851
|
Khil MS, Kolozsvary A, Apple M, Kim JH. Increased tumor cures using combined radiosurgery and BCNU in the treatment of 9l glioma in the rat brain. Int J Radiat Oncol Biol Phys 2000; 47:511-6. [PMID: 10802380 DOI: 10.1016/s0360-3016(00)00428-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE Radiosurgery refers to the delivery of high, single focused beams of ionizing radiation to defined intracranial lesions. 1,3 Bis[2-chloroethyl]-1-nitrosourea (BCNU) and cis-diammine-1, 1-cyclobutane-dicarboxylate platinum (II) (carboplatin) are commonly used cytotoxic agents for the treatment of malignant gliomas of the brain. Drug therapies have exhibited a modest enhanced cell killing when combined with radiation in experimental animal tumor systems. The purpose of the present study was to investigate the role of cytotoxic drugs, such as BCNU and carboplatin, in combination with a single high dose of radiosurgery on the tumor control rates of 9L tumors in the rat brain. METHODS AND MATERIALS Combined radiosurgery (25 Gy single dose) and/or chemotherapy (a single dose of BCNU, 7 mg/kg, i.p. 1.5 or 16 h prior to or 16 h after irradiation or a single dose of carboplatin, 30 mg/kg, administered either 1 h or 4 h prior to irradiation) was delivered 12 days after stereotactic tumor implantation. For dose escalation study, 4-10 mg/kg of BCNU was used. RESULTS The radiation alone group showed a dose-dependent survival. A single dose of 25 Gy to the control group resulted in an increase of the median survival time from 20 days to 42 days, but all animals died of the tumor in 50 days. A significant prolongation of the median survival time of animals was more than 100 days, resulting in animal cures of 50% or more when combined with radiosurgery (25 Gy) and BCNU (7 mg/kg). BCNU alone did not prolong the median survival time of the animal with the 9L brain tumor. In contrast, there was no survival improvement when the animals were treated with combined radiosurgery and carboplatin. None of the long-term surviving animals showed any significant brain tissue damage as evaluated by histopathology and clinical observations. CONCLUSION The data clearly suggest that the combined modalities of radiosurgery and concomitant BCNU represent an effective therapeutic regimen in the treatment of radioresistant human malignant gliomas of the brain. This study represents the first experimental report of the effectiveness of combined chemotherapy and radiosurgery.
Collapse
Affiliation(s)
- M S Khil
- Department of Radiation Oncology, Henry Ford Health System, Detroit, MI 48202-4689, USA.
| | | | | | | |
Collapse
|
852
|
Patel S, Breneman JC, Warnick RE, Albright RE, Tobler WD, van Loveren HR, Tew JM. Permanent iodine-125 interstitial implants for the treatment of recurrent glioblastoma multiforme. Neurosurgery 2000; 46:1123-8; discussion 1128-30. [PMID: 10807244 DOI: 10.1097/00006123-200005000-00019] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Brachytherapy with temporary implants may prolong survival in patients with recurrent glioblastoma multiforme (GBM), but it is associated with relatively high costs and morbidity. This study reports the time to progression and survival after permanent implantation of iodine-125 seeds for recurrent GBM and examines factors predictive of outcome. METHODS Forty patients with recurrent GBM were treated with maximal resection plus permanent placement of iodine-125 seeds into the tumor bed. A total dose of 120 to 160 Gy was administered, and patients were followed up with magnetic resonance imaging scans every 2 to 3 months. RESULTS Actuarial survival from the time of implantation was 47 weeks, with 7 of 40 patients still alive at a median of 59 weeks after implantation. Survival was significantly better for patients younger than 60 years, and a trend for longer survival was demonstrated with gross total resection and tumors with a low MIB-1 (a nuclear antigen present in all cell cycles of proliferating cells) staining index. Median time to progression was 25 weeks and, on multivariate analysis, was favorably influenced by gross total resection and patient age younger than 60 years. After implantation, 27 of 30 patients with failure had a local component to the failure. No patient developed symptoms attributable to radiation necrosis or injury. CONCLUSION Permanent iodine-125 implants for recurrent GBM result in survival comparable with that described in previous reports on temporary implants, but with less morbidity. Results are most favorable for patients who are younger than 60 years, and who undergo gross total resection. Despite this aggressive treatment, most patients die as a consequence of locally recurrent disease.
Collapse
Affiliation(s)
- S Patel
- The Neuroscience Institute, Division of Radiation Oncology, University of Cincinnati Medical Center, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
853
|
Postma TJ, Heimans JJ, Luykx SA, van Groeningen CJ, Beenen LF, Hoekstra OS, Taphoorn MJ, Zonnenberg BA, Klein M, Vermorken JB. A phase II study of paclitaxel in chemonaïve patients with recurrent high-grade glioma. Ann Oncol 2000; 11:409-13. [PMID: 10847458 DOI: 10.1023/a:1008376123066] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The prognosis of malignant gliomas remains poor. In recurrent disease, chemotherapy can be considered. PATIENTS AND METHODS In this phase II study we determined the anti-tumour efficacy of paclitaxel 200 mg/m2 in a three-hour intravenous infusion every three weeks in chemonaïve patients with recurrent high-grade glioma in terms of response, survival, and quality of life. RESULTS In 17 patients (14 glioblastoma multiforme, 3 anaplastic astrocytoma) 69 paclitaxel cycles were administered. Partial or complete responses were not observed. Stable disease for four to six months was observed in five patients (29%). Median time to progression and median survival were two and 10 months, respectively. Toxicity due to paclitaxel was as to be expected and minor in most cases. Quality of life and mood estimates appeared rather stable over time. CONCLUSIONS We conclude that three-weekly 200 mg/m2 paclitaxel chemotherapy for patients with recurrent high-grade gliomas did not lead to major complications or adverse effects on quality of life and mood. However, this therapy is of only very limited value in terms of response and survival in such patients.
Collapse
Affiliation(s)
- T J Postma
- Department of Neurology, University Hospital Vrije Universiteit Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
854
|
Affiliation(s)
- E Galanis
- Division of Medical Oncology, Mayo Clinic and Foundation, Rochester, MN 55905, USA
| | | |
Collapse
|
855
|
Benoit JP, Faisant N, Venier-Julienne MC, Menei P. Development of microspheres for neurological disorders: from basics to clinical applications. J Control Release 2000; 65:285-96. [PMID: 10699288 DOI: 10.1016/s0168-3659(99)00250-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Drug delivery to the central nervous system remains a challenging area of investigation for both basic and clinical neuroscientists. Numerous drugs are generally excluded from blood to brain transfer due to the negligible permeability of the brain capillary endothelial wall, which makes up the blood brain barrier in vivo. For several years, we have explored the potential applications of the microencapsulation of therapeutic agents to provide local controlled drug release in the central nervous system. Due to their size, these microparticles can be easily implanted by stereotaxy in discreet, precise and functional areas of the brain without damaging the surrounding tissue. This type of implantation avoids the inconvenient insertion of large implants by open surgery and can be repeated if necessary. We have established the compatibility of poly(lactide-co-glycolide) microspheres with brain tissues. Presently, the most developed applications concern Neurology and Neuro-oncology, with local delivery of neurotrophic factors and antimitotic drugs into neurodegenerative lesions and brain tumours, respectively. The drugs that had been encapsulated by our group included nerve growth factor (NGF), 5-fluorouracil (5-FU), idoxuridine and BCNU. Preclinical studies have been performed with each drug. Studies with NGF are reported as an example. A phase I/II clinical trial has been carried out in patients with newly diagnosed glioblastomas to assess the potentialities of 5-FU-loaded microspheres when intracranially implanted.
Collapse
Affiliation(s)
- J P Benoit
- UPRES EA 2169, Faculté de Pharmacie, Université d'Angers, Angers, France.
| | | | | | | |
Collapse
|
856
|
Bruce JN, Falavigna A, Johnson JP, Hall JS, Birch BD, Yoon JT, Wu EX, Fine RL, Parsa AT. Intracerebral clysis in a rat glioma model. Neurosurgery 2000; 46:683-91. [PMID: 10719865 DOI: 10.1097/00006123-200003000-00031] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Intracerebral clysis (ICC) is a new term we use to describe convection-enhanced microinfusion into the brain. This study establishes baseline parameters for preclinical, in vivo, drug investigations using ICC in a rat glioma model. METHODS Intracranial pressure was measured, with an intraparenchymal fiber-optic catheter, in male Fischer rats 10, 15, 20, and 25 days after implantation of C6 glioma cells in the right frontal lobe (n = 80) and in control rats without tumor (n = 20), before and during ICC. A 25% albumin solution (100 microl) was infused through an intratumoral catheter at 0.5, 1.0, 2.0, 3.0, and 4.0 microl/min. Infusate distribution was assessed by infusion of fluorescein isothiocyanate-dextran (Mr 20,000), using the aforementioned parameters (n = 36). Brains were sectioned and photographed under ultraviolet light, and distribution was calculated by computer analysis (NIH Image for Macintosh). Safe effective drug distribution was demonstrated by measuring tumor sizes and apoptosis in animals treated with N,N'-bis(2-chloroethyl)-N-nitrosourea via ICC, compared with untreated controls. Magnetic resonance imaging noninvasively confirmed tumor growth before treatment. RESULTS Intracranial pressure increased with tumor progression, from 5.5 mm Hg at baseline to 12.95 mm Hg on Day 25 after tumor cell implantation. Intracranial pressure during ICC ranged from 5 to 21 mm Hg and was correlated with increasing infusion volumes and increasing rates of infusion. No toxicity was observed, except at the higher ends of the tumor size and volume ranges. Fluorescein isothiocyanate-dextran distribution was greater with larger infusion volumes (30 microl versus 10 microl, n = 8, P < 0.05). No significant differences in distribution were observed when different infusion rates were compared while the volume was kept constant. At tolerated flow rates, the volumes of distribution were sufficient to promote adequate drug delivery to tumors. N,N'-Bis(2-chloroethyl)-N-nitrosourea treatment resulted in significant decreases in tumor size, compared with untreated controls. CONCLUSION The C6 glioma model can be easily modified to study aspects of interstitial delivery via ICC and the application of ICC to the screening of potential antitumor agents for safety and efficacy.
