SQU Med J, February 2011, Vol. 11, Iss. 1, pp. 5-28, Epub. 12th Feb 11 Invited Article Submitted 4th Aug 10 Revision ReQ. 5th Oct 10, Revision recd. 25th Oct 10 Accepted 8th Nov 10 Glia-derived neoplasms of the brain (gliomas) of the histological grade 3 (anaplastic astrocytoma, anaplastic oligoastrocytoma, and anaplastic oligodendroglioma) and grade 4 (glioblastoma multiforme, [GBM]) according to the morphology- based classification of the World Health Organization (WHO),1 represent different stages of the same fatal neoplastic disease of the central nervous system (CNS). WHO grade 3 and 4 gliomas are also known as malignant gliomas and are the most frequent intrinsic brain tumours in adults.2 According to the US central brain tumour registry, the annual rate of primary tumours of the CNS is Department of Neurosurgery, Klinikum Augsburg, D-86156 Augsburg, Germany. *Corresponding Author email: nikolai.rainov@klinikum-augsburg.de oÈf§]< �Í �œ� e �Å<›Ö̌Á̌÷<9ËÜrj÷]95%), without evidence of recurrence by day 30. Unconjugated toxin components (CRM-107, 454A12, or rRA) caused significant, but less potent tumour growth inhibition than the conjugated toxin.76 In a more recent study in 2002, Engebraaten et al. compared the efficacy of TransMIDTM with that of PE conjugated with the 425.3 antibody directed against EGFR.77 Mice with subcutaneous gliomas or rats with intracranial gliomas received different doses (1-10 µg) of TransMIDTM, or 425.3-PE, injected intratumourally in established tumours. Both toxins showed significant antitumour effects in subcutaneous tumours, but only 425.3-PE was effective in intracranial gliomas in rats. Intracerebral TransMIDTM was toxic in doses above 10 ng, while intracerebral 425.3-PE was tolerated up to a dose of 4 µg per animal. The authors concluded that both toxins have promising efficacy in brain tumour models, but that 425.3-PE is better tolerated and has a more specific activity at higher doses.77 In 1997, Laske et al. treated 18 patients with recurrent malignant glioma with intratumoural high flow interstitial microinfusion of TransMIDTM in a dose-escalating single arm phase 1 clinical trial.78 The drug was infused at a maximum flow rate of 4-10 µl/min at a toxin concentration of 0.1 µg/ml. Nine of the 15 evaluable patients responded to treatment by at least 50% reduction in tumour volume on MRI, including 2 complete responses. Reduction in tumour volume occurred no earlier than 1 month after completion of the first toxin infusion. In 4 patients, the response was not maximal until 6–14 months after the Nikolai G Rainov and Volkmar Heidecke review | 17 distribution of the large molecules of immunotoxin in the microenvironment of the tumour and/or brain, as well as non-specific associated toxicity to glial and neuronal cells.19 With the introduction of MRI-based functional imaging and 3D volumetric techniques in routine clinical practice, it will be possible to determine quantitatively brain tumour volumes and the volume of infusate in the tumour and normal brain.81,82 Diffusion tensor imaging (DTI) is a new MRI imaging technique sensitive to directional movements of water molecules induced by tissue barriers.83 CED makes use of the extracellular space of the brain as a natural pathway for the widespread distribution of agents infused in an aqueous solution, and therefore DTI could be used for imaging of CED delivery without the need for contrast agents. Further key developments in MR imaging and MR-related computer software as well as infusion catheter design and computerised simulation and delivery modelling within the brain may provide much needed new resources to be combined with the biologically targeted toxins.19 Further improvements in the molecular diagnostics of malignant glioma with genetic profiling of individual patients and tumours should allow for selection of subgroups of cases with high expression of the target receptor, who are most likely to benefit from the administration of a targeted immunotoxin. Clinical protocols have explored several points of presumed significance, such as the use institution, and consisting of either nitrosoureas, platinum compounds, temozolomide, procarbazine, or PCV (procarbazine, CCNU [lomustine], vincristine). The primary endpoint of the study was overall survival time. Adult patients eligible for enrolment into this trial had to be diagnosed with histologically confirmed GBM and had to have undergone conventional treatment, including surgery (biopsy or debulking) and/or radiation therapy and/or chemotherapy. Patients were required to have a recurrent and/or progressive tumour ≥1.0 cm and ≤4.0 cm in diameter.79,80 The trial was however prematurely terminated in 2007 after an interim conditional power analysis showed that it was extremely unlikely that TransMIDTM would meet the trial criteria for efficacy.80 No data from this clinical trial has yet been published. Conclusions and Future Developments - Immunotoxins Targeted toxins have shown considerable promise in phase 1 and 2 clinical trials with recurrent malignant gliomas. There are however some major obstacles that need to be overcome before targeted toxins may enter the mainstream of brain tumour therapies. The most important of them are the heterogeneity of target receptor expression in malignant glioma cells, the non-uniform and poorly predictable Figure 4a: Schematic diagrams of virus vectors. Schematic representation of a wild type retrovirus (RV) and a RV vector with the corresponding packaging cell line (VPC). Wild type RV has a double-stranded RNA genome which is converted to DNA by viral reverse transcriptase and then integrated into the host genome at non-specific sites. The genetically modified RV vector genome contains the cis-acting elements required for replication (long terminal repeats - LTR, packaging sequence - Ψ), but lacks the viral genes (capsid core proteins - gag, reverse transcriptase, integrase and protease - pol, and the envelope antigens - env), which are replaced by up to 8 kb of foreign coding sequences (transgene cassette, e.g. cDNA encoding HSV-tk). A selectable marker (e.g. neomycin resistance gene - neo) allows for antibiotic selection of cells expressing the RV vector. In order that RV vectors replicate, it is necessary to provide the missing viral genes in trans, e.g. expressed within a genetically engineered RV VPC. VPC are most frequently of murine fibroblast origin. Separation of the packaging function from the genetic material to be transferred ensures that a replication incompetent RV is generated and makes RV vectors biologically safe. Clinical Development of Experimental Therapies for Malignant Glioma 18 | SQU Medical Journal, February 2011, Volume 11, Issue 1 as oncolytic viruses.85 Most of the viruses employed in clinical trials are derived from a retrovirus (RV), adenovirus (AV), or herpes simplex virus type 1 (HSV1) [Figure 4a and b]. Other viruses, such as the Newcastle disease virus (NDV) or reovirus, have been also employed in clinical trials, but to a much lesser extent.19,20,86-88 The effects of virus-mediated local therapy of malignant gliomas have been investigated only since the 1990s. The initially favoured and best explored gene therapy approach included the use of a replication-disabled, genetically modified virus vector capable of insertion of a toxicity-inducing gene into tumour cells.89-91 Most frequently, the inserted gene (transgene) rendered the infected tumour cells and their clonal progeny differentially sensitive to treatment with a pro-drug.91,92 The gene/vector system most widely utilised in initial clinical trials was the HSV thymidine kinase (HSV- tk) gene transferred by a replication-incompetent RV vector which was released in situ by fibroblast- derived virus-producing cells (VPC) [Figure 5].93 The most severe limitation of clinical gene therapy with non-replicating viruses was, however, their inability to achieve sufficiently high levels of gene expression in sufficiently large numbers of target tumour cells to result in a clinical benefit.19,94,95A modified approach was therefore introduced in order to improve anti-tumour toxicity and virus distribution within the tumour mass and the surrounding normal brain. It employed conditionally replicating oncolytic viruses with a lytic life cycle instead of their non-replicating of different numbers of catheters, positioning of catheters, surgical resection of the tumour before or after toxin infusion, and single versus repeated infusion, but there is no clear answer to any of these questions. It remains unknown whether there are benefits from combining targeted toxins with chemotherapy and/or with fractionated external radiation. Protocols have investigated patients with recurrent or progressing GBM, and the role of targeted toxin infusion in newly diagnosed malignant glioma remains to be determined. Evidence of prolonged survival and late local recurrences in treated glioma patients has raised the question of periodically repeated application of toxin for extended control of local recurrence and progression. The randomised clinical trials were able to provide some answers as to the efficacy of the studied toxins and clinical protocols; however, more clinical research is needed to address the above mentioned global issues. g e n e t i c a l ly m o d i f i e d v i r u s e s Wild type viruses have been long known for their capability to destroy malignant tumour cells upon infection and intracellular replication. Genetic engineering of some of these viruses was, however, only recently done in an attempt to improve their utility as targeted gene transfer anti-cancer agents.84 Recombinant and usually non-replicating viruses transferring to tumour cells genes with toxicity- inducing intracellular effects are known as virus vectors, while replicating viruses with a lytic life cycle selectively destroying tumour cells are known Figure 4b: Schematic representation of the genome of a wild type adenovirus (AV) and recombinant AV vectors. The wild type AV genome is flanked by the inverted terminal repeats (ITR) and divided into E1A, E1B, E2, E3, and E4 regions, whose genes are expressed in a defined temporal sequence. It contains 36 kb of double-stranded DNA, of which several regions can be deleted to accommodate up to 10 kb of foreign DNA (e.g. insertion of a therapeutic transgene cassette in the E1 region, deletion of E3 region). Replication-deficient AV vectors are generated by placing the AV genome in a plasmid and replacing E1 with a transgene (e.g. HSV-tk), then transfecting the plasmid into a packaging cell line (VPC) that provides E1 functions in trans. Nikolai G Rainov and Volkmar Heidecke review | 19 the same method and RV-VPC preparation, Shand et al. treated 48 patients with recurrent GBM in a phase 1/2 study.105 The authors of both reports independently concluded that RV-VPC injections were safe. Gene therapy in both trials resulted in a prolonged delay of recurrence in individual cases. Izquierdo et al. conducted phase 1 studies in 7 patients with recurrent GBM. Five patients received a single intratumoural RV-VPC injection and 2 patients intraoperative RV-VPC injections, later followed by repeated RV-VPC applications into the tumour resection cavity.106,107 In 2003, Prados et al. treated 30 patients in a phase 1/2 study with recurrent GBM by repeated direct injection of RV- VPC, initially immediately after tumour resection in the walls of the resection cavity, and a week later via an intracavitary catheter.108 Fifty per cent of the patients experienced serious side effects possibly related to RV-VPC therapy. The median survival of all patients in the treatment group was 8.4 months. Puumalainen et al. showed low tumour transduction efficiency with a RV-VPC construct carrying the marker gene lacZ.109 The same study also evaluated AV-mediated marker gene transfer and found much higher transduction rates in some counterparts.96-100 n o n-r e p l i c at i n g r e t r o v i r u s Starting in the late 1990s, several clinical studies were published which investigated the use of RV-mediated HSV-tk gene transfer delivered by intratumoural injections of VPC and followed by systemic administration of ganciclovir (GCV).89,90,91,101 The earliest phase 1/2 clinical study was published by Ram et al. in 1997.93 These authors investigated, in 15 patients with recurrent malignant glioma, the tolerability and efficacy of RV-producing VPC (RV-VPC) delivered by a single stereotactic intratumoural injection. Treatment resulted in anti-tumour activity in 4 patients with relatively small tumours. One patient with GBM had a complete response with a recurrence-free survival of 50 months.93 The limited efficiency of histologically demonstrated RV-mediated gene transfer suggested the presence of a bystander effect conferring toxicity to non-transduced tumour cells.102,103 In 1998, Klatzmann et al. treated 12 patients with recurrent GBM in a phase 1/2 study by local injections of RV-VPC into the margins of the tumour cavity after tumour debulking.104 Using Figure 5: Schematic representation of the mode of action of recombinant retrovirus (RV) vector-mediated suicide gene therapy in patients with malignant glioma. Up to 20 ml of a suspension of viable RV-producing cells (VPC) carrying the transgene coding for herpes simplex virus (HSV) thymidine kinase (HSV-tk) are manually injected into the walls of the tumour resection cavity at the end of the surgical removal of a human glioblastoma multiforme (GBM). RV vectors, which have been genetically engineered to become replication-deficient, are produced in high titres by the implanted