Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
Clin Neuropathol ; 23(1): 34-43, 2004.
Article in English | MEDLINE | ID: mdl-14986932

ABSTRACT

OBJECTIVE: Extracranial subcutaneous masses involving the scalp and/or skull in young children are uncommon lesions that get excised by the neurosurgeon. Although the most common reported lesion is the dermoid cyst, our experience suggests that the spectrum of pathology in these lesions can present diagnostic challenges to the pathologist. MATERIAL: We reviewed 30 consecutive extracranial masses from 29 patients between July 1998 and June 2003. METHOD: Hematoxylin and eosin-stained sections were reviewed in all cases, and immunohistochemistry was performed in select cases. RESULTS: Twenty-three were within the scalp, 5 involved the scalp and skull and 2 were within the limits of the inner and outer tables of the skull. There were 8 dermoid cysts, 2 epidermoid cysts, 6 post-traumatic lesions including 3 calcified cephalhematomas and 3 pseudocysts, 5 vascular lesions including 3 capillary hemangiomas, 1 venous angioma and 1 lymphangioma, 2 cases of cranial fasciitis and 1 case each of benign teratoma, deep granuloma annulare, benign fibrous histiocytoma, congenital melanocytic nevus, hamartoma with ectopic meningothelial elements, cutaneous hyalinised ectopic meningioma and a meningocele with a fibrohistiocytic reaction. No lesions have recurred or exhibited malignant features. CONCLUSIONS: Surgical pathologists and neuropathologists should be aware that the differential diagnosis of "lumps and bumps on babie's heads" is quite varied and can be histologically challenging.


Subject(s)
Hamartoma/pathology , Head and Neck Neoplasms/pathology , Scalp/pathology , Skin Neoplasms/pathology , Skull Neoplasms/pathology , Skull/pathology , Child, Preschool , Craniocerebral Trauma/pathology , Diagnosis, Differential , Female , Head and Neck Neoplasms/congenital , Humans , Infant , Infant, Newborn , Male , Retrospective Studies , Scalp/injuries , Skin Neoplasms/congenital , Skull/injuries , Skull Neoplasms/congenital
3.
J Pediatr Surg ; 36(9): 1381-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11528610

ABSTRACT

BACKGROUND/PURPOSE: Diagnosis and management of the acute abdomen in patients with spina bifida (SB) can be problematic. There are at least 4 clinical factors that can predispose to the development of acute abdominal symptoms and signs, and patients with a thoracic level lesion can have a partially insensate abdomen. The authors analyzed their accumulated experience to determine the annual incidence of acute abdominal signs and symptoms in children and young adults with spina bifida, the differential diagnosis, the operative management, and the outcome. The pertinent literature was reviewed. METHODS: Cases were ascertained during a 10-year period at 1 institution and reviewed retrospectively. RESULTS: Twenty-two episodes of acute abdominal symptoms and signs in 19 children and young adults with SB were ascertained over 10 years at 1 institution, for an annual incidence of 0.74%. More patients had a thoracic level lesion (n = 12; 60%) than in the clinic population as a whole (27%; P =.04), but the gender distribution was similar (58% girls), as was the prevalence of ventriculoperitoneal shunts (VPS; 95%). The median age was 13 years (range, 1 year to 26 years). Hospitalization was necessary for 19 (86%) of the 22 episodes. The duration of symptoms before diagnosis was a median of 3 days (range, 1 to 14 days). Most patients (82%) presented with abdominal pain. Fever was present in 27%, shock in 23%, and peritoneal signs in 23%. There were 14 different final diagnoses, 10 (71%) of which were associated with a predisposing factor. Of the 22 episodes, 18 (82%) could be attributed to an underlying factor: (1) neurogenic bladder (9; 41%); (2) neurogenic bowel (3; 14%); (3) VPS (4; 18%); (4) complications from previous surgery (2; 9%). Thirteen patients (59%) underwent a total of 20 surgical procedures of 12 different kinds. Despite awareness of the complexities involved, 3 patients (14%) died: 1 from complications resulting from bladder perforation; 1 from urosepsis and shock; and 1 from peritonitis caused by VPS infection. CONCLUSION: The differential diagnosis of the acute abdomen in patients with SB is broad, conditions requiring surgery are frequently diagnosed, and the mortality rate is substantial, despite aggressive management.


