Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Breast Cancer Res Treat ; 98(1): 7-15, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16724166

ABSTRACT

Research into the interaction between the E. coli heat-stable enterotoxin (STh) and the guanylin receptor guanylate cyclase C (GC-C) has generated >100 synthetic analogs of the peptide, several of which have been investigated as imaging or therapeutic agents for colorectal cancers. The evidence presented here suggests that in addition to STh binding to GC-C expressing cell lines derived from human colon, STh also specifically binds to an as yet unidentified receptor expressed in high densities on the surface of cell lines derived from human breast cancers. In vitro whole-cell crosslinking studies using 125I-labeled F19-STh(1-19) demonstrate that the putative STh binding protein migrates as an approximately 120-125 kDa species by SDS-PAGE, significantly smaller than the glycosylated GC-C molecule found in the T84 human colon cancer cell line. RT-PCR using total RNA isolated from breast and colon cancer cell lines indicates that GC-C transcripts are undetectable in human breast cancer cell lines and abundant in human colon cancer cell lines. In vitro competitive binding studies using STh analogs and the estrogen receptor positive (ER+) T-47D cell line demonstrated IC50 values between 2.6 and 8.5 nM. Similar studies on the estrogen receptor negative (ER-) cell line MDA-MB-231 showed IC50's between 5.6 and 9.9 nM. Saturation binding analysis revealed receptor expression to fall between 40,000 and 120,000 sites per cell in these cell lines, receptor abundances equal to or greater than the abundance of GC-C in colorectal cancer cell lines. STh binding to these cells, although of similar affinity to STh binding to GC-C, is distinguishable from it on the basis of its ligand specificity. The characteristics of STh analogs as radiopharmaceutical agents were tested in an in vivo model utilizing T-47D human breast cancer cell xenografts in SCID mice. Clearance of STh analogs was rapid, primarily via renal excretion into the urine, with >85% ID excreted into the urine at 1 h p.i. Tumor uptake at 1 h p.i. in T-47D tumor cell xenografts was 0.67+/-0.23% ID/g, and was significantly decreased (p<0.05) upon co-administration of 4 mg/kg unlabeled STh. These results suggest that STh may find application for the imaging and treatment of breast cancer.


Subject(s)
Breast Neoplasms/therapy , Drug Screening Assays, Antitumor , Enterotoxins/therapeutic use , Escherichia coli/metabolism , Indium Radioisotopes/pharmacology , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Cross-Linking Reagents/pharmacology , Female , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Mice , Mice, SCID , Neoplasm Transplantation , Protein Binding
2.
J Nucl Med ; 46(1): 121-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15632042

ABSTRACT

UNLABELLED: The purpose of this study was to examine the therapeutic efficacy of (188)Re-(Arg(11))[Cys(3,4,10),d-Phe(7)]alpha-melanocyte-stimulating hormone(3-13) (CCMSH) in the B16/F1 murine melanoma- and TXM13 human melanoma-bearing mouse models. METHODS: (Arg(11))CCMSH was synthesized and labeled with (188)Re to form (188)Re-(Arg(11))CCMSH. B16/F1 melanoma-bearing mice were administrated 7.4 MBq, 22.2 MBq, and 2 x 14.8 MBq of (188)Re-(Arg(11))CCMSH via the tail vein. TXM13 melanoma-bearing mice were separately injected with 22.2 MBq, 2 x 14.8 MBq, and 37.0 MBq of (188)Re-(Arg(11))CCMSH through the tail vein. Two groups of 10 mice bearing either B16/F1 or TXM13 tumors were injected with saline as untreated controls. RESULTS: In contrast to the untreated control group, (188)Re-(Arg(11))CCMSH yielded rapid and lasting therapeutic effects in the treatment groups with either B16/F1 or TXM13 tumors. The tumor growth rate was reduced and the survival rate was prolonged in the treatment groups. Treatment with 2 x 14.8 MBq of (188)Re-(Arg(11))CCMSH significantly extended the mean life of B16/F1 tumor mice (P < 0.05), whereas the mean life of TXM13 tumor mice was significantly prolonged after treatment with 22.2-MBq and 37.0- MBq doses of (188)Re-(Arg(11))CCMSH (P < 0.05). High-dose (188)Re-(Arg(11))CCMSH produced no observed normal tissue toxicity. CONCLUSION: The therapy study results revealed that (188)Re-(Arg(11))CCMSH yielded significant therapeutic effects in both B16/F1 murine melanoma- and TXM13 human melanoma-bearing mouse models. (188)Re-(Arg(11))CCMSH appears to be a promising radiolabeled peptide for targeted radionuclide therapy of melanoma.


