumors developed 13-42 weeks after injection and short latency tumors were further distinguished as fast growing and GBM-like. Long latency tumors resembled slow-growing oligodendrogliomas and contained significantly less integrations as compared to short latency tumors. Several candidate genes tagged in this retroviral screen have known functions in neoplastic transformation and oncogenesis. Some candidates with a previously unknown function in tumorigenesis were found and their putative role in brain tumor formation will be discussed in this review. The results show that proviral tagging may be a useful tool in the search for candidate glioma genes.
Collapse
|
10
|
Abstract
The increasing number of elderly people in the world population has led to a parallel increase in the number of older cancer patients, with over 45% of all cancers in Europe occurring in patients >70 years of age. The increasing tendency to use oral chemotherapy is thus of interest in the elderly, given that both elderly patients and their physicians prefer to use less complex and toxic regimens when such treatments have equivalent efficacy to more complex regimens. However, data from studies designed to evaluate these therapies in the elderly are currently limited. Factors that must be considered before prescribing oral agents to this subset of patients include age-related physiological changes affecting clinical pharmacology, adherence, the patient's capability to self-administer medications, and safety issues concerning the older patient and his or her caregivers. The idea that elderly patients may benefit from the introduction of oral chemotherapy is very fashionable, but to date there is no proof that this approach is as effective as intravenous therapy in this age group, particularly since randomised trials are lacking. This review discusses these issues and reviews current information about the use of specific oral chemotherapeutic drugs for major neoplastic diseases in the elderly.
Collapse
Affiliation(s)
- Sara Lonardi
- Medical Oncology Unit, Cancer Center of the Veneto Region - IRCCS, Padova, Italy
| | | | | | | |
Collapse
|
11
|
Jones-Bolin S, Zhao H, Hunter K, Klein-Szanto A, Ruggeri B. The effects of the oral, pan-VEGF-R kinase inhibitor CEP-7055 and chemotherapy in orthotopic models of glioblastoma and colon carcinoma in mice. Mol Cancer Ther 2006; 5:1744-53. [PMID: 16891460 DOI: 10.1158/1535-7163.mct-05-0327] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CEP-7055, a fully synthetic, orally active N,N-dimethylglycine ester of CEP-5214, a C3-(isopropylmethoxy)-fused pyrrolocarbazole with potent pan-vascular endothelial growth factor receptor (VEGFR) kinase inhibitory activity, has recently completed phase I clinical trials in cancer patients. These studies evaluated the antitumor efficacy of CEP-7055 using orthotopic models of glioblastoma and colon carcinoma in combination with temozolomide, and irinotecan and oxaliplatin, respectively, for their effects on primary and metastatic tumor burden and median survival. Chronic administration of CEP-7055 (23.8 mg/kg/dose) and temozolomide resulted in improvement of median survival of nude mice bearing orthotopic human glioblastoma xenografts compared with temozolomide alone (261 versus 192 days, respectively; P < or = 0.02). Reductions in neurologic dysfunction, brain edema, hemorrhage, and intratumoral microvessel density (CD34 staining) were observed in glioma-bearing mice receiving CEP-7055 alone, temozolomide alone, and the combination of CEP-7055 and temozolomide relative to vehicle and to temozolomide monotherapy. The administration of CEP-7055 in combination with irinotecan (20 mg/kg/dose i.p. x 5 days), and to a lesser degree with oxaliplatin (10 mg/kg/dose i.v.), showed reductions on primary colon carcinoma and hepatic metastatic burden in the CT-26 tumor model relative to that achieved by irinotecan and oxaliplatin monotherapy. These data show the significant efficacy and tolerability of optimal efficacious doses of CEP-7055 when given in combination with temozolomide and irinotecan relative to monotherapy with these cytotoxic agents in preclinical orthotopic glioma and colon carcinoma models and lend support for the use of these treatment regimens in a clinical setting in patients with glioblastoma and colon carcinoma.