Collapse
Affiliation(s)
- J N Bruce
- Department of Neurological Surgery, Neurological Institute of New York, New York 10032, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
857
|
Abstract
The blood brain barrier (BBB) and the systemic toxicity of conventional chemotherapy present obstacles to the success of future blood-borne drug therapies of brain tumors. The work with polymer-encapsulated cancer drugs suggests an alternative and more focused treatment approach. Our experimental strategy integrates direct intracerebral drug delivery, sustained drug release from liposomes or polymer implants, and increased targeting of the drug either by chemically modifying the drug or by using tumor-specific carriers. This review will present some of the recent work on targeted drug delivery for brain cancer treatment.
Collapse
Affiliation(s)
- R L Gutman
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| | | | | |
Collapse
|
858
|
Engelhard HH. Tumor bed cyst formation after BCNU wafer implantation: report of two cases. SURGICAL NEUROLOGY 2000; 53:220-4. [PMID: 10773252 DOI: 10.1016/s0090-3019(00)00162-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Interstitial implantation of BCNU wafers is currently used to treat glioblastoma multiforme. Known complications of BCNU wafer implantation include abnormalities of wound healing (including CSF leak), edema formation, and intracranial infection. The purpose of this report is to alert neurosurgeons to an additional potential side effect: formation of a cystic mass within the implanted tumor bed. CASE PRESENTATIONS Two patients are described: a 54-year-old male, who presented with a large right parieto-occipital mass, and a 47-year-old woman with a large right frontal lobe tumor. Both tumors were found on initial craniotomy to be glioblastoma multiforme; both recurred rapidly despite radiation therapy. Patients were treated with a second craniotomy for tumor resection and placement of BCNU wafers. After implantation, the first patient did well for 6 weeks, then developed lethargy, headaches, and vomiting. CT scan showed a large cyst at the craniotomy site; this required reoperation for drainage. The second patient had a seizure, deterioration of mental status, and progressive hemiparesis 10 days after wafer implantation. CT scan again showed that a large cyst had formed in the area of the previous surgery; she also required reoperation. In each case, minimal tumor and no evidence of infection were found. Within a few more weeks, each patient succumbed to progressive disease. CONCLUSIONS The hypodense, roughly spherical cysts clearly demonstrated clinically significant mass effect, and required reoperation despite treatment with high-dose corticosteroids. Neurosurgeons should be alert to the possibility of tumor bed cyst formation in patients treated with interstitial BCNU wafers.
Collapse
Affiliation(s)
- H H Engelhard
- Department of Neurosurgery, The University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
859
|
Langer R. Biomaterials in drug delivery and tissue engineering: one laboratory's experience. Acc Chem Res 2000; 33:94-101. [PMID: 10673317 DOI: 10.1021/ar9800993] [Citation(s) in RCA: 434] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This Account reviews our laboratory's research in biomaterials. In one area, drug delivery, we discuss the development of materials that are capable of releasing macromolecules such as proteins and peptides, intelligent delivery systems based on magnetism or microchip technology, new degradable materials such as polyanhydrides, and noninvasive approaches for delivering molecules through the skin and lungs. A second area, tissue engineering, is also discussed. New polymer systems for creating cartilage, blood vessels, nerves, and other tissues are examined.
Collapse
Affiliation(s)
- R Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139-4307, USA
| |
Collapse
|
860
|
Abstract
BACKGROUND Patients with isolated supratentorial brain tumours, presumed to be primary on imaging, have two surgical management options - biopsy or resection. Surgical opinions appear to be equally divided when considering the relative risks and benefits of these two procedures. OBJECTIVES To estimate the clinical effectiveness of radical surgical resection compared to simple biopsy in patients with malignant glioma. SEARCH STRATEGY Electronic database searches of COCHRANE CONTROLLED TRIALS REGISTER (including the Cochrane Cancer Network Specialised Register of Trials), MEDLINE, CANCERLIT, EMBASE, BIOSIS and SCIENCE CITATION INDEX. Hand searching the references of all identified studies; hand searching the Journal of Neuro-Oncology over the previous 10 years, including all conference abstracts; personal communication. SELECTION CRITERIA Randomised and clinical controlled trials were included if they compared biopsy to resection, or looked at effect of extent of resection on survival, time to progression or quality of life, for malignant glioma patients of all ages. DATA COLLECTION AND ANALYSIS Studies were to be identified, critically appraised and data extracted by the author (SEM). For dichotomous data, Peto odds ratios (OR) with 95% confidence intervals (CI) were hoped to have been estimated. Normal continuous data were to have been summated using the weighted mean difference (WMD). MAIN RESULTS The electronic database search yielded 2100 citations. Of these, two articles were identified for possible inclusion, however both were excluded. The hand search and personal communication were similarly unproductive. No studies were included in the review and no data was synthesised. REVIEWER'S CONCLUSIONS Given that no qualifying studies were identified and because this is an important issue, both in terms of patient risk and benefit and health economics, the author feels it important to conduct a randomised controlled trial in this subject.
Collapse
Affiliation(s)
- S E Metcalfe
- c/o Edinburgh Centre of Neuro-Oncology, Western General Hospital, Crewe Road, Edinburgh, Scotland, UK, EH4 2XU.
| |
Collapse
|
861
|
Mairs RJ, Wideman CL, Angerson WJ, Whateley TL, Reza MS, Reeves JR, Robertson LM, Neshasteh-Riz A, Rampling R, Owens J, Allan D, Graham DI. Comparison of different methods of intracerebral administration of radioiododeoxyuridine for glioma therapy using a rat model. Br J Cancer 2000; 82:74-80. [PMID: 10638969 PMCID: PMC2363176 DOI: 10.1054/bjoc.1999.0879] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Auger electron emitting agent 5-[125I]iodo-2'-deoxyuridine (i.e. [125I]IUdR) holds promise for the treatment of residual glioma after surgery because this thymidine analogue kills only proliferating cells. However, malignant cells which are not synthesizing DNA during exposure to the radiopharmaceutical will be spared. To determine whether tumour incorporation of [125I]IUdR could be enhanced by protracted administration, we used a C6 cell line, growing in the brains of Wistar rats, as a glioma model and compared three methods of intracerebral delivery of [125I]IUdR. Twenty-four hours after administration of drug, autoradiography of brain sections demonstrated nuclear uptake of the radiopharmaceutical in cells throughout tumour while normal brain cells remained free of radioactivity. The [125I]IUdR labelling indices (% +/- s.e.m.) achieved were 6.2 (0.4) by single injection, 22.5 (4.1) using a sustained release polymer implant (poly(lactide-co-glycolide)) and 34.3 (2.0) by mini-osmotic pump. These results emphasize the need for a sustained delivery system as a prerequisite for effective treatment. These findings are also encouraging for the development of a sustained release system for radiolabelled IUdR for use in the treatment of intracranial tumours, particularly in the immediate postoperative setting.
Collapse
Affiliation(s)
- R J Mairs
- Department of Radiation Oncology, University of Glasgow, CRC Beatson Laboratories, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
862
|
|
863
|
Sampath P, Long DM, Brem H. The Hunterian Neurosurgical Laboratory: The First 100 Years of Neurosurgical Research. Neurosurgery 2000. [DOI: 10.1093/neurosurgery/46.1.184] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Prakash Sampath
- Department of Neurological Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Donlin M. Long
- Department of Neurological Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Henry Brem
- Department of Neurological Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
864
|
Cho KH, Hall WA, Gerbi BJ, Higgins PD, McGuire WA, Clark HB. Single dose versus fractionated stereotactic radiotherapy for recurrent high-grade gliomas. Int J Radiat Oncol Biol Phys 1999; 45:1133-41. [PMID: 10613305 DOI: 10.1016/s0360-3016(99)00336-3] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE To evaluate the efficacy of stereotactic radiotherapy (SRT) in patients with recurrent high-grade gliomas by comparing two different treatment regimens, single dose or fractionated radiotherapy. METHODS AND MATERIALS Between April 1991 and January 1998, 71 patients with recurrent high-grade gliomas were treated with SRT. Forty-six patients (65%) were treated with single dose radiosurgery (SRS) and 25 patients (35 %) with fractionated stereotactic radiotherapy (FSRT). For the SRS group, the median radiosurgical dose of 17 Gy was delivered to the median of 50% isodose surface (IDS) encompassing the target. For the FSRT group, the median dose of 37.5 Gy in 15 fractions was delivered to the median of 85% IDS. RESULTS Actuarial median survival time was 11 months for the SRS group and 12 months for the FSRT group (p = 0.3, log-rank test). Variables predicting longer survival were younger age (p = 0.006), lower grade (p = 0.0006), higher Karnofsky Performance Scale (KPS) (p = 0.0005), and smaller tumor volume (p = 0.02). Patients in the SRS group had more favorable prognostic factors, with median age of 48 years, KPS of 70, and tumor volume of 10 ml versus median age of 53 years, KPS of 60, and tumor volume of 25 ml in the FSRT group. Late complications developed in 14 patients in the SRS group and 2 patients in the FSRT group (p<0.05). CONCLUSION Given that FSRT patients had comparable survival to SRS patients, despite having poorer pretreatment prognostic factors and a lower risk of late complications, FSRT may be a better option for patients with larger tumors or tumors in eloquent structures. Since this is a nonrandomized study, further investigation is needed to confirm this and to determine an optimal dose/fractionation scheme.