Subject(s)
Drug Delivery Systems/methods , Melanoma/metabolism , Melanoma/radiotherapy , Rhenium/pharmacokinetics , Rhenium/therapeutic use , alpha-MSH/pharmacokinetics , alpha-MSH/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Metabolic Clearance Rate , Mice , Mice, Inbred C57BL , Organ Specificity , Radioisotopes/pharmacokinetics , Radioisotopes/therapeutic use , Radiopharmaceuticals/therapeutic use , Radiotherapy Dosage , Survival Analysis , Tissue Distribution , Treatment Outcome
3.
Mol Biol Rep ; 31(2): 121-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15293788

ABSTRACT

In vivo phage display is a new approach to acquire peptide molecules that bind stably to a given target. Phage peptide display libraries have been selected in mice and humans and numerous vasculature-targeting peptides have been reported. However, in vivo phage display has not typically produced molecules that extravasate to target specific organ or tumor antigens. Phage selections in animals have been performed for very short times without optimization for biodistribution or clearance rates to a particular organ. It is hypothesized that peptides that home to a desired antigen/organ can be obtained from in vivo phage experiments by optimization of incubation times, phage extraction and propagation procedures. To accomplish this goal, one must first gain a better understanding of the in vivo biodistribution and rate of clearance of engineered phage peptide display libraries. While the fate of wild type phage in rodents has been reported, the in vivo biodistribution of the commonly used engineered fd-tet M13 phage peptide display libraries (such as in the fUSE5 vector system) have not been well established. Here we report the biodistribution and clearance properties of fd-tet fifteen amino acid random peptide display libraries in fUSE5 phage in three common mouse models employed for drug discovery - CF-1, nude, and SCID mice.


Subject(s)
Bacteriophage M13/metabolism , Peptide Library , Animals , Bacteriophage M13/isolation & purification , Mice , Mice, Inbred Strains , Tissue Distribution
4.
Bioconjug Chem ; 15(4): 872-80, 2004.
Article in English | MEDLINE | ID: mdl-15264876

ABSTRACT

Three human Escherichia coli heat-stable peptide (STh) analogues, each containing a DOTA chelating group, were synthesized by SPPS and oxidative refolding and compared in in vitro and in vivo systems. One analogue, DOTA-F19-STh(1-19), contains an N-terminal DOTA group attached via an amide bond linkage to an STh moiety which is essentially wild-type except for a Tyr to Phe alteration at position 19 of the molecule. A second analogue, DOTA-R1,4,F19-STh(1-19), differs from the first in that asparagine residues in positions 1 and 4 have been altered to arginine residues in order to examine the effect of positively charged groups in the linker domain. A third analogue, DOTA-11AUN-F19-STh(1-19), differs from the first in that it incorporates an 11-aminoundecanoic acid spacer group between the DOTA group and the first asparagine residue. In vitro competitive binding assays utilizing T-84 human colon cancer cells demonstrated that significant alterations to the N-terminal region of the STh molecule were well tolerated and did not significantly affect binding affinity of STh for the guanylyl cyclase C (GC-C) receptor. Internalization and efflux studies of the indium-labeled species demonstrated that inclusion of positive charge in the linker moiety inhibits internalization of the compound within tumor cells. The characteristics of the three analogues were compared in an in vivo model utilizing T-84 human colon cancer cell xenografts in SCID mice. Clearance of all analogues was rapid, primarily via renal excretion into the urine, with >89% ID excreted into the urine at 1 h pi for all analogues. The 111In-DOTA-R1,4,F19-STh(1-19) and 111In-DOTA-11AUN-F19-STh(1-19) analogues both had longer residence times in the blood than did the 111In-DOTA-F19-STh(1-19) analogue, probably accounting for increased %ID/g values for tumors and nontarget tissues at 1 h pi. At 4 h pi, significant differences between analogues were only seen with respect to metabolic routes of excretion, indicating that increased blood residence time did not result in increased tumor residualization. Reduction of hepatic uptake of these compounds, however, could have significance in the development of agents for the imaging of hepatic metastases. The ability to manipulate in vivo pharmacodynamics and tumor uptake of radiolabeled STh peptides through modification of linker moieties is under continuing investigation in order to produce optimal imaging and therapeutic radiopharmaceuticals.


