Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Minim Invasive Neurosurg ; 54(3): 132-4, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21863522

ABSTRACT

INTRODUCTION: Neuroendoscopy is a rapidly growing field, but the risk of intraventricular hemorrhage is signficant. There are few novel ways of obtaining hemostasis for major hemorrhage described in the literature. PATIENT: A 5-year-old boy with a history of intraventricular hemorrhage and multi-cystic hydrocephalus presented with worsening brainstem compression from dilation of a 4 (th) ventricle cyst. RESULTS: During endoscopic surgery to fenestrate the cyst, arterial hemorrhage was emergently controlled with the hemostatic agent, FloSeal. CONCLUSION: To the best of the authors' knowledge, this is the first report of a patient with intraoperative intraventricular hemorrhage controlled with FloSeal.


Subject(s)
Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/surgery , Cerebral Ventricles/drug effects , Cerebral Ventricles/surgery , Gelatin Sponge, Absorbable/therapeutic use , Neuroendoscopy/methods , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/physiopathology , Cerebral Ventricles/pathology , Cerebral Ventricles/physiopathology , Child, Preschool , Cysts/etiology , Cysts/physiopathology , Cysts/surgery , Humans , Hydrocephalus/etiology , Hydrocephalus/physiopathology , Hydrocephalus/surgery , Male , Reoperation , Ventriculoperitoneal Shunt/adverse effects
2.
J Neurosurg ; 95(1): 145-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11453388

ABSTRACT

The history of the treatment for hydrocephalus dates back to the Fertile Crescent thousands of years ago. Despite three millennia of management, significant advances in the surgical treatment of the disease have been infrequent. During the 1950s, a milestone occurred at the Children's Hospital of Philadelphia, with the successful development of the first working shunt valve for the treatment of hydrocephalus. In this historical vignette, based on recent interviews with John Holter, D.Sc. (Hon) and Eugene Spitz. M.D., and on a review of the available literature, the authors narrate the exciting story of the development of the Spitz-Holter valve, which took place in Philadelphia during the early 1950s.


Subject(s)
Cerebrospinal Fluid Shunts/history , Hydrocephalus/history , Cerebrospinal Fluid Shunts/instrumentation , Eponyms , Equipment Design/history , History, 20th Century , Humans , Hydrocephalus/therapy , Male , Philadelphia
3.
J Neurosurg ; 91(6): 997-1004, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10584846

ABSTRACT

OBJECT: The goal of this study was to determine whether adenoviral vector-mediated expression of human wildtype p53 can enhance the radiosensitivity of malignant glioma cells that express native wild-type p53. The p53 gene is thought to function abnormally in the majority of malignant gliomas, although it has been demonstrated to be mutated in only approximately 30%. This has led to studies in which adenoviral transduction with wild-type human p53 has been investigated in an attempt to slow tumor cell growth. Recent studies suggest that reconstitution of wild-type p53 can render cells more susceptible to radiation-mediated death, primarily by p53-mediated apoptosis. METHODS: Rat RT2 glioma cells were analyzed for native p53 status by reverse transcriptase-polymerase chain reaction and sequence analysis and for p53 expression by Western blot analysis. Clonogenic survival and the terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assay were used to characterize RT2 cell radiosensitivity and apoptosis, respectively, with and without prior transduction with p53-containing and control adenoviral vectors. Animal survival length was monitored after intracerebral implantation with transduced and nontransduced RT2 cells, with and without cranial radiation. The RT2 cells were demonstrated to express native rat wild-type p53 and to markedly overexpress human p53 following adenoviral p53 transduction. The combination of p53 transduction followed by radiation resulted in marked decreases in RT2 cell survival and increases in apoptosis at radiation doses from 2 to 6 Gy. Animals receiving cranial radiation after intracerebral implantation with RT2 cells previously transduced with p53 survived significantly longer than control animals (p<0.01). CONCLUSIONS: The ability to enhance the radiosensitivity of malignant glioma cells that express wild-type p53 by using adenoviral transduction to induce overexpression of p53 offers hope for this approach as a therapeutic strategy, not only in human gliomas that express mutant p53, but also in those that express wild-type p53.


