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










Publication year range
1.
Cancers (Basel) ; 3(2): 1821-43, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-24212784

ABSTRACT

Evidence for the efficacy of targeted alpha therapy for the control of pancreatic cancer in preclinical models is reviewed. Results are given for in vitro pancreatic cancer cells and clusters and micro-metastatic cancer lesions in vivo. Two complementary targeting vectors are examined. These are the C595 monoclonal antibody that targets the MUC1 antigen and the PAI2 ligand that targets the uPA receptor. The expression of the tumor-associated antigen MUC-1 and the uPA receptor on three pancreatic cancer cell lines is reported for cell clusters, human mouse xenografts and lymph node metastases, as well as for human pancreatic cancer tissues, using immuno-histochemistry, confocal microscopy and flow cytometry. The targeting vectors C595 and PAI2 were labeled with the alpha emitting radioisotope 213Bi using the chelators cDTPA and CHX-A″ to form the alpha-conjugates (AC). Cell clusters were incubated with the AC and examined at 48 hours. Apoptosis was documented using the TUNEL assay. In vivo, the anti-proliferative effect for tumors was tested at two days post-subcutaneous cell inoculation. Mice were injected with different concentrations of AC by local or systemic administration. Changes in tumor progression were assessed by tumor size. MUC-1 and uPA are strongly expressed on CFPAC-1, PANC-1 and moderate expression was found CAPAN-1 cell clusters and tumor xenografts. The ACs can target pancreatic cells and regress cell clusters (~100 µm diameter), causing apoptosis in some 70-90 % of cells. At two days post-cell inoculation in mice, a single local injection of 74 MBq/kg of AC causes complete inhibition of tumor growth. Systemic injections of 111, 222 and 333 MBq/kg of alpha-conjugate caused significant tumor growth delay in a dose dependent manner after 16 weeks, compared with the non-specific control at 333 MBq/kg. Cytotoxicity was assessed by the MTS and TUNEL assays. The C595 and PAI2-alpha conjugates are indicated for the treatment of micro-metastatic pancreatic cancer with over-expression of MUC1 and uPA receptors in post-surgical patients with minimal residual disease. The observation of tumor regression in a Phase I clinical trial of targeted alpha therapy for metastatic melanoma indicates that alpha therapy can regress tumors by a process called tumor anti-vascular alpha therapy (TAVAT). As a consequence, this therapy could be indicated for the management of non-surgical pancreatic cancer tumors.

2.
Cancer Biol Ther ; 7(1): 76-80, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18347423

ABSTRACT

PURPOSE: Control of ovarian cancer (OC) ascites remains a major objective in post-surgical treatment. The aim of this study was to investigate the effect of targeted alpha therapy (TAT) for the control of ascites in an OC ascites mouse model; the biodistribution of (213)Bi-C595 and its long term toxicity. METHODS: The expression of tumor-associated antigen mucin-1 (MUC-1) in OVCAR3 ascites cells in mice and OC cancer tissues in patients was detected by indirect immmunostaining. The monoclonal antibody (MAb) C595 was labeled with (213)Bi using the chelator cDTPA to form the alpha-immunoconjugate (AIC). Mice were injected with different concentrations of AIC by i.p administration. Changes in tumor progression were assessed by measurement of the circumference of the abdomen. RESULTS: MUC-1 is strongly expressed in 73% of OC tissues. At 9 days post-cell inoculation in mice, a single injection of 355 MBq/kg of (213)Bi-C595 can prolong survival by 25 days. A high tumor: blood ratio (5.8) was found in biodistribution study. The maximum tolerance dose (MTD) was more than 1180 MBq/kg up to 21 weeks. CONCLUSIONS: C595 is a specific targeting vector for ovarian cancer cells, which show a high percentage of expression of MUC1. (213)Bi-C595 can effectively target and kill ovarian cancer cells in vitro and in vivo. (213)Bi-C595 is the recommended alpha conjugate for a Phase I clinical trial for ovarian cancer.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Bismuth/therapeutic use , Mucin-1/immunology , Ovarian Neoplasms/radiotherapy , Radioimmunotherapy , Alpha Particles , Animals , Cell Line, Tumor , Female , Humans , Kidney/metabolism , Mice , Mice, Inbred BALB C , Mucin-1/analysis , Ovarian Neoplasms/mortality , Radioimmunotherapy/adverse effects , Tissue Distribution , Xenograft Model Antitumor Assays
3.
Phys Med Biol ; 53(2): 319-28, 2008 Jan 21.
Article in English | MEDLINE | ID: mdl-18184988