VPC. GBM cells are highly mitotic and, after infection by RV, are able to produce most RV proteins, including HSV- tk. This enzyme converts the low-toxicity prodrug ganciclovir (GCV) into highly toxic metabolites, which block DNA replication during mitosis and render the tumour cells apoptotic. After repeated i.v. application of GCV to patients, all cells expressing HSV-tk are killed, including VPC. Cells not expressing HSV-tk may also be killed by the so-called “bystander effect” - transfer of toxic GCV metabolites from HSV-tk-expressing to HSV-tk-negative cells by direct cellular contact. Up to 10 non-expressing cells may be killed by one HSV-tk-expressing cells in cell culture. Clinical Development of Experimental Therapies for Malignant Glioma 20 | SQU Medical Journal, February 2011, Volume 11, Issue 1 The median survival of all patients was 10 months, while 5 patients were alive at 1 year and 1 patient at 3 years after therapy.111 Germano et al. carried out a dose-escalating phase 1 study in 11 patients with recurrent GBM.112 AV-HSV-tk doses of 2.5x1011-9x1011 virus particles were administered intraoperatively to the walls of the resection cavity immediately after tumour resection. The median survival of treated patients was 59 weeks. No major toxicities occurred, and the few serious AEs were not related to AV administration.112 Smitt et al. conducted another phase 1 dose escalation study in 14 patients with recurrent malignant glioma.113 Patients received 4.6x108-4.6x1011 virus particles injected manually in the walls of the tumour resection cavity at the end of tumour removal. Median overall survival was 4 months, however 4 patients survived for longer than 1 year following therapy. The treatment was safe and well tolerated. 113 Some groups were able to report significantly better clinical outcomes using the AV-HSV-tk approach. Sandmair et al. enrolled 21 patients with primary or recurrent malignant glioma in a comparative phase 1/2 study employing HSV-tk gene transfer by RV-VPC or by AV and comparing these with a third group receiving AV carrying only a marker gene, lacZ.114 The mean survival in the AV-HSV-tk group was significantly longer (15 months) than that of the other groups with mean survival of 7.4 months for RV-VPC and 8.3 months for AV-LacZ. No serious AE were reported in any of the groups. Building on these promising results, Immonen et al. conducted a randomised prospective phase 2 study to evaluate further the efficacy and safety of AV-HSV-tk in patients with primary or recurrent malignant glioma.115 Seventeen of the 36 patients received intraoperative injections of AV-HSV- tk (3x1010 pfu) into the walls of the surgically created resection cavity followed by GCV (5 mg/ kg twice daily for 14 days), and 19 patients had tumour surgery without AV injections. Standard radiotherapy was carried out in all patients with previously untreated tumours. The mean survival of patients in the AV-HSV-tk group was significantly longer (70.6 weeks) than in the control group (39.0 weeks) (P = 0.0095). An additional post hoc subgroup analysis excluding patients with anaplastic astrocytoma showed that there still was areas of the tumours. Harsh et al. performed a gene-marking and neuropathological study in 5 patients with recurrent GBM who received multiple stereotactically guided intratumoural injections of RV-VPC during a single surgical session.94 This study showed an extremely low gene transduction efficiency of the RV-VPC used. Finally, a large prospective randomised multicentre phase 3 clinical trial was carried out to allow definitive evaluation of RV-VPC treatment in patients with newly diagnosed GBM.101A total of 248 patients with previously untreated GBM were randomised to two groups and treated using either standard surgical resection and radiotherapy (n = 124) or standard therapy plus adjuvant RV-VPC injected in the walls of the resection cavity at tumour surgery (n = 124). Systemic administration of GCV (5 mg/kg i.v. twice daily) was started two weeks after surgery and continued for another 2 weeks. Somewhat surprisingly, this study found no significant difference in the progression-free, median, or 12-month survival of both the standard and the gene-therapy groups, although the approach was proven exceptionally safe. Based on the results of this trial, local therapy of malignant glioma with RV-VPC expressing HSV-TK has been largely abandoned.101 n o n-r e p l i c at i n g a d e n o v i r u s Studies with AV vectors were conducted and published, mostly in the late 1990s. Trask et al. conducted a phase 1 study of AV-HSV-tk in 12 patients with recurrent malignant glioma.110 A single stereotactic intratumoural injection was used to deliver AV doses of 2x109-2x1012 pfu. Adequate safety was reported with all doses except the highest, which caused significant CNS toxicity. Median survival in this study was only 4 months, however 3 patients showed long-term survival of 2 years or longer, which was interpreted as evidence for some local tumour control. Judy and Eck treated 13 patients with primary or recurrent malignant gliomas in a modified phase 1 study.111 Twelve patients received stereotactic intratumoural AV- HSV-tk injections and a week later underwent tumour mass resection with additional AV injections in the walls of the resection cavity. Total virus doses of 2x108-2x1011 pfu were used. Transient side effects, such as increased intracranial pressure (ICP), were noted only at the highest dose level. Nikolai G Rainov and Volkmar Heidecke review | 21 an acceptable safety profile.116 The results of the study have not been published yet, and final data still need to be provided. Preliminary information in the public space however states that the trial may be statistically underpowered and that Cerepro® has failed to show sufficient primary endpoint efficacy.117 o n c o ly t i c v i r u s e s Genetically modified and conditionally replicating AV and HSV have been the most frequently used viruses in early clinical studies with oncolytic viruses.87,89,97,99 Four phase 1 trials in recurrent malignant glioma have used intracerebrally inoculated G207 - a conditionally replicating HSV with defects in both ICP6 and ICP34.5 genes, which has demonstrated anti-tumour efficacy in preclinical studies in glioma.89,118,119 Markert et al. in 2000 carried out a dose escalation study treating 21 patients with recurrent malignant glioma using stereotactic intratumoural injections of G207 (up to 3x109 pfu total dose).89 MRI studies and neuropathological analysis suggested some anti-tumour activity of the treatment. The best responding 4 patients survived for a mean of 12.8 months, while the rest of the group had a mean survival of 6.2 months after therapy. Most importantly, this early trial showed that inoculation of a genetically engineered