Subject(s)
Apoptosis/radiation effects , Brain Neoplasms/genetics , Cell Survival/radiation effects , Genetic Therapy , Glioblastoma/genetics , Glioma/genetics , Transduction, Genetic , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/genetics , Adenoviridae/genetics , Animals , Apoptosis/genetics , Brain Neoplasms/radiotherapy , Cell Survival/genetics , Dose-Response Relationship, Radiation , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/radiotherapy , Glioma/radiotherapy , Humans , In Situ Nick-End Labeling , Neoplasm Transplantation , Rats , Rats, Inbred F344
4.
Surg Neurol ; 50(4): 367-75; discussion 375, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9817462

ABSTRACT

BACKGROUND: Direct infusion of therapeutic agents into the brain is a novel technique that has the potential for bypassing the blood-brain barrier and delivering high concentrations of therapeutic agents into the brain parenchyma. We have developed a model to characterize the distribution of Evans Blue (MW 960) and Blue Dextran (MW 2 x 10(6)) in rat brain using a positive pressure infusion system. METHODS: Evans Blue and Blue Dextran were infused in volumes of 20, 40, 60, 100, 140, and 180 microL into the caudate putamen of female Fischer rats over a period of 2 h with rates of infusion varying between 0.167 microL and 1.5 microL/min. During the infusions, the pressure generated in the infusion system and intracranial pressure were measured using a fiberoptic pressure monitoring system. After infusions, the volumes of distribution of the dye molecules were measured from 3-mm thick sections using video microscopy and computer image analysis. Histologic changes during the infusion were studied using snap freezing and hematoxylin/eosin staining of cryosections. RESULTS: Volumes of distribution for Evans Blue were greater than those for Blue Dextran. There was extensive spread of each dye in the ipsilateral hemisphere and also across the corpus callosum to the opposite hemisphere. Infusion/interstitial pressures peaked during the first 5 min of the infusion period, after which pressures dropped to a plateau value that remained relatively constant during the remainder of the infusion. Histologic findings suggest that this phenomenon is an important transition process that is likely to play a role in the pattern of distribution of macromolecules infused by this technique. No marked changes in intracranial pressure were noted during the infusion procedure. CONCLUSIONS: Direct positive pressure infusion into the brain has great potential in the treatment of brain tumors and other central nervous system disorders using both high and low molecular weight compounds (immunotoxins, protein conjugates, pharmacologic agents, oligonucleotides, and viral vectors).


Subject(s)
Brain/metabolism , Coloring Agents/pharmacokinetics , Animals , Blood-Brain Barrier , Coloring Agents/administration & dosage , Female , Male , Pressure , Rats
5.
Neurosurgery ; 41(4): 908-15, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9316053

ABSTRACT

OBJECTIVE: To improve the prognosis for primary malignant tumors of the central nervous system, new therapeutic strategies are needed. Antisense oligodeoxynucleotides (ODNs) offer the potential to block the expression of specific genes within cells. The proto-oncogene c-myc has long been implicated in the control of normal cell growth and its deregulation in the development of neoplasia. We therefore reasoned that a strategy using ODNs complementary to c-myc messenger ribonucleic acid would be a potent inhibitor of glioma cell proliferation. METHODS: A variety of antisense, sense, and scrambled (15-mer) phosphorothioate ODNs targeted to rat and human c-myc messenger ribonucleic acid were synthesized and added to the media of cultured RT-2 cells (a rat glioblastoma cell line). Cell growth was assessed by 3-[4,5-dimethylthiazol-2yl]-2,5-diphenyltetrazolium bromide dye assay 1 to 5 days after adding the ODNs. c-Myc protein expression was analyzed by Western blot analysis. The stability of the ODNs was confirmed by gel electrophoresis. RESULTS: Compared with cultures containing standard media, two of three antisense ODNs significantly inhibited the growth of glioma cells, whereas sense and scrambled sequence ODNs did not significantly affect cell growth at the concentrations tested. A human c-myc antisense sequence, which differed from the rat sequence by one base substitution, also had an inhibitory effect on RT-2 cells. Western blot analysis demonstrated that expression of immunoreactive c-Myc protein was also greatly reduced in the rat antisense ODN-treated cells (and not in sense-, scrambled-, or control-treated cells). The degree of reduction of c-Myc protein expression correlated well with the decrease in cell growth observed with several antisense ODNs. Phosphorothioate ODNs were stable in cell culture media for at least 5 days. CONCLUSION: These results suggest that c-Myc plays a critical role in glioma cell proliferation and demonstrate that antisense ODNs can suppress proto-oncogene expression and inhibit the proliferation of glioma cells. Our results indicate that the antiproliferative activity of these ODNs was mediated predominantly through sequence-specific antisense mechanisms, but that sequence-specific nonantisense effects may also contribute to the strongest effects demonstrated. These findings support a potential role for antisense strategies designed to inhibit c-myc expression in the treatment of malignant gliomas.