ABSTRACT

Ionizing radiation causes structural chromosomal aberrations, a proportion of which give rise to chromosome fragments without spindle attachment organelles. When a cell divides, some of these fragments are excluded from the main daughter nuclei and form small nuclei within the cytoplasm. The cytokinesis-block micronucleus assay allows these micronuclei (MN) to be counted, providing an in situ biological dosimeter. In this study, we evaluated the micronucleus frequency in peripheral blood lymphocytes after in vitro incubation with the alpha conjugates (213)BiI(3) and (213)Bi-9.2.27 (AIC). Lymphocytes were inoculated in vitro AIC for 3 h. Further, we report the first MN measurements in melanoma patients after targeted alpha therapy (TAT) with (213)Bi-9.2.27. Patients were injected with 260-360 MBq of AIC, and blood samples taken at 3 h, 2 weeks and 4 weeks post-treatment. Absorbed dose (MIRD) and effective total body dose (PED) were calculated. The MN frequency in lymphocytes was similar for equal in vitro incubation activities of (213)BiI(3) and (213)Bi-9.2.27 (P=0.5), indicating that there is no selective targeting of lymphocytes by the alpha conjugates. After inoculation with 10-1200 kBq mL-1 of AIC, there was a substantial activity-related increase in MN. The number of MN in the blood of treated patients peaked at 3 h post-TAT, slowly returning to baseline levels by 4 weeks. The mean photon equivalent dose (PED) is 0.43 Gy (SD 0.15) and the mean MIRD calculated absorbed dose is 0.11 Gy (SD 0.03), giving an RBE=4+/-0.4 for this study.


Subject(s)
Biological Assay/methods , Cytokinesis/radiation effects , Lymphocytes/cytology , Lymphocytes/radiation effects , Micronucleus Tests/methods , Radiometry/methods , Radiotherapy/methods , Aged , Aged, 80 and over , Alpha Particles/therapeutic use , Female , Humans , Male , Micronuclei, Chromosome-Defective/radiation effects , Middle Aged , Radiation Dosage , Relative Biological Effectiveness
4.
Cancer Biol Ther ; 6(6): 898-904, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17582216

ABSTRACT

OBJECTIVES: The plasminogen activator inhibitor type 2 (PAI2) when labelled with (213)Bi forms the (213)Bi-PAI2 alpha conjugate (AC). This AC has been shown to be efficacious in preclinical studies with breast, ovarian, prostate and pancreatic cancers. The objectives of this study were to investigate the pharmacokinetics and in vivo stability of (213)Bi-PAI2 in mice, its toxicity in mice and rabbits; and to determine whether a prior injection of a metal chelator (Ca-DTPA) or lysine can reduce toxicity by decreasing renal uptake. METHODS: Two chelators (CHX-A"-DTPA and cDTPA) were used for preparation of the (213)Bi-PAI2 conjugate, for intraperitoneal administration in mice and ear vein injection in rabbits. The mice were sacrificed at different time points for pharmacokinetic studies. Blood and organs were collected for toxicity studies for all groups. RESULTS: Both chelators were found to have similar %ID/g in the kidneys over four hours. Mice and rabbits did not show any short term toxicity over 13 weeks at 1420 MBq/kg and 120 MBq/kg (213)Bi-PAI2 respectively. Kidney uptake was decreased three fold by lysine. Radiation nephropathy was observed at 20-30 weeks in mice, leading to severe weight loss, whereas severe and widespread renal tubular necrosis was observed at 13 weeks in rabbits. CONCLUSIONS: Radiation nephropathy is the dose limiting toxicity observed in mice and rabbits. Lysine can reduce kidney uptake by three fold. Based on long-term monitoring, the maximum tolerance doses (MTD) are 350 and 120 MBq/kg for mice and rabbits respectively.


