Subscriber access provided by University Libraries, University of Memphis
Article
Metabolically-stabilized 68Ga-NOTA-bombesin for PET Imaging of prostate cancer and influence of protease inhibitor Phosphoramidon Susan Richter, Melinda Wuest, Cody N. Bergman, Stephanie Krieger, Buck E. Rogers, and Frank Wuest Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.5b00970 • Publication Date (Web): 14 Mar 2016 Downloaded from http://pubs.acs.org on March 22, 2016
Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.
Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.
Page 1 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Metabolically-stabilized 68Ga-NOTA-bombesin for PET Imaging of prostate
2
cancer and influence of protease inhibitor phosphoramidon
3 4 5 6
Susan Richter1, Melinda Wuest1, Cody N. Bergman1, Stephanie Krieger2,
7
Buck E. Rogers2, Frank Wuest1
8 9 10
1
11
Canada
12
2
13
USA
Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB T6G 2X4,
Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63108,
14 15 16 17 18
KEYWORDS:
19
phosphoramidon.
Bombesin, gastrin-releasing peptide (GRP) receptor,
68
Ga, prostate cancer, PET,
20 21 22 23
*Corresponding author: Frank Wuest, Department of Oncology, University of Alberta, 11560 University
24
Avenue, Edmonton, AB T6G 1Z2, Canada. Tel.: +1 (780) 989-8150; Fax: +1 (780) 432-8483. E-mail
25
address:
[email protected].
26
1
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 2 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
ABSTRACT
2
Peptide receptor-based targeted molecular imaging and therapy of cancer is on the current forefront
3
of nuclear medicine preclinical research and clinical practice. The frequent overexpression of gastrin-
4
releasing peptide (GRP) receptors in prostate cancer stimulated the development of radiolabeled
5
bombesin derivatives as high affinity peptide ligands for selective targeting of the GRP receptor. In this
6
study, we have evaluated a novel 68Ga-labeled bombesin derivative for PET imaging of prostate cancer
7
in vivo. In addition, we were interested in testing recently proposed “serve-and-protect” strategy to
8
improve metabolic stability of radiolabeled peptides in vivo and to enhance tumor uptake. GRP
9
receptor targeting peptides NOTA-BBN2 and
nat
Ga-NOTA-BBN2 demonstrated a characteristic
10
antagonistic profile and high binding affinity towards the GRP receptor in PC3 cells (IC50 4.6–8.2 nM).
11
Radiolabeled peptide 68Ga-NOTA-BBN2 was obtained from NOTA-BBN2 in radiochemical yields greater
12
than 62% (decay-corrected). Total synthesis time was 35 min, including purification using solid-phase
13
extraction.
14
peptidases in vivo within the investigated time frame of 60 min. Interestingly, metabolic stability was
15
not further enhanced in the presence of protease inhibitor phosphoramidon. Dynamic PET studies
16
showed high tumor uptake in both, PC3- and LNCaP-bearing BALB/c nude mice (SUV5min >0.6; SUV60min
17
>0.5). Radiotracer 68Ga-NOTA-BBN2 represents a novel radiometal-based bombesin derivative suitable
18
for GRP receptor targeting in PC3 and LNCaP mouse xenografts. Further increase of metabolic stability
19
in vivo and enhanced tumor uptake was not observed upon administration of protease inhibitor
20
phosphoramidon. This led to the conclusion that the recently proposed “serve-and-protect” strategy
21
may not be valid for peptides exhibiting favourable intrinsic metabolic stability in vivo.
68
Ga-NOTA-BBN2 exhibited favorable resistance against metabolic degradation by
22
2
ACS Paragon Plus Environment
Page 3 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
INTRODUCTION
2
Various
3
68
4
neuroendocrine tumors (NETs) through specific targeting of somatostatin receptors (sst2, sst3, and
5
sst5).1 Success of targeting NETs with small radiolabeled peptides for imaging and therapy prompted
6
the development of other peptide-based radiopharmaceuticals for targeting more common
7
malignancies such as prostate cancer.2 Prostate cancer is the most commonly diagnosed form of
8
cancer among men with an estimated 24,000 new cases and 4,100 deaths in Canada in 2015.3
9
A highly promising
68
Ga-octreotide peptide analogues (68Ga-DOTA-TOC,
68
Ga-DOTA-NOC,
68
Ga-DOTA-TATE and
Ga-HA-DOTA-TATE) represent an important breakthrough in the clinical management of patients with
68
Ga-labeled radiotracer for PET imaging of recurrent prostate cancer is 68
Ga-PSMA-HBED-CC.4 Diagnostic
10
peptidometic prostate-specific membrane antigen (PSMA) inhibitor
11
value of 68Ga-PSMA-HBED-CC has been demonstrated in 319 patients so far.4 Gastrin-releasing peptide
12
(GRP) receptors are attractive alternative targets for molecular imaging and peptide receptor
13
radionuclide therapy (PRRT) of prostate cancer. Molecular targeting of GRP receptors in prostate
14
cancer and other human malignancies like breast, colon and small-cell lung cancer (SCLC) is based on
15
elevated overexpression of GRP receptors in cancers compared to their rather low endogenous
16
expression levels in most other tissues and organs.5-7
17
Human prostate cancer cell lines PC3 and LNCaP are suitable models for studying GRP receptor-
18
mediated molecular targeting due to different GRP receptor levels. While PC3 cells express high
19
densities of GRP receptor (47,600 binding sites per cell) representing a late-stage prostate carcinoma,
20
whereas the lymph node metastasis-derived cell line LNCaP is described to possess lower densities of
21
the receptor (100 binding sites per cell) representing an early-stage prostate cancer.8,9 Both prostate
22
cancer cell lines also differ in their hormone sensitivity. PC3 cells are androgen-independent, but
23
LNCaP cells require androgens for growth.
24
Recently, we described metabolically stabilized bombesin analog QWAV-Sar-H-FA01010-Tle-NH2
25
(BBN2) for molecular imaging of GRP receptors in PC3 tumors. Peptide BBN2 was radiolabeled with
26
fluorine-18 via prosthetic group chemistry using [18F]SFB (N-succinimidyl-4-[18F]fluorobenzoate) or
27
glucose analogue [18F]FDG (2-deoxy-2-[18F]fluoro-D-glucose). Radiolabeled BBN2 was studied in PC3
28
tumors, and PET imaging confirmed favorable GRP receptor-specific tumor uptake and
29
radiopharmacological profile as an GRP receptor antagonist.10,11
3
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 4 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Here we report on the synthesis and evaluation of bombesin-derivative BBN2 decorated with
2
macrocyclic chelator NOTA for radiolabeling with gallium-68. Gallium-68 is a short-lived positron
3
emitter with a half-life of 68 min (Eβ+max = 1.9 MeV, β+ = 89%) that is available through a
4
generator (68Ge: half-life of 271 d).12
5
radiochemistry, and 68Ga provides high quality PET images due to its physical properties.13
6
In the past, several
7
examples include
8
NOTA- and
9
aminobenzoyl
68
68
68
68
68
Ge/68Ga-
Ga is compatible with straightforward and reproducible
Ga-labeled bombesins have been described in preclinical studies. Prominent
Ga-NOTA-P2-RM26 (68Ga-NOTA-PEG2-[DPhe6,Sta13,Leu14]-bombesin(6-14))14,
68
Ga-
Ga-NODAGA-MJ9 containing a 4-amino-1-carboxymethyl-piperidine linker or a Gly-4moiety,15
and
68
(68Ga-DOTA-4-amino-1-carboxymethyl-piperidine-
Ga-DOTA-RM2
10
[DPhe6,Sta13,Leu14]-bombesin(6-14).16 Pan et al. described GRP receptor targeting with
11
ATBBN (68Ga-NOTA-DPhe-Gln-Trp-Ala-Val-Gly-His-Leu-NHCH2CH3) in comparison with more hydrophilic
12
68
13
NHCH2CH3).17 GRP receptor agonist AMBA (DO3A-CH2CO-Gly-(4-aminobenzoyl)-bombesin(7-14) amide)
14
was decorated with different chelators such as NOTA and NODAGA and radiolabeled with
15
Radiotracer
16
imaging profile compared to metabolism-based targeting of prostate cancer with 18F-methylcholine.19
17
68
18
DOTA-4-amino-1-carboxymethylpiperidine-DPhe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2 and used in a
19
multicenter study for detection of primary and metastatic prostate carcinoma in 14 patients.20 Safety
20
profile and dosimetry of
21
GRP receptor pan-bombesin derivative
22
bombesin(6-14)amide in patients with gastrointestinal stromal tumors. However, all reported peptide-
23
based radiotracers included various challenges for a successful translation into clinical application. A
24
qualified candidate for clinical translation should possess the following characteristics: (1) high tumor
25
accumulation and retention, which is partially the result of a high affinity and high specific-binding of
26
the radiopeptide probe to its target and sufficient metabolic stability in vivo; (2) high tumor-to-
27
background ratios for favorable image contrast; and (3) fast clearance of radioactivity from non-target
28
organs and tissues to reduce background signal.
Ga-NOTA-
(68Ga-NOTA-Gly-Gly-Gly-Arg-Asp-Asn-DPhe-Gln-Trp-Ala-Val-Gly-His-Leu-
Ga-NOTA-MATBBN
68
68
68
Ga.18
Ga-AMBA was studied in human prostate cancer xenografts showing a superior PET
Ga-labeled bombesin peptides were also used in humans.
