Accepted Manuscript Synthesis and biological evaluation of phthalocyanine-peptide conjugate for EGFRtargeted photodynamic therapy and bioimaging Ligang Yu, Qiong Wang, Roy C.-H. Wong, Shirui Zhao, Dennis K.P. Ng, Pui-Chi Lo PII:
S0143-7208(18)32415-X
DOI:
https://doi.org/10.1016/j.dyepig.2018.11.055
Reference:
DYPI 7199
To appear in:
Dyes and Pigments
Received Date: 1 November 2018 Revised Date:
27 November 2018
Accepted Date: 28 November 2018
Please cite this article as: Yu L, Wang Q, Wong RC-H, Zhao S, Ng DKP, Lo P-C, Synthesis and biological evaluation of phthalocyanine-peptide conjugate for EGFR-targeted photodynamic therapy and bioimaging, Dyes and Pigments (2018), doi: https://doi.org/10.1016/j.dyepig.2018.11.055. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
Synthesis and biological evaluation of phthalocyanine-peptide conjugate for EGFR-targeted photodynamic therapy and bioimaging
RI PT
Ligang Yua, Qiong Wanga, Roy C.-H. Wongb, Shirui Zhaob, Dennis K. P. Ngb, PuiChi Loa,*
a
SC
Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
b
M AN U
Department of Chemistry, The Chinese University of Hong Kong, Shatin, N. T., Hong Kong, China
ABSTRACT: To improve the biocompatibility and tumor selectivity, we employed an epidermal growth factor receptor (EGFR) binding peptide (namely GE11, with a sequence of Tyr-His-Trp-
TE D
Tyr-Gly-Tyr-Thr-Pro-Gln-Asn-Val-Ile) as a tumor directing vector for the delivery of zinc(II) phthalocyanine for targeted photodynamic therapy and bioimaging. The photophysical properties, cellular uptake, in vitro cytotoxicity, and in vivo biodistribution of this phthalocyanine-peptide
EP
conjugate (namely Pc-GE11) have been evaluated. Pc-GE11 exhibited higher cellular uptake on EGFR-overexpressing epidermoid carcinoma A431 cells when compared with that on human
AC C
breast adenocarcinoma MCF7 cells (low EGFR expression). Moreover, pretreatment of A431 cells with GE11 peptide inhibited the cellular uptake of Pc-GE11 significantly and it exhibited exclusive light-activated cytotoxicity toward A431 cells. Furthermore, Pc-GE11 showed much higher tumor accumulation than the non-targeted control compound containing a random peptide sequence (Tyr-Trp-Gly-Pro-Asn-Ile-His-Tyr-Tyr-Thr-Gln-Val) after intravenous administration in A431-tumor bearing mice, indicating the potential application of this GE11 peptideconjugated photosensitizer for targeted photodynamic therapy and bioimaging. 1
ACCEPTED MANUSCRIPT
1. Introduction Photodynamic therapy (PDT) has long been used for the treatment of cancers and various
RI PT
diseases. It involves the combined action of a photosensitizer, molecular oxygen, and an appropriate wavelength of light to generate reactive oxygen species (ROS) for the eradication of cancer cells and tissues [1-3]. Currently, there are only few clinically approved PDT agents,
SC
including porfimer sodium, 5-aminolevulinic acid, temoporfin, verteporfin, talaporfrin, and padeliporfin [3]. However, the limited tumor selectivity and poor pharmacokinetics of these PDT
M AN U
agents have become major obstacles for their clinical application. To enhance the selectivity of PDT, “smart” photosensitizers have been developed, in which their photosensitizing properties are quenched through Foster resonance energy transfer or self-quenching mechanisms until they interact with specific cancer-associated stimuli [4-5]. Alternatively, photosensitizers can be conjugated with tumor-specific vectors, such as antibodies, peptides, carbohydrates, or other
TE D
small biomolecules, to enhance the tumor-targeting property [6-9]. Synthetic peptides have been widely used as the tumor-targeting ligands for cancer treatment and imaging [10-13]. With the
EP
aid of phage-display peptide library, a large number of peptide ligands have been identified which can specifically bind to different cancer cells, cancer-associated cell surface receptors, and
AC C
tumor vasculatures [14]. In addition to the high specificity, peptide-based drugs also have other advantages such as the ease of preparation and modification, non-immunogenicity, high tissue permeability, and rapid clearance from the body [15]. This approach has been widely used for the delivery of various agents such as chemotherapeutic drugs [16-18], radiopharmaceuticals [19-20], antisense oligonucleotides [21-23], and therapeutic nanoparticles [13, 24-25]. A substantial number of peptide-conjugated photosensitizers have been synthesized and evaluated for targeted PDT [26]. Peptide-conjugated photosensitizers usually exhibit less 2
ACCEPTED MANUSCRIPT
aggregation tendency and improved solubility in aqueous solution, hence enhancing the photosensitizing properties and therapeutic efficacy. Tumor homing peptides are one of the most interesting series which can target the receptors expressed on tumor tissues. Rahimipour et al.
RI PT
have conjugated protoporphyrin IX (PpIX) with a gonadotropin-releasing hormone (GnRH) agonist or antagonist to improve its selectivity toward cancerous cells that express GnRH receptors [27]. Another phthalocyanines conjugated with GnRH peptides have also been
SC
developed and they exhibited good antitumor efficacy on a GnRH receptors expressing murine breast cancer 4-T1 bearing mice [28]. A chlorin-type photosensitizer conjugated with the
M AN U
heptapeptide has also been developed to target the vascular endothelial growth factor (VEGF) co-receptor neuropilin-1, both in vitro and in vivo [29-30]. Several porphyrin, chlorin, and phthalocyanine-based photosensitizers conjugated with a linear or cyclic RGD motif have also been prepared and studied for their in vitro and in vivo selectivity and PDT efficacy toward
TE D
αvβ3-integrin expressing human endothelial cells [31-37]. Recently, a ruthenium-somatostatin photosensitizer has also been prepared for targeted PDT. It showed rapid cellular uptake by receptor-mediated endocytosis and potent photocytotoxicity on human lung cancer cells A549
EP
expressing somatostatin receptors [38].