Subject(s)
Chelating Agents/chemistry , Escherichia coli Proteins/chemistry , Hot Temperature , Animals , Cell Line, Tumor , Chelating Agents/metabolism , Chelating Agents/pharmacokinetics , Chromatography, High Pressure Liquid , Escherichia coli Proteins/administration & dosage , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/pharmacokinetics , Female , Humans , Indium Radioisotopes , Inhibitory Concentration 50 , Mice , Molecular Structure , Molecular Weight , Neoplasm Transplantation , Protein Binding , Protein Denaturation , Protein Renaturation , Tissue Distribution
5.
Nucl Med Biol ; 31(2): 213-23, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15013487

ABSTRACT

The radiolanthanides (149)Pm, (166)Ho, and (177)Lu possess a range of half-lives and alpha(-) beta(-) energies for targeted radiotherapy of cancer. (149)Pm-, (166)Ho-, and (177)Lu-DOTA-biotin were pretargeted to LS174T colorectal tumors in nude mice with CC49 scFvSA antibody-streptavidin fusion protein. Tumor uptakes of (149)Pm (22.9% ID/g), (166)Ho (30.2% ID/g), and (177)Lu (35.4% ID/g) peaked at 1-4 h. Rapid blood disappearance was accompanied by urinary excretion of 59-66% ID within 1 h. Biodistributions of these agents show promise for pretargeted radioimmunotherapy of cancer.


Subject(s)
Antibodies, Neoplasm/metabolism , Biotin/analogs & derivatives , Biotin/pharmacokinetics , Colonic Neoplasms/metabolism , Drug Delivery Systems/methods , Organometallic Compounds/pharmacokinetics , Radioimmunotherapy/methods , Radioisotopes/pharmacokinetics , Streptavidin/pharmacokinetics , Animals , Antibodies, Neoplasm/administration & dosage , Biotin/administration & dosage , Cell Line, Tumor , Colonic Neoplasms/radiotherapy , Holmium/administration & dosage , Holmium/pharmacokinetics , Humans , Immunoglobulin Fragments/administration & dosage , Immunoglobulin Fragments/metabolism , Injections, Intravenous , Lutetium/administration & dosage , Lutetium/pharmacokinetics , Metabolic Clearance Rate , Mice , Mice, Nude , Organ Specificity , Organometallic Compounds/administration & dosage , Promethium/administration & dosage , Promethium/pharmacokinetics , Radioisotopes/administration & dosage , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacokinetics , Streptavidin/administration & dosage , Tissue Distribution
6.
Bioconjug Chem ; 14(6): 1177-84, 2003.
Article in English | MEDLINE | ID: mdl-14624632

ABSTRACT

The purpose of this study was to evaluate the human MC1 receptor-mediated melanoma targeting properties of two metal cyclized alpha-MSH peptide analogues, (188)Re-(Arg(11))CCMSH and (188)Re-CCMSH. Initially, the presence and density of the MC1 receptor were determined on a bank of human melanoma cell lines. All eight human melanoma cell lines tested in this study displayed the MC1 receptor at a density of 900 to 5700 receptors per cell. Receptor affinity and biodistribution properties of (188)Re-(Arg(11))CCMSH and (188)Re-CCMSH were evaluated in a cultured TXM13 human melanoma-xenografted Scid mouse model. Biodistribution results demonstrated that 3.06 +/- 0.68 %ID/g of (188)Re-(Arg(11))CCMSH accumulated in the tumors 1 h postinjection and greater than 65% of the activity at 1 h postinjection remained in the tumors at 4 h after dose administration. Whole body clearance of (188)Re-(Arg(11))CCMSH was very rapid, with approximately 82% of injected dose cleared through urinary system at 4 h postinjection. There was very little activity in blood and major organs such as liver, lung, and muscle except for the kidney. (188)Re-CCMSH exhibited similar tumor uptake and retention in TXM13 human melanoma-xenografted Scid mice as (188)Re-(Arg(11))CCMSH. However, the kidney uptake value of (188)Re-CCMSH was two times higher than that of (188)Re-(Arg(11))CCMSH. The results of this study indicate that the MC1 receptor is present on the surface of a large number of human melanoma cells, which makes the MC1 receptor a good imaging or therapeutic target. Moreover, the biodistribution properties of (188)Re-(Arg(11))CCMSH and (188)Re-CCMSH highlight their potential as therapeutic agents for human melanoma.


