64Cu-Labeled Inhibitors of Prostate-Specific Membrane Antigen for PET Imaging of Prostate Cancer
- Sangeeta Ray Banerjee
- ,
- Mrudula Pullambhatla
- ,
- Catherine A. Foss
- ,
- Sridhar Nimmagadda
- ,
- Riccardo Ferdani
- ,
- Carolyn J. Anderson
- ,
- Ronnie C. Mease
- , and
- Martin G. Pomper
Abstract

Prostate-specific membrane antigen (PSMA) is a well-recognized target for identification and therapy of a variety of cancers. Here we report five 64Cu-labeled inhibitors of PSMA, [64Cu]3–7, which are based on the lysine–glutamate urea scaffold and utilize a variety of macrocyclic chelators, namely NOTA(3), PCTA(4), Oxo-DO3A(5), CB-TE2A(6), and DOTA(7), in an effort to determine which provides the most suitable pharmacokinetics for in vivo PET imaging. [64Cu]3–7 were prepared in high radiochemical yield (60–90%) and purity (>95%). Positron emission tomography (PET) imaging studies of [64Cu]3–7 revealed specific accumulation in PSMA-expressing xenografts (PSMA+ PC3 PIP) relative to isogenic control tumor (PSMA– PC3 flu) and background tissue. The favorable kinetics and high image contrast provided by CB-TE2A chelated [64Cu]6 suggest it as the most promising among the candidates tested. That could be due to the higher stability of [64Cu]CB-TE2A as compared with [64Cu]NOTA, [64Cu]PCTA, [64Cu]Oxo-DO3A, and [64Cu]DOTA chelates in vivo.
Introduction
Results
Chemical and Radiochemical Syntheses
Macrocyclic Chelator-Conjugated PSMA Inhibitors
Figure 1

Figure 1. Proposed structures of 64Cu-labeled inhibitors of PSMA.
Scheme 1

Scheme 2

Radiochemistry
yield (%) | Log Poct/w | Ki (nM) | ||
---|---|---|---|---|
3 | 2.84 | |||
[64Cu]3 | ∼65–70 | –1.17 | [63/65Cu]3 | 6.23 |
4 | 2.03 | |||
[64Cu]4 | ∼70–90 | –1.42 | [63/65Cu]4 | 10.76 |
5 | 3.10 | |||
[64Cu]5 | ∼75–85 | –1.56 | [63/65Cu]5 | 5.47 |
6 | 0.19 | |||
[64Cu]6A | ∼40–45 | –2.68 | [63/65Cu]6A | 3.98 |
[64Cu]6B | ∼30–35 | –2.31 | [63/65Cu]6B | 4.65 |
7 | 11.07 | |||
[64Cu]7 | ∼65–70 | ND | [63/65Cu ]7 | 13.26 |
In Vitro Binding
Small Animal PET-CT Imaging
Figure 2

Figure 2. Whole body PET-CT imaging of PSMA+ PC3 PIP and PSMA – PC3 flu tumor-bearing mice with compounds [64Cu]3, [64Cu]4, [64Cu]5, [64Cu]6B, and [64Cu]7 at 2.5 h postinjection, respectively. Mice were injected with ∼11.1 MBq (∼300 μCi) of radiotracer IV. PIP = PSMA+ PC3 PIP (solid arrow); flu = PSMA– PC3 flu (unfilled arrow); K= kidney; B = bladder; L = left; R = right. All images are decay-corrected and adjusted to the same maximum value.
Figure 3

Figure 3. Whole body PET-CT imaging of PC3 PIP and PC3 flu tumor bearing mice with [64Cu]3 at 20 min (left), 6 h (middle), 28 h (right). Abdominal radioactivity is primarily due to uptake within kidneys and bladder. PIP = PC3 PSMA+ PIP (solid arrow); flu = PC3 PSMA– flu (unfilled arrow); K= kidney; L = left; R = right, B = bladder. All images are decay-corrected and adjusted to the same maximum value.
Figure 4

Figure 4. Whole body PET-CT imaging of PC3 PIP and PC3 flu tumor bearing mice with [64Cu]6A (top row) and [64Cu]6B (bottom row) at 20 min, 2.5 h, 12 and 22 h postinjection. Abdominal radioactivity is primarily due to uptake within kidneys and bladder. PIP = PC3 PSMA+ PIP (solid arrow); flu = PC3 PSMA− flu (unfilled arrow); K= kidney; L = left; R = right, B = bladder. All images are decay-corrected and adjusted to the same maximum value.
Biodistribution
0.5 h | 1 h | 2 h | 4 h | |
---|---|---|---|---|
blood | 2.37 ± 0.90 | 1.12 ± 0.21 | 0.82 ± 0.20 | 0.45 ± 0.20 |
heart | 1.78 ± 0.41 | 1.39 ± 0.24 | 1.11 ± 0.13 | 0.71 ± 0.26 |
lung | 6.40 ± 0.59 | 5.99 ± 0.70 | 4.51 ± 0.97 | 2.49 ± 0.99 |
liver | 7.37 ± 0.40 | 7.50 ± 1.43 | 6.44 ± 0.83 | 4.18 ± 1.61 |
spleen | 28.02 ± 5.82 | 27.58 ± 10.05 | 12.33 ± 6.32 | 4.72 ± 1.49 |
fat | 1.78 ± 0.31 | 1.43 ± 0.38 | 0.76 ± 0.34 | 0.65 ± 0.75 |
kidney | 195.44 ± 29.82 | 233.39 ± 22.76 | 199.69 ± 48.44 | 108.05 ± 20.50 |
muscle | 0.57 ± 0.32 | 0.60 ± 0.14 | 0.34 ± 0.12 | 0.21 ± 0.10 |
small intestine | 4.60 ± 1.42 | 5.58 ± 1.43 | 3.68 ± 0.41 | 2.44 ± 1.30 |
large intestine | 4.51 ± 1.64 | 6.01 ± 2.54 | 3.80 ± 1.67 | 2.42 ± 1.19 |
bladder | 5.65 ± 2.77 | 2.48 ± 0.42 | 13.33 ± 8.35 | 1.70 ± 0.97 |
PC-3 PIP | 33.79 ± 9.68 | 29.11 ± 3.02 | 38.51 ± 5.68 | 20.64 ± 5.06 |
PC-3 flu | 2.40 ± 0.17 | 2.02 ± 0.18 | 1.92 ± 0.23 | 1.04 ± 0.37 |
PIP:flu | 14.03 ± 3.71 | 14.37 ± 0.43 | 19.96 ± 0.81 | 23.50 ± 4.65 |
PIP:blood | 15.11 ± 5.38 | 26.64 ± 5.12 | 48.75 ± 10.27 | 50.01 ± 15.54 |
Values expressed are in % ID/g ± standard deviation; n = 4 for all tissues.
0.5 h | 1 h | 2 h | 5 h | |
---|---|---|---|---|
blood | 2.50 ± 1.08 | 0.64 ± 0.24 | 0.26 ± 0.13 | 0.06 ± 0.05 |
heart | 0.92 ± 0.27 | 0.32 ± 0.08 | 0.13 ± 0.05 | 0.06 ± 0.01 |
lung | 2.70 ± 0.73 | 0.83 ± 0.11 | 0.45 ± 0.21 | 0.15 ± 0.01 |
liver | 0.73 ± 0.17 | 0.42 ± 0.04 | 0.30 ± 0.06 | 0.21 ± 0.03 |
stomach | 1.02 ± 0.47 | 0.48 ± 0.28 | 0.25 ± 0.06 | 0.10 ± 0.03 |
pancreas | 0.90 ± 0.23 | 0.31 ± 0.19 | 0.13 ± 0.05 | 0.05 ± 0.04 |
spleen | 5.80 ± 2.61 | 2.14 ± 0.58 | 0.65 ± 0.15 | 0.25 ± 0.07 |
fat | 0.87 ± 0.15 | 0.87 ± 1.32 | 0.19 ± 0.10 | 0.05 ± 0.05 |
kidney | 256.11 ± 75.72 | 148.60 ± 48.41 | 65.39 ± 21.16 | 3.54 ± 0.04 |
muscle | 0.33 ± 0.01 | 1.71 ± 2.29 | 0.17 ± 0.05 | 0.05 ± 0.02 |
small intestine | 0.85 ± 0.41 | 1.38 ± 1.89 | 0.26 ± 0.07 | 0.11 ± 0.10 |
large intestine | 0.88 ± 0.42 | 0.77 ± 0.65 | 0.30 ± 0.08 | 0.14 ± 0.05 |
bladder | 3.99 ± 0.26 | 7.06 ± 3.75 | 2.21 ± 0.57 | 1.43 ± 1.41 |
PC-3 PIP | 23.14 ± 2.20 | 29.50 ± 8.10 | 20.46 ± 2.90 | 18.69 ± 2.88 |
PC-3 flu | 1.29 ± 0.12 | 0.66 ± 0.26 | 0.26 ± 0.04 | 0.13 ± 0.03 |
PIP:flu | 19.36 ± 3.98 | 48.41 ± 14.25 | 79.40 ± 15.83 | 146.79 ± 32.30 |
PIP:blood | 9.13 ± 5.23 | 47.47 ± 7.24 | 101.17 ± 6.70 | 1123.96 ± 25.83 |
PIP:muscle | 69.35 ± 4.29 | 91.30 ± 93.17 | 132.99 ± 43.97 | 436.14 ± 202.71 |
Values expressed are in % ID/g ± standard deviation; n = 4 for all tissues.
[64Cu]3 | [64Cu]4 | [64Cu]6A | [64Cu]6B | |
---|---|---|---|---|
blood | 1.06 ± 0.29 | 1.87 ± 0.63 | 0.20 ± 0.03 | 0.20 ± 0.07 |
heart | 1.48 ± 0.14 | 2.71 ± 0.13 | 0.26 ± 0.10 | 0.35 ± 0.21 |
lung | 5.31 ± 0.74 | 10.16 ± 2.29 | 1.17 ± 0.61 | 0.89 ± 0.15 |
liver | 8.63 ± 0.92 | 17.04 ± 1.44 | 1.68 ± 0.38 | 1.63 ± 0.72 |
stomach | 3.88 ± 0.82 | 6.37 ± 0.78 | 0.62 ± 0.42 | 0.62 ± 0.17 |
pancreas | 2.22 ± 0.34 | 3.51 ± 0.67 | 0.46 ± 0.44 | 0.80 ± 0.92 |
spleen | 13.42 ± 1.18 | 9.27 ± 2.28 | 0.39 ± 0.30 | 0.90 ± 0.23 |
fat | 0.76 ± 0.77 | 1.18 ± 0.59 | 0.27 ± 0.26 | 0.05 ± 0.02 |
kidney | 125.40 ± 42.47 | 166.57 ± 42.39 | 23.55 ± 8.96 | 24.87 ± 11.26 |
muscle | 0.33 ± 0.08 | 0.71 ± 0.32 | 0.11 ± 0.05 | 0.12 ± 0.05 |
small intestine | 6.24 ± 1.46 | 10.95 ± 3.70 | 0.74 ± 0.32 | 0.68 ± 0.13 |
large intestine | 10.13 ± 6.83 | 10.10 ± 4.76 | 1.49 ± 0.75 | 1.28 ± 0.70 |
bladder | 12.47 ± 10.30 | 10.56 ± 6.20 | 7.98 ± 8.32 | 5.92 ± 4.94 |
PC-3 PIP | 16.58 ± 3.15 | 24.13 ± 10.06 | 11.10 ± 5.42 | 16.89 ± 5.73 |
PC-3 flu | 2.12 ± 0.25 | 3.88 ± 0.62 | 0.36 ± 0.09 | 0.40 ± 0.12 |
PIP:flu | 11.00 ± 0.73 | 6.22 ± 1.86 | 30.90 ± 7.92 | 41.75 ± 5.54 |
PIP:blood | 16.52 ± 5.14 | 12.87 ± 1.87 | 52.41 ± 17.34 | 86.97 ± 15.22 |
PIP:muscle | 46.98 ± 19.05 | 38.34 ± 21.69 | 97.31 ± 42.46 | 138.88 ± 30.86 |
Values expressed are in % ID/g ± standard deviation; n = 4 for all tissues.
Discussion
Conclusion
Experimental Section
Synthesis and Characterization of Compounds 1–7
9,16,24-Trioxo-1-thioxo-1-((4-(((S)-1,4,7-tris(carboxymethyl)-1,4,7-triazonan-2-yl)methyl)phenyl)amino)-2,8,17,23,25-pentaazaoctacosane-7,22,26,28-tetracarboxylic Acid, Compound 3
9,16,24-Trioxo-1-thioxo-1-((4-(((4S)-3,6,9-tris(carboxymethyl)-3,6,9,15-tetraazabicyclo[9.3.1]pentadeca-1(14),12-dien-4-yl)methyl)phenyl)amino)-2,8,17,23,25-pentaazaoctacosane-7,22,26,28-tetracarboxylic Acid, Compound 4
9,16,24-Trioxo-1-thioxo-1-((4-((4,7,10-tris(carboxymethyl)-1-oxa-4,7,10-triazacyclododecan-2-yl)methyl)phenyl)amino)-2,8,17,23,25-pentaazaoctacosane-7,22,26,28-tetracarboxylic Acid, Compound 5
(14S,29S,33S)-4,7-Dibenzyl-1-(11-(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecan-4-yl)-2,5,8,16,23,31-hexaoxo-3,6,9,15,24,30,32-heptaazapentatriacontane-14,29,33,35-tetracarboxylic Acid, Compound 6
Copper Complexes of Ligand 3, 4, 5, 6, and 7
64Cu Radiolabeling
NAALADase Assay
Cell Lines
Tumor Models
Small-Animal PET Imaging and Analysis
Ex Vivo Biodistribution
Data Analysis
Supporting Information
Detailed spectral data and supporting PET-CT blocking study image. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
We thank the University of Wisconsin Cyclotron Research Group for providing [64Cu]CuCl2. We also thank Nordion and Macrocyclics, Inc., for providing PCTA-Bn-SCN and Oxo-DO3A-Bn-SCN chelating agent, Drs. Cara Ferreira and Russell Redshaw for helpful discussion, and James Fox, Gilbert Green, and Dr. Yuchuan Wang for assistance with imaging and image analysis. We are grateful for the following sources of support: K25 CA148901, NIH NCI U54CA151838, NCI CA134675, and NCI R01 CA093375.
PSMA | prostate-specific membrane antigen |
GCPII | glutamate carboxypeptidase II |
NAALADase | N-acetylated-α-linked acidic dipeptidase |
PET | positron emission tomography |
SPECT | single photon emission computed tomography |
NOTA | 1,4,7-triazacyclononane-1,4,7-triacetic acid |
CB-TE2A | 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane |
PCTA | 3,6,9,15-tetraazabicyclo[9.3.1]-pentadeca-1(15),11,13-triene)-3,6,9-triacetic acid |
oxo-DO3A | oxa-4,7,1-tetraazacyclododecane-4,7,10-triacetic acid |
DOTA | 1,4,7,10-tetraazacyclododecane-N,N′,N″,N‴-tetraacetic acid |
References
This article references 69 other publications.
- 1Ghosh, A.; Heston, W. D. Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer J. Cell Biochem. 2004, 91, 528– 539Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsVWjs70%253D&md5=d5b2b495ac34449feb6a33e85b3de264Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancerGhosh, Arundhati; Heston, Warren D. W.Journal of Cellular Biochemistry (2004), 91 (3), 528-539CODEN: JCEBD5; ISSN:0730-2312. (Wiley-Liss, Inc.)A review. Prostate specific membrane antigen (PSMA), is a unique membrane bound glycoprotein, which is overexpressed manifold on prostate cancer as well as neovasculature of most of the solid tumors, but not in the vasculature of the normal tissues. This unique expression of PSMA makes it an important marker as well as a large extracellular target of imaging agents. PSMA can serve as target for delivery of therapeutic agents such as cytotoxins or radionuclides. PSMA has two unique enzymic functions, folate hydrolase and NAALADase and found to be recycled like other membrane bound receptors through clathrin coated pits. The internalization property of PSMA leads one to consider the potential existence of a natural ligand for PSMA. In this review we have discussed the regulation of PSMA expression within the cells, and significance of its expression in prostate cancer and metastasis.
- 2Rajasekaran, A. K.; Anilkumar, G.; Christiansen, J. J. Is prostate-specific membrane antigen a multifunctional protein? Am. J. Physiol., Cell Physiol. 2005, 288, C975– C981Google ScholarThere is no corresponding record for this reference.
- 3Chang, S. S.; Reuter, V. E.; Heston, W. D.; Gaudin, P. B. Comparison of anti-prostate-specific membrane antigen antibodies and other immunomarkers in metastatic prostate carcinoma Urology 2001, 57, 1179– 1183Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD387hslOrtg%253D%253D&md5=d07a48bed4ec39043f399679688bef1fComparison of anti-prostate-specific membrane antigen antibodies and other immunomarkers in metastatic prostate carcinomaChang S S; Reuter V E; Heston W D; Gaudin P BUrology (2001), 57 (6), 1179-83 ISSN:.OBJECTIVES: To compare the immunohistochemical properties of the 7E11 anti-prostate-specific membrane antigen (anti-PSMA) monoclonal antibody (mAb) with the recently developed anti-PSMA mAb, PM2J004.5, and with other common immunomarkers in metastatic prostate cancer. PSMA is a type II integral membrane glycoprotein highly expressed in prostate cancer cells. The mAb 7E11 is currently used in the radioisotopic evaluation of prostate cancer, and its immunohistochemical properties have been examined in primary prostate cancer specimens. METHODS: We examined 23 formalin-fixed, paraffin-embedded, metastatic prostate carcinoma specimens from various anatomic sites, including bone, lymph node, liver, lung, and soft tissue. Using the biotin-streptavidin method, we performed immunohistochemical reactions with the anti-PSMA mAbs 7E11 and PM2J004.5 and with antibodies to prostate-specific antigen and prostatic acid phosphatase. The immunoreactions were scored by pathologists unaware of the clinical and pathologic data according to a staining intensity scale and the percentage of cells stained. RESULTS: All four mAbs consistently stained the metastatic prostate cancer specimens. In 2 (8.7%) of 23 cases, however, the prostate-specific antigen immunoreaction was negative but the anti-PSMA mAbs had positive staining. Although 7E11 and PM2J004.5 had a similar staining intensity and percentage of cells stained for most specimens, in 3 (13%) of 23 specimens, 7E11 had less intense staining. None of the specimens were negative for all four antibodies. CONCLUSIONS: Anti-PSMA mAbs consistently immunoreacted with metastatic prostate cancer specimens and were positive in instances when prostate-specific antigen staining was negative. The anti-PSMA mAbs demonstrated similar staining patterns; however, in select cases, the PM2J004.5 mAb did show more intense staining. The anti-PSMA mAbs 7E11 and PM2J004.5 are useful in the pathologic evaluation of paraffin-embedded metastatic prostate cancer specimens.
- 4Wright, G. L., Jr.; Grob, B. M.; Haley, C.; Grossman, K.; Newhall, K.; Petrylak, D.; Troyer, J.; Konchuba, A.; Schellhammer, P. F.; Moriarty, R. Upregulation of prostate-specific membrane antigen after androgen-deprivation therapy Urology 1996, 48, 326– 334Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28zlt1OgsQ%253D%253D&md5=0bc36d48df2fd48360407a9f33267d3bUpregulation of prostate-specific membrane antigen after androgen-deprivation therapyWright G L Jr; Grob B M; Haley C; Grossman K; Newhall K; Petrylak D; Troyer J; Konchuba A; Schellhammer P F; Moriarty RUrology (1996), 48 (2), 326-34 ISSN:0090-4295.OBJECTIVES: To determine the expression of prostate-specific membrane antigen (PSMA) before and after androgen-deprivation therapy and to compare PSMA expression with prostate-specific antigen (PSA) expression. METHODS: We studied specimens from 20 patients with prostate cancer undergoing medical or surgical castration or combination androgen-deprivation therapy in whom matched pretreatment and post-treatment tissue specimens were available and 16 patients in whom only a post-treatment specimen was available. The expression of PSMA and PSA in the tissue specimens was determined by immunoperoxidase staining. The extent of staining was calculated by multiplying the percent of antigen-positive tumor cells by the staining intensity to arrive at a stain index for each biomarker. An in vitro study assessed the concentration of PSMA and PSA in extracts of LNCaP cells cultured in the presence or absence of androgen as determined by immunoassays and Western blot analysis. RESULTS: PSMA reactivity was found to be increased in 55% (11 of 20) of post-treatment primary tissues and 100% (4 of 4) of post-treatment metastatic specimens. In contrast, PSA expression was found to be decreased in 70% (14 of 20) of post-treatment primary and 100% (4 of 4) of post-treatment metastatic specimens. Neither type of androgen-deprivation treatment nor tissue sensitivity to androgen deprivation appeared to influence degree of biomarker expression. PSMA was found to be downregulated and PSA upregulated when LNCaP cells were cultured in the presence of testosterone or dihydrotestosterone. CONCLUSIONS: The enhanced expression of PSMA in tissues and LNCaP cells after androgen deprivation suggests that PSMA is upregulated in the majority of prostate carcinomas after androgen treatment. The high expression in metastatic tissues strongly suggests that PSMA may be a clinically useful target for antibody-and genetic-directed therapy of prostate cancer that recurs after androgen deprivation. The mechanism whereby androgens suppress the expression of PSMA, and the association of PSMA with the development of hormone-independent prostate cancers, will require further study.
- 5Evans, M. J.; Smith-Jones, P. M.; Wongvipat, J.; Navarro, V.; Kim, S.; Bander, N. H.; Larson, S. M.; Sawyers, C. L. Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 9578– 9582Google ScholarThere is no corresponding record for this reference.
- 6Perner, S.; Hofer, M. D.; Kim, R.; Shah, R. B.; Li, H.; Moller, P.; Hautmann, R. E.; Gschwend, J. E.; Kuefer, R.; Rubin, M. A. Prostate-specific membrane antigen expression as a predictor of prostate cancer progression Human Pathol. 2007, 38, 696– 701Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXkt1ajtLY%253D&md5=d26f9f8635836a05ab6bab48a802893dProstate-specific membrane antigen expression as a predictor of prostate cancer progressionPerner, Sven; Hofer, Matthias D.; Kim, Robert; Shah, Rajal B.; Li, Haojie; Moeller, Peter; Hautmann, Richard E.; Gschwend, Juergen E.; Kuefer, Rainer; Rubin, Mark A.Human Pathology (2007), 38 (5), 696-701CODEN: HPCQA4; ISSN:0046-8177. (Elsevier Inc.)Distinguishing aggressive prostate cancer from indolent disease represents an important clin. challenge, because current therapy may lead to overtreatment of men with limited disease. The prostate-specific membrane antigen (PSMA) is a membrane-bound glycoprotein that is highly restricted to the prostate. Previously, studies analyzing the expression of PSMA have found an up-regulation in correlation with prostate cancer, particularly in advanced cancer. This assocn. is ideal for an application as a prognostic marker. In the current study, we characterized PSMA expression in a high-risk cohort and evaluated its potential use as predictive marker of prostate-specific antigen (PSA) recurrence. PSMA expression was analyzed by immunohistochem. using tissue microarrays composed of tumor samples from 450 patients. Protein intensity was recorded using a semiautomated quant. microscope system (ACIS II; Clarient Chromavision Medical Systems, San Juan Capistrano, CA). PSMA expression levels differed significantly (P < .001) between benign prostatic tissue, localized prostate cancer, and lymph node metastases. Dividing the cohort into high- and low-PSMA expressing cancers based on the median area of pos. staining, we found that high PSMA levels were assocd. with significant increase of PSA recurrence (P = .004). This was independent of clin. parameters such as lymph node tumor burden (lymph node d., >20%; P < .001), extraprostatic extension (P = .017), seminal vesicle invasion (P < .001), and high Gleason score (8-10, P = .006). In a multivariate model, PSMA expression and metastases to pelvic lymph nodes were significantly assocd. with time to PSA recurrence (HR, 1.4; 95% confidence interval, 1.1-2.8, P = .017; and hazard ratio, 5; 95% confidence interval, 2.6-9.7, P < .001, resp.). In summary, PSMA is independently assocd. with PSA recurrence in a high-risk cohort and thus might provide insight into the addnl. use of adjuvant therapy. Validation on other cohorts is required.
- 7Sanna, V.; Pintus, G.; Roggio, A. M.; Punzoni, S.; Posadino, A. M.; Arca, A.; Marceddu, S.; Bandiera, P.; Uzzau, S.; Sechi, M. Targeted biocompatible nanoparticles for the delivery of (−)-epigallocatechin 3-gallate to prostate cancer cells J. Med. Chem. 2011, 54, 1321– 1332Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhslyiu7k%253D&md5=8a103de00d04093c1d00f9a8b24732faTargeted Biocompatible Nanoparticles for the Delivery of (-)-Epigallocatechin 3-Gallate to Prostate Cancer CellsSanna, Vanna; Pintus, Gianfranco; Roggio, Anna Maria; Punzoni, Stefania; Posadino, Anna Maria; Arca, Alessandro; Marceddu, Salvatore; Bandiera, Pasquale; Uzzau, Sergio; Sechi, MarioJournal of Medicinal Chemistry (2011), 54 (5), 1321-1332CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mol. targeted cancer therapy mediated by nanoparticles (NPs) is a promising strategy to overcome the lack of specificity of conventional chemotherapeutic agents. In this context, the prostate-specific membrane antigen (PSMA) has demonstrated a powerful potential for the management of prostate cancer (PCa). Cancer chemoprevention by phytochems. is emerging as a suitable approach for the treatment of early carcinogenic processes. Since (-)-epigallocatechin 3-gallate (EGCG) has shown potent chemopreventive efficacy for PCa, we designed and developed novel targeted NPs in order to selectively deliver EGCG to cancer cells. Herein, to explore the recent concept of "nanochemoprevention", we present a study on EGCG-loaded NPs consisting of biocompatible polymers, functionalized with small mols. targeting PSMA, that exhibited a selective in vitro efficacy against PSMA-expressing PCa cells. This approach could be beneficial for high risk patients and would fulfill a significant therapeutic need, thus opening new perspectives for novel and effective treatment for PCa.
- 8DiPippo, V. A.; Magargal, W. W.; Moorji, S. M.; Murga, J. D.; Olson, W. C. Antiandrogen modulation of prostate-specific membrane antigen (PSMA): dynamics and synergy with PSMA-targeted therapy ASCO Meet. Abstr. 2013, 31, e16007Google ScholarThere is no corresponding record for this reference.
- 9Petrylak, D. P.; Kantoff, P. W.; Mega, A. E.; Vogelzang, N. J.; Stephenson, J.; Fleming, M. T.; Stambler, N.; Petrini, M.; Blattman, S.; Israel, R. J. Prostate-specific membrane antigen antibody drug conjugate (PSMA ADC): a phase I trial in metastatic castration-resistant prostate cancer (mCRPC) previously treated with a taxane ASCO Meet. Abstr. 2013, 31, 5018Google ScholarThere is no corresponding record for this reference.
- 10Rotshteyn, Y.; Mercier, F.; Bruno, R.; Stambler, N.; Israel, R. J.; Wong, V. Correlation of PSMA ADC exposure with reduction in tumor growth rate determined using serial PSA measurements from a phase I clinical trial ASCO Meet. Abstr. 2013, 31, e16047Google ScholarThere is no corresponding record for this reference.
- 11Hrkach, J.; Von Hoff, D.; Mukkaram Ali, M.; Andrianova, E.; Auer, J.; Campbell, T.; De Witt, D.; Figa, M.; Figueiredo, M.; Horhota, A.; Low, S.; McDonnell, K.; Peeke, E.; Retnarajan, B.; Sabnis, A.; Schnipper, E.; Song, J. J.; Song, Y. H.; Summa, J.; Tompsett, D.; Troiano, G.; Van Geen Hoven, T.; Wright, J.; LoRusso, P.; Kantoff, P. W.; Bander, N. H.; Sweeney, C.; Farokhzad, O. C.; Langer, R.; Zale, S. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile Sci. Transl. Med. 2012, 4, 128ra39Google ScholarThere is no corresponding record for this reference.
- 12Hillier, S. M.; Maresca, K. P.; Femia, F. J.; Marquis, J. C.; Foss, C. A.; Nguyen, N.; Zimmerman, C. N.; Barrett, J. A.; Eckelman, W. C.; Pomper, M. G.; Joyal, J. L.; Babich, J. W. Preclinical evaluation of novel glutamate–urea–lysine analogues that target prostate-specific membrane antigen as molecular imaging pharmaceuticals for prostate cancer Cancer Res. 2009, 69, 6932– 6940Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVOmtLzE&md5=f222208c3c3fbcaa376306c49c9ec80bPreclinical Evaluation of Novel Glutamate-Urea-Lysine Analogues That Target Prostate-Specific Membrane Antigen as Molecular Imaging Pharmaceuticals for Prostate CancerHillier, Shawn M.; Maresca, Kevin P.; Femia, Frank J.; Marquis, John C.; Foss, Catherine A.; Nguyen, Nghi; Zimmerman, Craig N.; Barrett, John A.; Eckelman, William C.; Pomper, Martin G.; Joyal, John L.; Babich, John W.Cancer Research (2009), 69 (17), 6932-6940CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Prostate-specific membrane antigen (PSMA) is expressed in normal human prostate epithelium and is highly up-regulated in prostate cancer. We previously reported a series of novel small mol. inhibitors targeting PSMA. Two compds., MIP-1072, (S)-2-(3-((S)-1-carboxy-5-(4-iodobenzylamino)pentyl)ureido)pentanedioic acid, and MIP-1095, (S)-2-(3-((S)-1-carboxy-5-(3-(4-iodophenyl)ureido)pentyl)ureido)pentanedioic acid, were selected for further evaluation. MIP-1072 and MIP-1095 potently inhibited the glutamate carboxypeptidase activity of PSMA (Ki = 4.6 ± 1.6 nM and 0.24 ± 0.14 nM, resp.) and, when radiolabeled with 123I, exhibited high affinity for PSMA on human prostate cancer LNCaP cells (Kd = 3.8 ± 1.3 nM and 0.81 ± 0.39 nM, resp.). The assocn. of [123I]MIP-1072 and [123I]MIP-1095 with PSMA was specific; there was no binding to human prostate cancer PC3 cells, which lack PSMA, and binding was abolished by coincubation with a structurally unrelated NAALADase inhibitor, 2-(phosphonomethyl)pentanedioic acid (PMPA). [123I]MIP-1072 and [123I]MIP-1095 internalized into LNCaP cells at 37°C. Tissue distribution studies in mice showed 17.3 ± 6.3% (at 1 h) and 34.3 ± 12.7% (at 4 h) injected dose per g of LNCaP xenograft tissue, for [123I]MIP-1072 and [123I]MIP-1095, resp. [123I]MIP-1095 exhibited greater tumor uptake but slower washout from blood and nontarget tissues compared with [123I]MIP-1072. Specific binding to PSMA in vivo was shown by competition with PMPA in LNCaP xenografts, and the absence of uptake in PC3 xenografts. The uptake of [123I]MIP-1072 and [123I]MIP-1095 in tumor-bearing mice was corroborated by single-photon emission computed tomog./computed tomog. (SPECT/CT) imaging. PSMA-specific radiopharmaceuticals should provide a novel mol. targeting option for the detection and staging of prostate cancer.
- 13Mease, R. C.; Foss, C. A.; Pomper, M. G. PET imaging in prostate cancer: focus on prostate-specific membrane antigen Curr. Top. Med. Chem. 2013, 13, 951– 962Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXpslKhtb0%253D&md5=e49d20dfdfbb41921517c501ca5182e2PET imaging in prostate cancer: focus on prostate-specific membrane antigenMease, Ronnie C.; Foss, Catherine A.; Pomper, Martin G.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2013), 13 (8), 951-962CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)A review. Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. Positron emission tomog./computed tomog. (PET/CT) with emerging radiopharmaceuticals promises accurate staging of primary disease, restaging of recurrent disease, detection of metastatic lesions and, ultimately, for predicting the aggressiveness of disease. Prostate-specific membrane antigen (PSMA) is a well-characterized imaging biomarker of PCa. Because PSMA levels are directly related to androgen independence, metastasis and progression, PSMA could prove an important target for the development of new radiopharmaceuticals for PET. Preclin. data for new PSMA-based radiotracers are discussed and include new 89Zr- and 64Cu-labeled anti-PSMA antibodies and antibody fragments, 64Cu-labeled aptamers, and 11C-, 18F-, 68Ga-, 64Cu-, and 86Y-labeled low mol. wt. inhibitors of PSMA. Several of these agents, namely 68Ga- HBED-CC conjugate 15, 18F-DCFBC 8, and BAY1075553 are particularly promising, each having detected sites of PCa in initial clin. studies. These early clin. results suggest that PET/CT using PSMA-targeted agents, esp. with compds. of low mol. wt., will make valuable contributions to the management of PCa.