Among all these tumor-related receptors on the tumor cells, epidermal growth factor
AC C
receptors (EGFR) are particularly interested as they play essential roles in both normal physiological and cancerous conditions. EGFR are expressed in a variety of human tumors, including lung, colon, pancreas, breast, ovary, bladder, kidney, head and neck, and gliomas, therefore they serve as promising therapeutic targets [39-40]. Several anti-EGFR therapeutic agents, including monoclonal antibodies (e.g. cetuximab and panitumumab) and tyrosine kinase inhibitors (e.g. gefitinib, erlotinib, and lapatinib) have been developed [41]. Recently, EGFR-
3
ACCEPTED MANUSCRIPT
targeted GE11 peptide has been identified by phage display screening. It contains 12 amino acids with a peptide sequence of Tyr-His-Trp-Tyr-Gly-Tyr-Thr-Pro-Gln-Asn-Val-Ile and it can bind to EGFR competitively with EGF and mediate internalization to EGFR-expressing cancer cells [42].
RI PT
The GE11 peptide has been conjugated with radiolabeled and fluorescent probes [43-44], and several drug delivery systems to deliver doxorubicin, paclitaxel, gemcitabine, aminoflavone, and siRNA for the EGFR-targeted diagnosis and treatment of cancers [45-50]. More recently, several peptide-decorated
nano-photosensitizing
systems
encapsulating
SC
GE11
silicon(IV)
phthalocyanines and chlorin e6 have been explored for EGFR-targeted PDT [51-53]. In addition,
M AN U
chlorin e4 and zinc(II) phthalocyanine conjugated with GE11 peptide have been prepared, showing improved selectivity but weak absorption in the red region and only moderate PDT efficacy [54-56]. Therefore, there is still room for the development of photosensitizer-GE11 peptide conjugate for EGFR-targeted PDT with potent therapeutic efficacy. As an extension of
TE D
our exploration of peptide-based photosensitizers [34, 57], we report herein the synthesis, characterization, photophysical properties, and in vitro photodynamic activity of a zinc(II) phthalocyanine conjugated with GE11 peptide (Pc-GE11). The in vivo biodistribution of this
EP
conjugate has also been examined on tumor-bearing nude mice.
AC C
2. Results and discussion
2.1 Synthesis and characterization Scheme 1 shows the synthetic route of the Pc-GE11. The side-chain protected peptide resin was synthesized according to the standard 9-fluorenylmethoxycarbonyl (Fmoc) solid-phase peptide synthesis (SPPS) protocol [58] with commercially available N-α-Fmoc-protected amino acids and Sieber amide resin as the solid support. The 1,4-bis(triethylene glycol)-substituted
4
ACCEPTED MANUSCRIPT
carboxyl zinc(II) phthalocyanine (Pc-COOH), which exhibits a red-shifted absorption, enhanced 1
O2 generation, and reduced aggregation tendency as compared with unsubstituted zinc(II)
phthalocyanine (ZnPc), was synthesized according to the literature procedure [57]. Firstly, the
RI PT
Fmoc protecting group of the tyrosine was removed by 20% of piperidine in DMF, then the peptide-resin was reacted with Pc-COOH in the presence of 1-hydroxybenzotriazole (HOBt), O(benzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate (HBTU), and N,N-
SC
diisopropylethylamine (DIPEA). The dark-green resin obtained was treated with a solution containing trifluoroacetic acid (TFA) (88%), phenol (5%), triisopropylsilane (TIS) (2%) and H2O
M AN U
(5%) to remove all the protecting groups on the peptide sequence and cleave the peptide from the resin. The crude Pc-GE11 conjugate was purified by semi-preparative HPLC. The control compound (Pc-rGE11), which contains a random sequence peptide (Tyr-Trp-Gly-Pro-Asn-IleHis-Tyr-Tyr-Thr-Gln-Val), was also prepared by the same method. Both Pc-GE11 and Pc-
TE D
rGE11 were characterized by analytical HPLC and ESI mass spectrometry (Fig. S1-S4 in the Supporting Information). The purities of Pc-GE11 and Pc-rGE11 were found to be more than
AC C
EP
97% as determined by analytical high-performance liquid chromatography (HPLC).
5
ACCEPTED MANUSCRIPT
N
N
N
N
Zn
N
2. Pc-COOH, HOBt, HBTU, DIPEA
O
O
O
N
N
N
O
O
O
Pc-COOH
N
Zn
N
N
N
N
N
N
N
N
Zn
N
N
N
N
O
O
O
O
O
O
O
O
O
O
O
O
N N N
O C HN
N N
OH C O
TFA/phenol/TIS/H 2O (88:5:2:5)
O
O
N
SC
N
N
O
O
M AN U
N
O
O
RI PT
1. 20% piperidine in DMF
N N N
O C HN
Pc-GE11
TE D
Scheme 1. Synthetic route of zinc(II) phthalcyanine conjugated with GE11 peptide (Pc-GE11).