Subject(s)
Antineoplastic Agents/metabolism , Drug Delivery Systems , Melanoma, Experimental/drug therapy , Peptide Fragments/metabolism , Receptor, Melanocortin, Type 1/metabolism , Rhenium/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Humans , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Structure , Organotechnetium Compounds/chemistry , Organotechnetium Compounds/pharmacokinetics , Peptide Fragments/chemistry , Peptide Fragments/therapeutic use , Rhenium/pharmacokinetics , Structure-Activity Relationship , Tissue Distribution , Xenograft Model Antitumor Assays
7.
Cancer Res ; 63(14): 4082-8, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12874010

ABSTRACT

Bombesin (BBN), a 14 amino acid peptide, is an analogue of human gastrin-releasing peptide (GRP) that binds to GRP receptors (GRPrs) with high affinity and specificity. The GRPr is overexpressed on a variety of human cancer cells, including prostate, breast, lung, and pancreatic cancers. The specific aim of this study was to develop (99m)Tc(I)-radiolabled BBN analogues that maintain high specificity for the GRPr in vivo. A preselected synthetic sequence via solid phase peptide synthesis was designed to produce 2,3-diaminopropionic acid (Dpr)-BBN conjugates with the following general structure: Dpr-Ser-Ser-Ser-Gln-Trp-Ala-Val-Gly-His-Leu-Met-(NH(2)). The new BBN constructs were purified by reversed phase high-performance liquid chromatography. Electrospray mass spectrometry was used to characterize the nonmetallated BBN conjugates. Re(I)-BBN conjugates were prepared by the reaction of [Re(Br)(3)(CO)(3)](2-) and Dpr-Ser-Ser-Ser-Gln-Trp-Ala-Val-Gly-His-Leu-Met-(NH(2)) with gentle heating. Electrospray mass spectrometry was used to determine the molecular constitution of the new Re(I) conjugates. The (99m)Tc conjugates were prepared at the tracer level by preconjugation, postlabeling approach from the reaction of [(99m)Tc(H(2)O)(3)(CO)(3)](+) and corresponding ligand. The (99m)Tc and Re(I) conjugates behaved similarly under identical reversed phase high-performance liquid chromatography conditions. Results from in vitro and in vivo models demonstrated the ability of these derivatives to specifically target GRPrs on human, prostate, cancerous PC-3 cells.


Subject(s)
Bombesin/analogs & derivatives , Organotechnetium Compounds/chemical synthesis , Organotechnetium Compounds/pharmacokinetics , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptors, Bombesin/metabolism , beta-Alanine/analogs & derivatives , beta-Alanine/chemistry , Amino Acid Sequence , Animals , Bombesin/pharmacokinetics , Female , Humans , Isotope Labeling/methods , Male , Mice , Mice, Inbred ICR , Mice, SCID , Radionuclide Imaging , Spectrometry, Mass, Electrospray Ionization , beta-Alanine/pharmacokinetics
8.
J Nucl Med ; 44(5): 823-31, 2003 May.
Article in English | MEDLINE | ID: mdl-12732685