Subject(s)
Abdomen, Acute/diagnosis , Abdomen, Acute/epidemiology , Spinal Dysraphism/epidemiology , Adolescent , Adult , Age Distribution , Child , Child, Preschool , Comorbidity , Female , Humans , Incidence , Male , Prognosis , Risk Assessment , Risk Factors , Sex Distribution , Spinal Dysraphism/diagnosis , Spinal Dysraphism/surgery
4.
Histopathology ; 34(2): 154-62, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10064395

ABSTRACT

AIMS: We have reviewed immunohistochemically 17 paediatric medulloblastomas in order to determine if correlations exist that might be useful in subclassifying these tumours. METHODS AND RESULTS: The patient group included 11 children who had died (mean survival 13 months) and six still alive (followed for up to 10 years). Ten tumours were diffuse and six were nodular (one biopsy had only perivascular tumour). Of the 10 diffuse tumours, three were desmoplastic: of the six nodular tumours, all six were desmoplastic. All 17 tumours were synaptophysin-reactive: three nodular tumours were glial fibrillary acidic protein (GFAP)-reactive in the nodules (two of three S 100-reactive tumours were also GFAP-reactive). MIB-1 labelling indices (LI) ranged from 5 to 80%. Six tumours exhibited at least 1% LI against Tp53 (Mab D07 and/or Mab 1801). Eight cases were 100% bcl2-reactive with nine cases having an LI <80% ('low labelling'). All nine 'low labelling' bcl2 cases were TP53 non-reactive; all six Tp53-reactive cases were bcl2 100% reactive. Six of 10 patients with diffuse medulloblastomas survived 18 months or less while four of 10 are alive up to 10 years. In contrast, five of six patients with nodular neoplasms died within 48 months of diagnosis with one patient followed up for less than 1 year. CONCLUSIONS: Immunohistochemistry is a useful adjunct in characterizing subsets of paediatric medulloblastomas and confirms that larger co-operative studies may be fruitful in identifying a prognostic utility of a combined histochemical/immunohistochemical analysis on these tumours.


Subject(s)
Biomarkers, Tumor/metabolism , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Adolescent , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Loss of Heterozygosity , Male , Medulloblastoma/genetics , Medulloblastoma/mortality , Medulloblastoma/pathology , Reticulin/metabolism
5.
J Neurooncol ; 44(3): 233-41, 1999.
Article in English | MEDLINE | ID: mdl-10720203

ABSTRACT

The current study was designed to evaluate the toxicity and activity of Spartaject Busulfan, a microcrystalline preparation of busulfan, following its intrathecal administration into a nude rat model of human neoplastic meningitis. Animals were treated through permanent indwelling subarachnoid catheters. Human glioma D-456 MG growing in the subarachnoid space was treated with 8.1 micromol of intrathecal Spartaject Busulfan. Single-dose therapy was also subsequently compared with 4 doses of 8.1 and 2.0 micromol busulfan, respectively, against D-456 MG neoplastic meningitis. Additional experiments evaluated a saline control versus 8.1 micromol x 1, 6.2 micromol x 4 and 4.1 micromol x 4, respectively, against D-456 MG. A single dose of 8.1 micromol of intrathecal Spartaject Busulfan resulted in an increase in median survival of 61.7% compared with the saline control. In experiment 2, all busulfan treatments showed increases in median survival of 142.8% (8.1 micromol x 1), 52.3% (2.0 micromol x 4), and 23% (8.1 micromol x 4) (p < 0.001 for all groups) compared with the saline control. These results suggest that a narrow therapeutic dose range for both toxicity and activity has been defined for intrathecal busulfan in the treatment of human neoplastic meningitis in athymic nude rats. Although busulfan has only limited activity against solid tumors, the high doses achievable in the CSF following intrathecal administration coupled with the steep dose-response relationships of alkylating agents, provide rationale for further evaluation of this agent.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Glioblastoma/complications , Glioblastoma/drug therapy , Meningeal Neoplasms/complications , Meningeal Neoplasms/drug therapy , Meningitis/etiology , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Busulfan/administration & dosage , Busulfan/adverse effects , Drug Administration Schedule , Female , Injections, Spinal , Rats , Rats, Nude , Subarachnoid Space , Survival Analysis
6.
Clin Neuropathol ; 17(1): 55-60, 1998.
Article in English | MEDLINE | ID: mdl-9496542

ABSTRACT

Twenty-nine months after surgery, irradiation, and systemic chemotherapy for a pineal mixed germ cell tumor, an 11-year-old Caucasian male developed a 3 cm dural based nodule in the occipital lobe that proved to be a solitary fibrous tumor by immunohistochemical and ultrastructural examination. Differential diagnosis included fibrous meningioma, neurofibroma, Schwannoma, cranial fasciitis of infancy, and solitary fibrous tumor. A Masson trichrome stain revealed a prominent collagenous stroma and reticulin staining exhibited strong pericellular positivity. Immunohistochemical staining demonstrated diffuse vimentin and focal CD34 positivity of tumor cells. Ultrastructural examination revealed fibroblastic differentiation. These features are consistent with solitary fibrous tumor. Although we favor a radiation-induced origin for the neoplasm, alternative explanations for the tumor's origin include cerebrospinal fluid spread from the original germ cell tumor or a de novo neoplasm.