2.2 Spectroscopic and photosensitizing properties
EP
The electronic absorption and fluorescence spectra of Pc-GE11 were examined in DMF
AC C
(Fig. 1a) and compared with Pc-COOH. It can be seen that Pc-GE11 shows a Soret band at 338 nm, a vibronic band at 623 nm, and a Q-band at 691 nm. The Q-band absorbance of Pc-GE11 was weaker than that of Pc-COOH, suggesting certain degree of aggregation behavior of PcGE11 after the substitution of the GE11 peptide. Upon excitation at 610 nm, the fluorescence of Pc-GE11 at 708 nm was observed and it was also slightly weaker than that of Pc-COOH (Fig. 1b), which may be attributed to the aggregation or the quenching effect of the tryptophancontaining GE11 peptide [54-55]. The absorption and photophysical data are summarized in 6
ACCEPTED MANUSCRIPT
Table 1. To mimic the biological environment, we also measured their absorption, fluorescence emission and singlet oxygen (1O2) generation in phosphate buffered saline (PBS) being formulated with 0.05% Tween 80 (v/v). As shown in Fig. 2, both compounds exhibited very
RI PT
similar absorption and fluorescence spectra after formulated with surfactant in aqueous solution, suggesting that the GE11 peptide conjugation does not affect the core of the phthalocyanine and the quenching effect by the tryptophan moiety is not significant in this condition. The 1O2
SC
generation efficiency was also determined in PBS using 1,3-diphenylisobenzofuran (DPBF) as the scavenger. The concentration of the quencher was monitored spectroscopically at 415 nm
M AN U
during irradiation. As shown in Fig. S5 in the Supporting Information, both Pc-GE11 and PcCOOH could generate 1O2 efficiently and the efficiency is comparable to each other. It further proves that conjugation of GE11 peptide to the zinc(II) phthalocyanine do not interfere the
0.20 0.16
EP
0.12 0.08
AC C
Absorbance
(b)
Pc-COOH Pc-GE11
0.04 0.00
300
400
500
600
700
800
Wavelength (nm)
Fluorescence Intensity (a.u.)
(a)
TE D
photosensitizing properties.
2000
Pc-COOH Pc-GE11
1600 1200 800 400 0 640
660
680
700
720
740
760
780
800
Wavelength (nm)
Fig. 1. (a) Electronic absorption and (b) fluorescence spectra (excitation at 610 nm) of PcCOOH and Pc-GE11 in DMF (both at 1 µM).
7
ACCEPTED MANUSCRIPT
0.12
Absorbance
0.10 0.08 0.06 0.04 0.02 0.00 300
400
500
600
700
800
1000 Pc-COOH Pc-GE11
800
RI PT
(b)
Pc-COOH Pc-GE11
600 400 200 0 640
SC
0.14
Fluorescence Intensity (a.u.)
(a)
660
680
700
720
740
760
780
800
Wavelength (nm)
Wavelength (nm)
M AN U
Fig. 2. (a) Electronic absorption and (b) fluorescence spectra (excitation at 610 nm) of PcCOOH and Pc-GE11 in PBS (pH 7.4) formulated with 0.05% Tween 80 (both at 1 µM).
Table 1. Electronic absorption and photophysical data for Pc-GE11 and Pc-COOH in DMF.
Pc-COOH Pc-GE11
Φ Fb
Φ△c
688 (5.24)
705
0.14
0.63
691 (5.12)
708
0.10
0.48
Excited at 610 nm. b Relative to ZnPc in DMF as the reference (ΦF = 0.28). cRelative to ZnPc as
EP
a
λem (nm)a
λmax (nm) (log ε)
TE D
Compound
AC C
the reference (Φ∆ = 0.56 in DMF).
2.3 In vitro studies
To examine the targeting property of the Pc-GE11 conjugate, we examined the cellular uptake of this conjugate toward human epidermoid carcinoma A431 cells (with high expression of EGFR) and human breast carcinoma MCF7 cells (with low expression of EGFR) by flow cytometry. It is clear that the cellular uptake of Pc-GE11 is much higher on A431 cells when 8
ACCEPTED MANUSCRIPT
compared with MCF7 cells as shown in Fig. 3a. In addition, upon the addition of excess GE11 peptide (100 or 400 µM), the cellular uptake of Pc-GE11 was significantly inhibited on A431 cells and the trend is concentration-dependent. Pretreatment of 100 µM or 400 µM of GE11
RI PT
peptide on A431 cells could block 25 % or 50% of the cellular uptake of Pc-GE11, respectively (Fig. 3b). The competitive study clearly demonstrated the EGFR-mediated pathway for the internalization of Pc-GE11. In addition, we compared the cellular uptake of Pc-GE11 with the
SC
control substituted with scrambled peptide sequence Pc-rGE11. It is obvious that the cellular uptake of Pc-rGE11 was significantly lower than Pc-GE11 on A431 cells. The flow cytometric
M AN U
results are summarized in Fig. 4. Apart from using the flow cytometry, we also examined the cellular uptake of Pc-GE11 on A431 cells by confocal microscopy. As shown in Fig. 5, there is a
AC C
EP
TE D
strong intracellular fluorescence in the cytoplasm of the cells after treated with Pc-GE11 for 4 h.
Fig. 3. (a) Flow cytometric analysis of A431 and MCF7 cells after treated with Pc-GE11 (4 µM) for 4 h. (b) Flow cytometric analysis of A431 cells after pretreated with free GE11 peptide (100 or 400 µM) for 2 h, followed by incubation with Pc-GE11 (4 µM) for 4 h.
9
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
Fig. 4. Comparison on the intracellular fluorescence intensity of Pc-GE11 and Pc-rGE11 in A431 and MCF7 cells monitored by flow cytometry. Data are expressed as the mean±standard
AC C
EP
TE D
deviation of three independent experiments.
Fig. 5. Confocal microscopic images of A431 cells after treated with Pc-GE11 (4 µM) for 4 h.
10
ACCEPTED MANUSCRIPT
The dark- and photo-cytotoxicity of Pc-GE11 was examined on A431 cells. Fig. 6. shows the dose-dependent survival curves both in the absence and presence of light. It is clear that PcGE11 is non-cytotoxic in dark at the tested concentrations, while it exhibits good cytotoxicity
RI PT
upon illumination. The IC50, which defined as the concentration of the dye required to kill 50% of the cells, was found to be 14 µM. The light-activated cytotoxicity makes Pc-GE11 a
SC
promising PDT agent.
