Specific Drug Delivery to Cancer Cells with Double-Imprinted

Jul 3, 2018 - Specific Drug Delivery to Cancer Cells with Double-Imprinted Nanoparticles ..... to cells and free doxorubicin (at 100 nM concentration)...
2 downloads 0 Views 1016KB Size
Subscriber access provided by NEW MEXICO STATE UNIV

Communication

Specific drug delivery to cancer cells with doubleimprinted nanoparticles against EGFR membrane receptor Francesco Canfarotto, Larissa Lezina, António Guerreiro, Joanna Czulak, Alexey Petukhov, Alexandra Daks, Katarzyna Smolinska-Kempisty, Alessandro Poma, Sergey A. Piletsky, and Nickolai Barlev Nano Lett., Just Accepted Manuscript • DOI: 10.1021/acs.nanolett.7b03206 • Publication Date (Web): 03 Jul 2018 Downloaded from http://pubs.acs.org on July 3, 2018

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 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 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.

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 20 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

Nano Letters

“Specific drug delivery to cancer cells with double-imprinted nanoparticles against EGFR membrane receptor” Francesco Canfarotta #†, Larissa Lezina#‡⊥, António Guerreiro †, Joanna Czulak† Alexey Petukhov ⊥˚, Alexandra Daks ⊥, Katarzyna Smolinska-Kempisty ∥, Alessandro Poma §, Sergey Piletsky*∥, Nickolai A. Barlev *⊥@ #

These authors contributed equally to this work.



MIP Diagnostics Ltd., Fielding Johnson Building, University of Leicester, LE1 7RH, UK.



Department of Cancer Studies, University of Leicester, LE1 7RH. ∥ Department of Chemistry,

University of Leicester, LE1 7RH, UK. ⊥ Laboratory of Gene Expression and Regulation, Institute of Cytology, 194064 Saint-Petersburg, Russia, ˚Institute of Hematology, Almazov National Medical Research Centre, 197341, St Petersburg, Russia,

§

Chemistry Department,

University College London, WC1H 0AJ, UK, @Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141700, Russia. Russia *The correspondence should be sent to: [email protected] and [email protected]

KEYWORDS: molecular imprinting, nanoparticles, molecular recognition, membrane receptors, cancer, drug delivery

ACS Paragon Plus Environment

1

Nano Letters 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

Page 2 of 20

ACS Paragon Plus Environment

2

Page 3 of 20 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

Nano Letters

ABSTRACT

The Epidermal Growth Factor Receptor (EGFR), a tyrosine kinase receptor, is overexpressed in many tumours, including almost half of triple-negative breast cancers. The latter belong to a very aggressive and drug-resistant form of malignancy. Though humanized anti-EGFR antibodies can work efficiently against these cancers both as mono-therapy and in combination with genotoxic drugs, instability and high production costs are some of their known drawbacks in clinical use. In addition, the development of antibodies to target membrane proteins is a very challenging task. Accordingly, the main focus of the present work is the design of supramolecular agents for targeting of membrane proteins in cancer cells and hence, more specific drug delivery. These were produced using a novel double-imprinting approach based on the solid-phase method for preparation of molecularly imprinted polymer nanoparticles (nanoMIPs), which were loaded with doxorubicin and targeted towards a linear epitope of EGFR. Additionally, upon binding, doxorubicin-loaded anti-EGFR nanoMIPs elicited cytotoxicity and apoptosis only in those cells that overexpressed EGFR. Thus, this approach can provide a plausible alternative to conventional antibodies and sets up a new paradigm for therapeutic application of this class of materials against clinically relevant targets. Furthermore, nanoMIPs can promote the development of cell imaging tools against difficult targets such as membrane proteins.

