Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
J Immunol ; 167(8): 4560-5, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591784

ABSTRACT

A carcinoembryonic Ag (CEA)-based DNA vaccine encoding both CEA and CD40 ligand trimer achieved effective tumor-protective immunity against murine colon carcinoma in CEA-transgenic mice by activating both naive T cells and dendritic cells. Peripheral T cell tolerance to CEA was broken in a prophylactic model by this novel, dual-function DNA vaccine, whose efficacy was further enhanced by boosts with a recombinant Ab-IL-2 fusion protein (huKS1/4-IL-2). These conclusions are supported by four lines of evidence. First, a lethal challenge of MC38-CEA-KS Ag murine colon carcinoma cells was for the first time completely rejected in 100% of experimental animals treated by oral gavage of this DNA vaccine carried by attenuated Salmonella typhimurium, followed by five boosts with huKS1/4-IL-2. Second, specific activation of dendritic cells was indicated by their marked up-regulation in expression of costimulatory molecules B7.1 (CD80), B7.2 (CD86), and ICAM-1. Third, a decisive increase over control values was observed in both MHC class I Ag-restricted cytotoxicity of CTLs from successfully vaccinated mice and secretion of proinflammatory cytokines IFN-gamma and IL-12. Fourth, activation of CTLs was augmented, as indicated by up-regulation of activity markers LFA-1, CD25, CD28, and CD69. Taken together, these results suggest that a dual-function DNA vaccine encoding CEA and CD40 ligand trimer combined with tumor-targeted IL-2 may be a promising strategy for the rational development of DNA-based cancer vaccines for future clinical applications.


Subject(s)
CD40 Ligand/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoembryonic Antigen/therapeutic use , Carcinoma/prevention & control , Cell Adhesion Molecules , Colonic Neoplasms/prevention & control , Vaccines, DNA/therapeutic use , Animals , Antigens, CD/biosynthesis , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Neoplasm/therapeutic use , B7-1 Antigen/biosynthesis , B7-2 Antigen , CD40 Ligand/genetics , CD40 Ligand/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Epithelial Cell Adhesion Molecule , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/therapeutic use , Membrane Glycoproteins/biosynthesis , Mice , Mice, Transgenic , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/immunology , Vaccination
2.
Vaccine ; 20(3-4): 421-9, 2001 Nov 12.
Article in English | MEDLINE | ID: mdl-11672905

ABSTRACT

A DNA vaccine encoding human carcinoembryonic antigen (CEA) broke peripheral T-cell tolerance toward this tumor self-antigen expressed by Lewis lung carcinoma stably transduced with CEA in C57BL/6J mice transgenic for CEA. This vaccine, delivered by oral gavage with an attenuated strain of Salmonella typhimurium (SL7207), and boosted with an antibody-IL2 fusion protein, induced tumor-protective immunity mediated by MHC class I antigen-restricted CD8(+) T cells, resulting in eradication of subcutaneous tumors in 100% of mice and prevention of experimental pulmonary metastases in 75% of experimental animals. Both CTL and antigen-presenting dendritic cells were activated as indicated by a decisive increase in their respective activation markers CD2, CD25, CD28 as well as CD48 and CD80. The antitumor effects of this CEA-based DNA vaccine obtained in prophylactic settings, suggest that this approach could lead to the rational design of effective treatment modalities for human lung cancer.


Subject(s)
Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Vaccines, DNA/immunology , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Carcinoma, Lewis Lung/prevention & control , Carcinoma, Lewis Lung/secondary , Dendritic Cells/physiology , Histocompatibility Antigens Class I/immunology , Interleukin-12/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Vaccination , Vaccines, DNA/administration & dosage
3.
Clin Cancer Res ; 7(3 Suppl): 856s-864s, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11300483

ABSTRACT

Peripheral T-cell tolerance toward human carcinoembryonic self-antigen (CEA) was broken in CEA-transgenic C57BL/6J mice by an oral CEA-based DNA vaccine. This vaccine, delivered by the live, attenuated AroA- strain of Salmonella typhimurium (SL7207), induced tumor-protective immunity mediated by MHC class I-restricted CD8+ T cells. Activation of these T cells was indicated by increased secretion of proinflammatory cytokines IFN-gamma, interleukin (IL)-12 and granulocyte/macrophage-colony stimulating factor, as well as specific tumor rejection and growth suppression in vaccinated CEA-transgenic mice after a lethal challenge with murine MC38 colon carcinoma cells. These tumor cells were double transfected with CEA and the human epithelial cell adhesion molecule (Ep-CAM)/KSA and consequently served as a docking site for a recombinant antibody-IL2 fusion protein (KS1/4-IL2) recognizing KSA. Importantly, the efficacy of the tumor-protective immune response was markedly increased by boosts with this antibody-IL2 fusion protein, resulting in more effective tumor rejection coupled with increased expression of costimulatory molecules B7.2/B7.2 and intercellular adhesion molecule 1 (ICAM-1) on dendritic cells and intensified release of proinflammatory cytokines IFN-gamma, IL-12, and granulocyte/macrophage-colony stimulating factor from T cells of successfully vaccinated CEA-transgenic C57BL/6J mice. Increased T-cell activation mediated by boosts with KS1/4-IL2 fusion protein after tumor cell challenge was further indicated by expanded expression of T-cell activation markers CD25, CD28, CD69, and LFA-1. The application of such CEA-based DNA vaccines and its further improved versions may ultimately prove useful in combination therapies directed against human carcinomas expressing CEA self-antigens.