120
M AN U
Cell Viability (%)
100
in dark with light
80 60 40
0
TE D
20
0
4
8
12
16
20
24
28
32
[Pc-GE11] (µM)
Fig. 6. Cytotoxic effects of Pc-GE11 on A431 cells in the absence and presence of light (λ > 610
EP
nm, 18 mW cm-2, 32 J cm-2). Data are expressed as mean value ± standard error of the mean
AC C
(S.E.M.) of three independent experiments, each performed in quintuplicate.
2.4 In vivo bio-distribution To study the in vivo tumor targeting ability of Pc-GE11, nude mice bearing A431 tumor were intravenously injected with Pc-GE11 (1 nmol per gram of mouse) and its bio-distribution was monitored for 72 h continuously as reflected by the measurement of the whole body fluorescence intensity. The non-targeted compound Pc-rGE11 was also used as the control. Both Pc-GE11 11
ACCEPTED MANUSCRIPT
and Pc-rGE11 spread over the whole body after injection for 4 h and the fluorescence intensity in the mice decreased significantly after 10 h post-injection. The Pc-GE11 showed much better tumor-retention property after 24 h post-injection. Moreover, bright fluorescence of Pc-GE11
RI PT
was also observed in the tumor region during 24-72 h post-injection. However, the fluorescence intensity of Pc-rGE11 at the tumor site was not significant throughout the study. To evaluate the bio-distribution of both compounds in tumor and different organs, the mice were sacrificed 3
SC
days after intravenous injection. The tumor and organs (kidney, heart, liver, lung, spleen and skin) were harvested and their relative fluorescence intensities were then measured. As shown in Fig. 8,
M AN U
the fluorescence intensities of Pc-GE11 in different organs were similar with Pc-rGE11, however, the fluorescence intensity of Pc-GE11 in tumor was much higher than that of PcrGE11, further proving the elevated tumor accumulation by targeted delivery. All these results suggest that Pc-GE11 can accumulate in EGFR-overexpressed tumor and it is a promising
AC C
EP
TE D
fluorescent probe for the detection of EGFR-rich tumor.
12
SC
RI PT
ACCEPTED MANUSCRIPT
Pc-GE11 Pc-rGE11
100 80 60 40 20 0 10
20
30
40
50
60
70
TE D
0
M AN U
Normalized Integrated Intensity 2 (Counts per mm )
(c)
Time (h)
Fig. 7. Fluorescence images of A431 tumor-bearing nude mice before and after intravenous
EP
injection of (a) Pc-GE11 and (b) Pc-rGE11 over 3 days. (c) Changes in fluorescence intensity per unit area of the tumor after treated with Pc-GE11 and Pc-rGE11. Data are expressed as
AC C
mean value ± standard deviation (S.D.) of five mice.
13
ACCEPTED MANUSCRIPT
Pc-GE11 Pc-rGE11
30 25
RI PT
20
**
15 10 5
Tu m or
M AN U
Organs
Sk in
Ki dn ey (L ) Ki dn ey (R )
Lu ng
Sp le en
Li ve r
H ea rt
0
SC
Fluorescence Intensity (a.u.)
35
Fig. 8. Ex vivo fluorescence intensity of different organs. The mice were sacrificed 3 days after the treatment of Pc-GE11 or Pc-rGE11. Data are expressed as mean value ± standard deviation (S.D.) of five mice. Statistical significance of the fluorescence intensity at the tumor region: P
TE D
value **p < 0.01, compared between the Pc-GE11 group and Pc-rGE11 group is calculated through variance analysis by Microsoft excel.
EP
3. Conclusions
An EGFR-targeted zinc(II) phthalocyanine (Pc-GE11) has been synthesized and
AC C
characterized. The photophysical properties, cellular uptake, in vitro cytotoxicity and in vivo biodistribution of Pc-GE11 have been examined. Competitive assay showed that pretreatment with free GE11 peptide could block the cellular uptake of the conjugate in A431 cells, indicating the receptor-mediated internalization pathway. In addition, MTT assay showed that this conjugate was non-toxic in dark while it induced cytotoxicity upon irradiation. Pc-GE11 also exhibited preferential tumor accumulation in tumor-bearing mice. In summary, Pc-GE11 is a promising EGFR-targeted PDT agent and fluorescent probe. 14
ACCEPTED MANUSCRIPT
4. Experimental section
RI PT
4.1 Materials All the reactions were performed under an atmosphere of nitrogen. DMF was pre-dried with barium oxide and distilled under reduced pressure. n-Pentanol was distilled from sodium under reduced pressure. Tetrahydrofuran (THF) was dried by sodium and distilled under atmospheric
SC
pressure. All other solvents and reagents were of reagent grade and used as received. Chromatographic purifications were performed on silica gel (Macherey-Nagel, 230-400 mesh)
M AN U
columns with specific eluents. Size exclusion chromatography was carried out on Bio-Rad BioBeads S-X1 beads (200−400 mesh) with THF as the eluent. The GE11 peptide resin [FmocTyr(tBu)-His(Trt)-Trp(Boc)-Tyr(tBu)-Gly-Tyr(tBu)-Thr(Trt)-Pro-Gln(Trt)-Asn(Trt)-Val-Ileresin] and the scrambled sequence peptide resin [Fmoc-Tyr(tBu)-Trp(Boc)-Gly-Pro-Asn(Trt)-
TE D
Ile-His(Trt)-Tyr(tBu)-Tyr(tBu)-Thr(Trt)-Gln(Trt)-Val-resin] were prepared manually following SPPS protocol [58] with commercially available N-α-Fmoc-protected amino acids. Sieber amide
described [57].