Collapse
Affiliation(s)
- K H Cho
- Department of Therapeutic Radiology and Radiation Oncology, University of Minnesota Hospital and Clinic, Minneapolis 55455, USA.
| | | | | | | | | | | |
Collapse
|
865
|
Kreth FW, Berlis A, Spiropoulou V, Faist M, Scheremet R, Rossner R, Volk B, Ostertag CB. The role of tumor resection in the treatment of glioblastoma multiforme in adults. Cancer 1999. [DOI: 10.1002/(sici)1097-0142(19991115)86:10<2117::aid-cncr33>3.0.co;2-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
866
|
Uhrich KE, Cannizzaro SM, Langer RS, Shakesheff KM. Polymeric systems for controlled drug release. Chem Rev 1999; 99:3181-98. [PMID: 11749514 DOI: 10.1021/cr940351u] [Citation(s) in RCA: 1846] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- K E Uhrich
- Department of Pharmaceutical Science, University of Nottingham, Nottingham NG7-2RD, U.K
| | | | | | | |
Collapse
|
867
|
Abstract
It is important that before treatment is initiated, the precise diagnosis of brain tumor has been made and fits with the clinical, radiographic, and histologic findings. With low-grade gliomas, maximal surgical resection should be attempted. We delay radiation therapy unless the patient has significant neurologic symptoms that could be alleviated by treatment. Patients with glioblastoma multiforme (GBM) and anaplastic astrocytoma should undergo maximum surgical resection, postoperative radiation therapy, and adjuvant chemotherapy with bischloroethylnitrosourea (BCNU) or intensive procarbazine, chloroethylcyclohexylnitrosourea (CCNU), and vincristine (PCV) therapy, respectively. Patients with anaplastic oligodendroglioma (AO) and mixed anaplastic oligodendroglioma-astrocytoma (AOA) should undergo maximal surgical resection, postoperative radiation therapy, and intensive adjuvant PCV therapy. Meningiomas are usually managed with surgery alone; radiation therapy is used if the meningioma is malignant or recurs. Tamoxifen or hydroxyurea may be useful for recurrent meningiomas. Patients with primary central nervous system (CNS) lymphoma undergo biopsy; surgical resection of the tumor is not necessary. We treat immunocompetent patients with multimethod therapy. Patients with AIDS and primary CNS lymphoma receive radiation alone. Because the prognosis for primary CNS lymphomas has not changed significantly in the past 20 years, patients with these tumors should be enrolled in clinical trials if possible. Novel approaches, such as targeting angiogenesis, using signal transduction or invasion, or employing genes affecting growth control, are being pursued.
Collapse
|
868
|
Yapp DT, Lloyd DK, Zhu J, Lehnert S. Radiosensitization of a mouse tumor model by sustained intra-tumoral release of etanidazole and tirapazamine using a biodegradable polymer implant device. Radiother Oncol 1999; 53:77-84. [PMID: 10624857 DOI: 10.1016/s0167-8140(99)00123-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND AND PURPOSE Drug toxicities are often a limiting factor in long term treatment regimes used in conjunction with radiotherapy. If the drug could be localized to the tumor site and released slowly, then optimal, intra-tumoral drug concentrations could be achieved without the cumulative toxicity associated with repeated systemic drug dosage. In this paper we describe the use of a biodegradable polymer implant for sustained intra-tumoral release of high concentrations of drugs targeting hypoxic cells. MATERIALS AND METHODS The RIF-1 tumor was implanted subcutaneously or intramuscularly in C3H mice and irradiated with 60Co gamma rays. The drug delivery device was the co-polymer CPP-SA;20:80 into which the drug was homogeneously incorporated. The hypoxic radiosensitizer Etanidazole or the bioreductive drug Tirapazamine were delivered intra-tumorally by means of implanted polymer rods containing the drugs. Tumor growth delay (TGD) was used as the end point in these experiments. RESULTS Both Etanidazole and Tirapazamine potentiated the effects of acute and fractionated radiation in the intra-muscular tumors but neither drug was effective in sub-cutaneous tumors. Since both drugs target hypoxic cells we hypothesized that the lack of effect in the subcutaneous tumor was attributable to the smaller size of the hypoxic fraction in this tumor model. This was confirmed using the hypoxia marker EF5. CONCLUSIONS These results indicate that the biodegradable polymer implant is an effective vehicle for the intra-tumoral delivery of Etanidazole and Tirapazamine and that, in conjunction with radiation, this approach could improve treatment outcome in tumors which contain a sub-population of hypoxic, radioresistant cells.
Collapse
Affiliation(s)
- D T Yapp
- Department of Radiation Oncology, McGill University, Montréal, Quebec, Canada
| | | | | | | |
Collapse
|
869
|
Yuan X, Tabassi K, Williams JA. Implantable polymers for tirapazamine treatments of experimental intracranial malignant glioma. RADIATION ONCOLOGY INVESTIGATIONS 1999; 7:218-30. [PMID: 10492162 DOI: 10.1002/(sici)1520-6823(1999)7:4<218::aid-roi3>3.0.co;2-c] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Malignant gliomas remain refractory to intensive radiotherapy and cellular hypoxia enhances clinical radioresistance. Under hypoxic conditions, the benzotriazine di-N-oxide (3-amino-1,2,4-benzotriazine 1,4-dioxide) (tirapazamine) is reduced to yield a free-radical intermediate that results in DNA damage and cellular death. For extracranial xenografts, tirapazamine treatments have shown promise. We therefore incorporated tirapazamine into the synthetic, biodegradable polymer, measured the release, and tested the efficacy both alone and in combination with external beam radiotherapy in the treatment of experimental intracranial human malignant glioma xenografts. The [(poly(bis(p-carboxyphenoxy)-propane) (PCPP):sebacic acid (SA) (PCPP:SA ratio 20:80)] polymer was synthesized. The PCPP:SA polymer and solid tirapazamine were combined to yield proportions of 20% or 30% (wt/wt). Polymer discs (3 x 2 mm) (10 mg) were incubated (PBS, 37 degrees C), and the proportion of the drug released vs. time was recorded. Male nu/nu nude mice were anesthetized and received intracranial injections of 2 x 10(5) U251 human malignant glioma cells. For single intraperitoneal (i.p.) drug and/or external radiation treatments, groups of mice had i.p. 0.3 mmol/kg tirapazamine, 5 Gy cranial irradiation, or combined treatments on day 8 after inoculation. For fractionated drug and radiation treatments, mice had i.p. 0.15 mmol/kg tirapazamine, 5 Gy radiation, or combined treatments on days 8 and 9 after inoculation. For intracranial (i.c.) polymer treatments, mice had craniectomies and intracranial placement of polymer discs at the site of cellular inoculation. The maximally tolerated percentage loading of tirapazamine in the polymer.disc was determined. On day 7 after inoculation, groups of mice had i.c. empty or 3% tirapazamine alone or combined with radiation (5 Gy x 2 doses) or combined with i.p. drug (0.15 mmol/kg x 2 doses on days 8 and 9). Survival was recorded. Polymers showed controlled, protracted in vitro release for over 100 days. The 5 Gy x 1 treatment resulted in improved survival; 28.5 +/- 3.7 days (P = 0.01 vs. controls), while the single i.p. 0.3 mmol/kg tirapazamine treatment, 17.5 +/- 1.9 days (P = NS) and combined treatments; 21.5 +/- 5.0 days (P = NS) were not different. The fractionated treatments: 5 Gy x 2, i.p. 0.15 mmol/kg tirapazamine x 2 and the combined treatments resulted in improved survival: 44.5 +/- 3.9 (P < 0.001), 24.5 +/- 2.3 (P = 0.05) and 50.0 +/- 6.0 (P < 0.001), respectively. Survival after intracranial empty polymer was 16.5 +/- 3.0 days and increased to 31.0 +/- 3.0 (P = 0.003) days when combined with the 5 Gy x 2 treatment. The survival after the polymer bearing 3% tirapazamine alone vs. combined with radiation was not different. The combined 3% tirapazamine polymer, i.p. tirapazamine, and radiation treatments resulted in both early deaths and the highest long-term survivorship. The basis for potential toxicity is discussed. We conclude that implantable biodegradable polymers provide controlled intracranial release for treatment of experimental glioma. For treatment of malignant gliomas, the combination of continuous polymer-mediated delivery and fractionated systemic delivery of tirapazamine with external beam radiotherapy warrants further exploration.