Subject(s)
Cancer Vaccines , Carcinoembryonic Antigen/metabolism , Vaccines, DNA , Animals , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Colonic Neoplasms/metabolism , Cytokines/metabolism , Epithelial Cell Adhesion Molecule , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunoblotting , Intercellular Adhesion Molecule-1/metabolism , Interferon-gamma/metabolism , Interleukin-12/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmids/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , Up-Regulation
4.
Cancer Res ; 60(16): 4475-84, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969795

ABSTRACT

The purpose of this study was to engineer a bivalent single-chain anticarcinoembryonic antigen (CEA) antibody and an interleukin 2 (IL-2) fusion protein derivative for selective tumor targeting of cytokines. The variable domains of a high affinity anti-CEA antibody, T84.66, were used to form a single-gene-encoded antibody [single-chain variable fragment joined to the crystallizable fragment, Fc (scFvFc)]. The fusion protein (scFvFc.IL-2) consisted of mouse IL-2-fused to the COOH-terminal end of the scFvFc. The engineered proteins were assembled as complete molecules and were similar to the intact anti-CEA monoclonal antibody (Mab) in antigen-binding properties. Based on IL-2 content of the fusion protein, its ability to support proliferation of CTLL-2 cells was identical with that of IL-2. Despite a molecular size similar to that of the intact Mab, the blood clearance of the fusion protein was markedly faster than that of the intact Mab or scFvFc. Incubation of radiolabeled scFvFc.IL-2 but not the intact or scFvFc antibodies in mouse serum was accompanied by the appearance of complexes, suggesting that the latter may contribute to the accelerated clearance of the fusion protein. Biodistribution and tumor targeting studies were carried out in CEA-transgenic mice bearing CEA-positive murine tumors as well as the antigen-negative parental tumor. The bivalent anti-CEA scFvFc had tumor localization properties similar to those of the intact Mab. Although fusion of IL-2 to the COOH-terminal end of the bivalent scFvFc altered its pharmacokinetic properties, the fusion antibody was able to target tumors specifically. Maximum uptake of the intact Mab, scFvFc, and scFvFc.IL-2 in CEA-positive tumors was 29.3 +/- 5.0, 19.5 +/- 2.1, and 6.6 +/- 0.9% injected dose/g, respectively. Maximum tumor localization ratios (CEA-positive/CEA-negative tumor) were similar for all three antibody types (4.6-6.0), demonstrating the antigen specificity of the tumor targeting. Significant antigen-specific targeting to CEA-positive normal tissues of transgenic mice was not observed. Although the tumor-targeting properties of the fusion protein were low, the growth of CEA-expressing (P = 0.01) but not antigen-irrelevant (P = 0.22) syngeneic tumor cells was inhibited after treatment of transgenic mice with the anti-CEA-IL-2 antibody. Therapy of CEA-expressing tumors was improved after i.v. administration of the fusion protein (P = 0.0001). These studies indicate that anti-CEA antibody-directed cytokine targeting may offer an effective treatment for CEA-expressing carcinomas. The availability of an immunocompetent CEA transgenic mouse model will also help to determine the immunotherapeutic properties of these fusion proteins.


Subject(s)
Carcinoembryonic Antigen/immunology , Immunoglobulin Fragments/immunology , Immunotoxins/pharmacology , Interleukin-2/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Carcinoembryonic Antigen/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Drug Stability , Female , Genetic Engineering , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/metabolism , Immunotoxins/genetics , Immunotoxins/pharmacokinetics , Interleukin-2/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacokinetics , Tissue Distribution
5.
Cancer Immunol Immunother ; 49(6): 285-95, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10946810

ABSTRACT

In order to develop immunotherapy strategies that are based on eliciting immune responsiveness to the self-antigen, human carcinoembryonic antigen (CEA), we examined whether cytotoxic T lymphocyte (CTL) activity against CEA could be elicited in CEA-transgenic and nontransgenic mice. CEA-transgenic [C57BL/ 6-TGN(CEAGe)18FJP] and nontransgenic mice were primed with CEA-transfected syngeneic fibroblasts in combination with Corynebacterium parvum. Spleen cells from immunized mice were cultured with irradiated syngeneic MC-38 colon carcinoma cells transfected with CEA (MC-38.CEA) as stimulators prior to the measurement of CTL activity. Primed nontransgenic spleen cells showed augmented CTL activity against MC-38.CEA cells as compared with control parental MC-38 cells, nontransfected or transfected with vector only. Moreover, primed CEA transgenic spleen cells showed augmented CTL activity against MC-38.CEA cells that was similar to that observed in nontransgenic mice. All CTL clones derived from either transgenic or nontransgenic mice showed cross-reactivity with MC-38 cells expressing the CEA-related antigen, nonspecific cross-reacting antigen, but not biliary glycoprotein. CEA-specific CTL clones were not identified. Adoptive transfer of cloned CTL resulted in inhibition of MC-38.CEA but not MC-38.BGP tumor growth. Tumor cures were elicited in mice treated with a combination of cloned CTL and cyclophosphamide. Histopathological examination of CEA-expressing colons from either immunized mice or recipients of cloned CTL did not reveal any autoimmune reactions. These studies demonstrate that CTL recognizing cross-reactive class I epitopes on the CEA molecule can be induced in transgenic mice. The expression of these epitopes on tumor cells creates effective targets for CTL in vivo without inducing adverse reactions in CEA-expressing normal tissues. Since anti-CEA CTL have been generated in humans, CEA-transgenic mice may be a useful model to study vaccines that are based on CTL effector mechanisms.