ACS Paragon Plus Environment

3

Nano Letters 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

Page 4 of 20

MAIN TEXT Protein-protein interactions are central to most biological processes – from intercellular communication to programmed cell death – and represent a large and important class of targets for human therapeutics. The current excitement around therapeutic antibodies vividly illustrates the value of such targets, with a market value predicted to reach $125bn by 2020.1 As a compound class, antibodies are highly specific for their molecular antigen and are usually stable in human serum. On the other hand, antibodies suffer from complex manufacturing procedures, high production costs, lack of oral bioavailability, poor cell membrane permeability, increased patient morbidity and even humanized types can elicit immunogenic reactions.1-5 Also, whilst cell surface receptors are obvious antigens when attempting to target specific cell populations, in practice obtaining antibodies against such receptors is a very challenging task.6,7 In this article, we assess the potential of molecularly imprinted polymer nanoparticles (nanoMIPs) as an alternative to antibodies against cell membrane receptors and their potential in cell imaging and targeted drug delivery. When produced in nanoparticle format,8 MIPs exhibit a size compatible with biological applications and therefore can be used for labeling/targeting specific cell structures. Also, these nanoparticles can plausibly become a new class of therapeutic agent, which can address both extracellular protein targets, as well as intracellular proteins9 (currently inaccessible to antibodies). The nanoMIPs were produced using a solid-phase double-imprinting approach against two targets, a linear epitope of the extracellular domain of the epidermal growth factor receptor (EGFR) and against the cytotoxic agent doxorubicin. Consequently, nanoMIPs are capable of selectively recognizing EGFR-overexpressing cells, whilst possessing at the same time binding sites for loading the cytotoxic agent doxorubicin. EGFR was selected as target

ACS Paragon Plus Environment

4

Page 5 of 20 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

Nano Letters

protein because it is overexpressed in several malignancies, including breast, colorectal and lung cancers, and is one of the critical regulators of cell proliferation and invasiveness.10-13 The solid-phase method employed for the MIP preparation (Figure 1a) relies on the covalent immobilization of the primary template (a synthetic short EGFR peptide) on a solid support (e.g. glass beads). This support bearing the immobilized template is placed in contact with the monomer mixture, which also contains the drug to be incorporated (doxorubicin) as a secondary template in solution, then polymerization is initiated under conditions which promote the formation of polymer nanoparticles (Figure 1c and d). NanoMIPs obtained in this way are surface-imprinted against the primary template for accurate cell targeting, whilst possessing binding sites within their bulk for drug delivery (Figure 1a). The products obtained using this method are virtually free from the primary template (as it is immobilized),8, 14, 15 whereas other traditional production approaches require lengthy dialysis of the nanoMIPs to remove it.16,

17

NanoMIPs with homogeneous binding affinities, nanomolar dissociation constants and good specificity for all major target classes (from small molecules to peptides and proteins) have been produced by solid-phase imprinting16, 18, 19 and used in the development of sensors and assays.2024

Whereas the common approach to target specifically cancer cells is exerted via antibodies,25 very recently other research groups employed nanoMIPs to target overexpressed moieties exposed on the surface of cells. However, only saccharides (sialic and glucuronic acid) have been used as templates for successful generation of MIPs.26-28 Here, anti-EGFR nanoMIPs (EGFR-MIPs) were produced against a synthetic C-terminal linear peptide sequence (amino acids 418–435: SLNITSLGLRSLKEISDG) of the extracellular portion of the EGFR receptor (PDB ID: 1NQL) which is exposed at the protein surface (Figure 1b). A cysteine residue was added on the N-

