Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Skin Health Dis ; 1(1)2021 Mar.
Article in English | MEDLINE | ID: mdl-34541577

ABSTRACT

BACKGROUNDS: Folate Hydrolase-1 (FOLH1; PSMA) is a type II transmembrane protein, luminally expressed by solid tumour neo-vasculature. Monoclonal antibody (mAb), J591, is a vehicle for mAb-based brachytherapy in FOLH1+ cancers. Brachytherapy is a form of radiotherapy that involves placing a radioactive material a short distance from the target tissue (e.g., on the skin or internally); brachytherapy is commonly accomplished with the use of catheters, needles, metal seeds and antibody or small peptide conjugates. Herein, FOLH1 expression in primary (p) and metastatic (m) Merkel cell carcinoma (MCC) is characterized to determine its targeting potential for J591-brachytherapy. MATERIALS & METHODS: Paraffin sections from pMCC and mMCC were evaluated by immunohistochemistry for FOLH1. Monte Carlo simulation was performed using the physical properties of conjugated radioisotope lutetium-177. Kaplan-Meier survival curves were calculated based on patient outcome data and FOLH1 expression. RESULTS: Eighty-one MCC tumours were evaluated. 67% (54/81) of all cases, 77% (24/31) pMCC and 60% (30/50) mMCC tumours were FOLH1+. Monte Carlo simulation showed highly localized ionizing tracks of electrons emitted from the targeted neo-vessel. 42% (34/81) of patients with FOLH1+/- MCC had available survival data f or analysis. No significant differences in our limited data set were detected based on FOLH1 status (p = 0.4718; p = 0.6470), staining intensity score (p = 0.6966; p = 0.9841) or by grouping staining intensity scores (- and + vs. ++, +++, +++) (p = 0.8022; p = 0.8496) for MCC-specific survival or recurrence free survival, respectively. CONCLUSIONS: We report the first evidence of prevalent FOLH1 expression within MCC-associated neo-vessels, in 60-77% of patients in a large MCC cohort. Given this data, and the need for alternatives to immune therapies it is appropriate to explore the safety and efficacy o f FOLH1-targeted brachytherapy for MCC.

2.
Br J Cancer ; 91(1): 164-70, 2004 Jul 05.
Article in English | MEDLINE | ID: mdl-15188008

ABSTRACT

Interferon alpha (IFNalpha) is used to treat patients with advanced renal cell carcinoma (RCC) despite limited clinical benefit. IFNalpha can induce Fas receptor-mediated apoptosis by direct activation of pro-caspase-8 followed by activation of caspase-3. Alternative, indirect activation of caspase-3 via mitochondrial release of cytochrome c can occur and may explain the rescue from Fas-activated cell death by the antiapoptotic members of the Bcl-2 family. In this study, we examined G3139, a novel antisense compound targeting Bcl-2, in combination with IFNalpha. Human RCC lines (SK-RC-44 and SK-RC-07) were treated with IFNalpha, G3139 or a combination of the two. Fas-mediated cytotoxicity was induced by anti-Fas mAb, CH11. An analysis of Bcl-2, Fas and the cleavage of PARP was performed. IFNalpha induced Fas and Bcl-2 in SK-RC-44 and SK-RC-07. IFNalpha sensitised SK-RC-44 to anti-Fas and induced PARP cleavage confirming that IFNalpha has a cytotoxic effect on RCC lines by induction of the Fas antigen. Cytotoxicity was not evident in SK-RC-07 cells treated with IFNalpha. G3139 induced a specific downregulation of Bcl-2 in SK-RC-07 cells, which were then sensitised to anti-Fas after treatment with IFNalpha. Taken together, these results suggest that Fas-dependent pathways as well as alternative pathways, which can be inhibited by Bcl-2, exist in renal cell carcinoma. G3139 in combination with IFNalpha is a potential therapy in patients with metastatic renal cell carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/pathology , Interferon-alpha/pharmacology , Kidney Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Thionucleotides/pharmacology , fas Receptor/pharmacology , Carcinoma, Renal Cell/metabolism , Down-Regulation , Humans , Kidney Neoplasms/metabolism , Oligonucleotides, Antisense , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Cells, Cultured
3.
Prostate Cancer Prostatic Dis ; 5(1): 36-46, 2002.
Article in English | MEDLINE | ID: mdl-15195129