Subject(s)
Carcinoembryonic Antigen/physiology , Immunotherapy, Adoptive , Neoplasms, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Autoimmunity , Cell Line , Cloning, Molecular , Colonic Neoplasms/therapy , H-2 Antigens/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Int J Cancer ; 85(6): 751-6, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10709090

ABSTRACT

Mice transgenic for the carcinoembryonic (CEA) gene were used to study the biodistribution and tumor targeting of a radioiodinated monoclonal antibody (MAb), T84.66. The specificity of antibody uptake in tumors was assessed in mice bearing a CEA-transfected syngeneic tumor as well as the antigen-negative parental tumor. With high CEA-expressing tumors, the percent injected dose per gram (%ID/g) approached 30% at 48 hr. Tumor uptake in antigen-positive tumors was 5-8-fold higher than that observed in the antigen-negative parental tumors. Only antigen-positive tumors were visualized by immunoscintigraphy. The tumor targeting obtained in athymic nude mice bearing human tumor xenografts was similar to that observed with CEA-expressing murine tumors implanted in either athymic nude or transgenic mice. The degree of localization of CEA-transfected murine tumors was related with the level of antigen expression. Circulating antigen-radio-antibody complexes were not detected while blood clearance of radio-antibody was similar between transgenic and non-transgenic mice. With the exception of the large bowel, the distribution of radioiodinated MAb in normal tissues was similar in both CEA transgenic and non-transgenic mice. Increased localization of intact antibody was observed in the large bowel from transgenic mice, suggesting specific targeting to antigen-positive normal tissues. These results suggest that the CEA transgenic mouse model will be useful in the development of antibodies for radio-immunodetection and treatment of carcinomas expressing CEA.


Subject(s)
Carcinoembryonic Antigen , Colonic Neoplasms/diagnostic imaging , Radioimmunodetection , Animals , Antibodies, Monoclonal , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Disease Models, Animal , Female , Iodine Radioisotopes , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Neoplasm Transplantation , Radiopharmaceuticals , Transfection , Tumor Cells, Cultured
7.
J Nucl Med ; 39(12): 2097-104, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9867150

ABSTRACT

UNLABELLED: Chimeric T84.66 (cT84.66) is a high-affinity (1.16x10(11) M(-1)) IgG1 monoclonal antibody against carcinoembryonic antigen (CEA). The purpose of this pilot trial was to evaluate the tumor-targeting properties, biodistribution, pharmacokinetics and immunogenicity of 111In-labeled cT84.66 as a function of administered antibody protein dose. METHODS: Patients with CEA-producing colorectal cancers with localized disease or limited metastatic disease who were scheduled to undergo definitive surgical resection were each administered a single intravenous dose of 5 mg of isothiocyanatobenzyl diethylenetriaminepentaacetic acid-cT84.66, labeled with 5 mCi of 111In. Before receiving the radiolabeled antibody, patients received unlabeled diethylenetriaminepentaacetic acid-cT84.66. The amount of unlabeled antibody was 0, 20 or 100 mg, with five patients at each level. Serial blood samples, 24-hr urine collections and nuclear images were collected until 7 days postinfusion. Human antichimeric antibody response was assessed up to 6 mo postinfusion. RESULTS: Imaging of at least one known tumor site was performed in all 15 patients. Fifty-two lesions were analyzed, with an imaging sensitivity rate of 50.0% and a positive predictive value of 76.9%. The antibody detected tumors that were not detected by conventional means in three patients, resulting in a modification of surgical management. Interpatient variations in serum clearance rates were observed and were secondary to differences in clearance and metabolic rates of antibody and antibody:antigen complexes by the liver. Antibody uptake in primary tumors, metastatic sites and regional metastatic lymph nodes ranged from 0.4% to 134% injected dose/kg, resulting in estimated 90Y-cT84.66 radiation doses ranging from 0.3 to 193 cGy/mCi. Thirteen patients were evaluated 1-6 mo after infusion for human antichimeric antibody, and none developed a response. No major differences in tumor imaging, tumor uptake, pharmacokinetics or organ biodistribution were observed with increasing protein doses, although a trend toward increasing blood uptake and decreasing liver uptake was observed with increasing protein dose. CONCLUSION: Chimeric T84.66 demonstrated tumor targeting comparable to other radiolabeled intact anti-CEA monoclonal antibodies. Its immunogenicity after single administration was lower than murine monoclonal antibodies. These properties make 111In-cT84.66, or a lower molecular weight derivative, attractive for further evaluation as an imaging agent. Yttrium-90 dosimetry estimates predict potentially cytotoxic radiation doses to select tumor sites, which makes 90Y-cT84.66 also appropriate for further evaluation in Phase I radioimmunotherapy trials. Although clinically important changes in biodistribution, pharmacokinetics and tumor targeting with increasing protein doses of 111In-cT84.66 were not demonstrated, the results do suggest that antibody clearance from the blood is driven by hepatic uptake and metabolism, with more rapid blood clearance seen in patients with liver metastases. These patients with rapid clearance and potentially unfavorable biodistribution for imaging and therapy may, therefore, be a more appropriate subset in which to evaluate the role of administering higher protein doses. This underscores the need to further identify, characterize and understand those factors that influence the biodistribution and clearance of radiolabeled anti-CEA antibodies, to allow for better selection of patients for therapy and rational planning of radioimmunotherapy.


