Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
2.
Int J Pharm ; 191(1): 15-24, 1999 Nov 25.
Article in English | MEDLINE | ID: mdl-10556736

ABSTRACT

Sodium salts of medium-chain fatty acids, sodium caprate (C10) in particular, have been used as absorption-enhancing agents to promote transmucosal drug absorption. In this study, we conducted both in vitro and in vivo experiments to investigate the effects of C10 on intestinal permeabilities and mucosal morphology. Mucosal addition of C10 (13-25 mM) reduced the transepithelial electric resistance (TEER) of cultured monolayers of the human intestinal cell line Caco-2 by 40-65% and, upon removal of C10, a marked tendency of TEER recovery was recorded. C10 added mucosally at 13-50 mM increased the transports of mannitol and polyethylene glycol (PEG) 900 across Caco-2 in a dose-dependent manner. In contrast, the transport of a model D-decapeptide was maximally enhanced with 20-25 mM C10. No noticeable morphological alteration of the Caco-2 monolayers was observed after a 1-h mucosal pretreatment with C10. Co-delivery with C10 (0.05-0.5 mmol/kg) into the rat terminal ileum increased the D-decapeptide bioavailability (BA) dose-dependently. With 0.5 mmol/kg C10 co-administered, D-decapeptide percent BA was elevated from 2 to 11%. Following a 1-h incubation with 0.5 mmol/kg C10 (in liquid or powder form) non-invasively delivered into the rectal lumen, no signs of histological change in the rectal mucosa were detected. These results demonstrate that C10 can promote intestinal absorption of a small peptide without causing detrimental alterations of the intestinal mucosa. C10 thus seems to be a good candidate as an enhancing agent for improving the oral BA of small therapeutic peptides.


Subject(s)
Decanoic Acids/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Peptides/pharmacokinetics , Animals , Biological Availability , Biomarkers , Caco-2 Cells , Electrophysiology , Epithelium/physiology , Humans , Intestinal Mucosa/anatomy & histology , Male , Rats , Rats, Sprague-Dawley , Rectum/anatomy & histology , Rectum/drug effects
3.
Curr Opin Drug Discov Devel ; 2(2): 168-71, 1999 Mar.
Article in English | MEDLINE | ID: mdl-19649943

ABSTRACT

Controlled delivery of macromolecular drugs, such as peptides, proteins, oligonucleotides and polysaccharides remains a major challenge. While these drugs can be administered by subcutaneous injection or intravenous infusion, more convenient, painless drug delivery approaches are being developed. This mini-review will highlight the most recent innovations and trends in macromolecular drug delivery.

4.
J Pharm Sci ; 87(11): 1395-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9811496

ABSTRACT

DS-1, a modified Quillaja saponin, has recently been shown to promote the absorption of insulin and aminoglycoside antibiotics via the ocular and nasal route. The purpose of this study is to investigate the effect of DS-1 on intestinal permeability, the mechanism of its action, and reversibility of the effect. The permeation-enhancing activity of DS-1 was evaluated in cultured monolayers of the Caco-2 intestinal epithelial cells by examining its effect on the transepithelial electric resistance (TEER) and on transport of mannitol and a model D-decapeptide. Mucosal addition of DS-1 promptly reduced the TEER of the Caco-2 monolayers, and a propensity of recovery of the TEER was observed upon its removal. DS-1 added at 0.01-0.1% (w/v) increased the transports of both mannitol and D-decapeptide in a dose-dependent manner; a relatively "flat" concentration-dependence was seen at 0.1-0.2%. Visualization studies conducted by confocal laser scanning microscopy (CLSM) seem to suggest that DS-1 enhances the Caco-2 permeability mainly via a transcellular route. Histological examination failed to reveal noticeable morphological alterations in the cell monolayers pretreated with DS-1. The integrity of the Caco-2 monolayers, as assessed by their permeability to mannitol, was found to be recoverable following the mucosal pretreatment of DS-1. These results suggest that DS-1 is an efficacious intestinal permeation-enhancing agent with low adverse effect on the epithelial viability and barrier function.


