56 results on '"Maneval DC"'
Search Results
2. Abstract P1-03-09: Pegylated recombinant human hyaluronidase PH20 (PEGPH20) enhances efficacy of eribulin mesylate (HALAVEN®) in triple negative breast cancer xenografts
- Author
-
Bahn, JD, primary, DesJardins, C, additional, Condon, KB, additional, Fathallah, A, additional, Zimmerman, S, additional, Maneval, DC, additional, Littlefield, BA, additional, and Thompson, CB, additional
- Published
- 2016
- Full Text
- View/download PDF
3. RELATIONSHIP BETWEEN DOSE-RATE OF (6RS)LEUCOVORIN ADMINISTRATION, PLASMA-CONCENTRATIONS OF REDUCED FOLATES, AND POOLS OF 5,10-METHYLENETETRAHYDROFOLATES AND TETRAHYDROFOLATES IN HUMAN COLON ADENOCARCINOMA XENOGRAFTS
- Author
-
HOUGHTON, JA, WILLIAMS, LG, DEGRAAF, SSN, CHESHIRE, PJ, RODMAN, JH, MANEVAL, DC, WAINER, IW, JADAUD, P, and HOUGHTON, PJ
- Published
- 1990
4. Gene transfer into human bone marrow hematopoietic cells mediated by adenovirus vectors [see comments]
- Author
-
Watanabe, T, primary, Kuszynski, C, additional, Ino, K, additional, Heimann, DG, additional, Shepard, HM, additional, Yasui, Y, additional, Maneval, DC, additional, and Talmadge, JE, additional
- Published
- 1996
- Full Text
- View/download PDF
5. The safety of recombinant human hyaluronidase PH20 in nonclinical models: An overview of toxicology, pharmacology, and impact of anti-PH20 antibodies.
- Author
-
Nolan RP, Kang DW, Maneval DC, Knowles SP, LaBarre MJ, and Printz MA
- Subjects
- Animals, Female, Male, Antibodies immunology, Cell Adhesion Molecules, Hyaluronic Acid chemistry, Disease Models, Animal, Hyaluronoglucosaminidase administration & dosage, Hyaluronoglucosaminidase adverse effects, Hyaluronoglucosaminidase immunology, Recombinant Proteins administration & dosage, Recombinant Proteins adverse effects, Recombinant Proteins immunology
- Abstract
Hyaluronan (HA) is a glycosaminoglycan that forms a gel-like barrier in the subcutaneous (SC) space, limiting bulk fluid flow and the dispersion of SC-administered therapeutics. Recombinant human hyaluronidase PH20 (rHuPH20) facilitates the rapid delivery of co-administered therapeutics by depolymerizing HA in the SC space. Administration of rHuPH20 can induce the formation of anti-rHuPH20 antibodies, or anti-drug antibodies (ADAs), with the potential to bind endogenous PH20 hyaluronidase in the adult testes and epididymis. Using a variety of relevant animal models and multiple dose regimens of rHuPH20 across the full spectrum of animal development, we demonstrated that rHuPH20 administration resulted in the formation of ADAs. Although these ADAs can bind both the recombinant rHuPH20 enzyme and recombinant versions of animal model-specific hyaluronidases, they had no impact on fertility parameters (as measured by sperm concentration and motility, litter size, and litter viability) or fetal development. We present the result of our nonclinical studies in order of the developmental lifecycle, beginning with adults. Toxicology studies that extend beyond the standard package are also presented. These studies demonstrate the favorable safety profile of rHuPH20 and ADAs in nonclinical models. Additionally, we identified substantial safety margins for clinically relevant doses of rHuPH20., Competing Interests: Declaration of competing interest Authors are current or former employees and shareholders of Halozyme Therapeutics, Inc. Halozyme, Inc. is not the HALO stock., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
6. JNTX-101, a novel albumin-encapsulated gemcitabine prodrug, is efficacious and operates via caveolin-1-mediated endocytosis.
- Author
-
Cui T, Corrales-Guerrero S, Castro-Aceituno V, Nair S, Maneval DC, Monnig C, Kearney P, Ellis S, Raheja N, Raheja N, and Williams TM
- Abstract
Albumin is an attractive candidate carrier for the development of novel therapeutic drugs. Gemcitabine has been FDA approved for the treatment of solid tumors; however, new drugs that optimize gemcitabine delivery are not available for clinical use. The aim of this study was to test the efficacy of a novel albumin-encapsulated gemcitabine prodrug, JNTX-101, and investigate whether Cav-1 expression predicts the therapeutic efficacy of JNTX-101. We first determined the treatment efficacy of JNTX-101 in a panel of pancreatic/lung cancer cell lines and found that increases in Cav-1 expression resulted in higher uptake of albumin, while Cav-1 depletion attenuated the sensitivity of cells to JNTX-101. In addition, decreased Cav-1 expression markedly reduced JNTX-101-induced apoptotic cell death in a panel of cells, particularly in low-serum conditions. Furthermore, we tested the therapeutic efficacy of JNTX-101 in xenograft models and the role of Cav-1 in JNTX-101 sensitivity using a Tet-on-inducible tumor model in vivo . Our data suggest that JNTX-101 effectively inhibits cell viability and tumor growth, and that Cav-1 expression dictates optimal sensitivity to JNTX-101. These data indicate that Cav-1 correlates with JNTX-101 sensitivity, especially under nutrient-deprived conditions, and supports a role for Cav-1 as a predictive biomarker for albumin-encapsulated therapeutics such as JNTX-101., Competing Interests: C.M., P.K., S.E., D.C.M., Ni.R., and Ne.R. were employees of January Therapeutics when this work was performed., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
7. Dual Stromal Targeting Sensitizes Pancreatic Adenocarcinoma for Anti-Programmed Cell Death Protein 1 Therapy.
- Author
-
Blair AB, Wang J, Davelaar J, Baker A, Li K, Niu N, Wang J, Shao Y, Funes V, Li P, Pachter JA, Maneval DC, Dezem F, Plummer J, Chan KS, Gong J, Hendifar AE, Pandol SJ, Burkhart R, Zhang Y, Zheng L, and Osipov A
- Subjects
- Humans, Mice, Animals, Hyaluronoglucosaminidase pharmacology, Hyaluronoglucosaminidase therapeutic use, Hyaluronic Acid, Focal Adhesion Protein-Tyrosine Kinases, Cytokines pharmacology, Cell Death, Polyethylene Glycols therapeutic use, Tumor Microenvironment, Pancreatic Neoplasms, Pancreatic Neoplasms pathology, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal genetics, Liver Neoplasms drug therapy
- Abstract
Background & Aims: The stroma in pancreatic ductal adenocarcinoma (PDAC) contributes to its immunosuppressive nature and therapeutic resistance. Herein we sought to modify signaling and enhance immunotherapy efficacy by targeting multiple stromal components through both intracellular and extracellular mechanisms., Methods: A murine liver metastasis syngeneic model of PDAC was treated with focal adhesion kinase inhibitor (FAKi), anti-programmed cell death protein 1 (PD-1) antibody, and stromal hyaluronan (HA) degradation by PEGylated recombinant human hyaluronidase (PEGPH20) to assess immune and stromal modulating effects of these agents and their combinations., Results: The results showed that HA degradation by PEGPH20 and reduction in phosphorylated FAK expression by FAKi leads to improved survival in PDAC-bearing mice treated with anti-PD-1 antibody. HA degradation in combination with FAKi and anti-PD-1 antibody increases T-cell infiltration and alters T-cell phenotype toward effector memory T cells. FAKi alters the expression of T-cell modulating cytokines and leads to changes in T-cell metabolism and increases in effector T-cell signatures. HA degradation in combination with anti-PD-1 antibody and FAKi treatments reduces granulocytes, including granulocytic- myeloid-derived suppressor cells and decreases C-X-C chemokine receptor type 4 (CXCR4)-expressing myeloid cells, particularly the CXCR4-expressing granulocytes. Anti-CXCR4 antibody combined with FAKi and anti-PD-1 antibody significantly decreases metastatic rates in the PDAC liver metastasis model., Conclusions: This represents the first preclinical study to identify synergistic effects of targeting both intracellular and extracellular components within the PDAC stroma and supports testing anti-CXCR4 antibody in combination with FAKi as a PDAC treatment strategy., (Copyright © 2022 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
8. Risk Factors, Hyaluronidase Expression, and Clinical Immunogenicity of Recombinant Human Hyaluronidase PH20, an Enzyme Enabling Subcutaneous Drug Administration.
- Author
-
Printz MA, Sugarman BJ, Paladini RD, Jorge MC, Wang Y, Kang DW, Maneval DC, and LaBarre MJ
- Subjects
- Humans, Adult, Male, Mice, Animals, Epitopes, B-Lymphocyte, Leukocytes, Mononuclear, Recombinant Proteins metabolism, Testis metabolism, Antibodies, Risk Factors, RNA, Messenger, RNA-Directed DNA Polymerase, Hyaluronoglucosaminidase genetics, Hyaluronoglucosaminidase metabolism, Epitopes, T-Lymphocyte genetics
- Abstract
Multiple FDA-approved and clinical-development stage therapeutics include recombinant human hyaluronidase PH20 (rHuPH20) to facilitate subcutaneous administration. As rHuPH20-reactive antibodies potentially interact with endogenous PH20, we investigated rHuPH20 immunogenicity risk through hyaluronidase tissue expression, predicted B cell epitopes, CD4+ T cell stimulation indices and related these to observed clinical immunogenicity profiles from 18 clinical studies. Endogenous hyaluronidase PH20 expression in humans/mice was assessed by reverse transcriptase-polymerase chain reaction (RT-PCR), quantitative RT-PCR, and deep RNA-Seq. rHuPH20 potential T cell epitopes were evaluated in silico and confirmed in vitro. Potential B cell epitopes were predicted for rHuPH20 sequence in silico, and binding of polyclonal antibodies from various species tested on a rHuPH20 peptide microarray. Clinical immunogenicity data were collected from 2643 subjects. From 57 human adult and fetal tissues previously screened by RT-PCR, 22 tissue types were analyzed by deep RNA-Seq. Hyaluronidase PH20 messenger RNA expression was detected in adult human testes. In silico analyses of the rHuPH20 sequence revealed nine T cell epitope clusters with immunogenic potential, one cluster was homologous to human leukocyte antigen. rHuPH20 induced T cell activation in 6-10% of peripheral blood mononuclear cell donors. Fifteen epitopes in the rHuPH20 sequence had the potential to cross-react with B cells. The cumulative treatment-induced incidence of anti-rHuPH20 antibodies across clinical studies was 8.8%. Hyaluronidase PH20 expression occurs primarily in adult testes. Low CD4+ T cell activation and B cell cross-reactivity by rHuPH20 suggest weak rHuPH20 immunogenicity potential. Restricted expression patterns of endogenous PH20 indicate low immunogenicity risk of subcutaneous rHuPH20., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
9. Safety and pharmacokinetics of docetaxel in combination with pegvorhyaluronidase alfa in patients with non-small cell lung cancer.
- Author
-
Heineman T, Baumgart M, Nanavati C, Gabrail N, Van Wart SA, Mager DE, Maneval DC, Fathallah AM, and Sekulovich RE
- Subjects
- Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacokinetics, Carcinoma, Non-Small-Cell Lung diagnosis, Carcinoma, Non-Small-Cell Lung pathology, Docetaxel administration & dosage, Docetaxel pharmacokinetics, Dose-Response Relationship, Drug, Female, Humans, Hyaluronoglucosaminidase administration & dosage, Hyaluronoglucosaminidase pharmacokinetics, Lung Neoplasms diagnosis, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Non-Small-Cell Lung drug therapy, Docetaxel adverse effects, Hyaluronoglucosaminidase adverse effects, Lung Neoplasms drug therapy
- Abstract
This open-label, phase Ib study (NCT02346370) assessed the effect of pegvorhyaluronidase alfa (PVHA; PEGPH20) on the plasma pharmacokinetics (PKs) and safety of docetaxel in 15 patients with stage IIIB/IV non-small cell lung cancer (NSCLC). The docetaxel PK profile from this study was consistent with simulations from a published docetaxel population PK model, and did not demonstrate an effect of PVHA on docetaxel PK. A maximum a posteriori Bayesian fit of the literature PK model to the docetaxel PK appeared unbiased. Adverse events (AEs) were generally consistent with previous reports for docetaxel monotherapy in NSCLC, except for higher incidence of musculoskeletal events, including myalgias, with PVHA plus docetaxel. The most common AEs were fatigue (87%), muscle spasms (60%), and myalgia (53%). Four patients experienced thromboembolic events (27%), three leading to treatment discontinuation. PVHA appeared to demonstrate an acceptable safety profile when given with docetaxel without significantly changing the plasma PK of docetaxel in patients with stage IIIB/IV NSCLC., (© 2021 Halozyme Therapeutics Inc. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of the American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
10. A Phase I Study to Evaluate the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Recombinant Human Hyaluronidase PH20 Administered Intravenously in Healthy Volunteers.