oncolytic HSV virus in the human brain is safe, despite the concerns about possible encephalitis well known from its wild type counterpart.89 Rampling et al. (2000) conducted a first phase 1 dose escalation study using another selectively replicating HSV mutant with disrupted ICP34.5 genes known as HSV1716.120 Nine patients with recurrent malignant glioma received stereotactic intratumoural injections of HSV1716 up to a dose of 1x105 pfu. Four patients were alive for 14–24 months after virus injection. No signs of encephalitis or major neurological side effects were noted.120 Papanastassiou et al. (2002) used intratumoural injections of HSV1716 (1x105 pfu) in another phase 1 study in 12 patients with malignant glioma.87 The authors were able to demonstrate replication of virus in tumours resected 4–9 days after the HSV1716 injection. No toxicity was apparent. Additional data from the above studies were published separately and suggested intratumoural persistence and replication a significant survival benefit in GBM patients (55.3 versus 37.0 weeks, P = 0.0214). The treatment was well tolerated and showed no major side effects.115 A phase 3 randomised and standard care- controlled multicentre pivotal trial (also known as study 904) using the above AV-HSV-tk vector115 designated as sitimagene ceradenovec (Cerepro®, Arc Therapeutics Ltd., London, UK) has been carried out in patients with newly diagnosed malignant glioma.116 Patients were randomised to either standard care plus Cerepro® or standard care alone. Standard care was surgery and radiotherapy or surgery and radiotherapy followed by temozolomide, resulting in 4 treatment groups. This allowed comparison of the efficacy of Cerepro® and temozolomide in the same trial without denying patients the best possible standard care. The primary endpoint was survival, defined as time to death or re-intervention.116 The overall primary endpoint analysis in the ITT population (n = 236) compared Cerepro® with and without TMZ against controls with and without TMZ. It showed a 42 day improvement in median survival (310 days versus 268 days) in the two groups receiving Cerepro®. The improvement over standard care reached statistical significance (P < 0.032). At the primary endpoint, the group with Cerepro® and TMZ showed an improvement of 68% in median survival time compared with standard care (surgery and radiotherapy) controls (350 days versus 208 days). Against the same controls, treatment with Cerepro® alone showed an improved median survival trend approaching 50%, similar to those given treatment with TMZ alone after standard care (300 days and 307 days, respectively versus 208 days with standard care). Improvements in the combined Cerepro® and TMZ treatment group (n = 58) and TMZ alone group (n = 76) were significant (P <0.05). In the Cerepro® alone treatment group (n = 61), the effect was not statistically significant (P < 0.065). Of the total 53 patients still to report an event, only 7 are in the surgery and radiotherapy control group and thus confidence intervals and statistical significance levels in all treatment groups might be expected to improve with time. Whilst increases were observed in hemiparesis, aphasia and pyrexia following therapy, the serious AE reports for Cerepro® were in line with those in the previous studies, indicating that the virus has Clinical Development of Experimental Therapies for Malignant Glioma 22 | SQU Medical Journal, February 2011, Volume 11, Issue 1 manual injection of virus into tumours or tumour- invaded surrounding brain, as well as intrinsic barriers to intra- and peritumoural spread in a glioma-harbouring brain, such as cysts, fibrotic membranes, and tumour necrosis.84,97 Therapy with oncolytic viruses seems to hold more promise in the few early clinical trials than the therapy with non-replicating virus vectors, although both approaches were already proven to be safe and lacking major side effects.96-99, 119,122,126 However, further major advancements in virus designs, application modalities, and understanding of the interactions of the host immune system with the virus are clearly needed before oncolytic virus therapy of malignant brain tumours may enter routine clinical use. Final Conclusions and Future Developments of Drug Delivery Modalities to Brain Tumours The therapeutic success of any tumour-targeting and locally delivered agent will depend not only on its ability to kill tumour cells selectively, but to a high degree also on the delivery mode and distribution throughout a tumour and the surrounding normal brain tissue.19,84 Diffusion of particles (e.g. viruses) or large molecules (e.g. recombinant toxins) in tissue is a rather inefficient way of distribution and will depend not only on the concentration of the compound, but also on its size, molecular weight, polarity, and its avidity for the target receptor.40,92,127 To circumvent this limitation, distribution of agents by CED, a much more efficient and fast mode for interstitial delivery by high-flow infusion, has been studied in several animal models.100,127 Local delivery of targeted toxins by CED seems to be the best approach to circumvent the limitations of the blood-brain barrier (BBB) and to increase therapeutic efficacy by high local concentrations of drug. All clinical trials with targeted toxins have adopted CED as the delivery mode of choice.19 Virus particles can also be delivered by CED, although this modality has been studied mainly in animal experiments,20,84,127,128 while all human clinical trials carried out so far have employed bolus injections of virus or carrier suspensions.91,94,101,124 Intratumoural stereotactically guided injections can provide adequate virus delivery only to spatially of HSV1716, allowing the virus to kill tumour cells over extended periods of time. Analysis of tumour explants showed viral replication for up to 9 days after initial injection, and the amount of recovered virus exceeded the input dose in some tumour samples. In addition, 1 patient was free of tumour progression at nearly 3 years, and 2 patients were alive and stable after almost 4 years.121 Harrow et al. (2004) followed 12 patients with newly diagnosed or recurrent malignant glioma treated in a previous study.122 Three long-time survivors with GBM were clinically stable at 15–22 months following surgery and virus injection. No long-term toxicity was reported.122 ONYX-015, a naturally occurring AV mutant with deletion in the viral E1B gene, has been previously used in clinical trials for head and neck cancer and gastro-intestinal tract malignancies.97,123,124 The first multicentre phase 1 study using ONYX-015 in patients with recurrent malignant glioma was published by Chiocca et al. in 2004.124 Twenty-four patients received intraoperative doses of ONYX-015 (1x107-1x1010 