ABSTRACT

Limited options for the treatment of prostate cancer have spurred the search for new therapies. One innovative approach is the use of targeted alpha therapy (TAT) to inhibit cancer growth, using an alpha particle emitting radioisotope such as (213)Bi. Because of its short range and high linear energy transfer (LET), alpha-particles may be particularly effective in the treatment of cancer, especially in inhibiting the development of metastatic tumors from micro-metastases. Prostate-specific membrane antigen (PSMA) is expressed in prostate cancer cells and the neovasculature of a wide variety of malignant neoplasms including lung, colon, breast and others, but not in normal vascular endothelium. The expression is further increased in higher-grade cancers, metastatic disease and hormone-refractory prostate cancer (PCA). J591 is one of several monoclonal antibodies (mabs) to the extracellular domain of PSMA. Chelation of J591 mab with (213)Bi forms the alpha-radioimmunoconjugate (AIC). The objective of this preclinical study was to design an injectable AIC to treat human prostate tumors growing subcutaneously in mice. The anti-proliferative effects of AIC against prostate cancer were tested in vitro using the MTS assay and in vivo with the nude mice model. Apoptosis was documented using terminal deoxynucleotidyl transferase [TdT]-mediated deoxyuridinetriphosphate [dUTP] nick end-labeling (TUNEL) assay, while proliferative index was assessed using the Ki-67 marker. We show that a very high density of PSMA is expressed in an androgen-dependent human PCA cell line (LNCaP-LN3) and in tumor xenografts from nude mice. We also demonstrate that the AIC extensively inhibits the growth of LN3 cells in vitro in a concentration-dependent fashion, causing the cells to undergo apoptosis. Our in vivo studies showed that a local AIC injection of 50 microCi at 2 days post-cell inoculation gave complete inhibition of tumor growth, whereas results for a non-specific AIC were similar to those for untreated mice. Further, after 1 and 3 weeks post-tumor appearance, a single (100 microCi/100 microl) intra-lesional injection of AIC can inhibit the growth of LN3 tumor xenografts (volume<100 mm(3)) in nude mice. Tumors treated with AIC decreased in volume from a mean 46+/-14 mm(3) in the first week or 71+/-15 mm(3) in the third week to non-palpable, while in control mice treated with a non-specific AIC using the same dose, tumor volume increased from 42 to 590 mm(3). There were no observed side effects of the treatment. Because of its in vitro cytotoxicity and these anti-proliferative properties in vivo, the (213)Bi-J591 conjugate has considerable potential as a new therapeutic agent for the treatment of prostate cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Surface/immunology , Bismuth/therapeutic use , Glutamate Carboxypeptidase II/immunology , Prostatic Neoplasms/therapy , Radioimmunotherapy , Radioisotopes/therapeutic use , Alpha Particles , Animals , Apoptosis , Drug Evaluation, Preclinical , Flow Cytometry , Humans , Immunoconjugates/therapeutic use , Immunoenzyme Techniques , In Situ Nick-End Labeling , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology
4.
Science ; 294(5546): 1537-40, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11711678

ABSTRACT

A single, high linear energy transfer alpha particle can kill a target cell. We have developed methods to target molecular-sized generators of alpha-emitting isotope cascades to the inside of cancer cells using actinium-225 coupled to internalizing monoclonal antibodies. In vitro, these constructs specifically killed leukemia, lymphoma, breast, ovarian, neuroblastoma, and prostate cancer cells at becquerel (picocurie) levels. Injection of single doses of the constructs at kilobecquerel (nanocurie) levels into mice bearing solid prostate carcinoma or disseminated human lymphoma induced tumor regression and prolonged survival, without toxicity, in a substantial fraction of animals. Nanogenerators targeting a wide variety of cancers may be possible.


Subject(s)
Actinium/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/radiotherapy , Radioimmunotherapy/methods , Actinium/administration & dosage , Actinium/pharmacokinetics , Alpha Particles/therapeutic use , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Female , Half-Life , Heterocyclic Compounds, 1-Ring , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Linear Energy Transfer , Lymphoma/radiotherapy , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prostate-Specific Antigen/blood , Prostatic Neoplasms/radiotherapy , Survival Rate , Tumor Cells, Cultured
5.
Clin Cancer Res ; 7(10): 3276-81, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11595725

ABSTRACT

The antitumor effect of T cells is executed either through CD95 or Perforin (PFN)/Granzyme B (GrB) pathways. Induction of apoptosis by either mode requires activation of caspase family members. However, recent studies have suggested that cell death can proceed in the absence of caspase induction and apoptotic events. We investigated the contribution of CD95 and PFN/GrB-mediated cytotoxicity to apoptotic and necrotic mechanisms of cell death in human renal cell carcinoma. Although freshly isolated and cultured tumors expressed CD95 on their surface, they were resistant to CD95-mediated apoptosis. CD95 resistance coincided with decreased levels of FADD protein and diminished caspase-3-like activity. In contrast, we demonstrated that tumor cell death mediated by PFN/GrB can be achieved in the absence of functional caspase activity and is accompanied by a dramatic accumulation of nonapoptotic necrotic cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Carcinoma, Renal Cell/pathology , T-Lymphocytes, Cytotoxic/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/metabolism , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspases/metabolism , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Fas Ligand Protein , Fas-Associated Death Domain Protein , Granzymes , Humans , Jurkat Cells , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Necrosis , Perforin , Pore Forming Cytotoxic Proteins , Serine Endopeptidases/pharmacology , Tumor Cells, Cultured , fas Receptor/immunology , fas Receptor/metabolism
6.
J Urol ; 166(4): 1316-21, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11547065

ABSTRACT

PURPOSE: We provide an overview of advances in molecular based therapeutic strategies for prostate cancer and summarize the studies presented at the Society of Urologic Oncology Biotechnology Forum in 2000. MATERIALS AND METHODS: Three promising new treatment strategies are presented, and a critique of the advantages and limitations of each is offered by a leading expert in the field. RESULTS: Treatment results and the current state of dendritic cell based immunotherapy, monoclonal antibody therapy and anti-apoptotic treatment approaches are presented. CONCLUSIONS: Currently patients with advanced prostate carcinoma have expanded therapeutic options available in the form of molecular based phases II and III clinical trials.