Collapse
Affiliation(s)
- X Yuan
- Department of Oncology, Johns Hopkins Oncology Center, Baltimore, Maryland 21205, USA
| | | | | |
Collapse
|
870
|
Freeman TB, Vawter DE, Leaverton PE, Godbold JH, Hauser RA, Goetz CG, Olanow CW. Use of placebo surgery in controlled trials of a cellular-based therapy for Parkinson's disease. N Engl J Med 1999; 341:988-92. [PMID: 10498497 DOI: 10.1056/nejm199909233411311] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- T B Freeman
- University of South Florida, Tampa 33606, USA
| | | | | | | | | | | | | |
Collapse
|
871
|
Abrey LE, Rosenblum MK, Papadopoulos E, Childs BH, Finlay JL. High dose chemotherapy with autologous stem cell rescue in adults with malignant primary brain tumors. J Neurooncol 1999; 44:147-53. [PMID: 10619498 DOI: 10.1023/a:1006383400353] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
High dose chemotherapy (HDCT) with autologous (bone marrow or peripheral blood) stem cell rescue (ASCR) has had success in the treatment of some malignant pediatric brain tumors. We report a series of adults enrolled in one of three HDCT and ASCR protocols for malignant primary brain tumors. Overall toxic mortality was 18%; chemotherapy regimen, tumor type, and prior treatment did not predict transplant-related mortality. Patients over the age of 30 had a higher rate of toxic mortality. Patients with recurrent medulloblastoma had a significant improvement in long-term survival (median: 34 months) as compared with historical reports; two patients with glioblastoma survive beyond four years without progression, but overall, a significant improvement in long-term survival could not be demonstrated for malignant gliomas.
Collapse
Affiliation(s)
- L E Abrey
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | |
Collapse
|
872
|
Chen W, Lu DR. Carboplatin-loaded PLGA microspheres for intracerebral injection: formulation and characterization. J Microencapsul 1999; 16:551-63. [PMID: 10499836 DOI: 10.1080/026520499288753] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The purpose of this study is to prepare and characterize injectable carboplatin-loaded poly(D,L,-lactic-co-glycolic) acid copolymer (PLGA) microspheres for the intracerebral treatment of malignant glioma. The microspheres were prepared by an acetone/mineral oil emulsion and solvent evaporation method. Preparation variables were optimized and the following processing conditions resulted in the highest drug loading and best yields of the microspheres compared with those prepared with the other variables: the PLGA concentration was 8% (w/w) in the internal phase; the emulsifier (Span 80) concentration was 8% (w/w) in the external phase; the ratio of the internal phase: the external phase was 1:8; the stirring speed was 1500 rpm; the emulsion time was 15 min; the solvent evaporation time was 3.75 hr. Microspheres so prepared were analysed for size distribution, drug loading, in vitro release and morphological characteristics. The drug release in phosphate buffer solution started with a 10-day slow release period, followed by a fast near zero order release period from 12 to 22 days. The carboplatin release in brain homogenate was slower than in phosphate buffer solution. The morphological changes of the microspheres during the in vitro degradation correlated with the drug relase profile. In conclusion, the carboplatin-loaded PLGA microspheres were specifically prepared to meet the specification as an injectable and biodegradable brain implant.
Collapse
Affiliation(s)
- W Chen
- Department of Pharmaceutics, College of Pharmacy, University of Georgia, Athens 30602, USA
| | | |
Collapse
|
873
|
Abstract
This paper reports the development of three-dimensional simulations to study the effect of various factors on the delivery of 1-3-bis(2-chloroethyl)-1-nitrosourea (BCNU) to brain tumors. The study yields information on the efficacy of various delivery methods, and the optimal location of polymer implantation. Two types of drug deliveries, namely, systemic administration and controlled release from polymers, were simulated using fluid dynamics analysis package (FIDAP) to predict the temporal and spatial variation of drug distribution. Polymer-based delivery provides higher mean concentration, longer BCNU exposure time and reduced systemic toxicity than bolus injection. Polymer implanted in the core gives higher concentration of drug in both the core and viable zone than the polymer in the viable zone case. The penetration depth of BCNU is very short. This is because BCNU can get drained out of the system before diffusing to any appreciable distance. Since transvascular permeation is the dominant means of BCNU delivery, the interstitial convection has minor effect because of the extremely small transvascular Peclet number. The reaction of BCNU with brain tissues reduces the drug concentration in all regions and its effect increases with rate constant. The implantation of BCNU/ethylene-vinyl acetate copolymer (EVAc) matrix at the lumen of the viable zone immediately following the surgical removal of 80% of the tumor may be an effective treatment for the chemotherapy of brain tumors. The present study provides a quantitative examination on the working principle of Gliadel wafer for the treatment of brain tumors.
Collapse
Affiliation(s)
- C C Wang
- Department of Chemical Engineering, National University of Singapore, 10 Kent Ridge Crescent, Singapore, Singapore.
| | | | | | | |
Collapse
|
874
|
Muggli DS, Burkoth AK, Anseth KS. Crosslinked polyanhydrides for use in orthopedic applications: degradation behavior and mechanics. JOURNAL OF BIOMEDICAL MATERIALS RESEARCH 1999; 46:271-8. [PMID: 10380006 DOI: 10.1002/(sici)1097-4636(199908)46:2<271::aid-jbm17>3.0.co;2-x] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
High-strength, surface-eroding polymers were synthesized from methacrylated anhydride monomers of sebacic acid (MSA) and 1,6-bis(carboxyphenoxy) hexane (MCPH). These multifunctional monomers were photopolymerized using ultraviolet light to produce highly crosslinked polyanhydride networks. Through this approach, the crosslinking density of the resulting polymer network was used to control the final mechanical properties, while the degradation time scale was controlled by the chemical composition of the network. The combined hydrophobicity of the polymer backbone with the hydrolytically labile anhydride linkages led to surface-eroding networks, as confirmed by linear cumulative mass loss profiles as a function of degradation time for crosslinked polymer disks. By copolymerizing varying amounts of MSA and MCPH, the degradation rate of the final network was controlled from 2 days to 1 year. The tensile modulus of crosslinked poly(MSA) (1.4 GPa) was nearly an order of magnitude larger than that of linear poly(sebacic acid). In general, the mechanical properties of the crosslinked polyanhydrides networks were within ranges of those reported for cortical and trabecular bone. However, unlike bulk degrading polyesters such as poly(lactic acid), these surface eroding networks maintained >70% of their tensile modulus with 50% mass degradation.
Collapse
Affiliation(s)
- D S Muggli
- Department of Chemical Engineering, University of Colorado, Boulder 80309-0424, USA
| | | | | |
Collapse
|
875
|
Brem H, Lawson HC. The development of new brain tumor therapy utilizing the local and sustained delivery of chemotherapeutic agents from biodegradable polymers. Cancer 1999; 86:197-9. [PMID: 10421254 DOI: 10.1002/(sici)1097-0142(19990715)86:2<197::aid-cncr2>3.0.co;2-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
876
|
Menei P, Venier MC, Gamelin E, Saint-Andr� JP, Hayek G, Jadaud E, Fournier D, Mercier P, Guy G, Benoit JP. Local and sustained delivery of 5-fluorouracil from biodegradable microspheres for the radiosensitization of glioblastoma. Cancer 1999. [DOI: 10.1002/(sici)1097-0142(19990715)86:2<325::aid-cncr17>3.0.co;2-s] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
877
|
Newton HB, Turowski RC, Stroup TJ, McCoy LK. Clinical presentation, diagnosis, and pharmacotherapy of patients with primary brain tumors. Ann Pharmacother 1999; 33:816-32. [PMID: 10466912 DOI: 10.1345/aph.18353] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To briefly review the clinical presentation and diagnosis of patients with primary brain tumors, followed by an in-depth survey of the pertinent pharmacotherapy. DATA SOURCES A detailed search of the neurologic, neurosurgical, and oncologic literature for basic science research, clinical studies, and review articles related to chemotherapy and pharmacotherapy of primary brain tumors. STUDY SELECTION Relevant studies on tissue culture systems, animals, and humans examining the mechanisms of action, pharmacokinetics, clinical pharmacology, and treatment results of chemotherapeutic agents for primary brain tumors. In addition, studies of pharmacologic agents administered for supportive care and symptom control are reviewed. DATA SYNTHESIS Primary brain tumors derive from cells within the intracranial cavity and generally present with headache, seizure activity, cognitive changes, and weakness. They are diagnosed most efficiently with magnetic resonance imaging. After diagnosis, the most common supportive medications include corticosteroids, gastric acid inhibitors, and anticonvulsants. Chemotherapy is adjunctive treatment for patients with malignant tumors and selected recurrent or progressive benign neoplasms. In general, the most effective chemotherapeutic drugs are alkylating agents such as the nitrosoureas, procarbazine, cisplatin, and carboplatin. Other agents used include cyclophosphamide, methotrexate, vincristine, and etoposide. Angiogenesis inhibitors and gene therapy comprise some of the novel therapeutic strategies under investigation. CONCLUSIONS The efficacy of chemotherapy for primary brain tumors remains modest. Novel agents must be discovered that are more specific and attack tumor cells at the molecular level of tumorigenesis. Furthermore, strategies must be developed to counteract the pervasive problem of brain tumor chemoresistance.