- Author
-
Printz MA, Dychter SS, DeNoia EP, Harrigan R, Sugarman BJ, Zepeda M, Souratha J, Kang DW, and Maneval DC
- Abstract
Background: Recombinant human hyaluronidase PH20 (rHuPH20) is used in subcutaneous formulations (eg, RITUXAN HYCELA [rituximab and hyaluronidase human], HERCEPTIN HYLECTA [trastuzumab and hyaluronidase-oysk], PHESGO [pertuzumab/trastuzumab/hyaluronidase-zzxf], and Darzalex FASPRO [daratumumab and hyaluronidase-fihj]) to increase the dispersion and absorption of coadministered therapeutics. Although unlikely, subcutaneous products that include rHuPH20 could be mistaken for the intravenous formulation of the corresponding drugs (eg, RITUXAN [rituximab], HERCEPTIN [trastuzumab], and DARZALEX [daratumumab]). To understand the potential effects of inadvertent intravenous injection of rHuPH20, we investigated the safety profile, pharmacokinetics (PK), and pharmacodynamics (PD) of rHuPH20 administered intravenously., Objectives: This Phase I, open-label, single-center study in healthy volunteers was designed to assess the safety profile, tolerability, PK, and PD of rHuPH20 administered intravenously., Methods: Healthy volunteers received 5 mL intravenous infusion of either 10,000 U (n = 12) or 30,000 U (n = 12) rHuPH20 over 5 minutes. Blood samples for PK and PD analysis were obtained at baseline and at various times after initiation of infusion. Adverse events and laboratory parameters were measured to assess the safety profile and tolerability of the intravenous infusion. The PK of rHuPH20 was assessed using both an enzymatic assay and a mass-based immunoassay, and plasma hyaluronan concentrations were measured as a PD marker using an HPLC-MS/MS disaccharide assay., Results: All 24 volunteers (mean age = 36.5 years) completed the study, and no serious adverse events were reported in either treatment group. Overall, 2 adverse events (both Grade 1) were reported; catheter site pain in the 10,000 U group and hypotension in the 30,000 U group. Plasma concentrations of rHuPH20 increased during the 5-minute intravenous infusion (median t
max = 6 minutes from intravenous initiation) followed by a rapid plasma clearance (t1/2 ∼10 minutes from intravenous initiation). Plasma hyaluronan concentrations increased with dose and time (tmax range = 45‒120 minutes from intravenous initiation) and returned to baseline within 1 week of administration. Changes in both PK and PD measurements appeared proportional to dose., Conclusions: The study demonstrated that intravenous administration of up to 30,000 U rHuPH20 was well tolerated, rapidly cleared from the plasma, and did not appear to be associated with any serious adverse effects at doses used in subcutaneous therapeutic products. ( Curr Ther Res Clin Exp . 2020; 81)., Competing Interests: The study was sponsored by Halozyme Therapeutics, Inc, and the data are held by the company. Additional information about the study and/or datasets can be obtained by contacting Halozyme Therapeutics, 11388 Sorrento Valley Rd, San Diego, CA 92121.E-mail: publications@halozyme.com. The study sponsor was involved in the study design, data collection, data analysis, and preparation of the manuscript. Marie A. Printz and David W. Kang are employees of Halozyme Therapeutics, Inc. Samuel S. Dychter, Barry J. Sugarman, Monica Zepeda, Jennifer Souratha, Rena Harrigan, and Daniel C. Maneval were employees of Halozyme Therapeutics Inc. at the time of the study. The authors have indicated that they have no other conflicts of interest regarding the content of this article., (© 2020 The Authors.)- Published
- 2020
- Full Text
- View/download PDF
11. ENHANZE ® drug delivery technology: a novel approach to subcutaneous administration using recombinant human hyaluronidase PH20
- Author
-
Locke KW, Maneval DC, and LaBarre MJ
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal metabolism, Antigens, Surface administration & dosage, Antigens, Surface metabolism, Antineoplastic Agents, Immunological metabolism, Cell Adhesion Molecules metabolism, Drug Delivery Systems trends, Drug Therapy, Combination, Humans, Hyaluronoglucosaminidase metabolism, Immunoglobulins metabolism, Injections, Subcutaneous, Neoplasms drug therapy, Neoplasms metabolism, Recombinant Proteins administration & dosage, Recombinant Proteins metabolism, Antineoplastic Agents, Immunological administration & dosage, Cell Adhesion Molecules administration & dosage, Drug Delivery Systems methods, Hyaluronoglucosaminidase administration & dosage, Immunoglobulins administration & dosage
- Abstract
ENHANZE
® drug delivery technology is based on the proprietary recombinant human hyaluronidase PH20 enzyme (rHuPH20; Halozyme Therapeutics, Inc.) that facilitates the subcutaneous (SC) delivery of co-administered therapeutics. rHuPH20 works by degrading the glycosaminoglycan hyaluronan (HA), which plays a role in resistance to bulk fluid flow in the SC space, limiting large volume SC drug delivery, dispersion, and absorption. Co-administration of rHuPH20 with partner therapies can overcome administration time and volume barriers associated with existing SC therapeutic formulations, and has been shown to reduce the burden on patients and healthcare providers compared with intravenous formulations. rHuPH20 (as HYLENEX® recombinant) is currently FDA-approved for subcutaneous fluid administration for achieving hydration, to increase the dispersion and absorption of other injected drugs, and in subcutaneous urography for improving resorption of radiopaque agents. rHuPH20 is also co-formulated with two anticancer therapies, trastuzumab (i.e. Herceptin® SC) and rituximab (i.e. RITUXAN HYCELA® /RITUXAN® SC/MabThera® SC) and dosed sequentially with human immunoglobin to treat primary immunodeficiency (i.e. HyQvia® /HYQVIA® ). This article reviews pharmaceutical properties of rHuPH20, its current applications with approved therapeutics, and the potential for future developments.- Published
- 2019
- Full Text
- View/download PDF
12. The growth of a xenograft breast cancer tumor model with engineered hyaluronan-accumulating stroma is dependent on hyaluronan and independent of CD44.
- Author
-
Zhao C, Thompson BJ, Chen K, Gao F, Blouw B, Marella M, Zimmerman S, Kimbler T, Garrovillo S, Bahn J, Huang L, Huang Z, Shepard HM, Rosengren S, Thanos CD, and Maneval DC
- Abstract
Hyaluronan accumulation in the tumor microenvironment is associated with poor prognosis in several solid human cancers. To understand the role of stromal hyaluronan in tumor progression, we engineered 3T3HAS3, a hyaluronan-producing fibroblast cell line, by lentiviral transduction of Balb/c 3T3 cells with the human hyaluronan synthase 3 (HAS3) gene. 3T3HAS3 cells significantly enhanced tumor growth when co-grafted with MDA-MB-468 cells in nude mice. Immunohistochemical analysis of the xenograft tumors showed that MDA-MB-468 cells were surrounded by hyaluronan-accumulating stroma, closely resembling the morphology observed in human breast cancer specimens. Tumor growth of MDA-MB-468 + 3T3HAS3 co-grafts was greatly reduced upon hyaluronan degradation by lentiviral transduction of a human hyaluronidase gene in 3T3HAS3 cells, or by systemic administration of pegvorhyaluronidase alfa (PEGPH20). In contrast, the growth of the co-graft tumors was not inhibited when CD44 expression was reduced or ablated by small hairpin RNA-mediated CD44 knockdown in MDA-MB-468 cells, CD44 CRISPR knockout in 3T3HAS3 cells, or by grafting these cells in CD44 knockout nude mice. Collectively, these data demonstrate that tumor growth of an engineered xenograft breast cancer model with hyaluronan-accumulating stroma can be dependent on hyaluronan and independent of CD44., Competing Interests: conflicts of interest All authors are or have been employees and shareholders of Halozyme Therapeutics, Inc.
- Published
- 2019
- Full Text
- View/download PDF
13. Parallel Accumulation of Tumor Hyaluronan, Collagen, and Other Drivers of Tumor Progression.
- Author
-
Li X, Shepard HM, Cowell JA, Zhao C, Osgood RJ, Rosengren S, Blouw B, Garrovillo SA, Pagel MD, Whatcott CJ, Han H, Von Hoff DD, Taverna DM, LaBarre MJ, Maneval DC, and Thompson CB
- Subjects
- Animals, Carcinogenesis metabolism, Cell Line, Tumor, Collagen genetics, Humans, Hyaluronic Acid metabolism, Hyaluronoglucosaminidase chemistry, Hyaluronoglucosaminidase genetics, Mice, Neoplasms genetics, Neoplasms metabolism, Neoplasms pathology, Tumor Microenvironment genetics, Xenograft Model Antitumor Assays, Carcinogenesis genetics, Collagen metabolism, Hyaluronoglucosaminidase administration & dosage, Neoplasms therapy
- Abstract
Purpose: The tumor microenvironment (TME) evolves to support tumor progression. One marker of more aggressive malignancy is hyaluronan (HA) accumulation. Here, we characterize biological and physical changes associated with HA-accumulating (HA-high) tumors. Experimental Design: We used immunohistochemistry, in vivo imaging of tumor pH, and microdialysis to characterize the TME of HA-high tumors, including tumor vascular structure, hypoxia, tumor perfusion by doxorubicin, pH, content of collagen. and smooth muscle actin (α-SMA). A novel method was developed to measure real-time tumor-associated soluble cytokines and growth factors. We also evaluated biopsies of murine and pancreatic cancer patients to investigate HA and collagen content, important contributors to drug resistance. Results: In immunodeficient and immunocompetent mice, increasing tumor HA content is accompanied by increasing collagen content, vascular collapse, hypoxia, and increased metastatic potential, as reflected by increased α-SMA. In vivo treatment of HA-high tumors with PEGylated recombinant human hyaluronidase (PEGPH20) dramatically reversed these changes and depleted stores of VEGF-A165, suggesting that PEGPH20 may also diminish the angiogenic potential of the TME. Finally, we observed in xenografts and in pancreatic cancer patients a coordinated increase in HA and collagen tumor content. Conclusions: The accumulation of HA in tumors is associated with high tIP, vascular collapse, hypoxia, and drug resistance. These findings may partially explain why more aggressive malignancy is observed in the HA-high phenotype. We have shown that degradation of HA by PEGPH20 partially reverses this phenotype and leads to depletion of tumor-associated VEGF-A165. These results encourage further clinical investigation of PEGPH20. Clin Cancer Res; 24(19); 4798-807. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
14. Phase 1 trials of PEGylated recombinant human hyaluronidase PH20 in patients with advanced solid tumours.
- Author
-
Infante JR, Korn RL, Rosen LS, LoRusso P, Dychter SS, Zhu J, Maneval DC, Jiang P, Shepard HM, Frost G, Von Hoff DD, Borad MJ, and Ramanathan RK
- Abstract
This corrects the article DOI: 10.1038/bjc.2017.327.