68
68
Ga-BAY86-7548 was prepared from
Ga-BAY86-7548 was studied in healthy men.21 Another clinical study used 68
Ga-BZH3 (68Ga-DOTA-PEG2-[DTyr6,βAla11,Thi13,Nle14]-
4
ACS Paragon Plus Environment
Page 5 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Another special challenge with radiolabeled peptides is their limited metabolic stability in vivo.
2
Recently, the de Jong group has introduced a highly promising “serve-and-protect” strategy to increase
3
metabolic stability, bioavailability, prolonged circulation times and tumor-localization of various
4
radiopeptides containing a natural peptide sequence (somatostatin-based peptide
5
SS14, bombesin and gastrin derivatives
6
stabilized truncated CCK-minigastrin-analog
7
neutral endopeptidase (NEP) inhibitor phosphoramidon (PA).23-26
8
These studies discussed inhibition of neutral endopeptidase (NEP, EC 3.4.24.11), a key enzyme within
9
the metabolic degradation of peptides in general and bombesin in particular (cleavage between His12
10
and Leu13 residue)27,28. Inhibition of neutral endopeptidase through natural product protease inhibitor
11
phosphoramidon led to a significantly reduced metabolic degradation of radiopeptides in vivo.
12
The goal of the present study was the radiopharmacological investigation of novel bombesin-derivative
13
68
14
xenografts. The positive experience with our recently reported
15
high metabolic stability and favorable binding affinity towards the GRP receptor prompted us to extend
16
our research activities to the development of a
17
radiotracer for clinical translation. This work also included analysis of metabolic stability of 68Ga-NOTA-
18
BBN2 in vivo with and without co-administration of the endopeptidase inhibitor phosphoramidon to
19
test the “serve-and-protect” concept with intrinsic metabolically stable radiopeptides like 68Ga-NOTA-
20
BBN2 to enhance tumor uptake.
111
177
In-DOTA-PanSB1 and 111
111
In-DOTA-Ala1-
Lu-DOTA-GRP(13-27)) as well as
In-DOTA-MG11 in tumor sites based on co-injection of
Ga-NOTA-BBN2 as suitable radiopeptide for PET imaging of GRP receptors in prostate cancer
68
18
F-labeled BBN2 analogs in terms of
Ga-labeled BBN2 analog to develop a suitable
21
5
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 6 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
MATERIALS AND METHODS
2
Reagents. All chemicals were obtained from Sigma-Aldrich (St. Louis, MO, U.S.A.). Peptide synthesis
3
reagents were purchased from NovaBioChem. Fmoc-amino acid FA01010 ((4R,5S)-Fmoc-4-amino-5-
4
methylheptanoic acid) and linker Fmoc-Ava-OH were obtained from Polypeptide Inc. (San Diego, CA,
5
U.S.A). Stabilized bombesin peptide amide was synthesized via a combination of manual coupling
6
procedures and automated solid-phase peptide synthesis (SPPS) using the Syro I peptide synthesizer
7
(MultiSynTech/Biotage, Charlotte, NC, U.S.A). A 20 mCi (740 MBq) iThemba LABS 68Ge/68Ga-generator
8
from isoSolutions Inc. (Vancouver, B.C., Canada) was used as 68Ga source. Mass spectra were recorded
9
on an AB Sciex Voyager Elite matrix-assisted laser desorption ionisation mass spectrometer time-of-
10
flight (MALDI-MS TOF, AB Sciex, Foster City, CA, U.S.A.). Analytical HPLC was performed on a Shimadzu
11
system (Mandel Scientific, Guelph, ON, Canada) equipped with a DGU-20A5 degasser, a SIL-20A HT
12
autosampler, a LC-20AT pump, a SPD-M20A photo-diode-array detector and a Ramona Raytest
13
radiodetector using a Phenomenex Luna 10u C18(2) 100A, 250 x 4.6 mm column. Semi-preparative
14
HPLC was performed on a Gilson system (Mandel Scientific, Guelph, ON, Canada) with a 321 pump and
15
a 155 dual wavelength detector installed with a Phenomenex Jupiter 10u Proteo 90A, 250 x 10 mm,
16
4.5 µm C18 column. UV absorbance was monitored at a wavelength of 210 and 254 nm. Mobile phase
17
consisted of water/0.2%TFA as solvent A and acetonitrile as solvent B.
18
Human androgen-independent prostate cancer cell line PC3 (American Type Tissue Culture Centre,
19
Manassas, VA, U.S.A) was cultivated in 45% RPMI1640 Dulbecco’s modified Eagle’s medium (DMEM)
20
supplemented with 45% Ham's F-12 and 10% heat-inactivated fetal bovine serum (FBS) from Invitrogen
21
(Life Technologies Inc., Grand Island, NY, U.S.A). Human androgen-dependent prostate cancer cell line
22
LNCaP was obtained from ATCC (American Type Tissue Culture Centre, Manassas, VA, U.S.A). 125I-Tyr4-
23
BBN was obtained from PerkinElmer (Waltham, MA, U.S.A.). Cell-associated radioactivity was
24
measured on a 2480 automatic gamma counter WIZARD2® (PerkinElmer, Waltham, MA, U.S.A.).
25 26
Peptide Synthesis. Bombesin peptide Ava-Gln7-Trp8-Ala9-Val10-Sar11-His12-FA0101013-Tle14-NH2 (BBN2)
27
was synthesized prior to modification with bifunctional chelator pSCN-Bn-NOTA (S-2-(4-
28
Isothiocyanatobenzyl)-1,4,7-triazacyclononane-1,4,7-triacetic acid, Macrocyclics, Dallas, TX, U.S.A.) to
29
provide NOTA-BBN2 for radiolabeling with
68
Ga and the synthesis of non-radioactive reference 6
ACS Paragon Plus Environment
Page 7 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
nat
1
compound
Ga-NOTA-BBN2. Peptide synthesis of BBN2 was performed on an automated peptide
2
synthesizer (Syro I, MultiSynTech/Biotage, Charlotte, NC, U.S.A.) using the Fmoc-orthogonal solid phase
3
peptide synthesis starting from the Rink-amide MBHA resin (loading: 0.6 mmol/g). Amino acid
4
components of BBN2 are tert.-leucine (Tle), (4R,5S)-4-amino-5-methylheptanoic acid (FA01010),
5
histidine (His), sarcosine (Sar), valine (Val), alanine (Ala), tryptophan (Trp), glutamine (Gln) and linker
6
moiety 5-aminovaleric acid (Ava). Detailed description on the synthesis procedure, cleavage conditions
7
and analytical characterization of the bombesin sequence Ava-Gln7-Trp8-Ala9-Val10-Sar11-His12-
8
FA0101013-Tle14-NH2 (BBN2) can be found in a previously published manuscript.9
9 10
Synthesis of labeling precursor NOTA-BBN2. 5 mg (1 eq., 4.7 µmol) of Ava-Gln7-Trp8-Ala9-Val10-Sar11-
11
His12-FA0101013-Tle14-NH2 (BBN2) was dissolved in 200 µL of DMF in a LoBind Eppendorf tube before
12
3.5 mg (1.3 eq., 6.3 µmol) of pSCN-Bn-NOTA in DMF (150 µL) was added. The pH was adjusted to 9 by
13
the addition of 2.5 µL (4 eq., 18.8 µmol) of triethylamine (TEA). The reaction mixture was incubated at
14
50 oC for 3 h before it was subjected to semi-preparative HPLC purification. HPLC purification was
15
performed using a Phenomenex Jupiter 10u Proteo 90A, 250 x 10 mm, 4.5 µm C18 column at a flow
16
rate of 2 mL/min and a gradient of water/0.2% TFA as solvent A and acetonitrile as solvent B: 0-10 min
17
10% B, 25 min 50% B, 30-40 min 80% B, 40-45 min 90% B (tR = 30.8 min). HPLC solvent was reduced
18
under vacuum using a rotary evaporator and lyophilisation gave the chelator-modified peptide NOTA-
19
BBN2 as a white powder (6.2 mg, 4.1 µmol, 87% isolated yield). MW C72H108N18O16S 1512.8, measured
20
MALDI-MS (positive) m/z 1513.6 [M+H]+, 1535.5 [M+Na]+, 1551.5 [M+K]+.
21
Quality control was performed on an analytical Shimadzu HPLC system using a Phenomenex Luna 10u
22
C18(2) 100A, 250 × 4.6 mm column at a constant flow rate of 1 mL/min and the following gradient with
23
water/0.2% TFA as solvent A and acetonitrile as solvent B: 0-3 min 10% B, 10 min 30% B, 17 min 50% B,
24
23 min 70% B, 27-30 min 90% B (tR = 17.3 min, purity >99%).