ABSTRACT

UNLABELLED: Gastrin-releasing peptide (GRP) receptors have been shown to be expressed with high densities on several types of cancer cells including prostate, breast, small cell lung, and pancreas cancers. Bombesin (BBN) has been known to bind to GRP receptors with high affinity and specificity. The aim of these studies was to develop new (111)In-labeled BBN analogs having high tumor uptake and optimal pharmacokinetics for specific targeting of human prostate cancers. METHODS: A novel series of dodecanetetraacetic acid (DOTA)-X-BBN[7-14]NH(2) (X = 0, beta-Ala, 5-Ava, 8-Aoc, or 11-Aun) conjugates and their In(III)/(111)In complexes exhibiting high GRP-receptor-binding affinities were synthesized and characterized. RESULTS: In vitro competitive binding assays, using PC-3 androgen-independent human prostate cancer cells, demonstrated values of <2.5 nmol/L for inhibitory concentration of 50% for analogs with beta-Ala, 5-Ava, and 8-Aoc spacers. In vivo biodistribution studies of the (111)In-DOTA-X-BBN[7-14]NH(2) conjugates performed on CF-1 mice at 1 h after injection have revealed that the uptake of radioactivity in the pancreas, a GRP-receptor-expressing tissue, increased as a function of hydrocarbon spacer length (i.e., from 0.20 +/- 0.04 percentage injected dose [%ID] per gram for the analog with no spacer to a maximum of 26.97 +/- 3.97 %ID/g for the analog with 8-Aoc spacer). The radioactivity was cleared efficiently from the blood pool by excretion mainly through the renal/urinary pathway (e.g., 71.6 +/- 1.8 %ID at 1 h after injection for 8-Aoc spacer analog). In vivo pharmacokinetic studies of the (111)In-DOTA-8-Aoc-BBN[7-14]NH(2) conjugate conducted on PC-3 human prostate cancer-derived xenografts in SCID mice showed a specific uptake of radioactivity in tumor, with 3.63 +/- 1.11 %ID/g observed at 1 h after injection. High tumor-to-blood and tumor-to-muscle ratios of approximately 6:1 and 45:1, respectively, were achieved at 1 h after injection. Relative to the radioactivity observed in the tumor at 1 h after injection, 43%, 19%, and 9% of the radioactivity was retained at, respectively, 24, 48, and 72 h after injection. CONCLUSION: These studies showed that radiometallated DOTA-X-BBN[7-14]NH(2) constructs with hydrocarbon spacers ranging from 5 to 8 carbon atoms are feasible candidates for further development as diagnostic and therapeutic radiopharmaceuticals for patients with GRP-positive cancers.


Subject(s)
Indium Radioisotopes , Prostatic Neoplasms/diagnostic imaging , Radiopharmaceuticals , Receptors, Bombesin/analysis , Animals , Female , Humans , Male , Mice , Mice, Inbred ICR , Mice, SCID , Radionuclide Imaging , Tissue Distribution , Tumor Cells, Cultured
9.
Anticancer Res ; 23(1A): 63-70, 2003.
Article in English | MEDLINE | ID: mdl-12680195

ABSTRACT

BACKGROUND: Bombesin (BBN), a 14 amino acid peptide, is an analogue of human gastrin-releasing peptide (GRP) that binds to GRP receptors (GRPr) with high affinity and specificity. The GRPr is over-expressed on a variety of human cancer cells including prostate, breast, lung, and pancreatic cancers. The specific aim of this study was to develop a 188Re(I)-radiolabeled BBN analogue that maintains high specificity for the GRPr in vivo. MATERIALS AND METHODS: A preselected synthetic sequence via solid phase peptide synthesis (SPPS) was designed to produce a Dpr-BBN (Dpr = Diaminopropionic acid) conjugate with the following general structure: Dpr-X-Q-W-A-V-G-H-L-M-(NH2), where the spacer group, X = Serylserylserine. The new BBN-construct was purified by reversed phase-HPLC (RP-HPLC). The non-radioactive Re(I)-BBN conjugate was prepared by the reaction of [Re(Br)3(CO)3]2- and Dpr-SSS-bombesin(7-14)NH2 with heating. ES-MS was used to determine the molecular constitution of the non-metallated and metallated Re (I)--conjugates. The 188 Re-conjugate was prepared at the tracer level by the pre-conjugation, postlabeling approach from the reaction of [188Re(H2O)3(CO)3]+ and corresponding ligand. RESULTS: The 188Re- and non-radioactive Re(I)conjugate behaved similarly under identical RP-HPLC conditions. In vitro cell displacement assays showed that the new conjugate has an IC50 value of approximately 1 nM. In vitro cell binding assays showed that the new conjugate is rapidly internalized and exhibits long-term retention, demonstrating the agonistic efficacy of the radiolabel. In vivo targeting of human prostate, PC-3 tumor xenografts indicated uptake and retention of the new radioconjugate for time-point < or = 24 hours. CONCLUSION: Results from in vitro and in vivo models demonstrated the ability of these derivatives to specifically target GRP receptors on human, prostate and cancerous PC-3 cells. This new construct holds potential for the development of a therapeutic entity for the treatment of prostate cancer.


