Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Genes Dev ; 26(16): 1780-96, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22855790

ABSTRACT

Medulloblastoma (MB) is the most common pediatric CNS malignancy. We identify EAG2 as an overexpressed potassium channel in MBs across different molecular and histological subgroups. EAG2 knockdown not only impairs MB cell growth in vitro, but also reduces tumor burden in vivo and enhances survival in xenograft studies. Mechanistically, we demonstrate that EAG2 protein is confined intracellularly during interphase but is enriched in the plasma membrane during late G2 phase and mitosis. Disruption of EAG2 expression results in G2 arrest and mitotic catastrophe associated with failure of premitotic cytoplasmic condensation. While the tumor suppression function of EAG2 knockdown is independent of p53 activation, DNA damage checkpoint activation, or changes in the AKT pathway, this defective cell volume control is specifically associated with hyperactivation of the p38 MAPK pathway. Inhibition of the p38 pathway significantly rescues the growth defect and G2 arrest. Strikingly, ectopic membrane expression of EAG2 in cells at interphase results in cell volume reduction and mitotic-like morphology. Our study establishes the functional significance of EAG2 in promoting MB tumor progression via regulating cell volume dynamics, the perturbation of which activates the tumor suppressor p38 MAPK pathway, and provides clinical relevance for targeting this ion channel in human MBs.


Subject(s)
Cell Size , Ether-A-Go-Go Potassium Channels/metabolism , Medulloblastoma/physiopathology , Mitosis , Animals , COS Cells , Cell Cycle Checkpoints/genetics , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation/genetics , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , MAP Kinase Signaling System , Medulloblastoma/mortality , Mice , Survival Analysis
2.
Am J Pathol ; 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25451150

ABSTRACT

Available online October 16, 2014 This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

3.
Blood ; 121(5): 745-51, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23197589

ABSTRACT

UNLABELLED: Recurrent CNS lymphoma continues to be associated with poor outcomes in the rituximab era. Although IV rituximab mediates superior disease control of systemic non-Hodgkin lymphoma (NHL), it fails to completely eliminate the risk of meningeal recurrence, likely due to minimal CNS penetration. Given that rituximab acts synergistically with chemotherapy, we conducted the first phase 1 study of intraventricular immunochemotherapy in patients with recurrent CNS NHL. Fourteen patients received 10 mg or 25 mg intraventricular rituximab twice weekly for 4 weeks, with rituximab administered as monotherapy during the first treatment each week and rituximab administered in combination with methotrexate (MTX) during the second treatment each week. More than 150 doses were administered without serious toxicity. In a population with high-refractory CNS NHL, 75% of patients achieved complete cytologic responses and 43% achieved an overall complete response in CSF and/or brain parenchyma. Two patients achieved a first complete response of CNS NHL with intraventricular rituximab/MTX, including 1 with CNS lymphoma refractory to high-dose systemic and intrathecal MTX plus IV rituximab. We conclude that intraventricular rituximab in combination with MTX is feasible and highly active in the treatment of drug-resistant CNS NHL that is refractory or unresponsive to IV rituximab. KEY POINTS: Phase I study showed that intraventricular rituximab plus methotrexate is feasible and active in the treatment of refractory CNS lymphoma.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Lymphoma, Non-Hodgkin/drug therapy , Meningeal Neoplasms/drug therapy , Methotrexate/administration & dosage , Adult , Aged , Drug Resistance, Neoplasm/drug effects , Female , Humans , Infusions, Intraventricular , Lymphoma, Non-Hodgkin/pathology , Male , Meningeal Neoplasms/pathology , Meningeal Neoplasms/secondary , Middle Aged , Rituximab
4.
Clin Cancer Res ; 15(6): 1989-97, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19276270

ABSTRACT

PURPOSE: The prospect for advances in the treatment of patients with primary central nervous system lymphoma (PCNSL) is likely dependent on the systematic evaluation of its pathobiology. Animal models of PCNSL are needed to facilitate the analysis of its molecular pathogenesis and for the efficient evaluation of novel therapeutics. EXPERIMENTAL DESIGN: We characterized the molecular pathology of CNS lymphoma tumors generated by the intracerebral implantation of Raji B lymphoma cells in athymic mice. Lymphoma cells were modified for bioluminescence imaging to facilitate monitoring of tumor growth and response to therapy. In parallel, we identified molecular features of lymphoma xenograft histopathology that are evident in human PCNSL specimens. RESULTS: Intracerebral Raji tumors were determined to faithfully reflect the molecular pathogenesis of PCNSL, including the predominant immunophenotypic state of differentiation of lymphoma cells and their reactive microenvironment. We show the expression of interleukin-4 by Raji and other B lymphoma cell lines in vitro and by Raji tumors in vivo and provide evidence for a role of this cytokine in the M2 polarization of lymphoma macrophages both in the murine model and in diagnostic specimens of human PCNSL. CONCLUSION: Intracerebral implantation of Raji cells results in a reproducible and invasive xenograft model, which recapitulates the histopathology and molecular features of PCNSL, and is suitable for preclinical testing of novel agents. We also show for the first time the feasibility and accuracy of tumor bioluminescence in the monitoring of a highly infiltrative brain tumor.