- Published
- 2018
- Full Text
- View/download PDF
15. PH20 is not expressed in murine CNS and oligodendrocyte precursor cells.
- Author
-
Marella M, Ouyang J, Zombeck J, Zhao C, Huang L, Connor RJ, Phan KB, Jorge MC, Printz MA, Paladini RD, Gelb AB, Huang Z, Frost GI, Sugarman BJ, Steinman L, Wei G, Shepard HM, Maneval DC, and Lapinskas PJ
- Abstract
Objective: Expression of Spam1 /PH20 and its modulation of high/low molecular weight hyaluronan substrate have been proposed to play an important role in murine oligodendrocyte precursor cell (OPC) maturation in vitro and in normal and demyelinated central nervous system (CNS). We reexamined this using highly purified PH20., Methods: Steady-state expression of mRNA in OPCs was evaluated by quantitative polymerase chain reaction; the role of PH20 in bovine testicular hyaluronidase (BTH) inhibition of OPC differentiation was explored by comparing BTH to a purified recombinant human PH20 (rHuPH20). Contaminants in commercial BTH were identified and their impact on OPC differentiation characterized. Spam1 /PH20 expression in normal and demyelinated mouse CNS tissue was investigated using deep RNA sequencing and immunohistological methods with two antibodies directed against recombinant murine PH20., Results: BTH, but not rHuPH20, inhibited OPC differentiation in vitro. Basic fibroblast growth factor (bFGF) was identified as a significant contaminant in BTH, and bFGF immunodepletion reversed the inhibitory effects of BTH on OPC differentiation. Spam1 mRNA was undetected in OPCs in vitro and in vivo; PH20 immunolabeling was undetected in normal and demyelinated CNS., Interpretation: We were unable to detect Spam1 /PH20 expression in OPCs or in normal or demyelinated CNS using the most sensitive methods currently available. Further, "BTH" effects on OPC differentiation are not due to PH20, but may be attributable to contaminating bFGF. Our data suggest that caution be exercised when using some commercially available hyaluronidases, and reports of Spam1 /PH20 morphogenic activity in the CNS may be due to contaminants in reagents.
- Published
- 2017
- Full Text
- View/download PDF
16. Erratum to: Clinical Immunogenicity of rHuPH20, a Hyaluronidase Enabling Subcutaneous Drug Administration.
- Author
-
Rosengren S, Dychter SS, Printz MA, Huang L, Schiff RI, Schwarz HP, McVey JK, Drake FH, Maneval DC, Kennard DA, Frost GI, Sugarman BJ, and Muchmore DB
- Published
- 2015
- Full Text
- View/download PDF
17. Clinical Immunogenicity of rHuPH20, a Hyaluronidase Enabling Subcutaneous Drug Administration.
- Author
-
Rosengren S, Dychter SS, Printz MA, Huang L, Schiff RI, Schwarz HP, McVey JK, Drake FH, Maneval DC, Kennard DA, Frost GI, Sugarman BJ, and Muchmore DB
- Subjects
- Clinical Trials as Topic, Humans, Hyaluronoglucosaminidase immunology, Injections, Subcutaneous, Insulin administration & dosage, Recombinant Proteins immunology, Rituximab administration & dosage, Trastuzumab administration & dosage, Antibodies blood, Antibodies, Neutralizing blood, Hyaluronoglucosaminidase administration & dosage, Recombinant Proteins administration & dosage
- Abstract
Recombinant human PH20 hyaluronidase (rHuPH20) is used to facilitate dispersion of subcutaneously delivered fluids and drugs. This report summarizes rHuPH20 immunogenicity findings from clinical trials where rHuPH20 was co-administered with SC human immunoglobulin, trastuzumab, rituximab, or insulin. Plasma samples were obtained from evaluable subjects participating in ten different clinical trials as well as from healthy plasma donors. A bridging immunoassay and a modified hyaluronidase activity assay were used to determine rHuPH20-reactive antibody titers and neutralizing antibodies, respectively. rHuPH20-binding antibody populations from selected subjects with positive titers were affinity-purified and subjected to further characterization such as cross-reactivity with endogenous PH20. Among individual trials, the prevalence of pre-existing rHuPH20-reactive antibodies varied between 3 and 12%, excepting the primary immunodeficiency (PID) studies. Incidence of treatment-induced rHuPH20 antibodies was 2 to 18%, with the highest titers (81,920) observed in PID. No neutralizing antibodies were observed. Within most trials, the kinetics of antibody responses were comparable between pre-existing and treatment-induced antibody responses, although responses classified as persistent were more common in subjects with pre-existing titers. There was no association between antibody positivity and either local or systemic adverse events. Pre-existing and treatment-induced antibody populations were of similar immunoglobulin isotypes and cross-reacted to endogenous PH20 to similar extents. No cross-reactivity to PH20 paralogs was detected. rHuPH20 induces only modest immunogenicity which has no association with adverse events. In addition, antibodies purified from baseline-positive individuals are qualitatively similar to those purified from individuals developing rHuPH20-reactive antibodies following exposure to the enzyme.
- Published
- 2015
- Full Text
- View/download PDF
18. Tolerability and pharmacokinetic properties of ondansetron administered subcutaneously with recombinant human hyaluronidase in minipigs and healthy volunteers.
- Author
-
Dychter SS, Harrigan R, Bahn JD, Printz MA, Sugarman BJ, DeNoia E, Haughey DB, Fellows D, and Maneval DC
- Subjects
- Administration, Oral, Adult, Aged, Animals, Cross-Over Studies, Drug Therapy, Combination, Female, Humans, Hyaluronoglucosaminidase administration & dosage, Injections, Intramuscular, Injections, Intravenous, Injections, Subcutaneous, Male, Middle Aged, Ondansetron administration & dosage, Recombinant Proteins administration & dosage, Swine, Swine, Miniature, Therapeutic Equivalency, Hyaluronoglucosaminidase adverse effects, Hyaluronoglucosaminidase pharmacokinetics, Ondansetron adverse effects, Ondansetron pharmacokinetics, Recombinant Proteins adverse effects, Recombinant Proteins pharmacokinetics
- Abstract
Background: Subcutaneous ondansetron facilitated by recombinant human hyaluronidase PH20 (rHuPH20) is an alternative for treating nausea/vomiting in patients who cannot receive ondansetron by other routes of administration., Objective: Based on preclinical results in minipigs, a Phase I study was designed to assess the tolerability and pharmacokinetic properties of subcutaneous ondansetron + rHuPH20 compared with intramuscular, intravenous, or oral ondansetron monotherapy in healthy volunteers., Methods: In a crossover design, 3 minipigs were dosed with subcutaneous ondansetron 0.08 mg/kg + rHuPH20, or as intramuscular or intravenous monotherapy, for the evaluation of plasma ondansetron concentrations and local tolerability. In a randomized, open-label, 4-way crossover study, subjects received a randomized sequence of SC ondansetron 4 mg + rHuPH20, or ondansetron monotherapy IM (4 mg), IV (4 mg), or PO (8 mg), over 4 daily visits. Study participants included healthy volunteers aged 19 to 65 years with adequate venous access in both upper extremities and no history of QT-interval prolongation. Primary tolerability end points (administration-site observations, systemic adverse events [AEs], and subject-assessed pain) were assessed, and pharmacokinetic parameters (AUC, Cmax, Tmax, t½) were computed to compare relative rate and extent of systemic exposure. Results were described using summary statistics, and bioequivalence was determined with a linear mixed-effects model., Results: In the preclinical study, no adverse events or significant local reactions were observed. The Cmax (45.8 ng/mL at 0.08 hour) with subcutaneous administration + rHuPH20 was 83% greater and was achieved 68% faster than with intramuscular administration (Cmax = 25 ng/mL at 0.25 hour). In the clinical study, a total of 12 subjects (7 women, 5 men; white majority; mean age, 44.8) were randomized. The majority of AEs were at the injection site, mild in severity, and transient. After subcutaneous administration of ondansetron + rHuPH20, geometric mean Cmax was 35% higher than with intramuscular ondansetron, 43% lower than with intravenous ondansetron, and 126% higher than with oral ondansetron (corrected for dose). Bioequivalence tests demonstrated that systemic exposure after subcutaneous administration was similar to that after intramuscular or intravenous administration and significantly greater than that after oral administration., Conclusions: Subcutaneous ondansetron + rHuPH20 was generally well-tolerated. Subcutaneous dosing resulted in an extent of systemic exposure similar to that with intramuscular or intravenous dosing and greater than that with oral administration, and may be an option for clinical administration of ondansetron. ClinicalTrials.gov identifier: NCT01572012., (Copyright © 2014 Elsevier HS Journals, Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
19. Phase I trial of intravesical recombinant adenovirus mediated interferon-α2b formulated in Syn3 for Bacillus Calmette-Guérin failures in nonmuscle invasive bladder cancer.
- Author
-
Dinney CP, Fisher MB, Navai N, O'Donnell MA, Cutler D, Abraham A, Young S, Hutchins B, Caceres M, Kishnani N, Sode G, Cullen C, Zhang G, Grossman HB, Kamat AM, Gonzales M, Kincaid M, Ainslie N, Maneval DC, Wszolek MF, and Benedict WF
- Subjects
- Adenoviridae genetics, Administration, Intravesical, Adult, Aged, Aged, 80 and over, BCG Vaccine administration & dosage, Carcinoma, Transitional Cell mortality, Carcinoma, Transitional Cell pathology, Cholic Acids administration & dosage, Disaccharides administration & dosage, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Genetic Vectors, Humans, Interferon alpha-2, Interferon-alpha genetics, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Invasiveness pathology, Neoplasm Recurrence, Local pathology, Prognosis, Recombinant Proteins administration & dosage, Recombinant Proteins genetics, Risk Assessment, Survival Analysis, Treatment Failure, Treatment Outcome, Urinary Bladder Neoplasms mortality, Urinary Bladder Neoplasms pathology, Carcinoma, Transitional Cell therapy, Genetic Therapy methods, Interferon-alpha administration & dosage, Neoplasm Recurrence, Local mortality, Neoplasm Recurrence, Local therapy, Urinary Bladder Neoplasms therapy
- Abstract
Purpose: A phase I trial of intravesical recombinant adenovirus mediated interferon-α2b gene therapy (rAd-IFNα) formulated with the excipient SCH Syn3 was conducted in patients with nonmuscle invasive bladder cancer who had disease recurrence after treatment with bacillus Calmette-Guérin. The primary objective was to determine the safety of rAd-IFNα/Syn3. Secondary end points were demonstrated effective rAd-IFNα gene expression and preliminary evidence of clinical activity at 3 months., Materials and Methods: A total of 17 patients with recurrent nonmuscle invasive bladder cancer after bacillus Calmette-Guérin treatment were enrolled in the study. A single treatment of rAd-IFNα (3 × 10(9) to 3 × 10(11) particles per ml) formulated with the excipient Syn3 was administered. Patient safety was evaluated for 12 or more weeks. Efficacy of gene transfer was determined by urine IFNα protein concentrations. Preliminary drug efficacy was determined at 3 months., Results: Intravesical rAd-IFNα/Syn3 was well tolerated as no dose limiting toxicity was encountered. Urgency was the most common adverse event and all cases were grade 1 or 2. rAd-IFNα DNA was not detected in the blood. However, transient low serum IFNα and Syn3 levels were measured. High and prolonged dose related urine IFNα levels were achieved with the initial treatment. Of the 14 patients treated at doses of 10(10) or more particles per ml with detectable urine IFNα, 6 (43%) experienced a complete response at 3 months and 2 remained disease-free at 29.0 and 39.2 months, respectively., Conclusions: Intravesical rAd-IFNα/Syn3 was well tolerated with no dose limiting toxicity encountered. Dose dependent urinary IFNα concentrations confirmed efficient gene transfer and expression. Intravesical rAd-IFNα/Syn3 demonstrated clinical activity in nonmuscle invasive bladder cancer recurring after bacillus Calmette-Guérin., (Copyright © 2013 American Urological Association Education and Research, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
20. Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer.