Subject(s)
Colorectal Neoplasms/radiotherapy , Colorectal Neoplasms/surgery , Indium Radioisotopes/therapeutic use , Adult , Aged , Animals , Antibodies, Monoclonal/adverse effects , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , Female , Humans , Immunoglobulin G/adverse effects , Indium Radioisotopes/adverse effects , Indium Radioisotopes/pharmacokinetics , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Metabolic Clearance Rate , Mice , Middle Aged , Neoplasm Metastasis , Pilot Projects , Radiography , Radioimmunotherapy , Radionuclide Imaging , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/therapeutic use , Radiotherapy Dosage , Radiotherapy, Adjuvant , Sensitivity and Specificity , Tissue Distribution
9.
Cancer Res ; 58(7): 1469-77, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9537250

ABSTRACT

Mice transgenic for the human carcinoembryonic antigen (CEA) gene were prepared for use as a preclinical model for immunotherapy. A 32.6-kb fragment containing the complete human CEA gene and flanking sequences was isolated from a genomic cosmid clone and used to produce transgenic C57BL/6 mice. A homozygous line was established that was designated C57BL/6J-TgN(CEAGe)18FJP. Southern blot analysis showed that this line contained intact copies of the cosmid clone, with approximately 19 integrated copies at one chromosomal location. A mouse-human chimeric anti-CEA monoclonal antibody was used to examine CEA expression by immunohistochemical staining of frozen tissue sections. In the cecum and colon, approximately 20% of the luminal epithelial cells had strong cytoplasmic staining, whereas occasional glands showed intense staining. CEA was also expressed in gastric foveolar cells, whereas small intestine villi had only a few (<1%) positive cells. CEA was not found by immunohistochemistry in other tissues of the digestive tract, nor was it found in a wide range of other tissues or organs. Concordance in results was obtained between immunohistochemistry and analysis of tissue extracts by enzyme immunoassay. The lone exception was the testis, which was positive only by enzyme immunoassay. Expression of human CEA was not observed in tissues derived from nontransgenic mice. The fecal content of CEA in transgenic mice was approximately 100-fold less than that observed for humans. Circulating CEA was not detected. A CEA-transfected syngeneic murine colon carcinoma cell line, MC-38, was prepared that had stable expression of CEA in vitro and in vivo. The molecular size of CEA produced by CEA-transfected MC-38 cells and by the colon of transgenic mice was similar to that obtained with CEA purified from human colon tumors. Anti-CEA antibody appeared in nontransgenic but not transgenic mice bearing transfected MC-38 tumors. These findings demonstrate that CEA distribution and its properties in tissues of mice transgenic for the human CEA gene are similar to that observed in human tissues. As in humans, immune responsiveness to CEA, as reflected by antibody formation, was not detectable in transgenic mice bearing CEA-positive tumors. Thus, CEA transgenic mice may serve as a useful model for studying the efficacy and safety of various immunotherapy strategies directed at this tumor self-antigen.


Subject(s)
Cancer Vaccines/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Immunotherapy, Active , Mice, Transgenic/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Antibodies, Monoclonal , Blotting, Southern , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Carcinoembryonic Antigen/metabolism , Cell Division/physiology , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , DNA/genetics , DNA/isolation & purification , DNA/metabolism , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Transgenic/genetics , Tissue Distribution , Tumor Cells, Cultured
10.
J Nucl Med ; 38(12): 1951-9, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9430476