Subject(s)
Brain Neoplasms/genetics , Cell Division/drug effects , Glioblastoma/genetics , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Thionucleotides/pharmacology , Tumor Cells, Cultured/drug effects , Animals , Blotting, Western , Brain Neoplasms/pathology , Cell Division/genetics , Cell Line, Transformed , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/pathology , Humans , Proto-Oncogene Mas , Rats , Rats, Inbred F344
6.
Lymphokine Cytokine Res ; 12(1): 45-8, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8096153

ABSTRACT

Peripheral blood lymphocytes cultured with interleukin-2 (IL-2), which are referred to as lymphokine-activated killer (LAK) cells, develop the ability to lyse a wide variety of tumor cells but not normal cells. However, we report here that a cloned human natural killer cell line, NK3.3, was susceptible to lysis by human LAK cells. An IL-4-dependent NK3.3 subclone showed higher sensitivity to LAK-mediated lysis than the conditioned medium-dependent parent clone or the IL-2-dependent NK3.3 subclone. Cytokine-"starved" NK3.3 cells, which were cultured in medium lacking IL-2 or IL-4, exhibited a decreased susceptibility to LAK cell lysis. Periodate treatment of NK3.3 cells, which can induce nonspecific binding between effector cells and target cells through Schiff-base formation, enhanced the sensitivity to LAK, suggesting that adhesion molecules may be involved in this killing mechanism. The "starved" NK3.3 cells expressed lower levels of intracellular adhesion molecule 1 (ICAM-1); however, anti-ICAM-1 antibody could not inhibit the sensitivity of the NK3.3 clone to LAK lysis. These results suggest that ICAM-1 is not the major ligand in NK3.3 sensitivity, although the sensitivity correlates well with the magnitude of ICAM-1 expression.


Subject(s)
Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Natural/immunology , Antibodies , Binding, Competitive , Cell Adhesion Molecules/metabolism , Clone Cells/immunology , Cytotoxicity, Immunologic , Humans , Intercellular Adhesion Molecule-1 , Interleukin-2/immunology , Interleukin-4/immunology , Killer Cells, Natural/metabolism , Oxidation-Reduction
7.
Cancer Res ; 52(5): 1123-8, 1992 Mar 01.
Article in English | MEDLINE | ID: mdl-1737371

ABSTRACT

Interleukin 2 (IL-2) is a potent immunostimulant that causes the release of secondary cytokines and the production of lymphokine-activated killer cells. We investigated the cellular and cytokine responses to injection of recombinant human IL-2 into the human cerebrospinal fluid of 11 patients with metastatic tumors involving the spinal or cerebral leptomeninges. After initial intraventricular IL-2 administration (1.25 x 10(5) to 2 x 10(6) Cetus units/injection), cerebrospinal fluid samples were collected at intervals from 0 to 24 h. Enzyme-linked immunosorbent assay results indicated that IL-2 levels gradually decreased during the first 24 h, with an average t1/2 between 4 and 8 h. Induction of tumor necrosis factor alpha, interleukin 1 beta, interleukin 6, gamma-interferon, and interleukin 2 receptor (p55) was also assessed by enzyme-linked immunosorbent assay. Tumor necrosis factor alpha and interleukin 6 levels peaked at 2 to 4 h and 4 to 6 h, with concentrations between 71 to 1,714 pg/ml and 942 to 10,500 pg/ml, respectively. Interleukin 1 beta, gamma-interferon, and soluble IL-2 receptor peaked later, during 6 to 12 h; the levels achieved were 234 pg/ml, 25 NIH units/ml, and 207 units/ml, respectively. All cytokine concentrations returned to near baseline between 12 and 24 h; however, the soluble IL-2 receptor levels remained elevated. Additional observations included a rapid influx of neutrophilic leukocytes, followed by a prolonged presence of lymphocytes. These data indicate a broad and complex potential of the immune response in the central nervous system, as well as further define the cytokine cascade in response to IL-2 alone.


Subject(s)
Adenocarcinoma/cerebrospinal fluid , Interleukin-1/cerebrospinal fluid , Interleukin-2/pharmacology , Interleukin-6/cerebrospinal fluid , Melanoma/cerebrospinal fluid , Meningeal Neoplasms/cerebrospinal fluid , Receptors, Interleukin-2/metabolism , Tumor Necrosis Factor-alpha/cerebrospinal fluid , Adenocarcinoma/secondary , Adenocarcinoma/therapy , Adult , Female , Humans , Injections, Intraventricular , Interleukin-2/administration & dosage , Interleukin-2/cerebrospinal fluid , Interleukin-2/pharmacokinetics , Killer Cells, Lymphokine-Activated , Leukocyte Count , Lung Neoplasms , Lymphocyte Subsets , Male , Melanoma/secondary , Melanoma/therapy , Meningeal Neoplasms/secondary , Meningeal Neoplasms/therapy , Middle Aged
8.
Cancer Immunol Immunother ; 32(6): 391-9, 1991.
Article in English | MEDLINE | ID: mdl-1848799

