You are on page 1of 9

Journal of Controlled Release 182 (2014) 13–21

Contents lists available at ScienceDirect

Journal of Controlled Release


journal homepage: www.elsevier.com/locate/jconrel

Delivering wasp venom for cancer therapy


Miguel Moreno ⁎, Esther Zurita, Ernest Giralt ⁎⁎
Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Cytolytic peptides with potential therapeutic properties have appeared during the last three decades. However,
Received 10 February 2014 the use of these natural weapons is relatively narrow due to their non-specific cytolytic activity as well as their
Accepted 3 March 2014 rapid degradation and excretion when injected in blood. In order to rescue the use of these lytic peptides, we
Available online 12 March 2014
have designed pro-cytotoxic systems based on cytotoxic peptides conjugated to poly(L-glutamic acid) PGA poly-
mer through specific cleavage sequences that are sensitive over-expressed tumor proteases, such as the
Keywords:
Pro-cytotoxic systems
metalloproteinase-2 (MMP-2) or cathepsin B. The potent cytotoxic peptide tested here, Mitoparan, is inactive
Cytolytic peptides when conjugated to the polymer and then become active again once released through the tumor proteases. Fur-
Polyglutamic acid thermore, this pro-cytotoxic system was decorated by a particular targeting peptide which binds to HER2 recep-
Targeting peptides tors over-expressed in some types of breast tumor cells, thereby increasing the selective release of cytolytic
Overexpressed tumor proteases peptides inside tumor cell with exquisite spatiotemporal control. In this way, the system would improve the
HER2+ breast cancer maximum tolerated dose and the pharmacokinetic parameters of cytotoxic peptides in vivo.
© 2014 Elsevier B.V. All rights reserved.

1. Introduction Alternative types of carriers, such polymers, with suitable features could
improve the efficacy of delivery cytolytic peptides, overcoming some
The use of venoms in cancer therapy continues to be a challenge. drawbacks related to low activity, elaborate complexes and non-scalable
These natural weapons have been widely studied for the treatment of production. In fact, an increasing number of polymer therapeutics have
several immune-related diseases, and have recently entered preclinical started to move from basic science into clinical trials and, even into clini-
phases for cancer treatment [1]. However, the high toxicity of these po- cal practice [7]. Not only should the polymer in the drug delivery system
tential therapeutic peptides caused by non-specific lytic activity and be innocuous, hydrophilic, non-immunogenic and biodegradable, but also
their rapid degradation in blood make them of limited use in cancer should it have a long-circulating behavior and enhanced accumulation at
therapy. These peptides are between 10 and 50 residues in length, and the tumor site. In particular, the well-characterized poly(L-glutamic acid)
they show amphipathic properties. They have a propensity to interact (PGA), which has an attractive safety profile, has been widely used with
with membranes, oligomerizing on the cell surface so as to form tran- many kinds of cargo, such as low MW drugs [8] and, even, peptoids [9].
sient pores, thus causing cell death. Since free cytolytic peptides are In this study, we present a peptide-polymer design strategy to
not able to elicit a therapeutic benefit at a safe dose, they have to be obtain pro-cytotoxic systems based on lytic peptides conjugated to
targeted and delivered as pro-drugs. Previous studies report on how dif- PGA polymer through specific cleavage sites that are sensitive over-
ferent types of cytolytic peptides have been successfully conjugated to expressed tumor proteases, such as MMP-2 or cathepsin B (Fig. 1).
proteins through tumor-specific proteases, such as metalloproteinase- The potent cytotoxic peptides are inactive when conjugated to the poly-
2 (MMP-2) [2,3]. In addition, cytotoxic peptides have been carried by li- mer, but once released through the tumor proteases, they recover their
pidic systems, as in Yamada's work [4], where the peptide mastoparan activity. This strategy is thought to prevent the side effects that occur
was transported inside a transferrin-modified liposome. The most re- in vivo. Furthermore, this pro-cytotoxic carrier was decorated with pep-
cent case is the 26-mer cytotoxic peptide melittin, derived from bee tides able to specifically target tumor cells. In this way, the system
venom, which has been successfully targeted and delivered via fluoro- would improve the maximum tolerated dose and the pharmacokinetic
carbon nanocarriers to tumor models in vivo [5,6]. Further improve- parameters of cytotoxic peptides in vivo.
ments in order to increase the efficacy of the systems are needed.

2. Materials and methods


⁎ Correspondence to: M. Moreno, Institute for Research in Biomedicine (IRB Barcelona),
C\Baldiri I Reixac, 10, 08028 Barcelona, Spain. Tel.: +34 934037127. 2.1. Materials
⁎⁎ Correspondence to: E. Giralt, University of Barcelona, Institute for Research in
Biomedicine (IRB Barcelona), C\Baldiri I Reixac, 10, 08028 Barcelona, Spain.
E-mail addresses: miguel.moreno@irbbarcelona.org (M. Moreno), Fmoc-Nα-protected amino acids and poly(L-glutamic acid) (Npt-
ernest.giralt@irbbarcelona.org (E. Giralt). PGA(100)Na) were obtained from IRIS Biotech GmbH (Marktredwitz,

http://dx.doi.org/10.1016/j.jconrel.2014.03.005
0168-3659/© 2014 Elsevier B.V. All rights reserved.
14 M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21

Fig. 1. a) Synthetic strategy of the targeted pro-cytotoxic conjugate based on modified PGA polymer (PGAmod). PGA decoration with different reagents is used to prepare a soluble scaffold
onto which the targeting and pro-apoptotic peptides (Mitoparan) are attached. b) Schematic illustration showing the interaction of targeted pro-cytotoxic conjugates with HER2 positive
tumor cells, which overexpress MMP-2 on the plasmatic membrane (A) or cathepsin B inside lysosomes (B). These two enzymes will promote the release of Mitoparan, cutting through
specific cleavage sequences, transforming this pro-drug system into a toxic drug.