Subject(s)
Antineoplastic Agents/therapeutic use , Endocrine Gland Neoplasms/therapy , Germinoma/therapy , Meningeal Neoplasms/etiology , Pineal Gland , Child , Combined Modality Therapy , Diagnosis, Differential , Endocrine Gland Neoplasms/radiotherapy , Germinoma/radiotherapy , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male
7.
Med Pediatr Oncol ; 30(2): 75-80, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9403013

ABSTRACT

Seventeen patients less than or equal to 20 years of age with newly diagnosed (n = 10) or recurrent (n = 7) malignant gliomas (anaplastic astrocytoma and glioblastoma multiforme) were treated with cyclophosphamide in association with hematopoietic cytokines (GM-CSF or G-CSF). Cyclophosphamide was given at a dose of 2 g/m2 daily for 2 days at 4-week intervals. Toxicity consisted of grade IV neutropenia and thrombocytopenia in 95% and 48% of cycles, respectively. There were no cyclophosphamide-related cardiac, pulmonary, or urothelial toxicities observed. Four of 10 patients with newly diagnosed disease demonstrated responses (three complete and one partial responses; one CR was only of 2 months duration). None of the seven patients with recurrent tumors demonstrated a response. We conclude that high-dose cyclophosphamide warrants further evaluation in children with newly diagnosed malignant glioma.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Cyclophosphamide/therapeutic use , Glioblastoma/drug therapy , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male
8.
Neurosurgery ; 41(6): 1365-72; discussion 1372-3, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9402588

ABSTRACT

OBJECTIVE: The promise of immunotherapies developed against brain tumors in animal models has not been realized in human clinical trials. This may be because of the routine use of rodent tumors artificially induced by chemicals or viruses that do not accurately portray the intrinsic qualities of spontaneously arising human tumors and that often fail to incorporate the role of immunosuppressants, such as transforming growth factor-beta, that are secreted by human gliomas. From an astrocytoma that arose spontaneously in inbred VM/Dk mice, we have characterized a highly tumorigenic spontaneous murine astrocytoma cell line (SMA-560) that retains features of glial differentiation and naturally produces high levels of biologically active transforming growth factor-beta. We have used this model to determine whether cytokine production by tumor cells will inhibit intracerebral astrocytoma growth. METHODS: Packaging cell lines producing replication-incompetent retroviral vectors were used to transfect the SMA-560 cell line in vitro with the genes encoding the murine cytokines interleukin (IL)-2, IL-3, IL-4, IL-6, tumor necrosis factor-alpha, gamma-interferon, or granulocyte-macrophage colony-stimulating factor or the costimulatory molecule B7.1 (CD80). RESULTS: Mice challenged intracerebrally with 5000 untransfected SMA-560 cells all succumbed to tumor within 30 days, with a median survival of 25 days. In contrast, mice challenged with SMA-560 cells producing IL-2, IL-4, or tumor necrosis factor-alpha each had a more than 400% increase in median survival (P < 0.0001). In these groups, 78.3% (18 of 23 mice), 66.7% (10 of 15 mice), and 60% (6 of 10 mice) of the mice, respectively, remained alive without evidence of tumor for longer than 100 days after the initial tumor challenge. All other cytokines tested and the expression of B7.1 failed to result in an increase in median survival. CONCLUSION: Using a spontaneous astrocytoma model in an inbred mouse strain, we have shown that cytokine production by glial tumors can abrogate their tumorigenicity in vivo despite production of transforming growth factor-beta. These results predict that approaches directed at cytokine production within intracerebral astrocytomas may be efficacious in human trials and that the "immunological privilege" of the brain may not be absolute under such conditions.