- 14Cho, S. Y.; Gage, K. L.; Mease, R. C.; Senthamizhchelvan, S.; Holt, D. P.; Jeffrey-Kwanisai, A.; Endres, C. J.; Dannals, R. F.; Sgouros, G.; Lodge, M.; Eisenberger, M. A.; Rodriguez, R.; Carducci, M. A.; Rojas, C.; Slusher, B. S.; Kozikowski, A. P.; Pomper, M. G. Biodistribution, tumor detection, and radiation dosimetry of 18F–DCFBC, a low-molecular-weight inhibitor of prostate-specific membrane antigen, in patients with metastatic prostate cancer J. Nucl. Med. 2012, 53, 1883– 1891Google ScholarThere is no corresponding record for this reference.
- 15Afshar-Oromieh, A.; Malcher, A.; Eder, M.; Eisenhut, M.; Linhart, H. G.; Hadaschik, B. A.; Holland-Letz, T.; Giesel, F. L.; Kratochwil, C.; Haufe, S.; Haberkorn, U.; Zechmann, C. M. PET imaging with a [(68)Ga]gallium-labelled PSMA ligand for the diagnosis of prostate cancer: biodistribution in humans and first evaluation of tumour lesions Eur. J. Nucl. Med. Mol. Imaging 2012, 40, 486– 495Google ScholarThere is no corresponding record for this reference.
- 16Barrett, J. A.; Coleman, R. E.; Goldsmith, S. J.; Vallabhajosula, S.; Petry, N. A.; Cho, S.; Armor, T.; Stubbs, J. B.; Maresca, K. P.; Stabin, M. G.; Joyal, J. L.; Eckelman, W. C.; Babich, J. W. First-in-man evaluation of 2 high-affinity PSMA-avid small molecules for imaging prostate cancer J. Nucl. Med. 2013, 54, 380– 387Google ScholarThere is no corresponding record for this reference.
- 17Maresca, K. P.; Hillier, S. M.; Lu, G.; Marquis, J. C.; Zimmerman, C. N.; Eckelman, W. C.; Joyal, J. L.; Babich, J. W. Small molecule inhibitors of PSMA incorporating technetium-99m for imaging prostate cancer: effects of chelate design on pharmacokinetics Inorg. Chim. Acta 2012, 389, 168– 172Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XnslKmt74%253D&md5=2165975aa8528ada852cecc63bb0bd0cSmall molecule inhibitors of PSMA incorporating technetium-99m for imaging prostate cancer: Effects of chelate design on pharmacokineticsMaresca, Kevin P.; Hillier, Shawn M.; Lu, Genliang; Marquis, John C.; Zimmerman, Craig N.; Eckelman, William C.; Joyal, John L.; Babich, John W.Inorganica Chimica Acta (2012), 389 (), 168-175CODEN: ICHAA3; ISSN:0020-1693. (Elsevier B.V.)Single amino acid chelate (SAAC) systems for the complexation of the M(CO)3 moiety (M = Tc/Re) have been successfully incorporated into novel synthetic strategies for radiopharmaceuticals and evaluated in a variety of biol. applications. However, the lipophilicity of the first generation of 99mTc(CO)3 complexes has resulted in substantial hepatobiliary uptake when examd. either as lysine derivs. or integrated into biol. active small mols. and peptides. Here, we designed, synthesized, and evaluated novel polar functionalized imidazole derived SAAC systems (SAAC II) which have been chem. modified to promote overall 99mTc(CO)3L3 complex hydrophilicity with the intent of reducing non-target effects and enhancing renal clearance of prostate specific membrane antigen (PSMA) targeting small mols. The 99mTc-labeled compds. were prepd., purified, and evaluated for stability, lipophilicity, and tissue distribution in LNCaP xenograft mice. The Glu-urea-Lys-C11 analogs were prepd. with a variety of chelators to form (19R,23S)-1-(X)-2-((Y)methyl)-13,21-dioxo-2,14,20,22-tetraazapentacosane-19,23,25-tricarboxylic acid where X = Y = (methyl)pyridin-2-yl (6), X = Y = (methyl)-1H-imidazol-2-yl (7), X = (methyl)pyridin-2-yl, Y = carboxymethyl (8), X = Y = 1-(carboxymethyl)-1H-imidazol-2-yl (9), X = 1-(carboxymethyl)-1H-imidazol-2-yl, Y = carboxymethyl (10), and X = Y = 1-(1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl)-2-((1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl) (11). 99mTc labeling was achieved at ligand concns. as low as 10-6 M and the complexes were stable (>90%) for 24 h. These new SAAC II chelators were evaluated for their influence on binding of the Glu-urea-Lys-C11 analogs to PSMA-pos. LNCaP cells and compared to pyridine- and N-methylimidazole-contg. SAAC ligands. Tissue distribution of the 99mTc-complexes contg. the more polar chelators, 9 and 11, demonstrated decreased liver (<2% ID/g) and increased LNCaP tumor (>11% ID/g) accumulation at 1 h post-injection.
- 18Nedrow-Byers, J. R.; Moore, A. L.; Ganguly, T.; Hopkins, M. R.; Fulton, M. D.; Benny, P. D.; Berkman, C. E. PSMA-targeted SPECT agents: Mode of binding effect on in vitro performance Prostate 2013, 73, 355– 362Google ScholarThere is no corresponding record for this reference.
- 19Lu, G.; Maresca, K. P.; Hillier, S. M.; Zimmerman, C. N.; Eckelman, W. C.; Joyal, J. L.; Babich, J. W. Synthesis and SAR of 99mTc/Re-Labeled Small Molecule Prostate Specific Membrane Antigen Inhibitors with Novel Polar Chelates Biorg. Med. Chem. Lett. 2013, 23, 1557– 1563Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptlKjtQ%253D%253D&md5=0dbceebf79f273aafb5db22b912302f6Synthesis and SAR of 99mTc/Re-labeled small molecule prostate specific membrane antigen inhibitors with novel polar chelatesLu, Genliang; Maresca, Kevin P.; Hillier, Shawn M.; Zimmerman, Craig N.; Eckelman, William C.; Joyal, John L.; Babich, John W.Bioorganic & Medicinal Chemistry Letters (2013), 23 (5), 1557-1563CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Prostate specific membrane antigen (PSMA) is recognized as an attractive mol. target for the development of radiopharmaceuticals to image and potentially treat metastatic prostate cancer. A series of novel 99mTc/Re-tricarbonyl radiolabeled PSMA inhibitors were therefore synthesized by the attachment of glutamate-urea-lysine (Glu-urea-Lys) and glutamate-urea-glutamate (Glu-urea-Glu) pharmacophore to single amino acid chelate (SAAC) where the SAAC ligand was either bis(pyridin-2-ylmethyl)amino (DPA), bis((1-methyl-1H-imidazol-2-yl)methyl)amino (NMI), bis((1-(carboxymethyl)-1H-imidazol-2-yl)methyl)amino (CIM) or bis((1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl)methyl)amino (TIM). The in vitro binding affinity of the rhenium complexes was evaluated using PSMA-expressing human prostate cancer LNCaP cells. IC50 values ranged from 3.8 ± 2 to >2000 nM. A linker between the SAAC chelate and pharmacophore was required for high affinity binding. However, extending the length of the linker did not substantially improve binding. PSMA binding was also influenced by the nature of the SAAC chelate. One of the most potent compds., 23b (IC50 = 4.8 ± 2.7 nM), was radiolabeled with technetium tricarbonyl ({99mTc(CO)3}+) to afford the {99mTc(CO)3}+ complex in excellent yield and high purity. This effort led to the identification of a diverse series of promising high affinity {99mTc(CO)3}+ radiolabeled PSMA inhibitors.
- 20Nedrow-Byers, J. R.; Jabbes, M.; Jewett, C.; Ganguly, T.; He, H.; Liu, T.; Benny, P.; Bryan, J. N.; Berkman, C. E. A phosphoramidate-based prostate-specific membrane antigen-targeted SPECT agent Prostate 2012, 72, 904– 912Google ScholarThere is no corresponding record for this reference.
- 21Banerjee, S. R.; Foss, C. A.; Castanares, M.; Mease, R. C.; Byun, Y.; Fox, J. J.; Hilton, J.; Lupold, S. E.; Kozikowski, A. P.; Pomper, M. G. Synthesis and evaluation of technetium-99m- and rhenium-labeled inhibitors of the prostate-specific membrane antigen (PSMA) J. Med. Chem. 2008, 51, 4504– 4517Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXoslamsr4%253D&md5=e070e63a175f5d733d58021a6bfdf87dSynthesis and Evaluation of Technetium-99m- and Rhenium-Labeled Inhibitors of the Prostate-Specific Membrane Antigen (PSMA)Banerjee, Sangeeta R.; Foss, Catherine A.; Castanares, Mark; Mease, Ronnie C.; Byun, Youngjoo; Fox, James J.; Hilton, John; Lupold, Shawn E.; Kozikowski, Alan P.; Pomper, Martin G.Journal of Medicinal Chemistry (2008), 51 (15), 4504-4517CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The prostate-specific membrane antigen (PSMA) is increasingly recognized as a viable target for imaging and therapy of cancer. We prepd. seven 99mTc/Re-labeled compds. by attaching known Tc/Re chelating agents to an amino-functionalized PSMA inhibitor (lys-NHCONH-glu) with or without a variable length linker moiety. Ki values ranged from 0.17 to 199 nM. Ex vivo biodistribution and in vivo imaging demonstrated the degree of specific binding to engineered PSMA+ PC3 PIP tumors. PC3-PIP cells are derived from PC3 that have been transduced with the gene for PSMA. Despite demonstrating nearly the lowest PSMA inhibitory potency of this series, [99mTc(CO)3(L1)]+ (L1 = (2-pyridylmethyl)2N(CH2)4CH(CO2H)NHCO-(CH2)6CO-NH-lys-NHCONH-glu) showed the highest, most selective PIP tumor uptake, at 7.9 ± 4.0% injected dose per g of tissue at 30 min postinjection. Radioactivity cleared from nontarget tissues to produce a PIP to flu (PSMA-PC3) ratio of 44:1 at 120 min postinjection. PSMA can accommodate the steric requirements of 99mTc/Re complexes within PSMA inhibitors, the best results achieved with a linker moiety between the .vepsiln. amine of the urea lysine and the chelator.
- 22Kularatne, S. A.; Zhou, Z.; Yang, J.; Post, C. B.; Low, P. S. Design, synthesis, and preclinical evaluation of prostate-specific membrane antigen targeted (99m)Tc-radioimaging agents Mol. Pharmaceutics 2009, 6, 790– 800Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlsF2hs70%253D&md5=77c5b6f3776e761979de857d4299106eDesign, Synthesis, and Preclinical Evaluation of Prostate-Specific Membrane Antigen Targeted 99mTc-Radioimaging AgentsKularatne, Sumith A.; Zhou, Zhigang; Yang, Jun; Post, Carol B.; Low, Philip S.Molecular Pharmaceutics (2009), 6 (3), 790-800CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The high mortality and financial burden assocd. with prostate cancer can be partly attributed to a lack of sensitive screening methods for detection and staging of the disease. Guided by in silico docking studies using the crystal structure of PSMA, we designed and synthesized a series of PSMA-targeted 99mTc-chelate complexes for imaging PSMA-expressing human prostate cancer cells (LNCaP cell line). Of the six targeted radioimaging agents synthesized, three were found to bind LNCaP cells with low nanomolar affinity. Moreover, the same three PSMA-targeted imaging agents were shown to localize primarily to LNCaP tumor xenografts in nu/nu mice, with an av. of 9.8 ± 2.4% injected dose/g tissue accumulating in the tumor and only 0.11% injected dose/g tissue retained in the muscle at 4 h postinjection. Collectively, these high affinity, PSMA-specific radioimaging agents demonstrate significant potential for use in localizing prostate cancer masses, monitoring response to therapy, detecting prostate cancer recurrence following surgery, and selecting patients for subsequent PSMA-targeted chemotherapy.
- 23Zhang, Y.; DiFilipp, F.; Doke, A.; Huang, J.; Heston, W.; Huang, S. Preliminary micro-SPECT and biodistribution study of a novel Tc99m-labeled PSMA tracer derived from RBI1033 J. Nucl. Med. Meet. Abstr. 2012, 53 (Suppl 1) 1661Google ScholarThere is no corresponding record for this reference.
- 24Schafer, M.; Bauder-Wust, U.; Leotta, K.; Zoller, F.; Mier, W.; Haberkorn, U.; Eisenhut, M.; Eder, M. A dimerized urea-based inhibitor of the prostate-specific membrane antigen for 68Ga-PET imaging of prostate cancer EJNMMI Res. 2012, 2, 23Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38npsFSmuw%253D%253D&md5=186d67c326bb60367462b16b0deb419fA dimerized urea-based inhibitor of the prostate-specific membrane antigen for 68Ga-PET imaging of prostate cancerSchafer Martin; Bauder-Wust Ulrike; Leotta Karin; Zoller Frederic; Mier Walter; Haberkorn Uwe; Eisenhut Michael; Eder MatthiasEJNMMI research (2012), 2 (1), 23 ISSN:.UNLABELLED: BACKGROUND: Alternative positron-emission tomography (PET) probes like labeled inhibitors of the prostate-specific membrane antigen (PSMA) are of emerging clinical impact as they show the ability to image small lesions of recurrent prostate cancer. Here, the dimerization of the pharmacophore Glu-ureido-Lys via the 68Ga chelator N,N'-bis[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid (HBED-CC) was investigated to further improve the binding characteristics and pharmacokinetics. METHODS: The peptidomimetic structures were synthesized by solid-phase chemistry, and the resulting products were coupled with the respective 2,3,5,6-tetrafluorophenol esters of HBED-CC to form the monomeric reference and the dimeric Glu-ureido-Lys derivative. The binding properties were analyzed in competitive binding, internalization, and cell surface retention experiments. PET images and biodistribution data were obtained 1 h after injection in BALB/c nu/nu mice bearing LNCaP tumor xenografts. RESULTS: Cell binding data revealed significant better binding properties of the dimer (IC50 = 3.9 ± 1.8 nM; IC50 (monomer) = 12.1 ± 2.1 nM). The inhibition potency investigated by the enzyme-based NAALADase assay confirmed these results. Specific internalization in LNCaP cells was demonstrated for both, the monomer and dimer. As shown by efflux measurements, the dimeric compound was more effectively retained on the cell surface, resulting in advanced in vivo properties (T/BMonomer = 9.2; T/BDimer = 26.5). CONCLUSIONS: The dimeric [68Ga]7 is a promising imaging agent for PSMA-expressing tumors as it shows higher tumor uptake while observing more favorable background clearance. As compared to the respective monomer, the higher affinity and prolonged tumor retention additionally represent promising features and warrant further evaluation regarding 68Ga-PET imaging of PSMA expression.
- 25Eder, M.; Schafer, M.; Bauder-Wust, U.; Hull, W. E.; Wangler, C.; Mier, W.; Haberkorn, U.; Eisenhut, M. 68Ga-complex lipophilicity and the targeting property of a urea-based PSMA inhibitor for PET imaging Bioconjugate Chem. 2012, 23, 688– 697Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XivVCmtLo%253D&md5=4bb4cbba9f510ed2561b87a44bafa00a68Ga-Complex Lipophilicity and the Targeting Property of a Urea-Based PSMA Inhibitor for PET ImagingEder, Matthias; Schaefer, Martin; Bauder-Wuest, Ulrike; Hull, William-Edmund; Waengler, Carmen; Mier, Walter; Haberkorn, Uwe; Eisenhut, MichaelBioconjugate Chemistry (2012), 23 (4), 688-697CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Urea-based inhibitors of the prostate-specific membrane antigen (PSMA) represent low-mol.-wt. pepidomimetics showing the ability to image PSMA-expressing prostate tumors. The highly efficient, acyclic Ga(III) chelator N,N'-bis [2-hydroxy-5-(carboxyethyl)benzyl] ethylenediamine-N,N'- diacetic acid (HBED-CC) was introduced as a lipophilic side chain into the hydrophilic pharmacophore Glu-NH-CO-NH-Lys which was found favorable to interact with the PSMA "active binding site". This report describes the syntheses, in vitro binding analyses, and biodistribution data of the radiogallium labeled PSMA inhibitor Glu-NH-CO-NH-Lys(Ahx)-HBED-CC in comparison to the corresponding DOTA conjugate. The binding properties were analyzed using competitive cell binding and enzyme-based assays followed by internalization expts. Compared to the DOTA-conjugate, the HBED-CC deriv. showed reduced unspecific binding and considerable higher specific internalization in LNCaP cells. The 68Ga complex of the HBED-CC ligand exhibited higher specificity for PSMA expressing tumor cells resulting in improved in vivo properties. 68Ga labeled Glu-NH-CO-NH-Lys(Ahx)-HBED-CC showed fast blood and organ clearances, low liver accumulation, and high specific uptake in PSMA expressing organs and tumor. It could be demonstrated that the PET-imaging property of a urea-based PSMA inhibitor could significantly be improved with HBED-CC.
- 26Banerjee, S. R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Green, G.; Fox, J. J.; Horti, A.; Mease, R. C.; Pomper, M. G. 68Ga-labeled inhibitors of prostate-specific membrane antigen (PSMA) for imaging prostate cancer J. Med. Chem. 2010, 53, 5333– 5341Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnvVKqs7c%253D&md5=cbe8ccdc2d1833374546e68091632d9068Ga-Labeled Inhibitors of Prostate-Specific Membrane Antigen (PSMA) for Imaging Prostate CancerBanerjee, Sangeeta Ray; Pullambhatla, Mrudula; Byun, Youngjoo; Nimmagadda, Sridhar; Green, Gilbert; Fox, James J.; Horti, Andrew; Mease, Ronnie C.; Pomper, Martin G.Journal of Medicinal Chemistry (2010), 53 (14), 5333-5341CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Gallium-68 is a generator-produced radionuclide for positron emission tomog. (PET) that is being increasingly used for radiolabeling of tumor-targeting peptides. Compds. [68Ga]3 and [68Ga]6 are high-affinity urea-based inhibitors of the prostate-specific membrane antigen (PSMA) that were synthesized in decay-uncorrected yields ranging from 60% to 70% and radiochem. purities of more than 99%. Compd. [68Ga]3 demonstrated 3.78 ± 0.90% injected dose per g of tissue (%ID/g) within PSMA+ PIP tumor at 30 min postinjection, while [68Ga]6 showed a 2 h PSMA+ PIP tumor uptake value of 3.29 ± 0.77 %ID/g. Target (PSMA+ PIP) to nontarget (PSMA- flu) ratios were 4.6 and 18.3, resp., at those time points. Both compds. delineated tumor clearly by small animal PET. The urea series of imaging agents for PSMA can be radiolabeled with 68Ga, a cyclotron-free isotope useful for clin. PET studies, with maintenance of target specificity.
- 27Banerjee, S. R.; Pullambhatla, M.; Shallal, H.; Lisok, A.; Mease, R. C.; Pomper, M. G. A Modular Strategy to Prepare Multivalent Inhibitors of Prostate-Specific Membrane Antigen (PSMA) Oncotarget 2011, 2, 1244– 1253Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC387ntVehsQ%253D%253D&md5=ff90dd0090ea69db9c798ba6ebde318bA modular strategy to prepare multivalent inhibitors of prostate-specific membrane antigen (PSMA)Banerjee Sangeeta Ray; Pullambhatla Mrudula; Shallal Hassan; Lisok Ala; Mease Ronnie C; Pomper Martin GOncotarget (2011), 2 (12), 1244-53 ISSN:.We have developed a modular scaffold for preparing high-affinity, homo-multivalent inhibitors of the prostate-specific membrane antigen (PSMA) for imaging and therapy of prostate cancer (PCa). Our system contains a lysine-based (μ-, e-) dialkyne residue for incorporating a PSMA binding Lys-Glu urea motif exploiting click chemistry and a second lysine residue for subsequent modification with an imaging or therapeutic moiety. The utility of the multivalent scaffold was examined by synthesizing bivalent compounds 2 and 3 and comparing them with the monovalent analog 1. Determination of inhibition constants (Ki) revealed that bivalent 2 (0.2 nM) and 3 (0.08 nM) are significantly more potent (~ 5 fold and ~ 11 fold, respectively) inhibitors of PSMA than monovalent 1 (0.9 nM). A single photon emission computed tomography (SPECT)-CT imaging study of [111In]3 demonstrated high and specific uptake in PSMA+ PC-3 PIP tumor until at least 48 h post-injection, with rapid clearance from non-target tissues, including kidney. A biodistribution study revealed that [111In]3 demonstrated 34.0 ± 7.5 percent injected dose per gram of tissue in PSMA+ tumor at 24 h post-injection and was capable of generating target-to-non-target ratios of ~ 379 in PSMA+ PC-3 PIP tumors vs. isogenic PSMA-negative PC3-flu tumors in vivo. The click chemistry approach affords a convenient strategy toward multivalent PSMA inhibitors of enhanced affinity and superior pharmacokinetics for imaging.
- 28Banerjee, S. R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Foss, C. A.; Green, G.; Fox, J. J.; Lupold, S. E.; Mease, R. C.; Pomper, M. G. Sequential SPECT and optical imaging of experimental models of prostate cancer with a dual modality inhibitor of the prostate-specific membrane antigen Angew. Chem. 2011, 50, 9167– 9170Google ScholarThere is no corresponding record for this reference.
- 29Chen, Z.; Penet, M. F.; Nimmagadda, S.; Li, C.; Banerjee, S. R.; Winnard, P. T., Jr.; Artemov, D.; Glunde, K.; Pomper, M. G.; Bhujwalla, Z. M. PSMA-targeted theranostic nanoplex for prostate cancer therapy ACS Nano 2012, 6, 7752– 7762Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFGiu7jF&md5=5c9091dff6c26a165b597e4af3dd5032PSMA-Targeted Theranostic Nanoplex for Prostate Cancer TherapyChen, Zhihang; Penet, Marie-France; Nimmagadda, Sridhar; Li, Cong; Banerjee, Sangeeta R.; Winnard, Paul T.; Artemov, Dmitri; Glunde, Kristine; Pomper, Martin G.; Bhujwalla, Zaver M.ACS Nano (2012), 6 (9), 7752-7762CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Theranostic imaging, where diagnosis is combined with therapy, is particularly suitable for a disease that is as complex as cancer, esp. now that genomic and proteomic profiling can provide an extensive "fingerprint" of each tumor. With such information, theranostic agents can be designed to personalize treatment and minimize damage to normal tissue. Here we have developed a nanoplex platform for theranostic imaging of prostate cancer (PCa). In these proof-of-principle studies, a therapeutic nanoplex contg. multimodal imaging reporters was targeted to prostate-specific membrane antigen (PSMA), which is expressed on the cell surface of castrate-resistant PCa. The nanoplex was designed to deliver small interfering RNA (siRNA) along with a prodrug enzyme to PSMA-expressing tumors. Each component of the nanoplex was carefully selected to evaluate its diagnostic aspect of PSMA imaging and its therapeutic aspects of siRNA-mediated down-regulation of a target gene and the conversion of a prodrug to cytotoxic drug, using noninvasive multimodality imaging. Studies performed using two variants of human PC3-PCa cells and tumors, one with high PSMA expression level and another with negligible expression levels, demonstrated PSMA-specific uptake. In addn., down-regulation of the selected siRNA target, choline kinase (Chk), and the conversion of the nontoxic prodrug 5-fluorocytosine (5-FC) to cytotoxic 5-fluorouracil (5-FU) were also demonstrated with noninvasive imaging. The nanoplex was well-tolerated and did not induce liver or kidney toxicity or a significant immune response. The nanoplex platform described can be easily modified and applied to different cancers, receptors, and pathways to achieve theranostic imaging, as a single agent or in combination with other treatment modalities.
- 30Hao, G.; Kumar, A.; Dobin, T.; Oz, O. K.; Hsieh, J. T.; Sun, X. A multivalent approach of imaging probe design to overcome an endogenous anion binding competition for noninvasive assessment of prostate specific membrane antigen Mol. Pharmacol. 2013, 10, 2975– 2985Google ScholarThere is no corresponding record for this reference.
- 31Banerjee, S. R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Baidoo, K. E.; Brechbiel, M. W.; Mease, R. C.; Pomper, M. G. Preclinical Evaluation of 86Y-Labeled Inhibitors of Prostate Specific Membrane Antigen. J. Labelled Compd. Radiopharm. 2011, 54, Suppl S1, p S65.Google ScholarThere is no corresponding record for this reference.
- 32Holland, J. P.; Divilov, V.; Bander, N. H.; Smith-Jones, P. M.; Larson, S. M.; Lewis, J. S. 89Zr–DFO–J591 for immunoPET of prostate-specific membrane antigen expression in vivo J. Nucl. Med. 2010, 51, 1293– 1300Google ScholarThere is no corresponding record for this reference.
- 33Viola-Villegas, N.; Evans, H.; Bartlett, D.; Wu, A.; Lewis, J. Preclinical development of Zr-89 labeled anti-PSMA minibody and cys-diabody J Nucl. Med. Meet. Abstr. 2012, 53, 347Google ScholarThere is no corresponding record for this reference.
- 34Ray Banerjee, S.; Pullambhatla, M.; Foss, C. A.; Falk, A.; Byun, Y.; Nimmagadda, S.; Mease, R. C.; Pomper, M. G. Effect of chelators on the pharmacokinetics of (99m)Tc-labeled imaging agents for the prostate-specific membrane antigen (PSMA) J. Med. Chem. 2013, 56, 6108– 6121Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVWhtLjE&md5=3007d66e9dc898d5beb4039cd02ddae3Effect of Chelators on the Pharmacokinetics of 99mTc-Labeled Imaging Agents for the Prostate-Specific Membrane Antigen (PSMA)Ray Banerjee, Sangeeta; Pullambhatla, Mrudula; Foss, Catherine A.; Falk, Alexander; Byun, Youngjoo; Nimmagadda, Sridhar; Mease, Ronnie C.; Pomper, Martin G.Journal of Medicinal Chemistry (2013), 56 (15), 6108-6121CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Technetium-99m, the most commonly used radionuclide in nuclear medicine, can be attached to biol. important mols. through a variety of chelating agents, the choice of which depends upon the imaging application. The prostate-specific membrane antigen (PSMA) is increasingly recognized as an important target for imaging and therapy of prostate cancer (PCa). Three different 99mTc-labeling methods were employed to investigate the effect of the chelator on the biodistribution and PCa tumor uptake profiles of 12 new urea-based PSMA-targeted radiotracers. This series includes hydrophilic ligands for radiolabeling with the [99mTc(CO)3]+ core (L8-L10), traditional NxSy-based chelating agents with varying charge and polarity for the 99mTc-oxo core (L11-L18), and a 99mTc-organohydrazine-labeled radioligand (L19). 99mTc(I)-Tricarbonyl-labeled [99mTc]L8 produced the highest PSMA+ PC3 PIP to PSMA- PC3 flu tumor ratios and demonstrated the lowest retention in normal tissues including kidney after 2 h. These results suggest that choice of chelator is an important pharmacokinetic consideration in the development of 99mTc-labeled radiopharmaceuticals targeting PSMA.
- 35Wadas, T. J.; Wong, E. H.; Weisman, G. R.; Anderson, C. J. Copper chelation chemistry and its role in copper radiopharmaceuticals Curr. Pharm. Des. 2007, 13, 3– 16Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1Wjsrs%253D&md5=f79c91f1853f0188a9f129acc7902dc9Copper chelation chemistry and its role in copper radiopharmaceuticalsWadas, T. J.; Wong, E. H.; Weisman, G. R.; Anderson, C. J.Current Pharmaceutical Design (2007), 13 (1), 3-16CODEN: CPDEFP; ISSN:1381-6128. (Bentham Science Publishers Ltd.)A review. Mol. imaging is an important scientific discipline that plays a major role in clin. medicine and pharmaceutical development. While several imaging modalities including x-ray computed tomog. and magnetic resonance imaging generate high-resoln. anatomical images, positron emission tomog. (PET) and single photon emission computed tomog. offer insight into the physiol. processes that occur within a living organism. Of these two nuclear medicine imaging techniques, PET has advantages with respect to sensitivity and resoln., and this has led to the prodn. and development of many positron emitting radionuclides that include non-traditional radionuclides of the transition metals. Copper-64 (t1/2 = 12.7 h, β+: 17.4%, Eβ+max = 656 keV; β-: 39%, Eβ-max = 573 keV) has emerged as an important positron emitting radionuclide that has the potential for use in diagnostic imaging and radiotherapy. However, 64Cu must be delivered to the living system as a stable complex that is attached to a biol. targeting mol. for effective imaging and therapy. Therefore, significant research has been devoted to the development of ligands that can stably chelate 64Cu. This review discusses the necessary characteristics of an effective 64Cu chelator, while highlighting the development and evaluation of 64Cu-complexes attached to biol.-targeted ligands.
- 36Connett, J. M.; Anderson, C. J.; Guo, L. W.; Schwarz, S. W.; Zinn, K. R.; Rogers, B. E.; Siegel, B. A.; Philpott, G. W.; Welch, M. J. Radioimmunotherapy with a 64Cu-labeled monoclonal antibody: a comparison with 67Cu Proc. Natl. Acad. Sci. U. S. A. 1996, 93, 6814– 6818Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XjvFeit78%253D&md5=44ebb765573e81cab4eb53cc35c1249dRadioimmunotherapy with a 64Cu-labeled monoclonal antibody: a comparison with 67CuConnett, Judith M.; Anderson, Carolyn J.; Guo, Li-Wu; Schwarz, Sally W.; Zinn, Kurt R.; Rogers, Buck E.; Siegel, Barry A.; Philpott, Gordon; Welch, Michael J.Proceedings of the National Academy of Sciences of the United States of America (1996), 93 (13), 6814-6818CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)67Cu (t1/2 = 62 h) has demonstrated potential as a radionuclide for radioimmunotherapy, but limited availability severely restricts its widespread use. 64Cu (t1/2 = 12.8 h) has been shown to have comparable effectiveness in vitro and in vivo. The present study was undertaken to examine the therapeutic potential of 64Cu- and 67Cu-bromoacetamidobenzyl-1,4,8,11-tetraazacyclotetradecane-N,N',N'',N'''-tetraacetic acid (BAT)-2-iminothiolane (2IT)-1A3 (1A3 is a mouse anti-human colorectal cancer mAb) for treatment of GW39 human colon carcinoma carried in hamster thighs. Hamsters were injected with 64Cu- or 67C-BAT-2IT-1A3 or Cu-labeled nonspecific IgG (MOPC) or saline. Hamsters were killed 6-7 mo after therapy or when tumors were ≥10 g. Of the hamsters with small tumors (mean wt. 0.43 ± 0.25 g), 87.5% were disease-free 7 mo after treatment with 2 mCi (1 Ci = 37 GBq) of 64Cu-BAT-2IT-1A3 or 0.4 mCi of 67Cu-BAT-2IT-1A3. The mean tumor doses at these activities of 64Cu- and 67Cu-BAT-2IT-1A3 were 586 and 1269 rad (1 rad = 0.01 Gy), resp. In contrast, 76% of hamsters treated with 2 mCi of 64Cu-BAT-2IT-MOPC or 0.4 mCi of 67Cu-BAT-2IT-MOPC had to be killed before 6 mo because of tumor regrowth. When hamsters with larger tumors (mean wt. 0.66 ± 0.11 g) were treated with 64Cu- or 67Cu-BAT-2IT-1A3, survival was extended compared with controls, but only one animal remained tumor-free to 6 mo. These results demonstrate that 64Cu- and 67Cu-BAT-2IT-1A3 given in a single administered dose can eradicate small tumors without significant host toxicity, but addnl. strategies to deliver higher tumor doses will be needed for larger tumors.
- 37Blower, P. J.; Lewis, J. S.; Zweit, J. Copper radionuclides and radiopharmaceuticals in nuclear medicine Nucl. Med. Biol. 1996, 23, 957– 980Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXkslOqsQ%253D%253D&md5=39b9b14d56995b81d7c57f35e2fcdd82Copper radionuclides and radiopharmaceuticals in nuclear medicineBlower, Philip J.; Lewis, Jason S.; Zweit, JamalNuclear Medicine and Biology (1996), 23 (8), 957-980CODEN: NMBIEO; ISSN:0883-2897. (Elsevier)The chem., radiochem., radiobiol., and radiopharmacol. of radiopharmaceuticals contg. copper radionuclides are reviewed, with 210 refs. Copper radionuclides offer application in positron emission tomog., targeted radiotherapy, and single photon imaging. The chem. of copper is relatively simple and well-suited to radiopharmaceutical application. Current radiopharmaceuticals include biomols. labeled via bifunctional chelators primarily based on cyclic polyaminocarboxylates and polyamines, and pyruvaldehyde-bis(N4-methylthiosemicarbazone) (PTSM) and its analogs. The chem. of copper, of which only a fraction has yet been exploited, is likely to be applied more fully in the future.
- 38Guo, Y.; Parry, J. J.; Laforest, R.; Rogers, B. E.; Anderson, C. J. The role of p53 in combination radioimmunotherapy with 64Cu–DOTA–cetuximab and cisplatin in a mouse model of colorectal cancer J. Nucl. Med. 2013, 54, 1621– 1629Google ScholarThere is no corresponding record for this reference.