ABSTRACT

Outgrowth of tumor-infiltrating lymphocytes (TIL) from the human primary brain tumor glioblastoma multiforme was achieved by OKT3 initiation (10 ng/ml), followed by sustained expansion by interleukin-2 (IL-2; 200 U/ml). Tumor-infiltrating lymphocyte (TIL) initiation by this process was performed in parallel with the standard "IL-2-only" method. Of ten tumors, seven yielded TIL in response to OKT3/IL-2, whereas only three of these seven grew after initiation with IL-2 alone. On the basis of cell doubling times, at least 60 doublings, resulting in (hypothetically) up to 10(23) TIL from as few as 2 x 10(5) cells in tumor suspensions, could be achieved using OKT3/IL-2. OKT3-initiated TIL proliferated in culture for as long as 288 days, although senescence of some cultures occurred at as early as 73 days. Significant heterogeneity of lymphocytes infiltrating the fresh tumors and heterogeneity of resultant TIL phenotype and function were apparent, yet several common trends were noted. In all cases after OKT3 initiation, significant net growth was not apparent until approximately 14 days. In contrast, in the three samples that grew in response to IL-2 alone, log-phase growth was always observed earlier. During the early phase of the cultures, all TIL expressed some killing activity toward a broad spectrum of tumors, including the autologous tumor. No consistent preference of TIL for lysis of autologous tumor was observed. Glioblastoma multiforme TIL cultures contained a mixture of CD8+ and CD4+ cells, with few CD16+ or NKH-1+. Of the six TIL examined in detail for population phenotype in relationship to time in culture, four eventually became exclusively CD4+. Further analysis of these CD4+ TIL indicated that all were of the helper-inducer class, 4B4+ and 2H4-. Concurrent with the decline in CD8+ cells, a decline in the cytolytic activity of these TIL cultures occurred. Furthermore, in two TIL that remained CD8+, a decline in the cytolytic activity also occurred. Therefore, loss of killing activity was not merely a reflection of the major cell phenotype changes. These results indicate that the OKT3/IL-2 process provides an alternative to IL-2 alone for TIL initiation and growth, as well as providing a novel system for further analysis of tumor-derived lymphocyte and accessory cell functional potential.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/drug therapy , Cytotoxicity, Immunologic/drug effects , Glioblastoma/drug therapy , Interleukin-2/pharmacology , Lymphocytes, Tumor-Infiltrating/drug effects , Adult , Aged , Antigens, CD/analysis , Brain Neoplasms/immunology , Cell Division/drug effects , Drug Therapy, Combination , Female , Glioblastoma/immunology , Humans , Immunophenotyping , Male , Middle Aged , Muromonab-CD3 , Time Factors , Tumor Cells, Cultured
9.
J Neurosurg ; 69(3): 403-9, 1988 Sep.
Article in English | MEDLINE | ID: mdl-3261328

ABSTRACT

Short-term stimulation of nonantigen-primed peripheral blood mononuclear leukocytes with interleukin-2 generates a population of oncolytic effectors designated "lymphokine-activated killer" (LAK) cells. These LAK cells express potent lytic activity against a wide spectrum of fresh or cultured autochthonous (patient's own) and allogeneic (unrelated) tumors, yet specifically spare normal tissues. In this study, cells derived from primitive neuroectodermal tumors of the posterior fossa (PNET-PF) were examined for their sensitivity to LAK cytolysis utilizing an in vitro 4-hour chromium-51-release assay. Five early-passage cell lines, derived from primary PNET-PF, demonstrated significant sensitivity to LAK cell cytolysis. Lysis was equally effective in culture medium and cerebrospinal fluid. Three freshly excised PNET-PF exhibited similar susceptibility to lysis by autochthonous LAK cells. Greatly increased expansion of LAK cell cultures could be achieved by short-term stimulation with monoclonal anti-CD3 antibodies in addition to interleukin-2 activation. These findings constitute the preliminary in vitro foundations for potential intrathecal adoptive immunotherapy of PNET-PF with LAK cells.