Subject(s)
Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cytokines/metabolism , Animals , Carcinogenicity Tests , Female , Genetic Vectors , Histocompatibility Antigens Class I/analysis , Immunohistochemistry , Mice , Mice, Inbred Strains , Retroviridae/genetics , Transfection , Transforming Growth Factor beta/biosynthesis , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 93(19): 10399-404, 1996 Sep 17.
Article in English | MEDLINE | ID: mdl-8816812

ABSTRACT

Vaccination with cytokine-producing tumor cells generates potent immune responses against tumors outside the central nervous system (CNS). The CNS, however, is a barrier to allograft and xenograft rejection, and established tumors within the CNS have failed to respond to other forms of systemic immunotherapy. To determine what barriers the "immunologically privileged" CNS would pose to cytokine-assisted tumor vaccines and what cytokines would be most efficacious against tumors within the CNS, we irradiated B16 murine melanoma cells producing murine interleukin 2 (IL-2), IL-3, IL-4, IL-6, gamma-interferon, or granulocyte-macrophage colony stimulating factor (GM-CSF) and used these cells as subcutaneous vaccines against tumors within the brain. Under conditions where untransfected B16 cells had no effect, cells producing IL-3, IL-6, or GM-CSF increased the survival of mice challenged with viable B16 cells in the brain. Vaccination with B16 cells producing IL-4 or gamma-interferon had no effect, and vaccination with B16 cells producing IL-2 decreased survival time. GM-CSF-producing vaccines were also able to increase survival in mice with pre-established tumors. The response elicited by GM-CSF-producing vaccines was found to be specific to tumor type and to be abrogated by depletion of CD8+ cells. Unlike the immunity generated against subcutaneous tumors by GM-CSF, however, the effector responses generated against tumors in the CNS were not dependent on CD4+ cells. These data suggest that cytokine-producing tumor cells are very potent stimulators of immunity against tumors within the CNS, but effector responses in the CNS may be different from those obtained against subcutaneous tumors.


Subject(s)
Brain Neoplasms/immunology , Cancer Vaccines , Cytokines/biosynthesis , Melanoma, Experimental/immunology , T-Lymphocytes/immunology , Animals , Brain Neoplasms/prevention & control , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cell Line , Cytotoxicity, Immunologic , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Injections, Subcutaneous , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-3/biosynthesis , Interleukin-4/biosynthesis , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Recombinant Proteins/biosynthesis , Spleen/immunology , Transfection
10.
Genes Chromosomes Cancer ; 15(1): 54-63, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8824726

ABSTRACT

Loss of heterozygosity at specific chromosomal locations has been taken as evidence of a tumor suppressor gene located in that area. We performed a genomic allelotyping study on 46 childhood brain tumors of different histopathological types in order to identify and confirm common areas of deletion in different tumor types. Two hundred microsatellite DNA probes equally distributed over the 22 autosomes were applied, covering the genome in steps of approximately 25 cM. Our results confirm frequent loss of heterozygosity of chromosome arms 9q, 10q, 11p, 11q, 16q, and 22q in high-grade gliomas, medulloblastomas, and ependymomas. In addition, we found a new region of loss on chromosome segment 2p21-23 affected predominantly in high-grade gliomas and medulloblastomas.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , DNA, Satellite , Adolescent , Adult , Child , Child, Preschool , Female , Heterozygote , Humans , Infant , Male , Microsatellite Repeats , Polymerase Chain Reaction
11.
Clin Cancer Res ; 1(12): 1545-55, 1995 Dec.
Article in English | MEDLINE | ID: mdl-9815955

ABSTRACT

LMB-1 (B3-LysPE38) is an immunotoxin composed of the tumor-reactive monoclonal antibody B3 and a genetically engineered form of Pseudomonas exotoxin. Monoclonal antibody B3 reacts with a carbohydrate epitope that is found on a number of solid tumors (e.g., breast, ovarian, and lung carcinomas) that frequently invade the intrathecal space, causing neoplastic meningitis. The Pseudomonas exotoxin has been engineered to remove the binding domain to eliminate nonspecific binding. A model of human neoplastic meningitis using rats bearing the human epidermoid carcinoma A431 was used for therapeutic studies of immunotoxin LMB-1. Therapy was initiated 3 days after injection of the tumor cells, which was one third of the median survival time of untreated rats. A single intrathecal injection of 40 microgram increased median survival from 9 days with saline injection to 16 days (78%, P < 0.001), and a single dose of 200 microgram increased median survival to 25 days (188%, P < 0. 001). Three doses of 40 or 200 microgram given on days 3, 6, and 8 significantly increased the median survival of 9.5 days associated with saline injection to 40.5 days (326% increase) and 33.0 days (247% increase), respectively, with two long-term survivors (191-day survival) in each treatment group. LMB-1 had no therapeutic effect on the treatment of two B3 antigen-negative neoplastic meningitis models. Treatment of the antigen-positive A431 neoplastic meningitis with B3 alone or a nonspecific monoclonal, MOPC, coupled to the engineered Pseudomonas exotoxin produced no survival effects. Nontumor-bearing athymic rats showed no toxicity with a single dose of either 40 microgram or 200 microgram, or 3 doses of 40 microgram. However, when they were given three doses of 200 microgram, these rats showed weight loss and loss of neurological function, and two of eight animals died. These studies indicate that, in the range of the most therapeutically effective dosage, the immunotoxin LMB-1 is tolerated in the intrathecal space and should be considered for human intrathecal trials.


