1
|
Application of Convection-Enhanced Drug Delivery in the Treatment of Malignant Gliomas. World Neurosurg 2016; 90:172-178. [DOI: 10.1016/j.wneu.2016.02.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 02/01/2023]
|
2
|
Luther N, Cheung NKV, Dunkel IJ, Fraser JF, Edgar MA, Gutin PH, Souweidane MM. INTRAPARENCHYMAL AND INTRATUMORAL INTERSTITIAL INFUSION OF ANTI-GLIOMA MONOCLONAL ANTIBODY 8H9. Neurosurgery 2008; 63:1166-74; discussion 1174. [DOI: 10.1227/01.neu.0000334052.60634.84] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Neal Luther
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ira J. Dunkel
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Justin F. Fraser
- Department of Neurological Surgery, Weill Cornell Medical College, New York, New York
| | - Mark A. Edgar
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Philip H. Gutin
- Department of Neurological Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Mark M. Souweidane
- Department of Neurological Surgery, Weill Cornell Medical College, and Department of Neurological Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
3
|
Rousseau J, Boudou C, Barth RF, Balosso J, Estève F, Elleaume H. Enhanced survival and cure of F98 glioma-bearing rats following intracerebral delivery of carboplatin in combination with photon irradiation. Clin Cancer Res 2007; 13:5195-201. [PMID: 17726137 PMCID: PMC3390313 DOI: 10.1158/1078-0432.ccr-07-1002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The goal of the present study was to evaluate the efficacy of intracerebral (i.c.) administration of carboplatin by means of convection-enhanced delivery (CED) in combination with fractionated, external beam photon irradiation for the treatment of F98 glioma-bearing rats. EXPERIMENTAL DESIGN Carboplatin (20 microg/20 microL) was administrated i.c. by CED to F98 glioma-bearing rats, 13 days after stereotactic implantation of 10(3) tumor cells. One day following initiation of CED, a 24-Gy X-ray dose was administered in three daily fractions of 8 Gy each. Photon irradiation was carried out using either a conventional (6 MV) linear accelerator or a monochromatic synchrotron source (80 keV) at the European Synchrotron Radiation Facility. The primary end point of this study was overall survival. RESULTS The median survival times were 79 and 60 days and the corresponding percent increase in life spans were 182% and 114%, respectively, for the combination of carboplatin chemotherapy and irradiation with either 6-MV or 80-keV photons. A subset of long-term survivors (>200 days) were observed in both chemoradiotherapy groups: 16.6% and 8.3% for 6 MV and 80 keV, respectively. In contrast, the median survival times for 6-MV or 80-keV irradiated controls, chemotherapy alone, and untreated controls were 42, 51, 45, and 28 days, respectively. CONCLUSIONS Our results convincingly show the therapeutic efficacy of i.c. administration of carboplatin by means of CED in combination with either 6-MV or 80-keV photons. Further studies are warranted to optimize this combination of chemoradiotherapy for malignant gliomas.
Collapse
Affiliation(s)
- Julia Rousseau
- GIN, Grenoble Institut des Neurosciences
INSERM : U836CEAUniversité Joseph Fourier - Grenoble ICHU GrenobleUJF - Site Santé La Tronche BP 170 38042 Grenoble Cedex 9,FR
- ID17, Biomedical Beamline
ESRFGrenoble,FR
| | - Caroline Boudou
- GIN, Grenoble Institut des Neurosciences
INSERM : U836CEAUniversité Joseph Fourier - Grenoble ICHU GrenobleUJF - Site Santé La Tronche BP 170 38042 Grenoble Cedex 9,FR
| | - Rolf F. Barth
- Department of Pathology
Ohio State UniversityDivision of Biostatistics, Columbus, OH,US
| | - Jacques Balosso
- GIN, Grenoble Institut des Neurosciences
INSERM : U836CEAUniversité Joseph Fourier - Grenoble ICHU GrenobleUJF - Site Santé La Tronche BP 170 38042 Grenoble Cedex 9,FR
- ID17, Biomedical Beamline
ESRFGrenoble,FR
- CHU GrenobleFR
| | - François Estève
- GIN, Grenoble Institut des Neurosciences
INSERM : U836CEAUniversité Joseph Fourier - Grenoble ICHU GrenobleUJF - Site Santé La Tronche BP 170 38042 Grenoble Cedex 9,FR
- ID17, Biomedical Beamline
ESRFGrenoble,FR
- CHU GrenobleFR
| | - Hélène Elleaume
- GIN, Grenoble Institut des Neurosciences
INSERM : U836CEAUniversité Joseph Fourier - Grenoble ICHU GrenobleUJF - Site Santé La Tronche BP 170 38042 Grenoble Cedex 9,FR
- ID17, Biomedical Beamline
ESRFGrenoble,FR
- CHU GrenobleFR
- * Correspondence should be adressed to: Hélène Elleaume
| |
Collapse
|
4
|
