Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Neurochirurgie ; 63(5): 426-429, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27615154

ABSTRACT

Intramedullary spinal cavernoma is a rare vascular disease constituting 5-12% of all spinal vascular tumors. The clinical course is usually characterized either by an acute neurological deterioration, recurrent episodes of neurological deficits or by a slowly progressive neurological decline. Microsurgical removal is recommended when the symptoms become clinically relevant and the lesion appears accessible. In this article, we present a surgical technique to completely resect an intramedullary cavernoma with the aid of intraoperative electrophysiological monitoring and intraoperative real-time ultrasound guidance. A brief description of current management of this pathology is also presented.


Subject(s)
Hemangioma, Cavernous/surgery , Spinal Cord Neoplasms/surgery , Hemangioma, Cavernous/diagnostic imaging , Hemangioma, Cavernous/physiopathology , Humans , Microsurgery/methods , Monitoring, Intraoperative , Neurosurgical Procedures/methods , Spinal Cord Neoplasms/diagnostic imaging , Spinal Cord Neoplasms/physiopathology
3.
Ultraschall Med ; 36(2): 174-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25429625

ABSTRACT

PURPOSE: Brain shift and tissue deformation during surgery for intracranial lesions are the main actual limitations of neuro-navigation (NN), which currently relies mainly on preoperative imaging. Ultrasound (US), being a real-time imaging modality, is becoming progressively more widespread during neurosurgical procedures, but most neurosurgeons, trained on axial computed tomography (CT) and magnetic resonance imaging (MRI) slices, lack specific US training and have difficulties recognizing anatomic structures with the same confidence as in preoperative imaging. Therefore real-time intraoperative fusion imaging (FI) between preoperative imaging and intraoperative ultrasound (ioUS) for virtual navigation (VN) is highly desirable. We describe our procedure for real-time navigation during surgery for different cerebral lesions. MATERIALS AND METHODS: We performed fusion imaging with virtual navigation for patients undergoing surgery for brain lesion removal using an ultrasound-based real-time neuro-navigation system that fuses intraoperative cerebral ultrasound with preoperative MRI and simultaneously displays an MRI slice coplanar to an ioUS image. RESULTS: 58 patients underwent surgery at our institution for intracranial lesion removal with image guidance using a US system equipped with fusion imaging for neuro-navigation. In all cases the initial (external) registration error obtained by the corresponding anatomical landmark procedure was below 2 mm and the craniotomy was correctly placed. The transdural window gave satisfactory US image quality and the lesion was always detectable and measurable on both axes. Brain shift/deformation correction has been successfully employed in 42 cases to restore the co-registration during surgery. The accuracy of ioUS/MRI fusion/overlapping was confirmed intraoperatively under direct visualization of anatomic landmarks and the error was < 3 mm in all cases (100 %). CONCLUSION: Neuro-navigation using intraoperative US integrated with preoperative MRI is reliable, accurate and user-friendly. Moreover, the adjustments are very helpful in correcting brain shift and tissue distortion. This integrated system allows true real-time feedback during surgery and is less expensive and time-consuming than other intraoperative imaging techniques, offering high precision and orientation.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/surgery , Imaging, Three-Dimensional/instrumentation , Imaging, Three-Dimensional/methods , Intraoperative Complications/diagnosis , Intraoperative Complications/surgery , Intraoperative Period , Magnetic Resonance Imaging, Interventional/instrumentation , Magnetic Resonance Imaging, Interventional/methods , Multimodal Imaging/instrumentation , Multimodal Imaging/methods , Neuronavigation/instrumentation , Neuronavigation/methods , Preoperative Care , Ultrasonography, Interventional/instrumentation , Ultrasonography, Interventional/methods , User-Computer Interface , Adolescent , Adult , Aged , Brain Neoplasms/secondary , Cerebral Cortex/pathology , Cerebral Cortex/surgery , Child , Craniotomy/instrumentation , Craniotomy/methods , Equipment Design , Female , Humans , Male , Middle Aged , Sensitivity and Specificity
4.
J Neurooncol ; 106(3): 595-600, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21874383

