Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 6(1): 34-41, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10656429

ABSTRACT

Carcinoembryonic antigen (CEA) is a well-characterized oncofetal glycoprotein whose overexpression by human adenocarcinomas has been a target for cancer immunotherapy. Limited information is available regarding the ability of patients to mount an antibody response to this self-antigen following vaccination. Recombinant vaccinia viruses encoding full-length or internally deleted cDNAs for human CEA were used to vaccinate 32 patients with CEA-expressing adenocarcinomas, predominantly of colorectal origin. CEA-specific autoantibodies were induced by vaccination in 7 of 32 patients. None of the patients had CEA antibodies detected before vaccination. CEA specificity of the antibodies initially identified by ELISA was confirmed by competitive inhibition analysis as well as recognition of recombinant CEA produced in baculovirus-infected insect cell cultures and human cell cultures by Western blot. The CEA autoantibodies were predominantly IgG1, with a minority of patients also demonstrating IgM autoantibodies. CEA antibodies were of low titer and low avidity, based on competitive inhibition assays. These autoantibodies did not affect clinical serum CEA protein quantitation. Furthermore, elevated serum CEA levels commonly encountered in patients with advanced adenocarcinoma did not hinder detection of low avidity polyclonal CEA antibodies. CEA antibodies such as those induced in these pilot trials are projected to have modest antitumor activity. Thus, additional Phase I/II trials of recombinant vaccinia-CEA with alternative prime-boost approaches and/or augmentation strategies are warranted in an effort to enhance the frequency and avidity of CEA-specific autoantibodies and cytolytic T cells before Phase III trials.


Subject(s)
Adenocarcinoma/immunology , Autoantibodies/blood , Cancer Vaccines/immunology , Carcinoembryonic Antigen/immunology , Colorectal Neoplasms/immunology , Immunoglobulin G/blood , Vaccines, Synthetic/immunology , Adenocarcinoma/therapy , Antibodies, Monoclonal , Antibody Formation , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Colorectal Neoplasms/therapy , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/classification , Time Factors , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/therapeutic use , Vaccinia virus
2.
Clin Cancer Res ; 5(9): 2330-7, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499601

ABSTRACT

The principal objectives of this trial were twofold: (a) to examine the safety and relative efficacy of intradermal needle injection versus s.c. jet administration of a carcinoembryonic antigen (CEA)-encoding recombinant vaccinia virus (rV-CEA) over a limited dose range and (b) to evaluate CEA-specific immune responses or antitumor effects induced by rV-CEA vaccination. Patients were randomly assigned to one of two groups, depending upon the technique of vaccine administration. All 20 patients received two doses of 10(7) or 10(8) pfu of rV-CEA at a 4-week interval. Toxicity was limited to modest local inflammation at the inoculation site as well as low-grade fever and increased fatigue, each affecting fewer than 20% of the patients. No evidence of CEA-specific lymphoproliferation, interleukin 2 release, delayed-type hypersensitivity, or antibody response was observed. Thus, the efficacy comparison between the two administration techniques was based upon the induction of immune responses to the vaccinia virus vector. Both techniques induced vaccinia-specific lymphoproliferation, interleukin 2 release, and antibody responses of comparable magnitude and frequency as well as protected 80% of patients against pustule formation following vaccinia scarification. Thus, there is no compelling reason to recommend one administration technique over the other based upon toxicity or efficacy. We have selected s.c. jet injection for subsequent trials of rV-CEA based on the ability to accommodate larger injection volumes, enhanced standardization between clinicians, and avoidance of needles that could transmit the vaccine or blood-borne pathogens to health care workers. We recommend use of 10(8) pfu doses for subsequent trials of recombinant vaccinia virus vaccines based upon the favorable toxicity profile and more consistent local pustule formation indicative of an adequate inoculation of live virus. No objective clinical responses to the rV-CEA vaccine were observed among this population of patients with widely metastatic adenocarcinoma.