Subject(s)
ADP Ribose Transferases , Antibodies, Monoclonal/administration & dosage , Bacterial Toxins , Carcinoma/therapy , Exotoxins/administration & dosage , Immunotoxins/administration & dosage , Lewis Blood Group Antigens/immunology , Meningeal Neoplasms/therapy , Virulence Factors , Animals , Carcinoma/immunology , Carcinoma/pathology , Disease Models, Animal , Drug Screening Assays, Antitumor , Humans , Meningeal Neoplasms/immunology , Meningeal Neoplasms/pathology , Rats , Rats, Nude , Tumor Cells, Cultured , Pseudomonas aeruginosa Exotoxin A
12.
Cancer Res ; 55(17): 3803-9, 1995 Sep 01.
Article in English | MEDLINE | ID: mdl-7641197

ABSTRACT

Malignant gliomas will affect 15,000-17,000 Americans each year and carry a dismal prognosis. Adjuvant chemotherapy is hampered by inadequate drug delivery, systemic toxicity, and a markedly variable biological sensitivity. Intraarterial (i.a.) therapy may enhance selectivity by improving tumor drug delivery and reducing systemic toxicity. Using melphalan given i.a., we studied the therapy of intracranial human glioma xenografts in male athymic nude rats (mean weight, 300 g) which were inoculated intracerebrally with D-54 MG and D-456 MG. On Days 6 and 7 (D-54 MG) or Days 9 and 10 (D-456 MG), rats randomized by body weight and treated with single-dose melphalan given i.a. at 0.5 or 0.75 mg produced significantly higher median survival (D-54 MG, Days 33 and 32; D-456 MG, Days 52 and 54, respectively) compared with i.a. saline (D-54 MG, Day 14, P < 0.001; D-456 MG, Day 24, P = 0.000) or melphalan given i.v. at 0.75 mg and 0.9 mg (D-54 MG only; Day 19, P < 0.001; Day 23, P < 0.001, respectively) and at 0.5 and 0.75 mg (D-456 MG only; Day 26 for both doses, P = 0.00). Although a dose-dependent increase in median survival (D-54 MG, 0.25 mg, Day 18; 0.5 mg, Day 28.5; 0.75 mg, Day 32.5) was observed with i.a. administered melphalan, no significant difference was apparent between 0.5 and 0.75 mg in either tumor model (D-54 MG, P = 0.15; D-456 MG, P = 0.37). Toxicity studies in nontumor-bearing athymic rats yielded a maximum tolerated dose of 0.8 mg for i.a. administered melphalan. This dosage was superior in spite of different xenograft permeabilities (apparent mean blood-to-tissue transport [K] values for alpha-aminoisobutyric acid, 5.8 for D-54 MG and 1.3 for D-456 MG). Pharmacokinetic experiments demonstrated a significant first pass advantage for i.a. (versus i.v.) melphalan. The short plasma half-life, marked antiglioma activity, and lack of requirement for metabolic activation indicate that i.a. melphalan holds considerable promise for human glioma therapy.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Melphalan/administration & dosage , Animals , Brain/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Glioma/metabolism , Glioma/mortality , Humans , Injections, Intra-Arterial , Male , Melphalan/pharmacokinetics , Rats , Rats, Nude , Transplantation, Heterologous
13.
Oncogene ; 10(11): 2243-6, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7784070