Subject(s)
Prostatic Neoplasms/therapy , Antibodies, Monoclonal/therapeutic use , Apoptosis , Clinical Trials as Topic , Dendritic Cells , Humans , Immunotherapy/methods , Male , Neoplasm Staging , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology
7.
Cancer Res ; 61(5): 2008-14, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280760

ABSTRACT

A theoretical drawback to alpha-particle therapy with 213Bi is the short range of the particle track coupled with the short half-life of the radionuclide, thereby potentially limiting effective cytotoxicity to rapidly accessible, disseminated individual tumor cells (e.g., as in leukemia). In this work, a prostate carcinoma spheroid model was used to evaluate the feasibility of targeting micrometastatic clusters of tumor cells using 213Bi-labeled anti-prostate-specific membrane antigen (PSMA) antibody, J591. In prostate cancer, vascular dissemination of tumor cells or tumor cell clusters to the marrow constitutes an important step in the progression of this disease to widespread skeletal involvement, an incurable state. Such prevascularized clusters are ideal targets for radiolabeled antibodies because the barriers to antibody penetration that are associated with the capillary basal lamina have not yet formed. Beta- and gamma-emitting radionuclides such as 131I, which are widely used in radioimmunotherapy, are not expected to be effective when targeting single cells or small cell clusters. This is because the range of the emissions is one to two orders of magnitude greater than the target size, and the energy deposited per traversal is insufficient to produce any significant radiobiological effect. Spheroids of the prostate cancer cell line, LNCaP-LN3, were used as a model of prevascularized micrometastases; their response to an anti-PSMA antibody, J591, radiolabeled with the alpha-particle emitter 213Bi (T(1/2), 45.6 min.) has been measured. The time course of spheroid volume reductions was found to be sensitive to the initial spheroid volume. J591 labeled with 0.9 MBq/ml 213Bi resulted in a 3-log reduction in spheroid volume on day 33, relative to control, for spheroids with an initial diameter of 130 microm; 1.8 MBq/ml were required to achieve a similar response for spheroids with an initial diameter of 180 microm. Equivalent spheroid responses were observed after 12 Gy of acute external beam photon irradiation. Monte Carlo-based microdosimetric analyses of the 213Bi decay distribution in individual spheroids of 130-microm diameter yielded an average alpha-particle dose of 3.7 Gy to the spheroids, resulting in a relative biological effectiveness factor of 3.2 over photon irradiation. The activity concentrations used in the experiments were clinically relevant, and this work supports the possibility of using 213Bi-labeled antibodies not only for disseminated single tumor cells, as found in patients with leukemia, but also for micrometastatic tumor deposits up to 180 microm in diameter (1200 cells).


Subject(s)
Antigens, Surface , Bismuth/pharmacology , Carboxypeptidases/immunology , Immunotoxins/pharmacology , Prostatic Neoplasms/radiotherapy , Radioisotopes/pharmacology , Alpha Particles/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Cell Division/radiation effects , Glutamate Carboxypeptidase II , Humans , Immunotoxins/immunology , Immunotoxins/metabolism , Male , Microscopy, Confocal , Neoplasm Metastasis , Prostatic Neoplasms/immunology , Radioimmunotherapy , Spheroids, Cellular/immunology , Spheroids, Cellular/radiation effects , Tumor Cells, Cultured
8.
Mol Biol Cell ; 12(2): 279-95, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11179415

ABSTRACT

The cell adhesion molecule E-cadherin has been implicated in maintaining the polarized phenotype of epithelial cells and suppression of invasiveness and motility of carcinoma cells. Na,K-ATPase, consisting of an alpha- and beta-subunit, maintains the sodium gradient across the plasma membrane. A functional relationship between E-cadherin and Na,K-ATPase has not previously been described. We present evidence that the Na,K-ATPase plays a crucial role in E-cadherin-mediated development of epithelial polarity, and suppression of invasiveness and motility of carcinoma cells. Moloney sarcoma virus-transformed Madin-Darby canine kidney cells (MSV-MDCK) have highly reduced levels of E-cadherin and beta(1)-subunit of Na,K-ATPase. Forced expression of E-cadherin in MSV-MDCK cells did not reestablish epithelial polarity or inhibit the invasiveness and motility of these cells. In contrast, expression of E-cadherin and Na,K-ATPase beta(1)-subunit induced epithelial polarization, including the formation of tight junctions and desmosomes, abolished invasiveness, and reduced cell motility in MSV-MDCK cells. Our results suggest that E-cadherin-mediated cell-cell adhesion requires the Na,K-ATPase beta-subunit's function to induce epithelial polarization and suppress invasiveness and motility of carcinoma cells. Involvement of the beta(1)-subunit of Na,K-ATPase in the polarized phenotype of epithelial cells reveals a novel link between the structural organization and vectorial ion transport function of epithelial cells.


