Gastrin-Releasing Peptide Receptor- and Prostate ... - ACS Publications

Mar 15, 2018 - Thiol-Reactive Bifunctional Chelators for the Creation of Site-Selectively Modified Radioimmunoconjugates with Improved Stability...
21 downloads 4 Views 5MB Size
Article pubs.acs.org/bc

Cite This: Bioconjugate Chem. XXXX, XXX, XXX−XXX

Gastrin-Releasing Peptide Receptor- and Prostate-Specific Membrane Antigen-Specific Ultrasmall Gold Nanoparticles for Characterization and Diagnosis of Prostate Carcinoma via Fluorescence Imaging Marc Pretze,*,† Andreas Hien,‡ Matthias Rad̈ le,‡ Ralf Schirrmacher,§ Carmen Wan̈ gler,∥ and Björn Wan̈ gler*,† †

Molecular Imaging & Radiochemistry, Department of Clinical Radiology and Nuclear Medicine and ∥Biomedical Chemistry, Medical Faculty Mannheim of Heidelberg University, Mannheim 68167, Germany ‡ Institute of Process Control and Innovative Energy Conversion, Mannheim University of Applied Sciences, Mannheim 68163, Germany § Oncologic Imaging, Department of Oncology, University of Alberta, Edmonton 6820, Alberta, Canada S Supporting Information *

ABSTRACT: Gold nanoparticles (AuNPs) have widely been used for 70 years in cancer treatment, but only in the last 15 years has the focus been on specific AuNPs with homogeneous size and shape for various areas in science. They constitute a perfect platform for multifunctionalization and therefore enable the enhancement of target affinity. Here we report on the development of tumor specific AuNPs as diagnostic tools intended for the detection of prostate cancer via fluorescence imaging and positron emission tomography (PET). The AuNPs were further evaluated in vitro and in vivo and exhibited favorable diagnostic properties concerning tumor cell uptake, biodistribution, clearance, and tumor retention.



multimodal imaging techniques26 as well as for theranostic purposes.27−31 Many approaches are based on a phenomenon typically known as enhanced permeability and retention (EPR) due to passive extravasation of nanoparticles across the perforated vasculature of tumors.32 In this work, we report on the functionalization of targetspecific AuNPs for the diagnosis of different prostate cancers via near-infrared (NIR) fluorescence imaging. We show the straightforward synthesis and characterization of modified AuNPs with target-specific peptides (Bombesin (7−14) (BBN(7−14))33,34 and the Lys-urea-Glu motif (LUG)35) binding the gastrin-releasing peptide receptor (GRPR) and the prostate-specific membrane antigen (PSMA), respectively. The different AuNPs were tested for stability, avidity, and cell association, as well as fluorescence imaging in vitro. Furthermore, their biodistribution, tumor enrichment, and imaging in vivo were evaluated.

INTRODUCTION Gold nanoparticles (AuNPs) have been used for nearly 70 years for the therapy1−3 and diagnosis4 of different cancers. Early on, those colloidal solutions were unspecific and consisted of AuNPs of heterogeneous size distribution, restricting their use to interstitial brachytherapeutic applications.5 The development of methods for the synthesis of monodisperse AuNPs6 followed by surface-modification for enhanced stability and homogenization of AuNPs7−9 paved the way for further functionalization.10 The high affinity of sulfur toward gold surfaces and the formation of stable and covalent Au−S bonds11 enables a fast and facile functionalization of AuNPs with thiol-modified (bio)molecules. Although cytotoxic effects are known for citrate-capped AuNPs,12−14 polyethylene glycol-containing (PEG)ylated AuNPs exhibit toxic effects only at high concentrations of >3 g/kg.4,15,16 Furthermore, the PEGylation of the AuNPs leads to a higher bioavailability because the in vivo formation of a protein corona around the AuNP is hindered.17,18 Therefore, more target-specific AuNPs could be developed and labeled with small molecules,19 antibodies,20 peptides,21 natural products,22,23 radionuclides,24 and magnetic resonance imaging (MRI) relevant metals.25 Thus, AuNPs represent a perfect platform for multimerization of targetspecific effectors at their surface and modification for © XXXX American Chemical Society

Received: January 25, 2018 Revised: March 12, 2018 Published: March 15, 2018 A

DOI: 10.1021/acs.bioconjchem.8b00067 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

Figure 1. Synthetic pathway for differently functionalized NIR-AuNPs 4, 7, 8, and NODAGA-AuNPs 10, 11, and 12.