ABSTRACT

In 1991, a prospective phase II trial was initiated to evaluate the efficacy of treatment for adults with medulloblastoma (MB). After surgery, patients were staged with a neuroradiologic examination of the brain and neuroaxis and by cerebrospinal fluid cytology. All patients received three cycles of upfront cisplatinum (cisplatinum) and etoposide (VP16) chemotherapy followed by cranio-spinal radiation therapy. The current article reports on the long-term results from that trial. After a median follow-up of 14.9 years, among a total of 28 adults with MB, the overall progression-free survival and overall survival (OS) rates at 5 years were 57.6 and 80%, respectively. The median OS for the whole group of patients was 11.3 years. The observed toxicity was mainly hematological, with leukopenia and thrombocytopenia (16% of grades 3 and 4). In summary, in our small series of patients, the role of combination administration of CDDP + VP16 started before the initiation of radiotherapy in reducing recurrences, particularly distant recurrences, remains unclear. To know whether adding chemotherapy to craniospinal radiation in adult therapy increases relapse-free and overall survival, we must await the results of a larger randomized controlled clinical trial.


Subject(s)
Antineoplastic Agents/administration & dosage , Cerebellar Neoplasms/drug therapy , Cisplatin/administration & dosage , Etoposide/administration & dosage , Medulloblastoma/drug therapy , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/radiotherapy , Female , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Male , Medulloblastoma/mortality , Medulloblastoma/radiotherapy , Middle Aged , Survival Analysis
5.
Oncogene ; 28(15): 1807-11, 2009 Apr 16.
Article in English | MEDLINE | ID: mdl-19287454

ABSTRACT

Glioblastomas (GBMs) contain transformed, self-maintaining, multipotent, tumour-initiating cancer stem cells, whose identification has radically changed our perspective on the physiology of these tumours. Currently, it is unknown whether multiple types of transformed precursors, which display alternative sets of the complement of properties of true cancer stem cells, can be found in a GBM. If different subsets of such cancer stem-like cells (CSCs) do exist, they might represent distinct cell targets, with a differential therapeutic importance, also depending on their characteristics and lineage relationship. Here, we report the presence of two types of CSCs within different regions of the same human GBM. Cytogenetic and molecular analysis shows that the two types of CSCs bear quite diverse tumorigenic potential and distinct genetic anomalies, and, yet, derive from common ancestor cells. This provides critical information to unravel the development of CSCs and the key molecular/genetic components underpinning tumorigenicity in human GBMs.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Animals , Cell Line, Tumor , Chromosome Aberrations , Genome , Humans , Male , Mice , Mice, SCID , Middle Aged
6.
Nature ; 444(7120): 761-5, 2006 Dec 07.
Article in English | MEDLINE | ID: mdl-17151667

ABSTRACT

Transformed, oncogenic precursors, possessing both defining neural-stem-cell properties and the ability to initiate intracerebral tumours, have been identified in human brain cancers. Here we report that bone morphogenetic proteins (BMPs), amongst which BMP4 elicits the strongest effect, trigger a significant reduction in the stem-like, tumour-initiating precursors of human glioblastomas (GBMs). Transient in vitro exposure to BMP4 abolishes the capacity of transplanted GBM cells to establish intracerebral GBMs. Most importantly, in vivo delivery of BMP4 effectively blocks the tumour growth and associated mortality that occur in 100% of mice after intracerebral grafting of human GBM cells. We demonstrate that BMPs activate their cognate receptors (BMPRs) and trigger the Smad signalling cascade in cells isolated from human glioblastomas (GBMs). This is followed by a reduction in proliferation, and increased expression of markers of neural differentiation, with no effect on cell viability. The concomitant reduction in clonogenic ability, in the size of the CD133+ population and in the growth kinetics of GBM cells indicates that BMP4 reduces the tumour-initiating cell pool of GBMs. These findings show that the BMP-BMPR signalling system--which controls the activity of normal brain stem cells--may also act as a key inhibitory regulator of tumour-initiating, stem-like cells from GBMs and the results also identify BMP4 as a novel, non-cytotoxic therapeutic effector, which may be used to prevent growth and recurrence of GBMs in humans.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , AC133 Antigen , Animals , Antigens, CD/metabolism , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Glycoproteins/metabolism , Humans , Mice , Neoplastic Stem Cells/cytology , Peptides/metabolism , Signal Transduction/drug effects , Stem Cell Transplantation
7.
Acta Neurochir (Wien) ; 146(9): 995-1001; discussion 1001, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15340811

