Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
2.
J Exp Clin Cancer Res ; 38(1): 326, 2019 Jul 23.
Article in English | MEDLINE | ID: mdl-31337429

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is the second leading cause of cancer-related death in the Western population. The use in oncology of positron emission tomography/computed tomography (PET/CT) with emerging radiopharmaceuticals promises accurate staging of primary disease, restaging of recurrent disease and detection of metastatic lesions. Prostate-specific membrane antigen (PSMA) expression, directly related to androgen-independence, metastasis and progression, renders this tumour associate antigen a good target for the development of new radiopharmaceuticals for PET. Aim of this study was to demonstrate in a preclinical in vivo model (PSMA-positive versus PSMA-negative tumours) the targeting specificity and sensitivity of the anti-PSMA single-chain variable fragment (scFv) labelled with 124I. METHODS: The 124I-labeling conditions of the antibody fragment scFvD2B were optimized and assessed for purity and immunoreactivity. The specificity of 124I-scFvD2B was tested in mice bearing PSMA-positive and PSMA-negative tumours to assess both ex-vivo biodistribution and immune-PET. RESULTS: The uptake fraction of 124I-scFvD2B was very high on PSMA positive cells (range 75-91%) and highly specific and immuno-PET at the optimal time point, defined between 15 h and 24 h, provides a specific localization of lesions bearing the target antigen of interest (PSMA positive vs PSMA negative tumors %ID/g: p = 0.0198 and p = 0.0176 respectively) yielding a median target/background ratio around 30-40. CONCLUSIONS: Preclinical in vivo results of our immuno-PET reagent are highly promising. The target to background ratio is improved notably using PET compared to SPECT previously performed. These data suggest that, upon clinical confirmation of sensitivity and specificity, our anti-PSMA 124I-scFvD2B may be superior to other diagnostic modalities for PCa. The possibility to combine in patients our 124I-scFvD2B in multi-modal systems, such as PET/CT, PET/MR and PET/SPECT/CT, will provide quantitative 3D tomographic images improving the knowledge of cancer biology and treatment.


Subject(s)
Antigens, Surface/immunology , Glutamate Carboxypeptidase II/immunology , Prostatic Neoplasms/diagnosis , Radiopharmaceuticals/pharmacology , Single-Chain Antibodies/immunology , Animals , Antigens, Surface/pharmacology , Cell Line, Tumor , Glutamate Carboxypeptidase II/pharmacology , Humans , Immunoconjugates/immunology , Immunoconjugates/pharmacology , Iodine Radioisotopes/pharmacology , Male , Mice , Neoplasm Metastasis , Positron Emission Tomography Computed Tomography , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Radiopharmaceuticals/immunology , Single-Chain Antibodies/pharmacology , Tissue Distribution
3.
Eur J Cancer ; 49(9): 2223-32, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23433847

ABSTRACT

Prostate carcinoma is the most common non-cutaneous cancer in developed countries and represents the second leading cause of death. Early stage androgen dependent prostate carcinoma responds well to conventional therapies, but relatively few treatment options exist for patients with hormone-refractory prostate cancer. One of the most suitable targets for antibody-mediated approaches is prostate specific membrane antigen (PSMA) which is a well known tumour associated antigen. PSMA is a type II integral cell-surface membrane protein that is not secreted, and its expression density and enzymatic activity are increased progressively in prostate cancer compared to normal prostate epithelium, thereby making PSMA an ideal target for monoclonal antibody imaging and therapy. To obtain a small protein that can better penetrate tissue, we have engineered a single-chain variable fragment (scFv) starting from the variable heavy and light domains of the murine anti-PSMA monoclonal antibody D2B. scFvD2B was analysed in vitro for activity, stability, internalisation ability and in vivo for targeting specificity. Maintenance of function and immunoreactivity as well as extremely high radiolabelling efficiency and radiochemical purity were demonstrated by in vitro assays and under different experimental conditions. Despite its monovalent binding, scFvD2B retained a good strength of binding and was able to internalise around 40% of bound antigen. In vivo we showed its ability to specifically target only PSMA expressing prostate cancer xenografts. Due to these advantageous properties, scFvD2B has the potential to become a good theranostic reagent for early detection and therapy of prostate cancers.