(50 nm) has no impact on the specific tumor-targeting properties of the nanoparticle,38,39 because the big nanoparticles exhibit slower kinetics and therefore the biodistribution is strongly influenced by the nanoparticle itself and not by the tumor vector. The herein reported ultrasmall AuNP (80% confluency using 0.05% trypsin/EDTA solution. In contrast to PC3 and A431 cells, LNCaP cells were grown longer in well-plates before using them in in vitro assays (two days instead of one) and washed more carefully, since they grow less adherent. Bradford assays were performed after uptake, internalization, and avidity experiments. For cell uptake studies, Tris-Mn buffer was used (see SI for detailed recipe). Cells were seeded onto a 24 well plate 2−3 days prior to the experiment to obtain ∼1.6 × 106 cells per well. Cells were incubated with [68Ga]AuNPs (4.3 MBq/well) for a maximum of 5 h at 37 °C. Afterward, cells were washed carefully 3× with PBS and lysed 2× with 1 M NaOH for 10 min at 37 °C. The NaOH fractions were collected and measured in a gamma counter (2470 WIZARD2, PerkinElmer). Internalization Assay. Cells were seeded onto a 12 well plate 2−3 days prior to the experiment to reach ∼2.4 × 106 cells per well. Cells were incubated with [68Ga]AuNPs (4.4 MBq/well) for 4−5 h at 37 °C with and without blocking substance (60 μg BBN(7−14)/well (M = 940 g/mol) or 24 μg EG-LUG (EG = ethylene glycol) 14/well (M = 464 g/mol), ∼1000× excess each). After incubation, the cells were washed 3× with PBS, incubated 2× with glycine buffer (pH 2.8) for 5 min at 37 °C and the supernatant fraction was collected for gamma counting. Finally, the cells were lysed 2× with 1 M NaOH and the NaOH fractions were collected for gamma counting. For the different well sizes the recommended working volumes were used for incubation (2 mL for 12 well plate, 1 mL for 24 well plate). All cell experiments were performed in triplicate. Fluorescence Microscopy. Cells were seeded onto coverslips for 2 days, then washed with PBS and incubated for 24 h at 37 °C in 5% CO2 with the respective media containing different amounts of AuNPs (1−100 μg/mL). Afterward, the cells were washed with PBS and incubated with CellMask Orange-solution (1× working solution) for 15 min at 37 °C. Cells were fixed with 1:1 medium:4% formaldehyde in PBS for 2 min at ambient temperature and then with 4% formaldehyde in PBS for 15 min at ambient temperature. Cells were then washed 3× with PBS and coverslips were prepared onto an object plate with Sytox Green-solution (8.3 μM, 10 μL). Fluorescence microscopy was performed on a Leica TCS SP8 confocal microscope with lasers at λ = 488, 552, and 638 nm. Overlays of microscopies were generated using FIJI software (v1.50e). Avidity Studies. The avidities of the GRPR-specific or PSMA-specific AuNPs were determined similar to Fischer et al.53 In brief, cells were seeded on 12- or 24-well plates for 2 days. 125Iod-Tyr4-Bombesin (125I-BBN) (∼370 kBq, ∼81 GBq/ μmol) was purchased from PerkinElmer and 177Lu-PSMA-617 (∼972 MBq, ∼53.7 GBq/μmol) was obtained from University Medical Center Mainz. 125I-BBN (0.06 nM, 106.7 pg/mL) was added together with the GRPR-specific AuNP derivatives in medium in different concentrations (0.01−60 μg/mL). 177LuPSMA-617 (1 nM, 1.21 ng/mL) was added together with the PSMA-specific AuNP derivatives in medium in different concentrations (0.01−60 μg/mL). Cells were incubated with OptiMEM for 1 h at 37 °C, washed 3× with PBS and lysed 2× with 1 M NaOH. The NaOH fractions were collected and measured in the gamma counter. IC50 values were determined using Origin 8.1 software.