- 39Donnelly, P. S. The role of coordination chemistry in the development of copper and rhenium radiopharmaceuticals Dalton Trans. 2011, 40, 999– 1010Google ScholarThere is no corresponding record for this reference.
- 40Smith, S. V. Molecular imaging with copper-64 J. Inorg. Biochem. 2004, 98, 1874– 1901Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptlOktL0%253D&md5=b0972b1167a9b710b5c409d772941d95Molecular imaging with copper-64Smith, Suzanne V.Journal of Inorganic Biochemistry (2004), 98 (11), 1874-1901CODEN: JIBIDJ; ISSN:0162-0134. (Elsevier B.V.)A review. Mol. imaging is expected to change the face of drug discovery and development. The ability to link imaging to biol. for guiding therapy should improve the rate at which novel imaging technologies, probes, contrast agents, drugs and drug delivery systems can be transferred into clin. practice. Nuclear medicine imaging, in particular, positron emission tomog. (PET) allows the detection and monitoring of a variety of biol. and pathophysiol. processes, at tracer quantities of the radiolabeled target agents, and at doses free from pharmacol. effects. In the field of drug discovery and development, the use of radiotracers for radiolabelling target agents has now become one of the essential tools in identifying, screening and development of new target agents. In this regard, 64Cu (t1/2 = 12.7 h) has been identified as an emerging PET isotope. Its half-life is sufficiently long for radiolabelling a range of target agents and its ease of prodn. and adaptable chem. make it an excellent radioisotope for use in mol. imaging. This review describes recent advances, in the routes of 64Cu prodn., design and application of bi-functional ligands for use in radiolabelling with 64/67Cu2+, and their significance and anticipated impact on the field of mol. imaging and drug development.
- 41Wadas, T. J.; Wong, E. H.; Weisman, G. R.; Anderson, C. J. Molecular imaging of cancer with copper-64 radiopharmaceuticals and positron emission tomography (PET) Chem. Rev. 2010, 110, 2858– 2902Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXltFChtro%253D&md5=674cf315a0df26fc4407445bd35595d6Coordinating Radiometals of Copper, Gallium, Indium, Yttrium, and Zirconium for PET and SPECT Imaging of DiseaseWadas, Thaddeus J.; Wong, Edward H.; Weisman, Gary R.; Anderson, Carolyn J.Chemical Reviews (Washington, DC, United States) (2010), 110 (5), 2858-2902CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review.
- 42Shokeen, M.; Anderson, C. J. Coordinating radiometals of copper, gallium, indium, yttrium, and zirconium for PET and SPECT imaging of disease Acc. Chem. Res. 2009, 110, 832– 841Google ScholarThere is no corresponding record for this reference.
- 43De Silva, R. A.; Peyre, K.; Pullambhatla, M.; Fox, J. J.; Pomper, M. G.; Nimmagadda, S. Imaging CXCR4 expression in human cancer xenografts: evaluation of monocyclam 64Cu–AMD3465 J. Nucl. Med. 2011, 52, 986– 993Google ScholarThere is no corresponding record for this reference.
- 44Yuan, H.; Schroeder, T.; Bowsher, J. E.; Hedlund, L. W.; Wong, T.; Dewhirst, M. W. Intertumoral differences in hypoxia selectivity of the PET imaging agent 64Cu(II)-diacetyl-bis(N4-methylthiosemicarbazone) J. Nucl. Med. 2006, 47, 989– 998Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XmtlWmtrg%253D&md5=805152c3ada8861fa94ae0e0a28c16daIntertumoral differences in hypoxia selectivity of the PET imaging agent 64Cu(II)-diacetyl-Bis(N4-methylthiosemicarbazone)Yuan, Hong; Schroeder, Thies; Bowsher, James E.; Hedlund, Laurence W.; Wong, Terence; Dewhirst, Mark W.Journal of Nuclear Medicine (2006), 47 (6), 989-998CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine)Cu-Diacetyl-bis(N4-methylthiosemicarbazone) (Cu-ATSM) is a recently developed PET imaging agent for tumor hypoxia. However, its accuracy and reliability for measuring hypoxia have not been fully characterized in vivo. The aim of this study was to evaluate 64Cu-ATSM as a hypoxia PET marker by comparing autoradiog. distributions of 64Cu-ATSM with a well-established hypoxia marker drug, EF5. Methods: R3230 mammary adenocarcinomas (R3230Ac), fibrosarcomas (FSA), and 9L gliomas (9L) were used in the study. EF5 and Hoechst 33342, a vascular perfusion marker, were administered to the animal for immunohistochem. anal. 64Cu-ATSM microPET and autoradiog. were performed on the same animal. The tumor-to-muscle ratio (T/M ratio) and standardized uptake values (SUVs) were characterized for these 3 different types of tumors. Five types of images-microPET, autoradiog., EF5 immunostaining, Hoechst fluorescence vascular imaging, and hematoxylin-andeosin histol.-were superimposed, evaluated, and compared. Results: A significantly higher T/M ratio and SUV were seen for FSA compared with R3230Ac and 9L. Spatial correlation anal. between 64Cu-ATSM autoradiog. and EF5 immunostained images varied between the 3 tumor types. There was close correlation of 64Cu-ATSM uptake and hypoxia in R3230Ac and 9L tumors but not in FSA tumors. Interestingly, elevated 64Cu-ATSM uptake was obsd. in well-perfused areas in FSA, indicating a correlation between 64Cu-ATSM uptake and vascular perfusion as opposed to hypoxia. The same relationship was obsd. with 2 other hypoxia markers, pimonidazole and carbonic anhydrase IX, in FSA tumors. Breathing carbogen gas significantly decreased the hypoxia level measured by EF5 staining in FSA-bearing rats but not the uptake of 64Cu-ATSM. These results indicate that some other 64Cu-ATSM retention mechanisms, as opposed to hypoxia, are involved in this type of tumor. Conclusion: To our knowledge, this study is the first comparison between 64Cu-ATSM uptake and immunohistochem. in these 3 tumors. Although we have shown that 64Cu-ATSM is a valid PET hypoxia marker in some tumor types, but not for all, this tumor type-dependent hypoxia selectivity of 64Cu-ATSM challenges the use of 64Cu-ATSM as a universal PET hypoxia marker. Further studies are needed to define retention mechanisms for this PET marker.
- 45Liu, Z.; Li, Z. B.; Cao, Q.; Liu, S.; Wang, F.; Chen, X. Small-animal PET of tumors with (64)Cu-labeled RGD–bombesin heterodimer J. Nucl. Med. 2009, 50, 1168– 1177Google ScholarThere is no corresponding record for this reference.
- 46Dumont, R. A.; Deininger, F.; Haubner, R.; Maecke, H. R.; Weber, W. A.; Fani, M. Novel (64)Cu- and (68)Ga-labeled RGD conjugates show improved PET imaging of alpha(nu)beta(3) integrin expression and facile radiosynthesis J. Nucl. Med. 2011, 52, 1276– 1284Google ScholarThere is no corresponding record for this reference.
- 47Fani, M.; Del Pozzo, L.; Abiraj, K.; Mansi, R.; Tamma, M. L.; Cescato, R.; Waser, B.; Weber, W. A.; Reubi, J. C.; Maecke, H. R. PET of somatostatin receptor-positive tumors using 64Cu– and 68Ga–somatostatin antagonists: the chelate makes the difference J. Nucl. Med. 2011, 52, 1110– 1118Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvVKltro%253D&md5=3225eb0d38a7e33658a8a5168e914497PET of somatostatin receptor-positive tumors using 64Cu- and 68Ga-somatostatin antagonists: the chelate makes the differenceFani, Melpomeni; Del Pozzo, Luigi; Abiraj, Keelara; Mansi, Rosalba; Tamma, Maria Luisa; Cescato, Renzo; Waser, Beatrice; Weber, Wolfgang A.; Reubi, Jean Claude; Maecke, Helmut R.Journal of Nuclear Medicine (2011), 52 (7), 1110-1118CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine)Somatostatin-based radiolabeled peptides have been successfully introduced into the clinic for targeted imaging and radionuclide therapy of somatostatin receptor (sst)-pos. tumors, esp. of subtype 2 (sst2). The clin. used peptides are exclusively agonists. Recently, we showed that radiolabeled antagonists may be preferable to agonists because they showed better pharmacokinetics, including higher tumor uptake. Factors detg. the performance of radioantagonists have only scarcely been studied. Here, we report on the development and evaluation of four 64Cu or 68Ga radioantagonists for PET of sst2-pos. tumors. Methods: The novel antagonist p-Cl-Phe-cyclo(D-Cys-Tyr-D-4-amino-Phe(carbamoyl)-Lys-Thr-Cys)D-Tyr-NH2 (LM3) was coupled to 3 macrocyclic chelators, namely 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (CB-TE2A), 1,4,7-triazacyclononane, 1-glutaric acid-4,7-acetic acid (NODAGA), and DOTA. 64/natCu- and 68/natGa-NODAGA-LM3 were prepd. at room temp., and 64/natCu-CB-TE2A-LM3 and 68/natGa-DOTA-LM3 were prepd. at 95°C. Binding affinity and antagonistic properties were detd. with receptor autoradiog. and immunofluorescence microscopy using human embryonic kidney (HEK)-sst2 cells. In vitro internalization and dissocn. was evaluated using the same cell line. Biodistribution and small-animal PET studies were performed with HEK-sst2 xenografts. Results: All metallopeptides demonstrated antagonistic properties. The affinities depend on chelator and radiometal and vary about 10-fold; 68/natGa-NODAGA-LM3 has the lowest half maximal inhibitory concn. (1.3 ± 0.3 nmol/L). The biodistribution studies show impressive tumor uptake at 1 h after injection, particularly of 64Cu- and 68Ga-NODAGA-LM3 (∼40 percentage injected dose per g of tissue [%ID/g]), which were proven to be specific. Background clearance was fast and the tumor washout relatively slow for 64Cu-NODAGA-LM3 (∼15 %ID/g, 24 h after injection) and almost negligible for 64Cu-CB-TE2A-LM3 (26.9 ± 3.3 %ID/g and 21.6 ± 2.1 %ID/g, 4 and 24 h after injection, resp.). Tumor-to-normal-tissue ratios were significantly higher for 64Cu-NODAGA-LM3 than for 64Cu-CB-TE2A-LM3 (tumor-to-kidney, 12.8 ± 3.6 and 1.7 ± 0.3, resp.; tumor-to-muscle, 1,342 ± 115 and 75.2 ± 8.5, resp., at 24 h, P < 0.001). Small-animal PET shows clear tumor localization and high image contrast, esp. for 64Cu- and 68Ga-NODAGA-LM3. Conclusion: This article demonstrates the strong dependence of the affinity and pharmacokinetics of the somatostatin-based radioantagonists on the chelator and radiometal. 64Cu- and 68Ga-NODAGA-LM3 and 64Cu-CB-TE2A-LM3 are promising candidates for clin. translation because of their favorable pharmacokinetics and the high image contrast on PET scans.
- 48Liu, Z.; Li, Z. B.; Cao, Q.; Liu, S.; Wang, F.; Chen, X.; Small-animal, P. E. T. of tumors with (64)Cu-labeled RGD–bombesin heterodimer J. Nucl. Med. 2009, 50, 1168– 1177Google ScholarThere is no corresponding record for this reference.
- 49Rockey, W. M.; Huang, L.; Kloepping, K. C.; Baumhover, N. J.; Giangrande, P. H.; Schultz, M. K. Synthesis and radiolabeling of chelator–RNA aptamer bioconjugates with copper-64 for targeted molecular imaging Bioorg. Med. Chem. 2011, 19, 4080– 4090Google ScholarThere is no corresponding record for this reference.
- 50Hou, G. L.; Li, Y. H.; Zhang, Z. L.; Xiong, Y. H.; Chen, X. F.; Yao, K.; Liu, Z. W.; Han, H.; Qin, Z. K.; Zhou, F. J. A modified technique for neourethral anastomosis in orthotopic neobladder reconstruction Urology 2009, 74, 1145– 1149Google ScholarThere is no corresponding record for this reference.
- 51Cooper, M. S.; Ma, M. T.; Sunassee, K.; Shaw, K. P.; Williams, J. D.; Paul, R. L.; Donnelly, P. S.; Blower, P. J. Comparison of (64)Cu-complexing bifunctional chelators for radioimmunoconjugation: labeling efficiency, specific activity, and in vitro/in vivo stability Bioconjugate Chem. 2012, 23, 1029– 1039Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkvVyqurg%253D&md5=649071237104b7aaffb6d032acce51daComparison of 64Cu-Complexing Bifunctional Chelators for Radioimmunoconjugation: Labeling Efficiency, Specific Activity, and in Vitro/in Vivo StabilityCooper, Maggie S.; Ma, Michelle T.; Sunassee, Kavitha; Shaw, Karen P.; Williams, Jennifer D.; Paul, Rowena L.; Donnelly, Paul S.; Blower, Philip J.Bioconjugate Chemistry (2012), 23 (5), 1029-1039CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)High radiolabeling efficiency, preferably to high specific activity, and good stability of the radioimmunoconjugate are essential features for a successful immunoconjugate for imaging or therapy. In this study, the radiolabeling efficiency, in vitro stability, and biodistribution of immunoconjugates with eight different bifunctional chelators labeled with 64Cu were compared. The anti-CD20 antibody, rituximab, was conjugated to four macrocyclic bifunctional chelators (p-SCN-Bn-DOTA, p-SCN-Bn-Oxo-DO3A, p-SCN-NOTA, and p-SCN-PCTA), three DTPA derivs. (p-SCN-Bn-DTPA, p-SCN-CHX-A''-DTPA, and ITC-2B3M-DTPA), and a macrobicyclic hexamine (sarcophagine) chelator (sar-CO2H) = (1-NH2-8-NHCO(CH2)3CO2H)sar where sar = sarcophagine = 3,6,10,13,16,19-hexaazabicyclo[6.6.6]icosane. Radiolabeling efficiency under various conditions, in vitro stability in serum at 37 °C, and in vivo biodistribution and imaging in normal mice over 48 h were studied. All chelators except sar-CO2H were conjugated to rituximab by thiourea bond formation with an av. of 4.9 ± 0.9 chelators per antibody mol. Sar-CO2H was conjugated to rituximab by amide bond formation with 0.5 chelators per antibody mol. Efficiencies of 64Cu radiolabeling were dependent on the concn. of immunoconjugate. Notably, the 64Cu-NOTA-rituximab conjugate demonstrated the highest radiochem. yield (95%) under very dil. conditions (31 nM NOTA-rituximab conjugate). Similarly, sar-CO-rituximab, contg. 1/10th the no. of chelators per antibody compared to that of other conjugates, retained high labeling efficiency (98%) at an antibody concn. of 250 nM. In contrast to the radioimmunoconjugates contg. DTPA derivs., which demonstrated poor serum stability, all macrocyclic radioimmunoconjugates were very stable in serum with <6% dissocn. of 64Cu over 48 h. In vivo biodistribution profiles in normal female Balb/C mice were similar for all the macrocyclic radioimmunoconjugates with most of the activity remaining in the blood pool up to 48 h. While all the macrocyclic bifunctional chelators are suitable for mol. imaging using 64Cu-labeled antibody conjugates, NOTA and sar-CO2H show significant advantages over the others in that they can be radiolabeled rapidly at room temp., under dil. conditions, resulting in high specific activity.
- 52Moi, M. K.; Meares, C. F.; McCall, M. J.; Cole, W. C.; DeNardo, S. J. Copper chelates as probes of biological systems: stable copper complexes with a macrocyclic bifunctional chelating agent Anal. Biochem. 1985, 148, 249– 253Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2MXltlyjsLs%253D&md5=6f8bb1c6ff5effdf2b3bc2672e025802Copper chelates as probes of biological systems: stable copper complexes with a macrocyclic bifunctional chelating agentMoi, Min K.; Meares, Claude F.; McCall, Michael J.; Cole, William C.; DeNardo, Sally J.Analytical Biochemistry (1985), 148 (1), 249-53CODEN: ANBCA2; ISSN:0003-2697.The synthesis of a new bifunctional metal chelator, 6-(p-bromoacetamidobenzyl)-1,4,8,11-tetraazacyclotetradecane-N,N',N'',N'''-tetraacetic acid (TETA) is described, which can be covalently attached to proteins and which binds Cu stably in human serum under physiol. conditions. 67Cu chelates of p-nitrobenzyl-TETA, p-nitrobenzyl-EDTA, diethylenetriaminepentaacetic acid (DTPA)-N-butylamide, and DTPA, as well as conjugates of Cu chelates with mouse monoclonal antibody lym-1 were also prepd. and their stability in human serum was examd. In contrast to TETA, Cu chelates prepd. from analogs of EDTA or DTPA were not stable to prolonged incubation in human serum and rapidly lost Cu to serum albumin.
- 53Jones-Wilson, T. M.; Deal, K. A.; Anderson, C. J.; McCarthy, D. W.; Kovacs, Z.; Motekaitis, R. J.; Sherry, A. D.; Martell, A. E.; Welch, M. J. The in vivo behavior of copper-64-labeled azamacrocyclic complexes Nucl. Med. Biol. 1998, 25, 523– 530Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXlt1Wktrk%253D&md5=aa5cbc7f7ab5fb08f6659903f14f2fa3The in vivo behavior of copper-64-labeled azamacrocyclic complexesJones-Wilson, Teresa M.; Deal, Kim A.; Anderson, Carolyn J.; McCarthy, Deborah W.; Kovacs, Zoltan; Motekaitis, Ramunas J.; Sherry, A. Dean; Martell, Arthur E.; Welch, Michael J.Nuclear Medicine and Biology (1998), 25 (6), 523-530CODEN: NMBIEO; ISSN:0969-8051. (Elsevier Science Inc.)The use of copper radioisotopes in imaging and therapy applications has created a greater need for bifunctional chelates (BFCs) for complexing copper radioisotopes to biomols. It has been demonstrated that the charge and lipophilicity of the Cu-BFC complex has a significant effect on the in vivo behavior of the radiolabeled Cu-BFC-biomol. conjugate. To evaluate the effects of charge, stability, and macrocyclic backbone size on the biol. behavior of 64Cu complexes, a series of macrocyclic 64Cu complexes have been prepd., and the biodistributions of these agents were evaluated in normal Sprague-Dawley rats. Two macrocyclic backbones, dodecane and tetradecane, were evaluated; cyclen, DOTA, and DO2A were dodecane backbone derivs., and cyclam, TETA, and et-cyclam were tetradecane backbone derivs. The biodistributions of the 64Cu-labeled complexes correlated with differences in the size of the macrocycle backbone and the formal charge of the complex. All compds. showed uptake and clearance through the liver and kidneys; however, the pos. charged 64Cu complexes showed significantly higher uptake in both of these organs than did the neg. charged or neutral complexes. 64Cu-TETA, a neg. charged complex with the tetradecane backbone, had the most efficient clearance by 24 h' postinjection. These data suggest that neg. charged complexes may have more favorable clearance properties when used as BFCs.
- 54Garrison, J. C.; Rold, T. L.; Sieckman, G. L.; Figueroa, S. D.; Volkert, W. A.; Jurisson, S. S.; Hoffman, T. J. In vivo evaluation and small-animal PET/CT of a prostate cancer mouse model using 64Cu bombesin analogs: side-by-side comparison of the CB-TE2A and DOTA chelation systems J. Nucl. Med. 2007, 48, 1327– 1337Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVSktbzP&md5=6d3effae9572a90579e6590329a6b375In vivo evaluation and small-animal PET/CT of a prostate cancer mouse model using 64Cu bombesin analogs: side-by-side comparison of the CB-TE2A and DOTA chelation systemsGarrison, Jered C.; Rold, Tammy L.; Sieckman, Gary L.; Figueroa, Said Daibes; Volkert, Wynn A.; Jurisson, Silvia S.; Hoffman, Timothy J.Journal of Nuclear Medicine (2007), 48 (8), 1327-1337CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine)The BB2 receptor subtype, of the bombesin family of receptors, has been shown to be highly overexpressed in a variety of human tumors, including prostate cancer. Bombesin (BBN), a 14-amino acid peptide, has been shown to target the BB2 receptor with high affinity. 64Cu (half-life = 12.7 h, β+: 18%, Eβ+max = 653 keV; β-: 37%, Eβ-max = 578 keV) is a radioisotope that has clin. potential for application in both diagnostic imaging and radionuclide therapy. Recently, new chelation systems such as 1,4,8,11-tetraazabicyclo[6.6.2]hexadecane-4,11-diacetic acid (CB-TE2A) have been reported to significantly stabilize the 64Cu radiometal in vivo. The increased stability of the 64Cu-CB-TE2A chelate complex has been shown to significantly reduce nontarget retention compared with tetraazamacrocycles such as 1,4,7,10-tetraazacyclodoadecane-N,N',N",N'"-tetraacetic acid (DOTA). The aim of this study was to det. whether the CB-TE2A chelation system could significantly improve the in vivo stability of 64Cu bombesin analogs. The study directly compares 64Cu bombesin analogs using the CB-TE2A and DOTA chelation systems in a prostate cancer xenograft SCID (severely compromised immunodeficient) mouse model. Methods: The CB-TE2A-8-AOC-BBN(7-14)NH2 and DOTA-8-AOC-BBN(7-14) NH2 conjugates were synthesized and radiolabeled with 64Cu. The receptor-binding affinity and internalization profile of each metalated conjugate was evaluated using PC-3 cells. Pharmacokinetic and small-animal PET/CT studies were performed using female SCID mice bearing PC-3 xenografts. Results: In vivo BB2 receptor targeting was confirmed by tumor uptake values of 6.95 ± 2.27 and 4.95 ± 0.91 %ID/g (percentage injected dose per g) at the 15-min time point for the 64Cu-CB-TE2A and 64Cu-DOTA radioconjugates, resp. At the 24-h time point, liver uptake was substantially reduced for the 64Cu-CB-TE2A radioconjugate (0.21 ± 0.06 %ID/g) compared with the 64Cu-DOTA radioconjugate (7.80 ± 1.51 %ID/g). The 64Cu-CB-TE2A-8-AOC-BBN(7-14)NH2 radioconjugate demonstrated significant clearance, 98.60 ± 0.28 %ID, from the mouse at 24 h after injection. In contrast, only 67.84 ± 5.43 %ID of the 64Cu activity was excreted using the 64Cu-DOTA-8-AOC-BBN(7-14)NH2 radioconjugate because of nontarget retention. Conclusion: The pharmacokinetic and small-animal PET/CT studies demonstrate significantly improved nontarget tissue clearance for the 64Cu-CB-TE2A8-AOC-BBN(7-14)NH2. This is attributed to the improved in vivo stability of the 64Cu-CB-TE2A chelate complex as compared with the 64Cu-DOTA chelate complex.
- 55Sun, X.; Wuest, M.; Weisman, G. R.; Wong, E. H.; Reed, D. P.; Boswell, C. A.; Motekaitis, R.; Martell, A. E.; Welch, M. J.; Anderson, C. J. Radiolabeling and in vivo behavior of copper-64-labeled cross-bridged cyclam ligands J. Med. Chem. 2002, 45, 469– 477Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXptFKjs7s%253D&md5=95a2a757a82322ce1c46b6bcb323903bRadiolabeling and in vivo behavior of copper-64-labeled cross-bridged cyclam ligandsSun, Xiankai; Wuest, Melinda; Weisman, Gary R.; Wong, Edward H.; Reed, David P.; Boswell, C. Andrew; Motekaitis, Ramunas; Martell, Arthur E.; Welch, Michael J.; Anderson, Carolyn J.Journal of Medicinal Chemistry (2002), 45 (2), 469-477CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Macrocyclic chelators and their metal complexes have widespread applications in the biomedical sciences, including radiopharmaceutical chem. The use of copper radionuclides in radiopharmaceuticals is increasing. Macrocyclic chelators have been found to have enhanced in vivo stability over acyclic chelators such as EDTA and diethylenetriaminepentaacetic acid (DTPA). The currently used chelators of choice for labeling copper radionuclides to biol. mols. are analogs of TETA (1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid); however, recent reports have demonstrated evidence of in vivo instability of the radio-Cu(II)-TETA complexes. A new class of structurally reinforced macrocycles, the "cross-bridged" cyclam derivs., form highly stable complexes with Cu(II) that are resistant to dissocn. in strong acid. Here, we evaluate a series of 64Cu(II) cross-bridged macrocyclic complexes for biol. stability and in vivo behavior. The ligands evaluated include the parent ligand, 1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (1), and three 4,11-di-pendant arm derivs.: 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (2); 4,11-bis(N,N-diethyl-amidomethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (3); and 4,11-bis(amidoethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (4). Copper-64 formed complexes with ligands 1-4 in high radiochem. yields. The 64Cu-2 complex was neutral, while 64Cu complexes of 1, 3, and 4 were pos. charged. All complexes showed no decompn. in rat serum out to 24 h. Biodistribution expts. in Sprague-Dawley rats indicated that 64Cu-1, -3, and -4 were taken up by the liver and kidney and cleared slowly over 24 h, whereas 64Cu-2 cleared rapidly from all tissues. The rapid clearance of the 64Cu-2 complex from the blood and liver, as well as liver metab. expts. in rats, suggests that it is highly stable in vivo. A bifunctional chelator of 2 is a significant candidate for labeling copper radionuclides to biol. mols. for diagnostic imaging and targeted radiotherapy.
- 56Boswell, C. A.; Sun, X.; Niu, W.; Weisman, G. R.; Wong, E. H.; Rheingold, A. L.; Anderson, C. J. Comparative in vivo stability of copper-64-labeled cross-bridged and conventional tetraazamacrocyclic complexes J. Med. Chem. 2004, 47, 1465– 1474Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtFagtLw%253D&md5=542e8b853716a629b641740f102a48dbComparative in Vivo Stability of Copper-64-Labeled Cross-Bridged and Conventional Tetraazamacrocyclic ComplexesBoswell, C. Andrew; Sun, Xiankai; Niu, Weijun; Weisman, Gary R.; Wong, Edward H.; Rheingold, Arnold L.; Anderson, Carolyn J.Journal of Medicinal Chemistry (2004), 47 (6), 1465-1474CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The increased use of copper radioisotopes in radiopharmaceutical applications has created a need for bifunctional chelators (BFCs) that form stable radiocopper complexes and allow covalent attachment to biol. mols. The chelators most commonly utilized for labeling copper radionuclides to biomols. are analogs of 1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid (TETA); however, recent reports have communicated the instability of the radio-Cu(II)-TETA complexes in vivo. A class of bicyclic tetraazamacrocycles, the ethylene "cross-bridged" cyclam (CB-cyclam) derivs., form highly kinetically stable complexes with Cu(II) and therefore may be less susceptible to transchelation than their nonbridged analogs in vivo. Herein we report results on the relative biol. stabilities and identification of the resulting radiolabeled metabolites of a series of 64Cu-labeled macrocyclic complexes. Metab. studies in normal rat liver have revealed that the 64Cu complex of 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (64Cu-CB-TE2A) resulted in significantly lower values of protein-assocd. 64Cu than 64Cu-TETA [13 ± 6% vs 75 ± 9% at 4 h]. A similar trend was obsd. for the corresponding cyclen derivs., with the 64Cu complex of 4,10-bis(carboxymethyl)-1,4,7,10-tetraazabicyclo[5.5.2]tetradecane (64Cu-CB-DO2A) undergoing less transchelation than the 64Cu complex of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (64Cu-DOTA) [61 ± 14% vs 90.3 ± 0.5% protein assocd. 64Cu at 4 h]. These data indicate that the structurally reinforcing cross-bridge enhances in vivo stability by reducing metal loss to protein in both the cyclam and cyclen cross-bridged 64Cu complexes and that 64Cu-CB-TE2A is superior to 64Cu-CB-DO2A in that regard. These findings further suggest that a bifunctional chelator deriv. of CB-TE2A is a highly desirable alternative for labeling copper radionuclides to biol. mols. for diagnostic imaging and targeted radiotherapy.
- 57Sprague, J. E.; Peng, Y.; Sun, X.; Weisman, G. R.; Wong, E. H.; Achilefu, S.; Anderson, C. J. Preparation and biological evaluation of copper-64-labeled tyr3-octreotate using a cross-bridged macrocyclic chelator Clin. Cancer Res. 2004, 10, 8674– 8682Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtFGjsL7F&md5=645d85d633c7cc512392f9ec2c33d68dPreparation and biological evaluation of copper-64-labeled Tyr3-octreotate using a cross-bridged macrocyclic chelatorSprague, Jennifer E.; Peng, Yijie; Sun, Xiankai; Weisman, Gary R.; Wong, Edward H.; Achilefu, Samuel; Anderson, Carolyn J.Clinical Cancer Research (2004), 10 (24), 8674-8682CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Somatostatin receptors (SSTr) are expressed on many neuroendocrine tumors, and several radiotracers have been developed for imaging these types of tumors. For this reason, peptide analogs of somatostatin have been well characterized. Copper-64 (t1/2 = 12.7 h), a positron emitter suitable for positron emission tomog. (PET) imaging, was shown recently to have improved in vivo clearance properties when chelated by the cross-bridged tetraazamacrocycle 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo(6.6.2)hexadecane (CB-TE2A) compared with 1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid (TETA). CB-TE2A and TETA were conjugated to the somatostatin analog tyrosine-3-octreotate (Y3-TATE) for evaluation of CB-TE2A as a bifunctional chelator of 64Cu. The in vitro affinity of each compd. for SSTr was detd. using a homologous competitive binding assay. In vivo characteristics of both radiolabeled compds. were examd. in biodistribution and microPET studies of AR42J tumor-bearing rats. Cu-CB-TE2A-Y3-TATE (Kd = 1.7 nmol/L) and Cu-TETA-Y3-TATE (Kd = 0.7 nmol/L) showed similar affinities for AR42J derived SSTr. In biodistribution studies, nonspecific uptake in blood and liver was lower for 64Cu-CB-TE2A-Y3-TATE. Differences increased with time such that, at 4 h, blood uptake was 4.3-fold higher and liver uptake was 2.4-fold higher for 64Cu-TETA-Y3-TATE than for 64Cu-CB-TE2A-Y3-TATE. In addn., 4.4-times greater tumor uptake was detected with 64Cu-CB-TE2A-Y3-TATE than with 64Cu-TETA-Y3-TATE at 4 h postinjection. MicroPET imaging yielded similar results. CB-TE2A appears to be a superior in vivo bifunctional chelator of 64Cu for use in mol. imaging by PET or targeted radiotherapy due to both improved nontarget organ clearance and higher target organ uptake of 64Cu-CB-TE2A-Y3-TATE compared with 64Cu-TETA-Y3-TATE.
- 58Ait-Mohand, S.; Fournier, P.; Dumulon-Perreault, V.; Kiefer, G. E.; Jurek, P.; Ferreira, C. L.; Benard, F.; Guerin, B. Evaluation of 64Cu-labeled bifunctional chelate–bombesin conjugates Bioconjugate Chem. 2011, 22, 1729– 1735Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpsFSrtL8%253D&md5=b36324b077c22659f4525ed8d655e66bEvaluation of 64Cu-Labeled Bifunctional Chelate-Bombesin ConjugatesAit-Mohand, Samia; Fournier, Patrick; Dumulon-Perreault, Veronique; Kiefer, Garry E.; Jurek, Paul; Ferreira, Cara L.; Benard, Francois; Guerin, BrigitteBioconjugate Chemistry (2011), 22 (8), 1729-1735CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Several bifunctional chelates (BFCs) were investigated as carriers of 64Cu for PET imaging. The most widely used chelator for 64Cu labeling of BFCs is DOTA (1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tretraacetic acid), even though this complex exhibits only moderate in vivo stability. In this study, we prepd. a series of alternative chelator-peptide conjugates labeled with 64Cu, measured in vitro receptor binding affinities in human breast cancer T47D cells expressing the gastrin-releasing peptide receptor (GRPR) and compared their in vivo stability in mice. DOTA-, NOTA-(1,4,7-triazacyclononane-1,4,7-triacetic acid), PCTA-(3,6,9,15-tetraazabicyclo[9.3.1]pentadeca-1(15),11,13-triene-3,6,9-triacetic acid), and Oxo-DO3A-(1-oxa-4,7,10-triazacyclododecane-4,7,10-triacetic acid) peptide conjugates were prepd. using H2N-Aoc-[D-Tyr6,βAla11,Thi13,Nle14]bombesin(6-14) (BBN) as a peptide template. The BBN moiety was selected since it binds with high affinity to the GRPR, which is overexpressed on human breast cancer cells. A convenient synthetic approach for the attachment of aniline-BFC to peptides on solid support is also presented. To facilitate the attachment of the aniline-PCTA and aniline-Oxo-DO3A to the peptide via an amide bond, a succinyl spacer was introduced at the N-terminus of BBN. The partially protected aniline-BFC (p-H2N-Bn-PCTA(Ot-Bu)3 or p-H2N-Bn-DO3A(Ot-Bu)3) was then coupled to the resulting N-terminal carboxylic acid preactivated with DEPBT/ClHOBt on resin. After cleavage and purifn., the peptide-conjugates were labeled with 64Cu using [64Cu]Cu(OAc)2 in 0.1 M ammonium acetate buffer at 100 °C for 15 min. Labeling efficacy was >90% for all peptides; Oxo-DO3A-BBN was incubated an addnl. 150 min at 100 °C to achieve this high yield. Specific activities varied from 76 to 101 TBq/mmol. Competition assays on T47D cells showed that all BFC-BBN complexes retained high affinity for the GRPR. All BFC-BBN 64Cu-conjugates were stable for over 20 h when incubated at 37 °C in mouse plasma samples. However, in vivo, only 37% of the 64Cu/Oxo-DO3A complex remained intact after 20 h while the 64Cu/DOTA-BBN complex was completely demetalated. In contrast, both 64Cu/NOTA- and 64Cu/PCTA-BBN conjugates remained stable during the 20 h time period. Our results indicate that it is possible to successfully conjugate aniline-BFC with peptide on solid support. Our data also show that 64Cu-labeled NOTA- and PCTA-BBN peptide conjugates are promising radiotracers for PET imaging of many human cancers overexpressing the GRP receptor.