Subject(s)
Bombesin/analogs & derivatives , Bombesin/chemical synthesis , Bombesin/pharmacokinetics , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacokinetics , Radioisotopes/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptors, Bombesin/metabolism , Rhenium/chemistry , Animals , Bombesin/metabolism , Humans , Isotope Labeling/methods , Male , Mice , Mice, SCID , Organometallic Compounds/metabolism , Prostatic Neoplasms/metabolism , Radiopharmaceuticals/metabolism , Rhenium/metabolism , Tissue Distribution , Xenograft Model Antitumor Assays
10.
Nucl Med Biol ; 30(2): 101-9, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12623108

ABSTRACT

Bombesin (BBN), a 14 amino acid peptide, is an analogue of human gastrin releasing peptide (GRP) that binds to GRP receptors (GRPr) with high affinity and specificity. The GRPr is over expressed on a variety of human cancer cells including prostate, breast, lung, and pancreatic cancers. The specific aim of this study was to identify a BBN analogue that can be radiolabeled with (177)Lu and maintains high specificity for GRPr positive prostate cancer tumors in vivo. A preselected synthetic sequence via solid phase peptide synthesis (SPPS) was designed to produce a DOTA-BBN (DOTA = 1,4,7,10-tetraazacyclododecane-N,N',N",N"'-tetraacetic acid) conjugate with the following general structure: DOTA-X-Q-W-A-V-G-H-L-M-(NH(2)), where the spacer group, X = omega-NH(2)(CH(2))(7)COOH (8-Aoc). The BBN-construct was purified by reversed phase-HPLC (RP-HPLC). Electrospray Mass Spectrometry (ES-MS) was used to characterize both metallated and non-metallated BBN-conjugates. The new DOTA-conjugate was metallated with (177)Lu(III)Cl(3) or non-radioactive Lu(III)Cl(3). The (177)Lu(III)- and non-radiolabeled Lu(III)-conjugates exhibit the same retention times under identical RP-HPLC conditions. The (177)Lu-DOTA-8-Aoc-BBN[7-14]NH(2) conjugate was found to exhibit optimal pharmacokinetic properties in CF-1 normal mice. In vitro and in vivo models demonstrated the ability of the (177)Lu-DOTA-8-Aoc-BBN[7-14]NH(2) conjugate to specifically target GRP receptors expressed on PC-3 human prostate cancer cells.


Subject(s)
Biomarkers, Tumor/metabolism , Bombesin/pharmacokinetics , Peptide Fragments/pharmacokinetics , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/metabolism , Receptors, Bombesin/metabolism , Animals , Bombesin/blood , Bombesin/chemical synthesis , Female , Humans , Isotope Labeling/methods , Male , Metabolic Clearance Rate , Mice , Mice, Inbred ICR , Mice, SCID , Neoplasm Transplantation , Organ Specificity , Peptide Fragments/blood , Peptide Fragments/chemical synthesis , Prostatic Neoplasms/blood , Radiometry/methods , Radionuclide Imaging , Radiopharmaceuticals/blood , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
11.
Int J Cancer ; 101(5): 480-7, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12216078