EP
resin (Novabiochem®) was used as the solid support. Compound Pc-COOH was prepared as
AC C
4.2 Characterization
Reversed-phase HPLC was performed on a XBridge C18 column (5 µm, 4.6 mm × 150 mm) for analytical purpose or a XBridge C18 column (5 µm, 10 mm × 250 mm) for preparative purpose using a Waters system equipped with a 1525 binary pump and a Waters 2998 diode array detector. Electrospray ionization (ESI) mass spectra were recorded on a Thermo Finnigan MAT 95 XL mass spectrometer. UV−Vis and steady-state fluorescence spectra were taken on a Cary
15
ACCEPTED MANUSCRIPT
5G UV−vis−NIR spectrophotometer and a Hitachi F-7000 spectrofluorometer, respectively. The 1
O2 generation in PBS was measured by the method of chemical quenching of 1,3-
diphenylisobenzofuran (DPBF) [59]. The light source for singlet oxygen generation came from a
RI PT
300 W halogen lamp after passing through a water tank for cooling and a color glass filter (Newport) cut-on 610 nm.
SC
4.3 Synthesis of Pc-GE11
The Fmoc protecting group of tyrosine was first removed from the GE11 peptide resin (7.2 µmol)
M AN U
with piperidine (20%) in DMF, then the peptide resin was washed with DMF (5 mL × 2) and CH2Cl2 (5 mL × 2). A mixture of Pc-COOH (22 mg, 21.4 µmol), HOBt hydrate (3 mg, 19.6 µmol), and HBTU (8 mg, 21.1 µmol) in DMF (3 mL) was added to the peptide resin in the presence of DIPEA (10 µL). The mixture was bubbled with nitrogen overnight at room
TE D
temperature. The dark-green resin was washed with DMF and CH2Cl2 and then dried in vacuo. The resin was then treated with 1.5 mL of cocktail solution containing TFA (88%), phenol (5%), TIS (2%) and H2O (5%) to remove all the protecting groups on the peptide sequence and cleave
EP
the peptide from the resin. The crude Pc-GE11 conjugate was purified by semi-preparative HPLC. The conditions were set as follows: solvent A = acetonitrile; solvent B = 0.1% TFA in
AC C
distilled water; gradient: maintained at 30% A + 70% B in the first 5 min, then changed to 100% A in 15 min and maintained at 100% A for 3 min, then changed back to 30% A + 70% B in 3 min and maintained for another 5 min. The flow rate was fixed at 1 mL min-1 for analysis and 3.5 mL min-1 for purification. The purity of the conjugate was found to be >97% as determined by HPLC. HRMS (ESI): m/z calcd for C125H145N29O27Zn [M + 2H]2+, 1275.0087; found, 1275.0091.
16
ACCEPTED MANUSCRIPT
4.4 Synthesis of Pc-rGE11 The synthesis and purification of Pc-rGE11 was performed similarly according to the above procedure for Pc-GE11. The purity of the conjugate was found to be >97% as determined by
RI PT
HPLC. HRMS (ESI): m/z calcd for C125H143N29O27ZnNa2 [M + 2Na]2+, 1296.9906; found, 1296.9910.
SC
4.5 Cell lines and culture conditions
The human epidermoid carcinoma A431 (ATCC® CRL-1555™) and human breast
M AN U
adenocarcinoma MCF7 (ATCC® HTB-22™) cells were maintained in Dulbecco’s Modified Eagle Medium (DMEM, ThermoFisher, cat. no. 11965092). The medium was supplemented with fetal bovine serum (10%) and penicillin-streptomycin (100 units mL-1 and 100 µg mL-1
TE D
respectively). The cells were kept at 37 oC in a humidified 5% CO2 atmosphere.
4.6 Flow cytometric analysis
Approximately 5 x 105 A431 cells or MCF7 cells per well in 2 mL cell culture media were added
EP
in 35 mm Petri dish and incubated overnight. The cells were then treated with Pc-GE11 or PcrGE11 (4 µM) in growth medium (2 mL) for 4 h. The cells were rinsed with PBS (1 mL × 2) and
AC C
harvested by trypsin (0.5 mL, 5 min). The activity of the trypsin was then quenched by culture medium (1 mL). The cells were collected by centrifugation at 1800 rpm for 3 min. The pellet was washed with PBS (1 mL) and then subject to centrifugation. The cells were suspended in 1 mL of ice cold PBS in dark and their fluorescence (λex = 638 nm, λem = 687-737 nm) was measured by flow cytometer (Beckman Coulter CytoFLEX S Flow cytometer analyzer) with 104 cells counted in each sample. For the competitive assays, A431 cells were pre-incubated with
17
ACCEPTED MANUSCRIPT
GE11 peptide (100 µM or 400 µM) for 2 h, then Pc-GE11 (4 µM) was added and incubated for another 4 h.
RI PT
4.7 Fluorescence microscopic studies
About 6 × 105 A431 cells per well in 2 mL cell culture media were added in 35 mm glass-bottom confocal dishes (MatTek) and incubated overnight. Stock solution of Pc-GE11 (2 mM) was
SC
prepared in DMSO and it was further diluted to 4 µM with culture medium. The cells were then treated with Pc-GE11 (4 µM, 2 mL) for 4 h. The cells were rinsed with PBS (1 mL × 2) and
M AN U
observed with a confocal laser scanning microscope (Zeiss Laser Scanning Microscope LSM 880 NLO with Airyscan) equipped with a 633 nm laser (Helium-Neon 633 laser). The emission signals at 638-747 nm were collected.