Subject(s)
Intestines/drug effects , Saponins/pharmacology , Caco-2 Cells , Electrophysiology , Humans , Intestinal Absorption , Intestinal Mucosa/metabolism , Mannitol/metabolism , Quillaja
5.
J Drug Target ; 6(1): 37-43, 1998.
Article in English | MEDLINE | ID: mdl-9769019

ABSTRACT

Long-chain acylcarnitines, such as palmitoylcarnitine chloride (PCC), are endogenous compounds which have been shown to increase intestinal transport of small hydrophilic compounds (including some pharmaceutical agents) through the paracellular pathway. However, the size range of the compounds whose absorption can be improved by PCC has not been fully investigated. In the present study, we systematically examined the effect of PCC on the transport rate of a series of hydrophilic fluorescent model compounds of varying molecular weights (0.3-71.2 kD) across cultured monolayers of the human intestinal epithelial cells Caco-2. Mucosal addition of 100 or 200 microM PCC resulted in comparable time-dependent decreases in the transepithelial electric resistance (T1/2, approximately 15 min). PCC addition induced a striking increase in the transport of sodium fluorescein (Flu-Na; 0.3 kD) and a slight or moderate increase in transports of fluorescent compounds of 0.6-11 kD. The effect of PCC on transport of compounds with molecular weights of > or = 17 kD appeared to be negligible. Examination by confocal laser scanning microscopy clearly revealed dilated paracellular spaces in Caco-2 monolayers which had been mucosally pretreated with PCC, confirming that PCC increases intestinal permeability by opening a paracellular transport pathway. Our results suggest that PCC is particularly effective in enhancing intestinal absorption of small hydrophilic compound like Flu-Na and may also have limited use in promoting the transport of compounds of < or = 10 kD.


Subject(s)
Caco-2 Cells/drug effects , Caco-2 Cells/metabolism , Fluorescent Dyes/pharmacokinetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Palmitoylcarnitine/pharmacology , Cell Membrane Permeability , Humans , Molecular Weight
6.
Cytokine ; 7(1): 15-25, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7538333

ABSTRACT

The pleiotropic cytokine tumour necrosis factor-alpha (TNF) is thought to play a central role in infectious, inflammatory and autoimmune diseases. Critical to the understanding and management of TNF-associated pathology is the development of highly specific agents capable of modifying TNF activity. We evaluated the ability of a high affinity mouse/human chimeric anti-TNF monoclonal antibody (cA2) to neutralize the in vitro and in vivo biological effects of TNF. cA2 inhibited TNF-induced mitogenesis and IL-6 secretion by human fibroblasts, TNF-priming of human neutrophils, and the stimulation of human umbilical vein endothelial cells by TNF as measured by the expression of E-selectin, ICAM-1 and procoagulant activity. cA2 also specifically blocked TNF-induced adherence of human neutrophils to an endothelial cell monolayer. Receptor binding studies suggested that neutralization resulted from cA2 blocking of TNF binding to both p55 and p75 TNF receptors on the cells. In vivo, repeated administration of cA2 to transgenic mice that constitutively express human TNF reversed the cachectic phenotype and prevented subsequent mortality. These results demonstrated that cA2 effectively neutralized a broad range of TNF biological activities both in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Cachexia/prevention & control , Recombinant Fusion Proteins/immunology , Thromboplastin , Tumor Necrosis Factor-alpha/immunology , Animals , Blood Coagulation Factors/biosynthesis , Blood Coagulation Factors/genetics , Cachexia/physiopathology , Cell Adhesion/drug effects , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Division/drug effects , Cells, Cultured , E-Selectin , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Humans , Infliximab , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/metabolism , Mice , Mice, Transgenic , Neutralization Tests , Neutrophils/drug effects , Recombinant Proteins/pharmacology , Respiratory Burst/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/toxicity , Umbilical Veins
7.
J Infect Dis ; 168(6): 1429-35, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7504034