- Author
-
Jacobetz MA, Chan DS, Neesse A, Bapiro TE, Cook N, Frese KK, Feig C, Nakagawa T, Caldwell ME, Zecchini HI, Lolkema MP, Jiang P, Kultti A, Thompson CB, Maneval DC, Jodrell DI, Frost GI, Shepard HM, Skepper JN, and Tuveson DA
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Pancreatic Ductal blood supply, Carcinoma, Pancreatic Ductal mortality, Carcinoma, Pancreatic Ductal physiopathology, Cell Adhesion Molecules administration & dosage, Cell Adhesion Molecules pharmacology, Deoxycytidine administration & dosage, Deoxycytidine analogs & derivatives, Doxorubicin administration & dosage, Drug Resistance, Neoplasm drug effects, Hyaluronoglucosaminidase administration & dosage, Hyaluronoglucosaminidase pharmacology, Immunohistochemistry, Kaplan-Meier Estimate, Mice, Mice, Transgenic, Pancreatic Neoplasms blood supply, Pancreatic Neoplasms mortality, Pancreatic Neoplasms physiopathology, Recombinant Proteins administration & dosage, Recombinant Proteins pharmacology, Tissue Array Analysis, Treatment Outcome, Gemcitabine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor physiology, Carcinoma, Pancreatic Ductal drug therapy, Drug Delivery Systems, Drug Resistance, Neoplasm physiology, Hyaluronic Acid physiology, Pancreatic Neoplasms drug therapy
- Abstract
Objective: Pancreatic ductal adenocarcinoma (PDA) is characterised by stromal desmoplasia and vascular dysfunction, which critically impair drug delivery. This study examines the role of an abundant extracellular matrix component, the megadalton glycosaminoglycan hyaluronan (HA), as a novel therapeutic target in PDA., Methods: Using a genetically engineered mouse model of PDA, the authors enzymatically depleted HA by a clinically formulated PEGylated human recombinant PH20 hyaluronidase (PEGPH20) and examined tumour perfusion, vascular permeability and drug delivery. The preclinical utility of PEGPH20 in combination with gemcitabine was assessed by short-term and survival studies., Results: PEGPH20 rapidly and sustainably depleted HA, inducing the re-expansion of PDA blood vessels and increasing the intratumoral delivery of two chemotherapeutic agents, doxorubicin and gemcitabine. Moreover, PEGPH20 triggered fenestrations and interendothelial junctional gaps in PDA tumour endothelia and promoted a tumour-specific increase in macromolecular permeability. Finally, combination therapy with PEGPH20 and gemcitabine led to inhibition of PDA tumour growth and prolonged survival over gemcitabine monotherapy, suggesting immediate clinical utility., Conclusions: The authors demonstrate that HA impedes the intratumoral vasculature in PDA and propose that its enzymatic depletion be explored as a means to improve drug delivery and response in patients with pancreatic cancer.
- Published
- 2013
- Full Text
- View/download PDF
21. Herceptin.
- Author
-
Shepard HM, Jin P, Slamon DJ, Pirot Z, and Maneval DC
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal history, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Antineoplastic Agents chemistry, Antineoplastic Agents history, Antineoplastic Agents pharmacology, Breast Neoplasms immunology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Drug Design, Epidermal Growth Factor metabolism, Female, History, 20th Century, History, 21st Century, Humans, Immunotherapy history, Models, Molecular, Protein Conformation, Receptor, ErbB-2 chemistry, Receptor, ErbB-2 metabolism, Signal Transduction drug effects, Trastuzumab, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Immunotherapy methods, Receptor, ErbB-2 immunology
- Abstract
The biology of the human epidermal growth factor (EGF) receptor-2 (HER2) has been reviewed numerous times and provides an excellent example for developing a targeted cancer therapeutic. Herceptin, the FDA-approved therapeutic monoclonal antibody against HER2, has been used to treat over 150,000 women with breast cancer. However, the developmental history of Herceptin, the key events within the program that created pivotal decision points, and the reasons why decisions were made to pursue the monoclonal antibody approach have never been adequately described. The history of Herceptin is reviewed in a way which allows the experience to be shared for the purposes of understanding the drug discovery and development process. It is the objective of this review to describe the pivotal events and explain why critical decisions were made that resulted in the first therapeutic to successfully target tyrosine kinases in cancer. New approaches and future prospects for therapeutics targeting the HER family are also discussed.
- Published
- 2008
- Full Text
- View/download PDF
22. Enhancement of intravesical delivery with Syn3 potentiates interferon-alpha2b gene therapy for superficial bladder cancer.
- Author
-
Nagabhushan TL, Maneval DC, Benedict WF, Wen SF, Ihnat PM, Engler H, and Connor RJ
- Subjects
- Adenoviridae genetics, Administration, Intravesical, Animals, Cholic Acids therapeutic use, Disaccharides therapeutic use, Excipients therapeutic use, Genetic Therapy, Humans, Interferon alpha-2, Interferon-alpha therapeutic use, Recombinant Proteins, Urinary Bladder Neoplasms metabolism, Urothelium metabolism, Cholic Acids administration & dosage, Disaccharides administration & dosage, Excipients administration & dosage, Interferon-alpha administration & dosage, Interferon-alpha genetics, Urinary Bladder Neoplasms therapy
- Abstract
Intravesical administration of interferon alpha-2b protein (IFN) has been successfully used in the treatment of patients with superficial bladder tumors. Local dosing of IFN minimizes well-known systemic side effects of the drug, but exposure to bladder tumors is limited by the duration of instillation and transient concentrations achieved in the urothelium. Intravesical delivery of the gene encoding interferon results in an alternative strategy for IFN-based therapy of the disease, enabling sustained exposure of IFN protein that results from production by tumor and non-tumor cells in the urothelium. Efficient gene delivery and expression of IFN has been achieved using a recombinant adenovirus gene delivery system (rAd-IFN) in conjunction with the novel small molecule excipient Syn3. Studies with rAd-IFN/Syn3 in animal models result in urine concentrations of IFN that persisted for weeks and correlated with potent anti-tumor effects. The objective of this review is to communicate the rationale and preclinical findings that support ongoing clinical investigation of intravesical rAd-IFN/Syn3 in superficial bladder cancer.
- Published
- 2007
- Full Text
- View/download PDF
23. Sustained intravesical interferon protein exposure is achieved using an adenoviral-mediated gene delivery system: a study in rats evaluating dosing regimens.
- Author
-
Connor RJ, Anderson JM, Machemer T, Maneval DC, and Engler H
- Subjects
- Adenoviridae genetics, Animals, Dose-Response Relationship, Drug, Drug Carriers, Female, Interferon alpha-2, Rats, Rats, Sprague-Dawley, Recombinant Proteins, Interferon-alpha administration & dosage, Interferon-alpha genetics, Urinary Bladder
- Abstract
Objectives: To evaluate whether a recombinant replication-deficient adenovirus containing the secreted human interferon alpha-2b gene (rAd-IFN) could improve the tissue and urine levels of IFN protein by transducing the urothelium with the secreted human IFN-alpha gene. We also assessed whether varying the interval between rAd-IFN/Syn3 treatments would improve the duration and levels of gene expression., Methods: The rats received intravesical administration of rAd-IFN at varying concentrations in a formulation containing Syn3, an agent identified that facilitates passage of the adenovirus through the protective barrier of the bladder. Urine was collected daily for 7 days, and human IFN was measured in the urine by enzyme-linked immunosorbent assay. For the redosing studies, the animals received a second dose at varying intervals ranging from 1 to 7 days after the first dose or at longer intervals (30, 60, or 90 days)., Results: Rats that received intravesical administration of rAd-IFN in a Syn3 formulation expressed levels of human IFN protein in their urine at peak concentrations of 50,000 to 100,000 pg/mL, but were undetectable by 7 days. Expression was localized to the bladder with only minimal systemic exposure to IFN. Short-term redosing marginally improved the IFN urine concentrations, with maximal levels achieved when a second dose was administered 3 days after a first dose. Although gene expression was attenuated when a second dose was given 5 to 7 days after the first treatment, the levels and duration of IFN expression recovered when the interval was increased to 90 days., Conclusions: Intravesical treatment with rAd-IFN facilitates high levels of IFN transgene exposure and may be a new approach to treating superficial bladder cancer.
- Published
- 2005
- Full Text
- View/download PDF
24. p21WAF-1/Cip-1 gene therapy as an adjunct to glaucoma filtration surgery.
- Author
-
Atencio IA, Chen Z, Nguyen QH, Faha B, and Maneval DC
- Subjects
- Animals, Cyclin-Dependent Kinase Inhibitor p21, Humans, Cell Cycle Proteins genetics, Cell Cycle Proteins therapeutic use, Genetic Therapy methods, Glaucoma Drainage Implants adverse effects
- Abstract
Glaucoma is a blinding eye disease characterized by elevated intraocular pressure (IOP). Glaucoma filtration surgery (GFS) is designed to reduce IOP, but wound healing responses to the procedure can result in surgical failure. Anti-metabolites used in conjunction with GFS are commonly employed to control the wound healing response, but have unwanted side effects. This review describes the therapeutic potential of ocular gene therapy using an adenovirus vector containing the human p21WAF-1/Cip-1 gene (rAd-p21) to control unwanted wound healing post-GFS. Here, we summarize encouraging preclinical data in relevant models, and propose rAd-p21 gene therapy as an alternative to the currently used methods of wound healing modulation.
- Published
- 2004
25. Impact of human neutralizing antibodies on antitumor efficacy of an oncolytic adenovirus in a murine model.
- Author
-
Tsai V, Johnson DE, Rahman A, Wen SF, LaFace D, Philopena J, Nery J, Zepeda M, Maneval DC, Demers GW, and Ralston R
- Subjects
- Animals, Body Weight, Cell Line, Tumor, Disease Models, Animal, Dose-Response Relationship, Drug, Genetic Vectors, Humans, Mice, Mice, Nude, Mice, SCID, Neoplasm Transplantation, Polymerase Chain Reaction, Time Factors, beta-Galactosidase genetics, Adenoviridae genetics, Gene Transfer Techniques, Genetic Therapy methods
- Abstract
Purpose: The purpose of this study was to assess the impact of anti-adenovirus neutralizing antibodies (AdNAbs) on the distribution, tolerability, and efficacy of intravenously administered oncolytic adenovirus. A translational model was developed to evaluate the impact of humoral immunity on intravenous administration of oncolytic adenovirus in humans., Experimental Design: Initially, severe combined immunodeficient (SCID)/beige mice were passively immunized with various amounts of human sera to establish a condition of preexisting humoral immunity similar to humans. A replication-deficient adenovirus encoding beta-galactosidase (rAd-betagal) was injected intravenously into these mice. An AdNAb titer that mitigated galactosidase transgene expression was determined. A xenograft tumor-bearing nude mouse model was developed to assess how a similar in vivo titer would impact the activity of 01/PEME, an oncolytic adenovirus, after intravenous administration., Results: In SCID/beige mice, there was a dose dependence between AdNAbs and galactosidase transgene expression; 90% of transgene expression was inhibited when the titer was 80. A similar titer reconstituted in the nude mice with human serum, as was done in the SCID/beige mice, did not abrogate the antitumor efficacy of the replicating adenovirus after intravenous administration. Viral DNA increased in tumors over time., Conclusions: In intravenous administration, preexisting AdNAb titer of 80 significantly attenuated the activity of a 2.5 x 10(12) particles per kilogram dose of nonreplicating adenovirus; the same titer had no affect on the activity of an equivalent dose of replicating adenovirus. Our results suggest that a majority of patients with preexisting adenovirus immunity would be candidates for intravenous administration of oncolytic adenovirus.
- Published
- 2004
- Full Text
- View/download PDF
26. Intravesical Ad-IFNalpha causes marked regression of human bladder cancer growing orthotopically in nude mice and overcomes resistance to IFN-alpha protein.
- Author
-
Benedict WF, Tao Z, Kim CS, Zhang X, Zhou JH, Adam L, McConkey DJ, Papageorgiou A, Munsell M, Philopena J, Engler H, Demers W, Maneval DC, Dinney CP, and Connor RJ
- Subjects
- Administration, Intravesical, Animals, Caspases metabolism, Cell Death, Drug Resistance, Green Fluorescent Proteins genetics, Humans, Immunochemistry, Interferon alpha-2, Interferon-alpha therapeutic use, Mice, Mice, Nude, Neoplasm Transplantation, Nylons pharmacology, Recombinant Proteins, Transplantation, Heterologous, Tumor Cells, Cultured, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms pathology, Adenoviridae genetics, Genetic Therapy, Interferon-alpha genetics, Urinary Bladder Neoplasms therapy
- Abstract
We have produced prolonged, high local concentrations of interferon in vivo by intravesical instillation of adenoviruses encoding interferon-alpha (Ad-IFNalpha) together with the gene transfer-enhancing agent Syn3. We found sustained interferon protein levels for days, both in normal mouse urothelium and in human bladder cancer cells growing as superficial bladder tumors in nude mice using an orthotopic bladder model developed by us. Tumor burden in the bladder was determined utilizing cancer cells containing the green fluorescent protein. Marked tumor regression was observed following two 1-h exposures of Ad-IFNalpha/Syn3 and little or no cytotoxicity was detected in normal cells. Similar intravesical instillation of clinically relevant concentrations of IFN protein alone or Ad-IFNalpha without Syn3 was ineffective. Surprisingly, in vitro, Ad-IFNalpha also caused caspase-dependent death of bladder cancer cell lines that were resistant to high concentrations of IFN-alpha protein, including the cell line used in vivo. These findings demonstrate that Ad-IFNalpha can overcome resistance to IFN-alpha protein both in vitro and in vivo and support evaluation of intravesical Ad-IFNalpha/Syn3 for the treatment of superficial bladder cancer.
