U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Molecular Imaging and Contrast Agent Database (MICAD) [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2004-2013.

Cover of Molecular Imaging and Contrast Agent Database (MICAD)

Molecular Imaging and Contrast Agent Database (MICAD) [Internet].

Show details

Lys-cys-cys-tyr-ser-leu-(gly-ser-gly)-1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid-111In

111In-DOTA-(GSG)-KCCYSL

, PhD.

Author Information and Affiliations

Created: ; Last Update: December 12, 2007.

Chemical name: Lys-cys-cys-tyr-ser-leu-(gly-ser-gly)-1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid-111In
Abbreviated name: 111In-DOTA-(GSG)-KCCYSL
Synonym:
Agent Category: Peptide
Target: ErbB-2 extracellular domain
Target Category: Receptor-ligand binding
Method of detection: Single-photon emission computed tomography (SPECT) or planar gamma imaging
Source of signal: 111In
Activation: No
Studies:
  • Checkbox In vitro
  • Checkbox Rodents
.

Background

[PubMed]

The epidermal growth factor (EGF) and its receptor (EGFR) are known to play a role in the neoplastic transformation of cells (1). The EGFR is a tyrosine kinase receptor family that includes four members, EGFR and three other human EGF receptors (HERs) designated as HER2/ErbB2, HER3/ErbB3, and HER4/ErbB4. The four receptors constitute the HER-kinase axis and regulate cellular responses through complex receptor–ligand interactions. The various signal transduction pathways that respond to EGFR activation and mediate cell responses have been described by Brandt et al. (2). The various EGFRs are known to be amplified, mutated, or overexpressed in several cancers and are a target for the development of pharmaceutical agents that inhibit either the receptor or the signal transduction pathway (3). It has also been reported that the detection of circulating ErbB2-positive tumor cells indicates a poor prognosis for the cancer patient (4). A variety of inhibitors, including small molecules, antibodies, vaccines, and gene therapies, have been developed and tested against the EGFRs for the treatment of cancer (5, 6). However, to achieve therapeutic effects the receptor or signal transduction pathway must be involved in maintenance of the malignant phenotype because HER inhibitor therapy does not necessarily result in treatment of a cancer (7).

The use of antibodies alone against EGFR (e.g., cetuximab) and ErbB2 (e.g., trastuzumab) for the treatment of cancer has yielded only moderate results, and it has been observed that antibodies are most useful when used in combination with chemotherapy (8). In some cases cardiotoxicity has been reported in individuals treated with trastuzumab in combination with some chemotherapeutic agents (9). In an effort to develop better and safer anti-cancer drugs, investigators have attempted to use peptides that bind to receptors on tumors to treat the disease (10, 11). The development of peptide pharmaceuticals has also been pursued because, compared to the larger biomolecules such as antibodies, these molecules are easier to synthesize and have low immunogenicity, rapid blood clearance, and a higher uptake in tumor tissue (12). Using bacteriophage technology, a peptide of six amino acids, lys-cys-cys-tyr-ser-leu (KCCYSL), that specifically recognizes and binds the extracellular domain of ErbB2 was identified, and the investigators hypothesized that the peptide could be developed into an imaging and therapeutic agent against cancer cells that overexpress ErbB2 (13). As an extension to the earlier study, the tumor imaging and targeting ability of KCCYSL was evaluated after labeling with radioactive indium (111In). Using the labeled peptide, Kumar et al. performed in vitro and in vivo studies in human breast carcinoma cells that overexpress ErbB2 and in xenografted mice (14).

Synthesis

[PubMed]

The procedure used to identify and characterize KCCYSL from a phage display library as an ErbB2 extracellular domain binding peptide was described in detail by Karasseva et al. (13). The synthesis of 111In-labeled KCCYSL was described by Kumar et al. (14). The peptide was synthesized using solid-phase Fmoc technology on a peptide synthesizer. The bifunctional chelator 1,4,7,10-tetraazacyclododecane-N,N',N'',N'''-tetraacetic acid (DOTA) was linked with KCCYSL through a gly-ser-gly (GSG) spacer placed between the amino terminus of DOTA and the hexapeptide to obtain DOTA-(GSG)-KCCYSL. For use as control, a scrambled peptide version (KYLCSC) was also prepared similarly. The peptides were purified by reverse-phase high-pressure liquid chromatography (RP-HPLC) on a Vydac C18 column. The peptides were freeze-dried and stored at -20°C until required. Electrospray ionization-mass spectrometery was used to confirm the identity of the peptides. Details of how this technique was used to confirm the identity of the peptides were not provided in the publication.