Collapse
Affiliation(s)
- H B Newton
- Department of Neurology, The Ohio State University, Columbus 43210, USA.
| | | | | | | |
Collapse
|
878
|
Subach BR, Witham TF, Kondziolka D, Lunsford LD, Bozik M, Schiff D. Morbidity and Survival after 1,3-bis(2-chloroethyl)-1-Nitrosourea Wafer Implantation for Recurrent Glioblastoma: A Retrospective Case-matched Cohort Series. Neurosurgery 1999. [DOI: 10.1227/00006123-199907000-00004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
879
|
Subach BR, Witham TF, Kondziolka D, Lunsford LD, Bozik M, Schiff D. Morbidity and survival after 1,3-bis(2-chloroethyl)-1-nitrosourea wafer implantation for recurrent glioblastoma: a retrospective case-matched cohort series. Neurosurgery 1999; 45:17-22; discussion 22-3. [PMID: 10414561 DOI: 10.1097/00006123-199907000-00004] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
OBJECTIVE To determine the risks and survival benefit associated with implantation of an absorbable, 1,3-bis(2chloroethyl)-1-nitrosourea-impregnated polymer wafer, we prospectively studied patients with recurrent glioblastoma multiforme and compared them with a demographically matched cohort group. METHODS Over a 29-month period, 62 patients underwent operations. All had tumor growth despite standard treatment, a Karnofsky performance score of > or =70, and histopathological confirmation of glioblastoma. Seventeen patients underwent gross total resection with placement of 1,3-bis(2-chloroethyl)-1-nitrosourea wafers (wafer group) at a median 44 weeks from diagnosis (6 women, 11 men; median age, 56 years). A cohort group of 45 patients undergoing surgery for recurrent glioblastoma during the same time period, but not receiving wafers, was identified. Surgery was performed at a median 47 weeks from diagnosis (14 women, 31 men; median age, 54 years). RESULTS Within 6 weeks of surgery, 13 complications were identified in 8 patients in the wafer group. In the cohort group, 6 patients sustained 8 complications. We were unable to identify any survival advantage using Kaplan-Meier analysis. In the wafer group, median survival was 58 weeks from diagnosis and 14 weeks from wafer implantation. In the cohort group, median survival was 97 weeks from diagnosis and 50 weeks from operation. CONCLUSION 1,3-bis(2-chloroethyl)-1-Nitrosourea wafer implantation for recurrent glioblastoma was associated with a higher risk of postoperative complications, particularly those related to infection and wound healing. No clear survival benefit associated with wafer implantation was identified.
Collapse
Affiliation(s)
- B R Subach
- Department of Neurological Surgery, University of Pittsburgh Medical Center, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
880
|
|
881
|
Lichtenbeld HC, Ferarra N, Jain RK, Munn LL. Effect of local anti-VEGF antibody treatment on tumor microvessel permeability. Microvasc Res 1999; 57:357-62. [PMID: 10329263 DOI: 10.1006/mvre.1998.2140] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent studies have shown that systemic injection of anti-VEGF antibody into tumor-bearing mice results in decreases in tumor vascular permeability, vessel diameters, and tumor regression. Using a similar animal model, we have applied anti-VEGF antibody directly to the tumor tissue growing in transparent window chambers in SCID mice. Similar to the results obtained with systemic injection, vascular permeability was greatly reduced, but the response was reached at much lower concentrations with local application. Implications of these findings on local control of tumors are discussed.
Collapse
Affiliation(s)
- H C Lichtenbeld
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | |
Collapse
|
882
|
Domb AJ, Israel ZH, Elmalak O, Teomim D, Bentolila A. Preparation and characterization of carmustine loaded polyanhydride wafers for treating brain tumors. Pharm Res 1999; 16:762-5. [PMID: 10350022 DOI: 10.1023/a:1011995728760] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- A J Domb
- Department of Neurosurgery, Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
883
|
Mahoney MJ, Saltzman WM. Millimeter-scale positioning of a nerve-growth-factor source and biological activity in the brain. Proc Natl Acad Sci U S A 1999; 96:4536-9. [PMID: 10200297 PMCID: PMC16367 DOI: 10.1073/pnas.96.8.4536] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Toxicity prevents the systemic administration of many therapeutic proteins, and attempts at protein targeting via the circulatory system (i.e., "magic bullets") have failed in all but a few special cases. Direct administration at the target site is a logical alternative, particularly in the central nervous system, but the limits of direct administration have not been defined clearly. Nerve growth factor (NGF) enhances survival of cholinergic neurons and, therefore, has generated considerable interest for the treatment of Alzheimer's disease. We tested the effectiveness of local delivery by implanting small polymer pellets that slowly released NGF into the central nervous system of adult rats at controlled distances from a target site containing transplanted fetal cholinergic cells. NGF-releasing implants placed within 1-2 mm of the treatment site enhanced the biological function of cellular targets, whereas identical implants placed approximately 3 mm from the target site of treatment produced no beneficial effect. Effective NGF therapy required millimeter-scale positioning of the NGF source, and efficacy correlated with the spatial distribution of NGF concentration in the tissue; this result suggests that NGF must be delivered within several millimeters of the target to be effective in treating Alzheimer's disease. Because the human brain is divided into functional regions that are typically several centimeters in diameter and often irregular in shape, new methods for sculpting larger-scale drug fields are needed. We illustrate a concept, called pharmacotectonics, in which drug-delivery systems are arranged spatially in tissues to shape concentration fields for potent agents.
Collapse
Affiliation(s)
- M J Mahoney
- School of Chemical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
884
|
Bartus RT, Chen EY, Lynch G, Kordower JH. Cortical ablation induces a spreading calcium-dependent, secondary pathogenesis which can be reduced by inhibiting calpain. Exp Neurol 1999; 155:315-26. [PMID: 10072307 DOI: 10.1006/exnr.1998.7001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Many forms of acute brain injury are associated with a secondary, glutamate- and calcium-dependent cascade which greatly exacerbates the final damage. The calcium-dependent protease, calpain, has been implicated as an important variable in this pathogenic process. The present studies tested (i) if similar secondary degeneration occurs following surgical ablation of a discrete area within rat visual cortex, (ii) if activation of calpain contributes to the secondary degeneration by spreading into areas adjacent to the ablation, and (iii) if blocking calpain's proteolytic effects reduces the secondary degeneration attendant to the lesion. Antibodies selective for a protein fragment specifically generated by calpain were used to map areas in which the protease had been activated. Labeling was present 5 min after surgery in a narrow zone surrounding the ablated region. The volume of the immunopositive staining increased twofold within 24 h and fivefold by 48 h, at which time it was equivalent in size to the original lesion. This staining pattern significantly decreased in size at 5 days postsurgery. Application of calpain inhibitors to the ablation site immediately after surgery reduced the spread of calpain activation by approximately 80%. Following cortical ablation, the volume of the lateral geniculate nucleus ipsilateral to the cortical ablation shrank by 46 +/- 3% in control rats but only by 31 +/- 5% in animals given the calpain inhibitors. These results establish that (i) a secondary degenerative cascade is unleashed following discrete cortical surgery which expands into brain areas clearly outside the initial perturbation site, (ii) the gradual expansion of calpain activation contributes to the underlying secondary pathology, and (iii) blocking calpain activity substantially reduces atrophy in areas anatomically connected, but physically distal to the damaged zone. The possible utility of topical applications of calpain inhibitors, or analogously acting drugs, in minimizing the secondary effects of brain surgery is discussed.