ABSTRACT

OBJECTIVE: The object of this study is to describe a new method for performing craniotomies which obviates the need for burr holes, improves bony reconstruction, and reduces post-operative cosmetic deformities. Moreover, this technique provides excellent exposure of skull base structures and dural venous sinuses. METHODS: Craniotomies in varied locations are created with the use of a micro-oscillating saw and chisel. No burr holes are used and reconstruction with plates and screws is unnecessary. RESULTS: We initially applied our technique to approaches to the anterior skull base with combined craniofacial tumour resections. We have since performed over 2000 craniotomies of any size and shape in all supratentorial locations using the oscillating saw. CONCLUSIONS: We have found that our method creates better cosmetic results than standard techniques and is safer for craniotomies spanning dural venous sinuses. With experience, operating time was significantly reduced and costs were lowered because reconstruction with fixation devices was not needed.


Subject(s)
Craniotomy/instrumentation , Surgical Instruments , Brain Neoplasms/surgery , Humans , Skull Base/surgery , Skull Base Neoplasms/surgery
8.
Vet Pathol ; 39(3): 293-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12014493

ABSTRACT

Despite recent advances in surgical technology, resection is not an option for many brainstem tumors. Experimental models have played essential roles in examining new approaches to therapy. The objective of the present study was to generate models by determining coordinates for safe inoculation into the brainstem of mice and rats, and to establish whether the implantation of heterotopic cells would create reproducible survival curves. Morbidity and survival studies were used to map stereotactic coordinates allowing successful inoculation of tumor cells. Survival studies were used to investigate the time course of tumor growth. Tumor location was examined by light microscopy and magnetic resonance imaging. Mice survived injections of 2 microL of saline at interaural, lateral, and depth coordinates of -2.5, 1.0, and 3.5 mm and -1.5, 1.0, and 3.5 mm. Rats survived injections at interaural, lateral, and depth coordinates of -2.0, 2.0, and 7.0 mm and -3.0, 0, and 7.0 mm. Median survival of mice challenged with 5 x 10(5) EMT6 and 10(4) B16 tumor cells was 11 and 10 days, respectively. Median survival for rats challenged with 10(4) 9L and F98 cells was 14 and 13 days, respectively. The present study demonstrates a feasible approach to preparing models of brainstem tumors. Limitations of these models are discussed.


Subject(s)
Brain Stem Neoplasms/pathology , Disease Models, Animal , Tumor Cells, Cultured/transplantation , Animals , Female , Magnetic Resonance Imaging , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Rats, Nude , Survival Analysis
9.
Pharm Res ; 18(7): 899-906, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11496947

ABSTRACT

PURPOSE: The purpose of our study was to develop an injectable polymeric system for the long-term localized delivery of bioactive interleukin-2 for antitumor immunotherapy. METHODS: IL-2 was encapsulated into gelatin and chondroitin-6-sulfate using an aqueous-based complex coacervation. CTLL-2 cells were used to measure the bioactivity of released IL-2 and radiolabeled IL-2 was used for release studies in the rat brain and mouse liver. Antitumor efficacy studies were carried out in primary (9L gliosarcoma) and metastatic (B16-F10 melanoma) brain tumor models in rats and mice, respectively, as well as a murine liver tumor model (CT26 carcinoma). Survivors of the metastatic brain tumor challenge were rechallenged with tumor in the opposite lobe of the brain to confirm that antitumor immunologic memory had developed. RESULTS: Bioactive IL-2 was released for over 2 weeks in vitro and in vivo IL-2 release showed significant IL-2 levels for up to 21 days. Polymeric IL-2 microspheres injected intratumorally were statistically more effective in protecting animals challenged with fatal tumor doses in the brain and the liver than placebo or autologous tumor cells genetically engineered to secrete IL-2. Immunologic memory was induced following IL-2 microsphere therapy in the B16-F10 brain tumor model that was capable of protecting 42% of animals from a subsequent intracranial tumor challenge, suggesting that tumor destruction was mediated by the immune system. CONCLUSIONS: Local IL-2 therapy using novel polymeric carriers. aimed at stimulating long-lasting antitumor immunity, may provide an improved method of treating a variety of cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Biocompatible Materials/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems/methods , Interleukin-2/administration & dosage , Liver Neoplasms, Experimental/drug therapy , Animals , Biocompatible Materials/metabolism , Brain/drug effects , Brain/pathology , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Liver Neoplasms, Experimental/prevention & control , Melanoma, Experimental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microspheres , Polymers/administration & dosage , Polymers/metabolism , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
10.
J Neurosurg Sci ; 45(4): 216-9; discussion 219, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11912473