- Published
- 2004
- Full Text
- View/download PDF
27. Development of a formulation that enhances gene expression and efficacy following intraperitoneal administration in rabbits and mice.
- Author
-
Engler H, Machemer TR, Schluep T, Wen SF, Quijano E, Wills KN, Harper AE, Maneval DC, and Conroy SE
- Subjects
- Animals, Female, Gene Expression Regulation, Genetic Therapy, Genetic Vectors administration & dosage, Glucans therapeutic use, Glucose therapeutic use, Humans, Icodextrin, Injections, Intraperitoneal, Mice, Mice, Nude, Peritoneum enzymology, Rabbits, Rats, Rats, Sprague-Dawley, Transgenes genetics, beta-Galactosidase metabolism, Adenoviridae genetics, Glucans administration & dosage, Glucose administration & dosage, Ovarian Neoplasms metabolism, Ovarian Neoplasms therapy
- Abstract
We conducted a series of experiments to determine if intraperitoneal (IP) delivery of recombinant adenovirus (rAd)-based therapies is improved through carrier vehicle selection, and compared an icodextrin solution (a high molecular weight dextrin with a prolonged peritoneal cavity residence time) with a standardized phosphate buffered saline (PBS) delivery solution. In vitro, comparative adenovirus particle concentration determination (27 h) and bioactivity assay (24h) indicated equivalent compatibility with icodextrin or PBS. In vivo, rabbits treated IP (100 ml) with rAd-betagal 1 x 10(9) P/ml in icodextrin showed improved transgene expression throughout the peritoneal wall compared to rAd-betagal in PBS. In PC-3 tumor-bearing mice treated IP with 5 x 10(9) P/0.5 ml or 1 x 10(10) P/0.5 ml rAd-betagal, transgene expression was significantly enhanced (p < 0.01) with icodextrin compared to PBS in both tumor specimens and peritoneal wall. In subsequent studies we compared prolongation of survival in intraperitoneal PC-3 and MDAH-2774 human xenograft tumor models in nude mice using rAd-p53 in icodextrin or PBS in multi-dose ranging (1 x 10(8) to 1 x 10(10) P) experiments. The icodextrin formulation alone significantly increased rAd-p53 mediated survival (p < 0.05). In animals, these results show that IP rAd gene therapy can be improved with the use of icodextrin, and suggest that prolonged retention and distribution in the peritoneal cavity is an important factor.
- Published
- 2003
- Full Text
- View/download PDF
28. Syn3 provides high levels of intravesical adenoviral-mediated gene transfer for gene therapy of genetically altered urothelium and superficial bladder cancer.
- Author
-
Yamashita M, Rosser CJ, Zhou JH, Zhang XQ, Connor RJ, Engler H, Maneval DC, Karashima T, Czerniak BA, Dinney CP, and Benedict WF
- Subjects
- Animals, Female, Genetic Vectors, Humans, Mice, Mice, Nude, Swine, Tumor Cells, Cultured, Adenoviridae genetics, Cholic Acids administration & dosage, Disaccharides administration & dosage, Genetic Therapy, Transfection, Urothelium metabolism
- Abstract
Using our model to grow superficial human bladder cancer in the mouse bladder, we have found that the polyamide compound, Syn3, when injected intravesically for 1 hour at 1 mg/mL on two consecutive days, markedly increases rAd-beta-gal intravesical gene transfer and expression. This enhanced transgene expression was much greater than obtain by the use of 22% ethanol, which had previously been shown to increase intravesical adenoviral gene transfer, whereas little or no gene expression was seen with exposure to only rAd-beta-gal. beta-Galactosidase staining was seen in virtually every normal urothelial and superficial tumor cell present, including tumors that express little or no coxsackie-adenovirus receptors when Syn3 was present. High adenoviral-mediated gene transfer was also documented in the pig bladder using Syn3 in a similar protocol. Therefore, Syn3 may overcome the limitations of adequate intravesical adenoviral-mediated gene transfer and, when combined with an appropriate adenoviral-mediated gene, could offer an effective approach to the treatment of superficial bladder cancer and perhaps even genetically altered precursor lesions.
- Published
- 2002
- Full Text
- View/download PDF
29. Interferon-alpha2b secretion by adenovirus-mediated gene delivery in rat, rabbit, and chimpanzee results in similar pharmacokinetic profiles.
- Author
-
Demers GW, Sugarman BJ, Beltran JC, Westreich LN, Ahmed CM, Lau JY, Hong Z, Lanford RE, and Maneval DC
- Subjects
- Adenoviridae genetics, Animals, Dose-Response Relationship, Drug, Female, Genetic Vectors, Immunocompromised Host, Interferon alpha-2, Interferon-alpha genetics, Interferon-alpha immunology, Mice, Mice, SCID, Rabbits, Rats, Rats, Inbred BUF, Recombinant Proteins, Severe Combined Immunodeficiency genetics, Severe Combined Immunodeficiency immunology, Severe Combined Immunodeficiency metabolism, Species Specificity, Genetic Therapy methods, Interferon-alpha pharmacokinetics, Pan troglodytes metabolism
- Abstract
Gene delivery, with subsequent protein synthesis and secretion, in vivo has been proposed as an alternative way to deliver a therapeutic protein to the systemic circulation. Interferon-alpha (IFN) protein is effective in the treatment of viral and malignant diseases but has short serum half-life that requires frequent administration. An E1 region-deleted adenovirus vector encoding human IFN-alpha2b gene driven by the cytomegalovirus immediate early promoter (rAd-IFN) was generated to assess the serum concentration-time profiles of expressed IFN protein in animal models. Intravenous administration of rAd-IFN, normalized for body weight, resulted in dose-dependent serum IFN concentrations that persisted 8-40 days with similar concentration-time profiles in rats and rabbits. We sought to determine if serum concentration-time profiles in the rat and rabbit animal models would be predictive for a larger animal and would therefore be relevant models for potential dosing of human patients. Two chimpanzees (approximately 70 kg) dosed with rAd-IFN by intravenous administration normalized to body weight achieved serum IFN concentration-time profiles similar to those observed in rats and rabbits. The role of the immune response in limiting the persistence of transgene expression was highlighted by the persistence of serum IFN concentrations for over 200 days in beige/SCID immunodeficient mice. These studies suggest that serum concentration of secreted transgene products after gene delivery in small animal models may be highly predictive for larger species and will help define dosing strategies in human patients., ((c)2002 Elsevier Science (USA).)
- Published
- 2002
- Full Text
- View/download PDF
30. Intra-arterial delivery of p53-containing adenoviral vector into experimental brain tumors.
- Author
-
Abe T, Wakimoto H, Bookstein R, Maneval DC, Chiocca EA, and Basilion JP
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms pathology, Disease Models, Animal, Diuretics, Osmotic pharmacokinetics, Diuretics, Osmotic pharmacology, Female, Genes, erbB-1 physiology, Genetic Vectors, Glioma genetics, Glioma pathology, Humans, Infusions, Intra-Arterial, Lac Operon physiology, Mannitol pharmacokinetics, Mannitol pharmacology, Neoplasm Transplantation, Rats, Rats, Nude, Survival Rate, Tissue Distribution, Transduction, Genetic, Treatment Outcome, Tumor Suppressor Protein p53 metabolism, Adenoviridae genetics, Brain Neoplasms therapy, Genetic Therapy methods, Glioma therapy, Tumor Suppressor Protein p53 genetics
- Abstract
Human tumor xenografts established in athymic rat brains were used to determine the feasibility of intravascular delivery of tumor suppressor genes to brain tumors. Both tumor size and number were compared to characterize the effect of tumor burden on tumor transduction efficacy by a control LacZ-containing adenoviral vector. Experiments with tumors grown in vivo for either 3, 5, or 7 days demonstrated that 5-day-old tumors provided the best target for vector infection and transgene expression by this mode of administration. Intra-arterial mannitol facilitated transduction efficiency. Tumor burden did not seem to affect transduction, while tumor location appeared to be an important factor. Based on these results, intra-arterial infusion of a p53-containing adenoviral vector was carried out and resulted in significant retardation of brain tumor growth 3 days after administration. Effects at longer time points were not as significant. These findings indicate that intra-arterial administration of adenoviral vectors containing p53 is efficient and can result in changes in tumor size, but that long-term control of tumor growth may require multiple adenoviral treatments.
- Published
- 2002
- Full Text
- View/download PDF
31. Successful adenovirus-mediated wild-type p53 gene transfer in patients with bladder cancer by intravesical vector instillation.
- Author
-
Kuball J, Wen SF, Leissner J, Atkins D, Meinhardt P, Quijano E, Engler H, Hutchins B, Maneval DC, Grace MJ, Fritz MA, Störkel S, Thüroff JW, Huber C, and Schuler M
- Subjects
- Adenoviridae genetics, Administration, Intravesical, Adult, Aged, Aged, 80 and over, Cystectomy, DNA Primers, Dose-Response Relationship, Drug, Humans, Middle Aged, Neoplasm Invasiveness, Reverse Transcriptase Polymerase Chain Reaction, Urinary Bladder Neoplasms pathology, Gene Transfer Techniques, Genes, p53, Genetic Therapy, Genetic Vectors therapeutic use, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms therapy
- Abstract
Purpose: To study safety, feasibility, and biologic activity of adenovirus-mediated p53 gene transfer in patients with bladder cancer., Patients and Methods: Twelve patients with histologically confirmed bladder cancer scheduled for cystectomy were treated on day 1 with a single intratumoral injection of SCH 58500 (rAd/p53) at cystoscopy at one dose level (7.5 x 10(11) particles) or a single intravesical instillation of SCH 58500 with a transduction-enhancing agent (Big CHAP) at three dose levels (7.5 x 10(11) to 7.5 x 10(13) particles). Cystectomies were performed in 11 patients on day 3, and transgene expression, vector distribution, and biologic markers of transgene activity were assessed by molecular and immunohistochemical methods in tumors and normal bladder samples., Results: Specific transgene expression was detected in tissues from seven of eight assessable patients treated with intravesical instillation of SCH 58500 but in none of three assessable patients treated with intratumoral injection of SCH 58500. Induction of RNA and protein expression of the p53 target gene p21/WAF1 was demonstrated in samples from patients treated with SCH 58500 instillation at higher dose levels. Distribution studies after intravesical instillation of SCH 58500 revealed both high transduction efficacy and vector penetration throughout the whole urothelium and into submucosal tumor cells. No dose-limiting toxicity was observed, and side effects were local and of transient nature., Conclusion: Intravesical instillation of SCH 58500 combined with a transduction-enhancing agent is safe, feasible, and biologically active in patients with bladder cancer. Studies to evaluate the clinical efficacy of this treatment in patients with localized high-risk bladder cancer are warranted.
- Published
- 2002
- Full Text
- View/download PDF
32. Interferon alpha2b gene delivery using adenoviral vector causes inhibition of tumor growth in xenograft models from a variety of cancers.