Collapse
Affiliation(s)
- R T Bartus
- Alkermes Inc., Cambridge, Massachusetts, 02139, USA
| | | | | | | |
Collapse
|
885
|
Hudes RS, Corn BW, Werner-Wasik M, Andrews D, Rosenstock J, Thoron L, Downes B, Curran WJ. A phase I dose escalation study of hypofractionated stereotactic radiotherapy as salvage therapy for persistent or recurrent malignant glioma. Int J Radiat Oncol Biol Phys 1999; 43:293-8. [PMID: 10030252 DOI: 10.1016/s0360-3016(98)00416-7] [Citation(s) in RCA: 120] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
PURPOSE A phase I dose escalation of hypofractionated stereotactic radiotherapy (H-SRT) in recurrent or persistent malignant gliomas as a means of increasing the biologically effective dose and decreasing the high rate of reoperation due to toxicity associated with single-fraction stereotactic radiosurgery (SRS) and brachytherapy. MATERIALS AND METHODS From November 1994 to September 1996, 25 lesions in 20 patients with clinical and/or imaging evidence of malignant glioma persistence or recurrence received salvage H-SRT. Nineteen patients at the time of initial diagnosis had glioblastoma multiforme (GBM) and one patient had an anaplastic astrocytoma. All of these patients with tumor persistence or recurrence had received initial fractionated radiation therapy (RT) with a mean and median dose of 60 Gy (44.0-72.0 Gy). The median time from completion of initial RT to H-SRT was 3.1 months (0.7-45.5 months). Salvage H-SRT was delivered using daily 3.0-3.5 Gy fractions (fxs). Three different total dose levels were sequentially evaluated: 24.0 Gy/3.0 Gy fxs (five lesions), 30.0 Gy/3.0 Gy fxs (10 lesions), and 35.0 Gy/3.5 Gy fxs (nine lesions). Median treated tumor volume measured 12.66 cc (0.89-47.5 cc). The median ratio of prescription volume to tumor volume was 2.8 (1.4-5.0). Toxicity was judged by RTOG criteria. Response was determined by clinical neurologic improvement, a decrease in steroid dose without clinical deterioration, and/or radiologic imaging. RESULTS No grade 3 toxicities were observed and no reoperation due to toxicity was required. At the time of analysis, 13 of 20 patients had died. The median survival time from the completion of H-SRT is 10.5 months with a 1-year survival rate of 20%. Neurological improvement was found in 45% of patients. Decreased steroid requirements occurred in 60% of patients. Minor imaging response was noted in 22% of patients. Using Fisher's exact test, response of any kind correlated strongly to total dose (p = 0.0056). None of six lesions treated with 21 Gy or 24 Gy responded, whereas there was a 79% response rate among the 19 lesions treated with 30 or 35 Gy. Tumor volumes < or =20 cc were associated with a higher likelihood of response (p = 0.053). CONCLUSIONS H-SRT used in this cohort of previously irradiated patients with malignant glioma was not associated with the need for reoperation due to toxicity or grade 3 toxicity. This low toxicity profile and encouraging H-SRT dose-related response outcome justifies further evaluation and dose escalation.
Collapse
Affiliation(s)
- R S Hudes
- Department of Radiation Oncology, Wills Eye Hospital, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | |
Collapse
|
886
|
Emerich DF, Tracy MA, Ward KL, Figueiredo M, Qian R, Henschel C, Bartus RT. Biocompatibility of poly (DL-lactide-co-glycolide) microspheres implanted into the brain. Cell Transplant 1999; 8:47-58. [PMID: 10338275 DOI: 10.1177/096368979900800114] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The delivery of therapeutic molecules to the brain has been limited in part due to the presence of the blood-brain barrier. One potential solution is the implantation of biodegradable polymers with sustained release of drugs. Poly (DL-lactide-co-glycolide) (PLG) is a bioerodible polymer with a long and successful history of use as a suture material. More recently, PLG has been investigated for localized and sustained delivery of molecules into both peripheral sites and the brain. Despite its well-defined safety profile for parenteral applications, little information exists concerning the safety of PLG when implanted into the brain. To further characterize the biocompatibility of PLG in the brain, we examined the gliotic response following implants of PLG into the brains of rats. As a control, each animal received an injection of the suspension medium into the contralateral hemisphere. Following implantation, PLG was well tolerated. GFAP-positive astrocytes were observed throughout the cerebral cortex and striatum on both the implanted and control sides, with the reaction being greatest within the heavily myelinated fiber tracts of the corpus callosum. Quantitative analyses revealed that this reaction occurred within 1 h postsurgery, reached its peak at 1 week following surgery, and then decreased markedly by 1 month postsurgery. A minimal gliotic reaction was still present 1 year postsurgery but was localized to the needle tract. No differences in GFAP reactivity were seen between the polymer-implanted and control sides at any time point. Histological analysis determined that the majority of the PLG disappeared between 1 and 4 weeks. A set of parallel studies in which PLG samples were retrieved from the brain at various time points corroborated these findings and determined that the majority of PLG degraded within 2 weeks following implantation. Together, these results demonstrate that PLG is well tolerated following implantation into the CNS and that the astrocytic response to PLG is largely a consequence of the mechanical trauma that occurs during surgery. The biocompatibility of PLG implanted into the CNS provides further support for its use in a wide range of new therapeutic applications for sustained and localized drug delivery to the brain.
Collapse
Affiliation(s)
- D F Emerich
- Department of Pharmacology, Alkermes, Inc., Cambridge, MA 02139, USA
| | | | | | | | | | | | | |
Collapse
|
887
|
Larner J, Jane J, Laws E, Packer R, Myers C, Shaffrey M. A phase I-II trial of lovastatin for anaplastic astrocytoma and glioblastoma multiforme. Am J Clin Oncol 1998; 21:579-83. [PMID: 9856659 DOI: 10.1097/00000421-199812000-00010] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Malignant gliomas are thought to be highly dependent on the mevalonate pathway for cell growth. Lovastatin, a cholesterol-lowering drug, inhibits not only the rate-limiting step in the mevalonate pathway (hepatic hydroxymethyl glutaryl coenzyme A reductase), but also the prenylation of several key regulatory proteins including ras and the small guanosine triphosphate binding proteins. Therefore, from August 1994 through March 1996, 18 patients with either anaplastic glioma or glioblastoma multiforme were entered into a trial testing the safety of high-dose lovastatin with or without radiation. Although the response data is too premature to evaluate activity, the fact that high doses of lovastatin are well tolerated with concurrent radiation suggests that central nervous system toxicity will not be a significant limiting toxicity as more selective farnesyltransferase inhibitors are brought into the clinic as radiation sensitizers.
Collapse
Affiliation(s)
- J Larner
- Department of Therapeutic Radiology, University of Virginia Health Sciences Center, Charlottesville 22908, USA
| | | | | | | | | | | |
Collapse
|
888
|
Klatzmann D, Valéry CA, Bensimon G, Marro B, Boyer O, Mokhtari K, Diquet B, Salzmann JL, Philippon J. A phase I/II study of herpes simplex virus type 1 thymidine kinase "suicide" gene therapy for recurrent glioblastoma. Study Group on Gene Therapy for Glioblastoma. Hum Gene Ther 1998; 9:2595-604. [PMID: 9853526 DOI: 10.1089/hum.1998.9.17-2595] [Citation(s) in RCA: 130] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Despite extensive surgery for glioblastoma, residual tumor cells always lead to relapse. Gene therapy based on retrovirus-mediated gene transfer of herpes simplex virus type 1 thymidine kinase (HSV-1 TK), which specifically sensitizes dividing cells to ganciclovir (GCV) toxicity, may help eradicate such cells. During glioblastoma surgery, HSV-1 TK retroviral vector-producing cells (M11) were injected into the surgical cavity margins after tumor debulking. After a 7-day transduction period, GCV was administered for 14 days. Safety was assessed by clinical and laboratory evaluations, and efficacy was assessed by MRI-based relapse-free survival at month 4 and by overall survival. Twelve patients with recurrent glioblastoma were treated without serious adverse events related to M11 cell administration or GCV. Quality of life was not negatively influenced by this treatment. Overall median survival was 206 days, with 25% of the patients surviving longer than 12 months. At 4 months after treatment, 4 of 12 patients had no recurrence; their median overall survival was 528 days, compared with 194 days for patients with recurrence (p=0.03 by the log rank test). One patient is still free of detectable recurrence, steroid free and independent, 2.8 years after treatment. Thus, brain injections of M11 retroviral vector-producing cells for glioblastoma HSV-1 TK gene therapy were well tolerated and associated with significant therapeutic responses. These results warrant further development of this therapeutic strategy in brain tumor, including recurrent glioblastoma.
Collapse
Affiliation(s)
- D Klatzmann
- Department of Immunology, Hôpital Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
889
|
Kubek MJ, Liang D, Byrd KE, Domb AJ. Prolonged seizure suppression by a single implantable polymeric-TRH microdisk preparation. Brain Res 1998; 809:189-97. [PMID: 9853110 DOI: 10.1016/s0006-8993(98)00860-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Thyrotropin-releasing hormone (TRH; Protirelin) is an endogenous neuropeptide known to have anticonvulsant effects in several seizure models and in intractable epileptic patients. Like most neuropeptides, its duration of action may be limited by a lack of sustained site-specific bioavailability. To attempt to provide long-term delivery, we attached TRH to a biodegradable polyanhydride copolymer as a sustained-release carrier. Utilizing the rat kindling model of temporal lobe epilepsy, a single TRH microdisk implanted stereotaxically into the seizure focus (amygdala) significantly suppressed kindling expression when assessed by the number of stimulations required to reach each behavioral stage and to become fully kindled (8.63 +/- 0.92 vs. 16.17 +/- 1.37; Mean +/- S.E.M.). Two indices of seizure severity, afterdischarge duration (Mean +/- S.E.M., sec.) (stimulated amygdala [87.40 +/- 5.47 vs. 51.80 +/- 15.65] and unstimulated amygdala [89.60 +/- 5.55 vs. 48.67 +/- 15.8] and clonus duration (71.2 +/- 5.94 vs. 29.40 +/- 8.87; Mean +/- S.E.M., sec.), were also significantly reduced by a single polymeric-TRH implant. Fifty days after initiation of the study a significant reduction in clonus duration (53.90 +/- 3.27 vs. 40.09 +/- 4.14) still remained in the TRH-implanted groups. This report is the first to provide evidence in support of in situ microdisk pharmacotherapy for potential neuropeptide delivery in intractable epilepsy and possibly other neurological disorders.