25 nat
Ga-NOTA-BBN2. 1.5 mg (1 eq., 1.0 µmol) of NOTA-BBN2 was
26
Synthesis of reference compound
27
incubated with an excess (21 eq.) of natGaCl3 as Ga source in 100 mM NH4OAc buffer (pH 5.5). Prior to
28
the reaction with peptide NOTA-BBN2, natGaCl3 was challenged with EDTA each dissolved in 100 µL of
29
100 mM NH4OAc buffer (pH 5.5) for a 5 min incubation time at room temperature. 7
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 8 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
After adjusting the pH to 5.5 with 100 mM NH4OAc buffer the peptide Ga-EDTA mixture was allowed to
2
react for 48 h at ambient temperature. The reaction batch was purified via semi-preparative HPLC
3
performed on a Phenomenex Luna C18(2) 100A, 250 x 10 mm column using a gradient of water/0.2%
4
TFA as solvent A and acetonitrile as solvent B: 0-10 min 20% B, 25 min 50% B, 30-40 min 80% B and a
5
flow rate of 2 mL/min. The fractions containing the Ga-complex were collected, and the solvent was
6
reduced under vacuum using a rotary evaporator. Subsequent lyophilisation gave peptide natGa-NOTA-
7
BBN2 as white powder (1.2 mg, 0.8 µmol, 80% isolated yield). MW C72H105GaN18O16S 1578.7, measured
8
MALDI-MS (positive) m/z 1579.5 [M+H]+, 1601.5 [M+Na]+. Purity of natGa-NOTA-BBN2 was assessed on
9
an analytical Shimadzu HPLC system using a Phenomenex Luna 10u C18(2) 100A, 250 × 4.6 mm column
10
at a constant flow rate of 1 mL/min and the following gradient with water/0.2% TFA as solvent A and
11
acetonitrile as solvent B: 0-3 min 10% B, 10 min 30% B, 17 min 50% B, 23 min 70% B, 27-30 min 90% B
12
(tR = 17.0 min, purity > 96%).
13 14
Competitive Binding Assay. In vitro competitive binding of peptides was analyzed in human prostate
15
adenocarcinoma PC3 cells in triplicate as described before.9 Briefly, determination of the concentration
16
of half-maximum inhibition (IC50 values) was carried out as a competition against
17
(0.05 nM final concentration) using increasing concentrations of NOTA-BBN2 or
18
the range of 80 pM to 10,000 nM. After incubation for 2 h at 4 oC and several washing steps, cells were
19
harvested. Counts per minute (cpm) of cell-associated radioactivity was measured in a Wizard gamma
20
counter, decay-corrected and plotted versus log of peptide concentration to give the typical sigmoidal
21
dose-response curves.
125
I-Tyr4-bombesin
nat
Ga-NOTA-BBN2 in
22 23
Calcium Release Assay. Cells were plated on coverslips (Warner Instruments Cat #64-0701) treated
24
with 2 M HCl and 100 µg/mL Poly-D-lysine at 80% confluence 24 h before imaging. Media was removed
25
and cells were washed twice with calcium imaging buffer (140 mM NaCl, 4 mM KCl, 10 mM HEPES,
26
5 mM glucose, 1.3 mM MgSO4, 2.4 mM CaCl2) and incubated with 1 mL of calcium imaging buffer and
27
10 µL of Fura-2/Pluronic F-127 (Life Technologies Ltd., Invitrogen, Paisley, UK) for 30 min at 37 °C. Cells
28
were then washed 3 times with calcium imaging buffer and incubated with calcium imaging buffer at
29
37 °C for 30 min.
8
ACS Paragon Plus Environment
Page 9 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Cells were then positioned on the stage of an inverted fluorescence microscope and flushed with
2
calcium imaging buffer using a perfusion system. Images were obtained using a ORCA-R2 CCD camera
3
and Aquacosmos software (Hamamatsu Photonics, Japan) with alternating illumination at 340 and 380
4
nm. Fluorescence images were collected at 2 sec intervals through an objective lens (S Flour x 10/0.50
5
numerical aperture, Nikon) and an emission filter (470-550 nm). To test NOTA-BBN2, cells were flushed
6
with control agonist (1 nM Tyr4-BBN(1-14) for 30 sec to 2 min, then flushed with buffer and allowed to
7
return to baseline for 30 min. Cells were then perfused with NOTA-BBN2 (0.1 nM-100 nM) for 2 min
8
and then stimulated with 1 nM Tyr4-BBN(1-14). Cells were then allowed to return to baseline. Regions
9
of interest (ROIs) were selected based on cells with the best response. Changes in intracellular calcium
10
levels were determined by the Fura-2 ratio (F340/380) and changed to [Ca2+] using the formula Y=((R-
11
Rmin)/(Rmax-R)*(F380max/F380min))*Kd and the data were plotted using Prism v.6 (GraphPad Inc.)
12
software.
13 68
14
Radiolabeling of NOTA-BBN2 with Gallium-68. Radionuclide
15
(trace-metal grade) from the
16
Fuerstenfeldbruck, Germany). The high activity fraction of the 68Ga-eluate (2.5-2.7 mL) was collected in
17
the plastic reaction vessel while the rest of radioactivity was dispensed in the 20 mL syringe of the GRP
18
module dispenser unit. 600-900 µL (65-85 MBq) of the 68Ga-generator eluate was removed from the
19
plastic reaction vessel, diluted with 300-500 µL of 4 M NaOAc buffer (pH 9.4) to adjust pH to 5 and
20
reacted with 20 µg of NOTA-BBN2 in 20 µL of DI water for 8 min at room temperature in a LoBind
21
Eppendorf tube.
22
An EDTA challenge step for 10 min at room temperature followed by the addition of 12 µL of 10 mM
23
EDTA. The reaction mixture was diluted with 4 mL of PBS (pH 7.4) before purification via solid-phase
24
extraction (SPE) using a Sep-Pak tC18 Plus cartridge (Waters Corporation, Milford, MA, U.S.A.) (pre-
25
conditioned with 10 mL of MeCN and 10 mL of PBS). The cartridge-trapped
26
with 1.1 mL of EtOH. EtOH was evaporated at 85 oC in a stream of nitrogen before 68Ga-NOTA-BBN2
27
was re-dissolved in 100 µL of saline (0.9% w/v of NaCl). Typically, 22-52 MBq of
28
(radiochemical yield: 62±14% (decay-corrected)) was synthesized within 35±4 min (n=11).
68
Ga was eluted with ~5 mL of 1 N HCl
Ge/68Ga-generator via automated GRP module (Scintomics GmbH,
9
ACS Paragon Plus Environment
68
Ga-peptide was eluted
68
Ga-NOTA-BBN2
Molecular Pharmaceutics
Page 10 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
The 68Ga-labeled peptide was analyzed for radiochemical purity with radio-TLC on RP18 plates using 1
2
M NH4OAc/MeOH 10/90 (Rf = 0.1-0.2) and radio-HPLC using the Shimadzu HPLC system equipped with
3
a Phenomenex Luna 10u C18(2) 100A, 250 x 4.6 mm column at a constant flow rate of 1 mL/min and
4
the following gradient with water/0.2% TFA as solvent A and acetonitrile as solvent B: 0-3 min 10% B,
5
10 min 30% B, 17 min 50% B, 23 min 70% B, 27-30 min 90% B (tR = 17.4 min, radiochemical purity
6
>96%).
7 8
Determination of lipophilicity (68Ga-NOTA-BBN2). Lipophilicity was determined according to the
9
shake-flask method by determining the partition coefficient of the
68
Ga-labelled peptide in n-octanol
10
and PBS buffer (pH 7.4) as aqueous phase.29 The organic and the aqueous phase were pre-saturated
11
24 h before the actual start of the experiment. 500 µL of each layer were added to 2 MBq of
12
NOTA-BBN2 in a LoBind Eppendorf tube and the mixture was mixed vigorously for 3 min. The layers
13
were allowed to separate by centrifugation at 2000 rpm for 5 min. Aliquots of 100 µL were removed
14
from each phase and measured in a Wizard gamma counter (Wallac 1480 Wizard-3, Perkin-Elmer,
15
Woodbridge, Ontario, Canada). Calculated logD7.4 values are expressed as mean±SD from
16
3 experiments each performed in triplicate.
68
Ga-
17 18
Animal studies. All animal studies were carried out according to the guidelines of the Canadian Council
19
on Animal Care (CCAC) and approved by the Cross Cancer Institute Animal-Care Committee. In vivo
20
studies were done using normal BALB/c and male PC3 and LNCaP tumour-bearing BALB/c nude mice
21
(body weight: 20 - 24 g, Charles River Laboratories, Saint-Constant, QC, Canada).
22
For tumor xenografts, about 5-6x106 of PC3 cells in 100 µL of PBS or 20-25x106 of LNCaP cells in 200 µL
23
of PBS/Matrigel (50/50) were injected into the shoulder of male nude BALB/c mice subcutaneously.
24
Androgen-dependency of LNCaP tumors required addition of a 1.5 mg pellet containing
25
dehydroepiandrosterone DHEA (60 day release; Innovative Research of America, Sarasota, FL, U.S.A.),
26
which was implanted subcutaneously into the upper right flank at the same time when LNCaP cells
27
were injected. After 3-5 weeks PC3 tumors and after 6-8 weeks LNCaP tumors reached sizes of ∼ 300 -
28
500 mm3 and were used for the experiments described.