ABSTRACT

Pigmented villonodular synovitis (PVNS) is a disease of the joints which uncommonly involves the spine. We present a 70-year-old woman with radicular symptoms who was found to have a mass arising from a lumbar zygapophyseal joint with extension into the spinal canal. Following gross-total excision of the mass, histology revealed PVNS. One month after surgery, the patient had no symptoms and there was no evidence of residual or recurrent disease.


Subject(s)
Lumbar Vertebrae/diagnostic imaging , Spinal Diseases/diagnostic imaging , Spinal Diseases/pathology , Synovitis, Pigmented Villonodular/diagnostic imaging , Synovitis, Pigmented Villonodular/pathology , Aged , Decompression, Surgical , Female , Humans , Laminectomy , Leg , Lumbar Vertebrae/surgery , Pain/etiology , Radiography , Spinal Diseases/surgery , Synovitis, Pigmented Villonodular/complications , Synovitis, Pigmented Villonodular/surgery
11.
Neurosurgery ; 47(6): 1296-304; discussion 1304-5, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11126900

ABSTRACT

OBJECTIVE: Combined craniofacial resection has become the standard approach for malignant tumors involving the cribriform plate and anterior cranial fossa. Despite its widespread application, however, many surgeons agree that the procedure carries a risk of significant morbidity and even mortality. The purpose of this study was to analyze the experience at a single institution to determine the incidence of early postoperative complications encountered after combined craniofacial resection of tumors involving the cribriform plate and to provide information to improve management. METHODS: Between 1987 and 1997, 168 patients underwent combined craniofacial resection at the National Cancer Institute of Milan for tumors involving the cribriform plate. Patient charts, operative notes, follow-up clinic notes, radiographic studies, and pathology reports were analyzed. Morbidity encountered in the first 30 cases was compared with that encountered in the subsequent 138 cases. RESULTS: The most frequently encountered pathological findings were adenocarcinoma (53.6%), squamous cell carcinoma (17%), and esthesioneuroblastoma (9.8%). Eight patients (4.7%) died, 6 of whom were among the first 30 patients to undergo resection. Among patients with fatal complications were three with meningoencephalitis, three with intracranial hemorrhage, and one with myocardial infarction. Fifty patients (29.7%) had nonfatal morbidity; 16 of these patients were among the first 30 patients operated. Transient cerebrospinal fluid leakage was the most frequent adverse effect (9.5%); 12 patients (7.1%) had pneumocephalus, 3 (1.8%) had meningitis, 4 (2.4%) had wound infections, 3 (1.8%) experienced transient impairment of mental status, 3 (1.8%) had transient diplopia, 2 (1.2%) had diabetes insipidus, and 1 (0.6%) had bone flap necrosis. CONCLUSION: We observed a dramatic decrease in mortality and morbidity in patients who underwent combined craniofacial resection after the first 30 cases in our series. Improvement of specific aspects of surgical technique, such as more refined reconstructive methods and improved prophylactic antibiotic therapy, is at least partly responsible for this favorable trend.