Krauze MT, Noble CO, Kawaguchi T, Drummond D, Kirpotin DB, Yamashita Y, Kullberg E, Forsayeth J, Park JW, Bankiewicz KS. Convection-enhanced delivery of nanoliposomal CPT-11 (irinotecan) and PEGylated liposomal doxorubicin (Doxil) in rodent intracranial brain tumor xenografts. Neuro Oncol 2007; 9:393-403. [PMID: 17652269 PMCID: PMC1994096 DOI: 10.1215/15228517-2007-019] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We have previously shown that convection-enhanced delivery (CED) of highly stable nanoparticle/liposome agents encapsulating chemotherapeutic drugs is effective against intracranial rodent brain tumor xenografts. In this study, we have evaluated the combination of a newly developed nanoparticle/liposome containing the topoisomerase I inhibitor CPT-11 (nanoliposomal CPT-11 [nLs-CPT-11]), and PEGylated liposomal doxorubicin (Doxil) containing the topoisomerase II inhibitor doxorubicin. Both drugs were detectable in the CNS for more than 36 days after a single CED application. Tissue half-life was 16.7 days for nLs-CPT-11 and 10.9 days for Doxil. The combination of the two agents produced synergistic cytotoxicity in vitro. In vivo in U251MG and U87MG intracranial rodent xenograft models, CED of the combination was also more efficacious than either agent used singly. Analysis of the parameters involved in this approach indicated that tissue pharmacokinetics, tumor microanatomy, and biochemical interactions of the drugs all contributed to the therapeutic efficacy observed. These findings have implications for further clinical applications of CED-based treatment of brain tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Krystof S. Bankiewicz
- Address correspondence to Krystof S. Bankiewicz, Department of Neurological Surgery, MCB226, University of California, San Francisco, 1855 Folsom St., Mission Center Building, San Francisco, CA 94103-0555, USA (
)
| |
Collapse
|
5
|
Yamashita Y, Krauze MT, Kawaguchi T, Noble CO, Drummond DC, Park JW, Bankiewicz KS. Convection-enhanced delivery of a topoisomerase I inhibitor (nanoliposomal topotecan) and a topoisomerase II inhibitor (pegylated liposomal doxorubicin) in intracranial brain tumor xenografts. Neuro Oncol 2006; 9:20-8. [PMID: 17018695 PMCID: PMC1828110 DOI: 10.1215/15228517-2006-016] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Despite multimodal treatment options, the response and survival rates for patients with malignant gliomas remain dismal. Clinical trials with convection-enhanced delivery (CED) have recently opened a new window in neuro-oncology to the direct delivery of chemotherapeutics to the CNS, circumventing the blood-brain barrier and reducing systemic side effects. Our previous CED studies with liposomal chemotherapeutics have shown promising antitumor activity in rodent brain tumor models. In this study, we evaluated a combination of nanoliposomal topotecan (nLs-TPT) and pegylated liposomal doxorubicin (PLD) to enhance efficacy in our brain tumor models, and to establish a CED treatment capable of improving survival from malignant brain tumors. Both liposomal drugs decreased key enzymes involved in tumor cell replication in vitro. Synergistic effects of nLs-TPT and PLD on U87MG cell death were found. The combination displayed excellent efficacy in a CED-based survival study 10 days after tumor cell implantation. Animals in the control group and those in singleagent groups had a median survival of less than 30 days, whereas the combination group experienced a median survival of more than 90 days. We conclude that CED of two liposomal chemotherapeutics (nLs-TPT and PLD) may be an effective treatment option for malignant gliomas.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Krystof S. Bankiewicz
- Address correspondence to Krystof S. Bankiewicz, Department of Neurological Surgery, University of California, San Francisco, 1855 Folsom Street, Mission Center Building Room 226, San Francisco, CA 94103, USA (
)
| |
Collapse
|
6
|