ABSTRACT

Specific binding of two monoclonal IgM antibodies previously investigated as therapeutic agents for treating gram-negative septic shock, HA-1A and E5, was assessed with respect to lipid A and lipopolysaccharide (LPS). Both antibodies bound to lipid A; however, binding of HA-1A was significantly greater than that of E5 to LPS derived from rough strains of bacteria. Reciprocal competitive inhibition experiments supported the concept that HA-1A and E5 bind to distinct epitopes on lipid A. Further, competitive inhibition studies using a monoclonal anti-idiotype antibody with specificity for the variable region of HA-1A suggested that HA-1A and E5 do not share a common idiotype. Finally, studies using double-stranded DNA as antigen indicated that E5 but not HA-1A will bind to DNA. Collectively, these data indicate that HA-1A and E5 are different lipid A-specific antibodies that bind to distinct epitopes on lipid A.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes/immunology , Immunoglobulin M/immunology , Lipid A/immunology , Lipopolysaccharides/immunology , Animals , Antibody Specificity , Binding, Competitive , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Idiotypes , Mice
8.
Cytokine ; 5(4): 348-53, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8260601

ABSTRACT

HA-1A is a human monoclonal IgM antibody which recognizes the lipid A component of lipopolysaccharide (LPS). This antibody has reduced mortality in the septic shock syndrome resulting from Gram negative bacteria, in which many of the manifestations are considered to be due to cellular activation and secretion of cytokines, most notably TNF-alpha. However HA-1A does not directly neutralize LPS effectively in vitro, and studies reported to date have not defined its mechanism of action. Here we demonstrate that HA-1A, which in the presence of complement promotes immune adherence, may inhibit LPS action by facilitating its sequestration on red blood cells and clearance to an extent that cytokine production is reduced. Incubation of LPS at clinically significant (pg/ml) does with HA-1A at therapeutic levels (e.g. 10 micrograms/ml) and complement resulted in LPS association with erythrocyte CR1 receptors. This reduced the ability of the residual, free LPS by 50-70% to induce the secretion of TNF-alpha, IL-1 beta and IL-6 from normal blood mononuclear cells. This mechanism is likely to be operative in vivo, and could account for the protective effect of HA-1A, and its reduction of TNF-alpha production in vivo.


Subject(s)
Antibodies, Monoclonal/metabolism , Antigen-Antibody Complex/metabolism , Interleukin-1/biosynthesis , Interleukin-6/biosynthesis , Leukocytes, Mononuclear/immunology , Lipid A/immunology , Lipopolysaccharides/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis , Gram-Negative Bacteria , Humans , Immunoglobulin M/metabolism , Leukocytes, Mononuclear/drug effects , Lipid A/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/immunology , RNA, Messenger/analysis , RNA, Messenger/metabolism
9.
Infect Immun ; 61(5): 1756-63, 1993 May.
Article in English | MEDLINE | ID: mdl-8478065

ABSTRACT

Clinical data suggest that the human immunoglobulin M antiendotoxin antibody HA-1A reduced mortality in patients diagnosed with gram-negative bacteremia and bacteremia with shock. Previous studies have demonstrated that HA-1A binds to the lipid A domain of lipopolysaccharide (LPS). The present study evaluated the ability of HA-1A to interact with LPs isolated from various strains of gram-negative bacteria by using liquid-phase rate nephelometry and solid-phase immunoblotting assays. HA-1A formed immune complexes in solution with LPSs isolated from both rough and smooth gram-negative organisms. Western blot (immunoblot) analysis of these LPS preparations revealed that HA-1A bound to LPS isolated from rough gram-negative organisms and to a rough LPS-like component present in smooth LPS. HA-1A also bound to LPS-protein complexes found in certain commercial rough LPS preparations. Preincubation of HA-1A with lipid A completely blocked subsequent binding of HA-1A to LPS in both liquid- and solid-phase assay formats, suggesting that the interaction of HA-1A with LPS is through the lipid A domain. Evidence that the binding of HA-1A to LPS was mediated through the antigen-combining (Fv) region of the antibody was provided by the finding that a murine anti-idiotypic antibody to HA-1A inhibited binding. These findings suggested that the broad antiendotoxin reactivity exhibited by HA-1A appeared to be due to the ability of HA-1A to bind to the conserved lipid A moiety of LPSs derived from both smooth- and rough-phenotype gram-negative bacterial strains.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Toxins/immunology , Endotoxins/immunology , Gram-Negative Bacteria/immunology , Lipopolysaccharides/immunology , Antibodies, Monoclonal/immunology , Blotting, Western , Humans , Immunoglobulin M/immunology , In Vitro Techniques , Lipid A/immunology , Nephelometry and Turbidimetry
10.
J Infect Dis ; 167(4): 865-75, 1993 Apr.
Article in English | MEDLINE | ID: mdl-8450252