ASSOCIATED CONTENT

S Supporting Information *

The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.bioconjchem.8b00067. Synthesis data, electron microscopy (EM), dynamic light scattering (DLS), thermogravimetric analysis (TGA) measurements, confocal fluorescence microscopies, fluorescence imaging, NMR spectroscopy measurements, and ex vivo and in vivo data (PDF)



AUTHOR INFORMATION

Corresponding Authors

*E-mail: [email protected]. Phone: +49 621 383 6045. *E-mail: [email protected]. Phone: +49 621 383 5594. ORCID

Marc Pretze: 0000-0002-6432-5694 Ralf Schirrmacher: 0000-0002-7098-3036 Author Contributions

The manuscript was written through contributions of all authors. All authors have given approval to the final version of the manuscript. Notes

The authors declare no competing financial interest.



ACKNOWLEDGMENTS This work was supported by the Research Campus M2OLIE funded by the German Federal Ministry of Education and Research (BMBF, Funding Code 13GW0091B and 13GW0091E) and the “Zentrales Innovationsprogramm Mittelstand des Bundesministeriums für Wirtschaft und Energie” (AiF Project GmbH, Funding Code KF2035759AK3 and KF3086202AK3). We would like to thank Tobias Timmermann for measuring NMR spectra and Dr. Uwe Seibold for measuring MALDI-MS spectra and Dr. Mareike Roscher for fruitful discussion concerning the animal experiments. We like to thank Dr. Karsten Richter from the German Cancer Research Center (DKFZ) for measuring the EM. We also want to thank Prof. Dr. Wolfgang Schubert for using the TGA and Prof. Dr. Thorsten Röder for using the DLS at Mannheim University of Applied Sciences. We would like to thank Ms. G

DOI: 10.1021/acs.bioconjchem.8b00067 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry

(19) Shukla, R., Chanda, N., Zambre, A., Upendran, A., Katti, K., Kulkarni, R. R., Nune, S. K., Casteel, S. W., Smith, C. J., Vimal, J., Boote, E., Robertson, J. D., Kan, P., Engelbrecht, H., Watkinson, L. D., Carmack, T. L., Lever, J. R., Cutler, C. S., Caldwell, C., Kannan, R., et al. (2012) Laminin receptor specific therapeutic gold nanoparticles (198AuNP-EGCg) show efficacy in treating prostate cancer. Proc. Natl. Acad. Sci. U. S. A. 109, 12426−12431. (20) Kao, H. W., Lin, Y. Y., Chen, C. C., Chi, K. H., Tien, D. C., Hsia, C. C., Lin, W. J., Chen, F. D., Lin, M. H., and Wang, H. E. (2014) Biological characterization of cetuximab-conjugated gold nanoparticles in a tumor animal model. Nanotechnology 25, 295102. (21) Chanda, N., Kattumuri, V., Shukla, R., Zambre, A., Katti, K., Upendran, A., Kulkarni, R. R., Kan, P., Fent, G. M., Casteel, S. W., Smith, C. J., Boote, E., Robertson, J. D., Cutler, C., Lever, J. R., Katti, K. V., and Kannan, R. (2010) Bombesin functionalized gold nanoparticles show in vitro and in vivo cancer receptor specificity. Proc. Natl. Acad. Sci. U. S. A. 107, 8760−8765. (22) Kattumuri, V., Katti, K., Bhaskaran, S., Boote, E. J., Casteel, S. W., Fent, G. M., Robertson, D. J., Chandrasekhar, M., Kannan, R., and