29
10
ACS Paragon Plus Environment
Page 11 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Metabolic stability studies in vivo in the presence and absence of phosphoramidon. For metabolic
2
stability studies in vivo, normal BALB/c mice were anesthetized through inhalation of isoflurane in 40%
3
oxygen/ 60% nitrogen (gas flow 1 L/min) prior to i.v. radiotracer injection via the tail vein. Mice were
4
injected with 12-25 MBq of 68Ga-NOTA-BBN2. For metabolic stability in vivo in the presence of enzyme
5
inhibitor, normal BALB/c mice were co-injected with 300 µg of phosphoramidon (disodium salt, Sigma-
6
Aldrich, USA) in 50 µL of saline (10%EtOH) and the radiopeptide 68Ga-NOTA-BBN2 in 50 µL of saline.
7
Venous blood samples were collected at 5, 15, 30, and 60 min post injection via the mouse tail vein and
8
further processed. Blood cells were separated by centrifugation (13,000 rpm x 5 min). Supernatant was
9
removed and containing proteins were precipitated by addition of 2 volume parts of methanol (2v
10
MeOH/1v sample). Another centrifugation step (13,000 rpm x 5 min) was performed to obtain the
11
plasma in the supernatant. Fractions of blood cells, proteins and plasma were measured in a Wizard
12
gamma counter to determine radioactivity per sample. The clear plasma supernatant was injected onto
13
a Shimadzu HPLC system. The samples were analyzed using a Phenomenex Luna 10u C18(2) 100A, 250
14
x 4.6 mm column at a constant flow rate of 1 mL/min and the following gradient with water/0.2% TFA
15
as solvent A and acetonitrile as solvent B: 0-3 min 10% B, 10 min 30% B, 17 min 50% B, 23 min 70% B,
16
27-30 min 90% B.
17 68
18
Dynamic PET imaging studies. PET imaging of radiopeptide
Ga-NOTA-BBN2 was performed on a
19
INVEON PET/CT scanner (Siemens Preclinical Solutions, Knoxville, U.S.A.). Prior to radiotracer injection,
20
mice were anesthetized through inhalation of isoflurane in 40% oxygen/ 60% nitrogen (gas flow 1
21
L/min), and body temperature was kept constant at 37 oC. Mice were placed in a prone position into
22
the centre of the field of view.
23
A transmission scan for attenuation correction was not acquired. Mice were injected with 4-13 MBq of
24
68
25
For blocking studies, PC3 tumor-bearing BALB/c mice were pre-dosed with 300 µg of NOTA-BBN2 in
26
50 µL of saline 10 min prior to radiotracer injection. For enzyme inhibitor studies, PC3 tumor-bearing
27
BALB/c mice were co-injected with 300 µg of phosphoramidon (disodium salt, Sigma-Aldrich, U.S.A.) in
28
50 µL of saline (10% EtOH) and
29
over 60 min in 3D list mode.
Ga-NOTA-BBN2 in 150 µL of isotonic saline solution (0.9% w/v of NaCl) through a tail vein catheter.
68
Ga-NOTA-BBN2 in 50 µL of saline. Data acquisition was performed
11
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 12 of 37
Molecular Pharmaceutics
1 2 3 1 4 5 2 6 7 3 8 9 4 10 11 5 12 13 6 14 15 7 16 17 8 18 19 9 20 10 21 22 11 23 24 12 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
The dynamic list mode data were sorted into sinograms with 54 time frames (10x2, 8x5, 6x10, 6x20, 8x60, 10x120, 6x300s). The frames were reconstructed using maximum a posteriori (MAP) as reconstruction mode. No correction for partial volume effects was applied. The image files were processed using the ROVER v2.0.51 software (ABX GmbH, Radeberg, Germany). Masks defining 3D regions of interest (ROI) were set, and the ROIs were defined by thresholding. ROIs covered all visible tumor mass of the subcutaneous tumors, and the thresholds were defined by 50% of the maximum radioactivity uptake level. Mean standardized uptake values [SUVmean = (activity/mL tissue)/(injected activity/body weight), mL/g] were calculated for each ROI and time-activity curves (TAC) were generated. All semi-quantified PET data are presented as means ± SEM. Statistical differences were tested by Student's t test and were considered significant for P< 0.05.
12
ACS Paragon Plus Environment
Page 13 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
RESULTS
2
Radiosynthesis of
3
BBN2 functionalized with macrocyclic chelator NOTA (NOTA-BBN2) was prepared in high yields (87%)
4
and chemical purity (>99%) suitable for radiolabeling with gallium-68. Gallium-68 was obtained from a
5
20 mCi (740 MBq)
6
module. However, the labeling reaction of NOTA-BBN2, purification and isolation of 68Ga-NOTA-BBN2
7
was conducted manually. An outline of the radiosynthesis of 68Ga-NOTA-BBN2 is depicted in Figure 1.
68
Ga-NOTA-BBN2 and lipophilicity determination. Stabilized bombesin derivative
68
Ge/68Ga-generator that was eluted remotely controlled via the Scintomics GRP
8
((Figure 1))
9 10 11
NOTA-BBN2 (20 µg) was labeled with the 68Ga eluate adjusted to pH 5 with 4 M sodium actetate buffer
12
at room temperature. The reaction time was 18 min, including the ligand challenge step using acyclic
13
ligand EDTA (ethylenediaminetetraacetic acid) in excess. Labeling was quantitative after an 8 min
14
reaction time and the formed radiolabeled peptide remained stable after ligand challenge with EDTA.
15
Purification of the reaction mixture was performed using solid-phase extraction via a tC18 Plus
16
cartridge.
17
dissolving in saline for subsequent radiopharmaceutical studies.
18
decay-corrected radiochemical yields 62±14% (n=11) with >96% radiochemical purity. The total
19
synthesis time was 35±4 min. As starting activity was typically low (in the range of 60 - 90 MBq),
20
effective specific activity of 4 GBq/µmol was generated considering the present amount of 20 µg of
21
precursor peptide (NOTA-BBN2). Partition coefficient of
22
value of -2.10±0.01 in n-octanol and PBS (pH 7.4) which represents the lipophilicity of the peptide
23
radiotracer.
68
Ga-labeled peptide was eluted with EtOH and isolated by evaporation of EtOH and re-
68
68
Ga-NOTA-BBN2 was obtained in
Ga-NOTA-BBN2 was determined as logD
24 25
In vitro competitive binding assay and Ca2+ release assay. A radiometric competitive binding assay
26
was performed to test the inhibitory potency of natGa-NOTA-BBN2 and labeling precursor NOTA-BBN2
27
GRP receptor-expressing PC3 prostate cancer cells.
13
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 14 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Both peptides competed for binding to GRP receptors in a concentration-dependent manner at 4 oC for
2
2 h against GRP receptor-binding radioligand
3
peptide Tyr-BBN(1-14) served as internal reference.
4
Figure 2 displays generated sigmoidal concentration-response curves and derived IC50 values (half
5
maximum inhibition). Both peptides retained high affinity to the GRP receptor, and their inhibitory
6
potencies were in the same order of magnitude as endogenous-derived ligand Tyr4-BBN(1-14) with an
7
IC50 value of 2.4 nM. natGa-NOTA-BBN2 (IC50 value of 4.6 ± 1.2 nM) binds with somewhat higher affinity
8
to the GRP receptor compared to NOTA-BBN2 (IC50 value of 8.2 ± 0.2 nM).
125
I-Tyr-BBN(1-14) as tracer. GRP receptor-binding
9
((Figure 2))
10 11 12
Figure 3 shows the results for the Fura-2 based calcium release assay. This assay was used to elucidate
13
agonistic or antagonistic pharmacological profile NOTA-BBN2. The assay protocol included flushing of
14
the PC3 cells with control agonist (1 nM Tyr4-BBN(1-14)) for 30 s to 2 min in a first step to stimulate
15
intracellular calcium response (first peak) and re-adjustment to baseline level before cells were
16
perfused with NOTA-BBN2 using different concentrations (0.1 nM, 1 nM, 10 nM, 100 nM). Upon
17
subsequent addition of Tyr4-BBN(1-14), inhibition of calcium response (second peak) was observed at
18
10 and 100 nM of NOTA-BBN2, while a 1 nM concentration only partially inhibited the response. A
19
concentration of 0.1 nM had no effect at all. This result demonstrated that NOTA-BBN2 acts as an
20
antagonist on Tyr4-BBN(1-14) induced intracellular calcium release through GRP receptors. NOTA-
21
BBN2 inhibited the calcium response to 50% at a concentration of approximately 1 nM.
22
((Figure 3))
23 24 68
25
Dynamic PET imaging of
Ga-NOTA-BBN2 in PC3 and LNCaP tumor-bearing mice. Tumor-targeting
26
property of 68Ga-NOTA-BBN2 was studied in two different male prostate cancer xenografts (hormone-
27
dependent LNCaP and hormone-independent PC3) with dynamic PET imaging.
14
ACS Paragon Plus Environment
Page 15 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 4 displays PET images of PC3 and LNCaP tumor-bearing BALB/c mouse at 60 min after injection
2
of
3
tumor and muscle tissue.
68
Ga-NOTA-BBN2 as well as corresponding time-activity-curves (TACs) for radioactivity levels in
4
((Figure 4))
5 6 7
Radioactivity accumulation in PC3 tumor tissue resulted in a SUV of 0.56±0.05 and a SUV of 0.47±0.09
8
(both n=3) in LNCaP tumors after 60 min post injection. Tumor-to-muscle ratios in both xenograft
9
models were relatively high with 5.86±0.37 for PC3 and 5.79±1.02 for LNCaP. The elimination pathway 68
Ga-labeled peptide
68
10
of
Ga-NOTA-BBN2 was observed as predominately via kidneys towards final
11
radioactivity accumulation in the bladder. In addition, a negligible amount of radioactivity accumulated
12
in hepatobiliary organs with minor activity retention in liver and intestine. The visible ‘hot spot’ in close
13
proximity to the liver can be identified as the filled gallbladder.