ABSTRACT

UNLABELLED: Chimeric T84.66 (cT84.66) is a high-affinity (1.16 x 10[11] M[-1]) IgG1 monoclonal antibody (MAb) against carcinoembryonic antigen (CEA). This pilot trial evaluated the tumor-targeting properties, biodistribution, pharmacokinetics and immunogenicity of 111In-labeled cT84.66. METHODS: Patients with CEA-producing metastatic malignancies were administered a single intravenous dose of 5 mCi 111In-diethylenetriaminepentaacetic acid-cT84.66. Serial blood samples, 24-hr urine collections and nuclear images were collected up to 7 days postinfusion. Human antichimeric antibody response was assessed up to 6 mo postinfusion. RESULTS: Imaging of at least one known tumor site was observed in 14 of 15 (93%) patients. Seventy-four lesions were analyzed with an imaging sensitivity rate of 45.1% and a positive predictive value of 94.1%. In one patient, two additional bone metastases developed within 6 mo of antibody administration at sites initially felt to be falsely positive on scan. One patient developed a human antichimeric antibody response predominantly to the murine portion of the antibody. The antibody cleared serum with a median T(1/2alpha) of 6.53 hr and a T(1/2beta) of 90.87 hr. Interpatient variations in serum clearance rates were observed and were secondary to differences in clearance and metabolic rates of antibody-antigen complexes by the liver. One patient demonstrated very rapid clearance of antibody by the liver, which compromised antibody localization to the primary tumor. Antibody uptake in primary and metastatic tumors ranged from 0.5% to 10.5% injected dose/kg, resulting in estimated radiation doses ranging from 0.97 to 21.3 cGy/mCi 90Y. Antibody uptake in regional lymph nodes ranged from 1.3% to 377% injected dose/kg, resulting in estimated radiation doses ranging from 2.0 to 617 cGy/mCi 90Y. CONCLUSION: Chimeric T84.66 demonstrated tumor targeting that was comparable to that of other radiolabeled intact anti-CEA Mabs. Its immunogenicity after single administration was lower than murine Mabs. These properties make cT84.66 or a lower molecular weight derivative attractive for further evaluation as an imaging agent. These same properties also make it appropriate for future evaluation in Phase I therapy trials. Finally, a wide variation in the rate of antibody clearance was observed, with one patient demonstrating very slow clearance, resulting in the highest estimated marrow dose of the group, and one patient demonstrating unusually rapid clearance, resulting in poor antibody localization to tumor. Data from this study suggest that serum CEA levels, antibody-antigen complex clearance and, therefore, antibody clearance are influenced by both the production and clearance rates of CEA. This underscores the need to further identify, characterize and understand those factors that influence the biodistribution and clearance of radiolabeled anti-CEA antibodies to allow for better selection of patients for therapy and rational planning of radioimmunotherapy.


Subject(s)
Carcinoembryonic Antigen/immunology , Indium Radioisotopes/therapeutic use , Radioimmunodetection , Radioimmunotherapy , Adult , Aged , Animals , Female , Humans , Indium Radioisotopes/pharmacokinetics , Male , Mice , Middle Aged , Pilot Projects , Predictive Value of Tests , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Sensitivity and Specificity , Tissue Distribution , Yttrium Radioisotopes/pharmacokinetics , Yttrium Radioisotopes/therapeutic use
11.
Bioconjug Chem ; 7(5): 532-5, 1996.
Article in English | MEDLINE | ID: mdl-8889012

ABSTRACT

Boron neutron capture therapy, a binary form of cancer treatment, has the potential to deliver potent cytotoxic radiation to tumor cells with minimal collateral damage to normal tissues if methods for the selective accretion of elevated concentrations of boron-10 in tumor can be developed. In this regard, a monoclonal antibody with dual specificity, for both anionic boron cluster compounds (nido-carboranes) and a tumor-associated antigen (carcinoembryonic antigen, CEA), was produced. The specific binding of a nido-carborane to CEA-expressing tumor cells was achieved using this bispecific antibody. The ability of this bispecific antibody to concentrate selectively at tumor sites in vivo has also been demonstrated, thus suggesting its potential for sequestering boron-rich compounds in tumors.


Subject(s)
Antibodies, Bispecific , Boron Neutron Capture Therapy/methods , Colonic Neoplasms/radiotherapy , Drug Delivery Systems/methods , Humans , Models, Molecular , Tumor Cells, Cultured
12.
Cancer Res ; 55(23 Suppl): 5929s-5934s, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7493373

ABSTRACT

Chimeric T84.66 (cT84.66) is a high-affinity (5 x 10(10) M-1) anti-carcino-embryonic antigen (CEA) IgG1. In a recently completed pretherapy imaging trial, 111In-labeled cT84.66 demonstrated targeting of CEA-producing metastatic sites and low immunogenicity, with human antichimeric antibody (HACA) response in only 1 of 15 patients after a single administration. The purpose of the present study was to evaluate cT84.66-diethylenetriaminepentaacetic acid labeled with 90Y in a dose-escalation Phase I trial. Patients with metastatic CEA-producing malignancies received imaging doses of 5 mCi 111In-labeled cT84.66 first, followed 1-2 weeks later by 5 mg cT84.66 labeled with the therapeutic dose of 90Y. Immediately following the therapeutic infusion, diethylenetriaminepentaacetic acid was administered by continuous i.v. infusion over 3 days at 250 mg/m2 body surface area/24 h. Biodistribution, tumor targeting, absorbed radiation dose estimates, antibody clearance, and HACA response were evaluated through blood samples, 24-h urine collections, and nuclear images performed at serial time points after infusion. To date, three patients with metastatic colorectal cancer have been evaluated at the first dose level of 5 mCi/m2. No side effects were associated with antibody administration. Localization of the antibody to nonhepatic metastatic sites was observed. Size-exclusion high-performance liquid chromatography demonstrated the formation of CEA:antibody complexes in serum in all three patients. A significant variation among patients in the clearance rate of the antibody and complexes from blood to liver was seen, which resulted in a reciprocal relationship between estimated liver dose and red marrow dose. Patients who demonstrated faster clearance to liver demonstrated greater excretion of a low-molecular-weight metabolite through the urine. Two patients developed HACA response, which persisted at 4 months after therapy. At this first dose level, no tumor responses were seen and reversible grade 1 thrombocytopenia was observed in 2 patients. cT84.66 demonstrated effective localization in CEA-producing tumors. Its low immunogenicity after a single administration makes it attractive for further evaluation as a radioimmunotherapeutic agent. However, further evaluation is needed to determine whether its immunogenicity will remain low after multiple administrations. Additionally, in two of the three patients, we identified rapid clearance of the antibody to the liver. This underscores the need to identify, characterize, and understand further those factors that influence the biodistribution and clearance of anti-CEA antibodies to allow for better selection of patients for therapy and rational planning of radioimmunotherapy.