Subject(s)
Ethmoid Bone/surgery , Neurosurgical Procedures , Skull Neoplasms/surgery , Adult , Aged , Face/surgery , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Morbidity , Postoperative Complications/diagnosis , Postoperative Complications/mortality , Skull/surgery , Skull Neoplasms/diagnosis , Skull Neoplasms/mortality , Tomography, X-Ray Computed
12.
J Neurosurg ; 93(4): 693-7, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11014551

ABSTRACT

The synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome is a recently described, currently evolving clinical entity that groups together several idiopathic disorders of bone and skin formerly described under a variety of names. Among the spectrum of possible locations for the bone lesions, there is no previous report in the literature of primary involvement of the skull vault. A patient with primary involvement of the calvaria in the setting of SAPHO syndrome is described here, which, to the authors' knowledge, is the first report of such localization. The clinically and radiologically benign evolution of the different stages of the bone lesions is presented. The authors suggest that the SAPHO syndrome should be considered in the differential diagnosis of lytic, sclerotic, or hyperostotic lesions of the skull, particularly before considering invasive diagnostic procedures.


Subject(s)
Acquired Hyperostosis Syndrome/pathology , Skull/pathology , Adult , Diagnosis, Differential , Female , Humans , Osteitis/diagnosis , Osteitis/pathology
13.
Forum (Genova) ; 10(2): 152-65, 2000.
Article in English | MEDLINE | ID: mdl-10875976

ABSTRACT

Controlled delivery of chemotherapeutic agents by biodegradable polymers is a new strategy that has been added to the arsenal available for the treatment of malignant neoplasms. This approach is particularly suitable for the management of brain tumours because of the constraints imposed by the blood brain barrier (BBB). The use of polymers for local drug delivery minimises systemic toxicity, while achieving prolonged elevation of intratumoural drug concentrations that results in improved efficacy. In addition, this strategy broadens the spectrum of drugs available for the treatment of neoplasms in the central nervous system to include agents whose efficacy is significantly limited by systemic toxicity or inability to penetrate the BBB. In this review, we discuss the rationale and background for the use of this novel approach. We also summarise the clinical trials and laboratory investigations leading to the development of local delivery of anti-neoplastic agents from biodegradable polymers for the treatment of malignant gliomas.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioma/drug therapy , Animals , Biodegradation, Environmental , Delayed-Action Preparations , Drug Evaluation , Humans , Microspheres , Polymers
14.
J Neurosurg ; 92(3): 419-27, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10701528

ABSTRACT

OBJECT: Interleukin-12 (IL- 12) has potential for the treatment of tumors because it can stimulate an antitumor immune response and possesses antiangiogenic properties. In the study reported here, the authors investigated the therapeutic role of locally delivered IL-12 in a malignant brain tumor model. METHODS: After genetically engineering 9L gliosarcoma cells to express IL-12 (9L-IL12 cells), the authors used these cells as a source of locally delivered cytokine. First, they investigated the behavior of these cells, which were implanted with the aid of stereotactic guidance into the rat brain, by using serial magnetic resonance imaging and histopathological examination. Second, they assessed the antitumor efficacy of proliferating, as well as nonproliferating (irradiated), 9L-IL12 cells by implanting these cells in animals challenged by wild-type 9L gliosarcoma (9Lwt) cells. The IL-12 expression in brain regions injected with 9L-IL12 was confirmed by reverse transcription-polymerase chain reaction. Last, the authors explored whether animals treated with 9L-IL12 cells developed an antitumor immunological memory by rechallenging the survivors with a second injection of 9Lwt cells. The authors demonstrated that local delivery of IL-12 into the rat brain by genetically engineered cells significantly prolongs survival time in animals challenged intracranially with a malignant glioma. CONCLUSIONS: These findings support continued efforts to refine local delivery systems of IL-12 in an attempt to bring this therapy to clinical trials.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy , Gliosarcoma/therapy , Interleukin-12/genetics , Paracrine Communication/genetics , Animals , Brain/immunology , Brain/pathology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/physiology , Gliosarcoma/immunology , Gliosarcoma/pathology , Immunologic Memory/genetics , Immunotherapy , Interleukin-12/administration & dosage , Male , Neoplasm Transplantation , RNA, Messenger/genetics , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
15.
Neurosurg Focus ; 9(6): e4, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-16817687