Subject(s)
Cell Movement/physiology , Cell Polarity/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Actins/metabolism , Actins/ultrastructure , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/physiology , Cell Line/virology , Clone Cells , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Dogs , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Moloney murine sarcoma virus , Protein Subunits , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/genetics
9.
Mol Urol ; 4(3): 217-22;discussion 223, 2000.
Article in English | MEDLINE | ID: mdl-11062377

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a potential target in prostate cancer patients because it is very highly expressed and because it has been reported to be upregulated by androgen deprivation. This overview addresses the expression of the PSMA gene in terms of the promoter and enhancer and how that may play a role in gene therapy. We also review PSMA as a target for antibodies for imaging and treatment and the development of a novel hybrid T-cell receptor that combines the specificity of anti-PSMA antibodies with that of T-cell receptor activation when introduced into primary lymphocytes by retroviral-mediated gene transfer. We also discuss our recent findings on the expression of a PSMA-like gene and how that understanding allows specific targeting of PSMA.


Subject(s)
Antigens, Neoplasm/metabolism , Carboxypeptidases/metabolism , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/therapy , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Antigens, Surface/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carboxypeptidases/genetics , Carboxypeptidases/immunology , Enhancer Elements, Genetic , Enzyme Inhibitors/pharmacology , Female , Genetic Therapy , Glutamate Carboxypeptidase II , Humans , Male , Prodrugs/metabolism , Promoter Regions, Genetic , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/genetics , Receptors, Antigen, T-Cell/immunology , Tumor Cells, Cultured
10.
Cancer Res ; 60(21): 6095-100, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085533

ABSTRACT

A novel alpha-particle emitting monoclonal antibody construct targeting the external domain of prostate-specific membrane antigen (PSMA) was prepared and evaluated in vitro and in vivo. The chelating agent, N-[2-amino-3-(p-isothiocyanatophen-yl)propyl]-trans-cyclohexane-1, 2-diamine-N,N',N',N'',N''-pentaacetic acid, was appended to J591 monoclonal antibody to stably bind the 213Bi radiometal ion. Bismuth-213 is a short-lived (t 1/2 = 46 min) radionuclide that emits high energy alpha-particles with an effective range of 0.07-0.10 mm that are ideally suited to treating single-celled neoplasms and micrometastatic carcinomas. The LNCaP prostate cancer cell line had an estimated 180,000 molecules of PSMA per cell; J591 bound to PSMA with a 3-nM affinity. After binding, the radiolabeled construct-antigen complex was rapidly internalized into the cell, carrying the radiometal inside. [213Bi]J591 was specifically cytotoxic to LNCaP. The LD50 value of [213Bi]J591 was 220 nCi/ml at a specific activity of 6.4 Ci/g. The potency and specificity of [213Bi]J591 directed against LNCaP spheroids, an in vitro model for micrometastatic cancer, also was investigated. [213Bi]J591 effectively stopped growth of LNCaP spheroids relative to an equivalent dose of the irrelevant control [213Bi]HuM195 or unlabeled J591. Cytotoxicity experiments in vivo were carried out in an athymic nude mouse model with an i.m. xenograft of LNCaP cells. [213Bi]J591 was able to significantly improve (P < 0.0031) median tumor-free survival (54 days) in these experiments relative to treatment with irrelevant control [213Bi]HuM195 (33 days), or no treatment (31 days). Prostate-specific antigen (PSA) was also specifically reduced in treated animals. At day 51, mean PSA values were 104 ng/ml +/- 54 ng/ml (n = 4, untreated animals), 66 ng/ml +/- 16 ng/ml (n = 6, animals treated with [213Bi]HuM195), and 28 ng/ml +/- 22 ng/ml (n = 6, animals treated with [213Bi]J591). The reduction of PSA levels in mice treated with [213Bi]J591 relative to mice treated with [213Bi]HuM195 and untreated control animals was significant with P < 0.007 and P < 0.0136, respectively. In conclusion, a novel [213Bi]-radiolabeled J591 has been constructed that selectively delivers alpha-particles to prostate cancer cells for potent and specific killing in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bismuth/pharmacology , Immunotoxins/pharmacology , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy , Radioisotopes/pharmacology , Alpha Particles/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antigen-Antibody Complex/metabolism , Binding Sites , Cell Death/radiation effects , Humans , Immunotoxins/immunology , Immunotoxins/metabolism , Kinetics , Male , Mice , Mice, Nude , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Spheroids, Cellular/radiation effects , Substrate Specificity , Xenograft Model Antitumor Assays
11.
Cancer Res ; 60(18): 5237-43, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11016653