ACS Paragon Plus Environment

5

Nano Letters 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

Page 6 of 20

terminus of the epitope to allow its oriented immobilization on the solid-phase through a thiolreactive linker prior to nanoMIPs synthesis (see SI, Experimental Section for full details). The use of synthetic peptide epitopes as templates to produce imprinted polymers against proteins with a known sequence is potentially a novel and generic method that features significant advantages, as it does not require the preparation and purification of the full-length proteins, which are often difficult to produce in sufficient quantities and/or are unstable. To produce nanoMIPs, a mixture of monomers previously optimized specifically to imprint peptides and proteins in mild aqueous conditions was employed.15, 17, 29 The solid phase bearing the immobilized primary template was placed in contact with the monomer mixture (which contained the secondary template) and free-radical precipitation polymerization was initiated. Various monomers capable of establishing different molecular interactions with the epitope template were selected; these included: N-isopropylacrylamide (hydrogen bonding), N-tertbutylacrylamide (hydrophobic interactions), acrylic acid and N-(3-aminopropyl)methacrylamide (ionic interactions), whilst N,N′-methylenebisacrylamide was used as cross-linker. Due to the range of monomers used, this composition is also appropriate to imprint smaller molecules,[18] therefore the secondary template (doxorubicin) was simply added to the polymerization mixture. To visualize the nanoMIP interaction with the cancer cells, a fluorescent monomer, Nfluoresceinylacrylamide (N-fluo), was added to the monomer solution (see SI, Figure S1 for the fluorescence spectra). After polymerization, EGFR-nanoMIPs were obtained with an average hydrodynamic diameter in deionized water of 242 ± 13 nm, as measured by dynamic light scattering (DLS), and with approximately 150-200 nm when measured in dry state by TEM (see Figure SI, S2, and Figure 1c, 1d, respectively). NanoMIPs were tested by surface plasmon resonance (SPR) to determine

ACS Paragon Plus Environment

6

Page 7 of 20 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

Nano Letters

their affinity towards the primary template (the EGFR peptide epitope) as well as for the extracellular part oft he EGFR protein (aa 1-641). For control experiments, biotin-imprinted nanoparticles were prepared with the same monomer composition. Biotin was used as template because non-imprinted materials can not be produced using the solid-phase approach (the affinity separation step cannot be performed in the absence of an immobilised template). Biotin is sufficiently different from the EGFR epitope and is not naturally exposed on the cell surface, so these nano-MIPs can be considered for practical purposes as non-EGFR-imprinted materials. An equilibrium dissociation constant (KD) of 7.7 nM was obtained for the EGFR-nanoMIPs binding to the peptide, whilst a KD of 3.6 nM was obtained for the binding to the extracellular domain of the EGFR protein. Practically no binding of the EGFR peptide could be observed on the control nanoparticles (see SI, Figure S3 for the sensorgrams). To assess the efficacy of EGFR-nanoMIPs binding to their target region of EGFR in live cells, three breast cancer cell lines expressing different levels of EGFR (assessed by western blot, Figure 2a) were incubated with N-fluo-labelled nanoMIPs, followed by flow cytometry (FACS) analysis (Figure 2b and c). From the results shown in Figure 2 the breast cancer MDA-MB-468 cells, which expressed the highest amount of EGFR, also exhibited the highest binding of EGFRnanoMIPs (Figure 2c). Conversely, SKBR3 cells (that expressed virtually no EGFR) did not show any appreciable binding by the EGFR-nanoMIPs. To further assess the binding of fluorescent EGFR-nanoMIP, automated confocal microscopy was performed on MDA-MB-468 and SKBR-3 cells, respectively, expressing high and low levels of EGFR (Figure 2d, 2e). The nucleus was stained with DAPI (blue), and γ-tubulin (red) was employed to stain cell membrane. N-fluo-EGFR-nanoMIPs show green emission around 510 nm (SI, Figure S4). In agreement with the results obtained by FACS, we detected a strong fluorescent signal from the MIPs in MDA-

ACS Paragon Plus Environment

7

Nano Letters 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

Page 8 of 20

MB-468 cells overexpressing EGFR (Figure 2e), whereas almost no signal was observed in MDA-231 or SKBR3 cells (Figure. 2d and SI Figure S5). To further demonstrate that EGFRnanoMIPs bind specifically to the protein epitope in live cells, a competition assay with free peptide epitope in solution (tested at 10-fold range of concentrations: 0.1 and 1 µM) was performed using MDA-MB-468 cells (SI, Figure S6). The results showed a significant reduction of the amount of cell-bound EGFR-nanoMIPs detected by flow cytometry. However, the free peptide could not completely abolish the nanoMIP binding to the cells, arguing that some of the MIPs bind their target irreversibly. This is also suggested by the slow KOFF of the nanoMIPs bound to the EGFR protein, as detected by SPR (Figure S3). Importantly, the presence of the same peptide in the binding assay performed with SKBR-3 cells expressing low amount of EGFR had no obvious effect on already minuscule binding of EGFR-MIPs (compare SI Figure S6, right panel). Taken together, this indicates that EGFR-MIPs bind the EGFR epitope in a selective and competitive fashion. Collectively, these results demonstrate that the synthesized EGFR-nanoMIPs are specific for EGFR under physiological conditions, highlighting the potential use of nanoMIPs as convenient imaging tools to target specific membrane proteins in specific cell populations. Next, by employing EGFR-nanoMIPs we explored the possibility of delivering a cytotoxic drug (doxorubicin) specifically to MDA-MB-468 cells, enriched in EGFR. To this end, the drug was loaded