Subject(s)
Antibodies, Monoclonal , Antigens, Surface/immunology , Glutamate Carboxypeptidase II/immunology , Prostatic Neoplasms/diagnosis , Single-Chain Antibodies/chemistry , Animals , Antigen-Antibody Complex/immunology , Biomarkers, Tumor/immunology , DNA-Binding Proteins , Early Detection of Cancer , Enzyme Stability , Enzyme-Linked Immunosorbent Assay , Heterografts , Humans , Immunoglobulin G , Immunohistochemistry , Indicators and Reagents , Male , Mice , Protein Binding , Radioimmunoassay
4.
Gene Ther ; 10(12): 1018-25, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12776159

ABSTRACT

The alpha-folate receptor (FR) is selectively overexpressed in 90% of nonmucinous ovarian carcinomas, whereas no expression is detectable in normal ovarian surface epithelium (OSE). Indirect evidence suggests that FR expression is associated with tumor progression and affects cell proliferation. To evaluate better the role of FR, we developed an approach based on intracellular expression of single-chain (sc) antibodies (intrabody) to downmodulate membrane expression of FR in ovary cancer cells. IGROV-1 and SKOV3 ovarian carcinoma cell lines were transfected with an anti-FR intrabody. Transfectants and parental cells were tested for FR, integrins and anti-FR intrabody expression by fluorescence-activated cell sorting (FACS), reverse transcription and polymerase chain reaction (RT-PCR) and/or immunoblotting. Cell growth characteristics and adhesion properties were evaluated in liquid, semisolid and organotypic cultures. The anti-FR scFv inhibited FR expression from 60 to 99%. At physiological concentrations of folate, proliferation varied directly as a function of FR expression. FR downmodulation was accompanied by reduced colony-forming ability in soft agar, morphological change of the cells, significant enhanced adhesion to laminin or Matrigel, a two- to three-fold increase in alpha6beta4 integrin expression, and a marked reduction in laminin production. In three-dimensional organotypic cultures, anti-FR intrabody-transfected IGROV1 cells grew as a single-ordered layer, reminiscent of normal OSE growth in vivo. In conclusion, the anti-FR intrabody reverses the transformed phenotype in ovary cancer cells and may provide an efficient means to inhibit selectively the growth of these cells.


Subject(s)
Antibodies, Monoclonal/genetics , Carrier Proteins/immunology , Neoplasm Proteins/immunology , Ovarian Neoplasms/metabolism , Antibodies, Monoclonal/metabolism , Carrier Proteins/metabolism , Cell Adhesion , Cell Division , Cell Transformation, Neoplastic , Down-Regulation , Female , Folate Receptors, GPI-Anchored , Folic Acid/metabolism , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Neoplasm Proteins/metabolism , Ovarian Neoplasms/pathology , Phenotype , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Transfection , Tumor Cells, Cultured
5.
J Nucl Med ; 42(11): 1697-703, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696642