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a well-characterized cell surface antigen expressed by virtually all prostate cancers (PCas). PSMA has been successfully targeted in vivo with (111)In-labeled 7E11 monoclonal antibody (mAb; ProstaScint; Cytogen, Princeton, NJ), which binds to an intracellular epitope of PSMA. This work reports the in vitro characterization of three recently developed mAbs that bind the extracellular domain of PSMA (PSMAext). Murine mAbs J415, J533, J591, and 7E11 were radiolabeled with 131I and evaluated in competitive and saturation binding studies with substrates derived from LNCaP cells. J415 and J591 were conjugated to 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid labeled with (111)In. The uptake and cellular processing of these antibodies were evaluated in viable LNCaP cells. All four mAbs could be labeled with 131I up to a specific activity of 350 MBq/mg with no or little apparent loss of immunoreactivity. Competition assays revealed that J415 and J591 compete for binding to PSMAext antigen. J533 bound to a region close to the J591 binding epitope, but J533 did not interfere with J415 binding to PSMA. mAb 7E11 did not inhibit the binding of J415, J533, or J591 (or vice versa), consistent with earlier work that these latter mAbs bind PSMAext whereas 7E11 binds the intracellular domain of PSMA. Saturation binding studies demonstrated that J415 and J591 bound with a similar affinity (Kds 1.76 and 1.83 nM), whereas J533 had a lower affinity (Kd, 18 nM). In parallel studies, all four mAbs bound to a similar number of PSMA sites expressed by permeabilized cells (1,000,000-1,300,000 sites/cell). In parallel studies performed with viable LNCaP cells, J415, J533, and J591 bound to a similar number of PSMA sites (i.e., 600,000-800,000 sites/cell), whereas 7E11 bound only to a subpopulation of the available PSMA sites (95,000 sites/cell). This apparent binding of 7E11 to viable cells can be accounted for by a 5-7% subpopulation of permeabilized cells produced when the cells were trypsinized and suspended. Up to five DOTA chelates could be bound to either J415 or J591 without compromising immunoreactivity. A comparison of the cellular uptake and metabolic processing of the 131I- and (111)In-labeled antibodies showed a rapid elimination of 131I from the cell and a high retention of (111)In. All four mAbs recognized and bound to similar numbers of PSMAs expressed by ruptured LNCaP cells (i.e., the exposed intracellular and extracellular domains of PSMA). By comparison to J415 and J591, J533 had a lower binding affinity. Both J415 and J591 recognized and bound to the same high number of PSMAs expressed by intact LNCaP. By contrast, 7E11 bound to fewer sites expressed by intact LNCaP cells (i.e., the exposed extracellular domain of PSMA). Both J415 and J591 are promising mAbs for the targeting of viable PSMA-expressing tissue with diagnostic and therapeutic metallic radionuclides.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Carboxypeptidases/immunology , Immunoconjugates/immunology , Iodine Radioisotopes , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antibody Specificity , Antigens, Neoplasm/metabolism , Antigens, Surface/metabolism , Binding, Competitive , Carboxypeptidases/metabolism , Cell Membrane/metabolism , Chelating Agents/pharmacokinetics , Drug Stability , Glutamate Carboxypeptidase II , Humans , Immunoconjugates/metabolism , Immunoconjugates/pharmacokinetics , Indium Radioisotopes , Iodine Radioisotopes/therapeutic use , Isotope Labeling , Kinetics , Male , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , Quality Control , Tumor Cells, Cultured
12.
Curr Opin Urol ; 9(5): 391-5, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10579076

ABSTRACT

Over the past two decades, monoclonal antibody technology has had an increasing impact on clinical diagnostic and therapeutic options, and this is true in the realm of managing prostate cancer. Several targets such as prostate-specific antigen and prostatic acid phosphatase as well as, more recently, angiogenic antigens such as vascular endothelial growth factor have been examined for therapy. Prostate-specific membrane antigen, a type II integral membrane glycoprotein initially characterized by the monoclonal antibody 7E11, has shown promise. Recent evidence suggests that prostate-specific membrane antigen is also expressed in tumor-associated neovasculature of a wide variety of malignant neoplasms. With its expression in prostate secretory-acinar epithelium and the prostate and in the neovasculature associated with tumors, prostate-specific membrane antigen represents an excellent antigenic target for monoclonal antibody diagnostic and therapeutic options. As research continues, the role of monoclonal antibody imaging and therapy will become increasingly important in the management of prostate cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Prostate-Specific Antigen/immunology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Antibodies, Monoclonal/immunology , Antibody Specificity , Humans , Male , Organ Specificity
13.
Int J Cancer ; 83(4): 456-64, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10508479

ABSTRACT

The screening of cDNA expression libraries derived from human tumors with autologous antibody (SEREX) is a powerful method for defining the structure of tumor antigens recognized by the humoral immune system. Sixty-five distinct antigens (NY-REN-1 to NY-REN-65) reactive with autologous IgG were identified by SEREX analysis of 4 renal cancer patients and were characterized in terms of cDNA sequence, mRNA expression pattern, and reactivity with allogeneic sera. REN-9, -10, -19, and -26 have a known association with human cancer. REN-9 (LUCA-15) and REN-10 (gene 21) map to the small cell lung cancer tumor suppressor gene locus on chromosome 3p21.3. REN-19 is equivalent to LKB1/STK11, a gene that is defective in Peutz-Jeghers syndrome and cancer. REN-26 is encoded by the bcr gene involved in the [t(9:22)] bcr/abl translocation. Genes encoding 3 of the antigens in the series showed differential mRNA expression; REN-3 displays a pattern of tissue-specific isoforms, and REN-21 and REN-43 are expressed at a high level in testis in comparison to 15 other normal tissues. The other 62 antigens were broadly expressed in normal tissues. With regard to immunogenicity, 20 of the 65 antigens reacted only with autologous sera. Thirty-three antigens reacted with sera from normal donors, indicating that their immunogenicity is not restricted to cancer. The remaining 12 antigens reacted with sera from 5-25% of the cancer patients but not with sera from normal donors. Seventy percent of the renal cancer patients had antibodies directed against one or more of these 12 antigens. Our results demonstrate the potential of the SEREX approach for the analysis of the humoral immune response against human cancer.