ABSTRACT

HA-1A has been shown clinically to decrease mortality in septic patients with gram-negative bacteremia. In this study, the ability of HA-1A to augment the serum complement-dependent immune adherence of 125I-labeled Escherichia coli J5 lipopolysaccharide (LPS) to human erythrocytes (RBC) and polymorphonuclear leukocytes (PMNL) was evaluated. In vitro studies indicated three things: HA-1A mediates immune adherence of 125I-J5 LPS to human RBC and PMNL in a dose-dependent manner; under these conditions, high concentrations of LPS (400 ng/mL) could be specifically bound. Immune adherence occurs via the classical complement pathway as demonstrated by its calcium dependence; HA-1A-J5 LPS-C' immune complexes bound to CR1 on human RBC and PMNL. PMNL binding and internalization of immune complexes was demonstrated by trypsin stripping of externally bound immune complexes. These studies support the proposal that HA-1A can lower the bioavailability of endotoxin by mediating binding and potential clearance of LPS via human RBC through the reticuloendothelial system or via direct internalization by peripheral blood PMNL.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigen-Antibody Complex/metabolism , Complement Pathway, Classical , Endotoxins/metabolism , Erythrocytes/metabolism , Escherichia coli , Immunoglobulin M/pharmacology , Lipopolysaccharides/metabolism , Neutrophils/metabolism , Antibodies, Monoclonal, Humanized , Complement C3b/drug effects , Complement C3b/immunology , Complement C3b/metabolism , Complement C4b/drug effects , Complement C4b/metabolism , Dose-Response Relationship, Immunologic , Humans , Receptors, Complement/metabolism , Time Factors
11.
J Med Chem ; 36(8): 1024-31, 1993 Apr 16.
Article in English | MEDLINE | ID: mdl-8478902

ABSTRACT

In an effort to develop potent human purine nucleoside phosphorylase (PNP) inhibitors as immunosuppressive and chemotherapeutic agents, several 8-aminoguanine derivatives were synthesized and evaluated as potential PNP inhibitors. These studies were designed to investigate the hydrophobic effect of a substituent on the N-9 of the purine heterocycle and/or the C-5' positions. Compounds such as 8-aminoguanosine, guanosine, formycin B, and 8-aminoacyclovir containing a p-(fluorosulfonyl)benzoyl moiety were synthesized. The affinity of these compounds to erythrocytic PNP was determined and none of these compounds showed a better affinity than those of the parent compounds. However, we found that the effect of hydrophobicity at the N-9 and the C-5' positions might play an important role in binding to the active site of PNP. Thus, 8-amino-5'-deoxy-5'-(phenylthio)guanosine (19) was found to be the best inhibitor in this series of compounds with a Ki = 0.45 microM.


Subject(s)
Formycins/chemical synthesis , Guanine/analogs & derivatives , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Binding Sites , Formycins/chemistry , Formycins/pharmacology , Guanine/chemistry , Guanine/pharmacology , Humans , Kinetics , Purine-Nucleoside Phosphorylase/metabolism , Structure-Activity Relationship
12.
Infect Immun ; 61(2): 512-9, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8423081