ABSTRACT

The purpose of our study was to optimize melanoma tumor uptake of 188Re-CCMSH and reduce its nonspecific kidney retention. Nephrotoxicity is often a serious problem associated with targeted radiotherapy, therefore, increasing the tumor/kidney uptake ratio of 188Re-CCMSH is crucial for optimizing its therapeutic efficacy. Structural modification of the peptide and amino acid co-infusion were investigated as strategies to improve the tumor/kidney uptake ratio of 188Re-CCMSH. The substitution of Lys11 with Arg11 was examined to determine if removal of lysine from the peptide would improve kidney clearance without sacrificing tumor uptake. The pharmacokinetics of 188Re-CCMSH and 188Re-(Arg(11))CCMSH were determined in B16/F1 murine melanoma-bearing C57 mice. Tumor uptake values of (188)Re-CCMSH and 188Re-(Arg(11))CCMSH were 15.03 +/- 5.20% ID/g and 20.44 +/- 1.91% ID/g at 1 hr postinjection and 1.94 +/- 0.47% ID/g and 3.50 +/- 2.32% ID/g at 24 hr postinjection. Renal retention of 188Re-(Arg(11))CCMSH was 11.79 +/- 1.29 ID/g and 3.67 +/- 0.51 ID/g at 1 hr and 4 hr postinjection, which was a greater than 50% reduction compared to 188Re-CCMSH. The Arg for Lys substitution in 188Re-(Arg(11))CCMSH resulted in improved tumor uptake and reduced kidney retention. Renal retention of both 188Re-CCMSH and 188Re-(Arg(11))CCMSH were significantly reduced by co-injection of 20 mg of L-lysine, L-arginine and a combination of L-lysine:L-arginine. Tumor/kidney uptake values for 188Re-CCMSH and 188Re-(Arg(11))CCMSH were maximally reduced by 52.9% and 46.3%, respectively. However, even with amino acid co-injection, the tumor/kidney ratio of 188Re-CCMSH was lower than that of 188Re-(Arg(11))CCMSH. Improved tumor uptake and reduced kidney retention of 188Re-(Arg(11))CCMSH will facilitate targeted irradiation of melanoma tumors while minimizing the dose to the kidneys.


Subject(s)
Melanoma/radiotherapy , Radiopharmaceuticals/therapeutic use , Rhenium/pharmacokinetics , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacokinetics , Amino Acid Sequence , Biological Transport , Humans , Kidney/metabolism , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Oligopeptides/therapeutic use , Radiopharmaceuticals/pharmacokinetics , Rhenium/therapeutic use , Structure-Activity Relationship , Tissue Distribution , alpha-MSH/therapeutic use
12.
J Med Chem ; 45(14): 3048-56, 2002 Jul 04.
Article in English | MEDLINE | ID: mdl-12086490

ABSTRACT

Rhenium-cyclized CCMSH analogues are novel melanoma-targeting metallopeptides with high tumor uptake, long tumor retention, and low background in normal tissues, which make these metallopeptides an ideal structural motif for designing novel melanoma-targeting agents. ReCCMSH has been derivatized with a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelate so that it can be labeled with a wide variety of radionuclides for imaging and therapeutic applications. This study involved optimization of the in vivo biological properties of DOTA-ReCCMSH (S), through modification of the structure of the metallopeptide. Several DOTA-ReCCMSH analogues, Ac-Lys(DOTA)-ReCCMSH (4) DOTA-ReCCMSH(Arg(11)) (6), DOTA-ReCCMSH-OH (8), and DOTA-ReCCMSH-Asp-OH (10), were synthesized using solid phase peptide synthesis followed by rhenium cyclization. The IC(50) values of the metallopeptides were determined through competitive binding assays against (125)I-(Tyr(2))-NDP. Radiolabeling of the DOTA-rhenium-cyclized peptides with (111)In was carried out in NH(4)OAc (0.1 M; pH 5.5)-buffered solution for 30 min at 70 degrees C. The stability of the radiolabeled complexes was evaluated in 0.01 M, pH 7.4, phosphate-buffered saline/0.1% bovine serum albumin solution. After separation of the radiolabeled peptide from the unlabeled peptide by reverse phase high-performance liquid chromatography, the biodistribution of the radiolabeled complex was performed in C57 mice bearing B16/F1 murine melanoma tumors. All radiolabeled complexes showed fast blood clearance (2 h postinjection (pi): (111)In-S, 0.07 +/- 0.03% ID/g; (111)In-4, 0.09 +/- 0.06% ID/g; (111)In-6, 0.21 +/- 0.08% ID/g; (111)In-8, 0.11 +/- 0.10% ID/g; and (111)In-10, 0.05 +/- 0.03% ID/g), and their clearance was predominantly through the urine (4 h pi: 93.5 +/- 1.7, 87.8 +/- 6.5, 89.8 +/- 4.2, 93.3 +/- 1.1, and 93.8 +/- 1.8 (% ID) for (111)In-labeled S, 4, 6, 8, and 10, respectively). Tumor uptake values of 9.45 +/- 0.90, 6.01 +/- 2.36, 17.41 +/- 5.61, 9.27 +/- 0.68, and 7.32 +/- 2.09 (% ID/g) for (111)In-labeled S, 4, 6, 8, and 10, respectively, were observed at 4 h pi. The kidney uptake was 9.27 +/- 2.65% ID/g for (111)In-S, 19.02 +/- 2.63% ID/g for (111)In-4, 7.37 +/- 1.13% ID/g for (111)In-6, 8.70 +/- 0.88% ID/g for (111)In-8, and 8.13 +/- 1.47% ID/g for (111)In-10 at 4 h pi. Complex 6 showed high melanoma uptake and lower kidney uptake than the corresponding Lys(11) analogues, supporting 6 for further investigations as a potential therapeutic radiopharmaceutical.