Subject(s)
Antibodies, Neoplasm/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Aged , Antibody Specificity , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/isolation & purification , Blotting, Northern , Carcinoma, Renal Cell/genetics , Chromosome Mapping , Female , Gene Library , Humans , Kidney Neoplasms/genetics , Male , Middle Aged , Neoplasms/immunology , Organ Specificity , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Serologic Tests , Tumor Cells, Cultured
14.
Cancer ; 86(3): 492-7, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10430258

ABSTRACT

BACKGROUND: Using a reverse transcriptase-polymerase chain reaction (RT-PCR) assay, the authors previously determined the expression of MN/CA9 mRNA in renal cell carcinoma (RCC) and its absence in benign renal tissue. In the current study, the utility of an enhanced RT-PCR assay in the detection of renal carcinoma cells in the peripheral blood was assessed. METHODS: An enhanced MN/CA9 RT-PCR assay was applied to peripheral blood samples from a total of 96 patients. Forty-two patients had renal tumors, including 5 with benign renal lesions, 28 with localized RCC, and 9 with metastatic RCC. Fifty-four control patients without renal tumors were similarly tested. Pathologic staging for patients with localized cancer was T1N0M0 for 5, T2N0M0 for 9, and T3N0M0 for 14 patients. RESULTS: Cells expressing MN/CA9 were detected in 1 of 54 controls (1.8%) and in 18 of 37 cancer patients (49%). Thirteen of twenty eight patients (46%) with localized RCC and 5 of 9 (56%) with metastatic disease tested positive with the assay. No patient with a benign renal tumor exhibited MN/CA9 expression. All blood test results for patients with clear cell RCC were noted to be positive. No correlation was noted between MN/CA9 results and age, gender, or tumor grade. The differences in MN/CA9 results according to T classification were not statistically significant. CONCLUSIONS: The enhanced RT-PCR assay for MN/CA9 is a highly specific technique for detecting circulating renal carcinoma cells in the peripheral blood, and it may prove useful in the diagnosis and monitoring of RCC.


Subject(s)
Biomarkers, Tumor/analysis , Carbonic Anhydrases/analysis , Carcinoma, Renal Cell/chemistry , Isoenzymes/analysis , Kidney Neoplasms/chemistry , Neoplasm Proteins/analysis , Neoplastic Cells, Circulating/chemistry , Reverse Transcriptase Polymerase Chain Reaction/methods , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/secondary , Female , Humans , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplastic Cells, Circulating/pathology
15.
J Urol ; 162(2): 574-80, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10411090

ABSTRACT

PURPOSE: Multiple subtypes of renal cancer have been identified. Clear-cell renal cell carcinoma (RCC) is the most common subtype of RCC and one of the more aggressive. The goal of this study was to investigate in RCC the levels of Na,K-ATPase, an abundant enzyme in the kidney which is crucial for various kidney functions. Na,K-ATPase is a heterodimer consisting of a catalytic a-subunit and a glycosylated beta-subunit whose function is still not well-defined. MATERIALS AND METHODS: Fourteen clear-cell RCC specimens were studied. The levels of the Na,K-ATPase alpha and beta-subunits in normal kidney and RCC tissues were determined by immunoblot analysis. The localization of the alpha and beta-subunits was studied by immunofluorescence and laser scanning confocal microscopy. Na,K-ATPase activity was determined using a coupled-enzyme spectrophotometric assay. RESULTS: In normal kidney, the cells demonstrate an epithelial morphology with distinct basolateral plasma membrane localization of the alpha and beta-subunits. Conversely, the cells of the clear-cell RCC have lost their epithelial phenotype and the alpha and beta-subunits show a diffuse intracellular staining. Clear-cell RCC tumor cell lysates showed a consistent 95.6+/-2.8% (mean +/- SD) reduction in protein levels of beta-subunit relative to the levels in normal kidney. The alpha-subunit level in RCC lysates was generally near or above the levels relative to normal kidney. The reduced beta-subunit expression was accompanied by a significant reduction in the Na,K-ATPase activity in RCC membranes. CONCLUSIONS: These results suggest that the beta-subunit may regulate the Na,K-ATPase activity in vivo. Diminished Na,K-ATPase activity in conjunction with the reduced beta-subunit level is associated with the clear-cell RCC phenotype.