ABSTRACT

The effect of antibiotic exposure of phenotypically smooth gram-negative bacteria on binding by the human lipid A-reactive monoclonal antibody HA-1A (trademark of Centocor, Inc.) was examined by liquid-phase immunoassay and by dual-parameter flow cytometry (fluorescence-activated cell sorter [FACS]) analysis. HA-1A exhibited dose-dependent binding to untreated rough gram-negative bacteria such as the Escherichia coli D21F2 Re chemotype strain but little binding to untreated smooth strains such as E. coli O111:B4, or to gram-positive bacteria. However, overnight incubation of E. coli O111:B4 with inhibitory concentrations of ceftazidime produced dose-dependent enhancement of HA-1A binding. Similar augmentation of HA-1A binding was observed when other smooth strains were exposed to cell wall-active agents. Dual-parameter FACS analysis of E. coli O111:B4 exposed overnight to two times the MIC of ceftazidime revealed a decrease in forward light scatter, indicating a reduction in average cell size or bacterial fragmentation, accompanied by a striking increase in lipid A-inhibitable HA-1A binding. Moreover, ceftriaxone, but not gentamicin, produced a marked increase in propidium iodide uptake, indicating an increase in bacterial cell permeability, and a corresponding enhancement of HA-1A binding. Antibiotic-induced enhancement of HA-1A binding to smooth strains of gram-negative bacteria thus appears related to specific alterations in bacterial cell morphology resulting in exposure of the epitope recognized by HA-1A.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/immunology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/immunology , Lipid A/immunology , Ceftazidime/pharmacology , Humans
13.
Hum Antibodies Hybridomas ; 3(4): 191-200, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1477300

ABSTRACT

The use of murine anti-CD4 monoclonal antibodies (MAbs) has shown considerable promise for the treatment of allograft rejection and rheumatoid arthritis. We have constructed mouse-human anti-CD4 antibodies with the goal of increasing their clinical potential by decreasing immunogenicity and improving effector functions. The chimeric antibodies were constructed by cloning the heavy and light chain variable regions of M-T412, a murine antibody raised against the human CD4 antigen, and joining them to the human G1, G4, or kappa constant regions in mammalian expression vectors. After transfection into mouse myeloma cells, stable cell lines were isolated that secrete up to 140 micrograms/ml chimeric antibody in static culture. The chimeric antibodies were equivalent to the murine antibody in their binding characteristics and relative affinities. However, the chimeric M-T412 MAbs have enhanced activity when compared to the murine G2a MAb in mediating antibody-dependent cell-mediated cytotoxicity using human CD4+ target and effector cells.


Subject(s)
Antibodies, Monoclonal/biosynthesis , CD4 Antigens , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Cell Line , Cloning, Molecular , DNA/genetics , Genes, Immunoglobulin , Humans , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , T-Lymphocyte Subsets/immunology , Transfection
14.
Int J Rad Appl Instrum B ; 19(6): 669-77, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1522021

ABSTRACT

The in vitro labeling and stability of 99mTc-labeled antibody Fab' fragments prepared by a direct labeling technique were evaluated. Eight antibody fragments derived from murine IgG1 (N = 5), IgG2a (N = 2) and IgG3 (N = 1) isotypes were labeled with a preformed 99mTc-D-glucarate complex. No loss of radioactivity incorporation was observed for all the 99mTc-labeled antibody fragments after 24 h incubation at 37 degrees C. The 99mTc-labeled antibody fragments (IgG1, N = 2; IgG2a, N = 2; IgG3, N = 1) were stable upon challenge with DTPA, EDTA or acidic pH. Furthermore, using the affinity chromatography technique, two of the 99mTc-labeled antibody fragments displayed no loss of immunoreactivity after prolonged incubation in phosphate buffer up to 24 h at 37 degrees C. The bonding between 99mTc and antibody fragments was elucidated by challenging with a diamide ditholate (N2S2) compound. The Fab' with IgG2a isotype displayed tighter binding to 99mTc in comparison to the Fab' from IgG1 and IgG3 isotype in N2S2 challenge and incubation with human plasma. The in vivo biodistribution of five 99mTc-labeled fragments were evaluated in normal mice. In conclusion, the direct labeling method allows stable 99mTc labeling of antibody fragments from three of the major murine isotypes.