14 15
Blocking studies with NOTA-BBN2 in PC3 xenografts. Blocking studies were performed to demonstrate
16
specific binding of 68Ga-NOTA-BBN2 to the GRP receptor. Figure 5 shows results of radioactivity uptake
17
in PC3 tumors (and muscle uptake) after injection of 68Ga-NOTA-BBN2 in the presence and absence of
18
300 µg of NOTA-BBN2. Presence of 300 µg of NOTA-BBN2 resulted in significantly reduced tumor
19
uptake at 60 min p.i. which was visible in the PET image as well as in the TACs generated from tumor
20
ROI’s. A 40% blocking effect was demonstrated by measuring the SUV60min of 0.37±0.01 under control
21
conditions and the SUV60min of 0.23±0.01 (n=3) after injection of a pharmacological dose of NOTA-
22
BBN2. Tumor-to-muscle ratio was reduced from 4.22±0.57 to 2.99±0.34 (n=3) under blocking
23
conditions.
24 25
((Figure 5))
26 27
Effect of phosphoramidon on tumor uptake of 68Ga-NOTA-BBN2. Another focus of this study was to
28
investigate the influence of protease inhibitor phosphoramidon on metabolic stability and tumor
29
uptake according to the recently proposed “serve-and-protect” strategy.23
15
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 16 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
68
1
Interestingly, in our study PC3 tumor uptake of
Ga-NOTA-BBN2 after 60 min p.i. was not further
2
increased in the presence of 300 µg phosphoramidon (Figure 6).
3
((Figure 6))
4 5 6
Conversely, analysis of tumor uptake revealed an opposite trend towards a reduced tumor uptake in
7
the presence of the enzyme blocker. Determined SUV values after 60 min p.i. were reduced by ∼25%
8
from 0.50±0.06 (control) to 0.38±0.05 (n=3) under phosphoramidon treatment. This observation was
9
tumor specific since muscle clearance pattern was not impaired. Also the elimination pathway of 68Ga-
10
NOTA-BBN2 remained the same in phosphoramidon treated PC3-BALB/c mice.
11 68
Ga-NOTA-BBN2. In vivo metabolic stability of
12
Effect of phosphoramidon on metabolic stability of
13
68
14
radiopeptide and collection of blood samples at representative time points of 5, 15, 30 and 60 min.
15
Figure 7 summarizes the results of metabolic stability analysis of
16
samples in the presence and absence of 300 µg of phosphoramidon. Approximately 50% of
17
radiopeptide
18
remained stable over the remaining time course of the experiment resulting in ∼ 40% of intact 68Ga-
19
NOTA-BBN2 after 60 min p.i.. Surprisingly, presence of phosphoramidon did not increased plasma
20
stability of 68Ga-NOTA-BBN2 neither after 5 min nor after 60 min p.i.. This finding was in contradiction
21
to observed positive stability enhancing effects from recent literature studies.22 All detected
22
radiometabolites were of more hydrophilic nature than parent compound 68Ga-NOTA-BBN2. No 68Ga-
23
NOTA species was found which was indicative of a stable NOTA chelator-peptide linkage. However,
24
exact chemical nature of detected radiometabolites was not further determined.
Ga-NOTA-BBN2 was studied in normal BALB/c mice by intravenous administration of the
Ga-NOTA-BBN2 in mouse plasma
68
Ga-NOTA-BBN2 was metabolized within the first 10 min, while
25 26
68
((Figure 7))
27 28
16
ACS Paragon Plus Environment
68
Ga-labeled peptide
Page 17 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
DISCUSSION
2
The introduction of radiopeptides as high affinity and specific-targeting radiopharmaceuticals in
3
oncologic nuclear medicine has led to significant improvements and advancements in diagnosis and
4
therapy to enhance cancer patient care. Among many other radiopeptides, current research activities
5
on radiopeptides are directed towards the development of GRP receptor-targeting radiolabeled
6
bombesin derivatives possessing high metabolic stability, high tumor uptake and favorable
7
pharmacological profile like reduced hepatobiliary uptake and low background accumulation in vivo.
8
The present study introduced 68Ga-labeled bombesin derivative 68Ga-NOTA-BBN2 as novel radiotracer
9
for PET imaging of GRP receptors in prostate cancer. Our study described the radiosynthesis and 68
10
radiopharmacological evaluation of
Ga-NOTA-BBN2 in two prostate cancer xenograft models.
11
Moreover, we tested the influence of recently proposed protease inhibitor concept (“serve-and-
12
protect” strategy)23 on 68Ga-NOTA-BBN2 as a radiopeptide with intrinsic metabolic stability.
13
In a first step, in vitro pharmacology studies were performed with novel peptides NOTA-BBN2 and
14
nat
15
range (IC50 = 8.2 nM for NOTA-BBN2 and IC50 = 4.6 nM for
16
high affinity binding to the receptor. High binding affinity to the target (GRP receptor) is required for
17
high tumor uptake and retention of the radiopeptide. Modification with macrocyclic chelator NOTA
18
and the complexation with
19
binding affinities compared to earlier introduced fluorine-containing BBN2 peptides FBz-Ava-BBN2
20
(IC50 = 8.7 nM) and FDG-AOAc-BBN2 (IC50 = 16.5 nM)10,11 using the same competitive binding assay.
21
Furthermore, pharmacology of peptide interaction with Gαq protein-coupled GRP receptor was
22
determined with calcium release assay. Peptide agonists are characterized by internalization of the
23
formed ligand-receptor complex stimulating intracellular Ca2+ release resulting in Ca2+mediated second
24
messenger signaling. Antagonists bind to the receptor, but no internalization of the formed ligand-
25
receptor complex and hence to second messenger signaling cascade occurs. As observed during the
26
Fura-2 based intracellular calcium release assay, NOTA-BBN2 was able to reduce the stimulated
27
calcium signal using increasing concentrations of the peptide, which confirms antagonist properties of
28
studied bombesin derivatives NOTA-BBN2 and natGa-NOTA-BBN2.
Ga-NOTA-BBN2. Binding affinities towards the GRP receptor in PC3 cells were in the low nanomolar
nat
Ga3+ afforded compound
nat
Ga-NOTA-BBN2) which is important for
nat
Ga-NOTA-BBN2 which displayed higher
17
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 18 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
It was shown that GRP receptor antagonists demonstrated superior pharmacokinetic performance
2
compared to agonists based on higher receptor avidities and the absence of internalization giving the
3
benefit of avoiding side effects for the future development of peptide-based radiotherapeutics.15,30
4
Labeling precursor NOTA-BBN2 was prepared by solid-phase peptide synthesis and chelator
5
attachment in solution in high yields of over 80%. Reference compound
6
synthesized for identification purposes of the corresponding radiotracer
7
vitro pharmacology studies. Complexation of NOTA-BBN2 with gallium-68 was achieved using very low
8
peptide amounts of 20 µg, which equals 13 nmol of NOTA-BBN2. The pH of the generator eluate
9
([68Ga]Ga3+ in 1 N HCl) was adjusted with sodium acetate buffer to a pH of 5 to generate a reaction 68
68
nat
Ga-NOTA-BBN2 was
Ga-NOTA-BBN2 and for in
Ga3+ incorporation. Reaction at room temperature gave quantitative
10
medium preferable for
11
incorporation of the radiometal 68Ga3+. The radiosynthesis afforded radiochemically pure 68Ga-NOTA-
12
BBN2 within ∼35 min, in high reproducible radiochemical yields (decay-corrected) of ∼62% after
13
evaporation of EtOH and reformulation in saline. Radiosynthesis procedure is in alignment with the
14
rather short physical half-life of gallium-68 (68 min) and comparable to that of other
15
peptide syntheses described in the literature. A major difference of our described radiosynthesis in this
16
study is the performance of the 68Ga3+ incorporation into NOTA-BBN2 at room temperature compared
17
to the use of elevated temperatures as reported for various 68Ga-complexation reactions with NOTA.14-
18
17,31
19
BBN2 was further decreased to -2.1 compared to lipophilicity of [18F]SFB-labelled BBN2 (logP = +1.22)10
20
and [18F]FDG-labelled BBN2 (logP = -0.73).11
21
The more hydrophilic nature of 68Ga-NOTA-BBN2 directs the radiopeptide towards more favourable in
22
vivo radiopharmacokinetics as demonstrated by the more profound renal elimination route of
23
NOTA-BBN2 with only very little radioactivity in the liver.