pfu) injected manually in the walls of the surgical cavity immediately after tumour resection. The median survival time for all patients was 6.2 months. No definitive anti-tumour effect could be demonstrated, however none of the patients, at any virus dose, experienced serious AE and there was no dose-limiting neurological toxicity.124 Conclusions and Future Developments - Genetically Modified Viruses The clinical efficacy of local therapy for malignant glioma with non-replicating viruses has been so far rather disappointing, possibly with the exception of the studies with AV-HSV-tk.115,116 No clinical studies using non-replicating virus vectors are currently enrolling patients. Encouraging anti-tumour activity has been demonstrated in the some of the studies in malignant glioma treated with local injections of oncolytic AV and HSV.97-99,122 Systemic chemotherapy has been found to potentiate the anti-tumour effect of virus mediated oncolysis in other cancer types.123,125 Factors likely to be an issue with any type of oncolytic virus are the physical limitations of Nikolai G Rainov and Volkmar Heidecke review | 23 epidemiology, and end results; and National cancer data base. Neurooncol 1999; 1:205–11. 5. Swanson KR, Alvord EC Jr, Murray JD. Virtual brain tumours (gliomas) enhance the reality of medical imaging and highlight inadequacies of current therapy. Br J Cancer 2002; 86:14–8. 6. Burton EC, Prados MD. Malignant gliomas. Curr Treat Options Oncol 2000; 1:459–68. 7. Janus TJ, Kyritsis AP, Forman AD, Levin VA. Biology and treatment of gliomas. Ann Oncol 1992; 3:423– 33. 8. Salcman M. Malignant glioma management. Neurosurg Clin N Am 1990; 1:49–63. 9. Schiffer D, Cavalla P, Dutto A, Borsotti L. Cell proliferation and invasion in malignant gliomas. Anticancer Res 1997; 17:61–9. 10. Guha A, Mukherjee J. Advances in the biology of astrocytomas. Curr Opin Neurol 2004; 17:655–62. 11. Mitchell P, Ellison DW, Mendelow AD. Surgery for malignant gliomas: mechanistic reasoning and slippery statistics. Lancet Neurol 2005; 4:413–22. 12. Whittle IR. Surgery for gliomas. Curr Opin Neurol 2002; 5:663–9. 13. Giese A, Westphal M. Treatment of malignant glioma: a problem beyond the margins of resection. J Cancer Res Clin Oncol 2001; 127:217–25. 14. Hess CF, Schaaf JC, Kortmann RD, Schabet M, Bamberg M. Malignant glioma: patterns of failure following individually tailored limited volume irradiation. Radiother Oncol 1994; 30:146–9. 15. Giese A, Bjerkvig R, Berens ME, Westphal M. Cost of migration: invasion of malignant gliomas and implications for treatment. J Clin Oncol 2003; 21:1624–36. 16. DeAngelis LM. Brain tumors. N Engl J Med 2001; 344:114–23. 17 Schiff D, Shaffrey ME. Role of resection for newly diagnosed malignant gliomas. Expert Rev Anticancer Ther 2003; 3:621–30. 18 Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352:987–96. 19. Chiocca EA, Broaddus WC, Gillies GT, Visted T, Lamfers ML. Neurosurgical delivery of chemotherapeutics, targeted toxins, genetic and viral therapies in neuro-oncology. J Neurooncol 2004; 69:101–17. 20. Rainov NG, Kramm CM. Vector delivery methods and targeting strategies for gene therapy of brain tumors. Curr Gene Ther 2001; 1:367–83. 21. Brem H, Gabikian P. Biodegradable polymer implants to treat brain tumors. J Control Release 2001; 74:63– 7. limited areas of tumour, since the number of injection sites is limited for practical reasons by tumour size, length of surgery and increasing risk of haemorrhage.94 Direct intratumoural injections into the walls of the tumour resection cavity, although they can be performed under direct visual control and with multiple virus depots close to each other, have the same basic limitations as stereotactic procedures.93,101 Moreover, the depth of injection is limited to 10–15 mm from the visible resection border, which seems to be insufficient to reach tumour cells migrating away from the main mass.101 CED has been shown to improve significantly virus distribution in and around experimental brain tumours.127 Virus particles may be efficiently delivered by CED over large areas of tumour and surrounding brain and achieve widespread distribution and much larger coverage than with bolus injections.92,127,128 Since a large amount of clinical experience has become available in the clinical trials employing CED of targeted toxins, it seems a logical step that clinical protocols with CED of viruses should be implemented in future. Such trials may be able to combine the biological advantages of oncolytic viruses with the spatial distribution affordable by CED. c o n f l i c t o f i n t e r e s t NGR has received research grants from Neurocrine Inc. (San Diego, CA) and IVAX Inc. (Miami, FL). The authors have no financial interests in any of the biotechnology companies mentioned in the review. References 1. Kleihues P, Cavenee WK. Pathology and genetics of tumours of the nervous system. Lyon: IARC Press, 2000. 2. Ohgaki H, Kleihues P. Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas. J Neuropathol Exp Neurol 2005; 64:479–89. 3. Davis FG, Freels S, Grutsch J, Barlas S, Brem S. Survival rates in patients with primary malignant brain tumors stratified by patient age and tumor histological type: an analysis based on surveillance, epidemiology, and end results (SEER) data, 1973– 1991. J Neurosurg 1998; 88:1–10. 4. Davis FG, McCarthy BJ, Berger MS. Centralized databases available for describing primary brain tumor incidence, survival, and treatment: Central brain tumor registry of the United States; surveillance, Clinical Development of Experimental Therapies for Malignant Glioma 24 | SQU Medical Journal, February 2011, Volume 11, Issue 1 2009; 110:583–8. 34. Rhines LD, Sampath P, DiMeco F, Lawson HC, Tyler BM, Hanes J, et al. Local immunotherapy with interleukin-2 delivered from biodegradable polymer microspheres combined with interstitial chemotherapy: a novel treatment for experimental malignant glioma. Neurosurgery 2003; 52:872–80. 35. Pastan I, Chaudhary VK, Fitzgerald DJ. Recombinant toxins as novel therapeutic agents. Annu Rev Biochem 1992; 61:331–54. 36. Pastan I, Fitzgerald D. Recombinant toxins for cancer treatment. Science 1992; 254:1173–7. 37. Hall WA, Fodstad O. Immunotoxins and central nervous system neoplasia. J Neurosurg 1992; 76:1– 12. 38. Reiter Y. Recombinant immunotoxins in targeted cancer cell therapy. Adv Cancer Res 2001; 81:93–124. 39. Husain SR, Puri RK. Interleukin-13 receptor-directed cytotoxin for malignant glioma therapy: from bench to bedside. J Neurooncol 2003; 65:37–48. 40. Rustamzadeh E, Low WC, Vallera DA, Hall WA. Immunotoxin therapy for CNS tumor. J Neurooncol 2003; 64:101–16. 41. Gottstein C, Winkler U, Bohlen H, Diehl V, Engert A. Immunotoxins: is there a clinical value? Ann Oncol 5 Suppl 1994; 1:97–103. 42. Puri RK, Siegel JP. Interleukin-4 and cancer therapy. Cancer Invest 1993; 11:473–86. 