ABSTRACT

OBJECT: Local delivery of cytokines has been shown to have a potent antitumor activity against a wide range of malignant brain tumors. In this study, the authors examined the efficacy of treating central nervous system (CNS) tumors by transfecting poorly immunogenic B16/F10 melanoma cells with interleukin (IL)-2, IL-4, or granulocytemacrophage-colony stimulating factor (GM-CSF) gene, and using these cells to deliver the cytokine locally at the site of the CNS tumor. The object was to determine which cytokine would possess the greatest antitumor activity and to further elucidate its mechanism of action. METHODS: The transfected B16/F10 cells were irradiated to prevent replication and injected intracranially into C57BL/6 mice (10 mice per group) along with nonirradiated, nontransfected B16/F10 (wild-type) melanoma cells. Sixty percent of mice treated with IL-2 (p < 0.001 compared with control) and 10% treated with IL-4 (median survival = 31 days, p < 0.001 compared with control) were long term survivors (> 120 days). The median survival for animals treated with GM-CSF was 22 days with no long term survivors (p = 0.01 compared with control). Control animals that received only wild-type cells had a median survival of 18 days (range 15-20 days). Histopathological examination of brains from animals killed at different times showed minimal infiltration of tumor cells in the IL-2 group, moderate infiltration of tumor cells in the IL-4 group, and gross tumor invasion and tissue necrosis in the GM-CSF group. Animals treated with IL-2 showed a strong CD8 T cell-mediated response, whereas IL-4 evoked a prominent eosinophilic infiltrate in the area of the tumor. CONCLUSIONS: High levels of locally expressed IL-2 rather than IL-4 or GM-CSF stimulate a strong immunological cytotoxic antitumor response that leads to significant prolongation of survival in mice challenged with B16/F10 intracranial melanoma tumor cells. Consequently, IL-2 may be a superior candidate for use in paracrine immunotherapy.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Immunotherapy/methods , Melanoma/therapy , Paracrine Communication/immunology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Cell Line, Tumor , Eosinophils/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Survival Rate , Transfection
16.
Cancer Res ; 59(9): 2107-14, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10232596

ABSTRACT

Potent immune responses against malignant brain tumors can be elicited by paracrine intracranial (i.c.) immunotherapy with interleukin (IL)-2. Additionally, i.c. delivery of carmustine via biodegradable polymers has been shown to significantly prolong survival in both animal models and clinical trials. In this study, we show that the combination of paracrine immunotherapy, with nonreplicating genetically engineered tumor cells that produce IL-2, and local delivery of chemotherapy by biodegradable polymers prolongs survival in a synergistic manner in mice challenged intracranially with a lethal murine brain tumor. Animals receiving IL-2-transduced cells and polymers containing 10% 1,3-bis(2-chloroethyl)-1-nitrosourea had significantly improved survival compared with animals receiving IL-2-transduced cells or 10% 1,3-bis(2-chloroethyl)-1-nitrosourea alone. Median survival for the control group was 19 days. Survival in animals receiving IL-2-transduced cells and 1% carboplatin-containing polymers was also significantly improved compared with either therapy alone. Histopathological examination on day 14 of animals receiving combination treatment showed rare degenerating tumor cells. In addition to tissue necrosis surrounding the polymer, a marked inflammatory reaction was observed. In long-term survivors (all animals receiving combination treatment), no tumor was observed and the inflammatory reaction was completely resolved. The brains of animals receiving combination therapy showed both tissue necrosis due to local chemotherapy and strong inflammation due to paracrine immunotherapy. The demonstration of synergy between paracrine IL-2 and local i.c. delivery of antineoplastic drugs is novel and may provide a combined treatment strategy for use against both primary and metastatic i.c. tumors.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Carboplatin/therapeutic use , Carmustine/therapeutic use , Genetic Therapy , Interleukin-2/therapeutic use , Melanoma, Experimental/therapy , Tumor Cells, Cultured/transplantation , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/metabolism , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Biodegradation, Environmental , Brain Neoplasms/drug therapy , Carboplatin/administration & dosage , Carmustine/administration & dosage , Combined Modality Therapy , Decanoic Acids/administration & dosage , Delayed-Action Preparations , Drug Carriers , Drug Implants , Drug Screening Assays, Antitumor , Female , Interleukin-2/genetics , Interleukin-2/metabolism , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Polyesters/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Transfection , Tumor Cells, Cultured/metabolism
17.
Cancer Res ; 59(3): 645-52, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9973213