Katti, K. V. (2007) Gum arabic as a phytochemical construct for the stabilization of gold nanoparticles: in vivo pharmacokinetics and X-raycontrast-imaging studies. Small 3, 333−341. (23) Chanda, N., Kan, P., Watkinson, L. D., Shukla, R., Zambre, A., Carmack, T. L., Engelbrecht, H., Lever, J. R., Katti, K., Fent, G. M., Casteel, S. W., Smith, C. J., Miller, W. H., Jurisson, S., Boote, E., Robertson, J. D., Cutler, C., Dobrovolskaia, M., Kannan, R., and Katti, K. V. (2010) Radioactive gold nanoparticles in cancer therapy: therapeutic efficacy studies of GA-198AuNP nanoconstruct in prostate tumor-bearing mice. Nanomedicine (N. Y., NY, U. S.) 6, 201−209. (24) Zhu, J., Chin, J., Wängler, C., Wängler, B., Lennox, R. B., and Schirrmacher, R. (2014) Rapid 18F-labeling and loading of PEGylated gold nanoparticles for in vivo applications. Bioconjugate Chem. 25, 1143−1150. (25) Milne, M., Gobbo, P., McVicar, N., Bartha, R., Workentin, M. S., and Hudson, R. H. E. (2013) Water-soluble gold nanoparticles (AuNP) functionalized with a gadolinium(III) chelate via Michael addition for use as a MRI contrast agent. J. Mater. Chem. B 1, 5628− 5635. (26) Li, W., and Chen, X. (2015) Gold nanoparticles for photoacoustic imaging. Nanomedicine (London, U. K.) 10, 299−320. (27) Kim, M. S., Lee, E. J., Kim, J. W., Chung, U. S., Koh, W. G., Keum, K. C., and Koom, W. S. (2016) Gold nanoparticles enhance anti-tumor effect of radiotherapy to hypoxic tumor. Radiat. Oncol. J. 34, 230−238. (28) Zhang, X. D., Wu, D., Shen, X., Chen, J., Sun, Y. M., Liu, P. X., and Liang, X. J. (2012) Size-dependent radiosensitization of PEGcoated gold nanoparticles for cancer radiation therapy. Biomaterials 33, 6408−6419. (29) Her, S., Jaffray, D. A., and Allen, C. (2017) Gold nanoparticles for applications in cancer radiotherapy: Mechanisms and recent advancements. Adv. Drug Delivery Rev. 109, 84−101. (30) Black, K. C. L., Wang, Y., Luehmann, H. P., Cai, X., Xing, W., Pang, B., Zhao, Y., Cutler, C. S., Wang, L. V., Liu, Y., and Xia, Y. (2014) Radioactive 198Au-Doped Nanostructures with Different Shapes for In Vivo Analyses of Their Biodistribution, Tumor Uptake, and Intratumoral Distribution. ACS Nano 8, 4385−4394. (31) Cui, S., Yin, D., Chen, Y., Di, Y., Chen, H., Ma, Y., Achilefu, S., and Gu, Y. (2013) In vivo targeted deep-tissue photodynamic therapy based on near-infrared light triggered upconversion nanoconstruct. ACS Nano 7, 676−688. (32) Maeda, H., Fang, J., Inutsuka, T., and Kitamoto, Y. (2003) Vascular permeability enhancement in solid tumor: various factors, mechanisms involved and its implications. Int. Immunopharmacol. 3, 319−328. (33) Cornelio, D. B., Roesler, R., and Schwartsmann, G. (2007) Gastrin-releasing peptide receptor as a molecular target in experimental anticancer therapy. Ann. Oncol. 18, 1457−1466. (34) Pretze, M., Hien, A., Roscher, M., Richter, K., Rädle, M., Wängler, C., and Wängler, B. (2017) Efficient modification of GRPR-