For radiolabeling, the respective peptides were mixed with radioactive [111In]indium chloride in ammonium acetate buffer (pH 5.5) (14). The mixture was incubated at 85°C for 60 min, and the reaction was terminated by the addition of ethylenediamine tetraacetic acid to remove unreacted 111In. The labeled conjugates were then purified by RP-HPLC and flush-dried with nitrogen gas, and the pH was adjusted to 8.0 with sodium phosphate buffer in saline. Tris(2-carboxyethyl)phosphine hydrochloride was then added to the peptide stock solution to prevent oxidation of the cysteine thiols. Although the radiolabeling efficiency of the reaction was reported to be 40–50%, the specific activity, radiochemical yield, and purity of the product was not provided by the investigators (14). The labeled peptide was reported to be stable for 12 h in phosphate-buffered saline and for at least 1 h in mouse serum.

In Vitro Studies: Testing in Cells and Tissues

[PubMed]

The in vitro binding of 111In-DOTA(GSG)-KCCYSL was investigated in MDA-MB-435 cells that express ErbB2 receptors (14). For the same study, K-562 cells, which do not express ErbB2 receptors, were used as negative controls. The cells were incubated with the radiolabeled peptide for various times at 37°C, and binding of the label with MDA-MB-435 cells increased gradually and reached saturation by 2 h. Under the same conditions the K-562 cells bound little radioactivity. Neither cell line bound the scrambled version of the labeled peptide. In competition experiments with MDA-MB-435 cells, using an increasing concentration of cold DOTA(GSG)-KCCYSL, the binding of radioactivity decreased in a concentration-dependent manner, indicating a specificity of the labeled peptide for ErbB2 receptors (14). The 50% inhibitory concentration of the labeled peptide with these cells was determined to be 42.5 ± 2.67 nM.

To determine internalization of bound radioactivity, the MDA-MB-435 cells were incubated with the labeled peptide for different time periods and washed with ice-cold cell-binding medium to remove any unbound radioactivity. The cells were then incubated on ice with acetic acid in saline for 5 min and pelleted by centrifugation. Radioactivity was counted to determine the internalized (cell pellet) and the surface bound fraction (supernatant). The cell surface–associated label increased up to 2 h and ~11% of the total radioactivity was determined to be internalized during this period (P < 0.0001).

Animal Studies

Rodents

[PubMed]

The biodistribution of 111In-DOTA(GSG)-KCCYSL was investigated in severe combined immunodeficient mice that had MDA-MB-435 cells implanted in the shoulder (14). The radioactive peptide was injected into the mice through the tail vein, and the animals were euthanized in groups of three at different time points. The tissues of interest were collected and counted for incorporated radioactivity, and the uptake was expressed as a percentage of the injected radioactivity dose per gram (% ID/g). Distribution of the radiolabeled peptide was determined at various time points between 15 min and 24 h after administration (14). The tumor uptake of radioactivity decreased from 2.12 ± 0.32% ID/g at 30 min to 0.66 ± 0.11% ID/g at 2 h and to 0.33 ± 0.05% ID/g at 4 h. The blood radioactivity level was 3.16 ± 0.33% ID/g at 15 min after the injection and dropped to 0.13 ± 0.03% ID/g at 2 h. This indicated a rapid uptake of radioactivity by the tumor and a clearance from the blood during the same period. The tumor/blood ratio was 5.0 at the end of 2 h. A radioactivity peak of 10.51 ± 0.07% ID/g was observed in the kidneys at 15 min, and this subsequently decreased to 5–7% ID/g for the next 3 h after injection. This was probably because the radioactive peptide and its breakdown products were excreted primarily through the urinary route. A high level of radioactivity (3.18 ± 0.16% ID/g) was also detected in the lungs at 15 min after the injection, but this level decreased to 0.23 ± 0.17% ID/g at 4 h. The investigators suggested that the lungs showed a high amount of radioactivity probably because these organs are highly vascularized and have a high blood perfusion rate. During the same period, little radioactivity was observed in the other vital organs, muscle, or bone (14).

The specificity of tumor radioactivity uptake was evaluated by performing a competition experiment using unlabeled In-DOTA(GSG)-KCCYSL (14). The mice (n = 3) were injected with the unlabeled peptide (~0.07 μM) through the tail vein followed by administration of the labeled homolog 15 min later. The labeled peptide alone was administered to another group of mice (n = 3) to serve as the positive control. Results indicated that uptake of the radioactive peptide was blocked by 50% in tumors of mice injected with the non-radioactive peptide (P = 0.0012). Administration of the non-labeled peptide did not affect the percentage of radioactivity uptake per gram of tissue in the normal organs, including the kidneys and the lungs, of these animals (14). The investigators suggested that these observations indicate that uptake of the radiolabeled peptide by the tumor was receptor-mediated, but the label observed in the kidneys and the lungs was the result of non-specific uptake of the peptide (14).