- 59Ferreira, C. L.; Lamsa, E.; Woods, M.; Duan, Y.; Fernando, P.; Bensimon, C.; Kordos, M.; Guenther, K.; Jurek, P.; Kiefer, G. E. Evaluation of Bifunctional Chelates for the Development of Gallium-Based Radiopharmaceuticals Bioconjugate Chem. 2010, 21, 531– 536Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXit1Wrsr0%253D&md5=11d7e167a99c64067629f0bd4460d9bdEvaluation of Bifunctional Chelates for the Development of Gallium-Based RadiopharmaceuticalsFerreira, Cara L.; Lamsa, Eric; Woods, Michael; Duan, Yin; Fernando, Pasan; Bensimon, Corinne; Kordos, Myra; Guenther, Katharina; Jurek, Paul; Kiefer, Garry E.Bioconjugate Chemistry (2010), 21 (3), 531-536CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Ga radioisotopes, including the generator-produced positron-emitting isotope 68Ga (t1/2 = 68 min), are of increasing interest for the development of new radiopharmaceuticals. Bifunctional chelates (BFCs) that can be efficiently radiolabeled with Ga to yield complexes with good in vivo stability are needed. To this end, we undertook a systematic comparison of four BFCs contg. different chelating moieties: two novel BFCs, p-NO2-Bn-Oxo (1-oxa-4,7,10-triazacyclododecane-4,7,10-triacetic acid) and p-NO2-Bn-PCTA (3,6,9,15-tetraazabicyclo [9.3.1]pentadeca-1(15),11,13-triene-3,6,9-triacetic acid), and two more commonly used BFCs, p-NO2-Bn-DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) and p-NO2-Bn-NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid). Each BFC was compared with respect to radiolabeling conditions, radiochem. yield, stability, and in vivo clearance properties. p-NO2-Bn-PCTA, p-NO2-Bn-Oxo, and p-NO2-Bn-NOTA were all more efficiently radiolabeled with Ga compared to p-NO2-Bn-DOTA. p-NO2-Bn-DOTA required longer reaction time, higher concns. of BFC, or heating to obtain equiv. radiochem. yields. Better stability was obsd. for p-NO2-Bn-NOTA and p-NO2-Bn-PCTA compared to p-NO2-Bn-DOTA and p-NO2-Bn-Oxo, esp. with respect to transmetalation to transferrin. Ga-radiolabled p-NO2-Bn-Oxo was found to be kinetically labile and therefore unstable in vivo. Ga-radiolabeled p-NO2-Bn-NOTA and p-NO2-Bn-PCTA were relatively inert, while Ga-radiolabeled p-NO2-Bn-DOTA had intermediate stability, losing >20% of Ga in less than one hour when incubated with apo-transferrin. Similar stability differences were seen when incubating at pH 2. In vivo PET imaging and biodistribution studies in mice showed that 68Ga-radiolabeled p-NO2-Bn-PCTA, p-NO2-Bn-NOTA, and p-NO2-Bn-DOTA all cleared through the kidneys. While there was no statistical difference in the biodistribution results of 68Ga-radiolabeled p-NO2-Bn-PCTA and p-NO2-Bn-DOTA, 68Ga-radiolabeled p-NO2-Bn-NOTA cleared more rapidly from blood and muscle tissue but retained at up to 5 times higher activity in the kidneys.
- 60Ferreira, C. L.; Yapp, D. T.; Lamsa, E.; Gleave, M.; Bensimon, C.; Jurek, P.; Kiefer, G. E. Evaluation of novel bifunctional chelates for the development of Cu-64-based radiopharmaceuticals Nucl. Med. Biol. 2008, 35, 875– 882Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVaisLjI&md5=9013cd14b9030ed7df0ea9a4b33cbc30Evaluation of novel bifunctional chelates for the development of Cu-64-based radiopharmaceuticalsFerreira, Cara L.; Yapp, Donald T.; Lamsa, Eric; Gleave, Martin; Bensimon, Corinne; Jurek, Paul; Kiefer, Garry E.Nuclear Medicine and Biology (2008), 35 (8), 875-882CODEN: NMBIEO; ISSN:0969-8051. (Elsevier Inc.)Background: Currently available bifunctional chelates (BFCs) for attaching Cu-64 to a targeting mol. are limited by either their radiolabeling conditions or in vivo stability. With the goal of identifying highly effective BFCs, we compared the properties of two novel BFCs, 1-oxa-4,7,10-triazacyclododecane-S-5-(4-nitrobenzyl)-4,7,10-triacetic acid (p-NO2-Bn-Oxo) and 3,6,9,15-tetraazabicyclo[9.3.1]pentadeca-1(15),11,13-triene-S-4-(4-nitrobenzyl)-3,6,9-triacetic acid (p-NO2-Bn-PCTA), with the commonly used S-2-(4-nitrobenzyl)-1,4,7,10-tetraazacyclododecanetetraacetic acid (p-NO2-Bn-DOTA). Methods: p-NO2-Bn-DOTA, p-NO2-Bn-Oxo and p-NO2-Bn-PCTA were each radiolabeled with Cu-64 under various conditions to assess the reaction kinetics and robustness of the radiolabeling. Stability of each Cu-64 BFC complex was evaluated at low pH and in serum. Small animal positron emission tomog. imaging and biodistribution studies in mice were undertaken. Results: p-NO2-Bn-Oxo and p-NO2-Bn-PCTA possessed superior reaction kinetics compared to p-NO2-Bn-DOTA under all radiolabeling conditions; >98% radiochem. yields were achieved in <5 min at room temp. even when using near stoichiometric amts. of BFC. Under nonideal conditions, such as low or high pH, high radiochem. yields were still achievable with the novel BFCs. The radiolabeled compds. were stable in serum and at pH 2 for 48 h. The imaging and biodistribution of the Cu-64-radiolabeled BFCs illustrated differences between the BFCs, including preferential clearance via the kidneys for the p-NO2-Bn-PCTA Cu-64 complex. Conclusions: The novel BFCs facilitated efficient Cu-64 radiolabeling under mild conditions to produce stable complexes at potentially high specific activities. These BFCs may find wide utility in the development of Cu-64-based radiopharmaceuticals.
- 61Olszewski, R. T.; Bukhari, N.; Zhou, J.; Kozikowski, A. P.; Wroblewski, J. T.; Shamimi-Noori, S.; Wroblewska, B.; Bzdega, T.; Vicini, S.; Barton, F. B.; Neale, J. H. NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR J. Neurochem. 2004, 89, 876– 885Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksVyksLg%253D&md5=4ca607a68135b77ae0cd997eed5d2566NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluROlszewski, Rafal T.; Bukhari, Noreen; Zhou, Jia; Kozikowski, Alan P.; Wroblewski, Jarda T.; Shamimi-Noori, Susan; Wroblewska, Barbara; Bzdega, Tomasz; Vicini, Stefano; Barton, Franca B.; Neale, Joseph H.Journal of Neurochemistry (2004), 89 (4), 876-885CODEN: JONRA9; ISSN:0022-3042. (Blackwell Publishing Ltd.)Phencyclidine (PCP) administration elicits pos. and neg. symptoms that resemble those of schizophrenia and is widely accepted as a model for the study of this human disorder. Group II metabotropic glutamate receptor (mGluR) agonists have been reported to reduce the behavioral and neurochem. effects of PCP. The peptide neurotransmitter, N-acetylaspartylglutamate (NAAG), is a selective group II agonist. The authors synthesized and characterized a urea-based NAAG analog, ZJ43. This novel compd. is a potent inhibitor of enzymes, glutamate carboxypeptidase II (Ki = 0.8 nM) and III (Ki = 23 nM) that deactivate NAAG following synaptic release. ZJ43 (100 μM) does not directly interact with NMDA receptors or metabotropic glutamate receptors. Administration of ZJ43 significantly reduced PCP-induced motor activation, falling while walking, stereotypic circling behavior, and head movements. To test the hypothesis that this effect of ZJ43 was mediated by increasing the activation of mGluR3 via increased levels of extracellular NAAG, the group II mGluR selective antagonist LY341495 was co-administered with ZJ43 prior to PCP treatment. This antagonist completely reversed the effects of ZJ43. Addnl., LY341495 alone increased PCP-induced motor activity and head movements suggesting that normal levels of NAAG act to moderate the effect of PCP on motor activation via a group II mGluR. These data support the view that NAAG peptidase inhibitors may represent a new therapeutic approach to some of the components of schizophrenia that are modeled by PCP.
- 62Silver, D. A.; Pellicer, I.; Fair, W. R.; Heston, W. D.; Cordon-Cardo, C. Prostate-specific membrane antigen expression in normal and malignant human tissues Clin. Cancer Res. 1997, 3, 81– 95Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1M%252FjsFamtA%253D%253D&md5=06fb3a1b21fc371a5e42843ed558e5ebProstate-specific membrane antigen expression in normal and malignant human tissuesSilver D A; Pellicer I; Fair W R; Heston W D; Cordon-Cardo CClinical cancer research : an official journal of the American Association for Cancer Research (1997), 3 (1), 81-5 ISSN:1078-0432.Prostate-specific membrane antigen is a type II membrane protein with folate hydrolase activity produced by prostatic epithelium. The expression of this molecule has also been documented in extraprostatic tissues, including small bowel and brain. In the present study, an extensive immunohistochemical analysis was performed on a panel of well-characterized normal and malignant human tissues to further define the pattern of prostate-specific membrane antigen (PSMA) expression. Detectable PSMA levels were identified in prostatic epithelium, duodenal mucosa, and a subset of proximal renal tubules. A subpopulation of neuroendocrine cells in the colonic crypts also exhibited PSMA immunoreactivity. All other normal tissues, including cerebral cortex and cerebellum, had undetectable levels of PSMA. Thirty-three of 35 primary prostate adenocarcinomas and 7 of 8 lymph node metastases displayed tumor cell PSMA immunostaining. Eight of 18 prostate tumors metastatic to bone expressed PSMA. All of the other nonprostatic primary tumors studied had undetectable PSMA levels. However, intense staining was observed in endothelial cells of capillary vessels in peritumoral and endotumoral areas of certain malignancies, including 8 of 17 renal cell carcinomas, 7 of 13 transitional cell carcinomas, and 3 of 19 colon carcinomas. Extraprostatic PSMA expression appears to be highly restricted. Nevertheless, its diverse anatomical distribution implies a broader functional significance than previously suspected. The decrease in PSMA immunoreactivity noted in advanced prostate cancer suggests that expression of this molecule may be linked to the degree of tumor differentiation. The neoexpression of PSMA in endothelial cells of capillary beds in certain tumors may be related to tumor angiogenesis and suggests a potential mechanism for specific targeting of tumor neovasculature.
- 63Prasanphanich, A. F.; Nanda, P. K.; Rold, T. L.; Ma, L.; Lewis, M. R.; Garrison, J. C.; Hoffman, T. J.; Sieckman, G. L.; Figueroa, S. D.; Smith, C. J. [64Cu-NOTA-8-Aoc-BBN(7–14)NH2] targeting vector for positron-emission tomography imaging of gastrin-releasing peptide receptor-expressing tissues Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 12462– 12467Google ScholarThere is no corresponding record for this reference.
- 64Chen, Y.; Pullambhatla, M.; Foss, C. A.; Byun, Y.; Nimmagadda, S.; Senthamizhchelvan, S.; Sgouros, G.; Mease, R. C.; Pomper, M. G. 2-(3-{1-Carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pen tanedioic acid, [18F]DCFPyL, a PSMA-based PET imaging agent for prostate cancer Clin. Cancer Res. 2011, 17, 7645– 7653Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1CqtrnM&md5=dca0465b3b65f7b553a622e40a0c74632-(3-{1-Carboxy-5-[(6-[18F]Fluoro-Pyridine-3-Carbonyl)-Amino]-Pentyl}-Ureido)-Pentanedioic Acid, [18F]DCFPyL, a PSMA-Based PET Imaging Agent for Prostate CancerChen, Ying; Pullambhatla, Mrudula; Foss, Catherine A.; Byun, Youngjoo; Nimmagadda, Sridhar; Senthamizhchelvan, Srinivasan; Sgouros, George; Mease, Ronnie C.; Pomper, Martin G.Clinical Cancer Research (2011), 17 (24), 7645-7653CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)PURPOSE: We have synthesized and evaluated in vivo 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid, [18F]DCFPyL, as a potential imaging agent for the prostate-specific membrane antigen (PSMA). PSMA is upregulated in prostate cancer epithelia and in the neovasculature of most solid tumors. Exptl. Design: [18F]DCFPyL was synthesized in two steps from the p-methoxybenzyl (PMB) protected lys-C(O)-glu urea precursor using 6-[18F]fluoronicotinic acid tetrafluorophenyl ester ([18F]F-Py-TFP) for introduction of 18F. Radiochem. synthesis was followed by biodistribution and imaging with PET in immunocompromised mice using isogenic PSMA PC3 PIP and PSMA- PC3 flu xenograft models. Human radiation dosimetry ests. were calcd. using OLINDA/EXM 1.0. RESULTS: DCFPyL displays a Ki value of 1.1 ± 0.1 nmol/L for PSMA. [18F]DCFPyL was produced in radiochem. yields of 36%-53% (decay cor.) and specific radioactivities of 340-480 Ci/mmol (12.6-17.8 GBq/μmol, n = 3). In an immunocompromised mouse model [18F]DCFPyL clearly delineated PSMA+ PC3 PIP prostate tumor xenografts on imaging with PET. At 2 h postinjection, 39.4 ± 5.4 percent injected dose per g of tissue (%ID/g) was evident within the PSMA+ PC3 PIP tumor, with a ratio of 358:1 of uptake within PSMA+ PC3 PIP to PSMA- PC3 flu tumor placed in the opposite flank. At or after 1 h postinjection, minimal nontarget tissue uptake of [18F]DCFPyL was obsd. The bladder wall is the dose-limiting organ. CONCLUSIONS: These data suggest [18F]DCFPyL as a viable, new positron-emitting imaging agent for PSMA-expressing tissues. Clin Cancer Res; 17(24); 7645-53.
- 65Hillier, S. M.; Maresca, K. P.; Lu, G.; Merkin, R. D.; Marquis, J. C.; Zimmerman, C. N.; Eckelman, W. C.; Joyal, J. L.; Babich, J. W. 99mTc-Labeled Small-Molecule Inhibitors of Prostate-Specific Membrane Antigen for Molecular Imaging of Prostate Cancer J. Nucl. Med. 2013, 54, 1369– 1376Google ScholarThere is no corresponding record for this reference.
- 66Elsasser-Beile, U.; Reischl, G.; Wiehr, S.; Buhler, P.; Wolf, P.; Alt, K.; Shively, J.; Judenhofer, M. S.; Machulla, H. J.; Pichler, B. J. PET imaging of prostate cancer xenografts with a highly specific antibody against the prostate-specific membrane antigen J. Nucl. Med. 2009, 50, 606– 611Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1MzjsFKmtA%253D%253D&md5=b717818e3336ebe156e782f46f30aaecPET imaging of prostate cancer xenografts with a highly specific antibody against the prostate-specific membrane antigenElsasser-Beile Ursula; Reischl Gerald; Wiehr Stefan; Buhler Patrick; Wolf Philipp; Alt Karen; Shively John; Judenhofer Martin S; Machulla Hans-Jurgen; Pichler Bernd JJournal of nuclear medicine : official publication, Society of Nuclear Medicine (2009), 50 (4), 606-11 ISSN:0161-5505.UNLABELLED: Prostate-specific membrane antigen (PSMA), a transmembrane glycoprotein, is highly expressed by virtually all prostate cancers and is currently the focus of several diagnostic and therapeutic strategies. We have previously reported on the generation of several monoclonal antibodies (mAb) and antibody fragments that recognize and bind with high affinity to the extracellular domain of cell-adherent PSMA. This article reports the in vivo behavior and tumor uptake of the radiolabeled anti-PSMA mAb 3/A12 and its potential as a tracer for PET. METHODS: The mAb 3/A12 was conjugated with the chelating agent 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) and radiolabeled with (64)Cu. Severe combined immunodeficient mice bearing PSMA-positive C4-2 prostate carcinoma xenografts were used for small-animal PET imaging. Mice with PSMA-negative DU 145 tumors served as controls. For PET studies, each animal received 20-30 microg of radiolabeled mAb corresponding to an activity of 7.6-11.5 MBq. Imaging was performed 3, 24, and 48 h after injection. After the last scan, the mice were sacrificed and tracer in vivo biodistribution was measured by gamma-counting. RESULTS: Binding of the mAb 3/A12 on PSMA-expressing C4-2 cells was only minimally influenced by DOTA conjugation. The labeling efficiency using (64)Cu and DOTA-3/A12 was 95.3% +/- 0.3%. The specific activity after (64)Cu labeling was between 327 and 567 MBq/mg. After tracer injection, static small-animal PET images of mice with PSMA-positive tumors revealed a tumor-to-background ratio of 3.3 +/- 1.3 at 3 h, 7.8 +/- 1.4 at 24 h, and 9.6 +/- 2.7 at 48 h. In contrast, no significant tracer uptake occurred in the PSMA-negative DU 145 tumors. These results were confirmed by direct counting of tissues after the final imaging. CONCLUSION: Because of the high and specific uptake of (64)Cu-labeled mAb 3/A12 in PSMA-positive tumors, this ligand represents an excellent candidate for prostate cancer imaging and potentially for radioimmunotherapy.
- 67Alt, K.; Wiehr, S.; Ehrlichmann, W.; Reischl, G.; Wolf, P.; Pichler, B. J.; Elsasser-Beile, U.; Buhler, P. High-resolution animal PET imaging of prostate cancer xenografts with three different 64Cu-labeled antibodies against native cell-adherent PSMA Prostate 2010, 70, 1413– 1421Google ScholarThere is no corresponding record for this reference.
- 68Fischer, G.; Seibold, U.; Schirrmacher, R.; Wangler, B.; Wangler, C. (89)Zr, a radiometal nuclide with high potential for molecular imaging with PET: chemistry, applications and remaining challenges Molecules 2013, 18, 6469– 6490Google ScholarThere is no corresponding record for this reference.
- 69Cheng, Y.; Prusoff, W. H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50% inhibition (I50) of an enzymatic reaction Biochem. Pharmacol. 1973, 22, 3099– 3108Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE2cXhtVGgs7c%253D&md5=fe7175ca300d8f4085df87af37f8656cRelation between the inhibition constant K1) and the concentration of inhibitor which causes fifty per cent inhibition (I50) of an enzymic reactionCheng, Yung-Chi; Prusoff, William H.Biochemical Pharmacology (1973), 22 (23), 3099-108CODEN: BCPCA6; ISSN:0006-2952.The Ki and I50 values are equal where the kinetics are non- or uncompetitive, but not where they are competitive. The relation between the 2 values was analyzed for non- and uncompetitive bi- and monosubstrate reactions and for competitive bisubstrate reactions.
Cited By
This article is cited by 93 publications.
- Ina Hierlmeier, Amaury Guillou, Daniel F. Earley, Anthony Linden, Jason P. Holland, Mark D. Bartholomä. HNODThia: A Promising Chelator for the Development of 64Cu Radiopharmaceuticals. Inorganic Chemistry 2023, Article ASAP.
- Alexander M. Brown, Jana L. Butman, Raphael Lengacher, Natasha P. Vargo, Kirsten E. Martin, Angus Koller, Dariusz Śmiłowicz, Eszter Boros, Jerome R. Robinson. N,N-Alkylation Clarifies the Role of N- and O-Protonated Intermediates in Cyclen-Based 64Cu Radiopharmaceuticals. Inorganic Chemistry 2023, 62
(4)
, 1362-1376. https://doi.org/10.1021/acs.inorgchem.2c02907
- Brett A. Vaughn, Alexander M. Brown, Shin Hye Ahn, Jerome R. Robinson, Eszter Boros. Is Less More? Influence of the Coordination Geometry of Copper(II) Picolinate Chelate Complexes on Metabolic Stability. Inorganic Chemistry 2020, 59
(22)
, 16095-16108. https://doi.org/10.1021/acs.inorgchem.0c02314
- James M. Kelly, Shashikanth Ponnala, Alejandro Amor-Coarasa, Nicholas A. Zia, Anastasia Nikolopoulou, Clarence Williams, Jr., David J. Schlyer, Stephen G. DiMagno, Paul S. Donnelly, John W. Babich. Preclinical Evaluation of a High-Affinity Sarcophagine-Containing PSMA Ligand for 64Cu/67Cu-Based Theranostics in Prostate Cancer. Molecular Pharmaceutics 2020, 17
(6)
, 1954-1962. https://doi.org/10.1021/acs.molpharmaceut.0c00060
- Wojciech
G. Lesniak, Srikanth Boinapally, Sangeeta Ray Banerjee, Babak Behnam Azad, Catherine A. Foss, Chentian Shen, Ala Lisok, Bryan Wharram, Sridhar Nimmagadda, Martin G. Pomper. Evaluation of PSMA-Targeted PAMAM Dendrimer Nanoparticles in a Murine Model of Prostate Cancer. Molecular Pharmaceutics 2019, 16
(6)
, 2590-2604. https://doi.org/10.1021/acs.molpharmaceut.9b00181
- Christoph
A. Umbricht, Martina Benešová, Roger Hasler, Roger Schibli, Nicholas P. van der Meulen, Cristina Müller. Design and Preclinical Evaluation of an Albumin-Binding PSMA Ligand for 64Cu-Based PET Imaging. Molecular Pharmaceutics 2018, 15
(12)
, 5556-5564. https://doi.org/10.1021/acs.molpharmaceut.8b00712
- Swarbhanu Sarkar, Nikunj Bhatt, Yeong Su Ha, Phuong Tu Huynh, Nisarg Soni, Woonghee Lee, Yong Jin Lee, Jung Young Kim, Darpan N. Pandya, Gwang Il An, Kyo Chul Lee, Yongmin Chang, and Jeongsoo Yoo . High in Vivo Stability of 64Cu-Labeled Cross-Bridged Chelators Is a Crucial Factor in Improved Tumor Imaging of RGD Peptide Conjugates. Journal of Medicinal Chemistry 2018, 61
(1)
, 385-395. https://doi.org/10.1021/acs.jmedchem.7b01671
- Azadeh Pirisedigh, Véronique Blais, Samia Ait-Mohand, Khaled Abdallah, Brian J. Holleran, Richard Leduc, Yves L. Dory, Louis Gendron, and Brigitte Guérin . Synthesis and Evaluation of a 64Cu-Conjugate, a Selective δ-Opioid Receptor Positron Emission Tomography Imaging Agent. Organic Letters 2017, 19
(8)
, 2018-2021. https://doi.org/10.1021/acs.orglett.7b00575
- Ananda Kumar Kanduluru, Madduri Srinivasarao, and Philip S. Low . Design, Synthesis, and Evaluation of a Neurokinin-1 Receptor-Targeted Near-IR Dye for Fluorescence-Guided Surgery of Neuroendocrine Cancers. Bioconjugate Chemistry 2016, 27
(9)
, 2157-2165. https://doi.org/10.1021/acs.bioconjchem.6b00374
- Mariane Le Fur, Maryline Beyler, Nicolas Lepareur, Olivier Fougère, Carlos Platas-Iglesias, Olivier Rousseaux, and Raphaël Tripier . Pyclen Tri-n-butylphosphonate Ester as Potential Chelator for Targeted Radiotherapy: From Yttrium(III) Complexation to 90Y Radiolabeling. Inorganic Chemistry 2016, 55
(16)
, 8003-8012. https://doi.org/10.1021/acs.inorgchem.6b01135
- Amit Kumar, Tara Mastren, Bin Wang, Jer-Tsong Hsieh, Guiyang Hao, and Xiankai Sun . Design of a Small-Molecule Drug Conjugate for Prostate Cancer Targeted Theranostics. Bioconjugate Chemistry 2016, 27
(7)
, 1681-1689. https://doi.org/10.1021/acs.bioconjchem.6b00222
- Sangeeta Ray Banerjee, Zhengping Chen, Mrudula Pullambhatla, Ala Lisok, Jian Chen, Ronnie C. Mease, and Martin G. Pomper . Preclinical Comparative Study of 68Ga-Labeled DOTA, NOTA, and HBED-CC Chelated Radiotracers for Targeting PSMA. Bioconjugate Chemistry 2016, 27
(6)
, 1447-1455. https://doi.org/10.1021/acs.bioconjchem.5b00679
- Lye Lin Lock, Claudia D. Reyes, Pengcheng Zhang, and Honggang Cui . Tuning Cellular Uptake of Molecular Probes by Rational Design of Their Assembly into Supramolecular Nanoprobes. Journal of the American Chemical Society 2016, 138
(10)
, 3533-3540. https://doi.org/10.1021/jacs.6b00073
- Xing Yang, Ronnie C. Mease, Mrudula Pullambhatla, Ala Lisok, Ying Chen, Catherine A. Foss, Yuchuan Wang, Hassan Shallal, Hannah Edelman, Adam T. Hoye, Giorgio Attardo, Sridhar Nimmagadda, and Martin G. Pomper . [18F]Fluorobenzoyllysinepentanedioic Acid Carbamates: New Scaffolds for Positron Emission Tomography (PET) Imaging of Prostate-Specific Membrane Antigen (PSMA). Journal of Medicinal Chemistry 2016, 59
(1)
, 206-218. https://doi.org/10.1021/acs.jmedchem.5b01268
- Adrian V. Fuchs, Brian W.C. Tse, Amanda K. Pearce, Mei-Chun Yeh, Nicholas L. Fletcher, Steve S. Huang, Warren D. Heston, Andrew K. Whittaker, Pamela J. Russell, and Kristofer J. Thurecht . Evaluation of Polymeric Nanomedicines Targeted to PSMA: Effect of Ligand on Targeting Efficiency. Biomacromolecules 2015, 16
(10)
, 3235-3247. https://doi.org/10.1021/acs.biomac.5b00913
- Mengting Li, Sheng Wang, Qunshou Kong, Xinyi Cheng, Huanhuan Yan, Yao Xing, Xiaoli Lan, Dawei Jiang. Advances in macrocyclic chelators for positron emission tomography imaging. VIEW 2023, 4
(5)
https://doi.org/10.1002/VIW.20230042
- Mohammed Abusalem, Lucia Martiniova, Sarita Soebianto, Louis DePalatis, Gregory Ravizzini. Current Status of Radiolabeled Monoclonal Antibodies Targeting PSMA for Imaging and Therapy. Cancers 2023, 15
(18)
, 4537. https://doi.org/10.3390/cancers15184537
- Inki Lee, Min Hwan Kim, Kyongkyu Lee, Keumrok Oh, Hyunwoo Lim, Jae Hun Ahn, Yong Jin Lee, Gi Jeong Cheon, Dae Yoon Chi, Sang Moo Lim. Comparison of the Effects of DOTA and NOTA Chelators on 64Cu-Cudotadipep and 64Cu-Cunotadipep for Prostate Cancer. Diagnostics 2023, 13
(16)
, 2649. https://doi.org/10.3390/diagnostics13162649
- Kirsten E. Martin, Joseph A. Mattocks, Dariusz Śmiłowicz, Eduardo Aluicio-Sarduy, Jennifer N. Whetter, Jonathan W. Engle, Joseph A. Cotruvo, Eszter Boros. Radiolabeling and
in vivo
evaluation of lanmodulin with biomedically relevant lanthanide isotopes. RSC Chemical Biology 2023, 4
(6)
, 414-421. https://doi.org/10.1039/D3CB00020F
- Runze Wang, Bernard Ponsard, Hubert Wolterbeek, Antonia Denkova. Core–shell structured gold nanoparticles as carrier for 166Dy/166Ho in vivo generator. EJNMMI Radiopharmacy and Chemistry 2022, 7
(1)
https://doi.org/10.1186/s41181-022-00170-3
- Norah AlHokbany, Ibrahim AlJammaz, Basem AlOtaibi, Yousif AlMalki, Bander AlJammaz, Subhani M. Okarvi. Development of new copper-64 labeled rhodamine: a potential PET myocardial perfusion imaging agent. EJNMMI Radiopharmacy and Chemistry 2022, 7
(1)
https://doi.org/10.1186/s41181-022-00171-2
- Sairan Eom, Min Hwan Kim, Ranji Yoo, Goeun Choi, Joo Hyun Kang, Yong Jin Lee, Jin-Ho Choy. Dilute lattice doping of
64
Cu into 2D-nanoplates: its impact on radio-labeling efficiency and stability for target selective PET imaging. Journal of Materials Chemistry B 2022, 10
(45)
, 9389-9399. https://doi.org/10.1039/D2TB01165D
- Sarah D. Busslinger, Viviane J. Tschan, Olivia K. Richard, Zeynep Talip, Roger Schibli, Cristina Müller. [225Ac]Ac-SibuDAB for Targeted Alpha Therapy of Prostate Cancer: Preclinical Evaluation and Comparison with [225Ac]Ac-PSMA-617. Cancers 2022, 14
(22)
, 5651. https://doi.org/10.3390/cancers14225651
- Beshara Sheehan, Christina Guo, Antje Neeb, Alec Paschalis, Shahneen Sandhu, Johann S. de Bono. Prostate-specific Membrane Antigen Biology in Lethal Prostate Cancer and its Therapeutic Implications. European Urology Focus 2022, 8
(5)
, 1157-1168. https://doi.org/10.1016/j.euf.2021.06.006
- Thomas M. Jeitner, John W. Babich, James M. Kelly. Advances in PSMA theranostics. Translational Oncology 2022, 22 , 101450. https://doi.org/10.1016/j.tranon.2022.101450
- Chul-Hee Lee, Ilhan Lim, Sang-Keun Woo, Kwang Il Kim, Kyo Chul Lee, Kanghyon Song, Chang Woon Choi, Sang Moo Lim. The Feasibility of
64
Cu-PSMA I&T PET for Prostate Cancer. Cancer Biotherapy and Radiopharmaceuticals 2022, 37
(6)
, 417-423. https://doi.org/10.1089/cbr.2020.4189
- Marie-Christine Milot, Ophélie Bélissant Benesty, Véronique Dumulon-Perreault, Samia Ait-Mohand, Patrick O. Richard, Étienne Rousseau, Brigitte Guérin. 64Cu-DOTHA2-PSMA, a Novel PSMA PET Radiotracer for Prostate Cancer with a Long Imaging Time Window. Pharmaceuticals 2022, 15
(8)
, 996. https://doi.org/10.3390/ph15080996
- David G. Calatayud, Sotia Neophytou, Eleni Nicodemou, S. Giuseppe Giuffrida, Haobo Ge, Sofia I. Pascu. Nano-Theranostics for the Sensing, Imaging and Therapy of Prostate Cancers. Frontiers in Chemistry 2022, 10 https://doi.org/10.3389/fchem.2022.830133
- Truc T. Huynh, Sreeja Sreekumar, Cedric Mpoy, Buck E. Rogers. A comparison of 64Cu-labeled bi-terminally PEGylated A20FMDV2 peptides targeting integrin ανβ6. Oncotarget 2022, 13
(1)
, 360-372. https://doi.org/10.18632/oncotarget.28197
- B. Liu, R. Han, C. Yuan, H. Sun, Z. Chen, G. Tian, F. Shi, X. Zhang, P. Luo, H. Jia. Excitation functions of proton induced reactions on titanium and copper. Applied Radiation and Isotopes 2021, 173 , 109713. https://doi.org/10.1016/j.apradiso.2021.109713
- Andrew Siow, Renata Kowalczyk, Margaret A. Brimble, Paul W.R. Harris. Evolution of Peptide-Based Prostate-Specific Membrane Antigen (PSMA) Inhibitors: An Approach to Novel Prostate Cancer Therapeutics. Current Medicinal Chemistry 2021, 28
(19)
, 3713-3752. https://doi.org/10.2174/0929867327666201006153847
- Lachlan E. McInnes, Carleen Cullinane, Peter D. Roselt, Susan Jackson, Benjamin J. Blyth, Ellen M. van Dam, Nicholas A. Zia, Matthew J. Harris, Rodney J. Hicks, Paul S. Donnelly. Therapeutic Efficacy of a Bivalent Inhibitor of Prostate-Specific Membrane Antigen Labeled with