ABSTRACT

UNLABELLED: The chelating agent 1,4,7,10-tetraazacyclododecane-N,N', N",N"'-tetraacetic acid (DOTA) is used to label monoclonal antibodies (mAbs) and peptides with (90)Y. DOTA allows the generation of clinically useful stable metallic radioconjugates for the treatment of a variety of tumors, but its immunogenicity has remained controversial. In this study, we evaluated the immune response to DOTA in a preclinical mouse model and in patients entered in a clinical trial. METHODS: Sera were obtained from BALB/c mice injected intraperitoneally or subcutaneously with different doses and formulations of syngeneic and xenogeneic mAbs or peptide (murine mAb Mov19 [mM19]; its chimeric version; murine V/human C ChiMov19 [cM19]; or Tyr(3)-octreotide)-DOTA conjugates. Sera from patients with neuroendocrine tumors, enrolled in a protocol for somatostatin receptor-mediated radionuclide therapy with (90)Y-DOTA-D-Phe(1)-Tyr(3)-octreotide (DOTATOC), were also collected before and after each treatment. Levels and specificity of antibody response to relevant (Mov19, ChiMov19, or Tyr(3)-octreotide) and nonrelevant (human serum albumin) DOTA targets were tested by enzyme-linked immunosorbent assay and competition assays. An anti-DOTA mAb (IgG1) derived from a ChiMov19-DOTA immunized mouse was used, in a competitive radioimmunoassay, to determine the efficiency of DOTA presentation on the different carriers. RESULTS: Depending on the immunogenicity and dosage of the mAb, a specific anti-DOTA response was revealed in the preclinical system. However, DOTA-peptide conjugate induced no immune-detectable response against either chelator or carrier. DOTA was poorly presented on small peptides, as determined using the anti-DOTA mAb. CONCLUSION: A humoral response against DOTA is possible, but only as a consequence of the response elicited against the carrier. Octreotide was not immunogenic. Thus, (90)Y-DOTATOC can be considered a safe and useful tool for receptor-mediated radionuclide therapy of somatostatin receptor-overexpressing tumors.


Subject(s)
Antibody Formation/immunology , Biotin/analogs & derivatives , Biotin/immunology , Chelating Agents/chemistry , Organometallic Compounds/immunology , Radiopharmaceuticals/immunology , Animals , Antibodies, Monoclonal , Drug Carriers , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Neuroendocrine Tumors/radiotherapy , Radiopharmaceuticals/therapeutic use
6.
Cancer Immunol Immunother ; 49(12): 679-86, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11258794

ABSTRACT

The HER-2 antigen, which is overexpressed in many breast carcinomas, is an ideal target for monoclonal antibodies due to its low expression in normal tissue and its homogeneous distribution in the tumor mass. We have developed and characterized the murine MAb MGR6 against HER-2, which is able to inhibit proliferation of tumor cells overexpressing HER-2. On the basis of these preclinical results, phase I studies in breast carcinoma patients were conducted and radiolocalization data indicated an antibody half life which directly paralleled that of other whole antibodies and thus resulting in a limited in vivo diagnostic capacity. To obtain a smaller reagent with possibly improved in vivo properties, a single chain variable fragment (scFv) of the original MGR6-producing hybridoma was generated by phage display technology. Biologically active MGR6 scFv was purified rapidly and at high yield by metal affinity chromatography. Competition FACS and ELISA analyses identified an epitope on the HER-2 extracellular domain that was shared by the scFv and the parental MAb. BlAcore analysis indicated a Koff of 9.3 x 10(-4) s(-1), similar to that of the intact MGR6 MAb. Distribution and elimination half-lives of MGR6 scFv, calculated from in vivo preclinical evaluations, were much faster (13 min and 6.2 h, respectively) than previously published results for the intact MAb (mean t1/2beta of 46 h). This represents a theoretical improvement in pharmacokinetics with respect to the parental murine MAb and points to the potential for utilizing this fragment in redirecting therapeutic agents, such as radioisotopes, to different human carcinomas overexpressing HER-2.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Immunoglobulin Variable Region/immunology , Receptor, ErbB-2/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Gene Expression , Genes, myc/genetics , Genetic Vectors , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasm Transplantation , Peptide Library , Radioimmunoassay , Tissue Distribution
7.
Cancer Immunol Immunother ; 49(6): 296-304, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10946811