TE D
4.8 MTT assay
Approximately 3 × 104 A431 cells per well in 100 µL cell culture media were added to 96-well plates and incubated overnight. Pc-GE11 was first dissolved in DMSO to give 2 mM solution,
EP
which was diluted to different concentrations with culture medium. The cells were then treated with different concentrations of Pc-GE11 in growth medium for 4 h. The cells were rinsed with
AC C
PBS (100 µL × 2) and re-fed with 100 µL fresh media. The plate for dark cytotoxicity assay was directly plated in incubator overnight, while the other plate for photocytotoxicity testing was illuminated for 30 min at room temperature. The light source consisted of a 300 W halogen lamp, a water tank for cooling and a color glass filter (Newport, cut-on 610 nm). The fluence rate was 18 mW cm-2. An illumination of 30 min led to a total fluence of 32.4 J cm-2. After illumination, the cells were incubated overnight. Cell viability was determined by means of the colorimetric
18
ACCEPTED MANUSCRIPT
MTT assay.60 After rinsing the cells with PBS (100 µL), an 3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide (MTT) solution in PBS (3 mg ml-1, 50 µL) was added to each well followed by incubation for 4 h under the same conditions. DMSO (100 µL) was then added to
RI PT
each well to lyse the cells. The plates were placed on microplate reader (BioTek Synergy™ H1) at ambient temperature and the absorbance at 490 nm at each well was taken. The average absorbance of the blank wells, which did not contain the cells, was subtracted from the reading
SC
of the other wells. The cell viability was then determined by the equation: % Viability = [Σ(Ai/Acontrol × 100)]/n, where Ai is the absorbance of the ith data (i = 1, 2, …, n), Acontrol is the
M AN U
absorbance of the control wells, in which the drug was absent, and n is the number of data points.
4.9 In vivo study
Female Balb/c nude mice (20-25 g) were obtained from the Laboratory Animal Services Centre
TE D
at The Chinese University of Hong Kong. All animal experiments had been approved by the Animal Experimentation Ethics Committee of the University. The mice were kept under pathogen-free conditions with free access to food and water. A431 cells (5 × 106 cells in 200 µL)
EP
suspended in Hank’s balanced salt solution (HBSS) were inoculated subcutaneously on the back of the mice and waited until the tumors had grown to a size of 60-100 mm3. Pc-GE11 or Pc-
AC C
rGE11 (40 nmol) were first dissolved in DMSO (10 µL) and then diluted with distilled water (390 µL). These phthalocyanine solutions (200 µL, equivalent to 20 nmol) were intravenously injected into the tail vein of the tumor-bearing mice. In vivo fluorescence imaging was captured before and after the injection at different time points for 3 days with an Odyssey infrared imaging system (excitation wavelength at 680 nm, emission wavelength at ≥ 700 nm). The mice were sacrificed after 3 days and the organs were harvested. The ex vivo biodistribution of the
19
ACCEPTED MANUSCRIPT
compounds was determined using the fluorescence intensity obtained by the Odyssey infrared imaging system. The images were digitized and analyzed by the Odyssey imaging system
RI PT
software (no. 9201-500). Five mice were used for each compound.
Acknowledgements
SC
This work was supported by the City University of Hong Kong (ref. no. 7004585).
1.
Dolmans DEJGJ, Fukumura D, Jain RK. Photodynamic therapy for cancer. Nat Rev Cancer 2003; 3: 380-7.
2.
M AN U
References
Celli JP, Spring BQ, Rizvi I, Evans CL, Samkoe KS, Verma S, Pogue BW, Hasan T. Imaging and photodynamic therapy: mechanisms, monitoring, and optimization. Chem
3.
TE D
Rev 2010; 110: 2795-838.
van Straten D, Mashayekhi V, de Bruijn HS, Oliveira S, Robinson DJ. Oncologic photodynamic therapy: basic principles, current clinical status and future directions.
Lovell JF, Liu TWB, Chen J, Zheng G. Activatable photosensitizers for imaging and
AC C
4.
EP
Cancers 2017; 9: 1-54.
therapy. Chem Rev 2010; 110: 2839-57. 5.
Majumdar P, Nomula R, Zhao JZ. Activatable triplet photosensitizers: magic bullets for targeted photodynamic therapy. J Mater Chem C 2014; 2: 5982-97.
6.
Schmitt F, Juillerat-Jeanneret L. Drug targeting strategies for photodynamic therapy. AntiCancer Agents Med Chem 2012; 12: 500-25.
20
ACCEPTED MANUSCRIPT
7.
Giuntini F, Alonso CMA, Boyle RW. Synthetic approaches for the conjugation of porphyrins and related macrocycles to peptides and proteins. Photochem Photobiol Sci 2011; 10: 759-91. Verma S, Watt GM, Mai Z, Hasan T. Strategies for enhanced photodynamic therapy
RI PT
8.
effects. Photochem Photobiol 2007; 83: 996-1005. 9.
Lu J, Zhang W, Yuan L, Ma W, Li X, Lu W, Zhao Y, Chen G. One-pot synthesis of
SC
glycopolymer-porphyrin conjugate as photosensitizer for targeted cancer imaging and photodynamic therapy. Macromol Biosci 2014; 14: 340-6.
Shadidi M, Sioud M. Selective targeting of cancer cells using synthetic peptides. Drug Resist Updates 2003; 6: 363-71.
11.
M AN U
10.
Aina OH, Liu R, Sutcliffe JL, Marik J, Pan CX, Lam KS. From combinatorial chemistry to cancer-targeting peptides. Mol Pharmaceutics 2007; 4: 631-51. Ruoslahti E. Tumor penetrating peptides for improved drug delivery. Adv Drug Deliv Rev 2017; 110: 3-12.
13.
TE D
12.
David A. Peptide ligand-modified nanomedicines for targeting cells at the tumor
14.
EP
microenvironment. Adv Drug Deliv Rev 2017; 119: 120-42. Gray BP, Brown KC. Combinatorial peptide libraries: mining for cell-binding peptides.
15.
AC C
Chem Rev 2014; 114: 1020-81. Wu D, Gao Y, Qi Y, Chen L, Ma Y, Li Y. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett 2014; 351: 13-22. 16.
Arap W, Pasqualini R, Ruoslahti E. Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science 1998; 279: 377-80.
21
ACCEPTED MANUSCRIPT
17.