Subject(s)
Colorectal Neoplasms/radiotherapy , Radioimmunotherapy , Yttrium Radioisotopes/therapeutic use , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/immunology , Female , Humans , Immunoglobulin G/therapeutic use , Male , Middle Aged , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use
13.
Proc Natl Acad Sci U S A ; 92(15): 6986-90, 1995 Jul 18.
Article in English | MEDLINE | ID: mdl-7624355

ABSTRACT

As the first step of a research program aimed at developing a bispecific monoclonal antibody system for the delivery of boron-rich molecules to tumor cells for boron neutron capture therapy, monoclonal antibodies (mAbs) were produced against an anionic nido-carborane derivative, 4-[7,8-dicarbadodecahydroundecaborat(-1)-7-yl]butanoic acid. Two IgG subclass mAbs, designated HAW101 and HAW102, were identified that specifically bound the anionic nido-carborane hapten, as well as a variety of other anionic nido-carborane cage derivatives. By using surface plasmon resonance technology, the affinity constants of HAW101 and HAW102 were determined to be 1.9 x 10(9) and 6.8 x 10(8) M-1, respectively. A diverse array of 7-substituted and 7,8-disubstituted anionic nido-carborane derivatives reacted with the mAb HAW101 in competition ELISA, whereas anionic closo-polyhedral boranes showed negligible binding, suggesting a role for the open nido-carborane cage structure. These results suggest that mAbs such as HAW101, which bind anionic nido-carboranes, are useful in the development of bispecific mAbs for specific targeting and enhanced boron delivery to tumor sites.


Subject(s)
Antibody Specificity , Boranes/immunology , Boron Neutron Capture Therapy , Butyrates/immunology , Drug Carriers , Neoplasms/radiotherapy , Antibodies, Monoclonal , Binding, Competitive , Drug Design , Enzyme-Linked Immunosorbent Assay , Haptens/immunology , Humans , Structure-Activity Relationship
14.
Bioconjug Chem ; 5(6): 557-64, 1994.
Article in English | MEDLINE | ID: mdl-7873658

ABSTRACT

Antibodies conjugated to oligomeric carboranyl compounds have a high potential as target species for boron neutron capture therapy (BNCT) of solid tumors. As a first step toward developing conjugates with BNCT capabilities, an oligomeric nido-carboranyl phosphate diester (Kane, R. R., Dreschel, K., and Hawthorne, M.F. (1993) J. Am. Chem. Soc. 115, 8853-8854), CB10 (10 nido-carboranes containing 90 boron atoms) with a pseudo-5'-terminal amino group, was conjugated to the anticarcinoembryonic antigen antibody T84.66 and its F(ab') fragment. The homobifunctional linker disuccinimidyl suberate (DSS) was coupled to CB10 via its 5'-terminal amino group followed by removal of excess linker with organic solvent extraction and conjugation with intact antibody. Similarly, the heterobifunctional linker, m-maleimidobenzoyl-N-hydroxysuccinimide (MBS), was coupled to CB10 and conjugated to the hinge region sulfhydryl of the F(ab') fragment of T84.66. The extent of reaction was monitored by the mobility shift of CB10-antibody conjugate on native polyacrylamide gels and the increased susceptibility of the CB10-antibody conjugate to staining with silver nitrate. CB10 was also labeled with radioiodine (131I) in a solid phase reaction with iodogen and used in double-label studies with 125I-labeled antibody. Although free CB10 bound very tightly to gel filtration media such as Sephadex G-25, the CB10-antibody conjugate passed through freely. After separation of CB10-antibody conjugate from free CB10 on Sephadex G-25, molar incorporations of CB10 were calculated. At a molar ratio of 10:1 (CB10:T84.66), greater than 90% of T84.66 and 30% of its F(ab)' fragment were conjugated to CB10.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Boron Compounds/chemical synthesis , Boron Neutron Capture Therapy , Boron , Carcinoembryonic Antigen/immunology , Immunoglobulin Fab Fragments , Neoplasms/radiotherapy , Radioimmunotherapy , Thymine Nucleotides/chemical synthesis , Animals , Boron Compounds/pharmacokinetics , Boron Compounds/therapeutic use , Electrophoresis, Polyacrylamide Gel , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Succinimides/chemistry , Thymine Nucleotides/pharmacokinetics , Thymine Nucleotides/therapeutic use , Tissue Distribution
16.
Cancer Res ; 53(14): 3355-61, 1993 Jul 15.
Article in English | MEDLINE | ID: mdl-8324746