43. The PRECISE Trial: Study of IL13-PE38QQR compared to GLIADEL wafer in patients with recurrent glioblastoma multiforme. From: http:// clinicaltrials.gov, study identifier NCT00076986. Accessed: July 2010. 44. Puri RK, Leland P, Kreitman RJ, Pastan I. Human neurological cancer cells express interleukin-4 (IL- 4) receptors which are targets for the toxic effects of IL4-Pseudomonas exotoxin chimeric protein. Int J Cancer 1994; 58:574–81. 45. Kreitman RJ, Puri PK, Pastan I. A circularly permuted recombinant interleukin 4 toxin with increased activity. Proc Natl Acad Sci USA 1994; 91:6889–93. 46. Kreitman RJ, Puri RK, Pastan I. Increased antitumor activity of a circularly permuted IL-4-toxin in mice with IL-4 receptor bearing human carcinoma. Cancer Res 1995; 55:3357–63. 47. Frankel AE, Kreitman RJ, Sausville EA. Targeted toxins. Clin Cancer Res 2000; 6:326–34. 48. Puri RK, Hoon DS, Leland P, Snoy P, Rand RW, Pastan I, et al. Preclinical development of a recombinant toxin containing circularly permuted interleukin 4 and truncated Pseudomonas exotoxin for therapy of malignant astrocytoma. Cancer Res 1996; 56:5631– 7. 49. Joshi BH, Leland P, Asher A, Prayson RA, Varricchio F, Puri RK. In situ expression of interleukin-4 22. Gallia GL, Brem S, Brem H. Local treatment of malignant brain tumors using implantable chemotherapeutic polymers. J Natl Compr Canc Netw 2005; 3:721–8. 23. Brem H, Mahaley MS Jr, Vick NA, Black KL, Schold SC Jr, Burger PC, et al. Interstitial chemotherapy with drug polymer implants for the treatment of recurrent gliomas. J Neurosurg 1991; 74:441–6. 24. Brem H, Piantadosi S, Burger PC, Walker M, Selker R, Vick NA, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer Brain Tumor Treatment Group. Lancet 1995; 345:1008–12. 25. Westphal M, Hilt DC, Bortey E, Delavault P, Olivares R, Warnke PC, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neurooncol 2003; 5:79– 88. 26. Westphal M, Ram Z, Riddle V, Hilt D, Bortey E. Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir 2006; 148:269–75. 27. Fleming AB, Saltzman WM. Pharmacokinetics of the carmustine implant. Clin Pharmacokinet 2002; 41:403–19. 28. Brem H, Ewend MG, Piantadosi S, Greenhoot J, Burger PC, Sisti M. The safety of interstitial chemotherapy with BCNU-loaded polymer followed by radiation therapy in the treatment of newly diagnosed malignant gliomas: phase I trial. J Neurooncol 1995; 26:111–23. 29. Kleinberg LR, Weingart J, Burger P, Carson K, Grossman SA, Li K, et al. Clinical course and pathologic findings after Gliadel and radiotherapy for newly diagnosed malignant glioma: implications for patient management. Cancer Invest 2004; 22:1–9. 30. Valtonen S, Timonen U, Toivanen P, Kalimo H, Kivipelto L, Heiskanen O, et al. Interstitial chemotherapy with carmustine-loaded polymers for high-grade gliomas: a randomized double-blind study. Neurosurgery 1997; 41:44–8. 31. Olivi A, Grossman SA, Tatter S, Barker F, Judy K, Olsen J, et al. Dose escalation of carmustine in surgically implanted polymers in patients with recurrent malignant glioma: a new approaches to brain tumor therapy CNS consortium trial. J Clin Oncol 2003; 21:1845–9. 32. McGirt MJ, Brem H. Carmustine wafers (Gliadel) plus concomitant temozolomide therapy after resection of malignant astrocytoma: growing evidence for safety and efficacy. Ann Surg Oncol 2010; 17:1729–31. 33. McGirt MJ, Than KD, Weingart JD, Chaichana KL, Attenello FJ, Olivi A, et al. Gliadel (BCNU) wafer plus concomitant temozolomide therapy after primary resection of glioblastoma multiforme. J Neurosurg Nikolai G Rainov and Volkmar Heidecke review | 25 of head and neck: differential sensitivity to chimeric fusion proteins comprised of interleukin-13 and a mutated form of Pseudomonas exotoxin. Clin Cancer Res 2002; 8:1948–56. 61. Debinski W, Obiri NI, Powers SK, Pastan I, Puri RK. Human glioma cells overexpress receptors for interleukin 13 and are extremely sensitive to a novel chimeric protein composed of interleukin 13 and Pseudomonas exotoxin. Clin Cancer Res 1995; 1:1253–8. 62. Puri RK, Leland P, Obiri NI, Husain SR, Kreitman RJ, Haas GP, et al. Targeting of interleukin-13 receptor on human renal cell carcinoma cells by a recombinant chimeric protein composed of interleukin-13 and a truncated form of Pseudomonas exotoxin A (PE38QQR). Blood 1996; 87:4333–9. 63. Weingart J, Strauss LC, Grossman SA. Phase I/ II study: intratumoral infusion of IL13-PE38QQR cytotoxin for recurrent supratentorial malignant glioma. Neurooncol 2002; 4:379. 64. Kunwar S. Convection enhanced delivery of IL13- PE38QQR for treatment of recurrent malignant glioma: presentation of interim findings from ongoing phase 1 studies. Acta Neurochir Suppl 2003; 88:105–11. 65. Kunwar S, Prados M, Chang S, Berger MS, Lang FF, Piepmeier JM, et al. Peritumoral convection- enhanced delivery of IL13-PE38QQR in patients with recurrent malignant glioma - phase I interim results. Neurooncol 2003; 5:4. 66. Chang SM. Neuroradiographic changes following convection-enhanced delivery (CED) of the conjugated toxin IL13-PE38QQR following resection of recurrent malignant glioma. Neurooncol 2003; 5:4. 67. Prados MD, Lang FF, Stauss LC, Fleming CK, Aldape K, Kunwar S, et al. Intratumoral and intracerebral microinfusion of IL13-PE38QQR cytotoxin: phase I/II study of pre-and post-resection infusions in recurrent resectable maliganant glioma. Proc Am Soc Clin Oncol 2002; 21:2087. 68. Kunwar S, Chang S, Westphal M, Vogelbaum M, Sampson J, Barnett G, et al. Phase III randomized trial of CED of IL13-PE38QQR vs. Gliadel wafers for recurrent glioblastoma. Neuro Oncol 2010; 12:871– 81. 69. Weaver M, Laske DW. Transferrin receptor ligand- targeted toxin conjugate (Tf-CRM107) for therapy of malignant gliomas. J Neurooncol 2003; 65:3–13. 70. Johnson VG, Wilson D, Greenfield L, Youle RJ. The role of the diphtheria toxin receptor in cytosol translocation. J Biol Chem 1988; 263:1295–300. 71. Greenfield L, Johnson VG, Youle RJ. Mutations in diphtheria toxin separate binding from entry and amplify immunotoxin selectivity. Science 1987; 238:536–9. 72. Recht L, Torres CO, Smith TW, Raso V, Griffin TW. receptors in human brain tumors and cytotoxicity of a recombinant interleukin-4 (IL-4) cytotoxin in primary glioblastoma cell cultures. Cancer Res 2001; 61:8058–61. 50. Husain SR, Behari N, Kreitman RJ, Pastan I, Puri RK. Complete regression of established human glioblastoma tumor xenograft by interleukin-4 toxin therapy. Cancer Res 1998; 58:3649–53. 51. Weber F, Asher A, Bucholz R, Berger M, Prados M, Chang S, et al. Safety, tolerability, and tumor response of IL4-Pseudomonas exotoxin (NBI-3001) in patients with recurrent malignant glioma. J Neurooncol 2003; 64:125–37. 52. NBI-3001 followed by surgery in treating patients with recurrent glioblastoma multiforme. From: http://clinicaltrials.gov, study identifier NCT00014677. Accessed: July 2010. 