into

the

double-imprinted

EGFR-nanoMIPs

(“doxo-EGFR-nanoMIPs”).

Since

doxorubicin is a potent genotoxic drug, we expected that EGFR-expressing cells should preferentially be killed in comparison to cells treated with doxo-biotin-nanoMIPs, which cannot deliver the drug specifically to the cancer cells. The level of cell death was monitored by MTS and FACS analyses. In these experiments, “unloaded” EGFR-nanoMIPs showed no cytotoxic

ACS Paragon Plus Environment

8

Page 9 of 20 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

Nano Letters

effects, whilst doxo-loaded EGFR-nanoMIPs elicited an evident reduction in cell viability (Figure 3a). This effect was specific because SKBR-3 cells, which express low levels of EGFR, were insensitive to the treatment with doxo-loaded EGFR-nanoMIPs. Furthermore, as shown in Figure 3b, doxo-loaded EGFR-nanoMIPs augmented the level of cells in sub-G1 phase by more than 1.5 times compared to cells treated with doxo-loaded biotin-nanoMIPs or free doxorubicin. These results clearly demonstrate a more profound cell death in case of doxo-loaded EGFRnanoMIPs. To further confirm that the observed effects are due to specific binding of the nanoMIPs to EGFR, experiments were carried out with both doxo-loaded and unloaded EGFRnanoMIPs and breast cancer cell lines with high-levels (MDA-468) or low levels of EGFR (SKBR-3). Biotin-nanoMIPs were used as negative control. The results obtained suggest that the EGFR/nanoMIPs-mediated delivery of doxorubicin is specific (Figure S7) and leads to preferential killing of cells that overexpress EGFR (Figure S7). Noteworthy, neither biotin-MIPs nor the double-imprinted doxo-biotin-nanoMIPs affected significantly survival of MDA-MB-468 cells. Furthermore, EGFR-nanoMIPs without doxorubicin did not have any pronounced effect on survival of MDA-MB-468 cells, indicating that it is the targeted delivery of genotoxic drug, doxorubicin, that mediated cell death rather than mere binding of nanoMIPs to EGFR (Figure S8). To directly measure the intracellular levels of doxorubicin in MDA-MB-468 and SKBR-3 cells after the treatment with doxorubicin either bound to EGFR-nanoMIPs or in its free form we made use of the fact that doxorubicin exhibits auto-fluorescence at 480 nm, which can be detected with FACS. In fact, EGFR-nanoMIPs loaded with 0.1 µM of doxorubicin caused statistically higher retention of doxorubicin in EGFR high-MDA-MB-468 cells compared to the treatment with free doxorubicin. At the same time, EGFR-nanoMIPs loaded with 0.1 µM of