67
Cu. Journal of Nuclear Medicine 2021, 62
(6)
, 829-832. https://doi.org/10.2967/jnumed.120.251579
- Zeynep Talip, Francesca Borgna, Cristina Müller, Jiri Ulrich, Charlotte Duchemin, Joao P. Ramos, Thierry Stora, Ulli Köster, Youcef Nedjadi, Vadim Gadelshin, Valentin N. Fedosseev, Frederic Juget, Claude Bailat, Adelheid Fankhauser, Shane G. Wilkins, Laura Lambert, Bruce Marsh, Dmitry Fedorov, Eric Chevallay, Pascal Fernier, Roger Schibli, Nicholas P. van der Meulen. Production of Mass-Separated Erbium-169 Towards the First Preclinical in vitro Investigations. Frontiers in Medicine 2021, 8 https://doi.org/10.3389/fmed.2021.643175
- Mária Bodnár Mikulová, Peter Mikuš. Advances in Development of Radiometal Labeled Amino Acid-Based Compounds for Cancer Imaging and Diagnostics. Pharmaceuticals 2021, 14
(2)
, 167. https://doi.org/10.3390/ph14020167
- Dhanusha Sabanathan, Maria E. Lund, Douglas H. Campbell, Bradley J. Walsh, Howard Gurney. Radioimmunotherapy for solid tumors: spotlight on Glypican-1 as a radioimmunotherapy target. Therapeutic Advances in Medical Oncology 2021, 13 , 175883592110229. https://doi.org/10.1177/17588359211022918
- Nikhil Mayor, Niranjan J. Sathianathen, James Buteau, Samantha Koschel, Marta Antón Juanilla, Jada Kapoor, Arun Azad, Michael S. Hofman, Declan G. Murphy. Prostate‐specific membrane antigen theranostics in advanced prostate cancer: an evolving option. BJU International 2020, 126
(5)
, 525-535. https://doi.org/10.1111/bju.15143
- Nicholas P. van der Meulen, Roger Hasler, Zeynep Talip, Pascal V. Grundler, Chiara Favaretto, Christoph A. Umbricht, Cristina Müller, Gaia Dellepiane, Tommaso S. Carzaniga, Saverio Braccini. Developments toward the Implementation of 44Sc Production at a Medical Cyclotron. Molecules 2020, 25
(20)
, 4706. https://doi.org/10.3390/molecules25204706
- Lukas Greifenstein, Denise Späth, Jean Phillip Sinnes, Tilmann Grus, Frank Rösch. Mild and efficient
64
Cu labeling of perhydro-1, 4-diazepine derivatives for potential use with large peptides, proteins and antibodies. Radiochimica Acta 2020, 108
(7)
, 555-563. https://doi.org/10.1515/ract-2019-3167
- Mathieu L. Lepage, Hsiou‐Ting Kuo, Áron Roxin, Sungjoon Huh, Zhengxing Zhang, Rajaguru Kandasamy, Helen Merkens, Joel O. Kumlin, Alan Limoges, Stefan K. Zeisler, Kuo‐Shyan Lin, François Bénard, David M. Perrin. Toward
18
F‐Labeled Theranostics: A Single Agent that Can Be Labeled with
18
F,
64
Cu, or
177
Lu. ChemBioChem 2020, 21
(7)
, 943-947. https://doi.org/10.1002/cbic.201900632
- Thomas L. Andersen, Christina Baun, Birgitte B. Olsen, Johan H. Dam, Helge Thisgaard. Improving Contrast and Detectability: Imaging with [
55
Co]Co-DOTATATE in Comparison with [
64
Cu]Cu-DOTATATE and [
68
Ga]Ga-DOTATATE. Journal of Nuclear Medicine 2020, 61
(2)
, 228-233. https://doi.org/10.2967/jnumed.119.233015
- Brett A. Vaughn, Shin Hye Ahn, Eduardo Aluicio-Sarduy, Justin Devaraj, Aeli P. Olson, Jonathan Engle, Eszter Boros. Chelation with a twist: a bifunctional chelator to enable room temperature radiolabeling and targeted PET imaging with scandium-44. Chemical Science 2020, 11
(2)
, 333-342. https://doi.org/10.1039/C9SC04655K
- José Carlos dos Santos, Barbro Beijer, Ulrike Bauder-Wüst, Martin Schäfer, Karin Leotta, Matthias Eder, Martina Benešová, Christian Kleist, Frederik Giesel, Clemens Kratochwil, Klaus Kopka, Uwe Haberkorn, Walter Mier. Development of Novel PSMA Ligands for Imaging and Therapy with Copper Isotopes. Journal of Nuclear Medicine 2020, 61
(1)
, 70-79. https://doi.org/10.2967/jnumed.119.229054
- Nadeem Ahmed Lodhi, Ji Yong Park, Kyuwan Kim, Mi Kyung Hong, Young Joo Kim, Yun-Sang Lee, Gi Jeong Cheon, Keon Wook Kang, Jae Min Jeong. Synthesis and Evaluation of 99mTc-Tricabonyl Labeled Isonitrile Conjugates for Prostate-Specific Membrane Antigen (PSMA) Image. Inorganics 2020, 8
(1)
, 5. https://doi.org/10.3390/inorganics8010005
- Christoph A. Umbricht, Ulli Köster, Peter Bernhardt, Nadezda Gracheva, Karl Johnston, Roger Schibli, Nicholas P. van der Meulen, Cristina Müller. Alpha-PET for Prostate Cancer: Preclinical investigation using 149Tb-PSMA-617. Scientific Reports 2019, 9
(1)
https://doi.org/10.1038/s41598-019-54150-w
- Sangeeta Ray Banerjee, Vivek Kumar, Ala Lisok, Jian Chen, Il Minn, Mary Brummet, Srikanth Boinapally, Michael Cole, Ethel Ngen, Bryan Wharram, Cory Brayton, Robert F. Hobbs, Martin G. Pomper. 177Lu-labeled low-molecular-weight agents for PSMA-targeted radiopharmaceutical therapy. European Journal of Nuclear Medicine and Molecular Imaging 2019, 46
(12)
, 2545-2557. https://doi.org/10.1007/s00259-019-04434-0
- Xiaoxia Zhu, Xiaye Miao, Xinyue Qin, Xiaohong Zhu. Design of immunogens: The effect of bifunctional chelator on immunological response to chelated copper. Journal of Pharmaceutical and Biomedical Analysis 2019, 174 , 263-269. https://doi.org/10.1016/j.jpba.2019.06.001
- Niels J. Rupp, Christoph A. Umbricht, Daniele A. Pizzuto, Daniela Lenggenhager, Antonia Töpfer, Julian Müller, Urs J. Muehlematter, Daniela A. Ferraro, Michael Messerli, Grégoire B. Morand, Gerhard F. Huber, Daniel Eberli, Roger Schibli, Cristina Müller, Irene A. Burger. First Clinicopathologic Evidence of a Non–PSMA-Related Uptake Mechanism for
68
Ga-PSMA-11 in Salivary Glands. Journal of Nuclear Medicine 2019, 60
(9)
, 1270-1276. https://doi.org/10.2967/jnumed.118.222307
- Sagnik Sengupta, Mena Asha Krishnan, Sudeshna Chattopadhyay, Venkatesh Chelvam. Comparison of prostate‐specific membrane antigen ligands in clinical translation research for diagnosis of prostate cancer. Cancer Reports 2019, 2
(4)
https://doi.org/10.1002/cnr2.1169
- Clemens Kratochwil, Uwe Haberkorn, Frederik L. Giesel. Radionuclide Therapy of Metastatic Prostate Cancer. Seminars in Nuclear Medicine 2019, 49
(4)
, 313-325. https://doi.org/10.1053/j.semnuclmed.2019.02.003
- Hans-Jürgen Wester, Margret Schottelius. PSMA-Targeted Radiopharmaceuticals for Imaging and Therapy. Seminars in Nuclear Medicine 2019, 49
(4)
, 302-312. https://doi.org/10.1053/j.semnuclmed.2019.02.008
- Sebastian Hoberück, Gerd Wunderlich, Enrico Michler, Tobias Hölscher, Martin Walther, Danilo Seppelt, Ivan Platzek, Klaus Zöphel, Jörg Kotzerke. Dual‐time‐point
64
Cu‐PSMA
‐617‐
PET/CT
in patients suffering from prostate cancer. Journal of Labelled Compounds and Radiopharmaceuticals 2019, 62
(8)
, 523-532. https://doi.org/10.1002/jlcr.3745
- Nadine Leygue, Morgane Enel, Abdel Diallo, Béatrice Mestre‐Voegtlé, Chantal Galaup, Claude Picard. Efficient Synthesis of a Family of Bifunctional Chelators Based on the PCTA[12] Macrocycle Suitable for Bioconjugation. European Journal of Organic Chemistry 2019, 2019
(18)
, 2899-2913. https://doi.org/10.1002/ejoc.201900280
- Till Wüstemann, Uwe Haberkorn, John Babich, Walter Mier. Targeting prostate cancer: Prostate‐specific membrane antigen based diagnosis and therapy. Medicinal Research Reviews 2019, 39
(1)
, 40-69. https://doi.org/10.1002/med.21508
- Yeye Zhou, Jihui Li, Xin Xu, Man Zhao, Bin Zhang, Shengming Deng, Yiwei Wu. 64
Cu-based Radiopharmaceuticals in Molecular Imaging. Technology in Cancer Research & Treatment 2019, 18 , 153303381983075. https://doi.org/10.1177/1533033819830758
- Amandine Conte‐Daban, Maryline Beyler, Raphaël Tripier, Christelle Hureau. Kinetics Are Crucial When Targeting Copper Ions to Fight Alzheimer's Disease: An Illustration with Azamacrocyclic Ligands. Chemistry – A European Journal 2018, 24
(33)
, 8447-8452. https://doi.org/10.1002/chem.201801520
- Tilman Läppchen, Yvonne Kiefer, Jason P. Holland, Mark D. Bartholomä. In vitro and in vivo evaluation of the bifunctional chelator NODIA-Me in combination with a prostate-specific membrane antigen targeting vector. Nuclear Medicine and Biology 2018, 60 , 45-54. https://doi.org/10.1016/j.nucmedbio.2018.03.002
- Zhihao Zha, Karl Ploessl, Seok Rye Choi, Zehui Wu, Lin Zhu, Hank F. Kung. Synthesis and evaluation of a novel urea-based 68 Ga-complex for imaging PSMA binding in tumor. Nuclear Medicine and Biology 2018, 59 , 36-47. https://doi.org/10.1016/j.nucmedbio.2017.12.007
- Shankar Vallabhajosula, Berna D. Polack, John W. Babich. Molecular Imaging of Prostate Cancer: Radiopharmaceuticals for Positron Emission Tomography (PET) and Single-Photon Emission Computed Tomography (SPECT). 2018, 475-501. https://doi.org/10.1007/978-3-319-64096-9_27
- Christoph A. Umbricht, Martina Benešová, Raffaella M. Schmid, Andreas Türler, Roger Schibli, Nicholas P. van der Meulen, Cristina Müller. 44Sc-PSMA-617 for radiotheragnostics in tandem with 177Lu-PSMA-617—preclinical investigations in comparison with 68Ga-PSMA-11 and 68Ga-PSMA-617. EJNMMI Research 2017, 7
(1)
https://doi.org/10.1186/s13550-017-0257-4
- Sara Sheikhbahaei, Ali Afshar-Oromieh, Matthias Eiber, Lilja B. Solnes, Mehrbod S. Javadi, Ashley E. Ross, Kenneth J. Pienta, Mohamad E. Allaf, Uwe Haberkorn, Martin G. Pomper, Michael A. Gorin, Steven P. Rowe. Pearls and pitfalls in clinical interpretation of prostate-specific membrane antigen (PSMA)-targeted PET imaging. European Journal of Nuclear Medicine and Molecular Imaging 2017, 44
(12)
, 2117-2136. https://doi.org/10.1007/s00259-017-3780-7
- Lingzhou Zhao, Xiangyang Shi, Jinhua Zhao. Dendrimer-based contrast agents for PET imaging. Drug Delivery 2017, 24
(2)
, 81-93. https://doi.org/10.1080/10717544.2017.1399299
- Tadahiro Kin, Takaya Kawagoe, Shouhei Araki, Yukinobu Watanabe. Production of high-purity medical radio isotope
64
Cu with accelerator-based neutrons generated with 9 and 12 MeV deuterons. Journal of Nuclear Science and Technology 2017, 54
(10)
, 1123-1130. https://doi.org/10.1080/00223131.2017.1344585
- Aimen Zlitni, Melissa Yin, Nancy Janzen, Samit Chatterjee, Ala Lisok, Kathleen L. Gabrielson, Sridhar Nimmagadda, Martin G. Pomper, F. Stuart Foster, John F. Valliant, . Development of prostate specific membrane antigen targeted ultrasound microbubbles using bioorthogonal chemistry. PLOS ONE 2017, 12
(5)
, e0176958. https://doi.org/10.1371/journal.pone.0176958
- Xiaoping Xu, Jianping Zhang, Silong Hu, Simin He, Xiao Bao, Guang Ma, Jianmin Luo, Jingyi Cheng, Yingjian Zhang. 99m Tc-labeling and evaluation of a HYNIC modified small-molecular inhibitor of prostate-specific membrane antigen. Nuclear Medicine and Biology 2017, 48 , 69-75. https://doi.org/10.1016/j.nucmedbio.2017.01.010
- Aviral Singh, Harshad R. Kulkarni, Richard P. Baum. Imaging of Prostate Cancer Using 64 Cu-Labeled Prostate-Specific Membrane Antigen Ligand. PET Clinics 2017, 12
(2)
, 193-203. https://doi.org/10.1016/j.cpet.2016.12.001
- Eleni Gourni, Gjermund Henriksen. Metal-Based PSMA Radioligands. Molecules 2017, 22
(4)
, 523. https://doi.org/10.3390/molecules22040523
- Uwe Haberkorn, Matthias Eder, Klaus Kopka, John W. Babich, Michael Eisenhut. Role of Radiolabelled Small Molecules Binding to PSMA in Diagnosis and Therapy of Prostate Cancer. 2017, 51-58. https://doi.org/10.1007/978-3-319-57624-4_6
- Gerhard Glatting, Carmen Wängler, Björn Wängler. Physikalisch-technische Grundlagen und Tracerentwicklung in der Positronenemissionstomografie. 2017, 19-56. https://doi.org/10.1007/978-3-662-50468-0_2
- Mariane Le Fur, Maryline Beyler, Enikő Molnár, Olivier Fougère, David Esteban-Gómez, Gyula Tircsó, Carlos Platas-Iglesias, Nicolas Lepareur, Olivier Rousseaux, Raphaël Tripier. The role of the capping bond effect on pyclen
nat
Y
3+
/
90
Y
3+
chelates: full control of the regiospecific N-functionalization makes the difference. Chemical Communications 2017, 53
(69)
, 9534-9537. https://doi.org/10.1039/C7CC05088G
- Eleni Gourni, Luigi Del Pozzo, Mark Bartholomä, Yvonne Kiefer, Philipp T. Meyer, Helmut R. Maecke, Jason P. Holland. Radiochemistry and Preclinical PET Imaging of
68
Ga-Desferrioxamine Radiotracers Targeting Prostate-Specific Membrane Antigen. Molecular Imaging 2017, 16 , 153601211773701. https://doi.org/10.1177/1536012117737010
- Andreas Poschenrieder, Margret Schottelius, Markus Schwaiger, Horst Kessler, Hans-Jürgen Wester. The influence of different metal-chelate conjugates of pentixafor on the CXCR4 affinity. EJNMMI Research 2016, 6
(1)
https://doi.org/10.1186/s13550-016-0193-8
- James C Evans, Meenakshi Malhotra, John F Cryan, Caitriona M O'Driscoll. The therapeutic and diagnostic potential of the prostate specific membrane antigen/glutamate carboxypeptidase II (PSMA/GCPII) in cancer and neurological disease. British Journal of Pharmacology 2016, 173
(21)
, 3041-3079. https://doi.org/10.1111/bph.13576
- Ali Afshar-Oromieh, John W. Babich, Clemens Kratochwil, Frederik L. Giesel, Michael Eisenhut, Klaus Kopka, Uwe Haberkorn. The Rise of PSMA Ligands for Diagnosis and Therapy of Prostate Cancer. Journal of Nuclear Medicine 2016, 57
(Supplement 3)
, 79S-89S. https://doi.org/10.2967/jnumed.115.170720
- Eric Wehrenberg-Klee, N. Selcan Turker, Pedram Heidari, Benjamin Larimer, Dejan Juric, José Baselga, Maurizio Scaltriti, Umar Mahmood. Differential Receptor Tyrosine Kinase PET Imaging for Therapeutic Guidance. Journal of Nuclear Medicine 2016, 57
(9)
, 1413-1419. https://doi.org/10.2967/jnumed.115.169417
- Il Minn, Soo Min Koo, Hye Soo Lee, Mary Brummet, Steven P. Rowe, Michael A. Gorin, Polina Sysa-Shah, William D. Lewis, Hye-Hyun Ahn, Yuchuan Wang, Sangeeta Ray Banerjee, Ronnie C. Mease, Sridhar Nimmagadda, Mohamad E. Allaf, Martin G. Pomper, Xing Yang. [64Cu]XYIMSR-06: A dual-motif CAIX ligand for PET imaging of clear cell renal cell carcinoma. Oncotarget 2016, 7
(35)
, 56471-56479. https://doi.org/10.18632/oncotarget.10602
- Ali Afshar-Oromieh, Henrik Hetzheim, Wolfgang Kübler, Clemens Kratochwil, Frederik L. Giesel, Thomas A. Hope, Matthias Eder, Michael Eisenhut, Klaus Kopka, Uwe Haberkorn. Radiation dosimetry of 68Ga-PSMA-11 (HBED-CC) and preliminary evaluation of optimal imaging timing. European Journal of Nuclear Medicine and Molecular Imaging 2016, 43
(9)
, 1611-1620. https://doi.org/10.1007/s00259-016-3419-0
- Jessie R. Nedrow, Joseph D. Latoche, Kathryn E. Day, Jalpa Modi, Tanushree Ganguly, Dexing Zeng, Brenda F. Kurland, Clifford E. Berkman, Carolyn J. Anderson. Targeting PSMA with a Cu-64 Labeled Phosphoramidate Inhibitor for PET/CT Imaging of Variant PSMA-Expressing Xenografts in Mouse Models of Prostate Cancer. Molecular Imaging and Biology 2016, 18
(3)
, 402-410. https://doi.org/10.1007/s11307-015-0908-7
- Drishty Satpati, Ajit Shinto, K. K. Kamaleshwaran, Surekha Sane, Sharmila Banerjee. Convenient Preparation of [68Ga]DKFZ-PSMA-11 Using a Robust Single-Vial Kit and Demonstration of Its Clinical Efficacy. Molecular Imaging and Biology 2016, 18
(3)
, 420-427. https://doi.org/10.1007/s11307-016-0943-z
- Vikas Prasad, Ingo G. Steffen, Gerd Diederichs, Marcus R. Makowski, Peter Wust, Winfried Brenner. Biodistribution of [68Ga]PSMA-HBED-CC in Patients with Prostate Cancer: Characterization of Uptake in Normal Organs and Tumour Lesions. Molecular Imaging and Biology 2016, 18
(3)
, 428-436. https://doi.org/10.1007/s11307-016-0945-x
- Zhihang Chen, Marie-France Penet, Balaji Krishnamachary, Sangeeta R. Banerjee, Martin G. Pomper, Zaver M. Bhujwalla. PSMA-specific theranostic nanoplex for combination of TRAIL gene and 5-FC prodrug therapy of prostate cancer. Biomaterials 2016, 80 , 57-67. https://doi.org/10.1016/j.biomaterials.2015.11.048
- Laura K. van Dijk, Cheng‐Bin Yim, Gerben M. Franssen, Johannes H. A. M. Kaanders, Johan Rajander, Olof Solin, Tove J. Grönroos, Otto C. Boerman, Johan Bussink. PET of EGFR with
64
Cu‐cetuximab‐F(ab′)
2
in mice with head and neck squamous cell carcinoma xenografts. Contrast Media & Molecular Imaging 2016, 11
(1)
, 65-70. https://doi.org/10.1002/cmmi.1659
- Fan Pu, Mani Salarian, Shenghui Xue, Jingjuan Qiao, Jie Feng, Shanshan Tan, Anvi Patel, Xin Li, Kenza Mamouni, Khan Hekmatyar, Juan Zou, Daqing Wu, Jenny J. Yang. Prostate-specific membrane antigen targeted protein contrast agents for molecular imaging of prostate cancer by MRI. Nanoscale 2016, 8
(25)
, 12668-12682. https://doi.org/10.1039/C5NR09071G
- Uwe Haberkorn, Matthias Eder, Klaus Kopka, John W. Babich, Michael Eisenhut. New Strategies in Prostate Cancer: Prostate-Specific Membrane Antigen (PSMA) Ligands for Diagnosis and Therapy. Clinical Cancer Research 2016, 22
(1)
, 9-15. https://doi.org/10.1158/1078-0432.CCR-15-0820
- Eleni Gourni, Coline Canovas, Victor Goncalves, Franck Denat, Philipp T. Meyer, Helmut R. Maecke, . (R)-NODAGA-PSMA: A Versatile Precursor for Radiometal Labeling and Nuclear Imaging of PSMA-Positive Tumors. PLOS ONE 2015, 10
(12)
, e0145755. https://doi.org/10.1371/journal.pone.0145755
- Shaik Mohammad Naushad, Parvathaneni Shree Divyya, M. Janaki Ramaiah, Balraj Alex Stanley, S. Prasanna Lakshmi, J. Vishnupriya, Vijay Kumar Kutala. Clinical utility of genetic variants of glutamate carboxypeptidase II in predicting breast cancer and prostate cancer risk. Cancer Genetics 2015, 208
(11)
, 552-558. https://doi.org/10.1016/j.cancergen.2015.09.001
- Ali Afshar-Oromieh, Henrik Hetzheim, Clemens Kratochwil, Martina Benesova, Matthias Eder, Oliver C. Neels, Michael Eisenhut, Wolfgang Kübler, Tim Holland-Letz, Frederik L. Giesel, Walter Mier, Klaus Kopka, Uwe Haberkorn. The Theranostic PSMA Ligand PSMA-617 in the Diagnosis of Prostate Cancer by PET/CT: Biodistribution in Humans, Radiation Dosimetry, and First Evaluation of Tumor Lesions. Journal of Nuclear Medicine 2015, 56
(11)
, 1697-1705. https://doi.org/10.2967/jnumed.115.161299
- Zhengtao Qin. Recent advances of injectable radiopharmaceuticals for nuclear imaging and therapy: a new era in nuclear medicine. Materials Technology 2015, 30
(sup8)
, B250-B255. https://doi.org/10.1080/10667857.2015.1104829
- Punit Sharma, Rakesh Kumar, Abass Alavi. PET/Computed Tomography Using New Radiopharmaceuticals in Targeted Therapy. PET Clinics 2015, 10
(4)
, 495-505. https://doi.org/10.1016/j.cpet.2015.05.007
- Shanna Litau, Uwe Seibold, Alicia Vall‐Sagarra, Gert Fricker, Björn Wängler, Carmen Wängler. Comparative Assessment of Complex Stabilities of Radiocopper Chelating Agents by a Combination of Complex Challenge and in vivo Experiments. ChemMedChem 2015, 10
(7)
, 1200-1208. https://doi.org/10.1002/cmdc.201500132
- T. Maurer, H. Kübler, J.E. Gschwend, M. Eiber. Positronenemissionstomographie bei urologischen Tumorerkrankungen. Der Urologe 2015, 54
(7)
, 983-991. https://doi.org/10.1007/s00120-015-3868-2
- Ewen Bodio, Mohammed Boujtita, Karine Julienne, Patricia Le Saec, Sébastien G. Gouin, Jonathan Hamon, Eric Renault, David Deniaud. Synthesis and Characterization of a Stable Copper(I) Complex for Radiopharmaceutical Applications. ChemPlusChem 2014, 79
(9)
, 1284-1293. https://doi.org/10.1002/cplu.201402031
- Amanda K. Pearce, Barbara E. Rolfe, Pamela J. Russell, Brian W.-C. Tse, Andrew K. Whittaker, Adrian V. Fuchs, Kristofer J. Thurecht. Development of a polymer theranostic for prostate cancer. Polym. Chem. 2014, 5
(24)
, 6932-6942. https://doi.org/10.1039/C4PY00999A
- Robin C. Cumming, Dag Erlend Olberg, Julie L. Sutcliffe. Rapid
18
F-radiolabeling of peptides from [
18
F]fluoride using a single microfluidics device. RSC Adv. 2014, 4
(90)
, 49529-49534. https://doi.org/10.1039/C4RA10520F
Abstract
Figure 1
Figure 1. Proposed structures of 64Cu-labeled inhibitors of PSMA.
Scheme 1
Scheme 1. Synthesis of [64Cu]3–5Scheme 2
Scheme 2. Synthesis of [64Cu]6Figure 2
Figure 2. Whole body PET-CT imaging of PSMA+ PC3 PIP and PSMA – PC3 flu tumor-bearing mice with compounds [64Cu]3, [64Cu]4, [64Cu]5, [64Cu]6B, and [64Cu]7 at 2.5 h postinjection, respectively. Mice were injected with ∼11.1 MBq (∼300 μCi) of radiotracer IV. PIP = PSMA+ PC3 PIP (solid arrow); flu = PSMA– PC3 flu (unfilled arrow); K= kidney; B = bladder; L = left; R = right. All images are decay-corrected and adjusted to the same maximum value.
Figure 3
Figure 3. Whole body PET-CT imaging of PC3 PIP and PC3 flu tumor bearing mice with [64Cu]3 at 20 min (left), 6 h (middle), 28 h (right). Abdominal radioactivity is primarily due to uptake within kidneys and bladder. PIP = PC3 PSMA+ PIP (solid arrow); flu = PC3 PSMA– flu (unfilled arrow); K= kidney; L = left; R = right, B = bladder. All images are decay-corrected and adjusted to the same maximum value.
Figure 4
Figure 4. Whole body PET-CT imaging of PC3 PIP and PC3 flu tumor bearing mice with [64Cu]6A (top row) and [64Cu]6B (bottom row) at 20 min, 2.5 h, 12 and 22 h postinjection. Abdominal radioactivity is primarily due to uptake within kidneys and bladder. PIP = PC3 PSMA+ PIP (solid arrow); flu = PC3 PSMA− flu (unfilled arrow); K= kidney; L = left; R = right, B = bladder. All images are decay-corrected and adjusted to the same maximum value.
References
ARTICLE SECTIONSThis article references 69 other publications.
- 1Ghosh, A.; Heston, W. D. Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancer J. Cell Biochem. 2004, 91, 528– 539Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhsVWjs70%253D&md5=d5b2b495ac34449feb6a33e85b3de264Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancerGhosh, Arundhati; Heston, Warren D. W.Journal of Cellular Biochemistry (2004), 91 (3), 528-539CODEN: JCEBD5; ISSN:0730-2312. (Wiley-Liss, Inc.)A review. Prostate specific membrane antigen (PSMA), is a unique membrane bound glycoprotein, which is overexpressed manifold on prostate cancer as well as neovasculature of most of the solid tumors, but not in the vasculature of the normal tissues. This unique expression of PSMA makes it an important marker as well as a large extracellular target of imaging agents. PSMA can serve as target for delivery of therapeutic agents such as cytotoxins or radionuclides. PSMA has two unique enzymic functions, folate hydrolase and NAALADase and found to be recycled like other membrane bound receptors through clathrin coated pits. The internalization property of PSMA leads one to consider the potential existence of a natural ligand for PSMA. In this review we have discussed the regulation of PSMA expression within the cells, and significance of its expression in prostate cancer and metastasis.
- 2Rajasekaran, A. K.; Anilkumar, G.; Christiansen, J. J. Is prostate-specific membrane antigen a multifunctional protein? Am. J. Physiol., Cell Physiol. 2005, 288, C975– C981Google ScholarThere is no corresponding record for this reference.
- 3Chang, S. S.; Reuter, V. E.; Heston, W. D.; Gaudin, P. B. Comparison of anti-prostate-specific membrane antigen antibodies and other immunomarkers in metastatic prostate carcinoma Urology 2001, 57, 1179– 1183Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD387hslOrtg%253D%253D&md5=d07a48bed4ec39043f399679688bef1fComparison of anti-prostate-specific membrane antigen antibodies and other immunomarkers in metastatic prostate carcinomaChang S S; Reuter V E; Heston W D; Gaudin P BUrology (2001), 57 (6), 1179-83 ISSN:.OBJECTIVES: To compare the immunohistochemical properties of the 7E11 anti-prostate-specific membrane antigen (anti-PSMA) monoclonal antibody (mAb) with the recently developed anti-PSMA mAb, PM2J004.5, and with other common immunomarkers in metastatic prostate cancer. PSMA is a type II integral membrane glycoprotein highly expressed in prostate cancer cells. The mAb 7E11 is currently used in the radioisotopic evaluation of prostate cancer, and its immunohistochemical properties have been examined in primary prostate cancer specimens. METHODS: We examined 23 formalin-fixed, paraffin-embedded, metastatic prostate carcinoma specimens from various anatomic sites, including bone, lymph node, liver, lung, and soft tissue. Using the biotin-streptavidin method, we performed immunohistochemical reactions with the anti-PSMA mAbs 7E11 and PM2J004.5 and with antibodies to prostate-specific antigen and prostatic acid phosphatase. The immunoreactions were scored by pathologists unaware of the clinical and pathologic data according to a staining intensity scale and the percentage of cells stained. RESULTS: All four mAbs consistently stained the metastatic prostate cancer specimens. In 2 (8.7%) of 23 cases, however, the prostate-specific antigen immunoreaction was negative but the anti-PSMA mAbs had positive staining. Although 7E11 and PM2J004.5 had a similar staining intensity and percentage of cells stained for most specimens, in 3 (13%) of 23 specimens, 7E11 had less intense staining. None of the specimens were negative for all four antibodies. CONCLUSIONS: Anti-PSMA mAbs consistently immunoreacted with metastatic prostate cancer specimens and were positive in instances when prostate-specific antigen staining was negative. The anti-PSMA mAbs demonstrated similar staining patterns; however, in select cases, the PM2J004.5 mAb did show more intense staining. The anti-PSMA mAbs 7E11 and PM2J004.5 are useful in the pathologic evaluation of paraffin-embedded metastatic prostate cancer specimens.
- 4Wright, G. L., Jr.; Grob, B. M.; Haley, C.; Grossman, K.; Newhall, K.; Petrylak, D.; Troyer, J.; Konchuba, A.; Schellhammer, P. F.; Moriarty, R. Upregulation of prostate-specific membrane antigen after androgen-deprivation therapy Urology 1996, 48, 326– 334Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK28zlt1OgsQ%253D%253D&md5=0bc36d48df2fd48360407a9f33267d3bUpregulation of prostate-specific membrane antigen after androgen-deprivation therapyWright G L Jr; Grob B M; Haley C; Grossman K; Newhall K; Petrylak D; Troyer J; Konchuba A; Schellhammer P F; Moriarty RUrology (1996), 48 (2), 326-34 ISSN:0090-4295.OBJECTIVES: To determine the expression of prostate-specific membrane antigen (PSMA) before and after androgen-deprivation therapy and to compare PSMA expression with prostate-specific antigen (PSA) expression. METHODS: We studied specimens from 20 patients with prostate cancer undergoing medical or surgical castration or combination androgen-deprivation therapy in whom matched pretreatment and post-treatment tissue specimens were available and 16 patients in whom only a post-treatment specimen was available. The expression of PSMA and PSA in the tissue specimens was determined by immunoperoxidase staining. The extent of staining was calculated by multiplying the percent of antigen-positive tumor cells by the staining intensity to arrive at a stain index for each biomarker. An in vitro study assessed the concentration of PSMA and PSA in extracts of LNCaP cells cultured in the presence or absence of androgen as determined by immunoassays and Western blot analysis. RESULTS: PSMA reactivity was found to be increased in 55% (11 of 20) of post-treatment primary tissues and 100% (4 of 4) of post-treatment metastatic specimens. In contrast, PSA expression was found to be decreased in 70% (14 of 20) of post-treatment primary and 100% (4 of 4) of post-treatment metastatic specimens. Neither type of androgen-deprivation treatment nor tissue sensitivity to androgen deprivation appeared to influence degree of biomarker expression. PSMA was found to be downregulated and PSA upregulated when LNCaP cells were cultured in the presence of testosterone or dihydrotestosterone. CONCLUSIONS: The enhanced expression of PSMA in tissues and LNCaP cells after androgen deprivation suggests that PSMA is upregulated in the majority of prostate carcinomas after androgen treatment. The high expression in metastatic tissues strongly suggests that PSMA may be a clinically useful target for antibody-and genetic-directed therapy of prostate cancer that recurs after androgen deprivation. The mechanism whereby androgens suppress the expression of PSMA, and the association of PSMA with the development of hormone-independent prostate cancers, will require further study.