Subject(s)
Carcinoma, Renal Cell/enzymology , Kidney Neoplasms/enzymology , Sodium-Potassium-Exchanging ATPase/biosynthesis , Adult , Aged , Carcinoma, Renal Cell/pathology , Humans , Immunohistochemistry , Kidney Neoplasms/pathology , Middle Aged , Sodium-Potassium-Exchanging ATPase/analysis
16.
Cancer Res ; 59(13): 3192-8, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10397265

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a type II integral membrane glycoprotein that was initially characterized by the monoclonal antibody (mAb) 7E11. PSMA is highly expressed in prostate secretory-acinar epithelium and prostate cancer as well as in several extraprostatic tissues. Recent evidence suggests that PSMA is also expressed in tumor-associated neovasculature. We examined the immunohistochemical characteristics of 7E11 and those of four recently developed anti-PSMA mAbs (J591, J415, and Hybritech PEQ226.5 and PM2J004.5), each of which binds a distinct epitope of PSMA. Using the streptavidin-biotin method, we evaluated these mAbs in viable prostate cancer cell lines and various fresh-frozen benign and malignant tissue specimens. In the latter, we compared the localization of the anti-PSMA mAbs to that of the anti-endothelial cell mAb CD34. With rare exceptions, all five anti-PSMA mAbs reacted strongly with the neovasculature of a wide spectrum of malignant neoplasms: conventional (clear cell) renal carcinoma (11 of 11 cases), transitional cell carcinoma of the urinary bladder (6 of 6 cases), testicular embryonal carcinoma (1 of 1 case), colonic adenocarcinoma (5 of 5 cases), neuroendocrine carcinoma (5 of 5 cases), glioblastoma multiforme (1 of 1 cases), malignant melanoma (5 of 5 cases), pancreatic duct carcinoma (4 of 4 cases), non-small cell lung carcinoma (5 of 5 cases), soft tissue sarcoma (5 of 6 cases), breast carcinoma (5 of 6 cases), and prostatic adenocarcinoma (2 of 12 cases). Localization of the anti-PSMA mAbs to tumor-associated neovasculature was confirmed by CD34 immunohistochemistry in sequential tissue sections. Normal vascular endothelium in non-cancer-bearing tissue was consistently PSMA negative. The anti-PSMA mAbs reacted with the neoplastic cells of prostatic adenocarcinoma (12 of 12 cases) but not with the neoplastic cells of any other tumor type, including those of benign and malignant vascular tumors (0 of 3 hemangiomas, 0 of 1 hemangioendothelioma, and 0 of 1 angiosarcoma). The mAbs to the extracellular PSMA domain (J591, J415, and Hybritech PEQ226.5) bound viable prostate cancer cells (LNCaP and PC3-PIP), whereas the mAbs to the intracellular domain (7E11 and Hybritech PM2J004.5) did not. All five anti-PSMA mAbs reacted with fresh-frozen benign prostate secretory-acinar epithelium (28 of 28 cases), duodenal columnar (brush border) epithelium (11 of 11 cases), proximal renal tubular epithelium (5 of 5 cases), colonic ganglion cells (1 of 12 cases), and benign breast epithelium (8 of 8 cases). A subset of skeletal muscle cells was positive with 7E11 (7 of 7 cases) and negative with the other four anti-PSMA mAbs. PSMA was consistently expressed in the neovasculature of a wide variety of malignant neoplasms and may be an effective target for mAb-based antineovasculature therapy.


Subject(s)
Antigens, Surface , Carboxypeptidases/analysis , Carboxypeptidases/genetics , Neoplasms/blood supply , Neoplasms/enzymology , Neovascularization, Pathologic/enzymology , Prostate/enzymology , Antibodies, Monoclonal , Antigens, Neoplasm/analysis , Antigens, Neoplasm/genetics , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carboxypeptidases/immunology , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Female , Glutamate Carboxypeptidase II , Humans , Male , Neoplasms/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Testicular Neoplasms/blood supply , Testicular Neoplasms/enzymology , Testicular Neoplasms/pathology , Transfection , Tumor Cells, Cultured , Urinary Bladder Neoplasms/blood supply , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology
17.
Clin Cancer Res ; 5(5): 1219-29, 1999 May.
Article in English | MEDLINE | ID: mdl-10353760

ABSTRACT

Tumors may escape immune recognition and destruction through the induction of apoptosis in activated T lymphocytes. Results from several laboratories suggest that FasL (L/CD95L) expression in tumors may be responsible for this process. In this study of patients with renal cell carcinoma (RCC), we provide evidence for two mechanisms of T-cell apoptosis. One mechanism involves the induction of apoptosis via FasL expression in tumor cells. This is supported by several observations, including the fact that tumor cells in situ as well as cultured cell lines expressed FasL mRNA and protein by a variety of techniques. The FasL in RCC is functional because in coculture experiments, FasL+ tumors induced apoptosis in Fas-sensitive Jurkat T cells and in activated peripheral blood T cells but not in resting peripheral blood T cells. Most importantly, antibody to FasL partially blocked apoptosis of the activated T cells. Moreover, Fas was expressed by T cells derived from the peripheral blood (53% median) and tumor (44.3% median) of RCC patients. Finally, in situ staining for DNA breaks demonstrated apoptosis in a subset of T cells infiltrating renal tumors. These studies also identified a second mechanism of apoptosis in RCC patient peripheral T cells. Whereas these cells did not display DNA breaks when freshly isolated or after culture for 24 h in medium, peripheral blood T cells from RCC patients underwent activation-induced cell death after stimulation with either phorbol 12-myristate 13-acetate/ionomycin or anti-CD3/CD28 antibodies. Apoptosis mediated by exposure to FasL in tumor cells or through T-cell activation may contribute to the failure of RCC patients to develop an effective T-cell-mediated antitumor response.