ABSTRACT

Aberrant glycosylation is one of the most constant traits of malignant cells. The CaMBr1 hexasaccharide antigen, originally defined on the human breast carcinoma cell line MCF7, is expressed on some normal tissues but overexpressed in a high percentage of human breast, ovary, prostate and lung carcinomas. CaMBr1 overexpression is associated with poor prognosis. The epitope consists of the tetrasaccharide Fuc(alpha1-2)Ga1(beta1-3)GalNAc(beta1-3)Ga1alpha-O-spacer, which has recently become available as a synthetic oligosaccharide. Here we report the CaMBr1 tetrasaccharide conjugation to two different carrier proteins (CRM197 and KLH) and the evaluation of conjugate immunogenicity in mice following their administration in various vaccine formulations with two adjuvants (MPL-SE and Detox-PC). Radioimmunoassay to determine the level and isotype of anti-tetrasaccharide antibodies in mouse sera, and cytofluorimetric analysis and 51Cr-release assay on human tumor cells, to evaluate specificity of binding and complement-dependent lysis respectively, identified CaMBr1-CRM197, in association with the MPL-SE adjuvant, as the best vaccine formulation. This combination induced (1) production of tetrasaccharide-specific antibodies, with negligible side-effects; (2) antibodies with complement-mediated cytotoxic activity on human CaMBr1-positive cells and (3) a high titer of IgG1 detected in sera obtained 3 months after the first injection, indicating that the anti-tetrasaccharide antibody response was mediated by T cell activation. The availability of CaMBr1-glycoconjugate in the minimal and functional antigenic structure and the identification of an efficacious vaccine formulation opens the way to exploring the activity of this glycoconjugate in a clinical setting.


Subject(s)
ABO Blood-Group System/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Cancer Vaccines/immunology , Oligosaccharides/immunology , Animals , Dose-Response Relationship, Immunologic , Humans , Immunization , Immunoglobulin G/blood , Mice , Tumor Cells, Cultured
8.
Cancer Res ; 58(24): 5812-7, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9865740

ABSTRACT

We evaluated whether antibody response correlates with tumor therapy by cytokine gene-modified tumor cell vaccines. To characterize the antibody (Ab) response against a known antigen, colon carcinoma C26 cells and C26 variants engineered to produce interleukin (IL) 12 or IL-4 were further transduced to express the human tumor-associated antigen gp38 folate receptor (FR) alpha. Irradiated IL-12- and IL-4-producing C26/FR alpha cell vaccines cured 50 and 30% of mice bearing C26/FR alpha lung micrometastases. Treatment induced a rapid, CD4-dependent Ab production dominated by IgG2a and IgG1 in response to the IL-12 or IL-4 vaccine, respectively. In contrast, untreated tumor-bearing mice showed a late serological response dominated by IgM. Anti-FR alpha IgG1 and IgG2a were able to suppress tumor metastases upon passive transfer in vivo. Sera from mice cured by the IL-12 vaccine displayed a higher binding activity, a higher anti-FR alpha IgG2a content, and a higher complement-mediated tumor cell lysis in vitro compared to the sera from nonresponder mice. Such a correlation was not found in the sera of mice treated with the IL-4 vaccine. These data indicate that cytokine-producing tumor cell vaccines strongly influence antibody response, and that in the case of the IL-12-based vaccine, the Ab titer correlates with the therapeutic response, thus suggesting its use for monitoring the outcome of vaccination in cancer patients.


Subject(s)
Cancer Vaccines , Immunoglobulin G/biosynthesis , Interleukin-12/metabolism , Neoplasm Metastasis , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neoplasm/biosynthesis , Female , Interleukin-4/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Vaccination
9.
Cancer Res ; 58(18): 4146-54, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9751627