Subject(s)
Brain Neoplasms/pathology , Lymphoma/pathology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Cell Polarity , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Humans , Interleukin-4/genetics , Luminescent Measurements , Lymphoma/drug therapy , Lymphoma/immunology , Macrophages/physiology , Mice , Mice, Nude , STAT6 Transcription Factor/metabolism , Temozolomide , Tumor Suppressor Proteins/genetics
5.
JCI Insight ; 3(17)2018 09 06.
Article in English | MEDLINE | ID: mdl-30185663

ABSTRACT

Although initially responsive to androgen signaling inhibitors (ASIs), metastatic castration-resistant prostate cancer (mCRPC) inevitably develops and is incurable. In addition to adenocarcinoma (adeno), neuroendocrine prostate cancer (NEPC) emerges to confer ASI resistance. We have previously combined laser capture microdissection and phage antibody display library selection on human cancer specimens and identified novel internalizing antibodies binding to tumor cells residing in their tissue microenvironment. We identified the target antigen for one of these antibodies as CD46, a multifunctional protein that is best known for negatively regulating the innate immune system. CD46 is overexpressed in primary tumor tissue and CRPC (localized and metastatic; adeno and NEPC), but expressed at low levels on normal tissues except for placental trophoblasts and prostate epithelium. Abiraterone- and enzalutamide-treated mCRPC cells upregulate cell surface CD46 expression. Genomic analysis showed that the CD46 gene is gained in 45% abiraterone-resistant mCRPC patients. We conjugated a tubulin inhibitor to our macropinocytosing anti-CD46 antibody and showed that the resulting antibody-drug conjugate (ADC) potently and selectively kills both adeno and NEPC cell lines in vitro (sub-nM EC50) but not normal cells. CD46 ADC regressed and eliminated an mCRPC cell line xenograft in vivo in both subcutaneous and intrafemoral models. Exploratory toxicology studies of the CD46 ADC in non-human primates demonstrated an acceptable safety profile. Thus, CD46 is an excellent target for antibody-based therapy development, which has potential to be applicable to both adenocarcinoma and neuroendocrine types of mCRPC that are resistant to current treatment.


Subject(s)
Adenocarcinoma/metabolism , Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/metabolism , Membrane Cofactor Protein/metabolism , Neuroendocrine Tumors/metabolism , Prostate/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Androstenes/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/pharmacology , Antibody Affinity , Antigens, Neoplasm/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Female , Humans , Macaca fascicularis , Male , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/immunology , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Recombinant Fusion Proteins , Signal Transduction/drug effects , Therapeutics , Tumor Microenvironment , Xenograft Model Antitumor Assays
7.
J Clin Invest ; 126(12): 4640-4653, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27841764

ABSTRACT

Multiple myeloma is incurable by standard approaches because of inevitable relapse and development of treatment resistance in all patients. In our prior work, we identified a panel of macropinocytosing human monoclonal antibodies against CD46, a negative regulator of the innate immune system, and constructed antibody-drug conjugates (ADCs). In this report, we show that an anti-CD46 ADC (CD46-ADC) potently inhibited proliferation in myeloma cell lines with little effect on normal cells. CD46-ADC also potently eliminated myeloma growth in orthometastatic xenograft models. In primary myeloma cells derived from bone marrow aspirates, CD46-ADC induced apoptosis and cell death, but did not affect the viability of nontumor mononuclear cells. It is of clinical interest that the CD46 gene resides on chromosome 1q, which undergoes genomic amplification in the majority of relapsed myeloma patients. We found that the cell surface expression level of CD46 was markedly higher in patient myeloma cells with 1q gain than in those with normal 1q copy number. Thus, genomic amplification of CD46 may serve as a surrogate for target amplification that could allow patient stratification for tailored CD46-targeted therapy. Overall, these findings indicate that CD46 is a promising target for antibody-based treatment of multiple myeloma, especially in patients with gain of chromosome 1q.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/pharmacology , Immunoconjugates/pharmacology , Membrane Cofactor Protein/antagonists & inhibitors , Multiple Myeloma/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neoplasm/immunology , Cell Line, Tumor , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/immunology , Gene Dosage/immunology , Humans , Immunoconjugates/immunology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Mice , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
8.
Nat Neurosci ; 18(9): 1236-46, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258683