ABSTRACT

Overexpression of interleukin 4 (IL-4) can impair the tumorigenicity of glioma cells, but direct evidence of its antitumor efficacy after in vivo gene transfer into malignant gliomas has not been provided. To test this, we first injected into the brain of Sprague Dawley rats a 1:1 mixture of C6 rat glioblastoma cells and psi2.L4SN20 or E86.L4SN50 retroviral producer cells (RPCs), secreting 20 and 50 ng of IL-4/5 x 10(5) cells/48 h, respectively. Twenty-seven and 56% of rats receiving injections with these low- or medium-level IL-4 RPCs, respectively, survived tumor injection, whereas control rats died in about 1 month. E86.L4SN50 RPCs coinjected with 9L gliosarcoma cells into syngeneic Fischer 344 rats yielded similar results. A novel IL-4 RPC clone expressing higher levels of IL-4, E86.L4SN200, coinjected with 9L cells increased to 75% the fraction of long-term survivors and induced tumor regression in 50% of rats when injected into established 9L gliosarcomas. Cured rats developed an immunological memory because they rejected a challenge of wild-type 9L cells into the contralateral hemisphere. Magnetic resonance imaging was used to monitor 9L and C6 gliomas and gave direct evidence for tumor rejection in treated rats. Immunohistology showed inflammatory infiltrates in IL-4-treated tumors in which CD8+ T lymphocytes were more abundant, although CD4+ T lymphocytes, B lymphocytes, and macrophages were also present. Overall, these findings suggest that IL-4 gene transfer is a new, promising approach for treating malignant gliomas.


Subject(s)
Brain Neoplasms/therapy , Genetic Therapy/methods , Glioma/therapy , Interleukin-4/genetics , Animals , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , DNA, Complementary/metabolism , Female , Glioma/immunology , Glioma/metabolism , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Retroviridae/genetics
18.
Hum Gene Ther ; 8(11): 1345-53, 1997 Jul 20.
Article in English | MEDLINE | ID: mdl-9295129

ABSTRACT

The growth of U-87 or C6 gliomas co-implanted in nude mice with retroviral producer cells (VPC) expressing the herpes simplex virus-thymidine kinase (HSV-tk) gene is only partially impaired by treatment with ganciclovir (GCV). The effect of GCV is even less evident when C6 and VPC are co-implanted into the rat brain. Furthermore, tumors from C6 cells carrying the HSV-tk gene are not eradicated by GCV, although they remain sensitive to GCV when replated in vitro. These limits of the HSV-tk/GCV system in glioma gene therapy may be due to insufficient gene transfer and/or insufficient delivery of GCV to glioma cells. Combination of HSV-tk and one or more cytokines may improve the antitumor efficacy. Among cytokines, interleukin-4 (IL-4) has already been shown to be active against gliomas. In nude mice, GCV treatment inhibited tumor growth more effectively after co-injection of C6 cells with a mixture of VPC transducing IL-4 and HSV-tk genes than after co-injection with either IL-4 or HSV-tk VPC only. In immunocompetent Sprague-Dawley rats, co-injection of IL-4 VPC and C6 cells was also effective in inhibiting the growth of C6 brain tumors, 38% of the animals surviving for at least 2 months. Furthermore, increased and prolonged antitumor efficacy was obtained by transducing both IL-4 and HSV-tk genes.


Subject(s)
Antimetabolites/pharmacology , Ganciclovir/pharmacology , Genetic Therapy , Glioma/therapy , Interleukin-4/genetics , Simplexvirus/enzymology , Thymidine Kinase/genetics , Viral Proteins/genetics , Animals , Female , Gene Transfer Techniques , Genetic Vectors , Interleukin-4/metabolism , Interleukin-4/therapeutic use , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Retroviridae/genetics , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...