Subject(s)
Heterocyclic Compounds, 1-Ring/chemistry , Indium Radioisotopes , Organometallic Compounds/chemical synthesis , Peptides, Cyclic/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Rhenium , alpha-MSH/analogs & derivatives , alpha-MSH/chemical synthesis , Animals , Binding, Competitive , Isotope Labeling , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacokinetics , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured , alpha-MSH/chemistry , alpha-MSH/pharmacokinetics
13.
Bioconjug Chem ; 13(2): 224-31, 2002.
Article in English | MEDLINE | ID: mdl-11906259

ABSTRACT

New human Escherichia coli heat-stable peptide (ST(h)) analogues containing a DOTA chelating group were synthesized by sequential and selective formation of disulfides bonds in the peptide. This synthetic approach utilizes three orthogonal thiol-protecting groups, Trt, Acm, and t-Bu, to form three disulfide bonds by successive reactions using 2-PDS, iodine, and silyl chloride-sulfoxide systems. The DOTA-ST(h) conjugates exhibiting high guanylin/guanylate cyclase-C (GC-C) receptor binding affinities were obtained with >98% purity. In vitro competitive binding assays, employing T-84 human colon cancer cells, demonstrated the IC(50) values of <2 nM for GC-C receptor binding suggesting that the new synthetic ST(h) analogues are biologically active. In vitro stability studies of the (111)In-DOTA-Phe(19)-ST(h) conjugate incubated in human serum at 37 degrees C under 5% CO(2) atmosphere revealed that this conjugate is extremely stable with no observable decomposition at 24 h postincubation. HPLC analysis of mouse urine at 1 h pi of the (111)In-DOTA-Phe(19)-ST(h) conjugate showed only about 15% decomposition suggesting that the (111)In-DOTA-Phe(19)-ST(h) conjugate is highly stable, even under in vivo conditions. In vivo pharmacokinetic studies of the (111)In-DOTA-Phe(19)-ST(h) conjugate in T-84 human colon cancer derived xenografts in SCID mice conducted at 1 h pi showed an initial tumor uptake of 2.04 +/- 0.30% ID/g at 1 h pi with efficient clearance from the blood pool (0.23 +/- 0.14% ID/g, 1 h pi) by excretion mainly through the renal/urinary pathway (95.8 +/- 0.2% ID, 1 h pi). High tumor/blood, tumor/muscle, and tumor/liver ratios of approximately 9:1, 68:1, and 26:1, respectively, were achieved at 1 h pi The specific in vitro and in vivo uptake of the radioactivity by human colonic cancer cells highlights the potential of radiometalated-DOTA-ST(h) conjugates as diagnostic/therapeutic radiopharmaceuticals.


Subject(s)
Bacterial Toxins/chemical synthesis , Bacterial Toxins/pharmacokinetics , Colonic Neoplasms/drug therapy , Disulfides/metabolism , Enterotoxins/chemical synthesis , Enterotoxins/pharmacokinetics , Escherichia coli/chemistry , Heterocyclic Compounds, 1-Ring/chemistry , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Animals , Bacterial Toxins/metabolism , Bacterial Toxins/therapeutic use , Binding, Competitive , Chromatography, High Pressure Liquid , Colonic Neoplasms/metabolism , Enterotoxins/metabolism , Enterotoxins/therapeutic use , Escherichia coli Proteins , Female , Humans , Indium Radioisotopes/chemistry , Inhibitory Concentration 50 , Mice , Mice, SCID , Molecular Structure , Neoplasm Transplantation , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/therapeutic use , Tumor Cells, Cultured , Tumor Protein, Translationally-Controlled 1
SELECTION OF CITATIONS
SEARCH DETAIL