Subject(s)
Glucaric Acid/analogs & derivatives , Immunoglobulin Fragments , Isotope Labeling/methods , Organotechnetium Compounds , Organotechnetium Compounds/chemical synthesis , Chelating Agents/pharmacology , Drug Stability , Humans , Immunoglobulin Fragments/metabolism , Immunoglobulin G/metabolism , Immunotoxins/metabolism , Organotechnetium Compounds/metabolism , Organotechnetium Compounds/pharmacokinetics , Technetium , Tissue Distribution
15.
Bioconjug Chem ; 3(2): 126-31, 1992.
Article in English | MEDLINE | ID: mdl-1515466

ABSTRACT

A bispecific F(ab')2 monoclonal antibody which recognizes both the platelet GPIIb/IIIa receptor and human tissue plasminogen activator was produced to target tPA to platelets for enhancement of thrombolysis. A stable, thioether-cross-linked bispecific F(ab')2 (7E3 X P4B6) combining the GPIIb/IIIa-specific monoclonal antibody 7E3, which inhibits platelet aggregation, and a nonneutralizing anti-tPA monoclonal antibody (P4B6) was produced. This was performed by coupling each of the parental Fab' moieties with the homobifunctional cross-linker bis(maleimido methyl) ether (BMME). 7E3 X P4B6 was sequentially purified using gel-filtration chromatography and hydrophobic interaction (HIC) HPLC. HIC was shown to completely resolve each of the parental F(ab')2 species from the bispecific one. 7E3 X P4B6 was shown to retain completely each of the parental immunoreactivities in GPIIb/IIIa and tPA binding EIA's. The bispecific antibody inhibited platelet aggregation in vitro at levels comparable to those for 7E3 Fab. Recruitment of tPA activity to washed human platelets was demonstrated using the S-2251 chromogenic substrate assay. 7E3 X P4B6 recruited 12-fold more tPA to the washed platelets than a mixture of the parental F(ab')2 molecules used as controls.


Subject(s)
Antibodies, Monoclonal/chemistry , Blood Platelets/metabolism , Drug Carriers , Immunoglobulin Fab Fragments/chemistry , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Tissue Plasminogen Activator/pharmacokinetics , Antibodies, Monoclonal/immunology , Cross-Linking Reagents , Electrophoresis, Polyacrylamide Gel , Humans , Immunoglobulin Fab Fragments/immunology , In Vitro Techniques , Platelet Membrane Glycoproteins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Tissue Plasminogen Activator/chemistry , Tissue Plasminogen Activator/immunology
16.
Int J Clin Lab Res ; 22(1): 17-20, 1992.
Article in English | MEDLINE | ID: mdl-1633315

ABSTRACT

We have used a relatively new technology to increase the number of human lymphocytes that will react with human ovarian carcinoma cells. This technology, often called "retargeting of the immune system," can temporarily redirect the activity of immune cells that were originally committed to react with foreign substances other than cancer cells. In the example presented here, the antitumor effects of retargeted human T lymphocytes, collected from normal donors, were tested in immunodeficient mice with a human ovarian carcinoma line growing intraperitoneally. We retargeted T cells in vitro with a bispecific antibody that reacted with the T cell receptor complex and with a cell-surface antigen expressed by the ovarian carcinoma cells. Retargeted lymphocytes, injected intraperitoneally into mice 4 days after intraperitoneal injection of the tumor cells, impeded tumor growth and doubled the host survival time. These findings provide support for the concept that treatment of ovarian cancer patients with retargeted T cells could prove beneficial.


Subject(s)
Antibodies/therapeutic use , Cytotoxicity, Immunologic/immunology , Ovarian Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal , Antibody Specificity/immunology , Female , Lymphocyte Activation/immunology , Mice , Mice, Nude , Ovarian Neoplasms/mortality , Survival Rate , Tumor Cells, Cultured
17.
Cancer Res ; 51(20): 5716-21, 1991 Oct 15.
Article in English | MEDLINE | ID: mdl-1833054

ABSTRACT

A bispecific F(ab')2 fragment with anti-CD3 and antitumor specificity was used to target activated human peripheral blood lymphocytes (PBL) against OVCAR-3 human ovarian carcinoma cells growing i.p. in athymic mice. Mice were given injections of OVCAR-3 cells on day 0 and treated with i.p. injections of activated PBL coated with the [anti-CD3 (TR66) x antitumor (MOv18)] bispecific F(ab')2 on day 4, using an approximate effector:target ratio of 1:1. Treatment was evaluated for the ability either to block tumor growth at 15 days or to prolong survival of tumor-bearing mice. After 15 days, the incidence of mice with tumor growth was 20% among those given PBL coated with bispecific F(ab')2, whereas the incidence among mice untreated or treated with PBL alone or PBL with either parental antibody ranged from 80 to 94%. The mean survival time of tumor-bearing mice treated with PBL and bispecific F(ab')2 was 104 days, which was 3.5 times that of untreated mice and twice that of mice given PBL alone or PBL with either parental antibody. These results provide support for the concept that treatment of ovarian cancer patients with targeted T-cells could prove beneficial.