24
profile comparable to that of various PEGylated bombesin derivatives 68Ga-NOTA-PEG(n=2,3,4,6)-RM26
25
by Varasteh et al. (logD= -2.2, -2.4, -2.4, -2.5 for n = 2, 3, 4, 6). However, increase of the PEG linker
26
length had only a minor influence on a more favorable pharmacological profile of the radiopeptide.31
27
Dynamic PET studies of
28
results for preclinical molecular imaging of GRP receptors in prostate cancer. LNCaP tumors represent a
29
frequently used human prostate carcinoma model with functional androgen receptor and PSMA
Compared to our previously reported
68
18
F-labeled BBN2 derivatives, lipophilicity of
68
68
Ga-NOTA-
68
Ga-NOTA-
68
Ga-
Ga-NOTA-BBN2 exhibits a lipophilicity
Ga-NOTA-BBN2 in PC3 and LNCaP tumor-bearing mice showed promising
18
ACS Paragon Plus Environment
Page 19 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
expression which is a suitable model for metastatic and androgen-sensitive human prostate cancer.32
2
The small molecule size of
3
radioactivity from the blood and non-target tissues resulting in high tumor-to-muscle ratios of ∼5.8 to
4
5.9 after 60 min p.i.. The PC3 and LNCaP tumor is clearly visible on the left shoulder flank of each
5
animal after administration of radiopeptide 68Ga-NOTA-BBN2.
6
The obtained time-activity-curves are comparable for PC3 and LNCaP tumors with a slightly higher SUV
7
in the case of PC3 tumors. Washout of
8
compared to recently reported [18F]FDG-AOAc-BBN2.11
9
Our PET imaging results were comparable to the in vivo profile of
68
Ga-NOTA-BBN2 and its hydrophilic nature enables fast clearance of
68
Ga-NOTA-BBN2 from tumor tissue was less pronounced
68
Ga-labelled statine-bombesin
10
analog 68Ga-NOTA-MJ9 in PC3 mice.15 Furthermore, by using the two different tumor models for PET
11
imaging experiments, we were able to discuss GRP receptor expression with androgen receptor
12
expression in vivo. Our PET imaging data indicated that
13
independent of hormone receptor status of the tumor. Tumor uptake and tumor-to-muscle ratios are
14
comparable in both xenograft models. The observed slightly lower uptake in LNCaP-xenografts can be
15
attributed to the lower number of binding sites (GRP receptors) per cell in LNCaP cells in comparison to
16
PC3 cells.9 However, our result differed from the study recently described by Mansi et al. The authors
17
observed a 25% decreased tumor-to-muscle ratio in LNCaP compared to PC3 xenografts using GRP
18
receptor antagonist
19
Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2) after 60 min p.i..16
20
Moreover, tumor uptake of 68Ga-NOTA-BBN2 in PC3 BALB/c mice was 2-3 times higher (SUV60min 0.56)
21
compared to our recent results using various 18F-labeled BBN2 derivatives (SUV60min = 0.15 for [18F]FBz-
22
Ava-BBN2 and SUV60min = 0.27 for [18F]FDG-AOAc-BBN2).12 One possible explanation is the observed
23
higher binding affinity of
24
(16.5 nM).11 Excretion of 68Ga-NOTA-BBN2 from the body as determined by PET imaging experiments
25
occurred predominantly via the renal pathway as visible by radioactivity clearance through the kidneys
26
and radioactivity accumulation in the bladder. Only little radioactivity levels were found in the liver and
27
intestines.
68
68
Ga-NOTA-BBN2 uptake seems to be
Ga-RM2 (bombesin-derived, DOTA-4-amino-1-carboxymethyl-piperidine-DPhe-
nat
Ga-NOTA-BBN2 (4.6 nM) compared to FBz-BBN2 (8.7 nM) and FDG-BBN2
19
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 20 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
68
1
Specificity of
Ga-NOTA-BBN2 uptake in PC3 tumors through binding to GRP receptors was
2
demonstrated by blocking studies with NOTA-BBN2 (300 µg dose/animal). Tumor uptake was
3
significantly reduced in animals pre-dosed with NOTA-BBN2 in comparison to control animals.
4
An important possible limitation of using radiopeptides as radiopharmaceuticals for molecular imaging
5
and therapy is their frequently observed low metabolic stability in vivo based on their degradation by
6
various proteases present in the blood plasma and digestive system.
7
This problem has been tackled in the past by means of various chemical modifications (cyclization,
8
formation of pseudo-peptide bonds and peptide bond modification, incorporation of unnatural amino
9
acids). However, this chemistry-driven optimization requires particular fine-tuning as modifications
10
often result in the loss of binding affinity, reduced tumor uptake and unfavourable pharmacokinetics.
11
A simple and highly innovative approach was recently reported by the introduction of the “serve-and-
12
protect” strategy to address radiopeptide degradation through proteases. The “serve-and-protect”
13
concept was particularly focused on inhibition of neutral endopeptidase with natural product inhibitor
14
phosphoramidon. This led to impressive enhancement of in vivo metabolic stability and tumor uptake
15
of various radiopeptides labeled with long-lived radionuclides like In-111 and Lu-177. 23-25
16
Using GRP receptor antagonist
17
presence of phosphoramidon from 12 to 80% after 5 min p.i.. In addition, PC3 tumor uptake increased
18
from ∼4 to 21% ID/g after 4 h p.i. of
19
investigated native and stabilized truncated CCK2-receptor-targeting gastrin peptides labeled with In-
20
111 in the presence and absence of phosphoramidon. Furthermore, they demonstrated in situ
21
inhibition of neutral endopeptidase with phosphoramidon as promising tool to enhance diagnostic
22
efficacy through improved stability and tumor uptake.26
23
In our study, we observed that the majority of the enzymatic degradation of
24
occurs within the first 10 min p.i. and that ∼40% of the radiolabeled peptide remained intact after
25
60 min p.i.. This confirms the intrinsic stability of the BBN2 sequence.10,11 NOTA chelator-peptide
26
linkage via a thiourea bond also remained stable as no 68Ga-NOTA species was found in the HPLC traces
27
of plasma samples. However, co-injection of phosphoramidon with 68Ga-NOTA-BBN2 did not lead to an
28
increase in metabolic stability at any investigated time point resulting in ∼40% of intact radiopeptide
29
68
111
In-PanSB1, Nock et al. found an increase of plasma stability in the
111
In-PanSB1.23 In a very recent publication the same group also
Ga-NOTA-BBN2 after 60 min p.i..
20
ACS Paragon Plus Environment
68
Ga-NOTA-BBN2 itself
Page 21 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Evidently, in our PET study it seems that the application of protease inhibitor phosphoramidon did not
2
have a beneficial effect on a further stabilization of 68Ga-NOTA-BBN2 and hence an enhanced tumor
3
uptake.
4
In contrast, PC3 tumor uptake was lower in the phosphoramidon-treated mice. This is an interesting
5
finding for stabilized
6
uptake of 68Ga-NOTA-BBN2 is not consistent with the proposed “serve-and-protect” concept that has
7
also been successfully applied to stabilized truncated gastrin analogs for targeting cholecystokinin
8
subtype-2 (CCK2) receptor.26
9
In addition, we have also analyzed metabolic stability of
68
Ga-labeled bombesin derivative
68
Ga-NOTA-BBN2. The observed lower tumor
18
F-labeled BBN2 derivative [18F]FDG-AOAc68
10
BBN2 in vivo in the presence and absence of phosphoramidon. As for
11
increase of metabolic stability and tumor uptake was found (data not shown). We assume that no
12
further increase in metabolic stability and tumor uptake occurs due to the intrinsic high metabolic
13
stability of radiopeptides containing the BBN2 motif. This is in agreement with the authors’ report of
14
no measurable effect of phosphoramidon with metabolically-stabilized
15
Moreover, only longer-lived radionuclides like In-111 have been tested for the “serve-and-protect”
16
concept. Radiopeptides labeled with short-lived PET radionuclide such gallium-68 and fluorine-18 may
17
act differently to phosphoramidon inhibiton of proteases. However, this assumption has to be further
18
elucidated in the future.
19
Very recently, an article was published describing
20
bombesin(Sta13-Leu14) derivative, which was also tested towards the "serve-and-protect" concept.36
21
Most experiments presented in the article involved
22
stability data was reported for
23
(which does not possess the natural His-Leu segment) revealed slightly higher tumor uptake (2 fold)
24
upon PA treatment compared to non-treated controls after 60 min p.i.. However, the observed 2-fold
25
increase in tumor uptake in the case of stabilized radiopeptide 68Ga-JMV4168 is lower compared to the
26
data described in the original work describing the "serve-and-protect" concept when a 10 to 15 fold
27
increase in tumor uptake was reported.23
68
177
68
Ga-NOTA-BBN2, no further
Ga-JMV4168 as a
111
68
In-DTPA-octreotide.23
Ga-labeled stabilized
Lu-labeled radiopeptides. No in vivo metabolic
Ga-JMV4168. Biodistribution studies with stabilized
28
21
ACS Paragon Plus Environment
68
Ga-JMV4168
Molecular Pharmaceutics
Page 22 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
68
1
In summary,
Ga-NOTA-BBN2 represents a versatile peptide-based radiotracer to image GRP
2
receptors in prostate cancer in mice. The high intrinsic metabolic stability of the peptide backbone in
3
NOTA-BBN2 holds promise to be used within the theranostic concept by exchanging the diagnostic
4
radionuclide gallium-68 with the therapeutic radionuclide such as lutetium-177 for future peptide-
5
receptor radionuclide therapy of prostate cancer patients.37
6
22
ACS Paragon Plus Environment
Page 23 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
CONCLUSION
2
Gallium-68 labeled radiopeptide 68Ga-NOTA-BBN2 is a metabolically stabilized, antagonistic bombesin
3
analog which shows favorable pharmacokinetics and excellent tumor targeting properties for PET
4
imaging of GRP receptors in hormone dependent and independent prostate cancer models. The use of
5
the “serve-and-protect” concept through protease inhibition with phosphoramidon was applied to
6
radiopeptides labeled with short-lived positron emitters for the first time. However, no further
7
increase of metabolic stability and tumor uptake was found.