In vivo single-photon emission computed tomography of mice injected with 111In-DOTA(GSG)-KCCYSL showed that the radioactivity accumulated mainly in the xenografted human breast carcinoma cell tumors and the kidneys (14). Very low radioactivity was detected in the hepatobiliary or other systems of the animals. From these results the investigators concluded that 111In-DOTA(GSG)-KCCYSL was a promising peptide for the imaging of ErbB2-positive tumors.

Other Non-Primate Mammals

[PubMed]

No references are currently available.

Non-Human Primates

[PubMed]

No references are currently available.

Human Studies

[PubMed]

No references are currently available.

NIH Support

The imaging study reported in this chapter was funded by an NIH P50 Grant #CA103130.

References

1.
Gross M.E. , Shazer R.L. , Agus D.B. Targeting the HER-kinase axis in cancer Semin Oncol 200431Suppl 31:9–20. [PubMed: 15052539]
2.
Brandt B. , Meyer-Staeckling S. , Schmidt H. , Agelopoulos K. , Buerger H. Mechanisms of egfr gene transcription modulation: relationship to cancer risk and therapy response. Clin Cancer Res. 2006; 12 (24):7252–60. [PubMed: 17189396]
3.
Smith-Jones P.M. , Solit D.B. , Akhurst T. , Afroze F. , Rosen N. , Larson S.M. Imaging the pharmacodynamics of HER2 degradation in response to Hsp90 inhibitors. Nat Biotechnol. 2004; 22 (6):701–6. [PubMed: 15133471]
4.
Wulfing P. , Borchard J. , Buerger H. , Heidl S. , Zanker K.S. , Kiesel L. , Brandt B. HER2-positive circulating tumor cells indicate poor clinical outcome in stage I to III breast cancer patients. Clin Cancer Res. 2006; 12 (6):1715–20. [PubMed: 16551854]
5.
Dancey J.E. Recent advances of molecular targeted agents: opportunities for imaging. Cancer Biol Ther. 2003; 2 (6):601–9. [PubMed: 14688462]
6.
Hudis C.A. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007; 357 (1):39–51. [PubMed: 17611206]
7.
Bates S.E. , Fojo T. Epidermal growth factor receptor inhibitors: a moving target? Clin Cancer Res. 2005; 11 (20):7203–5. [PubMed: 16243788]
8.
Khalil M.Y. , Grandis J.R. , Shin D.M. Targeting epidermal growth factor receptor: novel therapeutics in the management of cancer. Expert Rev Anticancer Ther. 2003; 3 (3):367–80. [PubMed: 12820779]
9.
Seidman A. , Hudis C. , Pierri M.K. , Shak S. , Paton V. , Ashby M. , Murphy M. , Stewart S.J. , Keefe D. Cardiac dysfunction in the trastuzumab clinical trials experience. J Clin Oncol. 2002; 20 (5):1215–21. [PubMed: 11870163]
10.
Abraham J.M. , Sato F. , Cheng Y. , Paun B. , Kan T. , Olaru A. , Jin Z. , Yang J. , Agarwal R. , David S. , Hamilton J.P. , Ito T. , Mori Y. , Meltzer S.J. Novel Decapeptides that Bind Avidly and Deliver Radioisotope to Colon Cancer Cells. PLoS ONE. 2007; 2 (10):e964. [PMC free article: PMC1978517] [PubMed: 17912343]
11.
Nava-Parada P. , Emens L.A. GV-1001, an injectable telomerase peptide vaccine for the treatment of solid cancers. Curr Opin Mol Ther. 2007; 9 (5):490–7. [PubMed: 17932813]
12.
Landon L.A. , Zou J. , Deutscher S.L. Is phage display technology on target for developing peptide-based cancer drugs? Curr Drug Discov Technol. 2004; 1 (2):113–32. [PubMed: 16472251]
13.
Karasseva N.G. , Glinsky V.V. , Chen N.X. , Komatireddy R. , Quinn T.P. Identification and characterization of peptides that bind human ErbB-2 selected from a bacteriophage display library. J Protein Chem. 2002; 21 (4):287–96. [PubMed: 12168699]
14.
Kumar S.R. , Quinn T.P. , Deutscher S.L. Evaluation of an 111In-Radiolabeled Peptide as a Targeting and Imaging Agent for ErbB-2 Receptor Expressing Breast Carcinomas. Clin Cancer Res. 2007; 13 (20):6070–9. [PubMed: 17947470]

Views

Search MICAD

Limit my Search:


Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...