- 5Evans, M. J.; Smith-Jones, P. M.; Wongvipat, J.; Navarro, V.; Kim, S.; Bander, N. H.; Larson, S. M.; Sawyers, C. L. Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 9578– 9582Google ScholarThere is no corresponding record for this reference.
- 6Perner, S.; Hofer, M. D.; Kim, R.; Shah, R. B.; Li, H.; Moller, P.; Hautmann, R. E.; Gschwend, J. E.; Kuefer, R.; Rubin, M. A. Prostate-specific membrane antigen expression as a predictor of prostate cancer progression Human Pathol. 2007, 38, 696– 701Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXkt1ajtLY%253D&md5=d26f9f8635836a05ab6bab48a802893dProstate-specific membrane antigen expression as a predictor of prostate cancer progressionPerner, Sven; Hofer, Matthias D.; Kim, Robert; Shah, Rajal B.; Li, Haojie; Moeller, Peter; Hautmann, Richard E.; Gschwend, Juergen E.; Kuefer, Rainer; Rubin, Mark A.Human Pathology (2007), 38 (5), 696-701CODEN: HPCQA4; ISSN:0046-8177. (Elsevier Inc.)Distinguishing aggressive prostate cancer from indolent disease represents an important clin. challenge, because current therapy may lead to overtreatment of men with limited disease. The prostate-specific membrane antigen (PSMA) is a membrane-bound glycoprotein that is highly restricted to the prostate. Previously, studies analyzing the expression of PSMA have found an up-regulation in correlation with prostate cancer, particularly in advanced cancer. This assocn. is ideal for an application as a prognostic marker. In the current study, we characterized PSMA expression in a high-risk cohort and evaluated its potential use as predictive marker of prostate-specific antigen (PSA) recurrence. PSMA expression was analyzed by immunohistochem. using tissue microarrays composed of tumor samples from 450 patients. Protein intensity was recorded using a semiautomated quant. microscope system (ACIS II; Clarient Chromavision Medical Systems, San Juan Capistrano, CA). PSMA expression levels differed significantly (P < .001) between benign prostatic tissue, localized prostate cancer, and lymph node metastases. Dividing the cohort into high- and low-PSMA expressing cancers based on the median area of pos. staining, we found that high PSMA levels were assocd. with significant increase of PSA recurrence (P = .004). This was independent of clin. parameters such as lymph node tumor burden (lymph node d., >20%; P < .001), extraprostatic extension (P = .017), seminal vesicle invasion (P < .001), and high Gleason score (8-10, P = .006). In a multivariate model, PSMA expression and metastases to pelvic lymph nodes were significantly assocd. with time to PSA recurrence (HR, 1.4; 95% confidence interval, 1.1-2.8, P = .017; and hazard ratio, 5; 95% confidence interval, 2.6-9.7, P < .001, resp.). In summary, PSMA is independently assocd. with PSA recurrence in a high-risk cohort and thus might provide insight into the addnl. use of adjuvant therapy. Validation on other cohorts is required.
- 7Sanna, V.; Pintus, G.; Roggio, A. M.; Punzoni, S.; Posadino, A. M.; Arca, A.; Marceddu, S.; Bandiera, P.; Uzzau, S.; Sechi, M. Targeted biocompatible nanoparticles for the delivery of (−)-epigallocatechin 3-gallate to prostate cancer cells J. Med. Chem. 2011, 54, 1321– 1332Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhslyiu7k%253D&md5=8a103de00d04093c1d00f9a8b24732faTargeted Biocompatible Nanoparticles for the Delivery of (-)-Epigallocatechin 3-Gallate to Prostate Cancer CellsSanna, Vanna; Pintus, Gianfranco; Roggio, Anna Maria; Punzoni, Stefania; Posadino, Anna Maria; Arca, Alessandro; Marceddu, Salvatore; Bandiera, Pasquale; Uzzau, Sergio; Sechi, MarioJournal of Medicinal Chemistry (2011), 54 (5), 1321-1332CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mol. targeted cancer therapy mediated by nanoparticles (NPs) is a promising strategy to overcome the lack of specificity of conventional chemotherapeutic agents. In this context, the prostate-specific membrane antigen (PSMA) has demonstrated a powerful potential for the management of prostate cancer (PCa). Cancer chemoprevention by phytochems. is emerging as a suitable approach for the treatment of early carcinogenic processes. Since (-)-epigallocatechin 3-gallate (EGCG) has shown potent chemopreventive efficacy for PCa, we designed and developed novel targeted NPs in order to selectively deliver EGCG to cancer cells. Herein, to explore the recent concept of "nanochemoprevention", we present a study on EGCG-loaded NPs consisting of biocompatible polymers, functionalized with small mols. targeting PSMA, that exhibited a selective in vitro efficacy against PSMA-expressing PCa cells. This approach could be beneficial for high risk patients and would fulfill a significant therapeutic need, thus opening new perspectives for novel and effective treatment for PCa.
- 8DiPippo, V. A.; Magargal, W. W.; Moorji, S. M.; Murga, J. D.; Olson, W. C. Antiandrogen modulation of prostate-specific membrane antigen (PSMA): dynamics and synergy with PSMA-targeted therapy ASCO Meet. Abstr. 2013, 31, e16007Google ScholarThere is no corresponding record for this reference.
- 9Petrylak, D. P.; Kantoff, P. W.; Mega, A. E.; Vogelzang, N. J.; Stephenson, J.; Fleming, M. T.; Stambler, N.; Petrini, M.; Blattman, S.; Israel, R. J. Prostate-specific membrane antigen antibody drug conjugate (PSMA ADC): a phase I trial in metastatic castration-resistant prostate cancer (mCRPC) previously treated with a taxane ASCO Meet. Abstr. 2013, 31, 5018Google ScholarThere is no corresponding record for this reference.
- 10Rotshteyn, Y.; Mercier, F.; Bruno, R.; Stambler, N.; Israel, R. J.; Wong, V. Correlation of PSMA ADC exposure with reduction in tumor growth rate determined using serial PSA measurements from a phase I clinical trial ASCO Meet. Abstr. 2013, 31, e16047Google ScholarThere is no corresponding record for this reference.
- 11Hrkach, J.; Von Hoff, D.; Mukkaram Ali, M.; Andrianova, E.; Auer, J.; Campbell, T.; De Witt, D.; Figa, M.; Figueiredo, M.; Horhota, A.; Low, S.; McDonnell, K.; Peeke, E.; Retnarajan, B.; Sabnis, A.; Schnipper, E.; Song, J. J.; Song, Y. H.; Summa, J.; Tompsett, D.; Troiano, G.; Van Geen Hoven, T.; Wright, J.; LoRusso, P.; Kantoff, P. W.; Bander, N. H.; Sweeney, C.; Farokhzad, O. C.; Langer, R.; Zale, S. Preclinical development and clinical translation of a PSMA-targeted docetaxel nanoparticle with a differentiated pharmacological profile Sci. Transl. Med. 2012, 4, 128ra39Google ScholarThere is no corresponding record for this reference.
- 12Hillier, S. M.; Maresca, K. P.; Femia, F. J.; Marquis, J. C.; Foss, C. A.; Nguyen, N.; Zimmerman, C. N.; Barrett, J. A.; Eckelman, W. C.; Pomper, M. G.; Joyal, J. L.; Babich, J. W. Preclinical evaluation of novel glutamate–urea–lysine analogues that target prostate-specific membrane antigen as molecular imaging pharmaceuticals for prostate cancer Cancer Res. 2009, 69, 6932– 6940Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVOmtLzE&md5=f222208c3c3fbcaa376306c49c9ec80bPreclinical Evaluation of Novel Glutamate-Urea-Lysine Analogues That Target Prostate-Specific Membrane Antigen as Molecular Imaging Pharmaceuticals for Prostate CancerHillier, Shawn M.; Maresca, Kevin P.; Femia, Frank J.; Marquis, John C.; Foss, Catherine A.; Nguyen, Nghi; Zimmerman, Craig N.; Barrett, John A.; Eckelman, William C.; Pomper, Martin G.; Joyal, John L.; Babich, John W.Cancer Research (2009), 69 (17), 6932-6940CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Prostate-specific membrane antigen (PSMA) is expressed in normal human prostate epithelium and is highly up-regulated in prostate cancer. We previously reported a series of novel small mol. inhibitors targeting PSMA. Two compds., MIP-1072, (S)-2-(3-((S)-1-carboxy-5-(4-iodobenzylamino)pentyl)ureido)pentanedioic acid, and MIP-1095, (S)-2-(3-((S)-1-carboxy-5-(3-(4-iodophenyl)ureido)pentyl)ureido)pentanedioic acid, were selected for further evaluation. MIP-1072 and MIP-1095 potently inhibited the glutamate carboxypeptidase activity of PSMA (Ki = 4.6 ± 1.6 nM and 0.24 ± 0.14 nM, resp.) and, when radiolabeled with 123I, exhibited high affinity for PSMA on human prostate cancer LNCaP cells (Kd = 3.8 ± 1.3 nM and 0.81 ± 0.39 nM, resp.). The assocn. of [123I]MIP-1072 and [123I]MIP-1095 with PSMA was specific; there was no binding to human prostate cancer PC3 cells, which lack PSMA, and binding was abolished by coincubation with a structurally unrelated NAALADase inhibitor, 2-(phosphonomethyl)pentanedioic acid (PMPA). [123I]MIP-1072 and [123I]MIP-1095 internalized into LNCaP cells at 37°C. Tissue distribution studies in mice showed 17.3 ± 6.3% (at 1 h) and 34.3 ± 12.7% (at 4 h) injected dose per g of LNCaP xenograft tissue, for [123I]MIP-1072 and [123I]MIP-1095, resp. [123I]MIP-1095 exhibited greater tumor uptake but slower washout from blood and nontarget tissues compared with [123I]MIP-1072. Specific binding to PSMA in vivo was shown by competition with PMPA in LNCaP xenografts, and the absence of uptake in PC3 xenografts. The uptake of [123I]MIP-1072 and [123I]MIP-1095 in tumor-bearing mice was corroborated by single-photon emission computed tomog./computed tomog. (SPECT/CT) imaging. PSMA-specific radiopharmaceuticals should provide a novel mol. targeting option for the detection and staging of prostate cancer.
- 13Mease, R. C.; Foss, C. A.; Pomper, M. G. PET imaging in prostate cancer: focus on prostate-specific membrane antigen Curr. Top. Med. Chem. 2013, 13, 951– 962Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXpslKhtb0%253D&md5=e49d20dfdfbb41921517c501ca5182e2PET imaging in prostate cancer: focus on prostate-specific membrane antigenMease, Ronnie C.; Foss, Catherine A.; Pomper, Martin G.Current Topics in Medicinal Chemistry (Sharjah, United Arab Emirates) (2013), 13 (8), 951-962CODEN: CTMCCL; ISSN:1568-0266. (Bentham Science Publishers Ltd.)A review. Prostate cancer (PCa) is the second leading cause of cancer-related death in American men. Positron emission tomog./computed tomog. (PET/CT) with emerging radiopharmaceuticals promises accurate staging of primary disease, restaging of recurrent disease, detection of metastatic lesions and, ultimately, for predicting the aggressiveness of disease. Prostate-specific membrane antigen (PSMA) is a well-characterized imaging biomarker of PCa. Because PSMA levels are directly related to androgen independence, metastasis and progression, PSMA could prove an important target for the development of new radiopharmaceuticals for PET. Preclin. data for new PSMA-based radiotracers are discussed and include new 89Zr- and 64Cu-labeled anti-PSMA antibodies and antibody fragments, 64Cu-labeled aptamers, and 11C-, 18F-, 68Ga-, 64Cu-, and 86Y-labeled low mol. wt. inhibitors of PSMA. Several of these agents, namely 68Ga- HBED-CC conjugate 15, 18F-DCFBC 8, and BAY1075553 are particularly promising, each having detected sites of PCa in initial clin. studies. These early clin. results suggest that PET/CT using PSMA-targeted agents, esp. with compds. of low mol. wt., will make valuable contributions to the management of PCa.
- 14Cho, S. Y.; Gage, K. L.; Mease, R. C.; Senthamizhchelvan, S.; Holt, D. P.; Jeffrey-Kwanisai, A.; Endres, C. J.; Dannals, R. F.; Sgouros, G.; Lodge, M.; Eisenberger, M. A.; Rodriguez, R.; Carducci, M. A.; Rojas, C.; Slusher, B. S.; Kozikowski, A. P.; Pomper, M. G. Biodistribution, tumor detection, and radiation dosimetry of 18F–DCFBC, a low-molecular-weight inhibitor of prostate-specific membrane antigen, in patients with metastatic prostate cancer J. Nucl. Med. 2012, 53, 1883– 1891Google ScholarThere is no corresponding record for this reference.
- 15Afshar-Oromieh, A.; Malcher, A.; Eder, M.; Eisenhut, M.; Linhart, H. G.; Hadaschik, B. A.; Holland-Letz, T.; Giesel, F. L.; Kratochwil, C.; Haufe, S.; Haberkorn, U.; Zechmann, C. M. PET imaging with a [(68)Ga]gallium-labelled PSMA ligand for the diagnosis of prostate cancer: biodistribution in humans and first evaluation of tumour lesions Eur. J. Nucl. Med. Mol. Imaging 2012, 40, 486– 495Google ScholarThere is no corresponding record for this reference.
- 16Barrett, J. A.; Coleman, R. E.; Goldsmith, S. J.; Vallabhajosula, S.; Petry, N. A.; Cho, S.; Armor, T.; Stubbs, J. B.; Maresca, K. P.; Stabin, M. G.; Joyal, J. L.; Eckelman, W. C.; Babich, J. W. First-in-man evaluation of 2 high-affinity PSMA-avid small molecules for imaging prostate cancer J. Nucl. Med. 2013, 54, 380– 387Google ScholarThere is no corresponding record for this reference.
- 17Maresca, K. P.; Hillier, S. M.; Lu, G.; Marquis, J. C.; Zimmerman, C. N.; Eckelman, W. C.; Joyal, J. L.; Babich, J. W. Small molecule inhibitors of PSMA incorporating technetium-99m for imaging prostate cancer: effects of chelate design on pharmacokinetics Inorg. Chim. Acta 2012, 389, 168– 172Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XnslKmt74%253D&md5=2165975aa8528ada852cecc63bb0bd0cSmall molecule inhibitors of PSMA incorporating technetium-99m for imaging prostate cancer: Effects of chelate design on pharmacokineticsMaresca, Kevin P.; Hillier, Shawn M.; Lu, Genliang; Marquis, John C.; Zimmerman, Craig N.; Eckelman, William C.; Joyal, John L.; Babich, John W.Inorganica Chimica Acta (2012), 389 (), 168-175CODEN: ICHAA3; ISSN:0020-1693. (Elsevier B.V.)Single amino acid chelate (SAAC) systems for the complexation of the M(CO)3 moiety (M = Tc/Re) have been successfully incorporated into novel synthetic strategies for radiopharmaceuticals and evaluated in a variety of biol. applications. However, the lipophilicity of the first generation of 99mTc(CO)3 complexes has resulted in substantial hepatobiliary uptake when examd. either as lysine derivs. or integrated into biol. active small mols. and peptides. Here, we designed, synthesized, and evaluated novel polar functionalized imidazole derived SAAC systems (SAAC II) which have been chem. modified to promote overall 99mTc(CO)3L3 complex hydrophilicity with the intent of reducing non-target effects and enhancing renal clearance of prostate specific membrane antigen (PSMA) targeting small mols. The 99mTc-labeled compds. were prepd., purified, and evaluated for stability, lipophilicity, and tissue distribution in LNCaP xenograft mice. The Glu-urea-Lys-C11 analogs were prepd. with a variety of chelators to form (19R,23S)-1-(X)-2-((Y)methyl)-13,21-dioxo-2,14,20,22-tetraazapentacosane-19,23,25-tricarboxylic acid where X = Y = (methyl)pyridin-2-yl (6), X = Y = (methyl)-1H-imidazol-2-yl (7), X = (methyl)pyridin-2-yl, Y = carboxymethyl (8), X = Y = 1-(carboxymethyl)-1H-imidazol-2-yl (9), X = 1-(carboxymethyl)-1H-imidazol-2-yl, Y = carboxymethyl (10), and X = Y = 1-(1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl)-2-((1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl) (11). 99mTc labeling was achieved at ligand concns. as low as 10-6 M and the complexes were stable (>90%) for 24 h. These new SAAC II chelators were evaluated for their influence on binding of the Glu-urea-Lys-C11 analogs to PSMA-pos. LNCaP cells and compared to pyridine- and N-methylimidazole-contg. SAAC ligands. Tissue distribution of the 99mTc-complexes contg. the more polar chelators, 9 and 11, demonstrated decreased liver (<2% ID/g) and increased LNCaP tumor (>11% ID/g) accumulation at 1 h post-injection.
- 18Nedrow-Byers, J. R.; Moore, A. L.; Ganguly, T.; Hopkins, M. R.; Fulton, M. D.; Benny, P. D.; Berkman, C. E. PSMA-targeted SPECT agents: Mode of binding effect on in vitro performance Prostate 2013, 73, 355– 362Google ScholarThere is no corresponding record for this reference.
- 19Lu, G.; Maresca, K. P.; Hillier, S. M.; Zimmerman, C. N.; Eckelman, W. C.; Joyal, J. L.; Babich, J. W. Synthesis and SAR of 99mTc/Re-Labeled Small Molecule Prostate Specific Membrane Antigen Inhibitors with Novel Polar Chelates Biorg. Med. Chem. Lett. 2013, 23, 1557– 1563Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXptlKjtQ%253D%253D&md5=0dbceebf79f273aafb5db22b912302f6Synthesis and SAR of 99mTc/Re-labeled small molecule prostate specific membrane antigen inhibitors with novel polar chelatesLu, Genliang; Maresca, Kevin P.; Hillier, Shawn M.; Zimmerman, Craig N.; Eckelman, William C.; Joyal, John L.; Babich, John W.Bioorganic & Medicinal Chemistry Letters (2013), 23 (5), 1557-1563CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Prostate specific membrane antigen (PSMA) is recognized as an attractive mol. target for the development of radiopharmaceuticals to image and potentially treat metastatic prostate cancer. A series of novel 99mTc/Re-tricarbonyl radiolabeled PSMA inhibitors were therefore synthesized by the attachment of glutamate-urea-lysine (Glu-urea-Lys) and glutamate-urea-glutamate (Glu-urea-Glu) pharmacophore to single amino acid chelate (SAAC) where the SAAC ligand was either bis(pyridin-2-ylmethyl)amino (DPA), bis((1-methyl-1H-imidazol-2-yl)methyl)amino (NMI), bis((1-(carboxymethyl)-1H-imidazol-2-yl)methyl)amino (CIM) or bis((1-(2-(bis(carboxymethyl)amino)-2-oxoethyl)-1H-imidazol-2-yl)methyl)amino (TIM). The in vitro binding affinity of the rhenium complexes was evaluated using PSMA-expressing human prostate cancer LNCaP cells. IC50 values ranged from 3.8 ± 2 to >2000 nM. A linker between the SAAC chelate and pharmacophore was required for high affinity binding. However, extending the length of the linker did not substantially improve binding. PSMA binding was also influenced by the nature of the SAAC chelate. One of the most potent compds., 23b (IC50 = 4.8 ± 2.7 nM), was radiolabeled with technetium tricarbonyl ({99mTc(CO)3}+) to afford the {99mTc(CO)3}+ complex in excellent yield and high purity. This effort led to the identification of a diverse series of promising high affinity {99mTc(CO)3}+ radiolabeled PSMA inhibitors.
- 20Nedrow-Byers, J. R.; Jabbes, M.; Jewett, C.; Ganguly, T.; He, H.; Liu, T.; Benny, P.; Bryan, J. N.; Berkman, C. E. A phosphoramidate-based prostate-specific membrane antigen-targeted SPECT agent Prostate 2012, 72, 904– 912Google ScholarThere is no corresponding record for this reference.
- 21Banerjee, S. R.; Foss, C. A.; Castanares, M.; Mease, R. C.; Byun, Y.; Fox, J. J.; Hilton, J.; Lupold, S. E.; Kozikowski, A. P.; Pomper, M. G. Synthesis and evaluation of technetium-99m- and rhenium-labeled inhibitors of the prostate-specific membrane antigen (PSMA) J. Med. Chem. 2008, 51, 4504– 4517Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXoslamsr4%253D&md5=e070e63a175f5d733d58021a6bfdf87dSynthesis and Evaluation of Technetium-99m- and Rhenium-Labeled Inhibitors of the Prostate-Specific Membrane Antigen (PSMA)Banerjee, Sangeeta R.; Foss, Catherine A.; Castanares, Mark; Mease, Ronnie C.; Byun, Youngjoo; Fox, James J.; Hilton, John; Lupold, Shawn E.; Kozikowski, Alan P.; Pomper, Martin G.Journal of Medicinal Chemistry (2008), 51 (15), 4504-4517CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The prostate-specific membrane antigen (PSMA) is increasingly recognized as a viable target for imaging and therapy of cancer. We prepd. seven 99mTc/Re-labeled compds. by attaching known Tc/Re chelating agents to an amino-functionalized PSMA inhibitor (lys-NHCONH-glu) with or without a variable length linker moiety. Ki values ranged from 0.17 to 199 nM. Ex vivo biodistribution and in vivo imaging demonstrated the degree of specific binding to engineered PSMA+ PC3 PIP tumors. PC3-PIP cells are derived from PC3 that have been transduced with the gene for PSMA. Despite demonstrating nearly the lowest PSMA inhibitory potency of this series, [99mTc(CO)3(L1)]+ (L1 = (2-pyridylmethyl)2N(CH2)4CH(CO2H)NHCO-(CH2)6CO-NH-lys-NHCONH-glu) showed the highest, most selective PIP tumor uptake, at 7.9 ± 4.0% injected dose per g of tissue at 30 min postinjection. Radioactivity cleared from nontarget tissues to produce a PIP to flu (PSMA-PC3) ratio of 44:1 at 120 min postinjection. PSMA can accommodate the steric requirements of 99mTc/Re complexes within PSMA inhibitors, the best results achieved with a linker moiety between the .vepsiln. amine of the urea lysine and the chelator.
- 22Kularatne, S. A.; Zhou, Z.; Yang, J.; Post, C. B.; Low, P. S. Design, synthesis, and preclinical evaluation of prostate-specific membrane antigen targeted (99m)Tc-radioimaging agents Mol. Pharmaceutics 2009, 6, 790– 800Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlsF2hs70%253D&md5=77c5b6f3776e761979de857d4299106eDesign, Synthesis, and Preclinical Evaluation of Prostate-Specific Membrane Antigen Targeted 99mTc-Radioimaging AgentsKularatne, Sumith A.; Zhou, Zhigang; Yang, Jun; Post, Carol B.; Low, Philip S.Molecular Pharmaceutics (2009), 6 (3), 790-800CODEN: MPOHBP; ISSN:1543-8384. (American Chemical Society)The high mortality and financial burden assocd. with prostate cancer can be partly attributed to a lack of sensitive screening methods for detection and staging of the disease. Guided by in silico docking studies using the crystal structure of PSMA, we designed and synthesized a series of PSMA-targeted 99mTc-chelate complexes for imaging PSMA-expressing human prostate cancer cells (LNCaP cell line). Of the six targeted radioimaging agents synthesized, three were found to bind LNCaP cells with low nanomolar affinity. Moreover, the same three PSMA-targeted imaging agents were shown to localize primarily to LNCaP tumor xenografts in nu/nu mice, with an av. of 9.8 ± 2.4% injected dose/g tissue accumulating in the tumor and only 0.11% injected dose/g tissue retained in the muscle at 4 h postinjection. Collectively, these high affinity, PSMA-specific radioimaging agents demonstrate significant potential for use in localizing prostate cancer masses, monitoring response to therapy, detecting prostate cancer recurrence following surgery, and selecting patients for subsequent PSMA-targeted chemotherapy.
- 23Zhang, Y.; DiFilipp, F.; Doke, A.; Huang, J.; Heston, W.; Huang, S. Preliminary micro-SPECT and biodistribution study of a novel Tc99m-labeled PSMA tracer derived from RBI1033 J. Nucl. Med. Meet. Abstr. 2012, 53 (Suppl 1) 1661Google ScholarThere is no corresponding record for this reference.
- 24Schafer, M.; Bauder-Wust, U.; Leotta, K.; Zoller, F.; Mier, W.; Haberkorn, U.; Eisenhut, M.; Eder, M. A dimerized urea-based inhibitor of the prostate-specific membrane antigen for 68Ga-PET imaging of prostate cancer EJNMMI Res. 2012, 2, 23Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC38npsFSmuw%253D%253D&md5=186d67c326bb60367462b16b0deb419fA dimerized urea-based inhibitor of the prostate-specific membrane antigen for 68Ga-PET imaging of prostate cancerSchafer Martin; Bauder-Wust Ulrike; Leotta Karin; Zoller Frederic; Mier Walter; Haberkorn Uwe; Eisenhut Michael; Eder MatthiasEJNMMI research (2012), 2 (1), 23 ISSN:.UNLABELLED: BACKGROUND: Alternative positron-emission tomography (PET) probes like labeled inhibitors of the prostate-specific membrane antigen (PSMA) are of emerging clinical impact as they show the ability to image small lesions of recurrent prostate cancer. Here, the dimerization of the pharmacophore Glu-ureido-Lys via the 68Ga chelator N,N'-bis[2-hydroxy-5-(carboxyethyl)benzyl]ethylenediamine-N,N'-diacetic acid (HBED-CC) was investigated to further improve the binding characteristics and pharmacokinetics. METHODS: The peptidomimetic structures were synthesized by solid-phase chemistry, and the resulting products were coupled with the respective 2,3,5,6-tetrafluorophenol esters of HBED-CC to form the monomeric reference and the dimeric Glu-ureido-Lys derivative. The binding properties were analyzed in competitive binding, internalization, and cell surface retention experiments. PET images and biodistribution data were obtained 1 h after injection in BALB/c nu/nu mice bearing LNCaP tumor xenografts. RESULTS: Cell binding data revealed significant better binding properties of the dimer (IC50 = 3.9 ± 1.8 nM; IC50 (monomer) = 12.1 ± 2.1 nM). The inhibition potency investigated by the enzyme-based NAALADase assay confirmed these results. Specific internalization in LNCaP cells was demonstrated for both, the monomer and dimer. As shown by efflux measurements, the dimeric compound was more effectively retained on the cell surface, resulting in advanced in vivo properties (T/BMonomer = 9.2; T/BDimer = 26.5). CONCLUSIONS: The dimeric [68Ga]7 is a promising imaging agent for PSMA-expressing tumors as it shows higher tumor uptake while observing more favorable background clearance. As compared to the respective monomer, the higher affinity and prolonged tumor retention additionally represent promising features and warrant further evaluation regarding 68Ga-PET imaging of PSMA expression.
- 25Eder, M.; Schafer, M.; Bauder-Wust, U.; Hull, W. E.; Wangler, C.; Mier, W.; Haberkorn, U.; Eisenhut, M. 68Ga-complex lipophilicity and the targeting property of a urea-based PSMA inhibitor for PET imaging Bioconjugate Chem. 2012, 23, 688– 697Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XivVCmtLo%253D&md5=4bb4cbba9f510ed2561b87a44bafa00a68Ga-Complex Lipophilicity and the Targeting Property of a Urea-Based PSMA Inhibitor for PET ImagingEder, Matthias; Schaefer, Martin; Bauder-Wuest, Ulrike; Hull, William-Edmund; Waengler, Carmen; Mier, Walter; Haberkorn, Uwe; Eisenhut, MichaelBioconjugate Chemistry (2012), 23 (4), 688-697CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Urea-based inhibitors of the prostate-specific membrane antigen (PSMA) represent low-mol.-wt. pepidomimetics showing the ability to image PSMA-expressing prostate tumors. The highly efficient, acyclic Ga(III) chelator N,N'-bis [2-hydroxy-5-(carboxyethyl)benzyl] ethylenediamine-N,N'- diacetic acid (HBED-CC) was introduced as a lipophilic side chain into the hydrophilic pharmacophore Glu-NH-CO-NH-Lys which was found favorable to interact with the PSMA "active binding site". This report describes the syntheses, in vitro binding analyses, and biodistribution data of the radiogallium labeled PSMA inhibitor Glu-NH-CO-NH-Lys(Ahx)-HBED-CC in comparison to the corresponding DOTA conjugate. The binding properties were analyzed using competitive cell binding and enzyme-based assays followed by internalization expts. Compared to the DOTA-conjugate, the HBED-CC deriv. showed reduced unspecific binding and considerable higher specific internalization in LNCaP cells. The 68Ga complex of the HBED-CC ligand exhibited higher specificity for PSMA expressing tumor cells resulting in improved in vivo properties. 68Ga labeled Glu-NH-CO-NH-Lys(Ahx)-HBED-CC showed fast blood and organ clearances, low liver accumulation, and high specific uptake in PSMA expressing organs and tumor. It could be demonstrated that the PET-imaging property of a urea-based PSMA inhibitor could significantly be improved with HBED-CC.
- 26Banerjee, S. R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Green, G.; Fox, J. J.; Horti, A.; Mease, R. C.; Pomper, M. G. 68Ga-labeled inhibitors of prostate-specific membrane antigen (PSMA) for imaging prostate cancer J. Med. Chem. 2010, 53, 5333– 5341Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnvVKqs7c%253D&md5=cbe8ccdc2d1833374546e68091632d9068Ga-Labeled Inhibitors of Prostate-Specific Membrane Antigen (PSMA) for Imaging Prostate CancerBanerjee, Sangeeta Ray; Pullambhatla, Mrudula; Byun, Youngjoo; Nimmagadda, Sridhar; Green, Gilbert; Fox, James J.; Horti, Andrew; Mease, Ronnie C.; Pomper, Martin G.Journal of Medicinal Chemistry (2010), 53 (14), 5333-5341CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Gallium-68 is a generator-produced radionuclide for positron emission tomog. (PET) that is being increasingly used for radiolabeling of tumor-targeting peptides. Compds. [68Ga]3 and [68Ga]6 are high-affinity urea-based inhibitors of the prostate-specific membrane antigen (PSMA) that were synthesized in decay-uncorrected yields ranging from 60% to 70% and radiochem. purities of more than 99%. Compd. [68Ga]3 demonstrated 3.78 ± 0.90% injected dose per g of tissue (%ID/g) within PSMA+ PIP tumor at 30 min postinjection, while [68Ga]6 showed a 2 h PSMA+ PIP tumor uptake value of 3.29 ± 0.77 %ID/g. Target (PSMA+ PIP) to nontarget (PSMA- flu) ratios were 4.6 and 18.3, resp., at those time points. Both compds. delineated tumor clearly by small animal PET. The urea series of imaging agents for PSMA can be radiolabeled with 68Ga, a cyclotron-free isotope useful for clin. PET studies, with maintenance of target specificity.
- 27Banerjee, S. R.; Pullambhatla, M.; Shallal, H.; Lisok, A.; Mease, R. C.; Pomper, M. G. A Modular Strategy to Prepare Multivalent Inhibitors of Prostate-Specific Membrane Antigen (PSMA) Oncotarget 2011, 2, 1244– 1253Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC387ntVehsQ%253D%253D&md5=ff90dd0090ea69db9c798ba6ebde318bA modular strategy to prepare multivalent inhibitors of prostate-specific membrane antigen (PSMA)Banerjee Sangeeta Ray; Pullambhatla Mrudula; Shallal Hassan; Lisok Ala; Mease Ronnie C; Pomper Martin GOncotarget (2011), 2 (12), 1244-53 ISSN:.We have developed a modular scaffold for preparing high-affinity, homo-multivalent inhibitors of the prostate-specific membrane antigen (PSMA) for imaging and therapy of prostate cancer (PCa). Our system contains a lysine-based (μ-, e-) dialkyne residue for incorporating a PSMA binding Lys-Glu urea motif exploiting click chemistry and a second lysine residue for subsequent modification with an imaging or therapeutic moiety. The utility of the multivalent scaffold was examined by synthesizing bivalent compounds 2 and 3 and comparing them with the monovalent analog 1. Determination of inhibition constants (Ki) revealed that bivalent 2 (0.2 nM) and 3 (0.08 nM) are significantly more potent (~ 5 fold and ~ 11 fold, respectively) inhibitors of PSMA than monovalent 1 (0.9 nM). A single photon emission computed tomography (SPECT)-CT imaging study of [111In]3 demonstrated high and specific uptake in PSMA+ PC-3 PIP tumor until at least 48 h post-injection, with rapid clearance from non-target tissues, including kidney. A biodistribution study revealed that [111In]3 demonstrated 34.0 ± 7.5 percent injected dose per gram of tissue in PSMA+ tumor at 24 h post-injection and was capable of generating target-to-non-target ratios of ~ 379 in PSMA+ PC-3 PIP tumors vs. isogenic PSMA-negative PC3-flu tumors in vivo. The click chemistry approach affords a convenient strategy toward multivalent PSMA inhibitors of enhanced affinity and superior pharmacokinetics for imaging.