8
The possible convenient switch to lutetium-177 for radiotherapy offers the advantage to develop a
9
bombesin-based radiotheranostic probe for translation into the clinic. Clinical translation of bombesin-
10
based radiotheranostics would significantly contribute to the management of prostate cancer patients
11
to improve early diagnosis and therapy.
12 13 14 15 16
AUTHOR INFORMATION
17
Corresponding Author: Dr. Frank Wuest (
[email protected])
18 19 20
ACKNOWLEDGEMENTS
21
F.W. thanks the Dianne and Irving Kipnes Foundation for supporting this work. The authors like to
22
thank Gail Hipperson and Dan McGinn from the Vivarium of the Cross Cancer Institute (CCI) for general
23
animal handling.
24
23
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 24 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
REFERENCES
2
(1) Ambrosini, V.; Fanti S. 68Ga-DOTA-peptides in the diagnosis of NET. PET Clin. 2014, 9(1), 37-42.
3
(2) Jamous, M.; Haberkorn, U.; Mier W. Synthesis of peptide radiopharmaceuticals for the therapy and
4
diagnosis of tumor diseases. Molecules. 2013, 18(3), 3379-409.
5
(3) Canadian Cancer Society’s Advisory Committee on Cancer Statistics. Canadian Cancer Statistics
6
2015. Toronto, ON: Canadian Cancer Society; 2015, pp 17/39.
7
(4) Afshar-Oromieh, A.; Avtzi, E.; Giesel, F.L.; Holland-Letz, T.; Linhart, H.G.; Eder, M.; Eisenhut, M.;
8
Boxler, S.; Hadaschik, B.A.; Kratochwil, C.; Weichert, W.; Kopka, K.; Debus, J.; Haberkorn U. The
9
diagnostic value of PET/CT imaging with
68
Ga-labelled PSMA ligand HBED-CC in the diagnosis if
10
recurrent prostate cancer. Eur. J. Nucl. Med. Mol. Imaging 2015, 42(2), 197.
11
(5) Gonzalez N, Moody TW, Igarashi H, Ito T, Jensen RT. Bombesin-related peptides and their receptors:
12
recent advances in their role in physiology and disease states. Curr. Opin. Endocrinol. Diabetes Obes.
13
2008, 15(1), 58-64.
14
(6) Markwalder, R.; Reubi, J.C. Gastrin-releasing peptide receptors in the human prostate: relation to
15
neoplastic transformation. Cancer Res. 1999, 59(5), 1152-9.
16
(7) Mansi, R.; Fleischmann, A.; Mäcke, H.R.; Reubi, J.C. Targeting GRPR in urological cancers--from basic
17
research to clinical application. Nat. Rev. Urol. 2013, 10(4), 235-44.
18
(8) Maddalena, M.E.; Fox, J.; Chen, J.; Feng, W.; Cagnolini A.; Linder K.E.; Tweedle, M.F.; Nunn, A.D.;
19
Lantry, L.E.
20
prostate cancer models with low GRP-R expression. J. Nucl. Med. 2009, 50, 2017-24.
21
(9) Aprikian, A.G.; Han, K.; Chevalier, S.; Bazinet, M.; Viallet, J. Bombesin specifically induces
22
intracellular calcium mobilization via gastrin-releasing peptide receptors in human prostate cancer
23
cells. J. Mol. Endocrinol. 1996, 16, 297-306.
24
(10) Richter, S.; Wuest, M.; Krieger, S; Rogers, B.E.; Friebe, M.; Bergmann, R.; Wuest, F. Synthesis and
25
radiopharmacological evaluation of a high-affinity and metabolically stabilized
26
analogue for molecular imaging of gastrin-releasing peptide receptor-expressing prostate cancer. Nucl.
27
Med. Biol. 2013, 40, 1025-34.
177
Lu-AMBA Biodistribution, radiotherapeutic efficacy, imaging and autoradiography in
24
ACS Paragon Plus Environment
18
F-labled bombesin
Page 25 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
(11) Richter, S.; Wuest, M.; Bergman C.N.; Way, J.D.; Krieger, S; Rogers, B.E.; Wuest, F. Rerouting of
2
metabolic pathway of
3
26, 201-12.
4
(12) Sanchez-Crespo, A. Comparison of Gallium-68 and Fluorine-18 imaging characteristics in positron
5
emission tomography. Appl. Radiat. Isot. 2013, 76, 55-62.
6
(13) Velikyan, I. 68Ga-based radiopharmaceuticals: Production and application relationship. Molecules
7
2015, 20, 12913-43.
8
(14) Varasteh, Z.; Velikyan, I.; Lindeberg, G.; Sörensen, J.; Larhed, M.; Sandström, M.; Selvaraju, R.K.;
9
Malmberg, J.; Tolmachev, V.; Orlova, A. Synthesis and characterization of a high-affinity NOTA-
10
conjugated bombesin antagonist for GRPR-targeted tumor imaging. Bioconjug. Chem. 2013, 24(7),
11
1144-53.
12
(15) Gourni, E.; Mansi, R.; Jamous, M.; Waser, B.; Smerling, C.; Burian, A.; Buchegger, F.; Reubi, J.C.;
13
Maecke, H.R. N-terminal modifications improve the receptor affinity and pharmacokinetics of
14
radiolabeled peptidic gastrin-releasing peptide receptor antagonists: examples of
15
labeled peptides for PET imaging. J. Nucl. Med. 2014, 55(10), 1719-25.
16
(16) Mansi, R.; Wang, X.; Forrer, F.; Waser, B.; Cescato, R.; Graham, K.; Borkowski, S.; Reubi, J.C.;
17
Maecke, H.R. Development of a potent DOTA-conjugated bombesin antagonist for targeting GRPr-
18
positive tumours. Eur. J. Nucl. Med. Mol. Imaging. 2011, 38(1), 97-107.
19
(17) Pan, D.; Xu, Y.P.; Yang, R.H.; Wang, L.; Chen, F.; Luo, S.; Yang, M.; Yan, Y. A new (68)Ga-labeled BBN
20
peptide with a hydrophilic linker for GRPR-targeted tumor imaging. Amino Acids 2014, 46(6), 1481-9.
21
(18) Asti, M.; Iori, M.; Capponi, P.C.; Atti, G.; Rubagotti, S.; Martin, R.; Brennauer, A.; Müller, M.;
22
Bergmann, R.; Erba, P.A.; Versari, A. Influence of different chelators on the radiochemical properties of
23
a 68-Gallium labelled bombesin analogue. Nucl. Med. Biol. 2014, 41(1), 24-35.
24
(19) Schroeder, R.P.; van Weerden, W.M.; Krenning, E.P.; Bangma, C.H.; Berndsen, S.; Grievink-de Ligt,
25
C.H.; Groen, H.C.; Reneman, S.; de Blois, E.; Breeman, W.A.; de Jong, M. Gastrin-releasing peptide
26
receptor-based targeting using bombesin analogues is superior to metabolism-based targeting using
27
choline for in vivo imaging of human prostate cancer xenografts. Eur J. Nucl. Med. Mol. Imaging 2011,
28
38(7), 1257-66.
18
F-labeled peptides: The influence of prosthetic groups. Bioconj. Chem. 2015,
25
ACS Paragon Plus Environment
68
Ga- and
64
Cu-
Molecular Pharmaceutics
Page 26 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
(20) Kaehkoenen, E.; Jambor, I.; Kemppainen, J.; Lehtioe, K.; Groenroos, T.J.; Kuisma, A.; Luoto, P.;
2
Sipilae H.J.; Tolvanen, T.; Alanen, K.; Silen J.; Kallajoki, M.; Roivainen, A.; Schaefer, N.; Schibli, R.; Dragic
3
M.; Johayem, A.; Valencia, R.; Borkowski, S.; Minn, H. In vivo Imaging of prostate cancer using [68Ga]-
4
labeled bombesin analog BAY86-7548. Clin. Cancer Res. 2013, 19(19), 5434-5442.
5
(21) Roivainen, A.; Kähkönen, E..; Luoto, P.; Borkowski, S.; Hofmann, B.; Jambor, I.; Lehtiö, K.; Rantala,
6
T.; Rottmann, A.; Sipilä, H.; Sparks, R.; Suilamo, S.; Tolvanen, T.; Valencia, R.; Minn, H. Plasma
7
pharmacokinetics, whole-body distribution, metabolism, and radiation dosimetry of
8
antagonist BAY 86-7548 in healthy men. J. Nucl. Med. 2013, 54(6), 867-72.
9
(22) Dimitrakopoulou-Strauss, A.; Hohenberger, P.; Haberkorn, U.; Mäcke, H.R.; Eisenhut, M.; Strauss,
10
L.G. 68Ga-labeled bombesin studies in patients with gastrointestinal stromal tumors: comparison with
11
18
12
(23) Nock, B.A.; Maina, T.; Krenning, E.P.; de Jong, M. “To serve and protect”: Enzyme inhibitors as
13
radiopeptide escorts promote tumor targeting. J. Nucl. Med. 2014, 55, 121-127.