Kioi M, Husain SR, Croteau D, Kunwar S, Puri RK. Convection-enhanced delivery of interleukin-13 receptor-directed cytotoxin for malignant glioma therapy. Technol Cancer Res Treat 2006; 5:239-50. [PMID: 16700620 DOI: 10.1177/153303460600500307] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The treatment of patients with malignant brain tumors, in particular glioblastoma multiforme (GBM) is very challenging because of their diffuse infiltrative nature and the cytological heterogeneity. The median survival of patients with newly diagnosed GBM is only 12-15 months, and only 8-12% of them survive for two years. Novel approaches for brain tumor therapy are needed. Recently, targeted therapies have emerged as promising modality for cancer targeting. We have discovered that high affinity plasma membrane receptor for interleukin-13 (IL-13), an immune regulatory cytokine, is over-expressed in 60-80% of malignant brain tumors. To target these IL-13R, we generated a chimeric fusion protein, composed of human IL-13 and mutated Pseudomonas exotoxin (PE), termed IL-13 cytotoxin (IL13-PE), and tested its cytotoxicity to IL-13R-expressing GBM cells. IL-13 cytotoxin was highly potent and selective in killing IL-13R-expressing GBM cells. In contrast, normal cells including brain, immune, and endothelial cells were generally not affected by this cytotoxin due to no or low expression of IL-13R. In vivo pre-clinical studies for safety and toxicity were also performed in mice, rats, and monkeys, and IL-13 cytotoxin was found to be well tolerated by both systemic and intracerebral administrations. IL-13 cytotoxin was found to mediate remarkable efficacy in animal models of human brain tumors. Encouraged by these pre-clinical studies, four Phase 1/2 clinical trials in adult patients with recurrent malignant glioma have been completed. These clinical trials involved convection-enhanced delivery (CED) of IL-13 cytotoxin either intratumoral or intraparenchymal after resection of tumor. CED is a novel loco-regional drug delivery method for intracranial tumors that relies on a continuous pressure gradient to distribute drug into interstitial space. This route of IL-13 cytotoxin administration appears to be very well tolerated and have a good risk-benefit profile. Most recently, a randomized controlled Phase 3 clinical trial (PRECISE) with intraparenchymal IL-13 cytotoxin administration was completed and subjects are being monitored for safety and survival.
Collapse
Affiliation(s)
- Mitomu Kioi
- Tumor Vaccines and Biotechnology Branch, Division of Cellular and Gene Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, 29 Lincoln Drive MSC 4555, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
7
|
Saito R, Krauze MT, Noble CO, Drummond DC, Kirpotin DB, Berger MS, Park JW, Bankiewicz KS. Convection-enhanced delivery of Ls-TPT enables an effective, continuous, low-dose chemotherapy against malignant glioma xenograft model. Neuro Oncol 2006; 8:205-14. [PMID: 16723630 PMCID: PMC1871954 DOI: 10.1215/15228517-2006-001] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Treatment of malignant gliomas represents one of the most formidable challenges in oncology. The combination of surgery, radiation, and chemotherapy yields median survivals of less than one year. Here we demonstrate the use of a minimally invasive surgical technique, convection-enhanced delivery (CED), for local administration of a novel nanoparticle liposome containing topotecan. CED of this liposomal topotecan (Ls-TPT) resulted in extended brain tissue retention (t1/2 = 1.5 days), whereas free topotecan was rapidly cleared (t1/2 = 0.1 days) after CED. The favorable pharmacokinetic profile of extended topotecan release for about seven days, along with biodistribution featuring perivascular accumulation of the nanoparticles, provided, in addition to the known topoisomerase I inhibition, an effective antiangiogenic therapy. In the rat intracranial U87MG tumor model, vascular targeting of Ls-TPT with CED was associated with reductions in laminin expression and vascular density compared to free topotecan or control treatments. A single CED treatment on day 7 showed that free topotecan conferred no survival benefit versus control. However, Ls-TPT produced a significant (P = 0.0002) survival benefit, with six of seven complete cures. Larger U87MG tumors, where CED of Ls-TPT on day 12 resulted in one of six cures, indicated the necessity to cover the entire tumor with the infused therapeutic agent. CED of Ls-TPT was also efficacious in the intracranial U251MG tumor model (P = 0.0005 versus control). We conclude that the combination of a novel nanoparticle Ls-TPT and CED administration was very effective in treating experimental brain tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Krystof S. Bankiewicz
- Address correspondence to Krystof S. Bankiewicz, Department of Neurological Surgery, University of California at San Francisco, 1855 Folsom Street, Mission Center Building Room 226, San Francisco, CA 94103 (
)
| |
Collapse
|
8
|