ABSTRACT

To provide a new tool for the immunotherapy of human ovarian carcinoma, we constructed a fusion protein between interleukin-2 (IL-2) and the single-chain Fv (scFv) of MOV19, a monoclonal antibody directed against alpha-folate receptor (alpha-FR), known to be overexpressed on human nonmucinous ovarian carcinoma. This was accomplished by fusing the coding sequences in a single open reading frame and expressing the IL-2/MOV19 scFv chimera under the control of the murine immunoglobulin K promoter in J558L plasmacytoma cells. The design allowed the construction of a small molecule combining the specificity of MOV19 with the immunostimulatory activity of IL-2. This might improve the tissue penetration and distribution of the fusion protein within the tumor, reduce its immunogenicity, and avoid the toxicity related to the systemic administration of IL-2. The IL-2/MOV19 fusion protein was stable on purification from the cell supernatant and was biologically active. Importantly, this construct was able to target IL-2 onto the surface of alpha-FR-overexpressing tumor cells and stimulated the proliferation of the IL-2-dependent CTLL-2 cell line as well as that of human resting peripheral blood lymphocytes. In a syngeneic mouse model, IL-2/MOV19 scFv specifically targeted a-FR gene-transduced metastatic tumor cells without accumulating in normal tissues, due to its fast clearance from the body. Prolonged release of IL-2/MOV19 scFv by in vivo transplanted J558-EF6.1 producer cells protected 60% of mice from the development of lung metastases caused by an i.v. injection of a-FR gene-transduced tumor cells. Moreover, treatment with IL-2/MOV19 scFv, but not with recombinant IL-2, significantly reduced the volume of s.c. tumors. The pharmacokinetics and biological characteristics of IL-2/NMOV19 scFv might allow us to combine the systemic administration of this molecule with the adoptive transfer of in vitro retargeted T lymphocytes for the treatment of ovarian cancer, thereby providing local delivery of IL-2 without toxicity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carrier Proteins/immunology , Immunoglobulin Fragments/therapeutic use , Immunoglobulin Light Chains/therapeutic use , Immunotoxins/therapeutic use , Interleukin-2/therapeutic use , Ovarian Neoplasms/therapy , Receptors, Cell Surface , Recombinant Fusion Proteins/therapeutic use , Animals , Female , Folate Receptors, GPI-Anchored , Humans , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred BALB C
10.
J Nucl Med ; 38(9): 1378-81, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9293791

ABSTRACT

UNLABELLED: Radioimmunodetection of tumor can be improved by introducing a two-step system in which radiolabeled streptavidin is administrated after the injection of a biotinylated monoclonal antibody (MAb) (two-step) or radiolabeled biotin is injected after biotinylated MAb and avidin (three-step). The anti-carcinoembryonic antigen (CEA) MAb FO23C5 has been recently exploited in a three-step protocol based on the avidin-biotin system. The anti-folate receptor (FR) MAb MOv18 has proven suitable for radioimmunodetection of ovarian cancer using directly radiolabeled MAb or in a two-step method. In this study, we analyzed the suitability of MOv18 in a three-step protocol in ovarian carcinoma patients and the internalization events after formation of the MOv18-avidin complex. METHODS: Selected patients with documented metastatic lesions were enrolled in a three-step radioimaging analysis with biotinylated MOv18 and FO23C5, avidin and (111)In-labeled biotin. Two-step internalization experiments were conducted in vitro with MOv18 and MOv19 MAbs on the FR-overexpressing IGROV1 cell line and with the anti-CEA MAb FO23C5 on the LS174T cell line. Cells were incubated sequentially with biotinylated MAb and 125I-labeled streptavidin or with 125I-biotinylated MAb and cold streptavidin. RESULTS: In the in vivo study, SPECT revealed the majority of metastatic lesions in patients injected with biotinylated MOv18; however, the tumor-to-background ratio was relatively low. In the in vitro study, a consistent internalization was induced by antigen-biotinylated MAb-streptavidin complex formation at the cell surface in both antigenic systems analyzed. However, the extent of internalization was lower in the CEA model. CONCLUSION: The internalization ability of avidin suggests its potential clinical application for delivering toxic agents in a two-step approach (biotinylated MAb + avidin conjugate). The suitability of a given MAb for three-step clinical applications (biotinylated MAb + avidin + biotin) should be previously investigated by using appropriate in vitro experiments.


Subject(s)
Antibodies, Monoclonal , Avidin , Bacterial Proteins , Iodine Radioisotopes , Radioimmunodetection , Avidin/administration & dosage , Bacterial Proteins/administration & dosage , Biotin , Carcinoembryonic Antigen/biosynthesis , Carrier Proteins/immunology , Colonic Neoplasms/immunology , Female , Folate Receptors, GPI-Anchored , Humans , Indium Radioisotopes , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Receptors, Cell Surface/immunology , Streptavidin , Tomography, Emission-Computed, Single-Photon , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...