ABSTRACT

The purpose of this study was to determine if human colon cancer cells transduced with monoclonal antibody (MAb) genes become sensitive to immune destruction through coexpression of both the MAb and its reactive antigen. Murine retroviral expression vectors were constructed with the heavy or light chain genes of an anti-human colon carcinoma MAb, D612, that mediates antibody-dependent cell-mediated cytotoxicity (ADCC). Transduction of D612 MAb genes into the D612 antigen-positive (> 95%) human colon carcinoma cell line, LS-174T, was carried out by sequential cocultivation with PA317 packaging cells producing infectious virions containing the light or heavy chain expression vectors. Six cultures survived drug selection, two of which were found to have elevated levels of both light and heavy immunoglobulin chain activity in their supernatants. IgG secretion levels (24 h) were 1-2 ng/1 x 10(6) cells. Low but definite antigen reactivity was also present in supernatants obtained from these LS-174T transductants. Immunocytochemical staining of transduced tumor cells revealed that > 95% of the cells were positive for IgG expression. Thus, LS-174T transductants were capable of producing both the D612 MAb and D612-reactive antigen. Analysis of transductants by flow cytometry further revealed that > 95% of the cells had murine immunoglobulin on their surfaces. ADCC mediated by human natural killer cells against nontransduced tumor cells was observed when the latter cells were co-cultivated in the presence of transductants producing both D612 heavy and light chains but not in the presence of tumor cells transduced with light chain only. LS-174T cells transduced with both D612 heavy and light chain genes were more sensitive to cytotoxicity mediated by natural killer cells than were light chain gene only transductants. ADCC contributed to the greater sensitivity of the former transductants to cytotoxicity based on its inhibition by anti-FcR gamma III antibody. Thus, these studies demonstrate that tumor cells transduced with genes encoding for MAbs that can participate in ADCC reactions are able to sensitize nontransduced tumor cells to immune destruction as well as to direct killer cells against themselves. These studies may lead to a new immunotherapeutic approach for the treatment of cancer based on MAb gene therapy.


Subject(s)
Antibodies, Monoclonal/metabolism , Antibodies, Neoplasm/metabolism , Antibody-Dependent Cell Cytotoxicity/immunology , Colonic Neoplasms/immunology , Immunoglobulin G/analysis , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/genetics , Antibodies, Neoplasm/analysis , Antibodies, Neoplasm/genetics , Colonic Neoplasms/metabolism , Flow Cytometry , Humans , Immunoglobulin Heavy Chains/analysis , Immunoglobulin Light Chains/analysis , Immunohistochemistry , Transfection
17.
Immunol Lett ; 36(2): 179-85, 1993 May.
Article in English | MEDLINE | ID: mdl-8102353

ABSTRACT

cDNA encoding the human IL gene (580 bp), inserted into a retroviral expression vector carrying neomycin resistance selective marker, was introduced into HT-29 human colon carcinoma cells by lipofection. Interleukin-6 activity was measured by ELISA and bioassay using B9 cells. Interleukin-6 secreted by transfected HT-29 cells was shown to be biologically active. The expression of the human tumor associated antigen CEA (carcinoembryonic antigen), HLA classes I and II, and ICAM-1 antigens in the transfected HT-29 cells were also analyzed by flow cytometry. Significant enhancement in the expression of CEA but not in the expression of HLA class I, HLA class II and ICAM-1 antigens, was observed in the transfected HT-29 cells as compared to the parental HT-29 cells. These results provide experimental evidence that enhancement of tumor antigen expression on tumor cells can be induced by IL-6 gene transfection, and suggest another potential role for the use of IL-6 gene transfer in the immunotherapy of human cancers.


Subject(s)
Antigens, Neoplasm/biosynthesis , Carcinoma/pathology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes , Interleukin-6/genetics , Antigens, Neoplasm/genetics , Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Carcinoma/immunology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Colorectal Neoplasms/immunology , DNA/genetics , Flow Cytometry , HLA Antigens/biosynthesis , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Intercellular Adhesion Molecule-1 , Interleukin-6/biosynthesis , Interleukin-6/physiology , Liposomes , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Receptors, Interleukin-6 , Transfection , Tumor Cells, Cultured
18.
Int J Biol Markers ; 6(2): 83-90, 1991.
Article in English | MEDLINE | ID: mdl-1716293

ABSTRACT

Enzyme-labeled monoclonal antibodies (MAbs) were used in an immunohistochemical, dual-staining study of 10 colon adenocarcinomas. MAbs B72.3 and COL-4, reactive with the high molecular weight tumor-associated glycoprotein-72 (TAG-72) antigen and carcinoembryonic antigen (CEA), respectively, were labeled with horseradish peroxidase or alkaline phosphatase. Dual staining using the two MAbs on a single tissue section (formalin-fixed, paraffin-embedded) showed that greater numbers of carcinoma cells could be detected by using the combination of the two MAbs than could be detected by use of either MAb alone. In many tumors, some carcinoma cells reacted with MAb B72.3, some reacted with MAb COL-4, and some cells reacted with both MAbs. Only 1 of 10 carcinomas showed greater than 75% reactive cells when stained with each MAb individually. In 9 of 10 cases, however, greater than 75% of cells reacted when the combination of MAbs was used. Cell surface and cytoplasmic patterns of reactivity were observed with both MAbs while some pools of extracellular mucin were composed of both TAG-72 and CEA. This study supports the rationale for the use of a combination of anti-TAG-72 and anti-CEA MAbs for in vitro immunologic detection and potential in vivo immunodiagnostic and immunotherapeutic applications for these MAbs in colon adenocarcinoma patients.