Subject(s)
Adenocarcinoma/secondary , Adenocarcinoma/therapy , Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/genetics , Vaccinia virus/genetics , Adenocarcinoma/immunology , Adult , Aged , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/administration & dosage , Carcinoembryonic Antigen/biosynthesis , Carcinoembryonic Antigen/immunology , Female , Humans , Injections, Intradermal , Injections, Subcutaneous , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Male , Middle Aged , Vaccinia virus/immunology
3.
Clin Cancer Res ; 4(11): 2903-12, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9829759

ABSTRACT

In preparation for a Phase I trial of DNA immunization against carcinoembryonic antigen (CEA) in patients with colorectal carcinoma, we have produced a single plasmid DNA encoding CEA and hepatitis B surface antigen (HBsAg) under transcriptional regulatory control of two separate cytomegalovirus promoters within separate eukaryotic expression cassettes, designated pCEA/HBsAg. Hepatitis B surface antigen was included to provide an internal positive control for the efficacy of this immunization strategy without regard to the issue of breaking tolerance to a self-antigen. In the present work, we sought to examine the immunogenicity of this plasmid in a nonhuman primate model with close phylogenetic relationship to humans. Groups of pig-tailed macaques were immunized with pCEA/ HBsAg by i.m. injection or particle bombardment of the skin according to a dose and schedule thought to be optimal for the respective technique of DNA immunization. Both administration techniques produced humoral and lympho-proliferative responses of comparable magnitude. However, delayed type hypersensitivity to CEA and CEA-specific interleukin-2 release were observed only in the i.m. group, suggesting a qualitative difference in the character of the immune response elicited by the two techniques of DNA immunization. The antibody responses to CEA and HBsAg were surprisingly persistent in that all immunized animals maintained moderate antibody titers against both antigens for more than 15 months after the last boost. No toxicity was observed during 2 years of follow-up, including no measurable levels of anti-DNA antibody. This antitumor immunization strategy is presently being examined in patients with metastatic colorectal carcinoma using pCEA/HBsAg administered by i.m. injection.


Subject(s)
Cancer Vaccines/administration & dosage , Carcinoembryonic Antigen/immunology , Polynucleotides/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Antibodies/blood , Antibodies/immunology , Antibody Formation/drug effects , Cancer Vaccines/immunology , Cancer Vaccines/toxicity , Female , Hepatitis B Surface Antigens/immunology , Immunity, Cellular/drug effects , Immunization , Interleukin-2/metabolism , Lymphocyte Activation , Macaca nemestrina , Polynucleotides/immunology , Polynucleotides/toxicity , Vaccines, DNA/immunology , Vaccines, DNA/toxicity
4.
Immunopharmacology ; 40(1): 1-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9776473

ABSTRACT

The antiproliferative effect of BCX-34 was tested in normal human peripheral blood mononuclear cells (PBMCs) induced to proliferate with OKT3, tetanus toxoid, the mixed lymphocyte reaction, or IL-2. In the case of OKT3, tetanus toxoid, or the MLR the IC50s ranged between 0.7 and 4 microM. With IL-2, the IC50 was 14.6 microM. In T-cells purified by rosetting the IC50 with IL-2 was 0.62 microM. In CD4 or CD8 cells obtained by magnetic activated cell sorting the IC50s with IL-2 were 0.24 and 0.62 microM, respectively. BCX-34 inhibition of proliferation in human PBMCs may not depend entirely upon the accumulation of intracellular dGTP because tetanus toxoid-induced proliferation was inhibited in the absence of deoxyguanosine and was not reversed by deoxycytidine. BCX-34 did not inhibit IL-2 release from PBMCs and did not alter PBMC viability. The results of these studies show that BCX-34 is a potent inhibitor of normal human T-cell proliferation induced by antigenic or IL-2 stimulation. BCX-34 in normal human T-cells has a deoxyguanosine-independent mechanism to suppress in vitro proliferation. BCX-34 appears to have little effect on T-cell viability. The data suggest that BCX-34 may be useful in the treatment of T-cell proliferative disorders.


Subject(s)
Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Immunosuppressive Agents/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , T-Lymphocytes/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Guanine/pharmacology , Humans , Interleukin-2/metabolism , Interleukin-2/pharmacology , Lymphocyte Activation/drug effects , Lymphocyte Culture Test, Mixed , Muromonab-CD3/pharmacology , T-Lymphocytes/immunology , Tetanus Toxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...