53. Hunter T. The epidermal growth factor receptor gene and its product. Nature 1984; 311:414–6. 54. Wong AJ, Bigner SH, Bigner DD, Kinzler KW, Hamilton SR, Vogelstein B. Increased expression of the epidermal growth factor receptor gene in malignant gliomas is invariably associated with gene amplification. Proc Natl Acad Sci USA 1987; 84:6899–903. 55. Humphrey PA, Wong AJ, Vogelstein B, Friedman HS, Werner MH, Bigner DD, et al. Amplification and expression of the epidermal growth factor receptor gene in human glioma xenographs. Cancer Res 1988; 48:2231–8. 56. Liberman TA, Nusbaum HR, Razon N, Kris R, Lax I, Soreq H, et al. Amplification, enhanced expression, and possible rearrangement of EGF receptor gene in primary human brain tumors of glial origin. Nature 1985; 313:144–7. 57. Sampson JH, Akabani G, Archer GE, Bigner DD, Berger MS, Friedman AH, et al. Progress report of a phase I study of the intracerebral microinfusion of a recombinant chimeric protein composed of transforming growth factor (TGF)-alpha and a mutated form of the Pseudomonas exotoxin termed PE-38 (TP-38) for the treatment of malignant brain tumors. J Neurooncol 2003; 65:27–35. 58. Weber F. Preliminary results of a phase II study of TP- 38 immunotoxin delivered via convection-enhanced delivery to patients with recurrent glioblastoma multiforme: report of the TP-38 study group. Ann Meeting German Soc Neurosurg. From: http://www. egms.de/en/meetings/dgnc2005/ 05dgnc0094.htm. Accessed: July 2010. 59. Joshi BH, Plautz GE, Puri RK. Interleukin-13 receptor alpha chain: a novel tumor-associated transmembrane protein in primary explants of human malignant gliomas. Cancer Res 2000; 60:1168–72. 60. Joshi BH, Kawakami K, Leland P, Puri RK. Heterogeneity in interleukin-13 receptor expression and subunit structure in squamous cell carcinoma Clinical Development of Experimental Therapies for Malignant Glioma 26 | SQU Medical Journal, February 2011, Volume 11, Issue 1 Retrovirus therapy of tumors with activated Ras pathway. Science 1998; 282:1332–4. 87. Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D, Nicoll J, et al. The potential for efficacy of the modified (ICP 34.5-) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: a proof of principle study. Gene Ther 2002; 9:398–406. 88. Rainov NG, Kramm CM, Aboody-Guterman K, Chase M, Ueki K, Louis DN, et al. Retrovirus- mediated gene therapy of experimental brain neoplasms using the herpes simplex virus-thymidine kinase/ganciclovir paradigm. Cancer Gene Ther 1996; 3:99–106. 89. Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, Hunter WD, et al. Conditionally replicating herpes simplex virus mutant, G207 for the treatment of malignant glioma: results of a phase I trial. Gene Ther 2000; 7:867–74. 90. Oldfield EH, Ram Z, Culver KW, Blaese RM, DeVroom HL, Anderson WF, et al. Gene therapy for the treatment of brain tumors using intra-tumoral transduction with the thymidine kinase gene and intravenous ganciclovir. Hum Gene Ther 1993; 4:39– 69. 91. Rainov NG, Ren H. Gene therapy for human malignant brain tumors. Cancer J 2003; 9:180–8. 92. Muldoon LL, Kroll RA, Pagel MA, Roman- Goldstein S, Fiamengo SA, Neuwelt EA. Delivery of therapeutic genes to brain and intracerebral tumors. In: Chiocca EA, Breakefield XO, Eds. Gene therapy for neurological disorders and brain tumors. Boston: Humana Press, 1997. Pp. 128–39. 93. Ram Z, Culver KW, Oshiro EM, Viola JJ, DeVroom HL, Otto E, et al. Therapy of malignant brain tumors by intratumoral implantation of retroviral vector- producing cells. Nat Med 1997; 3:1354–61. 94. Harsh GR, Deisboeck TS, Louis DN, Hilton J, Colvin M, Silver JS, et al. Thymidine kinase activation of ganciclovir in recurrent malignant gliomas: a gene- marking and neuropathological study. Neurosurgery 2000; 92:804–11. 95. Ram Z, Barnett G, Vogelbaum M, Constantini S, Lillehei K, Sherman J, et al. Pre-operative infusion of IL13-PE38QQR cytotoxin by convection-enhanced delivery (CED) in recurrent malignant glioma: A phase I/II study. Proc Am Soc Clin Oncol 2003; 22:403. 96. Andreansky S, Soroceanu L, Flotte ER, Chou J, Markert JM, Gillespie GY, et al. Evaluation of genetically engineered herpes simplex viruses as oncolytic agents for human malignant brain tumors. Cancer Res 1997; 57:1502–9. 97. Chiocca EA. Oncolytic viruses. Nat Cancer Rev 2002; 2:938–51. 98. Dirven C, Van Beusechem V, Lamfers M, Grill J, Gerritsen WR, Vandertop WP. Oncolytic Transferrin receptor in normal and neoplastic brain tissue: implications for brain tumor immunotherapy. J Neurosurg 1990; 72:941–5. 73. Jefferies WA, Brandon MR, Hunt SV, Williams AF, Gatter KC, Mason DY. Transferrin receptor on endothelium of brain capillaries. Nature 1984; 312:162–3. 74. Johnson VG, Wrobel C, Wilson D, Zovickian J, Greenfield L, Oldfield EH. et al. Improved tumor- specific immunotoxins in the treatment of CNS and leptomeningeal neoplasia. J Neurosurg 1989; 70:240–8. 75. Hall WA, Godal A, Juell S, Fodstad O. In vitro efficacy of transferrin-toxin conjugates against glioblastoma multiforme. J Neurosurg 1992; 76:838–44. 76. Laske DW, Ilercil O, Akbasak A, Youle RJ, Oldfield EH. Efficacy of direct intratumoral therapy with targeted protein toxins for solid human gliomas in nude mice. J Neurosurg 1994; 80:520–6. 77. Engebraaten O, Hjortland GO, Juell S, Hirschberg H, Fodstad O. Intratumoral immunotoxin treatment of human malignant brain tumors in immunodeficient animals. Int J Cancer 2002; 97:846–52. 78. Laske DW, Youle RJ, Oldfield EH. Tumor regression with regional distribution of the targeted toxin TF- CRM107 in patients with malignant brain tumors. Nat Med 1997; 3:1362–8. 79. Study of therapy with TransMID™ compared to best standard of care in patients with glioblastoma multiforme. From: http://clinicaltrials.gov, study identifier NCT00083447, see also NCT00088400 and NCT00087230. Accessed: July 2010. 80. Celtic Pharma Terminates Transmid Trial KSB311R/ C I I I / 0 0 1 . Fr o m : http : / / w w w. d r u g s . co m / n e w s / celtic-pharma-terminates-transmid-trial-ksb311r- ciii-001-5246.html. Accessed: July 2010. 81. Briellmann RS, Pell GS, Wellard RM, Mitchell LA, Abbott DF, Jackson GD. MR imaging of epilepsy: state of the art at 1.5 T and potential of 3 T. Epileptic Disord 2003; 5:3–20. 82. Tummala RP, Chu RM, Liu H, Truwit CL, Hall WA. Application of diffusion tensor imaging to magnetic- resonance-guided brain tumor resection. Ped Neurosurg 2003; 39:39–43. 83. Cruz Junior LC, Sorensen AG. Diffusion tensor magnetic resonance imaging of brain tumors. Neurosurg Clin N Am 2005; 16:115–34. 84. Zlokovic BV, Apuzzo ML. Cellular and molecular neurosurgery: pathways from concept to reality - part II: vector systems and delivery methodologies for gene therapy of the central nervous system. Neurosurgery 1997; 40:805–13. 