Subject(s)
Adenocarcinoma/chemistry , Antigens, Neoplasm/analysis , Carcinoembryonic Antigen/analysis , Colonic Neoplasms/chemistry , Glycoproteins/analysis , Immunoenzyme Techniques , Adenocarcinoma/diagnosis , Alkaline Phosphatase , Antibodies, Monoclonal , Colonic Neoplasms/diagnosis , Horseradish Peroxidase , Humans , Staining and Labeling
19.
Cancer Res ; 51(3): 926-34, 1991 Feb 01.
Article in English | MEDLINE | ID: mdl-1988133

ABSTRACT

Monoclonal antibody (MAb) D612 recognizes an antigen expressed on the cell surface of normal and malignant gastrointestinal epithelium. It is a murine IgG2a/kappa which has been previously shown to mediate killing of human colon carcinoma cells using human effector cells (which could be enhanced in the presence of interleukin-2). Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) analyses of MAb D612 immunoprecipitates of extracts of L-[35S]methionine-, L-[3H]leucine-, and D-[3H]glucosamine-labeled human colon carcinoma cells showed that the D612 antigen is a Mr 48,000 glycoprotein. Similar estimates of molecular mass were obtained from SDS-PAGE analyses of MAb D612 immuno-precipitates of radioiodinated extracts of surgically resected colon carcinoma and adjacent normal colonic mucosa. D612 antigen was not detectable in immunoprecipitates of supernatant media from radiolabeled cell cultures, suggesting that the antigen is not readily shed from the surface of cultured cells. The D612 antigen was shown to be clearly distinct from previously described gastrointestinal carcinoma-associated glycoproteins: the D612 antigen shows a migration pattern of SDS-PAGE distinct from those of the antigens recognized by MAbs KS1/4 and GA733, and reciprocal immunodepletion analyses of D-[3H]glucosamine-labeled colon carcinoma cells utilizing MAbs D612 and GA733 revealed no cross-reactivity between these antibodies. Similarly, competitive binding studies between MAbs 17-1A and KS1/4 and MAb D612 revealed no similarity between the epitopes recognized by MAb D612 and MAbs 17-1A and KS1/4. MAbs D612 and 17-1A were also titered in immunoperoxidase staining assays on serial frozen sections of normal and malignant colon. MAb D612 showed a higher titer of immunostaining reactivity with both normal and malignant colon than did MAb 17-1A. MAb D612 showed roughly equivalent immunostaining titers against normal and malignant colon; whereas MAb 17-1A showed higher titer of immunostaining reactivity against the normal colon tissue than against the malignant colon. Flow cytometric analysis of phosphatidylinositol-specific phospholipase C-treated colon carcinoma cells revealed no loss of D612 antigen from the cell surface, suggesting that the mechanisms of attachment of the D612 antigen to the cell surface does not involve linkage to a phosphatidylinositol glycan.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Antibodies, Monoclonal , Antigens, Neoplasm/analysis , Carcinoma/immunology , Colorectal Neoplasms/immunology , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/isolation & purification , Humans , Melanoma/immunology , Molecular Weight , Precipitin Tests , Tumor Cells, Cultured/immunology
20.
J Immunother (1991) ; 10(1): 2-12, 1991 Feb.
Article in English | MEDLINE | ID: mdl-2012797

ABSTRACT

The tumoricidal properties of an anti-human colon carcinoma monoclonal antibody (MAb), designated D612 (IgG2a), alone and in combination with IL-2-activated human lymphocytes were investigated in athymic mice bearing LS-174T colon tumor xenografts. Treatment of mice bearing LS-174T tumors (1 day, s.c.) with a single i.v. dose of 400 micrograms of D612 alone resulted in a significant inhibition of tumor growth. Lower doses of D612 had an intermediate effect on tumor growth. Similar inhibition of tumor growth was obtained when D612 was administered in three doses of 400 or 800 micrograms each during the first week after tumor implantation. Mouse macrophages but not splenocytes mediated antibody-dependent cellular cytotoxicity with D612, suggesting that tumor inhibition was due to the participation of host macrophages with D612. Human lymphokine-activated killer (LAK) cells were generated by incubating human peripheral blood mononuclear cells (PBLs) from normal donors with 100 U/ml of IL-2 for 24 h. An administration of human LAK cells did not significantly inhibit the growth of the human xenograft tumor. Adoptive transfer of a single dose of human LAK cells (2 x 10(7), i.v.) into mice treated with a suboptimal dose of D612 (200 micrograms) significantly inhibited tumor growth compared to that obtained with either D612 or LAK cells alone. Similar results were obtained with three doses of D612 plus human LAK cells although there was a tendency for multiple doses of LAK cells alone to show some antitumor effects. LAK cells or PBLs had similar antitumor activities when used in conjunction with D612. When larger established tumors were treated, single or multiple doses of D612 or LAK cells alone were without effect; however, LAK cells plus D612 elicited significant growth inhibition. These results demonstrate that the tumoricidal properties of LAK cells and the D612 MAb can be augmented when used together in the immunotherapy of human colon cancer xenografts.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Colonic Neoplasms/therapy , Immunotherapy , Killer Cells, Lymphokine-Activated/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Antigens, Neoplasm/immunology , Colonic Neoplasms/immunology , Female , Humans , Interleukin-2/pharmacology , Macrophages/immunology , Mice , Mice, Nude , Neoplasm Transplantation , Spleen/immunology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...