- Author
-
Iqbal Ahmed CM, Johnson DE, Demers GW, Engler H, Howe JA, Wills KN, Wen SF, Shinoda J, Beltran J, Nodelman M, Machemer T, Maneval DC, Nagabhushan TL, and Sugarman BJ
- Subjects
- Animals, Blotting, Western, Cell Division, Cytomegalovirus genetics, DNA Primers chemistry, Female, Humans, Interferon alpha-2, Interferon-alpha metabolism, Mice, Mice, Inbred BALB C, Mice, Inbred ICR, Mice, Nude, Mice, SCID, Neoplasm Metastasis, Neoplasms, Experimental mortality, Recombinant Proteins, Reverse Transcriptase Polymerase Chain Reaction, Survival Rate, Transplantation, Heterologous, Tumor Cells, Cultured, Adenoviridae genetics, Genetic Therapy methods, Genetic Vectors, Interferon-alpha genetics, Neoplasms, Experimental therapy
- Abstract
A recombinant adenovirus expressing human interferon alpha2b driven by the cytomegalovirus promoter, IACB, was shown to produce and secrete biologically active protein in vitro and in vivo. Intravenous administration of IACB in Buffalo rats resulted in circulating levels of biologically active human interferon at 70,000 international units/mL for up to 15 days. Distribution of interferon protein after IACB administration was different from that seen with the subcutaneous delivery of interferon protein. Higher levels of interferon protein were observed in liver and spleen after IACB delivery compared to protein delivery. The antitumor efficacy of IACB, as measured by suppression of tumor growth, was tested in athymic nude mice bearing established human tumor xenografts from different types of human cancer. Subcutaneous tumors most responsive to the intratumoral administration of IACB ranked as U87MG (glioblastoma) and K562 (chronic myelogenous leukemia), followed by Hep 3B (hepatocellular carcinoma) and LN229 cells (glioblastoma). Intravenous administration of IACB in animals bearing U87MG or Hep 3B xenografts was also effective in suppressing tumor growth, although to a lesser extent than the intratumoral administration. IACB was also tested in a metastatic model in beige/SCID mice generated with H69 (small cell lung carcinoma) cells and was found to prolong survival in tumor-bearing animals. This suggested that interferon gene delivery can be effective in suppressing tumor growth in a wide variety of cells.
- Published
- 2001
- Full Text
- View/download PDF
33. Purging of human breast cancer cells from stem cell products with an adenovirus containing p53.
- Author
-
Hirai M, Kelsey LS, Vaillancourt M, Maneval DC, Watanabe T, and Talmadge JE
- Subjects
- Breast Neoplasms pathology, Breast Neoplasms therapy, Cell Death genetics, Cell Division genetics, Cell Separation methods, Clone Cells pathology, Coculture Techniques, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Growth Inhibitors genetics, Growth Inhibitors toxicity, Hematopoietic Stem Cells metabolism, Humans, Tumor Cells, Cultured, Tumor Stem Cell Assay, Tumor Suppressor Protein p53 biosynthesis, Adenoviridae genetics, Breast Neoplasms genetics, Breast Neoplasms virology, Hematopoietic Stem Cells pathology, Tumor Suppressor Protein p53 genetics
- Abstract
Tumor cell contamination of stem cell products can contribute to tumor relapse following high-dose chemotherapy and stem cell rescue. Numerous techniques have been used to remove the tumor cells from stem cell products with the objective of prolonging relapse-free survival. However, to date these techniques have been relatively ineffectual and/or toxic to hematopoietic stem and progenitor cells. The differential infectivity of adenovirus (Adv) vectors for breast cancer cells, compared with hematopoietic cells, has suggested that Adv-p53 might provide an effective purging strategy. To facilitate the use of Adv-p53 as a clinical strategy, we undertook studies to determine the parameters necessary for optimal stem cell product purging. The parameters studied were the particle number to nucleated cell ratio, the duration of coincubation, the incubation volume, and the presence or absence of hematopoietic progenitor cells. We have found that these parameters are interdependent and conclude that a 4-hour coincubation with an Adv-p53 particle to nucleated cell ratio of 2000:1 with 2 x 10(8) nucleated cells/mL is optimal for tumor cell purging. Furthermore, this appeared to be a safe procedure, with total loss of clonogenic growth of breast cancer cells as well as no significant effect on progenitor cell function as determined by granulocyte-macrophage colony-forming unit assays.
- Published
- 2000
- Full Text
- View/download PDF
34. Ethanol improves adenovirus-mediated gene transfer and expression to the bladder epithelium of rodents.
- Author
-
Engler H, Anderson SC, Machemer TR, Philopena JM, Connor RJ, Wen SF, and Maneval DC
- Subjects
- Animals, Female, Rats, Rats, Sprague-Dawley, Urothelium drug effects, Adenoviridae, Ethanol pharmacology, Gene Transfer Techniques, Solvents pharmacology, Urinary Bladder drug effects
- Abstract
Objectives: Replication deficient adenoviral vectors (rAds) are used as gene delivery systems that can efficiently transduce a variety of tissues and may be appropriate vectors to develop gene therapeutics for many urologic applications. However, the bladder epithelium has been shown to be highly resistant to transgene expression after intracystic administration. A potential explanation for this low gene transfer efficiency may be the protective structure of the urothelium. Since this protective barrier can be disrupted by organic solvents, we assessed whether ethanol co-administration can enhance adenovirus-mediated transgene expression., Methods: Normal and bladder tumor-bearing rats received a single intracystic administration of rAd encoding beta-galactosidase (rAd-beta gal) or p53 (rAd-p53). rAd was administered in a saline solution or in solutions with increasing concentrations of ethanol. Transgene expression was evaluated in the bladder tissues., Results: A dramatic increase in urothelial beta-galactosidase transgene expression was achieved by rAd-beta gal administered in a 22% ethanol solution. Transgene expression was enhanced in normal urothelium and in superficial bladder tumors. p53 transgene expression was similarly enhanced., Conclusions: Co-administration of 22% ethanol enhanced local rAd-mediated transgene expression in the normal and neoplastic bladder epithelium in rodents. Improvement of rAd-mediated transgene expression is progress toward local gene delivery to the urothelium and may enable local gene therapy for superficial bladder cancer or other bladder diseases.
- Published
- 1999
- Full Text
- View/download PDF
35. Adenovirus p53 purging for human breast cancer stem cell products.
- Author
-
Hirai M, Kelsey LS, Maneval DC, Vaillancourt M, and Talmadge JE
- Subjects
- Antineoplastic Agents therapeutic use, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Division, Humans, Kinetics, Temperature, Tumor Cells, Cultured, Adenoviridae genetics, Bone Marrow Purging methods, Bone Marrow Transplantation, Breast Neoplasms therapy, Genes, p53
- Abstract
Tumor cell (TC) contamination of stem cell products can contribute to relapse after high dose chemotherapy and stem cell rescue. A new purging technology using replication-deficient recombinant adenovirus (Adv) containing the p53 tumor suppressor gene (Adv-p53) has been suggested to reduce tumor contamination of autologous stem cell product. We demonstrate herein a safe and effective Adv-p53 purging procedure using four human breast cancer TC lines. Multiple parameters need to be achieved to successfully purge stem cell products, including a high cell:virus ratio, a small incubation volume, a long incubation time and 37 degrees C rather than room temperature. These parameters are all interrelated and equally important for the inhibition of TC clonogenic growth. In our studies, we also observed that Adv could nonspecifically inhibit TC clonogenic growth, although Adv-p53 treatment led to a significantly greater inhibition of clonogenic growth by cells expressing mutated p53. The presence of peripheral stem cell (PSC) products was found to decrease the effect of Adv-p53 on TC clonogenic growth, suggesting that PSC products could compete with TC for infection by recombinant Adv. However, X-Gal staining after incubation with Adv containing-galactosidase demonstrated that PSC products were 2, 000-fold more resistant to Adv infection than TC. We conclude that a 4-hour incubation of stem cell products (2 x 10(8)/ml) with 4 x 10(11) Adv-p53 particles is sufficient to completely purge TC with no effect on hematopoietic cell function.
- Published
- 1999
- Full Text
- View/download PDF
36. A recombinant adenoviral vector expressing full-length human retinoblastoma susceptibility gene inhibits human tumor cell growth.
- Author
-
Demers GW, Harris MP, Wen SF, Engler H, Nielsen LL, and Maneval DC
- Subjects
- Animals, Cell Division drug effects, Cell Division genetics, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, G1 Phase drug effects, G1 Phase genetics, Genetic Vectors genetics, Humans, L-Lactate Dehydrogenase drug effects, L-Lactate Dehydrogenase metabolism, Mice, Mice, Nude, Neoplasms, Experimental drug therapy, Neoplasms, Experimental genetics, Recombinant Proteins genetics, Recombinant Proteins metabolism, Recombinant Proteins pharmacology, Retinoblastoma metabolism, Tumor Cells, Cultured drug effects, Adenoviridae genetics, Antineoplastic Agents pharmacology, Genetic Therapy methods, Genetic Vectors pharmacology, Retinoblastoma genetics
- Abstract
As a prelude to considering retinoblastoma (RB) gene therapy for cancer, a series of human tumor cell lines with either full-length, mutated, or undetectable RB protein were treated with recombinant adenovirus encoding RB (ACNRB). Both RB protein expression and the cytotoxic and antiproliferative effects of ACNRB treatment were evaluated. While the transgene expression of a reporter virus encoding the beta-galactosidase enzyme (rAd-beta-gal) varied among cell lines, the reintroduction and expression of the RB gene resulted in a pronounced inhibition of cellular proliferation in RB-altered cell lines. An antiproliferative response was observed with control adenovirus treatment in some cell lines. ACNRB treatment did not cause detectable cytotoxicity in either RB+ or RB-altered cells. Dose-dependent cytostasis was observed in RB- cell lines. In vivo tumor suppression was observed in a breast xenograft model subsequent to the treatment of established tumors with ACNRB. These data support a role for RB gene therapy of tumors with RB mutations and provide a basis for the further evaluation of ACNRB gene therapy of human cancer.
- Published
- 1998
37. p53 gene therapy in a rat model of hepatocellular carcinoma: intra-arterial delivery of a recombinant adenovirus.
- Author
-
Anderson SC, Johnson DE, Harris MP, Engler H, Hancock W, Huang WM, Wills KN, Gregory RJ, Sutjipto S, Wen SF, Lofgren S, Shepard HM, and Maneval DC
- Subjects
- Animals, Apoptosis, Carcinoma, Hepatocellular metabolism, Defective Viruses, Female, Gene Expression, Hepatic Artery, Humans, Infusions, Intra-Arterial, Infusions, Intravenous, Liver Neoplasms, Experimental metabolism, Neoplasm Proteins genetics, Rats, Transgenes, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Adenoviridae, Carcinoma, Hepatocellular therapy, Genes, p53, Genetic Therapy methods, Genetic Vectors administration & dosage, Liver Neoplasms, Experimental therapy, Neoplasm Proteins metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
p53 tumor suppressor gene therapy has been proposed for cancers characterized by inactivation of p53 function, and successful therapy will require efficient strategies for gene delivery. To maximize transgene expression in tumors, a clinical strategy has been proposed to treat neoplasms in the liver via hepatic artery administration of a recombinant adenovirus encoding wild-type p53 (rAd-p53). We have developed a syngeneic rat model using a p53mut hepatocellular carcinoma cell line (McA-RH7777) that results in multifocal liver tumor nodules to provide experimental support for this strategy. Treatment of McA-RH7777 cells with rAd-p53 in vitro resulted in efficient transgene expression, growth suppression, and apoptosis. Intrahepatic artery dosing with rAd-p53 or an adenovirus encoding beta-galactosidase (rAd-betagal) increased transgene expression in tumor tissue and decreased systemic exposure when compared with i.v. dosing. Daily hepatic artery dosing of rAd-p53 suppressed tumor growth when compared with untreated rats or animals treated with rAd-betagal. These data demonstrate the potential for arterial gene delivery to tumors using recombinant adenoviruses, and support continued investigation of rAd-p53 gene therapy for liver malignancies.
- Published
- 1998
38. Enhancement of adenovirus-mediated gene transfer to human bone marrow cells.
- Author
-
Watanabe T, Kelsey L, Ageitos A, Kuszynski C, Ino K, Heimann DG, Varney MT, Shepard HM, Vaillancourt MT, Maneval DC, and Talmadge JE
- Subjects
- Adenoviruses, Human physiology, Cells, Cultured, Colony-Forming Units Assay, Culture Media, Genes, Reporter, Genes, p53, Hematopoiesis, Hematopoietic Cell Growth Factors pharmacology, Hematopoietic Stem Cells metabolism, Humans, Safety, beta-Galactosidase genetics, Adenoviruses, Human genetics, Bone Marrow Purging methods, Gene Transfer Techniques, Genetic Vectors genetics, Hematopoietic Stem Cells virology, Transfection methods
- Abstract
Adenovirus infection of CD34+ hematopoietic stem/progenitor cells is dependent on the multiplicity of infection (MOI), time of incubation, the volume in which the co-incubation occurs and the presence or absence of growth factors. Studies revealed that a brief co-incubation (1-8 hours), resulted in low levels of transgene expression, suggesting that adenovirus infection of CD34+ cells occurs slowly, and optimal transduction requires a 24 hour exposure to adenovirus. Infection by Ad/beta-gal or Ad/p53 at a MOI of 500:1 provided a high transduction efficiency but inhibited hematopoietic function. However, treatment at a MOI of 50-100 resulted in efficient transduction (10.7-15.7% positive) without detectable toxicity. Secondary proof of adenovirus transgene expression was demonstrated by detection of mRNA for p53 in Ad/p53 infected stem cells. We conclude that a 24 hour exposure to recombinant adenovirus encoding p53 or beta-gal, at a MOI of 50-100 is optimal for in vitro gene transfer to BM cells and has no significant effect on hematopoietic function. Adenovirus-mediated transduction of BM cells can also be modulated by growth factors (IL-3, GM-CSF and G-CSF) with improved gene delivery and maintenance of hematopoietic function. In summary, adenovirus vectors can be used to transiently transduce stem cells, and conditions have been defined to maximize expression and limit inhibitory effects on CD34+ cells. These data support continued investigation of this vector for local cytokine delivery and purging of stem cell products.