Kouodom MN, Ronconi L, Celegato M, Nardon C, Marchio L, Dou QP, Aldinucci D, Formaggio F, Fregona D. Toward the selective delivery of chemotherapeutics into tumor cells by targeting peptide transporters: tailored gold-based anticancer peptidomimetics. J
18.
RI PT
Med Chem 2012; 55: 2212-26.
Guan H, McGuire MJ, Li S, Brown KC. Peptide-targeted polyglutamic acid doxorubicin
19: 1813-21. 19.
Okarvi SM. Peptide-based radiopharmaceuticals and cytotoxic conjugates: potential tools
M AN U
against cancer. Cancer Treat Rev 2008; 34: 13-26. 20.
Tweedle MF. Peptide-targeted diagnostics and radiotherapeutics. Acc Chem Res 2009; 42: 958-68.
21.
SC
conjugates for the treatment of alpha(v)beta(6)-positive cancers. Bioconjugate Chem 2008;
Ming X. Cellular delivery of siRNA and antisense oligonucleotides via receptor-mediated
22.
TE D
endocytosis. Expert Opin Drug Delivery 2011; 8: 435-49.
Taskova M, Madsen CS, Jensen KJ, Hansen LH, Vester B, Astakhova K. Antisense oligonucleotides internally labeled with peptides show improved target recognition and
23.
EP
stability to enzymatic degradation. Bioconjugate Chem 2017; 28: 768-74. Favretto ME, Brock R. Stereoselective uptake of cell-penetrating peptides is conserved in
24.
AC C
antisense oligonucleotide polyplexes. Small 2015; 11: 1414-7. Farkhani SM, Valizadeh A, Karami H, Mohammadi S, Sohrabi N, Badrzadeh F. Cell penetrating peptides: efficient vectors for delivery of nanoparticles, nanocarriers, therapeutic and diagnostic molecules. Peptides 2014; 57: 78-94. 25.
Ruoslahti E. Peptides as targeting elements and tissue penetration devices for nanoparticles. Adv Mater 2012; 24: 3747-56.
22
ACCEPTED MANUSCRIPT
26.
Han K, Ma ZY, Han HY. Functional peptide-based nanoparticles for photodynamic therapy. J Mater Chem B 2018; 6: 2-38.
27.
Rahimipour S, Ben-Aroya N, Ziv K, Chen A, Fridkin M, Koch Y. Receptor-mediated
analogues. J Med Chem 2003; 46: 3965-74. 28.
RI PT
targeting of a photosensitizer by its conjugation to gonadotropin-releasing hormone
Xu P, Jia Y, Yang Y, Chen J, Hu P, Chen Z, Huang M. Photodynamic oncotherapy
29.
SC
mediated by gonadotropin-releasing hormone receptors. J Med Chem 2017; 60: 8667-72. Tirand L, Frochot C, Vanderesse R, Thomas N, Trinquet E, Pinel S, Viriot ML, Guillemin
M AN U
F, Barberi-Heyob M. A peptide competing with VEGF165 binding on neuropilin-1 mediates targeting of a chlorin-type photosensitizer and potentiates its photodynamic activity in human endothelial cells. J Controlled Release 2006; 111: 153-64. 30.
Thomas N, Bechet D, Becuwe P, Tirand L, Vanderesse R, Frochot C, Guillemin F,
TE D
Barberi-Heyob M. Peptide-conjugated chlorin-type photosensitizer binds neuropilin-1 in vitro and in vivo. J Photochem Photobiol B Biol 2009; 96: 101-8. 31.
Frochot C, Stasio BD, Vanderesse R, Belgy MJ, Dodeller M, Guillemin F, Viriot ML,
EP
Barberi-Heyob M. Interest of RGD-containing linear or cyclic peptide targeted tetraphenylchlorin as novel photosensitizers for selective photodynamic activity. Bioorg
32.
AC C
Chem 2007; 35: 205-20.
Srivatsan A, Ethirajan M, Pandey SK, Dubey S, Zheng X, Liu TH, Shibata M, Missert J, Morgan J, Pandey R. K. Conjugation of cRGD peptide to chlorophyll a based photosensitizer (HPPH) alters its pharmacokinetics with enhanced tumor-imaging and photosensitizing (PDT) efficacy. Mol Pharmaceutics 2011; 8: 1186-97.
23
ACCEPTED MANUSCRIPT
33.
You H, Yoon HE, Jeong PH, Ko H, Yoon JH, Kim YC. Pheophorbide-a conjugates with cancer-targeting moieties for targeted photodynamic cancer therapy. Bioorg Med Chem 2015; 23: 1453-62. Ke MR, Ng DKP, Lo PC. Synthesis and in vitro photodynamic activities of an integrin-
RI PT
34.
targeting cRGD-conjugated zinc(II) phthalocyanine. Chem Asian J 2014; 9: 554-61. 35.
Luan L, Fang W, Liu W, Tian M, Ni Y, Chen X, Yu X. Phthalocyanine-cRGD conjugate:
SC
synthesis, photophysical properties and in vitro biological activity for targeting photodynamic therapy. Org Biomol Chem 2016; 14: 2985-92.
Zheng BY, Yang XQ, Zhao Y, Zheng QF, Ke MR, Lin T, Chen RX, Ho KKK, Kumar N,
M AN U
36.
Huang JD. Synthesis and photodynamic activities of integrin-targeting silicon(IV) phthalocyanine-cRGD conjugates. Eur J Med Chem 2018; 155: 24-33. 37.
Liu Q, Pang M, Tan S, Wang J, Chen Q, Wang K, Wu W, Hong Z. Potent peptide-
310-20. 38.
TE D
conjugated silicon phthalocyanines for tumor photodynamic therapy. J Cancer 2018; 9:
Wang T, Zabarska N, Wu Y, Lamla M, Fischer S, Monczak K, Ng DYW, Rau S, Weil T. selective
ruthenium-somatostatin
photosensitizer
for
cancer
targeted
EP
Receptor
photodynamic applications. Chem Commun 2015; 51: 12552-5. Wieduwilt MJ, Moasser MM. The epidermal growth factor receptor family: biology
AC C
39.
driving targeted therapeutics. Cell Mol Life Sci 2008; 65: 1566-84. 40.