ACS Paragon Plus Environment

9

Nano Letters 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

Page 10 of 20

doxorubicin produced much weaker retention of doxorubicin in EGFR low-SKBR-3 cells, which was comparable to free doxorubicin. These results are consistent with other data demonstrating that double-imprinted nano-MIPs can be used as specific vehicles for targeted delivery of drugs (Figure S9). We reasoned that if EGFR-nanoMIPs loaded with doxorubicin caused statistically significant accumulation of the genotoxic drug, then we should also see an increased level of DNA damage in these cells manifested by ɣ-H2Ax staining. MDA-MB-468 cells were incubated with unloaded or loaded with doxorubicin (0,1 µM) EGFR-MIPs or biotin-MIPs (used as control). Subsequently, cells were stained with anti-phospho-H2A.X antibodies followed by confocal microscopy analysis (Figure S10). Indeed, we observed statistically significant augmentation of nuclear ɣ-H2Ax staining when cells were incubated with doxorubicin-loaded EGFR-nanoMIPs, but not with doxorubicin-loaded biotin-nanoMIPs. Finally, we undertook initial attempts to investigate the effect of EGFR-nanoMIPs binding on EGFR signaling. As consequence of EGFR interaction with its ligand, the receptor undergoes dimerization and subsequent internalization resulting in its accumulation in cytoplasmic lysosomes. Thus, we treated MDA-MB-468 cells with EGFR-MIPs in the presence or absence of EGF (Supplementary Fig. S12). We showed that EGFR-MIPs binding with the target receptor even in the absence of specific ligand caused accumulation of EGFR in cytoplasm suggesting that EGFR undergoes endocytosis upon interaction with EGFR-MIPs. Interestingly, we did not observe any significant effect on the downstream activation of EGFR-dependent kinases, e.g. Akt1 (Supplementary Fig. S11). However, when cells were incubated with EGFR-nanoMIPs loaded with doxorubicin we did observe activation of Erk1/2, which is consistent with the previously published data on participation of Erk1/2 in cell stress response.

ACS Paragon Plus Environment

10

Page 11 of 20 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

Nano Letters

In summary, in this study we demonstrated that nanoMIPs prepared via a double-imprinting method against a synthetic epitope of a membrane receptor can specifically recognize a native protein and selectively deliver a drug payload to the corresponding cell targets. As such, nanoMIPs can be considered as a new plausible alternative to conventional antibodies both in respect to the imaging and/or therapeutic tools against various clinically relevant targets. AUTHOR INFORMATION Corresponding Authors *To whom correspondence should be addressed, email: [email protected] and [email protected] CONFLICTS OF INTEREST Dr Canfarotta is employed by MIP Diagnostics Ltd. and therefore would like to disclose a competing financial interest.

ACKNOWLEDGMENTS A.P. thanks the UCL Department of Anatomy (Dr Mark Turmaine) for help with TEM and the NC3Rs for the award of a David Sainsbury Fellowship. A.D., A.P., and N B. appreciate the support of RSF grant 14-15-00816.

REFERENCES (1) Ecker, D. M.; Jones, S. D.; Levine, H. L., mAbs 2015, 7 (1), 9-14. (2) Berg, T., Angewandte Chemie - International Edition 2003, 42 (22), 2462-2481.

ACS Paragon Plus Environment

11

Nano Letters 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

Page 12 of 20

(3) Cochran, A. G., Current Opinion in Chemical Biology 2001, 5 (6), 654-659. (4) Gadek, T. R.; Nicholas, J. B., Biochemical Pharmacology 2003, 65 (1), 1-8. (5) Sharma, S. K.; Ramsey, T. M.; Bair, K. W., Current Medicinal Chemistry - Anti-Cancer Agents 2002, 2 (2), 311-330. (6) Hamakubo, T.; Kusano-Arai, O.; Iwanari, H., Biochimica et Biophysica Acta - Proteins and Proteomics 2014, 1844 (11), 1920-1924. (7) Doranz, B. J.; Banik, S. S. R., Genetic Engineering and Biotechnology News 2015, 35 (15), 20-21. (8) Poma, A.; Guerreiro, A.; Whitcombe, M. J.; Piletska, E. V.; Turner, A. P. F.; Piletsky, S. A., Advanced Functional Materials 2013, 23 (22), 2821-2827. (9) Canfarotta, F.; Waters, A.; Sadler, R.; McGill, P.; Guerreiro, A.; Papkovsky, D.; Haupt, A.; Piletsky S., Biocompatibility and internalization of molecularly imprinted nanoparticles. Nano Research, 2016, 9, 3463–3477 (10) Kwapiszewski, R.; Pawlak, S. D.; Adamkiewicz, K., Targeted Oncology 2016, 1-14. (11) Hanna, D. L.; Lenz, H. J., Expert Review of Clinical Pharmacology 2016, 9 (8), 10911108. (12) Woitok, M.; Klose, D.; Niesen, J.; Richter, W.; Abbas, M.; Stein, C.; Fendel, R.; Bialon, M.; Püttmann, C.; Fischer, R.; Barth, S.; Kolberg, K., Cancer Letters 2016, 381 (2), 323-330. (13) Ohba, T.; Toyokawa, G.; Osoegawa, A.; Hirai, F.; Yamaguchi, M.; Taguchi, K. I.; Seto, T.; Takenoyama, M.; Ichinose, Y.; Sugio, K., Surgery Today 2016, 46 (9), 1091-1098.