Subject(s)
Bismuth/pharmacokinetics , Bismuth/toxicity , Plasminogen Activator Inhibitor 2/pharmacokinetics , Plasminogen Activator Inhibitor 2/toxicity , Radioimmunotherapy/methods , Radioisotopes/toxicity , Alpha Particles , Animals , Chelating Agents/pharmacology , Female , Kidney/pathology , Medical Oncology/methods , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Rabbits , Radioisotopes/chemistry , Time Factors
5.
Prostate ; 66(16): 1753-67, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16955401

ABSTRACT

BACKGROUND: Micrometastasis is a major problem for prostate cancer (CaP) patients. Our study investigated the therapeutic potential of multiple targeted alpha-therapy (MTAT) in the treatment of CaP micrometastases (spheroids) using (213)Bi-labeled multiple targeted alpha-radioimmunoconjugates. METHODS: The expression of multiple tumor-associated antigens (TAAs) on frozen sections of human fresh CaP tissues and spheroids cultured from DU 145 and LNCaP-LN3 CaP cell lines was detected by immunohistochemistry and flow cytometry. Targeting vectors were two monoclonal antibodies (MAbs), and plasminogen activator inhibitor type 2 (PAI2) that binds to cell surface urokinase plasminogen activator (uPA). These vectors were labeled with (213)Bi using standard methodology. DU 145 and LNCaP-LN3 spheroids were incubated with different activities of test and control alpha-conjugates (ACs), and spheroid growth was measured for volume change and growth delay over a 50-day period using light microscopy. RESULTS: TAAs were expressed heterogeneously on frozen sections from human CaP tissues and CaP spheroids. MTAT combining three ACs (one-third dose of each) with an activity of 6.4 MBq/ml completely targeted small DU 145 and LNCaP-LN3 spheroids (diameter <100 microm) and slightly regressed the growth of medium spheroids (180-200 microm); MTAT with 2.2 or 4.8 MBq/ml activities delayed the growth of tumor spheroids. CONCLUSIONS: Our results suggest that the cytotoxicity of MTAT to CaP spheroids is highly dependent on antigenic expression, concentration of radioactivity and spheroid size. MTAT may be a potent therapeutic agent for micrometastases, effectively targeting small CaP cell clusters, and overcoming the heterogeneous expression of targeted antigens.


Subject(s)
Alpha Particles/therapeutic use , Antibodies, Monoclonal/administration & dosage , Bismuth/administration & dosage , Immunoconjugates/administration & dosage , Prostatic Neoplasms/radiotherapy , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Cell Line, Tumor , Flow Cytometry , Humans , Immunoconjugates/immunology , Immunohistochemistry , Male , Neoplasm Metastasis , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Spheroids, Cellular
6.
Cancer Lett ; 234(2): 176-83, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-15961220

ABSTRACT

The vectors PAI2, C595 and Herceptin target the membrane-bound uPA, MUC1 and HER2 antigens expressed by cancer cells, respectively. The expression of these receptors was tested in the ovarian cancer cell line OVCAR-3; MUC-1 was strongly expressed (3+), uPA moderately expressed (2+), but HER2 was negative (-). The alpha-emitting radionuclide Bismuth-213 was chelated with these targeting vectors to form alpha conjugates (ACs), the cytotoxicity of which were tested with OVCAR-3 cells. The PAI2 and C595 ACs are highly cytotoxic to the ovarian monolayer cancer cells and cell clusters in a concentration-dependent fashion and cause morphological changes of treated cancer cells, inducing apoptosis. These ACs are potential candidates for the control of ovarian cancer at the minimum residual disease (MRD) stage.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Bismuth/administration & dosage , Drug Delivery Systems/methods , Ovarian Neoplasms/drug therapy , Plasminogen Activator Inhibitor 2/administration & dosage , Radioisotopes/administration & dosage , Alpha Particles/therapeutic use , Antibodies, Monoclonal, Humanized , Female , Humans , Immunohistochemistry , Mucin-1/metabolism , Ovarian Neoplasms/metabolism , Radioisotopes/metabolism , Receptor, ErbB-2 , Spheroids, Cellular/drug effects , Trastuzumab , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
7.
Cancer Biol Ther ; 4(8): 848-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16082185