Collapse
Affiliation(s)
- M J Kubek
- Department of Anatomy, Indiana University School of Medicine, Indianapolis 46202-5120, USA.
| | | | | | | |
Collapse
|
890
|
Reza MS, Whateley TL. Iodo-2'-deoxyuridine (IUdR) and 125IUdR loaded biodegradable microspheres for controlled delivery to the brain. J Microencapsul 1998; 15:789-801. [PMID: 9818956 DOI: 10.3109/02652049809008261] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this work was to develop sustained local release systems for radioiodinated iodo-2'-deoxyuridine (125IUdR) from biodegradable polymeric microspheres to facilitate the controlled delivery of 125IUdR to brain tumours. The selective uptake of IUdR into the cell nucleus results in cell disruption over the short range of the low energy Auger electrons. The biodegradable microspheres can be precisely implanted in the brain by stereotactic techniques and the IUdR within the microspheres is protected from degradation and thus a sustained source of radiolabelled IUdR is available in the vicinity of the residual tumour cells. Poly(lactic-co-glycolic acid), PLGA (85:15), microspheres containing cold IUdR and the Auger-electron emitter 125I, as 125IUdR were prepared using the O/W, O/O and W/O/W emulsion-solvent evaporation methods. The W/O/W emulsion method was most effective in achieving good drug loading with the use of bovine plasma in the internal water phase. Also effective in improving the drug loading was the use of 20% acetone in the dichloromethane and the presence of Span 40 in the organic phase. Electrolytes (NaCl and IUdR) in the external aqueous phase also improved drug loading. After an initial rapid release from the microspheres, a sustained release was observed over 15 days for the 'cold' IUdR. The sustained release portions of the release curves showed Higuchi (t1/2), diffusion controlled release kinetics. The radiolabelled IUdR microspheres showed a burst release effect of 30-40% followed by a sustained release over 35 days.
Collapse
Affiliation(s)
- M S Reza
- Department of Pharmaceutical Sciences, University of Strathclyde, Glasgow, UK
| | | |
Collapse
|
891
|
Ewend MG, Sampath P, Williams JA, Tyler BM, Brem H. Local delivery of chemotherapy prolongs survival in experimental brain metastases from breast carcinoma. Neurosurgery 1998; 43:1185-93. [PMID: 9802862 DOI: 10.1097/00006123-199811000-00093] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Despite improved systemic control of metastatic breast cancer, the incidence of brain metastases from breast carcinoma continues to rise, in part because most systemically administered agents have poor central nervous system penetration. Therefore, as a method of optimizing drug delivery into the central nervous system, we studied the safety and efficacy of chemotherapy delivered locally via biodegradable polymers in a mouse model of breast carcinoma metastases to the brain. METHODS The chemotherapeutic agents carmustine (BCNU), carboplatin, and camptothecin were incorporated into controlled release polymers and tested individually against intracranial challenges of EMT-6 breast tumor in BALB/c female mice. For each drug, four groups were tested: Group 1, empty polymer (no drug); Group 2, external beam radiotherapy (XRT) alone; Group 3, local chemotherapy from biodegradable polymer alone; and Group 4, local chemotherapy and XRT together. Polymers were implanted 5 days after intracranial tumor inoculation; XRT was administered on Days 7 through 9 (300 cGy/d). RESULTS BCNU polymer alone (n = 10; median survival time, >200 d; P < 0.0001) and BCNU and XRT together (n = 10; median survival time, 41 d; P = 0.02) significantly improved survival in mice with intracranial EMT-6 breast cancer in comparison with control animals (n = 20; median survival time, 17 d). Carboplatin and camptothecin, either with or without XRT, and XRT alone did not have any significant effect on survival. CONCLUSION Local delivery of BCNU with biodegradable polymers can significantly prolong survival in a murine model of intracranial metastatic breast cancer. Surgical resection and placement of BCNU polymers into the resection cavity may decrease the incidence of local recurrence of breast cancer metastases with minimal morbidity.
Collapse
Affiliation(s)
- M G Ewend
- Division of Neurological Surgery, University of North Carolina, Chapel Hill, USA
| | | | | | | | | |
Collapse
|
892
|
COMMENTS. Neurosurgery 1998. [DOI: 10.1097/00006123-199811000-00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
893
|
|
894
|
Kong Q, Kleinschmidt-DeMasters BK, Lillehei KO. Intralesionally implanted cisplatin plus systemic carmustine for the treatment of brain tumor in rats. J Surg Oncol 1998; 69:76-82. [PMID: 9808509 DOI: 10.1002/(sici)1096-9098(199810)69:2<76::aid-jso6>3.0.co;2-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND OBJECTIVES The benefit of conventional chemotherapy for the treatment of malignant brain tumors, although limited, is real. A major obstacle in the treatment of these lesions is the ability to deliver drug across the blood-brain barrier (BBB). Local drug implantation, circumventing the BBB, has been a useful strategy for treatment of intracranial lesions, and may work synergistically with systemic chemotherapy. To test this hypothesis, either intraperitoneal (i.p.) carmustine or cisplatin was combined with the intracranial (i.c.) administration of polymer-delivered cisplatin in rats with intracranial tumors. METHODS AND RESULTS 9L gliosarcoma tumor cells (5 x 10(3)) were administered through a right frontal lobe cannula in rats 7 days prior to treatments. Cisplatin-loaded biodegradable polymer was then administered via the cannula, with free cisplatin or carmustine injected i.p. Animals were monitored for 60 days post-treatment. In experiment 1, i.c. cisplatin at a dose of 0.5, 1.0, 2.0, and 4.0 mg/m2 resulted in a mean survival time of 34 +/- 3, 39 +/- 14, 47 +/- 11, and 31 +/- 20 days (MST +/- SD), respectively, compared to 26 +/- 4 days in the control group and 30 +/- 7 days in the group treated with 50 mg/m2 i.p. free cisplatin. In experiment 2, i.p. free cisplatin at 25, 40, 50, and 100 mg/m2 resulted in a MST of 28 +/- 3, 30 +/- 4, 32 +/- 3, and 14 +/- 8 days, respectively, compared to 26 +/- 1 days in the control group. In experiment 3, the MST in the groups treated with 0.5 mg/m2 of i.c. cisplatin, 25 mg/m2 of i.p. cisplatin, 10 mg/kg of i.p. carmustine, i.c. cisplatin (0.5 mg/m2) plus i.p. cisplatin (25 mg/m2), and i.c. cisplatin (0.5 mg/m2) plus i.p. carmustine (10 mg/kg) was 30 +/- 4 days (P > 0.05), 28 +/- 2 (P > 0.05), 36 +/- 4 (P < 0.01), 32 +/- 3 (P < 0.01), and 50 +/- 11 days (P < 0.01), respectively, compared to the tumor control group (26 +/- 1 days). Long-term survivors (29%) were seen only in the i.c. cisplatin plus i.p. carmustine group. Additive toxicity was not observed. CONCLUSIONS Intralesional polymer-delivered (i.c.) cisplatin plus systemic (i.p.) carmustine is highly effective for the treatment of intracranial 9L gliosarcoma in tumors.
Collapse
Affiliation(s)
- Q Kong
- Department of Surgery, University of Colorado Health Sciences Center, Denver 80262, USA.
| | | | | |
Collapse
|
895
|
Williams JA, Yuan X, Dillehay LE, Shastri VR, Brem H, Williams JR. Synthetic, implantable polymers for local delivery of IUdR to experimental human malignant glioma. Int J Radiat Oncol Biol Phys 1998; 42:631-9. [PMID: 9806525 DOI: 10.1016/s0360-3016(98)00258-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Recently, polymeric controlled delivery of chemotherapy has been shown to improve survival of patients with malignant glioma. We evaluated whether we could similarly deliver halogenated pyrimidines to experimental intracranial human malignant glioma. To address this issue we studied the in vitro release from polymers and the in vivo drug delivery of IUdR to experimental human U251 glioblastoma xenografts. METHODS AND MATERIALS In vitro: To measure release, increasing (10%, 30%, 50%) proportions of IUdR in synthetic [(poly(bis(p-carboxyphenoxy)-propane) (PCPP):sebacic acid (SA) polymer discs were serially incubated in buffered saline and the supernatant fractions were assayed. In vivo: To compare local versus systemic delivery, mice bearing flank xenografts had intratumoral or contralateral flank IUdR polymer (50% loading) treatments. Mice bearing intracranial (i.c.) xenografts had i.c. versus flank IUdR polymer treatments. Four or 8 days after implantation of polymers, mice were sacrificed and the percentage tumor cells that were labeled with IUdR was measured using quantitative microscopic immunohistochemistry. RESULTS In vitro: Increasing percentage loadings of IUdR resulted in higher percentages of release: 43.7 + 0.1, 70.0 + 0.2, and 90.2 + 0.2 (p < 0.001 ANOVA) for the 10%, 30%, and 50% loadings, respectively. In vivo: For the flank tumors, both the ipsilateral and contralateral IUdR polymers resulted in similarly high percentages labeling of the tumors versus time. For the ipsilateral IUdR polymers, the percentage of tumor cellular labeling after 4 days versus 8 days was 45.8 +/- 7.0 versus 40.6 +/- 3.9 (p = NS). For the contralateral polymer implants, the percentage of tumor cellular labeling were 43.9 +/- 10.1 versus 35.9 +/- 5.2 (p = NS) measured 4 days versus 8 days after implantation. For the i.c. tumors treated with extracranial IUdR polymers, the percentage of tumor cellular labeling was low: 13.9 +/- 8.8 and 11.2 +/- 5.7 measured 4 and 8 days after implantation. For the i.c. tumors having the i.c. IUdR polymers, however, the percentage labeling was comparatively much higher: 34.3 +/- 4.9 and 35.3 +/- 4.0 on days 4 and 8, respectively. For the i.c. tumors, examination of the percentage cellular labeling versus distance from the implanted IUdR polymer showed that labeling was highest closest to the polymer disc. CONCLUSION Synthetic, implantable biodegradable polymers provide the local, controlled release of IUdR and result in the high, local delivery of IUdR to experimental intracranial human malignant glioma. This technique holds promise for the local delivery of IUdR for radiosensitization of human brain tumors.