Subject(s)
Cerebellar Neoplasms/pathology , Interleukin-2/pharmacology , Killer Cells, Natural/drug effects , Leukocytes, Mononuclear/drug effects , Medulloblastoma/pathology , Adult , Cerebellar Neoplasms/immunology , Child , Child, Preschool , Cytotoxicity, Immunologic , Female , Humans , Immunotherapy/methods , In Vitro Techniques , Infant , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Male , Medulloblastoma/immunology
10.
Cancer Res ; 48(8): 2184-92, 1988 Apr 15.
Article in English | MEDLINE | ID: mdl-3258186

ABSTRACT

When peripheral blood lymphocytes (PBL) are incubated with interleukin 2 (IL 2), a novel cytotoxic lymphocyte subpopulation, termed lymphokine activated killers (LAK), arises. LAK are functionally defined as IL 2 responsive cells demonstrating major histocompatibility antigen-unrestricted cell-mediated cytotoxicity against fresh solid tumors and other natural killer cell-resistant and -sensitive tumor targets in the absence of prior antigen priming. Flow cytometric analysis of IL 2 activated PBL using forward and right angle light scatter and fluorescence intensity identified the emergence of a large, optically dense, autofluorescent cell population which paralleled the generation of LAK activity. These unique IL 2 induced lymphocytes have been named giant autofluorescent lymphocytes (GAL). These cells are readily distinguished from the small nonfluorescent lymphocytes (SNL) observed in fresh PBL, unstimulated cultured PBL, and those cells remaining after incubation with IL 2 which have not acquired GAL characteristics. In this investigation, LAK cultures were sorted on days 4, 5, and 6 into GAL and SNL populations and were tested for oncolytic activity against the natural killer-resistant Daudi and RC-1 tumor targets. Against these targets, lymphocytes from non-IL 2 activated PBL or the sorted SNL population expressed less than 2% of the oncolytic activity (measured in lytic units) exhibited by GAL effectors. The SNL and GAL populations were cultured in IL 2 for up to 48 h following the sorting procedure and then reassayed for tumor cytolytic activity. During this culture period, GAL but not SNL continued to express LAK killing against natural killer-resistant tumor targets. Using gamma-irradiation to prevent further cell cycling, it was shown that the functional half-life of the LAK effector was approximately 8.5 h. Therefore, the cytotoxicity expressed by the sorted GAL population after 48 h in culture (equivalent to five functional half-lives) must be expressed by progeny of the originally plated lymphocytes. These results indicate that in addition to the LAK effector, the GAL population contains a self-sustaining, recycling intermediate responsible for generating new LAK. Our data indicate that analysis of IL 2 activated PBL using GAL light-scattering properties has application in phenotyping LAK, monitoring of cellular kinetics, cell sorting, and enrichment of the LAK effector population, and in the clinical monitoring of IL 2 therapy.


Subject(s)
Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Cell Cycle , Cytotoxicity, Immunologic/drug effects , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Killer Cells, Natural/drug effects , Light , Lymphocytes/classification , Lymphocytes/immunology , Lymphokines/pharmacology , Phenotype , Scattering, Radiation
11.
Cell Immunol ; 111(1): 235-46, 1988 Jan.
Article in English | MEDLINE | ID: mdl-3123071

ABSTRACT

Human lymphocytes respond to IL-2 with the generation of MHC-unrestricted oncolytic activity. This function has been named lymphokine-activated killing (LAK). To investigate the mechanism by which IL-2 activates and maintains LAK, we have examined the role(s) of IL-2 cell surface receptors. Removal or blockade of unstimulated lymphocytes expressing the IL-2 receptor Tac does not preclude the acquisition of LAK function. Therefore, a non-Tac IL-2 receptor was proposed to be involved in LAK generation. Using direct 125I-IL-2 binding to Tac-negative LAK precursors suggested the existence of such an alternate IL-2 receptor. Chemical crosslinking of 125I-IL-2 to Tac-depleted lymphocytes followed by SDS-PAGE determined that the size of the non-Tac-binding protein was approximately 75 kDa. Tac-negative lymphocytes activated by a limited IL-2 pulse which was insufficient for detectable Tac upregulation indicated that an initial non-Tac pathway was involved in functional differentiation. The development of lytic function, Tac upregulation, and cellular proliferation was prohibited by trypsin, a treatment shown also to eliminate 125I-IL-2 binding to Tac-negative lymphocytes. The Tac antigen, although not involved in the initial generation of LAK, is involved in the proliferative maintenance of this lytic function.


Subject(s)
Cytotoxicity, Immunologic , Interleukin-2/pharmacology , Killer Cells, Natural/immunology , Lymphocyte Activation , Receptors, Immunologic/physiology , Cell Differentiation , Cell Division , Humans , Interleukin-2/metabolism , Killer Cells, Natural/cytology , Receptors, Interleukin-2 , Trypsin
SELECTION OF CITATIONS
SEARCH DETAIL
...