ABSTRACT

PURPOSE: The aim of this study was to investigate the effect of targeted alpha therapy for the control of in vitro pancreatic cancer cell clusters and micrometastatic cancer lesions in vivo. METHODS: The expression of tumor-associated antigen MUC-1 on three pancreatic cancer cell clusters and animal xenografts was detected by indirect immmunostaining. Monoclonal antibodies C595 (test) and A2 (non-specific control) were labeled with 213Bi using the chelator CHX.A" to form the alpha-immunoconjugate (AIC). Cell clusters were incubated with AIC and examined at 48 h. Apoptosis was documented using the TUNEL assay. In vivo, an antiproliferative effect for tumors was tested at two days post-subcutaneous cell inoculation. Mice were injected with different concentrations of AIC by regional or systemic administration. Changes in tumor progression were assessed by tumor size. RESULTS: MUC-1 is strongly expressed on CFPAC-1, PANC-1 and moderate expression was found CAPAN-1 cell clusters and tumor xenografts. The AICs can target and kill cancer cell clusters (100 mm) in vitro. Some 73-81 % of cells were TUNEL positive cells in the clusters after incubation with AIC. At two days post- cell inoculation in mice, a single local injection of 74 and 148 MBq/kg of AIC causes complete inhibition of tumor growth. Systemic injections of 111, 222 and 333 MBq/kg of AIC cause significant tumor growth delay after 16 weeks, compared with the nonspecific control providing 333 MBq/kg after 16 weeks. CONCLUSIONS: CFPAC-1, PANC-1 and CAPAN-1 pancreatic cancer cell clusters and pancreatic tumor xenografts show high expression of the MUC-1 target antigen. 213Bi-C595 can specifically target and regress pancreatic cancer cell clusters in vitro, and delay and inhibit tumor growth in vivo. 213Bi-C595 may be a useful agent for the treatment of micrometastatic pancreatic cancer with overexpression of MUC 1 antigen in post-surgical patients with minimal residual disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Isothiocyanates/therapeutic use , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/radiotherapy , Pentetic Acid/analogs & derivatives , Radioimmunotherapy , Animals , Antigens, Neoplasm , Bismuth/therapeutic use , Cell Line, Tumor , Humans , Mice , Mucin-1 , Mucins/analysis , Mucins/immunology , Pancreatic Neoplasms/chemistry , Pentetic Acid/therapeutic use , Xenograft Model Antitumor Assays
8.
Clin Exp Metastasis ; 22(7): 575-86, 2005.
Article in English | MEDLINE | ID: mdl-16475028

ABSTRACT

PURPOSE: The urokinase plasminogen activator (uPA) and its receptor (uPAR) are expressed by pancreatic cancer cells and can be targeted by the plasminogen activator inhibitor type 2 (PAI2). We have labeled PAI2 with (213)Bi to form the alpha conjugate (AC), and have studied its in vitro cytotoxicity and in vivo efficacy. METHODS AND MATERIALS: The expression of uPA/uPAR on pancreatic cell lines, human pancreatic cancer tissues, lymph node metastases, and mouse xenografts were detected by immunohistochemistry, confocal microscopy, and flow cytometry. Cytotoxicity was assessed by the MTS and TUNEL assay. At 2 days post-cancer cell subcutaneous inoculation, mice were injected with AC by local or systemic injection. RESULTS: uPA/uPAR is strongly expressed on pancreatic cancer cell lines and cancer tissues. The AC can target and kill cancer cells in vitro in a concentration-dependent fashion. Some 90% of TUNEL positive cells were found after incubation with 1.2 MBq/ml of AC. A single local injection of approximately 222 MBq/kg 2 days post-cell inoculation can completely inhibit tumor growth over 12 weeks, and an intraperitoneal injection of 111 MBq/kg causes significant tumor growth delay. CONCLUSIONS: (213)Bi-PAI2 can specifically target pancreatic cancer cells in vitro and inhibit tumor growth in vivo. (213)Bi-PAI2 may be a useful agent for the treatment of post-surgical pancreatic cancer patients with minimum residual disease.