Yoshida D, Takahashi H, Teramoto A. Inhibition of glioma angiogenesis and invasion by SI-27, an anti-matrix metalloproteinase agent in a rat brain tumor model. Neurosurgery 2004; 54:1213-20; discussion 1220-1. [PMID: 15113477 DOI: 10.1227/01.neu.0000119237.46690.c6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2003] [Accepted: 12/17/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The matrix metalloproteinase (MMP) inhibitor SI-27 has undergone extensive development because of its effectiveness against glioma invasion and angiogenesis. However, previous studies have been performed in vitro. The present work investigates the potential of SI-27 to inhibit tumor invasion, slow angiogenesis, and prolong survival in rodent brain tumor models. METHODS Stable enhanced green fluorescent protein-expressing clones of a human malignant glioma cell line, U251MG, were stereotactically xenografted into the periphery of the anterior striatum and corpus callosum of Fischer 944 rats after immunosuppression with cyclosporin A. SI-27 (1 or 10 mg/kg) or carrier solution was administered on three successive days by intraperitoneal injection, and tumor invasion and angiogenesis were assessed 3 weeks later by quantitative image analysis. This was performed on whole brain sections analyzed either by direct observation of enhanced green fluorescent protein-expressing glioma cells or by additional immunohistochemistry to detect the endothelial cells with anti-factor VIII monoclonal antibody. In situ zymography on frozen sections was used to detect MMP activity. RESULTS The group receiving a total of 30 mg/kg showed a statistically significant (P < 0.001) increase in survival time compared with the controls receiving carrier (median survival, 47.3 versus 32.6 d). There was also a decrease in MMP activity, tumor cell invasion, and neovascularization. In contrast, animals given 3 mg/kg did not show these differences. CONCLUSION Systemic administration of the anti-MMP agent SI-27 is effective in the treatment of glioma in an animal model.
Collapse
Affiliation(s)
- Daizo Yoshida
- Department of Neurosurgery, Nippon Medical School, Tokyo, Japan.
| | | | | |
Collapse
|
9
|
Chen TC, Su S, Fry D, Liebes L. Combination therapy with irinotecan and protein kinase C inhibitors in malignant glioma. Cancer 2003; 97:2363-73. [PMID: 12712458 DOI: 10.1002/cncr.11307] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The topoisomerase-I inhibitor irinotecan (CPT-11) is currently used in Phase I/II trials for the treatment of patients with recurrent malignant gliomas. Protein kinase C (PKC) inhibitors such as high-dose tamoxifen and hypericin also have been used in the treatment of malignant gliomas. The current study examined the role of PKC inhibitors as chemosensitizers for CPT-11 and their proposed mechanism of action. Two glioma cell lines (A-172 and U-87) and one primary glioma cell culture (LA-567) were used. Proliferation ((3)H-thymidine) and cytotoxicity (methylthiotetrazole) studies were performed using CPT-11 (0-100 microM) alone, 7-ethyl-10-hydroxy camptothecin (SN-38) (0-1000 nM) alone or in the presence of a PKC inhibitor, tamoxifen (10 microM), hypericin (10 microM), calphositin C (400 nM), or staurosporine (10 nM). The terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling(TUNEL) assay was used to determine apoptosis as the mechanism of cytotoxicity; alterations in bcl-2 and bax expression were determined using Western blot analysis. Conversion of CPT-11 to SN-38 by glioma cells was determined using high-performance liquid chromatography (HPLC) analysis. Increasing CPT-11 and SN-38 concentrations induced cytotoxic morphologic changes, decreased proliferation, and increased cytotoxicity on all glioma cell lines tested. These changes were increased in the presence of a PKC inhibitor. The mechanism of the cytotoxicity was determined to be apoptosis by the TUNEL assay. The combination of a PKC inhibitor with CPT-11 or SN-38 led to decreased expression of the antiapoptotic protein bcl-2, and increased expression of the proapoptotic protein bax. HPLC analysis demonstrated conversion of CPT-11 to SN-38 by glioma cells. A combination of CPT-11 or SN-38 with a PKC inhibitor was found to lead to a decrease in proliferation and an increase in apoptosis in malignant glioma cells. The induction of apoptosis was secondary to a decrease in bcl-2 and an increase in bax expression. Glioma cells are capable of converting CPT-11 to SN-38 by intrinsic tumor carboxylesterases.
Collapse
Affiliation(s)
- Thomas C Chen
- Department of Neurosurgery, University of Southern California, School of Medicine, Los Angeles, California 90033, USA.
| | | | | | | |
Collapse
|