Collapse
Affiliation(s)
- Susan Jones-Bolin
- Oncology Research, Cephalon, Inc., 145 Brandywine Parkway, West Chester, PA 19380, USA
| | | | | | | | | |
Collapse
|
12
|
Yang SH, Kim MK, Lee TK, Lee KS, Jeun SS, Park CK, Kang JK, Kim MC, Hong YK. Temozolomide chemotherapy in patients with recurrent malignant gliomas. J Korean Med Sci 2006; 21:739-44. [PMID: 16891823 PMCID: PMC2729901 DOI: 10.3346/jkms.2006.21.4.739] [Citation(s) in RCA: 12] [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] [Indexed: 11/22/2022] Open
Abstract
Numerous studies have demonstrated the clinical activity of temozolomide, a second-generation alkylating agent, against malignant brain tumors, however, its activity has not been reported in an Asian population. This study analyzed the efficacy and toxicity of temozolomide in 25 adult patients with recurrent or progressive malignant gliomas after surgery and standard radiation therapy with or without chemotherapy, enrolled in our institution since July 2000. Sixteen patients had glioblastoma multiforme (GBM), six with anaplastic astrocytoma, and three with anaplastic oligodendroglioma. Of the 25 patients, 3 (12%) achieved a complete response (CR), 8 (32%) achieved a partial response (PR), 6 (24%) had stable disease (SD), and 8 (32%) had progressive disease (PD). Two patients achieved a CR, 4 patients achieved a PR, 3 patients had SD and 7 patients had PD in GBM, and 1 patient achieved a CR, 4 patients achieved a PR, 3 patients had SD, 1 patient had PD in the non-GBM patients. Median progression free survival was 8 weeks in GBM and 22 weeks in the non-GBM patients. The median overall survival of each group was 17 weeks and 28 weeks. Temozolomide demonstrated moderate activity in recurrent and progressive malignant gliomas without serious toxicity.
Collapse
Affiliation(s)
- Seung-Ho Yang
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Moon-Kyu Kim
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Tae-Kyu Lee
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Kwan-Sung Lee
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Chun-Kun Park
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Joon-Ki Kang
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Moon-Chan Kim
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| | - Yong-Kil Hong
- Department of Neurosurgery, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
13
|
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: 13] [Impact Index Per Article: 0.7] [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
|
14
|
Trivedi RN, Almeida KH, Fornsaglio JL, Schamus S, Sobol RW. The role of base excision repair in the sensitivity and resistance to temozolomide-mediated cell death. Cancer Res 2005; 65:6394-400. [PMID: 16024643 DOI: 10.1158/0008-5472.can-05-0715] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
DNA-alkylating agents have a central role in the curative therapy of many human tumors; yet, resistance to these agents limits their effectiveness. The efficacy of the alkylating agent temozolomide has been attributed to the induction of O6-MeG, a DNA lesion repaired by the protein O6-methylguanine-DNA methyltransferase (MGMT). Resistance to temozolomide has been ascribed to elevated levels of MGMT and/or reduced mismatch repair. However, >80% of the DNA lesions induced by temozolomide are N-methylated bases that are recognized by DNA glycosylases and not by MGMT, and so resistance to temozolomide may also be due, in part, to robust base excision repair (BER). We used isogenic cells deficient in the BER enzymes DNA polymerase-beta (pol-beta) and alkyladenine DNA glycosylase (Aag) to determine the role of BER in the cytotoxic effect of temozolomide. Pol-beta-deficient cells were significantly more susceptible to killing by temozolomide than wild-type or Aag-deficient cells, a hypersensitivity likely caused by accumulation of BER intermediates. RNA interference-mediated pol-beta suppression was sufficient to increase temozolomide efficacy, whereas a deficiency in pol-iota or pol-lambda did not increase temozolomide-mediated cytotoxicity. Overexpression of Aag (the initiating BER enzyme) triggered a further increase in temozolomide-induced cytotoxicity. Enhanced Aag expression, coupled with pol-beta knockdown, increased temozolomide efficacy up to 4-fold. Furthermore, loss of pol-beta coupled with temozolomide treatment triggered the phosphorylation of H2AX, indicating the activation of the DNA damage response pathway as a result of unrepaired lesions. Thus, the BER pathway is a major contributor to cellular resistance to temozolomide and its efficacy depends on specific BER gene expression and activity.
Collapse
Affiliation(s)
- Ram N Trivedi
- Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania 15213-1863, USA
| | | | | | | | | |
Collapse
|
15
|
The results of postoperative irradiation in malignant glioma patients. Rep Pract Oncol Radiother 2005. [DOI: 10.1016/s1507-1367(05)71079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
16
|
Abstract
The current standard of care for patients with high-grade glioma is resection followed by radiotherapy. Adjuvant chemotherapy is not widely accepted because of the low sensitivity of gliomas to traditional antineoplastic agents, the poor penetration of most drugs across the blood-brain barrier, and the significant systemic toxicity associated with current agents. However, nitrosoureas and, subsequently, temozolomide (Temodar [US], Temodal [international]; Schering-Plough Corporation, Kenilworth, NJ), a novel alkylating agent, cross the blood-brain barrier and have activity against gliomas. Nitrosoureas have been studied in phase III trials in the adjuvant setting. In individual trials, chemotherapy did not increase median survival but did increase the proportion of patients surviving >/=18 months by 15%. Only with large meta-analyses did the addition of chemotherapy achieve a statistically significant improvement in median survival. Currently there is no means of identifying which patients will benefit from adjuvant chemotherapy, but nitrosoureas and temozolomide are well tolerated in most patients, justifying the administration of adjuvant chemotherapy to all newly diagnosed patients with malignant glioma.
Collapse
Affiliation(s)
- Lisa M DeAngelis
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|