ABSTRACT

Over 20% of the drugs for treating human diseases target ion channels, but no cancer drug approved by the US Food and Drug Administration (FDA) is intended to target an ion channel. We found that the EAG2 (Ether-a-go-go 2) potassium channel has an evolutionarily conserved function for promoting brain tumor growth and metastasis, delineate downstream pathways, and uncover a mechanism for different potassium channels to functionally cooperate and regulate mitotic cell volume and tumor progression. EAG2 potassium channel was enriched at the trailing edge of migrating medulloblastoma (MB) cells to regulate local cell volume dynamics, thereby facilitating cell motility. We identified the FDA-approved antipsychotic drug thioridazine as an EAG2 channel blocker that reduces xenografted MB growth and metastasis, and present a case report of repurposing thioridazine for treating a human patient. Our findings illustrate the potential of targeting ion channels in cancer treatment.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Drug Delivery Systems/methods , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/physiology , Evolution, Molecular , Thioridazine/administration & dosage , Animals , Brain Neoplasms/diagnosis , COS Cells , Chlorocebus aethiops , Drosophila , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods , Young Adult
9.
Clin Cancer Res ; 20(4): 1029-41, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24190981

ABSTRACT

PURPOSE: To elucidate the mechanistic basis for efficacy of intrathecal rituximab. We evaluated complement activation as well as the pharmacokinetics of intraventricular rituximab in patients who participated in two phase 1 multicenter studies. EXPERIMENTAL DESIGN: We evaluated complement activation as a candidate mediator of rituximab within the central nervous system (CNS). Complement C3 and C5b-9 were quantified by ELISA in serial cerebrospinal fluid (CSF) specimens after intraventricular rituximab administration. We determined rituximab concentration profiles in CSF and serum. A population three- compartment pharmacokinetic model was built to describe the disposition of rituximab following intraventricular administration. The model was derived from results of the first trial and validated with results of the second trial. RESULTS: Complement C3 and C5b-9 were reproducibly activated in CSF after intraventricular rituximab. Ectopic expression of C3 mRNA and protein within CNS lymphoma lesions was localized to myeloid cells. Constitutive high C3 activation at baseline was associated with adverse prognosis. A pharmacokinetic model was built, which contains three distinct compartments, to describe the distribution of rituximab within the neuroaxis after intraventricular administration. CONCLUSIONS: We provide the first evidence of C3 activation within the neuroaxis with intraventricular immunotherapy and suggest that complement may contribute to immunotherapeutic responses of rituximab in CNS lymphoma. Penetration of rituximab into neural tissue is supported by this pharmacokinetic model and may contribute to efficacy. These findings have general implications for intraventricular immunotherapy. Our data highlight potential innovations to improve efficacy of intraventricular immunotherapy both via modulation of the innate immune response as well as innovations in drug delivery.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Central Nervous System Neoplasms/cerebrospinal fluid , Complement Activation , Lymphoma/cerebrospinal fluid , Neoplasm Recurrence, Local/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Agents/administration & dosage , Central Nervous System Neoplasms/drug therapy , Central Nervous System Neoplasms/immunology , Clinical Trials, Phase I as Topic , Complement C3a/cerebrospinal fluid , Complement C5b/cerebrospinal fluid , Female , Humans , Injections, Intraventricular , Injections, Spinal , Lymphoma/drug therapy , Lymphoma/immunology , Male , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Rituximab , Tissue Distribution
10.
Clin Cancer Res ; 18(4): 1146-55, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22228634