ABSTRACT

The high incidence of loss of chromosome 10 alleles in glioblastoma multiforme suggests the presence on this chromosome of a tumor suppressor gene that is important in glioma tumorigenesis and progression. Our initial deletion mapping studies using restriction fragment length polymorphism markers indicated a common deletion region in 10q24-qter. In an attempt to localize the deleted region further, we screened a panel of 117 gliomas for loss of heterozygosity for chromosome 10 loci using 10 microsatellite markers. Seventeen tumors showed partial loss of a copy of chromosome 10 and were further analysed using 28 additional microsatellite markers. Of these, 10 had terminal deletion in the q arm, three had deletions in both p and q arms, two contained interstitial deletion in 10q and two carried deletions in 10p. In the 15 tumors with deletions in 10q, the minimal overlapping deletion region was in distal 10q between markers D10S587 and D10S216. Loci D10S587 and D10S216 are approximately mapped to a 5 cM region in 10q25.1.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 10 , Glioma/genetics , Heterozygote , Humans , Tumor Cells, Cultured
14.
Proc Natl Acad Sci U S A ; 92(7): 2765-9, 1995 Mar 28.
Article in English | MEDLINE | ID: mdl-7708720

ABSTRACT

LMB-7 [B3(Fv)-PE38] is a single-chain immunotoxin constructed from the murine monoclonal antibody B3 and a truncated from of Pseudomonas exotoxin PE38. Antibody B3 recognizes a carbohydrate epitope found on solid tumors that frequently invade the intrathecal space and cause neoplastic meningitis. We tested the therapeutic value of intrathecally administered LMB-7 by using a model of human neoplastic meningitis in athymic rats. This model is representative of a clinical situation in that antibody B3 cross-reacts with a number of normal tissues that can be used to monitor potential systemic toxicity. Treatment was begun 3 days after A431 tumor implantation. Without treatment, the animals median survival was 10 days. Intrathecal administration of 10 micrograms of LMB-7 in 40 microliters on days 3, 5, and 7 produced 4 of 10 and 8 of 10 long-term survivors (> 170 days) in two experiments. Of the long-term survivors, 2 of 4 and 7 of 8 survivors had no microscopic evidence of tumor and were considered histologic cures. Lack of significant toxicity in the effective dose range and specificity make LMB-7 an excellent candidate for intrathecal treatment of neoplastic meningitis in humans.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Immunotoxins/therapeutic use , Meningitis/drug therapy , Animals , Antibodies, Monoclonal , Exotoxins , Humans , Immunotoxins/administration & dosage , Immunotoxins/toxicity , Injections, Spinal , Meningitis/etiology , Mice , Rats , Rats, Nude , Time Factors , Transplantation, Heterologous , Tumor Cells, Cultured
15.
J Comput Assist Tomogr ; 19(1): 153-5, 1995.
Article in English | MEDLINE | ID: mdl-7822537

ABSTRACT

We report development of an intracranial epidermoid cyst 2 years after a depressed skull fracture. The epidermoid cyst is presumed to be the result of introduction of epidermal elements at the time of trauma. Post-traumatic intracranial epidermoid cysts are rare and appear to be less common than those occurring in the spine.


Subject(s)
Brain Diseases/diagnostic imaging , Brain Diseases/etiology , Epidermal Cyst/diagnostic imaging , Epidermal Cyst/etiology , Skull Fractures/complications , Adolescent , Humans , Intracranial Aneurysm/diagnostic imaging , Intracranial Aneurysm/etiology , Male , Time Factors , Tomography, X-Ray Computed
16.
J Neurooncol ; 24(1): 39-45, 1995.
Article in English | MEDLINE | ID: mdl-8523074

ABSTRACT

DNA derived from medulloblastoma biopsies was analyzed to determine if deletions of the 17p region, mutations of the TP53 gene, or amplification of the c-myc, N-myc, EGFR (epidermal growth factor receptor), or MDM2 (murine double-minute-2) genes was indicative of a poor prognosis. Loss of heterozygosity for 17p, observed in 8/28 (29%) paired samples, was associated with a shortened survival period (p = 0.045 by the logrank test). TP53 mutations occurred in 2/46 (4.3%) tumor samples. c-myc Amplification was seen in 3/43 (6.9%) cases, while none of the tumors contained amplified N-myc, EGFR, or MDM2 genes. These results demonstrate that, while only rare medulloblastomas contain TP53 gene mutations or amplification of the c-myc gene, loss of heterozygosity on chromosome 17p is indicative of a significantly worse prognosis among patients with these tumors. Further, these results provide a strong impetus for a prospective analysis of loss of heterozygosity in a cooperative group setting, which would include tumor staging, a selection of treatment modalities, and multivariate analyses.