Mitsudomi T, Yatabe Y. Epidermal growth factor receptor in relation to tumor development: EGFR gene and cancer. FEBS Journal 2010; 277: 301-8.
41.
Seshacharyulu P, Ponnusamy MP, Haridas D, Jain M, Ganti AK, Batra SK. Targeting the EGFR signaling pathway in cancer therapy. Expert Opin Ther Targets 2012; 16: 15-31.
24
ACCEPTED MANUSCRIPT
42.
Genta I, Chiesa E, Colzani B, Modena T, Conti B, Dorati R. GE11 Peptide as an active targeting agent in antitumor therapy: a minireview. Pharmaceutics 2018; 10: 1-14 (doi: 10.3390/pharmaceutics10010002). Yu HM, Chen JH, Lin KL, Lin WJ. Synthesis of
68
Ga-labeled NOTA-RGD-GE11
RI PT
43.
heterodimeric peptide for dual integrin and epidermal growth factor receptor-targeted tumor imaging. J Label Compd Radiopharm 2015; 58: 299-303.
Zhao N, Williams TM, Zhou Z, Fronczek FR, Sibrian-Vazquez M, Jois SD, Vicente MGH.
SC
44.
Synthesis of BODIPY-Peptide Conjugates for Fluorescence Labeling of EGFR
45.
M AN U
Overexpressing Cells. Bioconjugate Chem 2017; 28: 1566-79.
Liang X, Shi B, Wang K, Fan M, Jiao D, Ao J, Song N, Wang C, Gu J, Li Z. Development of self-assembling peptide nanovesicle with bilayers for enhanced EGFR-targeted drug and gene delivery. Biomaterials 2016; 82: 194-207.
Milane L, Duan Z, Amiji M. Therapeutic efficacy and safety of paclitaxel/lonidamine
TE D
46.
loaded EGFR-targeted nanoparticles for the treatment of multi-drug resistant cancer. Plos One 2011; 6: e24075.
Mondal G, Kumar V, Shukla SK, Singh PK, Mahato RI. EGFR-targeted polymeric mixed
EP
47.
micelles carrying gemcitabine for treating pancreatic cancer. Biomacromolecules 2016; 17:
48.
AC C
301-13.
Chen G, Ma B, Wang Y, Xie R, Li C, Dou K, Gong S. CuS-based theranostic micelles for NIR-controlled combination chemotherapy and photothermal therapy and photoacoustic imaging. ACS Appl Mater Interfaces 2017; 9: 41700-11.
25
ACCEPTED MANUSCRIPT
49.
Brinkman AM, Chen G, Wang Y, Hedman CJ, Sherer NM, Havighurst TC, Gong S, Xu W. Aminoflavone-loaded EGFR-targeted unimolecular micelle nanoparticles exhibit anticancer effects in triple negative breast cancer. Biomaterials 2016; 101: 20-31. Chen G, Wang Y, Xie R, Gong S. Tumor-targeted pH/redox dual-sensitive unimolecular
RI PT
50.
nanoparticles for efficient siRNA delivery. J Controlled Release 2017; 259: 105-14. 51.
Master AM, Livingston M, Oleinick NL, Gupta AS. Optimization of a nanomedicine-based
SC
silicon phthalocyanine 4 photodynamic therapy (Pc 4-PDT) strategy for targeted treatment of EGFR-overexpressing cancers. Mol Pharmaceutics 2012; 9: 2331-8. Master A, Malamas A, Solanki R, Clausen DM, Eiseman JL, Gupta AS. A cell-targeted
M AN U
52.
photodynamic nanomedicine strategy for head and neck cancers. Mol Pharmaceutics 2013; 10: 1988-97. 53.
Chu WY, Tsai MH, Peng CL, Shih YH, Luo TY, Yang SJ, Shieh MJ. pH-Responsive
TE D
nanophotosensitizer for an enhanced photodynamic therapy of colorectal cancer overexpressing EGFR. Mol Pharmaceutics 2018; 15: 1432-44. 54.
Kim J, Chae J, Kim JS, Goh SH, Choi Y. Photosensitizer-conjugated tryptophan-
513: 584-90.
Kim J, Won Y, Goh SH, Choi Y. A redox-responsive theranostic agent for target-specific
AC C
55.
EP
containing peptide ligands as new dual-targeted theranostics for cancers. Int J Pharm 2016:
fluorescence imaging and photodynamic therapy of EGFR-overexpressing triple-negative breast cancers. J Mater Chem B 2016; 4: 6787-90. 56.
Ongarora BG, Fontenot KR, Hu X, Sehgal I, Satyanarayana-Jois SD, Vicente MGH. Phthalocyanine-peptide conjugates for epidermal growth factor receptor targeting. J Med Chem 2012; 55: 3725-38.
26
ACCEPTED MANUSCRIPT
57.
Ke MR, Yeung SL, Fong WP, Ng DKP, Lo PC. A phthalocyanine-peptide conjugate with high in vitro photodynamic activity and enhanced in vivo tumor-retention property. Chem Eur J 2012; 18: 4225-33. Atherton E, Sheppard RC. Solid phase peptide synthesis: a practical approach, IRL Press: Oxford, 1989.
59.
Maree
MD,
Kuznetsova
N,
Nyokong
T.
Silicon
RI PT
58.
octaphenoxyphthalocyanines:
SC
photostability and singlet oxygen quantum yields. J Photochem Photobiol A 2001; 140: 117-25.
M AN U
Tada H, Shiho O, Kuroshima K, Koyama M, Tsukamoto K. An improved colorimetric
EP
TE D
assay for interleukin-2. J Immunol Methods 1986; 93: 157-65.
AC C
60.
27