- Published
- 1998
- Full Text
- View/download PDF
39. P53 tumor suppressor gene therapy for cancer.
- Author
-
Nielsen LL and Maneval DC
- Subjects
- Combined Modality Therapy, Genetic Vectors, Humans, Neoplasms genetics, Genes, p53, Genetic Therapy, Neoplasms therapy
- Abstract
The last two decades have led to a greater understanding of the genetic basis of human malignancy. Although numerous genetic alterations have been detected in cancer, activation of oncogenes and inactivation of cell cycle regulators (e.g., tumor suppressor genes) are now known to play a critical role in the progression of the disease. Therapeutic strategies based on specific molecular alterations in cancer include reintroduction of wild-type tumor suppressor function to cells lacking the gene. p53 gene therapy provides an attractive strategy to test the potential clinical feasibility of this approach. Alterations in p53 function are present in approximately half of all malignancies, and expression of wild-type p53 can result in apoptosis in human tumor cells. This review summarizes current investigations with p53 gene therapy, highlighting the preclinical efforts with adenoviral, retroviral, and lipid-based gene delivery systems. A comprehensive review of the various clinical targets suggested for p53 gene therapy is presented together with challenges and prospects for future clinical investigation.
- Published
- 1998
40. p53-mediated accumulation of hypophosphorylated pRb after the G1 restriction point fails to halt cell cycle progression.
- Author
-
Linke SP, Harris MP, Neugebauer SE, Clarkin KC, Shepard HM, Maneval DC, and Wahl GM
- Subjects
- Carcinoma, Non-Small-Cell Lung, Cell Cycle radiation effects, Cyclin-Dependent Kinase Inhibitor p21, Cyclins metabolism, Diploidy, Enzyme Inhibitors metabolism, Fibroblasts, G1 Phase, Gamma Rays, Humans, Lung Neoplasms, Oncogene Proteins, Viral biosynthesis, Papillomaviridae genetics, Phosphorylation, Recombinant Proteins biosynthesis, Repressor Proteins biosynthesis, S Phase, Tumor Cells, Cultured, Tumor Suppressor Protein p53 biosynthesis, Cell Cycle physiology, Cyclin-Dependent Kinases antagonists & inhibitors, Cyclins biosynthesis, Retinoblastoma Protein metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
This study analyses whether the inability of p53 to induce G1 arrest after the restriction point relates to an inability to modulate pRb phosphorylation. Transient p53 overexpression in normal human diploid fibroblasts and p53-deficient cancer cells led to increased levels of the cyclin-dependent kinase inhibitor p21 cip1/Waf1/Sdi1 and an accumulation of hypophosphorylated pRb in cells growing asynchronously and in cells synchronized in late G1 or M. Similarly, gamma-irradiation of asynchronous, late-G1, or S phase fibroblasts led to an increase in hypophosphorylated pRb. Experiments with fibroblasts expressing the HPV16 E6 protein indicated that accumulation of hypophosphorylated pRb required functional p53. Progression into and through S phase was not altered by the presence of hypophosphorylated pRb in late G1, consistent with the failure of p53 to mediate G1 arrest in cells that are past the restriction point. These data indicate that accumulation of hypophosphorylated pRb has significantly different effects on cell cycle progression in early G1 versus late G1 or S phase.
- Published
- 1997
- Full Text
- View/download PDF
41. Adenovirus-mediated p53 gene transfer suppresses growth of human glioblastoma cells in vitro and in vivo.
- Author
-
Köck H, Harris MP, Anderson SC, Machemer T, Hancock W, Sutjipto S, Wills KN, Gregory RJ, Shepard HM, Westphal M, and Maneval DC
- Subjects
- Adenoviridae genetics, Animals, Apoptosis, Cell Division genetics, Glioblastoma genetics, Glioblastoma metabolism, Glioblastoma pathology, Humans, L-Lactate Dehydrogenase metabolism, Mice, Mice, Inbred BALB C, Mice, Nude, Rats, Tumor Cells, Cultured, Gene Transfer Techniques, Genes, p53 genetics, Genetic Therapy, Glioblastoma therapy
- Abstract
Alterations in the p53 tumor-suppressor gene occur in 35-60% of human glioblastomas, and re-introduction of p53 can suppress neoplastic growth. To evaluate the potential for p53 gene therapy of glioblastoma, we have analyzed the response of human glioblastoma cell lines in vitro and in vivo to experimental therapy with replication-deficient recombinant adenoviruses encoding wild-type p53 (rAd-p53). Western blot analyses showed high-level expression of p53 protein after treatment with rAd-p53, and transgene expression was dependent on promoter strength. A p53-specific dose-dependent inhibition of in vitro cellular proliferation was observed in 5 of 6 cell lines, and growth inhibition corresponded to adenovirus-mediated gene transfer and expression. p53-specific cell death was quantitated by release of the lactate dehydrogenase enzyme. Fragmentation of DNA into nucleosomal oligomers and the occurrence of a hypodiploid cell population detected by flow cytometry provided evidence for apoptosis. Studies in nude mice demonstrated that ex vivo infection with rAd-p53 suppressed the tumorigenic potential of human glioblastoma cells. Furthermore, direct injection of rAd-p53 into established s.c. xenografts inhibited tumor growth. Our observations suggest that re-introduction of wild-type p53 may have potential clinical utility for gene therapy of glioblastoma.
- Published
- 1996
- Full Text
- View/download PDF
42. Adenoviral-mediated p53 tumor suppressor gene therapy of human ovarian carcinoma.
- Author
-
Mujoo K, Maneval DC, Anderson SC, and Gutterman JU
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma virology, Animals, Carcinogenicity Tests, Female, Genetic Vectors, Humans, Mice, Mice, Nude, Ovarian Neoplasms genetics, Ovarian Neoplasms virology, Transduction, Genetic, Tumor Cells, Cultured, Adenocarcinoma therapy, Adenoviridae genetics, Genes, p53, Genetic Therapy, Ovarian Neoplasms therapy
- Abstract
Mutations of p53 gene are reported in 50-60% of human cancers and reintroduction of wild-type p53 can suppress cell proliferation. In this study, replication deficient recombinant adenovirus encoding wild-type p53 (ACN53) under the control of the human cytomegalovirus (CMV) promoter was constructed. A specific incorporation of the p53 gene with ACN53 reduced 3 (deleted p53 gene) cells was observed. ACN53 reduced the colony-forming ability of SK-OV-3 cells 72-216 h after single infection. A highly aggressive ovarian xenograft model was established in which animals die between 25-45 days. A localization study with the adenovirus-containing beta galactosidase reporter gene showed effective gene transfer in the tumor tissues. Ex vivo treatment of SK-OV-3 cells with ACN53 followed by injection into nude mice, increased the survival of the p53 treated mice by more than 50% compared with control animals. Gene therapy with ACN53 in intraperitoneal model of SK-OV-3 cells in two independent experiments revealed that there were some long-term survivors in the group of mice [2/5 (66 and 120 days) and [2/8 (166 and 423 days)] treated with ACN53. These findings demonstrate the potential of the p53 tumor suppressor gene therapy in human ovarian carcinoma.
- Published
- 1996
43. Adenovirus-mediated p53 gene transfer inhibits growth of human tumor cells expressing mutant p53 protein.
- Author
-
Harris MP, Sutjipto S, Wills KN, Hancock W, Cornell D, Johnson DE, Gregory RJ, Shepard HM, and Maneval DC
- Subjects
- Animals, Cell Division drug effects, Cell Division genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms physiopathology, Female, Gene Transfer Techniques, Genetic Vectors, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Mutation, Recombination, Genetic, Survival Rate, Transduction, Genetic, Tumor Cells, Cultured, Adenoviridae genetics, Colorectal Neoplasms therapy, Genes, p53 genetics, Genetic Therapy methods, Tumor Suppressor Protein p53 biosynthesis
- Abstract
Human malignancies are often characterized by mutations of the p53 tumor suppressor gene. In a large proportion of cases, the mutation results in production of an altered protein that can bind and inactivate the wild-type gene product. This "dominant-negative" activity of mutant p53 molecules may limit the utility of p53 gene therapy of cancer. Using replication-deficient recombinant adenoviruses (rAd-p53) as a p53 gene delivery system, we evaluated the effects of p53 reintroduction on a series of 45 human cell lines containing wild-type, mutated, or no p53 protein. Results indicate a p53-specific, dose-dependent, and promoter-specific growth inhibition of a majority of p53-altered cell lines that correlates with the degree of adenovirus transgene expression. Similar effects were not observed on cells containing wild-type p53. rAd-p53 inhibited the growth of cells expressing various mutant p53 proteins including those characterized as "dominant negative mutants", and the antiproliferative effects were not abrogated by high levels of endogenous mutated p53 protein. In vivo, rAd-p53 also suppressed tumor growth and increased survival of nude mice bearing tumors that express mutant p53. These results support a role for p53 gene therapy of cancer, including malignancies harboring mutations in this tumor suppressor gene.
- Published
- 1996
44. Gene therapy for hepatocellular carcinoma: chemosensitivity conferred by adenovirus-mediated transfer of the HSV-1 thymidine kinase gene.
- Author
-
Wills KN, Huang WM, Harris MP, Machemer T, Maneval DC, and Gregory RJ
- Subjects
- Animals, Carcinoma, Hepatocellular genetics, Cell Division drug effects, Ganciclovir pharmacology, Genetic Vectors pharmacology, Humans, Mice, Neoplasms, Experimental drug therapy, Neoplasms, Experimental genetics, Thymidine Kinase pharmacology, alpha-Fetoproteins biosynthesis, alpha-Fetoproteins genetics, Carcinoma, Hepatocellular therapy, Drug Resistance, Neoplasm genetics, Genetic Therapy, Herpesvirus 1, Human genetics, Thymidine Kinase genetics
- Abstract
We have investigated whether adenovirally mediated gene transfer of the herpes simplex thymidine kinase gene to human hepatocellular carcinoma (HCC) cell lines can sensitize these cells to the prodrug ganciclovir and thereby provide a therapeutic option for this intractable cancer. Two replication-deficient adenoviruses encoding for the herpes simplex virus type-1 (HSV) thymidine kinase (TK) gene were generated in which expression of TK is under the control of either the human cytomegalovirus immediate early promoter (CMV) or the human alpha-fetoprotein (AFP) promoter/enhancer. We demonstrate that the combination of adenovirally mediated TK gene transfer and ganciclovir treatment effectively inhibits proliferation and causes cell death of HCC cells in vitro and that in vivo TK gene transfer and ganciclovir treatment inhibits hepatocellular tumor growth in a mouse model of this cancer. Furthermore, we show that expression of the TK gene can be restricted to those HCCs that express the tumor marker AFP through the incorporation of the AFP enhancer/promoter within an adenoviral vector.