Collapse
Affiliation(s)
- J A Williams
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | | | | | | | | |
Collapse
|
896
|
Pollina J, Plunkett RJ, Ciesielski MJ, Lis A, Barone TA, Greenberg SJ, Fenstermaker RA. Intratumoral infusion of topotecan prolongs survival in the nude rat intracranial U87 human glioma model. J Neurooncol 1998; 39:217-25. [PMID: 9821107 DOI: 10.1023/a:1005954121521] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Topotecan is a topoisomerase (topo) I inhibitor with promising activity in preclinical studies. We hypothesized that low-dose intratumoral delivery of topotecan would be highly effective for gliomas. Human glioma cell lines (U87, U138 and U373) displayed different sensitivities to topotecan (IC50 range: 0.037 microM to 0.280 microM) in cell culture. The most resistant of the glioma cell lines (U87) was implanted stereotactically into the brains of nude rats. Twelve days later, at which time tumor diameter measured 2 to 2.5 mm, animals were randomized to three groups: group I, intratumoral topotecan infused via osmotic pump (n = 12); group II, intratumoral saline infusion (n = 7); and group III, no treatment (n = 10). Animals were sacrificed when signs of deterioration developed, or at 60 days. Animals in group I had a mean survival time (MST) of > 55 days (range = 40-60); whereas, those in groups II and III had MST of 26.1 (range = 21-31) and 26.5 (range = 20-30) days, respectively. The differences in survival between group I and each of the other groups were statistically significant (p < 0.0001; Logrank Mantel-Cox). None of the animals that survived 60 days had histological evidence of residual tumor at sacrifice. Measurement of topotecan levels in normal brain revealed cytotoxic concentrations up to 4.5 mm from the site of infusion. This study demonstrates that intratumoral topotecan delivered via an osmotic pump prolongs survival in the U87 human glioma model.
Collapse
Affiliation(s)
- J Pollina
- Department of Neurosurgery, S.U.N.Y., Buffalo School of Medicine and Biomedical Sciences, New York, USA
| | | | | | | | | | | | | |
Collapse
|
897
|
Affiliation(s)
- E H Oldfield
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA
| | | |
Collapse
|
898
|
Wheldon TE, Mairs RJ, Rampling RP, Barrett A. Modelling the enhancement of fractionated radiotherapy by gene transfer to sensitize tumour cells to radiation. Radiother Oncol 1998; 48:5-13. [PMID: 9756166 DOI: 10.1016/s0167-8140(98)00022-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND PURPOSE Several strategies now exist for the use of gene transfer methodologies to sensitize tumour cells to radiation. These include the transfection of genes synthesizing cytokines, p53 gene replacement and methods based on the use of HSV-tk and gancyclovir. Very recently, the sequencing of radioprotector or repair genes, such as ATM, Ku80 and XRCC2, has made it possible to consider the design of gene transfer strategies resulting in protector gene knock-out. Selectivity of transfected gene expression might be achieved by use of tissue-specific promoters or by the trophism of viral vectors. The purpose of this study was to evaluate the probable efficacy of such strategies. METHODS We have modelled gene transfer-mediated radiosensitization of tumour cells during radiotherapy, focusing on anti-protector gene strategies, to explore the role of transfection frequency, sensitizing efficacy, transfection stability, untransfectable subpopulations, the timing of gene therapy and the treatment schedule structure. RESULTS We predict a substantial therapeutic benefit of gene transfer treatment (with at least weekly transfection) which modifies cellular radiosensitivity by a factor of 1.5 or more, despite modest efficiency of cellular transfection (e.g. 50%), transient retention of the transfected gene (e.g. 2-day half-life) and the existence of a small minority (e.g. 1%) of untransfectable cells. CONCLUSIONS The analysis shows repeated administration of gene transfer treatment to be obligatory and implies that the existence of untransfectable minority subpopulations (i.e. cells inaccessible to the vector) will be the major limiting factor in therapy. Experimental work is needed to confirm these predictions before clinical studies begin.
Collapse
Affiliation(s)
- T E Wheldon
- Department of Radiation Oncology, University of Glasgow, CRC Beatson Laboratories, UK
| | | | | | | |
Collapse
|
899
|
Plautz GE, Barnett GH, Miller DW, Cohen BH, Prayson RA, Krauss JC, Luciano M, Kangisser DB, Shu S. Systemic T cell adoptive immunotherapy of malignant gliomas. J Neurosurg 1998; 89:42-51. [PMID: 9647171 DOI: 10.3171/jns.1998.89.1.0042] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT To determine the feasibility, toxicity, and potential therapeutic benefits of systemic adoptive immunotherapy, 10 patients with progressive primary or recurrent malignant glioma received this treatment. Adoptive immunotherapy, the transfer of immune T lymphocytes, is capable of mediating the regression of experimental brain tumors in animal models. In animal models, lymph nodes (LNs) that drain the tumor vaccine site are a rich source of tumor-immune T cells. METHODS In this clinical study, patients were inoculated intradermally with irradiated autologous tumor cells and granulocyte macrophage-colony stimulating factor as an adjuvant. Cells from draining inguinal LNs, surgically resected 7 days after vaccination, were stimulated sequentially with staphylococcal enterotoxin A and anti-CD3, and a low dose of interleukin-2 (60 IU/ml) was used to expand the stimulated cells. The maximum cell proliferation was 350-fold over 10 days of culture. The activated cells were virtually all T cells consisting of various proportions of CD4 and CD8 cells. These cells were given to patients by intravenous infusion at doses ranging from 9 x 10(8) to 1.5 x 10(11). There were no Grade 3 or 4 toxicities associated with the treatment. Following T-cell transfer therapy, radiographic regression that lasted at least 6 months was demonstrated in two patients with recurrent tumors. One patient demonstrated stable disease that has lasted for more than 17 months. The remaining patients had progressive disease; however, four of the eight patients with recurrent tumor remain alive more than 1 year after surgery for recurrence. Three patients required intervention with corticosteroid agents or additional surgery approximately 1 month following cell transfer. CONCLUSIONS These intriguing clinical observations warrant further trials to determine whether this approach can provide therapeutic benefits and improve survival.
Collapse
Affiliation(s)
- G E Plautz
- Department of Neurological Surgery, Center for Surgery Research, The Cleveland Clinic Foundation, Ohio 44195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
900
|
Wiranowska M, Ransohoff J, Weingart JD, Phelps C, Phuphanich S, Brem H. Interferon-containing controlled-release polymers for localized cerebral immunotherapy. J Interferon Cytokine Res 1998; 18:377-85. [PMID: 9660244 DOI: 10.1089/jir.1998.18.377] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Controlled-release ethylene-vinyl acetate copolymers (EVAc), which were used previously for the in vivo intracerebral delivery of chemotherapeutics, were evaluated as a possible route of localized intracerebral delivery of interferon (IFN). Natural mouse IFN-alpha/beta (Mu-IFN-alpha/beta) was incorporated into polymers at 5% or 10% by weight with 2 x 10(4) U or 4 x 10(4) U, respectively. In vitro and in vivo studies of the release of Mu-IFN-alpha/beta from EVAc polymers showed the released IFN to be biologically active, as determined by the inhibition assay of viral cytopathic effect (CPE). Evaluation of the in vitro kinetics of release showed that most of the IFN activity was released in the first 4 days, with the rest being released thereafter. The in vivo kinetic release of Mu-IFN-alpha/beta from intracerebrally implanted polymers showed that most of the IFN activity was released within 24 h after polymer implantation in the hemisphere ipsilateral to the polymer. This IFN activity gradually decreased over the next 72 h, with a significant linear trend (p < 0.0001). The hemisphere contralateral to the implanted polymer showed no significant levels of IFN activity throughout the 4 days of evaluation. By contrast, blood levels of IFN increased from day 1 to day 4, showing a significant linear trend (p = 0.0125), with IFN levels on day 4 being significantly higher (p < 0.05) than on day 1 after polymer implant. This study demonstrates the feasibility of intracranial controlled local delivery of IFN using a polymer delivery device.
Collapse
Affiliation(s)
- M Wiranowska
- Department of Anatomy, College of Medicine, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa 33612, USA.
| | | | | | | | | | | |
Collapse
|