Subject(s)
Cerebellar Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17 , Heterozygote , Medulloblastoma/genetics , Adolescent , Adult , Case-Control Studies , Cerebellar Neoplasms/mortality , Child , Child, Preschool , DNA Probes , ErbB Receptors/genetics , Female , Genes, Tumor Suppressor , Genes, myc , Humans , Infant , Male , Medulloblastoma/mortality , Microsatellite Repeats , Point Mutation , Polymerase Chain Reaction , Prognosis
17.
Cancer Res ; 54(17): 4710-4, 1994 Sep 01.
Article in English | MEDLINE | ID: mdl-8062269

ABSTRACT

We report the activity and toxicity of intrathecal melphalan in the treatment of human neoplastic meningitis in the subarachnoid space of athymic nude rats. Animals received injections via chronic indwelling subarachnoid catheters with 5 x 10(5) or 5 x 10(6) TE-671 human rhabdomyosarcoma cells or 5 x 10(6) D-54 MG human glioma cells and were treated with melphalan on days 8, 5, or 5, respectively. Melphalan toxicity in nontumor-bearing rats was assessed at single doses of a 2.0, 3.0, 4.0, or 5.0 mM solution, with clinical and histological evidence of neurotoxicity observed at the 4.0 and 5.0 mM levels. Multiple-dose toxicity studies using a dosing schedule of twice a week for two weeks with a 0.25, 0.5, 0.75, 1.0, 1.5, or 2 mM solution revealed dose-dependent clinical and histological evidence for toxicity at all dosages. Treatment of TE-671 with a single dose of 2.0 mM intrathecal melphalan produced an increase in median survival of 442% compared with saline controls (P < 0.003). Comparison of a single dose of 1.0 or 2.0 mM melphalan with a multiple dose regimen at 0.25 or 0.5 mM melphalan in the treatment of TE-671 revealed increases in median survival of 50% for 1.0 mM, 57% for 2.0 mM, 79% for 0.5 mM, and 111% for 0.25 mM concentrations. Comparison of a single dose of 1 mM melphalan with multiple doses of 0.25 mM melphalan in the treatment of D-54 MG revealed an increase in median survival of 475+% for each of the regimens. Intrathecal melphalan may be an important new addition in the treatment of neoplastic meningitis and is currently being evaluated clinically in a Phase 1 trial.


Subject(s)
Brain Neoplasms/drug therapy , Glioma/drug therapy , Melphalan/administration & dosage , Meningitis/drug therapy , Rhabdomyosarcoma/drug therapy , Animals , Brain Neoplasms/mortality , Demyelinating Diseases/chemically induced , Disease Models, Animal , Drug Screening Assays, Antitumor , Female , Glioma/mortality , Humans , Injections, Spinal , Melphalan/adverse effects , Meningitis/etiology , Meningitis/mortality , Rats , Rats, Nude , Rhabdomyosarcoma/mortality , Subarachnoid Space , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Surg Neurol ; 40(4): 278-83, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8211637

ABSTRACT

Supratentorial embryonal neuroepithelial tumors are undifferentiated neoplasms. We have used this term in preference to the controversial classification primitive neuroectodermal tumors (PNET). These lesions in children are malignant neoplasms which are usually fatal within 2 years of diagnosis in spite of therapy with surgery, radiotherapy, and chemotherapy. We have adopted an aggressive approach to the treatment of these tumors with surgical resection, hyperfractionated craniospinal irradiation of 30.6-43.9 Gy followed by a tumor boost to a total dose of 50-63.7 Gy, and adjuvant chemotherapy with cyclophosphamide, vincristine, and cis-platinum. We have treated five children, aged 4-18 years, with this approach. In contrast to the results reported in the literature, four children are alive without evidence of tumor from 4.3 to 8.0 years following diagnosis. One has suffered a tumor relapse at 2.3 years following diagnosis but remains alive. The basis of our therapeutic strategy for childhood supratentorial embryonal neuroepithelial tumors and the implications of our clinical results are discussed.


Subject(s)
Cerebellar Neoplasms/radiotherapy , Cerebellar Neoplasms/surgery , Chemotherapy, Adjuvant , Neoplasms, Germ Cell and Embryonal/radiotherapy , Neoplasms, Germ Cell and Embryonal/surgery , Adolescent , Brain/radiation effects , Cerebellar Neoplasms/drug therapy , Child , Child, Preschool , Combined Modality Therapy , Female , Humans , Male , Neoplasms, Germ Cell and Embryonal/drug therapy , Spinal Cord/radiation effects , Tomography, X-Ray Computed
19.
Cancer Res ; 53(10 Suppl): 2338-43, 1993 May 15.
Article in English | MEDLINE | ID: mdl-8485721