ABSTRACT

PURPOSE: We evaluated a novel therapy for primary central nervous system lymphoma (PCNSL) with induction immunochemotherapy with high-dose methotrexate, temozolomide, and rituximab (MT-R) followed by intensive consolidation with infusional etoposide and high-dose cytarabine (EA). In addition, we evaluated the prognostic value of the minimum apparent diffusion coefficient (ADC(min)) derived from diffusion-weighted MRI (DW-MRI) in patients treated with this regimen. EXPERIMENTAL DESIGN: Thirty-one patients (median age, 61 years; median Karnofsky performance score, 60) received induction with methotrexate every 14 days for 8 planned cycles. Rituximab was administered the first 6 cycles and temozolomide administered on odd-numbered cycles. Patients with responsive or stable central nervous system (CNS) disease received EA consolidation. Pretreatment DW-MRI was used to calculate the ADC(min) of contrast-enhancing lesions. RESULTS: The complete response rate for MT-R induction was 52%. At a median follow-up of 79 months, the 2-year progression-free and overall survival were 45% and 58%, respectively. For patients receiving EA consolidation, the 2-year progression-free and overall survival were 78% and 93%, respectively. EA consolidation was also effective in an additional 3 patients who presented with synchronous CNS and systemic lymphoma. Tumor ADC(min) less than 384 × 10(-6) mm(2)/s was significantly associated with shorter progression-free and overall survival. CONCLUSIONS: MT-R induction was effective and well tolerated. MT-R followed by EA consolidation yielded progression-free and overall survival outcomes comparable to regimens with chemotherapy followed by whole-brain radiotherapy consolidation but without evidence of neurotoxicity. Tumor ADC(min) derived from DW-MRI provided better prognostic information for PCNSL patients treated with the MTR-EA regimen than established clinical risk scores.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/drug therapy , Diffusion Magnetic Resonance Imaging , Lymphoma/diagnosis , Lymphoma/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/adverse effects , Brain/pathology , Central Nervous System Neoplasms/mortality , Consolidation Chemotherapy , Cytarabine/administration & dosage , Cytarabine/adverse effects , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/analogs & derivatives , Etoposide/administration & dosage , Etoposide/therapeutic use , Female , Humans , Immunotherapy , Male , Methotrexate/administration & dosage , Methotrexate/adverse effects , Middle Aged , Prognosis , Remission Induction , Rituximab , Survival Analysis , Temozolomide , Treatment Outcome
12.
ACS Nano ; 4(2): 978-84, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20121209

ABSTRACT

Protease activity measurement has broad application in drug screening, diagnosis and disease staging, and molecular profiling. However, conventional immunopeptidemetric assays (IMPA) exhibit low fluorescence signal-to-noise ratios, preventing reliable measurements at lower concentrations in the clinically important picomolar to nanomolar range. Here, we demonstrated a highly sensitive measurement of protease activity using a nanoplasmonic resonator (NPR). NPRs enhance Raman signals by 6.1 x 10(10) times in a highly reproducible manner, enabling fast detection of proteolytically active prostate-specific antigen (paPSA) activities in real-time, at a sensitivity level of 6 pM (0.2 ng/mL) with a dynamic range of 3 orders of magnitude. Experiments on extracellular fluid (ECF) from the paPSA-positive cells demonstrate specific detection in a complex biofluid background. This method offers a fast, sensitive, accurate, and one-step approach to detect the proteases' activities in very small sample volumes.


Subject(s)
Enzyme Assays/instrumentation , Nanotechnology , Prostate-Specific Antigen/metabolism , Cell Line, Tumor , Enzyme Assays/methods , Humans , Kinetics , Male , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/enzymology , Time Factors
13.
J Clin Oncol ; 26(1): 96-105, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18056677

ABSTRACT

PURPOSE: Elucidation of the CSF proteome may yield insights into the pathogenesis of CNS disease. We tested the hypothesis that individual CSF proteins distinguish CNS lymphoma from benign focal brain lesions. METHODS: We used a liquid chromatography/mass spectrometry-based method to differentially quantify and identify several hundred CSF proteins in CNS lymphoma and control patients. We used enzyme-linked immunosorbent assay (ELISA) to confirm one of these markers in an additional validation set of 101 cases. RESULTS: Approximately 80 CSF proteins were identified and found to be present at significantly different concentrations, both higher and lower, in training and test studies, which were highly concordant. To further validate these observations, we defined in detail the expression of one of these candidate biomarkers, antithrombin III (ATIII). ATIII RNA transcripts were identified within CNS lymphomas, and ATIII protein was localized selectively to tumor neovasculature. Determination of ATIII concentration by ELISA was significantly more accurate (> 75% sensitivity; > 98% specificity) than cytology in the identification of cancer. Measurement of CSF ATIII levels was found to potentially enhance the ability to diagnose and predict outcome. CONCLUSION: Our findings demonstrate for the first time that proteomic analysis of CSF yields individual biomarkers with greater sensitivity in the identification of cancer than does CSF cytology. We propose that the discovery of CSF protein biomarkers will facilitate early and noninvasive diagnosis in patients with lesions not amenable to brain biopsy, as well as provide improved surrogates of prognosis and treatment response in CNS lymphoma and brain metastasis.