ACS Paragon Plus Environment

12

Page 13 of 20 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

Nano Letters

(14) Canfarotta, F.; Poma, A.; Guerreiro, A.; Piletsky, S., Nature Protocols 2016, 11 (3), 443455. (15) Poma, A.; Guerreiro, A.; Caygill, S.; Moczko, E.; Piletsky, S., RSC Advances 2014, 4 (8), 4203-4206. (16) Ambrosini, S.; Beyazit, S.; Haupt, K.; Tse Sum Bui, B., Chemical Communications 2013, 49 (60), 6746-6748. (17) Hoshino, Y.; Koide, H.; Urakami, T.; Kanazawa, H.; Kodama, T.; Oku, N.; Shea, K. J., Journal of the American Chemical Society 2010, 132 (19), 6644-6645. (18) Cáceres, C.; Canfarotta, F.; Chianella, I.; Pereira, E.; Moczko, E.; Esen, C.; Guerreiro, A.; Piletska, E.; Whitcombe, M. J.; Piletsky, S. A., Analyst 2016, 141 (4), 1405-1412. (19) Moczko, E.; Poma, A.; Guerreiro, A.; Perez De Vargas Sansalvador, I.; Caygill, S.; Canfarotta, F.; Whitcombe, M. J.; Piletsky, S., Nanoscale 2013, 5 (9), 3733-3741. (20) Basozabal, I.; Guerreiro, A.; Gomez-Caballero, A.; Aranzazu Goicolea, M.; Barrio, R. J., Biosensors and Bioelectronics 2014, 58, 138-144. (21) Chianella, I.; Guerreiro, A.; Moczko, E.; Caygill, J. S.; Piletska, E. V.; De Vargas Sansalvador, I. M. P.; Whitcombe, M. J.; Piletsky, S. A., Analytical Chemistry 2013, 85 (17), 8462-8468. (22) Shutov, R. V.; Guerreiro, A.; Moczko, E.; De Vargas-Sansalvador, I. P.; Chianella, I.; Whitcombe, M. J.; Piletsky, S. A., Small 2014, 10 (6), 1086-1089.

ACS Paragon Plus Environment

13

Nano Letters 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

Page 14 of 20

(23) Garcia-Mutio, D.; Gomez-Caballero, A.; Guerreiro, A.; Piletsky, S.; Goicolea, M. A.; Barrio, R. J., Chemical 2016, in press. DOI: 10.1016/j.snb.2016.02.018. (24) Garcia-Mutio, D.; Guerreiro, A.; Gomez-Caballero, A.; Gutierrez-Climente, R.; Piletsky, S.; Goicolea, M. A.; Barrio, R. J. Procedia Engineering, 2015, 1132-1136. (25) Cho, H. S.; Dong, Z.; Pauletti, G. M.; Zhang, J.; Xu, H.; Gu, H.; Wang, L.; Ewing, R. C.; Huth, C.; Wang, F.; Shi, D., ACS Nano 2010, 4 (9), 5398-5404. (26) Kunath, S.; Panagiotopoulou, M.; Maximilien, J.; Marchyk, N.; Sänger, J.; Haupt, K., Advanced Healthcare Materials 2015, 4 (9), 1322-1326. (27) Shinde, S.; El-Schich, Z.; Malakpour, A.; Wan, W.; Dizeyi, N.; Mohammadi, R.; Rurack, K.; Gjörloff Wingren, A.; Sellergren, B., Journal of the American Chemical Society 2015, 137 (43), 13908-13912. (28) Yin, D.; Wang, S.; He, Y.; Liu, J.; Zhou, M.; Ouyang, J.; Liu, B.; Chen, H. Y.; Liu, Z., Chemical Communications 2015, 51 (100), 17696-17699. (29) Hoshino, Y.; Kodama, T.; Okahata, Y.; Shea, K. J., Journal of the American Chemical Society 2008, 130 (46), 15242-15243.