Anne-Maria Suhr and Ms. Stephanie Riester for their technical assistance in the in vitro and in vivo experiments.



ABBREVIATIONS AuNP, gold nanoparticle; CT, computer tomography; TEM, transmission electron microscopy; DLS, dynamic light scattering; TGA, thermogravimetric analysis; NMR, nuclear magnetic resonance spectroscopy; LUG, Lys-urea-Glu motif; BBN, bombesin; PSMA, prostate-specific membrane antigen



REFERENCES

(1) Sheppard, C. W., Goodell, J. P. B., and Hahn, P. F. (1947) Colloidal gold containing the radioactive isotope Au198 in the selective internal radiation therapy of diseases of the lymphoid system. J. Lab. Clin. Med. 12, 1437−1441. (2) Flocks, R. H., Kerr, H. D., Elkins, H. B., and Culp, D. (1952) Treatment of carcinoma of the prostate by interstitial radiation with radio-active gold (Au 198): a preliminary report. J. Urol. 68, 510−522. (3) Goldie, H., Watkins, F. B., Powell, C., and Hahn, P. F. (1952) Effect of Colloidal Au198 on the Growth Cycle of Leukemic Cells and on the Survival of Their Host. Cancer Res. 12, 92−99. (4) Hainfeld, J. F., Slatkin, D. N., Focella, T. M., and Smilowitz, H. M. (2006) Gold nanoparticles: a new X-ray contrast agent. Br. J. Radiol. 79, 248−253. (5) Sherman, A. I., and Ter-Pogossian, M. (1953) Lymph-node concentration of radioactive colloidal gold following interstitial injection. Cancer 6, 1238−1240. (6) Frens, G. (1973) Controlled Nucleation for the Regulation of the Particle Size in Monodisperse Gold Suspensions. Nature, Phys. Sci. 241, 20−22. (7) Brust, M., Walker, M., Bethell, D., Schiffrin, D. J., and Whyman, R. (1994) Synthesis of Thiol-derivatised Gold Nanoparticles in a Twophase Liquid-Liquid System. J. Chem. Soc., Chem. Commun. 0, 801− 802. (8) Turkevich, J. (1985) Colloidal Gold. Part I - Historical and preparative aspects, morphology and structure. Gold Bull. 18, 86−91. (9) Waters, C. A., Mills, A. J., Johnson, K. A., and Schiffrin, D. J. (2003) Purification of dodecanethiol derivatised gold nanoparticles. Chem. Commun., 540−541. (10) Bielinska, A., Eichman, J. D., Lee, I., Baker, J. R., Jr., and Balogh, L. (2002) Imaging {Au0-PAMAM} gold-dendrimer nanocomposites in cells. J. Nanopart. Res. 4, 395−403. (11) Häkkinen, H. (2012) The gold-sulfur interface at the nanoscale. Nat. Chem. 4, 443−455. (12) Li, M., Al-Jamal, K. T., Kostarelos, K., and Reineke, J. (2010) Physiologically based pharmacokinetic modeling of nanoparticles. ACS Nano 4, 6303−6317. (13) Pompa, P. P., Vecchio, G., Galeone, A., Brunetti, V., Sabella, S., Maiorano, G., Falqui, A., Bertoni, G., and Cingolani, R. (2011) In vivo toxicity assessment of gold nanoparticles in Drosophila melanogaster. Nano Res. 4, 405−413. (14) Vecchio, G., Galeone, A., Brunetti, V., Maiorano, G., Sabella, S., Cingolani, R., and Pompa, P. P. (2012) Concentration-dependent, size-independent toxicity of citrate capped AuNPs in Drosophila melanogaster. PLoS One 7, e29980. (15) Connor, E. E., Mwamuka, J., Gole, A., Murphy, C. J., and Wyatt, M. D. (2005) Gold nanoparticles are taken up by human cells but do not cause acute cytotoxicity. Small 1, 325−327. (16) Verma, A., Uzun, O., Hu, Y., Hu, Y., Han, H. S., Watson, N., Chen, S., Irvine, D. J., and Stellacci, F. (2008) Surface-structureregulated cell-membrane penetration by monolayer-protected nanoparticles. Nat. Mater. 7, 588−595. (17) Cui, M., Liu, R., Deng, Z., Ge, G., Liu, Y., and Xie, L. (2014) Quantitative study of protein coronas on gold nanoparticles with different surface modifications. Nano Res. 7, 345−352. (18) Dai, Q., Walkey, C., and Chan, W. C. (2014) Polyethylene glycol backfilling mitigates the negative impact of the protein corona on nanoparticle cell targeting. Angew. Chem., Int. Ed. 53, 5093−5096. H