Subject(s)
Biomarkers, Tumor/cerebrospinal fluid , Brain Neoplasms/cerebrospinal fluid , Lymphoma/cerebrospinal fluid , Neoplasm Proteins/cerebrospinal fluid , Adolescent , Adult , Aged , Aged, 80 and over , Antithrombin III/genetics , Antithrombin III/metabolism , Brain Neoplasms/pathology , Case-Control Studies , Chromatography, Liquid , Diagnosis, Differential , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoblotting , Immunoenzyme Techniques , Leukemia, Myeloid/cerebrospinal fluid , Leukemia, Myeloid/pathology , Lymphoma/pathology , Lymphoma, B-Cell/cerebrospinal fluid , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell, Marginal Zone/cerebrospinal fluid , Lymphoma, B-Cell, Marginal Zone/pathology , Lymphoma, Large B-Cell, Diffuse/cerebrospinal fluid , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Non-Hodgkin/cerebrospinal fluid , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Proteomics , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Survival Rate
14.
Biol Chem ; 384(2): 257-66, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12675519

ABSTRACT

Membrane type serine protease 1 (MT-SP1) is a representative member of a large family of related enzymes known as type II transmembrane serine proteases or membrane type serine proteases. MT-SP1 has been implicated in the selective proteolysis of key extracellular substrates but its physiological role is still not fully understood. MT-SP1 expression at the protein and RNA level has been previously examined by nonquantitative methods such as in situ hybridization, Northern blotting and immunohistochemistry. To establish an introductory understanding of the quantitative mRNA expression of MT-SP1 and to correlate these levels with urokinase-type plasminogen activator receptor (uPAR), a key component of extracellular proteolysis, quantitative RT-PCR was carried out. RNA expression was analyzed in 34 human cancer cell lines, 26 human tissues and 18 primary human breast cancer tissue samples. MT-SP1 mRNA is highly expressed in many breast, ovarian, prostate and colon cancer cell lines and normal human tissues of endodermal origin. At the transcript level, MT-SP1 shows a highly statistically significant correlation (Pearson's product moment correlation r = 0.784, p < 0.001) with uPAR in human breast cancer tissue. The exact role of MT-SP1 in concert with proteins such as uPAR and other members of the plasminogen activator cascade has yet to be ascertained. However, the significant correlation between MT-SP1 and uPAR transcript levels in this initial study suggests further work to establish the role of MT-SP1 as a possible prognostic, diagnostic or therapeutic target for breast cancer.


Subject(s)
Membrane Proteins/biosynthesis , Serine Endopeptidases/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Transformed , Epithelial Cells/cytology , Epithelial Cells/enzymology , Female , Humans , Membrane Proteins/genetics , Middle Aged , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Reverse Transcriptase Polymerase Chain Reaction/methods , Serine Endopeptidases/genetics , Tissue Distribution , Transcription, Genetic , Tumor Cells, Cultured
15.
Blood ; 101(2): 466-8, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12393404

ABSTRACT

Most lymphomas that involve the central nervous system are B-cell neoplasms that express the cell surface molecule CD20. After intravenous administration, rituximab can be reproducibly measured in the cerebrospinal fluid (CSF) in patients with primary central nervous system lymphoma; however, the CSF levels of rituximab are approximately 0.1% of serum levels associated with therapeutic activity in patients with systemic non-Hodgkin lymphoma. Because lymphomatous meningitis is a frequent complication of non-Hodgkin lymphoma, we have conducted an analysis of the safety and pharmacokinetics of direct intrathecal administration of rituximab using cynomolgus monkeys. No significant acute or delayed toxicity, neurologic or otherwise, was detected. Pharmacokinetic analysis suggests that drug clearance from the CSF is biphasic, with a terminal half-life of 4.96 hours. A phase 1 study to investigate the safety and pharmacokinetics of intrathecal rituximab in patients with recurrent lymphomatous meningitis will be implemented based on these findings.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Central Nervous System Neoplasms/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Animals , Antibodies, Monoclonal/toxicity , Antibodies, Monoclonal, Murine-Derived , Area Under Curve , Drug Evaluation, Preclinical , Female , Half-Life , Injections, Spinal , Macaca fascicularis , Metabolic Clearance Rate , Rituximab
SELECTION OF CITATIONS
SEARCH DETAIL