14
(24) Marsouvanidis, P.J.; Melis, M.; de Blois, E.; Breeman, W.A.; Krenning, E.P.; Maina, T.; Nock, B.A.; de
15
Jong, M. In vivo enzyme inhibition improves the targeting of [177Lu]DOTA-GRP(13-27) in GRPR-positive
16
tumors in mice. Cancer Biother. Radiopharm. 2014, 29, 359-67.
17
(25) Kaloudi, A.; Nock, B.A.; Krenning, E.P.; Maina, T.; de Jong, M. Radiolabeled gastrin/CCK analogs in
18
tumor diagnosis - towards higher stability and improved tumor targeting. Q. J. Nucl. Med. Mol. Imaging
19
2015, ahead of print.
20
(26) Kaloudi, A.; Nock, B.A; Lymperis, E.; Sallegger, W.; Krenning, E.P.; de Jong, M.; Maina, T. In vivo
21
inhibition of neutral endopeptidase enhances the diagnostic potential of truncated gastrin
22
radioligands. Nucl. Med. Biol. 2015, 42(11), 824-32.
23
(27) Höhne, A.; Mu, L.; Honer, M.; Schubiger, P.A.; Ametamey, S.M.; Graham, K. et al. Synthesis,
24
labeling, and in vitro and in vivo studies of bombesin peptides modified with silicon-based building
25
blocks. Bioconjug. Chem. 2008, 19, 1871–9.
26
(28) Mu, L.; Honer, M.; Becaud, J.; Martic, M.; Schubiger, P.A.; Ametamey, S.M. et al. In vitro and in
27
vivo characterization of novel
28
Bioconjug Chem. 2010, 21, 1864–71.
68
Ga bombesin
F-FDG. J. Nucl. Med. 2007, 48(8), 1245-50.
18
111
In-
18
F-
F-labeled bombesin analogues for targeting GRPR-positive tumors.
26
ACS Paragon Plus Environment
Page 27 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
(29) Waterhouse, R.N. Determination of lipophilicity and its use as a predictor of blood-brain-barrier
2
penetration of molecular imaging agents. Mol. Imaging Biol. 2003, 5, 376-89.
3
(30) Cescato, R.; Maina, T.; Nock, B.; Nikolopoulou, A.; Charalambidis, D.; Piccand, V.; Reubi, J.C.
4
Bombesin receptor antagonists may be preferable to agonists for tumor targeting. J. Nucl. Med. 2008,
5
49(2), 318-26.
6
(31) Varasteh, Z.; Rosenström, U.; Velikyan, I.; Mitran, B.; Altai, M.; Honarvar, H.; Rosestedt, M.;
7
Lindeberg, G.; Sörensen, J.; Larhed, M.; Tolmachev, V.; Orlova, A. The effect of mini-PEG-based spacer
8
length on binding and pharmacokinetic properties of a 68Ga-labeled NOTA-conjugated antagonistic
9
analog of bombesin. Molecules 2014, 19, 10455-72.
10
(32) Sato, N.; Gleave, M.E.; Bruchovsky, N.; Rennie, P.S.; Beraldi, E.; Sullivan, L.D. A metastatic and
11
androgen-sensitive human prostate cancer model using intraprostatic inoculation of LNCaP cells in
12
SCID mice. Cancer Res. 1997, 57, 1584-9.
13
(33) Varasteh, Z.; Mitran, B.; Rosenström, U.; Velikyan, I.; Rosestedt, M.; Lindeberg, G.; Sörensen, J.;
14
Larhed, M.; Tolmachev, V.; Orlova, A. The effect of macrocyclic chelators on the targeting properties of
15
the 68Ga-labeled gastrin releasing peptide receptor antagonist PEG2-RM26. Nucl. Med. Biol. 2015, 42,
16
446-54.
17
(34) Velikyan, I. Continued rapid growth in
18
Comp. Radiopharm. 2015, 58, 99-121.
19
(35) Velikyan, I. Prospective of ⁶⁸Ga-radiopharmaceutical development. Theranostics 2013, 4, 47-80.
20
(36) Chatalic, K.L.S.; Konijnenberg, M; Nonnekens, J.; de Blois, E.; Hoeben, S.; de Ridder, C.; Brunel, L.;
21
Fehrentz, J.-A.; Martinez, J.; van Gent, D.C.; Nock, B.A.; Maina, T.; van Weerden, W. M.; de Jong, M. In
22
vivo stabilization of a gastrin-releasing peptide receptor antagonist enhances PET imaging and
23
radionuclide therapy of prostate cancer on preclinical studies. Theranostics 2016, 6(1), 104-117.
24
(37) Baum, R.P.; Kulkarni, H.R. THERANOSTICS: From Molecular Imaging Using Ga-68 Labeled Tracers
25
and PET/CT to Personalized Radionuclide Therapy - The Bad Berka Experience. Theranostics 2012, 2(5),
26
437-47.
68
Ga applications: update 2013 to June 2014. J. Labelled
27
27
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 28 of 37
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure legends
2 3
Figure 1. Structure of stabilized bombesin derivative NOTA-BBN2 and radiosynthesis of 68Ga-NOTA-
4
BBN2.
5 nat
6
Figure 2. Binding of NOTA-BBN2 and
Ga-NOTA-BBN2 towards gastrin-releasing peptide (GRP)
7
receptor was determined in a competitive binding assay in prostate cancer cell line PC3 using 125I-Tyr4-
8
BBN as a radioligand. Sigmoidal concentration-response curves and IC50 values obtained from the
9
competitive binding assay are presented on top as mean ± SEM from n=3 (triplicates).
10 11
Figure 3. Results from the intracellular calcium-release assay are shown on the bottom. Increasing
12
concentrations (0.1-100 nM) of NOTA-BBN2 resulted in a reduced intracellular calcium release induced
13
by stimulation with the GRP receptor agonist 125I-Tyr4-BBN.
14 15
Figure 4. Representative PET images (maximum intensity projection) of PC3 tumor (left) and LNCaP
16
tumor-bearing BALB/c mouse (right) 60 min after injection of 68Ga-NOTA-BBN2. Corresponding time-
17
activity curves (TACs) show radioactivity levels in both tumors (PC3: black, LNCaP: gray) compared to
18
muscle tissue as SUV and mean ± SEM from n = 3 experiments.
19 68
20
Figure 5. Representative PET images (maximum intensity projection) of
Ga-NOTA-BBN2 in PC3
21
tumor-bearing BALB/c mouse in the absence (left image) and in the presence (right image) of blocking
22
agent NOTA-BBN2 (300 µg). Time-activity curves (TACs) on the right show the blocking effect in PC3
23
tumor compared to muscle tissue over time as SUV and mean ± SEM from n =3 experiments.
24 25
Figure 6. Effect of protease inhibitor phosphoramidon (PA, 300 µg) on 68Ga-NOTA-BBN2 tumor uptake
26
in PC3 BALB/c nude miceobtained with PET. Left: control and right: in the presence of PA - PET as
27
maximum intensity projections. Corresponding time-activity curves (TACs) on the right show
28
radioactivity levels in PC3 tumors and muscle tissue over time as SUV and mean ± SEM from n = 3-4
29
experiments. 28
ACS Paragon Plus Environment
Page 29 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
68
1
Figure 7. In vivo metabolic stability of
Ga-NOTA-BBN2 in the presence (white bars)and absence of
2
protease inhibitor phosphoramidon (gray bars) over 60 min p.i.. Data was obtained from n = 3
3
experiments and is displayed as percentage of intact 68Ga-labeled peptide (mean±SEM) for every time
4
point.
5 6
29
ACS Paragon Plus Environment
Molecular Pharmaceutics Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 1
2
3 4
30
ACS Paragon Plus Environment
Page 30 of 37
Page 31 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 2
2
3 4
31
ACS Paragon Plus Environment
Molecular Pharmaceutics Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 3
2
3 4
32
ACS Paragon Plus Environment
Page 32 of 37
Page 33 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 4
2
3 4
33
ACS Paragon Plus Environment
Molecular Pharmaceutics Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 5
2
3 4
34
ACS Paragon Plus Environment
Page 34 of 37
Page 35 of 37
Molecular Pharmaceutics
Molecular Pharmaceutics
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
1
Figure 6
2
3 4
35
ACS Paragon Plus Environment
Molecular Pharmaceutics
Page 36 of 37
Molecular Pharmaceutics
1
Figure 7
% of intact Ga-labelled peptide
2
100
68
Ga-NOTA-BBN2 (n = 3)
68
Ga-NOTA-BBN2 + 300 µ g PA (n = 3)
75 50 25
68
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
0 5
15
30
time p.i. [min] 3 4
36
ACS Paragon Plus Environment
60
Page 37 of 37
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
For Table of Contents Use Only
Metabolically-stabilized 68Ga-NOTA-bombesin for PET Imaging of prostate cancer and influence of protease inhibitor phosphoramidon Susan Richter, Melinda Wuest, Cody N. Bergman, Stephanie Krieger, Buck E. Rogers, Frank Wuest
ACS Paragon Plus Environment