DOI: 10.1021/acs.bioconjchem.8b00067 Bioconjugate Chem. XXXX, XXX, XXX−XXX

Article

Bioconjugate Chemistry specific gold nanoparticles for fluorescence imaging of prostate carcinoma. J. Labelled Compd. Radiopharm. 60, S601. (35) Eder, M., Schäfer, M., Bauder-Wüst, U., Hull, W. E., Wängler, C., Mier, W., Haberkorn, U., and Eisenhut, M. (2012) 68Ga-complex lipophilicity and the targeting property of a urea-based PSMA inhibitor for PET imaging. Bioconjugate Chem. 23, 688−697. (36) Zhu, J., Waengler, C., Lennox, R. B., and Schirrmacher, R. (2012) Preparation of water-soluble maleimide-functionalized 3 nm gold nanoparticles: a new bioconjugation template. Langmuir 28, 5508−5512. (37) Francis, G. L. (2010) Albumin and mammalian cell culture: implications for biotechnology applications. Cytotechnology 62, 1−16. (38) Mier, W., Babich, J., and Haberkorn, U. (2014) Is nano too big? Eur. J. Nucl. Med. Mol. Imaging 41, 4−6. (39) Kiessling, F., Mertens, M. E., Grimm, J., and Lammers, T. (2014) Nanoparticles for imaging: top or flop? Radiology 273, 10−28. (40) Dvorak, H. F., Nagy, J. A., Dvorak, J. T., and Dvorak, A. M. (1988) Identification and characterization of the blood vessels of solid tumors that are leaky to circulating macromolecules. Am. J. Pathol. 133, 95−109. (41) Cai, Q. Y., Yu, P., Besch-Williford, C., Smith, C. J., Sieckman, G. L., Hoffman, T. J., and Ma, L. (2013) Near-infrared fluorescence imaging of gastrin releasing peptide receptor targeting in prostate cancer lymph node metastases. Prostate 73, 842−854. (42) Chen, H., Wan, S., Zhu, F., Wang, C., Cui, S., Du, C., Ma, Y., and Gu, Y. (2014) A fast tumor-targeting near-infrared fluorescent probe based on bombesin analog for in vivo tumor imaging. Contrast Media Mol. Imaging 9, 122−134. (43) Ma, L., Yu, P., Veerendra, B., Rold, T. L., Retzloff, L., Prasanphanich, A., Sieckman, G., Hoffman, T. J., Volkert, W. A., and Smith, C. J. (2007) In vitro and in vivo evaluation of alexa fluor 680bombesin[7−14]NH2 peptide conjugate, a high-affinity fluorescent probe with high selectivity for the gastrinreleasing peptide receptor. Mol. Imaging 6, 7290.2007.00013. (44) Juran, S., Walther, M., Stephan, H., Bergmann, R., Steinbach, J., Kraus, W., Emmerling, F., and Comba, P. (2009) Hexadentate bispidine derivatives as versatile bifunctional chelate agents for copper(II) radioisotopes. Bioconjugate Chem. 20, 347−359. (45) Tang, C., and Pang, Z. (2009) Synthesis of small peptides and their use as bifunctional chelating agents in diagnostic radiopharmaceuticals. Chin. J. Chem. 27, 195−201. (46) Kuchar, M., Pretze, M., Kniess, T., Steinbach, J., Pietzsch, J., and Loser, R. (2012) Site-selective radiolabeling of peptides by 18Ffluorobenzoylation with [18F]SFB in solution and on solid phase: a comparative study. Amino Acids 43, 1431−1443. (47) Licha, K., Riefke, B., Ntziachristos, V., Becker, A., Chance, B., and Semmler, W. (2000) Hydrophilic cyanine dyes as contrast agents for near-infrared tumor imaging: Synthesis, photophysical properties and spectroscopic characterization. Photochem. Photobiol. 72, 392−398. (48) Licha, K., Hessenius, C., Becker, A., Henklein, P., Bauer, M., Wisniewski, S., Wiedenmann, B., and Semmler, W. (2001) Synthesis, characterization, and biological properties of cyanine-labeled somatostatin analogues as receptor-targeted fluorescent probes. Bioconjugate Chem. 12, 44−50. (49) Montet, X., Yuan, H., Weissleder, R., and Josephson, L. (2006) Enzyme-based visualization of receptor-ligand binding in tissues. Lab. Invest. 86, 517−525. (50) Wu, X., Gong, S., Roy-Burman, P., Lee, P., and Culig, Z. (2013) Current mouse and cell models in prostate cancer research. Endocr.Relat. Cancer 20, R155−70. (51) Yang, Y. S., Zhang, X. Z., Xiong, Z. M., and Chen, X. Y. (2006) Comparative in vitro and in vivo evaluation of two Cu-64-labeled bombesin analogs in a mouse model of human prostate adenocarcinoma. Nucl. Med. Biol. 33, 371−380. (52) Laidler, P., Dulińska, J., Lekka, M., and Lekki, J. (2005) Expression of prostate specific membrane antigen in androgenindependent prostate cancer cell line PC-3. Arch. Biochem. Biophys. 435, 1−14.

(53) Fischer, G., Lindner, S., Litau, S., Schirrmacher, R., Wängler, B., and Wängler, C. (2015) Next Step toward optimization of GRP receptor avidities: Determination of the minimal distance between BBN(7−14) units in peptide homodimers. Bioconjugate Chem. 26, 1479− 1483. (54) Eggener, S. E., Stern, J. A., Jain, P. M., Oram, S., Ai, J., Cai, X., Roehl, K. A., and Wang, Z. (2006) Enhancement of intermittent androgen ablation by ″off-cycle″ maintenance with finasteride in LNCaP prostate cancer xenograft model. Prostate 66, 495−502.

I

DOI: 10.1021/acs.bioconjchem.8b00067 Bioconjugate Chem. XXXX, XXX, XXX−XXX