ASSOCIATED CONTENT Supporting Information. Experimental methods, fluorescence spectra of the nanoMIPs, results of DLS and SPR analysis, confocal microscopy images of fluorescent nanoMIPs in MDA-468 and SKBR-3 cells after 2 and 24 h incubation and FACS analysis of EGFR nanoMIPs in breast

ACS Paragon Plus Environment

14

Page 15 of 20 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

Nano Letters

cancer cells expressing low levels of EGFR, peptide competition assay between EGFRnanoMIPs and MDA-MB-468 cells, effects of biotin-nanoMIPs and EGFR-nano-MIPs loaded and unloaded with doxorubicin on survival of MDA-MB-468 and SKBR 3cells, concentrationdependent effect of EGFR-nanoMIPs loaded with different amounts of doxorubicin on survival of MDA-MB-468 cells, incorporation levels of intracellular doxorubicin in MDA-468 and SKBR-3 cells treated with EGFR-nano-MIPs loaded with doxorubicin versus free doxorubicin.

Figure 1. (a) Scheme of the solid-phase synthesis process for double-imprinted nanoMIPs using a peptide epitope of EGFR as primary template attached to the solid phase and doxorubicin as secondary template in solution. (b) 3D structure of EGFR showing in blue is the sequence of the peptide template used for nanoMIPs fabrication and in yellow – antigenic region for the

ACS Paragon Plus Environment

15

Nano Letters 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

Page 16 of 20

therapeutic antibody Cetuximab which targets the same receptor. (c, d) TEM images of EGFR nanoMIPs at 20000× magnification (c) and 80000× magnification (d). Scale bars are respectively 1 µm and 200 nm.

ACS Paragon Plus Environment

16

Page 17 of 20 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

Nano Letters

Figure 2. Analysis of binding specificity for fluorescent N-fluo-EGFR-nanoMIPs in breast cancer cells. (a) FACS analysis of binding of EGFR-nanoMIPs to three cell lines expressing different levels of EGFR. Bars represent standard deviation for 3 replicates. FACS analysis on (b) SKBR-3 (expressing low amounts of EGFR) and (c) MDA-MB-468 cells (expressing high amounts of EGFR), demonstrating binding of EGFR-nanoMIPs to be proportional to amount of EGFR. Confocal microscopy on (d) SKBR-3 and (e) MDA-MB-468 cells, confirming the specific binding of EGFR-nanoMIPs (green) to the target protein.

ACS Paragon Plus Environment

17

Nano Letters 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

a

Page 18 of 20

b

c

Figure 3. Toxicity assays. (a) MTS test performed on MDA-MB-468 and SKBR-3 cells treated with EGFR-nanoMIPs either loaded with doxorubicin (doxo-EGFR-MIPs) or unloaded (EGFRMIPs). The control represents cells incubated in the absence of nanoMIPs. (b) Increase of the level of MDA-MB-468 cells in sub-G1 phase, due to the binding of doxo-EGFR-nanoMIP and doxo-biotin-nanoMIPs to cells, and free doxorubicin (at 100 nM concentration) analyzed by FACS. Prior to incubation with cells, the amount of doxorubicin loaded into both types of nanoMIPs (EGFR and biotin) and added to the cells was adjusted and assessed to be equal by fluorescence, and was 95 nM. The control represents cells incubated in the absence of nanoMIPs.

ACS Paragon Plus Environment

18

Page 19 of 20 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

Nano Letters

Statistical analysis was done by one-way ANOVA. All treated samples are compared with the untreated control, ** P < 0.01. (c) Schematic representation of the binding of nanoMIPs to EGFR at on the cell surface showing the targeted release of doxorubicin.

ACS Paragon Plus Environment

19

Nano Letters 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

Page 20 of 20

Table of contents graphic

ACS Paragon Plus Environment

20