- 28Banerjee, S. R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Foss, C. A.; Green, G.; Fox, J. J.; Lupold, S. E.; Mease, R. C.; Pomper, M. G. Sequential SPECT and optical imaging of experimental models of prostate cancer with a dual modality inhibitor of the prostate-specific membrane antigen Angew. Chem. 2011, 50, 9167– 9170Google ScholarThere is no corresponding record for this reference.
- 29Chen, Z.; Penet, M. F.; Nimmagadda, S.; Li, C.; Banerjee, S. R.; Winnard, P. T., Jr.; Artemov, D.; Glunde, K.; Pomper, M. G.; Bhujwalla, Z. M. PSMA-targeted theranostic nanoplex for prostate cancer therapy ACS Nano 2012, 6, 7752– 7762Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtFGiu7jF&md5=5c9091dff6c26a165b597e4af3dd5032PSMA-Targeted Theranostic Nanoplex for Prostate Cancer TherapyChen, Zhihang; Penet, Marie-France; Nimmagadda, Sridhar; Li, Cong; Banerjee, Sangeeta R.; Winnard, Paul T.; Artemov, Dmitri; Glunde, Kristine; Pomper, Martin G.; Bhujwalla, Zaver M.ACS Nano (2012), 6 (9), 7752-7762CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Theranostic imaging, where diagnosis is combined with therapy, is particularly suitable for a disease that is as complex as cancer, esp. now that genomic and proteomic profiling can provide an extensive "fingerprint" of each tumor. With such information, theranostic agents can be designed to personalize treatment and minimize damage to normal tissue. Here we have developed a nanoplex platform for theranostic imaging of prostate cancer (PCa). In these proof-of-principle studies, a therapeutic nanoplex contg. multimodal imaging reporters was targeted to prostate-specific membrane antigen (PSMA), which is expressed on the cell surface of castrate-resistant PCa. The nanoplex was designed to deliver small interfering RNA (siRNA) along with a prodrug enzyme to PSMA-expressing tumors. Each component of the nanoplex was carefully selected to evaluate its diagnostic aspect of PSMA imaging and its therapeutic aspects of siRNA-mediated down-regulation of a target gene and the conversion of a prodrug to cytotoxic drug, using noninvasive multimodality imaging. Studies performed using two variants of human PC3-PCa cells and tumors, one with high PSMA expression level and another with negligible expression levels, demonstrated PSMA-specific uptake. In addn., down-regulation of the selected siRNA target, choline kinase (Chk), and the conversion of the nontoxic prodrug 5-fluorocytosine (5-FC) to cytotoxic 5-fluorouracil (5-FU) were also demonstrated with noninvasive imaging. The nanoplex was well-tolerated and did not induce liver or kidney toxicity or a significant immune response. The nanoplex platform described can be easily modified and applied to different cancers, receptors, and pathways to achieve theranostic imaging, as a single agent or in combination with other treatment modalities.
- 30Hao, G.; Kumar, A.; Dobin, T.; Oz, O. K.; Hsieh, J. T.; Sun, X. A multivalent approach of imaging probe design to overcome an endogenous anion binding competition for noninvasive assessment of prostate specific membrane antigen Mol. Pharmacol. 2013, 10, 2975– 2985Google ScholarThere is no corresponding record for this reference.
- 31Banerjee, S. R.; Pullambhatla, M.; Byun, Y.; Nimmagadda, S.; Baidoo, K. E.; Brechbiel, M. W.; Mease, R. C.; Pomper, M. G. Preclinical Evaluation of 86Y-Labeled Inhibitors of Prostate Specific Membrane Antigen. J. Labelled Compd. Radiopharm. 2011, 54, Suppl S1, p S65.Google ScholarThere is no corresponding record for this reference.
- 32Holland, J. P.; Divilov, V.; Bander, N. H.; Smith-Jones, P. M.; Larson, S. M.; Lewis, J. S. 89Zr–DFO–J591 for immunoPET of prostate-specific membrane antigen expression in vivo J. Nucl. Med. 2010, 51, 1293– 1300Google ScholarThere is no corresponding record for this reference.
- 33Viola-Villegas, N.; Evans, H.; Bartlett, D.; Wu, A.; Lewis, J. Preclinical development of Zr-89 labeled anti-PSMA minibody and cys-diabody J Nucl. Med. Meet. Abstr. 2012, 53, 347Google ScholarThere is no corresponding record for this reference.
- 34Ray Banerjee, S.; Pullambhatla, M.; Foss, C. A.; Falk, A.; Byun, Y.; Nimmagadda, S.; Mease, R. C.; Pomper, M. G. Effect of chelators on the pharmacokinetics of (99m)Tc-labeled imaging agents for the prostate-specific membrane antigen (PSMA) J. Med. Chem. 2013, 56, 6108– 6121Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtVWhtLjE&md5=3007d66e9dc898d5beb4039cd02ddae3Effect of Chelators on the Pharmacokinetics of 99mTc-Labeled Imaging Agents for the Prostate-Specific Membrane Antigen (PSMA)Ray Banerjee, Sangeeta; Pullambhatla, Mrudula; Foss, Catherine A.; Falk, Alexander; Byun, Youngjoo; Nimmagadda, Sridhar; Mease, Ronnie C.; Pomper, Martin G.Journal of Medicinal Chemistry (2013), 56 (15), 6108-6121CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Technetium-99m, the most commonly used radionuclide in nuclear medicine, can be attached to biol. important mols. through a variety of chelating agents, the choice of which depends upon the imaging application. The prostate-specific membrane antigen (PSMA) is increasingly recognized as an important target for imaging and therapy of prostate cancer (PCa). Three different 99mTc-labeling methods were employed to investigate the effect of the chelator on the biodistribution and PCa tumor uptake profiles of 12 new urea-based PSMA-targeted radiotracers. This series includes hydrophilic ligands for radiolabeling with the [99mTc(CO)3]+ core (L8-L10), traditional NxSy-based chelating agents with varying charge and polarity for the 99mTc-oxo core (L11-L18), and a 99mTc-organohydrazine-labeled radioligand (L19). 99mTc(I)-Tricarbonyl-labeled [99mTc]L8 produced the highest PSMA+ PC3 PIP to PSMA- PC3 flu tumor ratios and demonstrated the lowest retention in normal tissues including kidney after 2 h. These results suggest that choice of chelator is an important pharmacokinetic consideration in the development of 99mTc-labeled radiopharmaceuticals targeting PSMA.
- 35Wadas, T. J.; Wong, E. H.; Weisman, G. R.; Anderson, C. J. Copper chelation chemistry and its role in copper radiopharmaceuticals Curr. Pharm. Des. 2007, 13, 3– 16Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXht1Wjsrs%253D&md5=f79c91f1853f0188a9f129acc7902dc9Copper chelation chemistry and its role in copper radiopharmaceuticalsWadas, T. J.; Wong, E. H.; Weisman, G. R.; Anderson, C. J.Current Pharmaceutical Design (2007), 13 (1), 3-16CODEN: CPDEFP; ISSN:1381-6128. (Bentham Science Publishers Ltd.)A review. Mol. imaging is an important scientific discipline that plays a major role in clin. medicine and pharmaceutical development. While several imaging modalities including x-ray computed tomog. and magnetic resonance imaging generate high-resoln. anatomical images, positron emission tomog. (PET) and single photon emission computed tomog. offer insight into the physiol. processes that occur within a living organism. Of these two nuclear medicine imaging techniques, PET has advantages with respect to sensitivity and resoln., and this has led to the prodn. and development of many positron emitting radionuclides that include non-traditional radionuclides of the transition metals. Copper-64 (t1/2 = 12.7 h, β+: 17.4%, Eβ+max = 656 keV; β-: 39%, Eβ-max = 573 keV) has emerged as an important positron emitting radionuclide that has the potential for use in diagnostic imaging and radiotherapy. However, 64Cu must be delivered to the living system as a stable complex that is attached to a biol. targeting mol. for effective imaging and therapy. Therefore, significant research has been devoted to the development of ligands that can stably chelate 64Cu. This review discusses the necessary characteristics of an effective 64Cu chelator, while highlighting the development and evaluation of 64Cu-complexes attached to biol.-targeted ligands.
- 36Connett, J. M.; Anderson, C. J.; Guo, L. W.; Schwarz, S. W.; Zinn, K. R.; Rogers, B. E.; Siegel, B. A.; Philpott, G. W.; Welch, M. J. Radioimmunotherapy with a 64Cu-labeled monoclonal antibody: a comparison with 67Cu Proc. Natl. Acad. Sci. U. S. A. 1996, 93, 6814– 6818Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28XjvFeit78%253D&md5=44ebb765573e81cab4eb53cc35c1249dRadioimmunotherapy with a 64Cu-labeled monoclonal antibody: a comparison with 67CuConnett, Judith M.; Anderson, Carolyn J.; Guo, Li-Wu; Schwarz, Sally W.; Zinn, Kurt R.; Rogers, Buck E.; Siegel, Barry A.; Philpott, Gordon; Welch, Michael J.Proceedings of the National Academy of Sciences of the United States of America (1996), 93 (13), 6814-6818CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)67Cu (t1/2 = 62 h) has demonstrated potential as a radionuclide for radioimmunotherapy, but limited availability severely restricts its widespread use. 64Cu (t1/2 = 12.8 h) has been shown to have comparable effectiveness in vitro and in vivo. The present study was undertaken to examine the therapeutic potential of 64Cu- and 67Cu-bromoacetamidobenzyl-1,4,8,11-tetraazacyclotetradecane-N,N',N'',N'''-tetraacetic acid (BAT)-2-iminothiolane (2IT)-1A3 (1A3 is a mouse anti-human colorectal cancer mAb) for treatment of GW39 human colon carcinoma carried in hamster thighs. Hamsters were injected with 64Cu- or 67C-BAT-2IT-1A3 or Cu-labeled nonspecific IgG (MOPC) or saline. Hamsters were killed 6-7 mo after therapy or when tumors were ≥10 g. Of the hamsters with small tumors (mean wt. 0.43 ± 0.25 g), 87.5% were disease-free 7 mo after treatment with 2 mCi (1 Ci = 37 GBq) of 64Cu-BAT-2IT-1A3 or 0.4 mCi of 67Cu-BAT-2IT-1A3. The mean tumor doses at these activities of 64Cu- and 67Cu-BAT-2IT-1A3 were 586 and 1269 rad (1 rad = 0.01 Gy), resp. In contrast, 76% of hamsters treated with 2 mCi of 64Cu-BAT-2IT-MOPC or 0.4 mCi of 67Cu-BAT-2IT-MOPC had to be killed before 6 mo because of tumor regrowth. When hamsters with larger tumors (mean wt. 0.66 ± 0.11 g) were treated with 64Cu- or 67Cu-BAT-2IT-1A3, survival was extended compared with controls, but only one animal remained tumor-free to 6 mo. These results demonstrate that 64Cu- and 67Cu-BAT-2IT-1A3 given in a single administered dose can eradicate small tumors without significant host toxicity, but addnl. strategies to deliver higher tumor doses will be needed for larger tumors.
- 37Blower, P. J.; Lewis, J. S.; Zweit, J. Copper radionuclides and radiopharmaceuticals in nuclear medicine Nucl. Med. Biol. 1996, 23, 957– 980Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXkslOqsQ%253D%253D&md5=39b9b14d56995b81d7c57f35e2fcdd82Copper radionuclides and radiopharmaceuticals in nuclear medicineBlower, Philip J.; Lewis, Jason S.; Zweit, JamalNuclear Medicine and Biology (1996), 23 (8), 957-980CODEN: NMBIEO; ISSN:0883-2897. (Elsevier)The chem., radiochem., radiobiol., and radiopharmacol. of radiopharmaceuticals contg. copper radionuclides are reviewed, with 210 refs. Copper radionuclides offer application in positron emission tomog., targeted radiotherapy, and single photon imaging. The chem. of copper is relatively simple and well-suited to radiopharmaceutical application. Current radiopharmaceuticals include biomols. labeled via bifunctional chelators primarily based on cyclic polyaminocarboxylates and polyamines, and pyruvaldehyde-bis(N4-methylthiosemicarbazone) (PTSM) and its analogs. The chem. of copper, of which only a fraction has yet been exploited, is likely to be applied more fully in the future.
- 38Guo, Y.; Parry, J. J.; Laforest, R.; Rogers, B. E.; Anderson, C. J. The role of p53 in combination radioimmunotherapy with 64Cu–DOTA–cetuximab and cisplatin in a mouse model of colorectal cancer J. Nucl. Med. 2013, 54, 1621– 1629Google ScholarThere is no corresponding record for this reference.
- 39Donnelly, P. S. The role of coordination chemistry in the development of copper and rhenium radiopharmaceuticals Dalton Trans. 2011, 40, 999– 1010Google ScholarThere is no corresponding record for this reference.
- 40Smith, S. V. Molecular imaging with copper-64 J. Inorg. Biochem. 2004, 98, 1874– 1901Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptlOktL0%253D&md5=b0972b1167a9b710b5c409d772941d95Molecular imaging with copper-64Smith, Suzanne V.Journal of Inorganic Biochemistry (2004), 98 (11), 1874-1901CODEN: JIBIDJ; ISSN:0162-0134. (Elsevier B.V.)A review. Mol. imaging is expected to change the face of drug discovery and development. The ability to link imaging to biol. for guiding therapy should improve the rate at which novel imaging technologies, probes, contrast agents, drugs and drug delivery systems can be transferred into clin. practice. Nuclear medicine imaging, in particular, positron emission tomog. (PET) allows the detection and monitoring of a variety of biol. and pathophysiol. processes, at tracer quantities of the radiolabeled target agents, and at doses free from pharmacol. effects. In the field of drug discovery and development, the use of radiotracers for radiolabelling target agents has now become one of the essential tools in identifying, screening and development of new target agents. In this regard, 64Cu (t1/2 = 12.7 h) has been identified as an emerging PET isotope. Its half-life is sufficiently long for radiolabelling a range of target agents and its ease of prodn. and adaptable chem. make it an excellent radioisotope for use in mol. imaging. This review describes recent advances, in the routes of 64Cu prodn., design and application of bi-functional ligands for use in radiolabelling with 64/67Cu2+, and their significance and anticipated impact on the field of mol. imaging and drug development.
- 41Wadas, T. J.; Wong, E. H.; Weisman, G. R.; Anderson, C. J. Molecular imaging of cancer with copper-64 radiopharmaceuticals and positron emission tomography (PET) Chem. Rev. 2010, 110, 2858– 2902Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXltFChtro%253D&md5=674cf315a0df26fc4407445bd35595d6Coordinating Radiometals of Copper, Gallium, Indium, Yttrium, and Zirconium for PET and SPECT Imaging of DiseaseWadas, Thaddeus J.; Wong, Edward H.; Weisman, Gary R.; Anderson, Carolyn J.Chemical Reviews (Washington, DC, United States) (2010), 110 (5), 2858-2902CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review.
- 42Shokeen, M.; Anderson, C. J. Coordinating radiometals of copper, gallium, indium, yttrium, and zirconium for PET and SPECT imaging of disease Acc. Chem. Res. 2009, 110, 832– 841Google ScholarThere is no corresponding record for this reference.
- 43De Silva, R. A.; Peyre, K.; Pullambhatla, M.; Fox, J. J.; Pomper, M. G.; Nimmagadda, S. Imaging CXCR4 expression in human cancer xenografts: evaluation of monocyclam 64Cu–AMD3465 J. Nucl. Med. 2011, 52, 986– 993Google ScholarThere is no corresponding record for this reference.
- 44Yuan, H.; Schroeder, T.; Bowsher, J. E.; Hedlund, L. W.; Wong, T.; Dewhirst, M. W. Intertumoral differences in hypoxia selectivity of the PET imaging agent 64Cu(II)-diacetyl-bis(N4-methylthiosemicarbazone) J. Nucl. Med. 2006, 47, 989– 998Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XmtlWmtrg%253D&md5=805152c3ada8861fa94ae0e0a28c16daIntertumoral differences in hypoxia selectivity of the PET imaging agent 64Cu(II)-diacetyl-Bis(N4-methylthiosemicarbazone)Yuan, Hong; Schroeder, Thies; Bowsher, James E.; Hedlund, Laurence W.; Wong, Terence; Dewhirst, Mark W.Journal of Nuclear Medicine (2006), 47 (6), 989-998CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine)Cu-Diacetyl-bis(N4-methylthiosemicarbazone) (Cu-ATSM) is a recently developed PET imaging agent for tumor hypoxia. However, its accuracy and reliability for measuring hypoxia have not been fully characterized in vivo. The aim of this study was to evaluate 64Cu-ATSM as a hypoxia PET marker by comparing autoradiog. distributions of 64Cu-ATSM with a well-established hypoxia marker drug, EF5. Methods: R3230 mammary adenocarcinomas (R3230Ac), fibrosarcomas (FSA), and 9L gliomas (9L) were used in the study. EF5 and Hoechst 33342, a vascular perfusion marker, were administered to the animal for immunohistochem. anal. 64Cu-ATSM microPET and autoradiog. were performed on the same animal. The tumor-to-muscle ratio (T/M ratio) and standardized uptake values (SUVs) were characterized for these 3 different types of tumors. Five types of images-microPET, autoradiog., EF5 immunostaining, Hoechst fluorescence vascular imaging, and hematoxylin-andeosin histol.-were superimposed, evaluated, and compared. Results: A significantly higher T/M ratio and SUV were seen for FSA compared with R3230Ac and 9L. Spatial correlation anal. between 64Cu-ATSM autoradiog. and EF5 immunostained images varied between the 3 tumor types. There was close correlation of 64Cu-ATSM uptake and hypoxia in R3230Ac and 9L tumors but not in FSA tumors. Interestingly, elevated 64Cu-ATSM uptake was obsd. in well-perfused areas in FSA, indicating a correlation between 64Cu-ATSM uptake and vascular perfusion as opposed to hypoxia. The same relationship was obsd. with 2 other hypoxia markers, pimonidazole and carbonic anhydrase IX, in FSA tumors. Breathing carbogen gas significantly decreased the hypoxia level measured by EF5 staining in FSA-bearing rats but not the uptake of 64Cu-ATSM. These results indicate that some other 64Cu-ATSM retention mechanisms, as opposed to hypoxia, are involved in this type of tumor. Conclusion: To our knowledge, this study is the first comparison between 64Cu-ATSM uptake and immunohistochem. in these 3 tumors. Although we have shown that 64Cu-ATSM is a valid PET hypoxia marker in some tumor types, but not for all, this tumor type-dependent hypoxia selectivity of 64Cu-ATSM challenges the use of 64Cu-ATSM as a universal PET hypoxia marker. Further studies are needed to define retention mechanisms for this PET marker.
- 45Liu, Z.; Li, Z. B.; Cao, Q.; Liu, S.; Wang, F.; Chen, X. Small-animal PET of tumors with (64)Cu-labeled RGD–bombesin heterodimer J. Nucl. Med. 2009, 50, 1168– 1177Google ScholarThere is no corresponding record for this reference.
- 46Dumont, R. A.; Deininger, F.; Haubner, R.; Maecke, H. R.; Weber, W. A.; Fani, M. Novel (64)Cu- and (68)Ga-labeled RGD conjugates show improved PET imaging of alpha(nu)beta(3) integrin expression and facile radiosynthesis J. Nucl. Med. 2011, 52, 1276– 1284Google ScholarThere is no corresponding record for this reference.
- 47Fani, M.; Del Pozzo, L.; Abiraj, K.; Mansi, R.; Tamma, M. L.; Cescato, R.; Waser, B.; Weber, W. A.; Reubi, J. C.; Maecke, H. R. PET of somatostatin receptor-positive tumors using 64Cu– and 68Ga–somatostatin antagonists: the chelate makes the difference J. Nucl. Med. 2011, 52, 1110– 1118Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvVKltro%253D&md5=3225eb0d38a7e33658a8a5168e914497PET of somatostatin receptor-positive tumors using 64Cu- and 68Ga-somatostatin antagonists: the chelate makes the differenceFani, Melpomeni; Del Pozzo, Luigi; Abiraj, Keelara; Mansi, Rosalba; Tamma, Maria Luisa; Cescato, Renzo; Waser, Beatrice; Weber, Wolfgang A.; Reubi, Jean Claude; Maecke, Helmut R.Journal of Nuclear Medicine (2011), 52 (7), 1110-1118CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine)Somatostatin-based radiolabeled peptides have been successfully introduced into the clinic for targeted imaging and radionuclide therapy of somatostatin receptor (sst)-pos. tumors, esp. of subtype 2 (sst2). The clin. used peptides are exclusively agonists. Recently, we showed that radiolabeled antagonists may be preferable to agonists because they showed better pharmacokinetics, including higher tumor uptake. Factors detg. the performance of radioantagonists have only scarcely been studied. Here, we report on the development and evaluation of four 64Cu or 68Ga radioantagonists for PET of sst2-pos. tumors. Methods: The novel antagonist p-Cl-Phe-cyclo(D-Cys-Tyr-D-4-amino-Phe(carbamoyl)-Lys-Thr-Cys)D-Tyr-NH2 (LM3) was coupled to 3 macrocyclic chelators, namely 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (CB-TE2A), 1,4,7-triazacyclononane, 1-glutaric acid-4,7-acetic acid (NODAGA), and DOTA. 64/natCu- and 68/natGa-NODAGA-LM3 were prepd. at room temp., and 64/natCu-CB-TE2A-LM3 and 68/natGa-DOTA-LM3 were prepd. at 95°C. Binding affinity and antagonistic properties were detd. with receptor autoradiog. and immunofluorescence microscopy using human embryonic kidney (HEK)-sst2 cells. In vitro internalization and dissocn. was evaluated using the same cell line. Biodistribution and small-animal PET studies were performed with HEK-sst2 xenografts. Results: All metallopeptides demonstrated antagonistic properties. The affinities depend on chelator and radiometal and vary about 10-fold; 68/natGa-NODAGA-LM3 has the lowest half maximal inhibitory concn. (1.3 ± 0.3 nmol/L). The biodistribution studies show impressive tumor uptake at 1 h after injection, particularly of 64Cu- and 68Ga-NODAGA-LM3 (∼40 percentage injected dose per g of tissue [%ID/g]), which were proven to be specific. Background clearance was fast and the tumor washout relatively slow for 64Cu-NODAGA-LM3 (∼15 %ID/g, 24 h after injection) and almost negligible for 64Cu-CB-TE2A-LM3 (26.9 ± 3.3 %ID/g and 21.6 ± 2.1 %ID/g, 4 and 24 h after injection, resp.). Tumor-to-normal-tissue ratios were significantly higher for 64Cu-NODAGA-LM3 than for 64Cu-CB-TE2A-LM3 (tumor-to-kidney, 12.8 ± 3.6 and 1.7 ± 0.3, resp.; tumor-to-muscle, 1,342 ± 115 and 75.2 ± 8.5, resp., at 24 h, P < 0.001). Small-animal PET shows clear tumor localization and high image contrast, esp. for 64Cu- and 68Ga-NODAGA-LM3. Conclusion: This article demonstrates the strong dependence of the affinity and pharmacokinetics of the somatostatin-based radioantagonists on the chelator and radiometal. 64Cu- and 68Ga-NODAGA-LM3 and 64Cu-CB-TE2A-LM3 are promising candidates for clin. translation because of their favorable pharmacokinetics and the high image contrast on PET scans.
- 48Liu, Z.; Li, Z. B.; Cao, Q.; Liu, S.; Wang, F.; Chen, X.; Small-animal, P. E. T. of tumors with (64)Cu-labeled RGD–bombesin heterodimer J. Nucl. Med. 2009, 50, 1168– 1177Google ScholarThere is no corresponding record for this reference.
- 49Rockey, W. M.; Huang, L.; Kloepping, K. C.; Baumhover, N. J.; Giangrande, P. H.; Schultz, M. K. Synthesis and radiolabeling of chelator–RNA aptamer bioconjugates with copper-64 for targeted molecular imaging Bioorg. Med. Chem. 2011, 19, 4080– 4090Google ScholarThere is no corresponding record for this reference.
- 50Hou, G. L.; Li, Y. H.; Zhang, Z. L.; Xiong, Y. H.; Chen, X. F.; Yao, K.; Liu, Z. W.; Han, H.; Qin, Z. K.; Zhou, F. J. A modified technique for neourethral anastomosis in orthotopic neobladder reconstruction Urology 2009, 74, 1145– 1149Google ScholarThere is no corresponding record for this reference.
- 51Cooper, M. S.; Ma, M. T.; Sunassee, K.; Shaw, K. P.; Williams, J. D.; Paul, R. L.; Donnelly, P. S.; Blower, P. J. Comparison of (64)Cu-complexing bifunctional chelators for radioimmunoconjugation: labeling efficiency, specific activity, and in vitro/in vivo stability Bioconjugate Chem. 2012, 23, 1029– 1039Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XkvVyqurg%253D&md5=649071237104b7aaffb6d032acce51daComparison of 64Cu-Complexing Bifunctional Chelators for Radioimmunoconjugation: Labeling Efficiency, Specific Activity, and in Vitro/in Vivo StabilityCooper, Maggie S.; Ma, Michelle T.; Sunassee, Kavitha; Shaw, Karen P.; Williams, Jennifer D.; Paul, Rowena L.; Donnelly, Paul S.; Blower, Philip J.Bioconjugate Chemistry (2012), 23 (5), 1029-1039CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)High radiolabeling efficiency, preferably to high specific activity, and good stability of the radioimmunoconjugate are essential features for a successful immunoconjugate for imaging or therapy. In this study, the radiolabeling efficiency, in vitro stability, and biodistribution of immunoconjugates with eight different bifunctional chelators labeled with 64Cu were compared. The anti-CD20 antibody, rituximab, was conjugated to four macrocyclic bifunctional chelators (p-SCN-Bn-DOTA, p-SCN-Bn-Oxo-DO3A, p-SCN-NOTA, and p-SCN-PCTA), three DTPA derivs. (p-SCN-Bn-DTPA, p-SCN-CHX-A''-DTPA, and ITC-2B3M-DTPA), and a macrobicyclic hexamine (sarcophagine) chelator (sar-CO2H) = (1-NH2-8-NHCO(CH2)3CO2H)sar where sar = sarcophagine = 3,6,10,13,16,19-hexaazabicyclo[6.6.6]icosane. Radiolabeling efficiency under various conditions, in vitro stability in serum at 37 °C, and in vivo biodistribution and imaging in normal mice over 48 h were studied. All chelators except sar-CO2H were conjugated to rituximab by thiourea bond formation with an av. of 4.9 ± 0.9 chelators per antibody mol. Sar-CO2H was conjugated to rituximab by amide bond formation with 0.5 chelators per antibody mol. Efficiencies of 64Cu radiolabeling were dependent on the concn. of immunoconjugate. Notably, the 64Cu-NOTA-rituximab conjugate demonstrated the highest radiochem. yield (95%) under very dil. conditions (31 nM NOTA-rituximab conjugate). Similarly, sar-CO-rituximab, contg. 1/10th the no. of chelators per antibody compared to that of other conjugates, retained high labeling efficiency (98%) at an antibody concn. of 250 nM. In contrast to the radioimmunoconjugates contg. DTPA derivs., which demonstrated poor serum stability, all macrocyclic radioimmunoconjugates were very stable in serum with <6% dissocn. of 64Cu over 48 h. In vivo biodistribution profiles in normal female Balb/C mice were similar for all the macrocyclic radioimmunoconjugates with most of the activity remaining in the blood pool up to 48 h. While all the macrocyclic bifunctional chelators are suitable for mol. imaging using 64Cu-labeled antibody conjugates, NOTA and sar-CO2H show significant advantages over the others in that they can be radiolabeled rapidly at room temp., under dil. conditions, resulting in high specific activity.
- 52Moi, M. K.; Meares, C. F.; McCall, M. J.; Cole, W. C.; DeNardo, S. J. Copper chelates as probes of biological systems: stable copper complexes with a macrocyclic bifunctional chelating agent Anal. Biochem. 1985, 148, 249– 253Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaL2MXltlyjsLs%253D&md5=6f8bb1c6ff5effdf2b3bc2672e025802Copper chelates as probes of biological systems: stable copper complexes with a macrocyclic bifunctional chelating agentMoi, Min K.; Meares, Claude F.; McCall, Michael J.; Cole, William C.; DeNardo, Sally J.Analytical Biochemistry (1985), 148 (1), 249-53CODEN: ANBCA2; ISSN:0003-2697.The synthesis of a new bifunctional metal chelator, 6-(p-bromoacetamidobenzyl)-1,4,8,11-tetraazacyclotetradecane-N,N',N'',N'''-tetraacetic acid (TETA) is described, which can be covalently attached to proteins and which binds Cu stably in human serum under physiol. conditions. 67Cu chelates of p-nitrobenzyl-TETA, p-nitrobenzyl-EDTA, diethylenetriaminepentaacetic acid (DTPA)-N-butylamide, and DTPA, as well as conjugates of Cu chelates with mouse monoclonal antibody lym-1 were also prepd. and their stability in human serum was examd. In contrast to TETA, Cu chelates prepd. from analogs of EDTA or DTPA were not stable to prolonged incubation in human serum and rapidly lost Cu to serum albumin.
- 53Jones-Wilson, T. M.; Deal, K. A.; Anderson, C. J.; McCarthy, D. W.; Kovacs, Z.; Motekaitis, R. J.; Sherry, A. D.; Martell, A. E.; Welch, M. J. The in vivo behavior of copper-64-labeled azamacrocyclic complexes Nucl. Med. Biol. 1998, 25, 523– 530Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXlt1Wktrk%253D&md5=aa5cbc7f7ab5fb08f6659903f14f2fa3The in vivo behavior of copper-64-labeled azamacrocyclic complexesJones-Wilson, Teresa M.; Deal, Kim A.; Anderson, Carolyn J.; McCarthy, Deborah W.; Kovacs, Zoltan; Motekaitis, Ramunas J.; Sherry, A. Dean; Martell, Arthur E.; Welch, Michael J.Nuclear Medicine and Biology (1998), 25 (6), 523-530CODEN: NMBIEO; ISSN:0969-8051. (Elsevier Science Inc.)The use of copper radioisotopes in imaging and therapy applications has created a greater need for bifunctional chelates (BFCs) for complexing copper radioisotopes to biomols. It has been demonstrated that the charge and lipophilicity of the Cu-BFC complex has a significant effect on the in vivo behavior of the radiolabeled Cu-BFC-biomol. conjugate. To evaluate the effects of charge, stability, and macrocyclic backbone size on the biol. behavior of 64Cu complexes, a series of macrocyclic 64Cu complexes have been prepd., and the biodistributions of these agents were evaluated in normal Sprague-Dawley rats. Two macrocyclic backbones, dodecane and tetradecane, were evaluated; cyclen, DOTA, and DO2A were dodecane backbone derivs., and cyclam, TETA, and et-cyclam were tetradecane backbone derivs. The biodistributions of the 64Cu-labeled complexes correlated with differences in the size of the macrocycle backbone and the formal charge of the complex. All compds. showed uptake and clearance through the liver and kidneys; however, the pos. charged 64Cu complexes showed significantly higher uptake in both of these organs than did the neg. charged or neutral complexes. 64Cu-TETA, a neg. charged complex with the tetradecane backbone, had the most efficient clearance by 24 h' postinjection. These data suggest that neg. charged complexes may have more favorable clearance properties when used as BFCs.