85. Pulkkanen KJ, Yla-Herttuala S. Gene therapy for malignant glioma: current clinical status. Mol Ther 2005; 12:585–98. 86. Coffey MC, Strong JE, Forsyth PA, Lee PW. Nikolai G Rainov and Volkmar Heidecke review | 27 HD, Wyde PR, Goodman JC. et al. Phase I study of adenoviral delivery of the HSV-tk gene and ganciclovir administration in patients with current malignant brain tumors. Mol Ther 2000; 1:195–203. 111. Judy KD, Eck SL. The use of suicide gene therapy for the treatment of malignancies of the brain. In: Lattime EC, Stanton LC, Eds. Gene therapy of cancer. San Diego, CA: Academic Press, 2002. Pp. 505–12. 112. Germano IM, Fable J, Gultekin SH, Silvers A. Adenovirus/herpes simplex–thymidine kinase/ ganciclovir complex: preliminary results of a phase I trial in patients with recurrent malignant gliomas. J Neurooncol 2003; 65:279–89. 113. Smitt PS, Driesse M, Wolbers J, Kros M, Avezaat C. Treatment of relapsed malignant glioma with an adenoviral vector containing the herpes simplex thymidine kinase gene followed by ganciclovir. Mol Ther 2003; 7:851–8. 114. Sandmair AM, Loimas S, Puranen P, Immonen A, Kossila M, Puranen M, et al. Thymidine kinase gene therapy for human malignant glioma, using replication-deficient retroviruses or adenoviruses. Hum Gene Ther 2000; 11:2197–205. 115. Immonen A, Vapalahti M, Tyynela K, Hurskainen H, Sandmair A, Vanninen R, et al. AdvHSV-tk gene therapy with intravenous ganciclovir improves survival in human malignant glioma: a randomised, controlled study. Mol Ther 2004; 10:967–72. 116. Preliminary Cerepro® Phase III results meet primary endpoint - Secondary endpoints yet to be established with 45% of patients still alive. From: http://www.dg- gt.de/docs/CereproPhase.pdf. Accessed: July 2010. 117. Mitchell P. Ark’s gene therapy stumbles at the finish line. Nat Biotech 2010; 28:183–4. 118. Mineta T, Rabkin SD, Yazaki T, Hunter WD, Martuza RL. Attenuated multi-mutated herpes simplex virus-1 for the treatment of malignant gliomas. Nat Med 1995; 1:938–43. 119. Markert JM, Parker JN, Gillespie GY, Whitley RJ. Genetically engineered human herpes simplex virus in the treatment of brain tumours. Herpes 2001; 8:17–22. 120. Rampling R, Cruickshank G, Papanastassiou V, Nicoll J, Hadley D, Brennan D, et al. Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther 2000; 7:859–66. 121. Harland J, Papanastassiou V, Brown SM. HSV1716 persistence in primary human glioma cells in vitro. Gene Ther 2002; 9:1194–8. 122. Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser M, et al. HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: safety data and long-term survival. Gene Ther 2004; 11:1648–58. 123. Khuri FR, Nemunaitis J, Ganly I, Arseneau J, Tannock IF, Romel L, et al. A controlled trial of intratumoral adenoviruses for treatment of brain tumours. Expert Opin Biol Ther 2002; 2:943–52. 99. Heise C, Kirn DH. Replication-selective adenoviruses as oncolytic agents. J Clin Invest 2000; 105:847–51. 100. Baxter LT, Jain RK. Transport of fluid and macromolecules in tumors I. Role of interstitial pressure and convection. Microvasc Res 1989; 37:77–102. 101. Rainov NG. A phase III clinical evaluation of herpes simplex virus type 1 thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and radiation in adults with previously untreated glioblastoma multiforme. Hum Gene Ther 2000; 11:2389–401. 102. Freeman SM, Abboud CN, Whartenby KA, Packman CH, Koeplin DS, Moolten FL, et al. The “bystander effect”: tumor regression when a fraction of the tumor mass is genetically modified. Cancer Res 1993; 53:5274–83. 103. Moolten FL. Tumor chemosensitivity conferred by inserted herpes thymidine kinase genes: paradigm for a prospective cancer control strategy. Cancer Res 1986; 46:5276–81. 104. Klatzmann D, Valery CA, Bensimon G, Marro B, Boyer O, Mokhtari K, et al. A phase I/II study of herpes simplex virus type 1 thymidine kinase “suicide” gene therapy for recurrent glioblastoma. Hum Gene Ther 1998; 9:2595–604. 105. Shand N, Weber F, Mariani L, Bernstein M, Gianella- Borradori A, Long Z, et al. A phase 1-2 clinical trial of gene therapy for recurrent glioblastoma multiforme by tumor transduction with the herpes simplex thymidine kinase gene followed by ganciclovir. Hum Gene Ther 1999; 10:2325–35. 106. Izquierdo M, Cortes ML, Martin V, de Felipe P, Izquierdo JM, Pérez-Higueras A, et al. Gene therapy in brain tumours: implications of the size of glioblastoma on its curability. Acta Neurochir Suppl 19097; 68:111–7. 107. Izquierdo M, Martin V, de Felipe P, Izquierdo JM, Pérez-Higueras A, Cortés ML, et al. Human malignant brain tumor response to herpes simplex thymidine kinase (HSVtk)/ganciclovir gene therapy. Gene Ther 1996; 3:491–5. 108. Prados MD, McDermott M, Chang SM, Wilson CB, Fick J, Culver KW, et al. Treatment of progressive or recurrent glioblastoma multiforme in adults with herpes simplex virus thymidine kinase gene vector- producer cells followed by intravenous ganciclovir administration: a phase I/II multi-institutional trial. J Neurooncol 2003; 65:269–78. 109. Puumalainen A, Vapalahti M, Agrawal RS, Kossila M, Laukkanen J, Lehtolainen P, et al. ß-galactosidase gene transfer to human malignant glioma in vivo using replication-deficient retroviruses and adenoviruses. Hum Gene Ther 1998; 9:1769–74. 110. Trask TW, Trask RP, Aguilar-Cordova E, Shine Clinical Development of Experimental Therapies for Malignant Glioma 28 | SQU Medical Journal, February 2011, Volume 11, Issue 1 126. Jiang H, Gomez-Manzano C, Lang FF, Alemany R, Fueyo J. Oncolytic adenovirus: preclinical and clinical studies in patients with human malignant gliomas. Curr Gene Ther 2009; 9:422–7. 127. Bankiewicz KS, Eberling JL, Kohutnicka M, Jagust W, Pivirotto P, Bringas J, et al. Convection-enhanced delivery of AAV vector in parkinsonian monkeys; in vivo detection of gene expression and restoration of dopaminergic function using pro-drug approach. Exp Neurol 2000; 164:2–14. 128. Nilaver G, Muldoon LL, Kroll RA, Pagel MA, Breakefield XO, Davidson BL, et al. Delivery of herpesvirus and adenovirus to nude rat intracerebral tumors after osmotic blood-brain barrier disruption. Proc Natl Acad Sci USA 1995; 92:9829– 33. ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med 2000; 6:879–85. 124. Chiocca EA, Abbed KM, Tatter S, Louis DN, Hochberg FH, Barker F, et al. A phase I open-label, dose-escalation, multi-institutional trial of injection with an E1B-attenuated adenovirus, ONYX-015, into the peritumoral region of recurrent malignant gliomas, in the adjuvant setting. Mol Ther 2004; 10:958–66. 125. Nemunaitis J, Khuri F, Ganly I, Arseneau J, Posner M, Vokes E, et al. Phase II trial of intratumoral administration of ONYX-015, a replication-selective adenovirus, in patients with refractory head and neck cancer. J Clin Oncol 2001; 19:289–98.