- Published
- 1995
45. Recombinant adenovirus-mediated gene transfer to genitourinary epithelium in vitro and in vivo.
- Author
-
Bass C, Cabrera G, Elgavish A, Robert B, Siegal GP, Anderson SC, Maneval DC, and Curiel DT
- Subjects
- Animals, Carcinoma, Transitional Cell genetics, Carcinoma, Transitional Cell pathology, Epithelium metabolism, Epithelium virology, Female, Genes, Reporter, Humans, Luciferases biosynthesis, Luciferases genetics, Male, Mice, Mice, Inbred BALB C, Rats, Rats, Sprague-Dawley, Transfection, Tumor Cells, Cultured, Urinary Bladder metabolism, Urinary Bladder virology, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms pathology, beta-Galactosidase biosynthesis, beta-Galactosidase genetics, Adenoviruses, Human genetics, Carcinoma, Transitional Cell therapy, Genetic Therapy methods, Genetic Vectors, Recombinant Fusion Proteins biosynthesis, Urinary Bladder pathology, Urinary Bladder Neoplasms therapy
- Abstract
Transitional cell carcinoma (TCC) of the bladder is associated with characterized lesions in dominant and recessive oncogenes. The understanding of the molecular basis of tumorigenesis in these instances makes possible the application of gene therapy strategies for TCC. In this regard, the ability to directly access the epithelium of the genitourinary (GU) tract via the urethra provides a practical means to implement these various gene therapy approaches. We thus explored vector strategies to accomplish direct in vivo transduction of GU epithelium. Initially, three human (HT 1197, HT 1376, T24) and one mouse (MBT-2) TCC cell lines were transduced using a recombinant adenoviral vector expressing the firefly luciferase reporter gene, rAd-CMV-Luc. In these studies, reporter gene expression was found to be significantly elevated above background for all four cell lines. Of note, the TCC cell lines HT 1197 and HT 1376 showed expression levels comparable with the cervical carcinoma cell line HeLa, a cell line previously shown to be highly susceptible to recombinant adenovirus-mediated gene transduction. An in vitro time course for T24 and MBT-2 using rAd-CMV-Luc showed peak expression 1 day after transduction for the T24 line and 3 days after transduction for the MBT-2 line, with detectable levels of expression persisting for at least 7 days. As a next step, human and mouse primary tissue deriving from the GU epithelium were transduced using rAd-CMV-Luc. In this assay, luciferase expression levels significantly above background were observed in both instances.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1995
46. Development and characterization of recombinant adenoviruses encoding human p53 for gene therapy of cancer.
- Author
-
Wills KN, Maneval DC, Menzel P, Harris MP, Sutjipto S, Vaillancourt MT, Huang WM, Johnson DE, Anderson SC, and Wen SF
- Subjects
- Animals, Base Sequence, Carcinoma, Small Cell pathology, Cytomegalovirus genetics, DNA Replication, DNA, Complementary genetics, DNA, Recombinant genetics, DNA, Viral genetics, Female, Humans, Lung Neoplasms pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Molecular Sequence Data, Neoplasm Transplantation, Neoplasms genetics, Promoter Regions, Genetic, Recombinant Fusion Proteins, Transplantation, Heterologous, Tumor Cells, Cultured, Tumor Suppressor Protein p53 deficiency, Tumor Suppressor Protein p53 genetics, Adenoviruses, Human genetics, Carcinoma, Small Cell therapy, Defective Viruses genetics, Genes, p53, Genetic Therapy, Genetic Vectors, Neoplasms therapy
- Abstract
We have constructed recombinant human adenoviruses that express wild-type human p53 under the control of either the Ad 2 major late promoter (MLP) or the human cytomegalovirus (CMV) immediate early gene promoter. Each construct replaces the Ad 5 E1a and E1b coding sequences necessary for viral replication with the p53 cDNA and MLP or CMV promoter. These p53/Ad recombinants are able to express p53 protein in a dose-dependent manner in infected human cancer cells. Tumor suppressor activity of the expressed p53 protein was assayed by several methods. [3H]Thymidine incorporation assays showed that the recombinant adenoviruses were capable of inhibiting DNA synthesis in a p53-specific, dose-dependent fashion. Ex vivo treatment of Saos-2 tumor cells, followed by injection of the treated cells into nude mice, led to complete tumor suppression using the MLP/p53 recombinant. Following a single injection of CMV/p53 recombinant adenovirus into the peritumoral space surrounding an in vivo established tumor derived from a human small cell lung carcinoma cell line (NIH-H69), we were able to detect p53 mRNA in the tumors at 2 and 7 days post-injection. Continued treatment of established H69 tumors with MLP/p53 recombinant led to reduced tumor growth and increased survival time compared to control treated animals. These results indicate that recombinant adenoviruses expressing wild-type p53 may be useful vectors for gene therapy of human cancer.
- Published
- 1994
- Full Text
- View/download PDF
47. Measurement of Tc-99m DTPA serum clearance for estimating glomerular filtration rate in children with cancer.
- Author
-
Rodman JH, Maneval DC, Magill HL, and Sunderland M
- Subjects
- Adolescent, Child, Child, Preschool, Female, Humans, Male, Glomerular Filtration Rate, Neoplasms physiopathology, Technetium Tc 99m Pentetate blood
- Abstract
Clearance of the radiopharmaceutical Tc-99m DTPA estimated from blood samples with no urine collection can provide a reliable estimate of glomerular filtration rate (GFR) in adults, but has not been well studied in children. The disposition of Tc-99m DTPA was determined in 17 children with cancer, and the influence of binding and study design on the estimates for serum clearance were evaluated. Nine blood samples were obtained over 6 hours in each patient, and serum was assayed for total and free Tc-99m. Free Tc-99m DTPA was determined by ultrafiltration. Estimates of clearance derived from a two-compartment model for ultrafiltrable Tc-99m DTPA were determined from all nine measured concentrations, and these results served as a reference value for GFR in each subject. Total Tc-99m DTPA concentrations also were best described by a two-compartment model, but the median total clearance (35 ml/min) was significantly (p < 0.01) lower than the ultrafiltrate clearance (58 ml/min). The effect of a simplified sampling schedule was assessed from clearance estimates based on a 3-point subset of the ultrafiltrable data. The median clearance of 69 ml/minute was significantly higher (p < 0.01) than the reference GFR. However with a correction factor included to account for the positive bias arising from the limited sampling schedule, the reference estimates for GFR were well predicted (r2 = 0.99) with no significant bias. Ultrafiltrable Tc-99m DTPA serum clearance using a limited sampling schedule can provide a consistent and clinically feasible estimate of glomerular filtration rate in children, but binding in serum and study design are potentially important confounding factors.
- Published
- 1993
48. Relationship between dose rate of [6RS]Leucovorin administration, plasma concentrations of reduced folates, and pools of 5,10-methylenetetrahydrofolates and tetrahydrofolates in human colon adenocarcinoma xenografts.
- Author
-
Houghton JA, Williams LG, de Graaf SS, Cheshire PJ, Rodman JH, Maneval DC, Wainer IW, Jadaud P, and Houghton PJ
- Subjects
- Adenocarcinoma blood, Animals, Colonic Neoplasms blood, Female, Floxuridine administration & dosage, Floxuridine blood, Floxuridine pharmacology, Half-Life, Humans, Injections, Intravenous, Leucovorin administration & dosage, Leucovorin blood, Mice, Mice, Inbred CBA, Thymidylate Synthase blood, Time Factors, Adenocarcinoma metabolism, Colonic Neoplasms metabolism, Floxuridine metabolism, Leucovorin pharmacology, Tetrahydrofolates blood, Thymidylate Synthase antagonists & inhibitors
- Abstract
[6RS]Leucovorin (5-formyltetrahydrofolate; 5-CHO-H4PteGlu) administered in different regimens in combination with 5-fluorouracil (FUra) has increased the response rates to FUra in patients with colon adenocarcinoma. Using preclinical models of human colon adenocarcinomas as xenografts in immune-deprived mice, the effect of the rate of administration of racemic [6RS]leucovorin on the concentration-time profile of reduced folates in plasma, size of intratumor pools of 5,10-methylenetetrahydrofolates (CH2-H4PteGlun) and tetrahydrofolates (H4PteGlun), and the distribution of their polyglutamate species have been examined. Bolus injection i.v., or 4-h or 24-h infusion of [6RS]leucovorin (500 mg/m2) yielded similar concentration profiles of the biologically active [6S] and inactive [6R] isomers of 5-CHO-H4-PteGlu and 5-methyltetrahydrofolate (5-CH3-H4PteGlu) in mouse plasma to those previously reported in humans, but with more rapid elimination half-lives (t1/2 = 11 to 16 min, 23 to 41 min, and 30 to 35 min, respectively). Thus, reduced folates remained elevated in plasma during the period of [6RS]leucovorin administration. In HxELC2 and HxGC3 tumors, pools of CH2-H4PteGlun and H4PteGlun were increased from 350% to 700% of control, but only during [6RS]leucovorin infusion. Intracellular levels subsequently declined rapidly, similar to the loss of reduced folates from plasma. Increasing the rate of [6RS]leucovorin delivery by decreasing the time for administration from a 24-h to a 4-h infusion did not further increase the intratumor pools of CH2-H4PteGlun and H4PteGlun, suggesting saturation in the cellular metabolism of [6RS]leucovorin. In HxGC3 tumors, CH2-H4PteGlu4-5 were elevated more rapidly than in line HxELC2, which accumulated predominantly a shorter chain length species following i.v. bolus injection. During the 4-h infusion schedule, di- and triglutamate species in particular accumulated in both tumors with no elevation in CH2-H4PteGlu5 until the infusion was discontinued, when this species increased as the shorter chain length forms were declining. However, during the 24-h infusion of [6RS]leucovorin, CH2-H4PteGlu3-5 were elevated in tumors. Since these species have been reported to increase the binding affinity of [6-3H]5-fluorodeoxyuridine monophosphate ([6-3H]FdUMP) to thymidylate synthase, and intratumor pools of CH2-H4PteGlun and H4PteGlun were elevated during the 24-h infusion of [6RS]leucovorin, this was considered to be the preferred schedule for administration.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1990
49. Measurement of skin-to-kidney distance in children: implications for quantitative renography.
- Author
-
Maneval DC, Magill HL, Cypess AM, and Rodman JH
- Subjects
- Body Constitution, Child, Female, Humans, Kidney diagnostic imaging, Male, Reference Values, Skin, Tomography, X-Ray Computed, Kidney anatomy & histology, Radioisotope Renography
- Abstract
Variation in skin-to-kidney center distance has been shown to have a significant influence on quantification of renal function with the gamma camera. Several techniques to compensate for this variability have been proposed in adults, yet it has been suggested that depth correction is not necessary for quantitative renography in children. Skin-to-kidney center distances were measured from computed tomograms in 53 supine pediatric patients. Nearly 40% of the kidneys examined varied more than 1 cm from the average renal depth, and 8% deviated more than 2 cm. Right kidney depth differed from left kidney depth by more than 1 cm in less than 10% of the patients. Measurements were in agreement with regression equations based on lateral scintigraphy in children, but were consistently underestimated by nomograms developed for skin-to-kidney center distance in adults. Failure to recognize interindividual variability in skin-to-kidney center distance can introduce significant errors in quantitative pediatric renography.
- Published
- 1990
50. A kinetic model for 99mTc-DMSA in the rat.
- Author
-
Maneval DC, D'Argenio DZ, and Wolf W
- Subjects
- Animals, Female, Injections, Intravenous, Kidney diagnostic imaging, Organotechnetium Compounds administration & dosage, Radionuclide Imaging, Rats, Rats, Inbred Strains, Succimer administration & dosage, Technetium Tc 99m Dimercaptosuccinic Acid, Tissue Distribution, Kidney metabolism, Organotechnetium Compounds pharmacokinetics, Succimer pharmacokinetics, Sulfhydryl Compounds pharmacokinetics
- Abstract
A pharmacokinetic model was developed for the renal imaging agent 99mTc-DMSA in anesthetized rats, which incorporated data from serial measurements of blood and urine simultaneously with dynamic images obtained over an 8-h period. Animals which received a 10 mg/kg dose of unlabeled DMSA immediately before 99mTc-DMSA injection had a significantly reduced kidney accumulation and greater urinary elimination of 99mTc than animals which received the radiopharmaceutical alone. The kidney clearance was also significantly lower in rats receiving unlabeled DMSA, but no significant difference was determined between the urine clearance estimates of the two animal groups. Because the increase in the amount eliminated in the urine was not coupled with a significant change in urine clearance, it would appear that unlabeled DMSA saturated the kidney uptake mechanism(s) of 99mTc-DMSA without modifying the urinary clearance process. This interpretation is consistent with the hypothesis that renal handling of 99mTc-DMSA is governed by both glomerular filtration and peritubular capillary uptake. The simultaneous acquisition of blood, urine and non invasive image data allows for a comprehensive and informative model of the physiological disposition of 99mTc-DMSA.
- Published
- 1990
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.