Subject(s)
Apoptosis/physiology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/cytology , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , T-Lymphocytes, Cytotoxic/cytology , Apoptosis/drug effects , Blood Cells/immunology , Carcinoma, Renal Cell/blood , DNA Fragmentation , Fas Ligand Protein , Humans , In Situ Nick-End Labeling , Ionomycin/pharmacology , Jurkat Cells/immunology , Kidney Neoplasms/blood , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Muromonab-CD3/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , T-Lymphocytes, Cytotoxic/immunology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , fas Receptor/physiology
18.
Cancer Res ; 59(12): 2838-42, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10383143

ABSTRACT

The antitumor effect of immuno- and chemotherapeutic agents is executed through stimulation of apoptotic programs in susceptible cells. Apoptosis induced in tumor cells requires activation of members of the caspase family of proteases. Deficient expression or activation of caspases may account in part for the failure of many current anticancer therapies. However, recent studies suggest that cell death can proceed in the absence of caspases. We investigated the susceptibility of human renal cell carcinoma (RCC) lines to two distinct modes of cell death, apoptosis and necrosis. RCC lines displayed almost complete resistance to apoptosis in response to the intracellular zinc chelator, N,N,N'N'-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN), which instead induced dramatic accumulation of nonapoptotic necrotic cells. Conversely, TPEN was a potent inducer of apoptosis in caspase-competent normal kidney cells (NK-72) and Jurkat T lymphocytes. Resistance to apoptosis in RCC lines correlated with almost complete loss of caspase-3 expression and variable down-regulation of caspase-7, caspase-8, and caspase-10. These data may explain the resistance of RCC to drugs inducing apoptosis and have important consequences for further attempts to manipulate tumor cell death.


Subject(s)
Carcinoma, Renal Cell/pathology , Caspases/metabolism , Kidney Neoplasms/pathology , Apoptosis , Carcinoma, Renal Cell/enzymology , Caspases/deficiency , Cholinesterase Inhibitors/pharmacology , Ethylenediamines/pharmacology , Humans , Jurkat Cells , Kidney Neoplasms/enzymology , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Necrosis , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Tumor Cells, Cultured
19.
Neoplasia ; 1(2): 123-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10933046

ABSTRACT

The expression of immunoglobulin-based artificial receptors in normal T lymphocytes provides a means to target lymphocytes to cell surface antigens independently of major histocompatibility complex restriction. Such artificial receptors have been previously shown to confer antigen-specific tumoricidal properties in murine T cells. We constructed a novel zeta chain fusion receptor specific for prostate-specific membrane antigen (PSMA) termed Pz-1. PSMA is a cell-surface glycoprotein expressed on prostate cancer cells and the neovascular endothelium of multiple carcinomas. We show that primary T cells harvested from five of five patients with different stages of prostate cancer and transduced with the Pz-1 receptor readily lyse prostate cancer cells. Having established a culture system using fibroblasts that express PSMA, we next show that T cells expressing the Pz-1 receptor release cytokines in response to cell-bound PSMA. Furthermore, we show that the cytokine release is greatly augmented by B7.1-mediated costimulation. Thus, our findings support the feasibility of adoptive cell therapy by using genetically engineered T cells in prostate cancer patients and suggest that both CD4+ and CD8+ T lymphocyte functions can be synergistically targeted against tumor cells.


Subject(s)
Antigens, Surface , Carboxypeptidases/metabolism , Cytokines/biosynthesis , Gene Transfer Techniques , Prostatic Neoplasms/immunology , T-Lymphocytes/metabolism , 3T3 Cells , Aged , Aged, 80 and over , Animals , CD28 Antigens/metabolism , Cell Separation , Coculture Techniques , Epithelial Cells/metabolism , Fibroblasts/metabolism , Flow Cytometry , Glutamate Carboxypeptidase II , Humans , Male , Mice , Middle Aged , Prostatic Neoplasms/metabolism , Recombinant Fusion Proteins/metabolism , Retroviridae/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology
20.
Cancer Metastasis Rev ; 18(4): 483-90, 1999.
Article in English | MEDLINE | ID: mdl-10855791

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a cell surface glycoprotein that is expressed by prostate epithelial cells. PSMA-specific monoclonal antibodies have been utilized to characterize the biologic function and in vivo biodistribution of PSMA. PSMA is an attractive target protein for monoclonal antibody directed imaging or therapeutics for prostate cancer since its expression is relatively restricted to prostate epithelial cells and is over-expressed in prostate cancer, including in advanced stages. Currently, clinical usage of PSMA specific monoclonal antibodies has been limited to diagnostic immunohistochemistry and imaging of patients with prostate cancer. Novel applications for these antibodies will be discussed.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Surface , Carboxypeptidases/immunology , Neoplasms/drug therapy , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Antibody Specificity , Antigens, Neoplasm/immunology , Glutamate Carboxypeptidase II , Humans , Immunohistochemistry , Male , Neoplasms/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...