- 54Garrison, J. C.; Rold, T. L.; Sieckman, G. L.; Figueroa, S. D.; Volkert, W. A.; Jurisson, S. S.; Hoffman, T. J. In vivo evaluation and small-animal PET/CT of a prostate cancer mouse model using 64Cu bombesin analogs: side-by-side comparison of the CB-TE2A and DOTA chelation systems J. Nucl. Med. 2007, 48, 1327– 1337Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhtVSktbzP&md5=6d3effae9572a90579e6590329a6b375In vivo evaluation and small-animal PET/CT of a prostate cancer mouse model using 64Cu bombesin analogs: side-by-side comparison of the CB-TE2A and DOTA chelation systemsGarrison, Jered C.; Rold, Tammy L.; Sieckman, Gary L.; Figueroa, Said Daibes; Volkert, Wynn A.; Jurisson, Silvia S.; Hoffman, Timothy J.Journal of Nuclear Medicine (2007), 48 (8), 1327-1337CODEN: JNMEAQ; ISSN:0161-5505. (Society of Nuclear Medicine)The BB2 receptor subtype, of the bombesin family of receptors, has been shown to be highly overexpressed in a variety of human tumors, including prostate cancer. Bombesin (BBN), a 14-amino acid peptide, has been shown to target the BB2 receptor with high affinity. 64Cu (half-life = 12.7 h, β+: 18%, Eβ+max = 653 keV; β-: 37%, Eβ-max = 578 keV) is a radioisotope that has clin. potential for application in both diagnostic imaging and radionuclide therapy. Recently, new chelation systems such as 1,4,8,11-tetraazabicyclo[6.6.2]hexadecane-4,11-diacetic acid (CB-TE2A) have been reported to significantly stabilize the 64Cu radiometal in vivo. The increased stability of the 64Cu-CB-TE2A chelate complex has been shown to significantly reduce nontarget retention compared with tetraazamacrocycles such as 1,4,7,10-tetraazacyclodoadecane-N,N',N",N'"-tetraacetic acid (DOTA). The aim of this study was to det. whether the CB-TE2A chelation system could significantly improve the in vivo stability of 64Cu bombesin analogs. The study directly compares 64Cu bombesin analogs using the CB-TE2A and DOTA chelation systems in a prostate cancer xenograft SCID (severely compromised immunodeficient) mouse model. Methods: The CB-TE2A-8-AOC-BBN(7-14)NH2 and DOTA-8-AOC-BBN(7-14) NH2 conjugates were synthesized and radiolabeled with 64Cu. The receptor-binding affinity and internalization profile of each metalated conjugate was evaluated using PC-3 cells. Pharmacokinetic and small-animal PET/CT studies were performed using female SCID mice bearing PC-3 xenografts. Results: In vivo BB2 receptor targeting was confirmed by tumor uptake values of 6.95 ± 2.27 and 4.95 ± 0.91 %ID/g (percentage injected dose per g) at the 15-min time point for the 64Cu-CB-TE2A and 64Cu-DOTA radioconjugates, resp. At the 24-h time point, liver uptake was substantially reduced for the 64Cu-CB-TE2A radioconjugate (0.21 ± 0.06 %ID/g) compared with the 64Cu-DOTA radioconjugate (7.80 ± 1.51 %ID/g). The 64Cu-CB-TE2A-8-AOC-BBN(7-14)NH2 radioconjugate demonstrated significant clearance, 98.60 ± 0.28 %ID, from the mouse at 24 h after injection. In contrast, only 67.84 ± 5.43 %ID of the 64Cu activity was excreted using the 64Cu-DOTA-8-AOC-BBN(7-14)NH2 radioconjugate because of nontarget retention. Conclusion: The pharmacokinetic and small-animal PET/CT studies demonstrate significantly improved nontarget tissue clearance for the 64Cu-CB-TE2A8-AOC-BBN(7-14)NH2. This is attributed to the improved in vivo stability of the 64Cu-CB-TE2A chelate complex as compared with the 64Cu-DOTA chelate complex.
- 55Sun, X.; Wuest, M.; Weisman, G. R.; Wong, E. H.; Reed, D. P.; Boswell, C. A.; Motekaitis, R.; Martell, A. E.; Welch, M. J.; Anderson, C. J. Radiolabeling and in vivo behavior of copper-64-labeled cross-bridged cyclam ligands J. Med. Chem. 2002, 45, 469– 477Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXptFKjs7s%253D&md5=95a2a757a82322ce1c46b6bcb323903bRadiolabeling and in vivo behavior of copper-64-labeled cross-bridged cyclam ligandsSun, Xiankai; Wuest, Melinda; Weisman, Gary R.; Wong, Edward H.; Reed, David P.; Boswell, C. Andrew; Motekaitis, Ramunas; Martell, Arthur E.; Welch, Michael J.; Anderson, Carolyn J.Journal of Medicinal Chemistry (2002), 45 (2), 469-477CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Macrocyclic chelators and their metal complexes have widespread applications in the biomedical sciences, including radiopharmaceutical chem. The use of copper radionuclides in radiopharmaceuticals is increasing. Macrocyclic chelators have been found to have enhanced in vivo stability over acyclic chelators such as EDTA and diethylenetriaminepentaacetic acid (DTPA). The currently used chelators of choice for labeling copper radionuclides to biol. mols. are analogs of TETA (1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid); however, recent reports have demonstrated evidence of in vivo instability of the radio-Cu(II)-TETA complexes. A new class of structurally reinforced macrocycles, the "cross-bridged" cyclam derivs., form highly stable complexes with Cu(II) that are resistant to dissocn. in strong acid. Here, we evaluate a series of 64Cu(II) cross-bridged macrocyclic complexes for biol. stability and in vivo behavior. The ligands evaluated include the parent ligand, 1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (1), and three 4,11-di-pendant arm derivs.: 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (2); 4,11-bis(N,N-diethyl-amidomethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (3); and 4,11-bis(amidoethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (4). Copper-64 formed complexes with ligands 1-4 in high radiochem. yields. The 64Cu-2 complex was neutral, while 64Cu complexes of 1, 3, and 4 were pos. charged. All complexes showed no decompn. in rat serum out to 24 h. Biodistribution expts. in Sprague-Dawley rats indicated that 64Cu-1, -3, and -4 were taken up by the liver and kidney and cleared slowly over 24 h, whereas 64Cu-2 cleared rapidly from all tissues. The rapid clearance of the 64Cu-2 complex from the blood and liver, as well as liver metab. expts. in rats, suggests that it is highly stable in vivo. A bifunctional chelator of 2 is a significant candidate for labeling copper radionuclides to biol. mols. for diagnostic imaging and targeted radiotherapy.
- 56Boswell, C. A.; Sun, X.; Niu, W.; Weisman, G. R.; Wong, E. H.; Rheingold, A. L.; Anderson, C. J. Comparative in vivo stability of copper-64-labeled cross-bridged and conventional tetraazamacrocyclic complexes J. Med. Chem. 2004, 47, 1465– 1474Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtFagtLw%253D&md5=542e8b853716a629b641740f102a48dbComparative in Vivo Stability of Copper-64-Labeled Cross-Bridged and Conventional Tetraazamacrocyclic ComplexesBoswell, C. Andrew; Sun, Xiankai; Niu, Weijun; Weisman, Gary R.; Wong, Edward H.; Rheingold, Arnold L.; Anderson, Carolyn J.Journal of Medicinal Chemistry (2004), 47 (6), 1465-1474CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The increased use of copper radioisotopes in radiopharmaceutical applications has created a need for bifunctional chelators (BFCs) that form stable radiocopper complexes and allow covalent attachment to biol. mols. The chelators most commonly utilized for labeling copper radionuclides to biomols. are analogs of 1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid (TETA); however, recent reports have communicated the instability of the radio-Cu(II)-TETA complexes in vivo. A class of bicyclic tetraazamacrocycles, the ethylene "cross-bridged" cyclam (CB-cyclam) derivs., form highly kinetically stable complexes with Cu(II) and therefore may be less susceptible to transchelation than their nonbridged analogs in vivo. Herein we report results on the relative biol. stabilities and identification of the resulting radiolabeled metabolites of a series of 64Cu-labeled macrocyclic complexes. Metab. studies in normal rat liver have revealed that the 64Cu complex of 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo[6.6.2]hexadecane (64Cu-CB-TE2A) resulted in significantly lower values of protein-assocd. 64Cu than 64Cu-TETA [13 ± 6% vs 75 ± 9% at 4 h]. A similar trend was obsd. for the corresponding cyclen derivs., with the 64Cu complex of 4,10-bis(carboxymethyl)-1,4,7,10-tetraazabicyclo[5.5.2]tetradecane (64Cu-CB-DO2A) undergoing less transchelation than the 64Cu complex of 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (64Cu-DOTA) [61 ± 14% vs 90.3 ± 0.5% protein assocd. 64Cu at 4 h]. These data indicate that the structurally reinforcing cross-bridge enhances in vivo stability by reducing metal loss to protein in both the cyclam and cyclen cross-bridged 64Cu complexes and that 64Cu-CB-TE2A is superior to 64Cu-CB-DO2A in that regard. These findings further suggest that a bifunctional chelator deriv. of CB-TE2A is a highly desirable alternative for labeling copper radionuclides to biol. mols. for diagnostic imaging and targeted radiotherapy.
- 57Sprague, J. E.; Peng, Y.; Sun, X.; Weisman, G. R.; Wong, E. H.; Achilefu, S.; Anderson, C. J. Preparation and biological evaluation of copper-64-labeled tyr3-octreotate using a cross-bridged macrocyclic chelator Clin. Cancer Res. 2004, 10, 8674– 8682Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtFGjsL7F&md5=645d85d633c7cc512392f9ec2c33d68dPreparation and biological evaluation of copper-64-labeled Tyr3-octreotate using a cross-bridged macrocyclic chelatorSprague, Jennifer E.; Peng, Yijie; Sun, Xiankai; Weisman, Gary R.; Wong, Edward H.; Achilefu, Samuel; Anderson, Carolyn J.Clinical Cancer Research (2004), 10 (24), 8674-8682CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Somatostatin receptors (SSTr) are expressed on many neuroendocrine tumors, and several radiotracers have been developed for imaging these types of tumors. For this reason, peptide analogs of somatostatin have been well characterized. Copper-64 (t1/2 = 12.7 h), a positron emitter suitable for positron emission tomog. (PET) imaging, was shown recently to have improved in vivo clearance properties when chelated by the cross-bridged tetraazamacrocycle 4,11-bis(carboxymethyl)-1,4,8,11-tetraazabicyclo(6.6.2)hexadecane (CB-TE2A) compared with 1,4,8,11-tetraazacyclotetradecane-1,4,8,11-tetraacetic acid (TETA). CB-TE2A and TETA were conjugated to the somatostatin analog tyrosine-3-octreotate (Y3-TATE) for evaluation of CB-TE2A as a bifunctional chelator of 64Cu. The in vitro affinity of each compd. for SSTr was detd. using a homologous competitive binding assay. In vivo characteristics of both radiolabeled compds. were examd. in biodistribution and microPET studies of AR42J tumor-bearing rats. Cu-CB-TE2A-Y3-TATE (Kd = 1.7 nmol/L) and Cu-TETA-Y3-TATE (Kd = 0.7 nmol/L) showed similar affinities for AR42J derived SSTr. In biodistribution studies, nonspecific uptake in blood and liver was lower for 64Cu-CB-TE2A-Y3-TATE. Differences increased with time such that, at 4 h, blood uptake was 4.3-fold higher and liver uptake was 2.4-fold higher for 64Cu-TETA-Y3-TATE than for 64Cu-CB-TE2A-Y3-TATE. In addn., 4.4-times greater tumor uptake was detected with 64Cu-CB-TE2A-Y3-TATE than with 64Cu-TETA-Y3-TATE at 4 h postinjection. MicroPET imaging yielded similar results. CB-TE2A appears to be a superior in vivo bifunctional chelator of 64Cu for use in mol. imaging by PET or targeted radiotherapy due to both improved nontarget organ clearance and higher target organ uptake of 64Cu-CB-TE2A-Y3-TATE compared with 64Cu-TETA-Y3-TATE.
- 58Ait-Mohand, S.; Fournier, P.; Dumulon-Perreault, V.; Kiefer, G. E.; Jurek, P.; Ferreira, C. L.; Benard, F.; Guerin, B. Evaluation of 64Cu-labeled bifunctional chelate–bombesin conjugates Bioconjugate Chem. 2011, 22, 1729– 1735Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpsFSrtL8%253D&md5=b36324b077c22659f4525ed8d655e66bEvaluation of 64Cu-Labeled Bifunctional Chelate-Bombesin ConjugatesAit-Mohand, Samia; Fournier, Patrick; Dumulon-Perreault, Veronique; Kiefer, Garry E.; Jurek, Paul; Ferreira, Cara L.; Benard, Francois; Guerin, BrigitteBioconjugate Chemistry (2011), 22 (8), 1729-1735CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Several bifunctional chelates (BFCs) were investigated as carriers of 64Cu for PET imaging. The most widely used chelator for 64Cu labeling of BFCs is DOTA (1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tretraacetic acid), even though this complex exhibits only moderate in vivo stability. In this study, we prepd. a series of alternative chelator-peptide conjugates labeled with 64Cu, measured in vitro receptor binding affinities in human breast cancer T47D cells expressing the gastrin-releasing peptide receptor (GRPR) and compared their in vivo stability in mice. DOTA-, NOTA-(1,4,7-triazacyclononane-1,4,7-triacetic acid), PCTA-(3,6,9,15-tetraazabicyclo[9.3.1]pentadeca-1(15),11,13-triene-3,6,9-triacetic acid), and Oxo-DO3A-(1-oxa-4,7,10-triazacyclododecane-4,7,10-triacetic acid) peptide conjugates were prepd. using H2N-Aoc-[D-Tyr6,βAla11,Thi13,Nle14]bombesin(6-14) (BBN) as a peptide template. The BBN moiety was selected since it binds with high affinity to the GRPR, which is overexpressed on human breast cancer cells. A convenient synthetic approach for the attachment of aniline-BFC to peptides on solid support is also presented. To facilitate the attachment of the aniline-PCTA and aniline-Oxo-DO3A to the peptide via an amide bond, a succinyl spacer was introduced at the N-terminus of BBN. The partially protected aniline-BFC (p-H2N-Bn-PCTA(Ot-Bu)3 or p-H2N-Bn-DO3A(Ot-Bu)3) was then coupled to the resulting N-terminal carboxylic acid preactivated with DEPBT/ClHOBt on resin. After cleavage and purifn., the peptide-conjugates were labeled with 64Cu using [64Cu]Cu(OAc)2 in 0.1 M ammonium acetate buffer at 100 °C for 15 min. Labeling efficacy was >90% for all peptides; Oxo-DO3A-BBN was incubated an addnl. 150 min at 100 °C to achieve this high yield. Specific activities varied from 76 to 101 TBq/mmol. Competition assays on T47D cells showed that all BFC-BBN complexes retained high affinity for the GRPR. All BFC-BBN 64Cu-conjugates were stable for over 20 h when incubated at 37 °C in mouse plasma samples. However, in vivo, only 37% of the 64Cu/Oxo-DO3A complex remained intact after 20 h while the 64Cu/DOTA-BBN complex was completely demetalated. In contrast, both 64Cu/NOTA- and 64Cu/PCTA-BBN conjugates remained stable during the 20 h time period. Our results indicate that it is possible to successfully conjugate aniline-BFC with peptide on solid support. Our data also show that 64Cu-labeled NOTA- and PCTA-BBN peptide conjugates are promising radiotracers for PET imaging of many human cancers overexpressing the GRP receptor.
- 59Ferreira, C. L.; Lamsa, E.; Woods, M.; Duan, Y.; Fernando, P.; Bensimon, C.; Kordos, M.; Guenther, K.; Jurek, P.; Kiefer, G. E. Evaluation of Bifunctional Chelates for the Development of Gallium-Based Radiopharmaceuticals Bioconjugate Chem. 2010, 21, 531– 536Google Scholar59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXit1Wrsr0%253D&md5=11d7e167a99c64067629f0bd4460d9bdEvaluation of Bifunctional Chelates for the Development of Gallium-Based RadiopharmaceuticalsFerreira, Cara L.; Lamsa, Eric; Woods, Michael; Duan, Yin; Fernando, Pasan; Bensimon, Corinne; Kordos, Myra; Guenther, Katharina; Jurek, Paul; Kiefer, Garry E.Bioconjugate Chemistry (2010), 21 (3), 531-536CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Ga radioisotopes, including the generator-produced positron-emitting isotope 68Ga (t1/2 = 68 min), are of increasing interest for the development of new radiopharmaceuticals. Bifunctional chelates (BFCs) that can be efficiently radiolabeled with Ga to yield complexes with good in vivo stability are needed. To this end, we undertook a systematic comparison of four BFCs contg. different chelating moieties: two novel BFCs, p-NO2-Bn-Oxo (1-oxa-4,7,10-triazacyclododecane-4,7,10-triacetic acid) and p-NO2-Bn-PCTA (3,6,9,15-tetraazabicyclo [9.3.1]pentadeca-1(15),11,13-triene-3,6,9-triacetic acid), and two more commonly used BFCs, p-NO2-Bn-DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) and p-NO2-Bn-NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid). Each BFC was compared with respect to radiolabeling conditions, radiochem. yield, stability, and in vivo clearance properties. p-NO2-Bn-PCTA, p-NO2-Bn-Oxo, and p-NO2-Bn-NOTA were all more efficiently radiolabeled with Ga compared to p-NO2-Bn-DOTA. p-NO2-Bn-DOTA required longer reaction time, higher concns. of BFC, or heating to obtain equiv. radiochem. yields. Better stability was obsd. for p-NO2-Bn-NOTA and p-NO2-Bn-PCTA compared to p-NO2-Bn-DOTA and p-NO2-Bn-Oxo, esp. with respect to transmetalation to transferrin. Ga-radiolabled p-NO2-Bn-Oxo was found to be kinetically labile and therefore unstable in vivo. Ga-radiolabeled p-NO2-Bn-NOTA and p-NO2-Bn-PCTA were relatively inert, while Ga-radiolabeled p-NO2-Bn-DOTA had intermediate stability, losing >20% of Ga in less than one hour when incubated with apo-transferrin. Similar stability differences were seen when incubating at pH 2. In vivo PET imaging and biodistribution studies in mice showed that 68Ga-radiolabeled p-NO2-Bn-PCTA, p-NO2-Bn-NOTA, and p-NO2-Bn-DOTA all cleared through the kidneys. While there was no statistical difference in the biodistribution results of 68Ga-radiolabeled p-NO2-Bn-PCTA and p-NO2-Bn-DOTA, 68Ga-radiolabeled p-NO2-Bn-NOTA cleared more rapidly from blood and muscle tissue but retained at up to 5 times higher activity in the kidneys.
- 60Ferreira, C. L.; Yapp, D. T.; Lamsa, E.; Gleave, M.; Bensimon, C.; Jurek, P.; Kiefer, G. E. Evaluation of novel bifunctional chelates for the development of Cu-64-based radiopharmaceuticals Nucl. Med. Biol. 2008, 35, 875– 882Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhsVaisLjI&md5=9013cd14b9030ed7df0ea9a4b33cbc30Evaluation of novel bifunctional chelates for the development of Cu-64-based radiopharmaceuticalsFerreira, Cara L.; Yapp, Donald T.; Lamsa, Eric; Gleave, Martin; Bensimon, Corinne; Jurek, Paul; Kiefer, Garry E.Nuclear Medicine and Biology (2008), 35 (8), 875-882CODEN: NMBIEO; ISSN:0969-8051. (Elsevier Inc.)Background: Currently available bifunctional chelates (BFCs) for attaching Cu-64 to a targeting mol. are limited by either their radiolabeling conditions or in vivo stability. With the goal of identifying highly effective BFCs, we compared the properties of two novel BFCs, 1-oxa-4,7,10-triazacyclododecane-S-5-(4-nitrobenzyl)-4,7,10-triacetic acid (p-NO2-Bn-Oxo) and 3,6,9,15-tetraazabicyclo[9.3.1]pentadeca-1(15),11,13-triene-S-4-(4-nitrobenzyl)-3,6,9-triacetic acid (p-NO2-Bn-PCTA), with the commonly used S-2-(4-nitrobenzyl)-1,4,7,10-tetraazacyclododecanetetraacetic acid (p-NO2-Bn-DOTA). Methods: p-NO2-Bn-DOTA, p-NO2-Bn-Oxo and p-NO2-Bn-PCTA were each radiolabeled with Cu-64 under various conditions to assess the reaction kinetics and robustness of the radiolabeling. Stability of each Cu-64 BFC complex was evaluated at low pH and in serum. Small animal positron emission tomog. imaging and biodistribution studies in mice were undertaken. Results: p-NO2-Bn-Oxo and p-NO2-Bn-PCTA possessed superior reaction kinetics compared to p-NO2-Bn-DOTA under all radiolabeling conditions; >98% radiochem. yields were achieved in <5 min at room temp. even when using near stoichiometric amts. of BFC. Under nonideal conditions, such as low or high pH, high radiochem. yields were still achievable with the novel BFCs. The radiolabeled compds. were stable in serum and at pH 2 for 48 h. The imaging and biodistribution of the Cu-64-radiolabeled BFCs illustrated differences between the BFCs, including preferential clearance via the kidneys for the p-NO2-Bn-PCTA Cu-64 complex. Conclusions: The novel BFCs facilitated efficient Cu-64 radiolabeling under mild conditions to produce stable complexes at potentially high specific activities. These BFCs may find wide utility in the development of Cu-64-based radiopharmaceuticals.
- 61Olszewski, R. T.; Bukhari, N.; Zhou, J.; Kozikowski, A. P.; Wroblewski, J. T.; Shamimi-Noori, S.; Wroblewska, B.; Bzdega, T.; Vicini, S.; Barton, F. B.; Neale, J. H. NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluR J. Neurochem. 2004, 89, 876– 885Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksVyksLg%253D&md5=4ca607a68135b77ae0cd997eed5d2566NAAG peptidase inhibition reduces locomotor activity and some stereotypes in the PCP model of schizophrenia via group II mGluROlszewski, Rafal T.; Bukhari, Noreen; Zhou, Jia; Kozikowski, Alan P.; Wroblewski, Jarda T.; Shamimi-Noori, Susan; Wroblewska, Barbara; Bzdega, Tomasz; Vicini, Stefano; Barton, Franca B.; Neale, Joseph H.Journal of Neurochemistry (2004), 89 (4), 876-885CODEN: JONRA9; ISSN:0022-3042. (Blackwell Publishing Ltd.)Phencyclidine (PCP) administration elicits pos. and neg. symptoms that resemble those of schizophrenia and is widely accepted as a model for the study of this human disorder. Group II metabotropic glutamate receptor (mGluR) agonists have been reported to reduce the behavioral and neurochem. effects of PCP. The peptide neurotransmitter, N-acetylaspartylglutamate (NAAG), is a selective group II agonist. The authors synthesized and characterized a urea-based NAAG analog, ZJ43. This novel compd. is a potent inhibitor of enzymes, glutamate carboxypeptidase II (Ki = 0.8 nM) and III (Ki = 23 nM) that deactivate NAAG following synaptic release. ZJ43 (100 μM) does not directly interact with NMDA receptors or metabotropic glutamate receptors. Administration of ZJ43 significantly reduced PCP-induced motor activation, falling while walking, stereotypic circling behavior, and head movements. To test the hypothesis that this effect of ZJ43 was mediated by increasing the activation of mGluR3 via increased levels of extracellular NAAG, the group II mGluR selective antagonist LY341495 was co-administered with ZJ43 prior to PCP treatment. This antagonist completely reversed the effects of ZJ43. Addnl., LY341495 alone increased PCP-induced motor activity and head movements suggesting that normal levels of NAAG act to moderate the effect of PCP on motor activation via a group II mGluR. These data support the view that NAAG peptidase inhibitors may represent a new therapeutic approach to some of the components of schizophrenia that are modeled by PCP.
- 62Silver, D. A.; Pellicer, I.; Fair, W. R.; Heston, W. D.; Cordon-Cardo, C. Prostate-specific membrane antigen expression in normal and malignant human tissues Clin. Cancer Res. 1997, 3, 81– 95Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADyaK1M%252FjsFamtA%253D%253D&md5=06fb3a1b21fc371a5e42843ed558e5ebProstate-specific membrane antigen expression in normal and malignant human tissuesSilver D A; Pellicer I; Fair W R; Heston W D; Cordon-Cardo CClinical cancer research : an official journal of the American Association for Cancer Research (1997), 3 (1), 81-5 ISSN:1078-0432.Prostate-specific membrane antigen is a type II membrane protein with folate hydrolase activity produced by prostatic epithelium. The expression of this molecule has also been documented in extraprostatic tissues, including small bowel and brain. In the present study, an extensive immunohistochemical analysis was performed on a panel of well-characterized normal and malignant human tissues to further define the pattern of prostate-specific membrane antigen (PSMA) expression. Detectable PSMA levels were identified in prostatic epithelium, duodenal mucosa, and a subset of proximal renal tubules. A subpopulation of neuroendocrine cells in the colonic crypts also exhibited PSMA immunoreactivity. All other normal tissues, including cerebral cortex and cerebellum, had undetectable levels of PSMA. Thirty-three of 35 primary prostate adenocarcinomas and 7 of 8 lymph node metastases displayed tumor cell PSMA immunostaining. Eight of 18 prostate tumors metastatic to bone expressed PSMA. All of the other nonprostatic primary tumors studied had undetectable PSMA levels. However, intense staining was observed in endothelial cells of capillary vessels in peritumoral and endotumoral areas of certain malignancies, including 8 of 17 renal cell carcinomas, 7 of 13 transitional cell carcinomas, and 3 of 19 colon carcinomas. Extraprostatic PSMA expression appears to be highly restricted. Nevertheless, its diverse anatomical distribution implies a broader functional significance than previously suspected. The decrease in PSMA immunoreactivity noted in advanced prostate cancer suggests that expression of this molecule may be linked to the degree of tumor differentiation. The neoexpression of PSMA in endothelial cells of capillary beds in certain tumors may be related to tumor angiogenesis and suggests a potential mechanism for specific targeting of tumor neovasculature.
- 63Prasanphanich, A. F.; Nanda, P. K.; Rold, T. L.; Ma, L.; Lewis, M. R.; Garrison, J. C.; Hoffman, T. J.; Sieckman, G. L.; Figueroa, S. D.; Smith, C. J. [64Cu-NOTA-8-Aoc-BBN(7–14)NH2] targeting vector for positron-emission tomography imaging of gastrin-releasing peptide receptor-expressing tissues Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 12462– 12467Google ScholarThere is no corresponding record for this reference.
- 64Chen, Y.; Pullambhatla, M.; Foss, C. A.; Byun, Y.; Nimmagadda, S.; Senthamizhchelvan, S.; Sgouros, G.; Mease, R. C.; Pomper, M. G. 2-(3-{1-Carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pen tanedioic acid, [18F]DCFPyL, a PSMA-based PET imaging agent for prostate cancer Clin. Cancer Res. 2011, 17, 7645– 7653Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1CqtrnM&md5=dca0465b3b65f7b553a622e40a0c74632-(3-{1-Carboxy-5-[(6-[18F]Fluoro-Pyridine-3-Carbonyl)-Amino]-Pentyl}-Ureido)-Pentanedioic Acid, [18F]DCFPyL, a PSMA-Based PET Imaging Agent for Prostate CancerChen, Ying; Pullambhatla, Mrudula; Foss, Catherine A.; Byun, Youngjoo; Nimmagadda, Sridhar; Senthamizhchelvan, Srinivasan; Sgouros, George; Mease, Ronnie C.; Pomper, Martin G.Clinical Cancer Research (2011), 17 (24), 7645-7653CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)PURPOSE: We have synthesized and evaluated in vivo 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid, [18F]DCFPyL, as a potential imaging agent for the prostate-specific membrane antigen (PSMA). PSMA is upregulated in prostate cancer epithelia and in the neovasculature of most solid tumors. Exptl. Design: [18F]DCFPyL was synthesized in two steps from the p-methoxybenzyl (PMB) protected lys-C(O)-glu urea precursor using 6-[18F]fluoronicotinic acid tetrafluorophenyl ester ([18F]F-Py-TFP) for introduction of 18F. Radiochem. synthesis was followed by biodistribution and imaging with PET in immunocompromised mice using isogenic PSMA PC3 PIP and PSMA- PC3 flu xenograft models. Human radiation dosimetry ests. were calcd. using OLINDA/EXM 1.0. RESULTS: DCFPyL displays a Ki value of 1.1 ± 0.1 nmol/L for PSMA. [18F]DCFPyL was produced in radiochem. yields of 36%-53% (decay cor.) and specific radioactivities of 340-480 Ci/mmol (12.6-17.8 GBq/μmol, n = 3). In an immunocompromised mouse model [18F]DCFPyL clearly delineated PSMA+ PC3 PIP prostate tumor xenografts on imaging with PET. At 2 h postinjection, 39.4 ± 5.4 percent injected dose per g of tissue (%ID/g) was evident within the PSMA+ PC3 PIP tumor, with a ratio of 358:1 of uptake within PSMA+ PC3 PIP to PSMA- PC3 flu tumor placed in the opposite flank. At or after 1 h postinjection, minimal nontarget tissue uptake of [18F]DCFPyL was obsd. The bladder wall is the dose-limiting organ. CONCLUSIONS: These data suggest [18F]DCFPyL as a viable, new positron-emitting imaging agent for PSMA-expressing tissues. Clin Cancer Res; 17(24); 7645-53.
- 65Hillier, S. M.; Maresca, K. P.; Lu, G.; Merkin, R. D.; Marquis, J. C.; Zimmerman, C. N.; Eckelman, W. C.; Joyal, J. L.; Babich, J. W. 99mTc-Labeled Small-Molecule Inhibitors of Prostate-Specific Membrane Antigen for Molecular Imaging of Prostate Cancer J. Nucl. Med. 2013, 54, 1369– 1376Google ScholarThere is no corresponding record for this reference.
- 66Elsasser-Beile, U.; Reischl, G.; Wiehr, S.; Buhler, P.; Wolf, P.; Alt, K.; Shively, J.; Judenhofer, M. S.; Machulla, H. J.; Pichler, B. J. PET imaging of prostate cancer xenografts with a highly specific antibody against the prostate-specific membrane antigen J. Nucl. Med. 2009, 50, 606– 611Google Scholar66https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1MzjsFKmtA%253D%253D&md5=b717818e3336ebe156e782f46f30aaecPET imaging of prostate cancer xenografts with a highly specific antibody against the prostate-specific membrane antigenElsasser-Beile Ursula; Reischl Gerald; Wiehr Stefan; Buhler Patrick; Wolf Philipp; Alt Karen; Shively John; Judenhofer Martin S; Machulla Hans-Jurgen; Pichler Bernd JJournal of nuclear medicine : official publication, Society of Nuclear Medicine (2009), 50 (4), 606-11 ISSN:0161-5505.UNLABELLED: Prostate-specific membrane antigen (PSMA), a transmembrane glycoprotein, is highly expressed by virtually all prostate cancers and is currently the focus of several diagnostic and therapeutic strategies. We have previously reported on the generation of several monoclonal antibodies (mAb) and antibody fragments that recognize and bind with high affinity to the extracellular domain of cell-adherent PSMA. This article reports the in vivo behavior and tumor uptake of the radiolabeled anti-PSMA mAb 3/A12 and its potential as a tracer for PET. METHODS: The mAb 3/A12 was conjugated with the chelating agent 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) and radiolabeled with (64)Cu. Severe combined immunodeficient mice bearing PSMA-positive C4-2 prostate carcinoma xenografts were used for small-animal PET imaging. Mice with PSMA-negative DU 145 tumors served as controls. For PET studies, each animal received 20-30 microg of radiolabeled mAb corresponding to an activity of 7.6-11.5 MBq. Imaging was performed 3, 24, and 48 h after injection. After the last scan, the mice were sacrificed and tracer in vivo biodistribution was measured by gamma-counting. RESULTS: Binding of the mAb 3/A12 on PSMA-expressing C4-2 cells was only minimally influenced by DOTA conjugation. The labeling efficiency using (64)Cu and DOTA-3/A12 was 95.3% +/- 0.3%. The specific activity after (64)Cu labeling was between 327 and 567 MBq/mg. After tracer injection, static small-animal PET images of mice with PSMA-positive tumors revealed a tumor-to-background ratio of 3.3 +/- 1.3 at 3 h, 7.8 +/- 1.4 at 24 h, and 9.6 +/- 2.7 at 48 h. In contrast, no significant tracer uptake occurred in the PSMA-negative DU 145 tumors. These results were confirmed by direct counting of tissues after the final imaging. CONCLUSION: Because of the high and specific uptake of (64)Cu-labeled mAb 3/A12 in PSMA-positive tumors, this ligand represents an excellent candidate for prostate cancer imaging and potentially for radioimmunotherapy.
- 67Alt, K.; Wiehr, S.; Ehrlichmann, W.; Reischl, G.; Wolf, P.; Pichler, B. J.; Elsasser-Beile, U.; Buhler, P. High-resolution animal PET imaging of prostate cancer xenografts with three different 64Cu-labeled antibodies against native cell-adherent PSMA Prostate 2010, 70, 1413– 1421Google ScholarThere is no corresponding record for this reference.
- 68Fischer, G.; Seibold, U.; Schirrmacher, R.; Wangler, B.; Wangler, C. (89)Zr, a radiometal nuclide with high potential for molecular imaging with PET: chemistry, applications and remaining challenges Molecules 2013, 18, 6469– 6490Google ScholarThere is no corresponding record for this reference.
- 69Cheng, Y.; Prusoff, W. H. Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50% inhibition (I50) of an enzymatic reaction Biochem. Pharmacol. 1973, 22, 3099– 3108Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE2cXhtVGgs7c%253D&md5=fe7175ca300d8f4085df87af37f8656cRelation between the inhibition constant K1) and the concentration of inhibitor which causes fifty per cent inhibition (I50) of an enzymic reactionCheng, Yung-Chi; Prusoff, William H.Biochemical Pharmacology (1973), 22 (23), 3099-108CODEN: BCPCA6; ISSN:0006-2952.The Ki and I50 values are equal where the kinetics are non- or uncompetitive, but not where they are competitive. The relation between the 2 values was analyzed for non- and uncompetitive bi- and monosubstrate reactions and for competitive bisubstrate reactions.
Supporting Information
Supporting Information
ARTICLE SECTIONSDetailed spectral data and supporting PET-CT blocking study image. This material is available free of charge via the Internet at http://pubs.acs.org.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.