Subject(s)
Bismuth/therapeutic use , Neoplasm Metastasis/radiotherapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/radiotherapy , Plasminogen Activator Inhibitor 2/therapeutic use , Radioisotopes/therapeutic use , Animals , Cell Line, Tumor , Cell Survival/radiation effects , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Microscopy, Confocal , Radiopharmaceuticals/therapeutic use , Transplantation, Heterologous , Urokinase-Type Plasminogen Activator/metabolism
9.
Int J Radiat Oncol Biol Phys ; 60(3): 896-908, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15465208

ABSTRACT

PURPOSE: Control of metastatic prostate cancer (CaP) is an elusive objective. Some 30% of patients with clinically localized CaP will develop micrometastatic disease. Defining the expression of tumor-associated antigens on CaP will enable appropriate selection of therapeutic targets. METHODS AND MATERIALS: The expression of tumor-associated antigens on CaP cell lines (PC-3, DU 145, and LNCaP-LN3) was detected by immunohistochemistry and flow cytometry. Test and control alpha-conjugates were prepared using monoclonal antibodies, an inhibitor, plasminogen activator inhibitor type 2, that binds to the cell-membrane-bound protease, urokinase plasminogen activator, and a control protein labeled with (213)Bi using standard methods. These were used singly or together against three different CaP cell lines in vitro. The cytotoxicity of the alpha-conjugates was assessed using the [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt] (MTS) assay. RESULTS: The PC-3 and DU 145 cancer cell lines expressed antigens that bind monoclonal antibodies BLCA-38 and #394 (mouse anti-human urokinase plasminogen activator B-chain) but not J591. The LNCaP-LN3 cells bound J591 but not #394 or BLCA-38. For the PC-3, DU 145, and LNCaP-LN3 cell lines, multiple-targeted alpha-therapy combining four alpha-conjugates (one-quarter doses of each) gave D(0) (37% cell survival) values of 15, 17, and 27 microCi/mL compared with those of the controls of 272, 289, and 281 microCi/mL, respectively. CONCLUSION: Metastatic prostate cancer-associated antigens recognized by multiple monoclonal antibodies are potential targets for alpha-therapy. Multiple-targeted alpha-therapy produced cytotoxicity specific to three CaP cell lines and may form the basis of treatment for micrometastatic CaP, overcoming the heterogeneity of expression of the targeted antigens.


Subject(s)
Antigens, Neoplasm/analysis , Bismuth/therapeutic use , Prostatic Neoplasms/immunology , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy/methods , Radioisotopes/therapeutic use , Antibodies, Monoclonal/therapeutic use , Apoptosis , Cell Line, Tumor/immunology , Cell Proliferation/radiation effects , Humans , Immunoconjugates/therapeutic use , Male , Plasminogen Activator Inhibitor 2/therapeutic use , Prostatic Neoplasms/pathology
10.
Cancer Lett ; 205(2): 161-71, 2004 Mar 18.
Article in English | MEDLINE | ID: mdl-15036648

ABSTRACT

HER-2 has been implicated in the oncogenesis of human prostate cancer (CaP) and is the target of a new treatment for metastatic breast cancer using the humanised monoclonal antibody (MAb) trastuzumab (Herceptin). In this study, a novel alpha-particle emitting [213Bi]Herceptin construct, which targets the HER-2 extracellular domain on CaP cells, was prepared and evaluated in vitro. We used immunocytochemistry, flow cytometry and Western blot analysis to examine the expression of HER-2 in a panel of established human CaP cell lines, used the MTS assay to evaluate the cytotoxicity of 213Bi-Herceptin on these cell lines and the TUNEL assay to document apoptosis. The results indicate that LNCaP-LN3 cells express high levels of HER-2 protein, in contrast, DU 145 cells express low to medium levels, and PC-3 cells express an undetectable level of HER-2 protein. 213Bi-Herceptin was specifically cytotoxic to LNCaP-LN3 cells in a concentration-dependent fashion, cause the cells to undergo apoptosis, whereas DU 145 showed an HER-2 level-dependent response to 213Bi-Herceptin cytotoxicity. In contrast, PC-3 cells were resistant to 213Bi-Herceptin-induced cytotoxicity. The 213Bi-Herceptin induced apoptosis in LNCaP-LN3 cells could be inhibited by incubation with unlabeled Herceptin. Our results suggest that 213Bi-Herceptin alpha-conjugate might be a promising new agent for the treatment of preangiogenic cancer cell clusters or micro-metastases with high levels of HER-2 expression.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Apoptosis/radiation effects , Bismuth/therapeutic use , Prostatic Neoplasms/radiotherapy , Radioimmunotherapy , Radioisotopes/therapeutic use , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/pathology , Receptor, ErbB-2/analysis , Trastuzumab
SELECTION OF CITATIONS
SEARCH DETAIL
...