ABSTRACT

The addition of chemotherapy, notably using nitrosoureas, in the treatment of patients with glioblastoma multiforme has resulted in only modest improvements in long-term patient survival over the use of surgical intervention and irradiation alone. Intraarterial (i.a.) chemotherapy offers the potential benefit of increasing tumor drug delivery because of first-pass drug uptake, while minimizing systemic drug levels and toxicity. We have now investigated the i.a. therapy of intracerebral human glioma xenografts in athymic rats with 4-hydroperoxycyclophosphamide (4-HC), a preactivated derivative of cyclophosphamide. Athymic male rats were given intracerebral injections of the human glioma line D-54 MG. On Day 5 after injection, the rats were randomized (n = 8-10) by body weight (mean weight, approximately 300 g). In one set of experiments, each group received either i.v. saline, i.a. saline, 6 mg i.a. 4-HC, 6 mg i.v. 4-HC (6 mg), or 12 mg i.v. 4-HC. Intraarterial 4-HC produced significant increases in median survival (Day 24) compared with i.a. saline controls (140% increase), equivalent doses given i.v. (71% increase), and twice the equivalent dose given i.v. (50% increase) (by Wilcoxon rank sum analysis, P < 0.05 is statistically significant). The i.a. maximum tolerated dose was subsequently determined to be approximately 12.5 mg in non-tumor-bearing rats. Further experiments demonstrated a dose-response increase in survival for i.a. dosages of 6, 9, and 12.5 mg with significant improvement when compared with saline controls and 12.5 mg i.v. Pharmacokinetic experiments also demonstrated a significant first-pass uptake advantage for i.a. (versus i.v.) administered 4-HC. The short plasma half-life and marked antiglioma activity of 4-HC, without the need for hepatic activation, suggest a therapeutic application of this drug in the i.a. treatment of brain tumors.


Subject(s)
Brain Neoplasms/drug therapy , Cyclophosphamide/analogs & derivatives , Glioma/drug therapy , Animals , Body Weight/drug effects , Brain Neoplasms/metabolism , Cyclophosphamide/pharmacokinetics , Cyclophosphamide/pharmacology , Cyclophosphamide/toxicity , Dose-Response Relationship, Drug , Glioma/metabolism , Humans , Injections, Intra-Arterial , Injections, Intravenous , Male , Mice , Mice, Nude , Rats , Rats, Nude , Transplantation, Heterologous
20.
Cancer Res ; 50(6): 1954-9, 1990 Mar 15.
Article in English | MEDLINE | ID: mdl-2306744

ABSTRACT

Neoplastic meningitis can result from leptomeningeal dissemination of a variety of cancers. We now report the development of animal models of human neoplastic meningitis and activity of intrathecal 4-hydroperoxycyclophosphamide (4-HC) against the human rhabdomyosarcoma cell line TE-671 and the human glioma cell line D-54 MG grown in the subarachnoid space of athymic rats. The injection of 5 x 10(5) TE-671 or D-54 MG cells resulted in leptomeningeal tumor growth from the base of the brain to the cauda equina. Daily weights and neurological examinations revealed progressive neurological deficits and weight loss, with death occurring between Days 21 and 27 for TE-671 and Days 14 and 26 for D-54 MG. 4-HC toxicity in non-tumor-bearing rats was assessed at dose levels of 2.0, 10.0, 15.0, and 20.0 mM, with clinical and histological evidence of neurotoxicity observed at the 2 highest dose levels. Intrathecal treatment with 4-HC on Day 8 following injection of TE-671 resulted in an increase in median survival of 20% (P = 0.04) at 1.0 mM 4-HC and 41% (P less than 0.001) at 2.5 mM 4-HC. Intrathecal treatment with 4-HC (2.5 mM) on Day 5 following injection of D-54 MG resulted in an increase in median survival of 23% (P = 0.009). These studies show the usefulness of the athymic rat model of human neoplastic meningitis and demonstrate the efficacy in vivo of intrathecally administered 4-HC against a human glioma and a human rhabdomyosarcoma cell line and the lack of toxicity at therapeutic levels of 4-HC in normal athymic rats.


Subject(s)
Antineoplastic Agents/administration & dosage , Cyclophosphamide/analogs & derivatives , Meningeal Neoplasms/drug therapy , Meningitis/drug therapy , Spinal Cord Neoplasms/drug therapy , Animals , Cell Line , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Female , Humans , Injections, Spinal , Meningeal Neoplasms/pathology , Meningitis/etiology , Neoplasm Transplantation , Rats , Rats, Nude , Rhabdomyosarcoma/drug therapy , Spinal Cord Neoplasms/pathology , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...