Subject(s)
Immunoglobulin Fab Fragments/immunology , Ovarian Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Female , Humans , Immunotherapy, Adoptive , Mice , Mice, Nude , Ovarian Neoplasms/mortality , Specific Pathogen-Free Organisms
18.
Int J Rad Appl Instrum B ; 18(5): 483-97, 1991.
Article in English | MEDLINE | ID: mdl-1917518

ABSTRACT

The 323/A3 murine monoclonal antibody, initially described as reactive to breast carcinomas, is found by immunohistological analyses to have broad cross reactivity with adenocarcinomas of diverse histologic origin. The 323/A3 antigen is similar to the tumor-associated 17-1A antigen as revealed by immunoblot and cross-competition cell binding studies. We have investigated the potential use of the 323/A3 monoclonal antibody for tumor imaging as a Fab' molecule labeled with 99mTc. In vitro studies demonstrate that 323/A3 Fab' has high affinity (2-3 x 10(9) M-1) with no significant loss of immunoreactivity compared to the intact IgG. In vivo studies demonstrate that 99mTc 323/A3 Fab' can rapidly detect human breast and colon tumor xenografts growing in athymic nude mice. Distinct breast tumor visualization is observed as early as 1 h post intravenous administration with the 99mTc 323/A3 Fab'. Distinct colon tumor visualization is observed by 3 h (the earliest time point imaged). Tumor-to-blood ratios are higher for 99mTc 323/A3 Fab' than with a 99mTc-labeled nonspecific isotype-matched Fab' antibody. These results suggest that 99mTc 323/A3 Fab' can detect 17-1A antigen and may have potential clinical utility for the rapid diagnostic imaging of adenocarcinomas.


Subject(s)
Adenocarcinoma/diagnosis , Antibodies, Monoclonal , Immunoglobulin Fab Fragments , Technetium , Adenocarcinoma/immunology , Animals , Blotting, Northern , Breast Neoplasms/immunology , Colonic Neoplasms/immunology , Evaluation Studies as Topic , Female , Humans , Hybridomas , Immunohistochemistry , Isotope Labeling , Mice , Mice, Nude , Peptide Fragments/immunology , Tumor Cells, Cultured/immunology
20.
Brain Res ; 527(2): 335-41, 1990 Sep 17.
Article in English | MEDLINE | ID: mdl-2253038

ABSTRACT

Neurons in the tuberomammillary nucleus (TM) of the rat hypothalamus were immunolabelled for the enzyme adenosine deaminase (ADA) and investigated by electron microscopic immunohistochemical techniques. ADA-immunoreactivity was distributed throughout the somal and dendritic cytoplasm of TM neurons and in the karyoplasm of most, but not all of these neurons. Immunoreactive axons were rarely observed within the tightly packed cell clusters of the TM subdivisions examined. Dense deposition of immunoreaction product together with reasonable preservation of morphological detail facilitated identification of immunoreaction product together with reasonable preservation of morphological detail facilitated identification of immunoreactive profiles and allowed characterization of the ultrastructural features of labelled neurons and the relationships of these with each other and with surrounding unlabelled neuronal and glial elements. Immunolocalization of ADA therefore represents a reliable and convenient method for the identification of TM neurons in EM studies of their ultrastructure and synaptic interactions.


Subject(s)
Adenosine Deaminase/analysis , Hypothalamus, Posterior/enzymology , Neurons/enzymology , Animals , Axons/ultrastructure , Dendrites/ultrastructure , Histamine/physiology , Hypothalamus, Posterior/ultrastructure , Immunoenzyme Techniques , Male , Microscopy, Immunoelectron , Neurons/ultrastructure , Rats , Rats, Inbred Strains , Synapses/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...