Germany). The Fmoc-Rink-Amide AM polystyrene resin was pur- 2.2. Peptide synthesis
chased from CBL-PATRAS (Patras, Greece). Coupling reagents: 2-
(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium tetrafluoroborate Peptides were synthesized by solid phase synthesis using the 9-
(TBTU) was from Albatros Chem, Inc. (Montreal, Canada). Trifluoroacetic fluorenylmethoxycarbonyl/tert-butyl (Fmoc/tBu) strategy. Fmoc-
acid (TFA) was purchased from Scharlab S.L. (Barcelona, Spain). Rink-Amide AM polystyrene resin, Nα-Fmoc-protected amino acids
Piperidine, dimethylformamide (DMF), dichloromethane (DCM) (3 eq)/TBTU(3 eq.), and DIEA(6 eq.) were used. The Fmoc protecting
and acetonitrile (MeCN) were from SDS (Peypin, France). N,N- group was cleaved by treatment with a solution of 20% piperidine in
Diisopropylethylamine (DIEA) was obtained from Merck (Darmstadt, DMF. For labeling with 5(6)-carboxyfluorescein (CF) of the dye-
Germany). Tri-isopropylsilane (TIS) was from Fluka (Buchs, peptides 2, 3, and 9, firstly, Nα-Fmoc-Lys(Alloc)-OH was incorporated
Switzerland). Lissamine rhodamine B sulfonyl chloride (Sulrhodamine at the desired position. For the Alloc group removal, the resin was treat-
B) was obtained from Acros (Somerville, NJ). Methoxy PEG Amine, ed with Pd(PPh3)4 and PhSiH3 (3 × 15 min) and then washed with DMF.
HCl Salt, MW 5000 was from JenKem Technology USA Inc. LysoTracker® Afterwards, 2 treatments of 30 min with a solution of sodium N,N′
Green DND-26 and Red DND-99 were supplied by Invitrogen (Carlsbad, diethyldithiocarbamate in DMF were performed. Finally, the CF was in-
CA). Cathepsin B from human liver, Sephadex® G-25, 5(6)-carboxyflu- corporated as it was a Nα-Fmoc-protected amino acid. Peptides were
orescein (CF), tetrakis(triphenylphosphine)palladium(0), (Pd(PPh3)4), cleaved from the resin by treatment with 95% TFA, 2.5% TIS, 2.5%
phenylsilane (PhSiH3) and ethanolamine were obtained from Sigma- water for 3 h and detected by analytical RP-HPLC Waters 996 photodi-
Aldrich (St. Louis, USA). Recombinant Human Matrix Metalloproteinase ode array detector equipped with the Waters 2695 separation module,
2 (MMP-2) was purchase from Sina Biological Inc. (Beijing, China). the Symmetry column (C 18, 5 mm, 4.6P150 mm) and the Millennium
M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21 15

software. Peptides were purified by semipreparative RP-HPLC (Waters added, and multilamellar vesicles were obtained. Large unilamellar ves-
2487 Dual l Absorbance Detector equipped with a Waters 2700 Sample icles (LUVs) with a mean diameter of 200 nm were prepared from the
Manager, a Waters 600 Controller, a Waters Fraction Collector, a Sym- multilamellar vesicles by the LiposoFast device from Avestin, Inc.,
metry column [C 18, 5 mm, 30P100 mm] and Millennium chromatogra- using polycarbonate filters with a pore size of 0.2 μm (Nuclepore
phy manager software) (see Supplementary data, Fig. S1). Peptides Corp., Cambridge, CA, USA). Breakdown of the vesicle membrane leads
were characterized by MALDI-TOF mass spectrometry (Voyager-DE RP to content leakage, i.e., CF fluorescence. Non-encapsulated CF was sepa-
MALDI-TOF, PE Biosystems with a N2-laser of 337 nm) and high- rated from the vesicle suspension through a Sephadex G-25 filtration
resolution ESI-MS model (LTQ-FT Ultra, Thermo Scientific) (see Supple- column eluted with buffer containing 10 mM HEPES, 150 mM NaCl,
mentary data, Table S1). and 0.1 mM EDTA, pH 7.4. Leakage of intraliposomal CF was assayed
by treating the probe-loaded liposomes (final lipid concentration,
2.3. Synthesis of modified polyglutamic acid polymers (PGAmod) 0.125 mM) with the appropriate amount of peptide or conjugate in Co-
star 3797 round-bottom 96-well plates, each well containing a final vol-
The pH of a solution of poly(L-glutamic acid) (PGA) sodium was ume of 100 μl. Leakage was measured at various peptide-to-lipid ratios.
lowered to obtain its proton form. After centrifugation, the precipitate The micro titer plate was incubated at RT for 1 h to induce dye leakage.
was lyophilized and dehydrated in a vacuum drying oven in the pres- Leakage was measured at various peptide-to-lipid ratios. Changes in
ence of phosphorus pentoxide. The protonated PGA form (50 mg), N- fluorescence intensity were recorded using the FL600 fluorescence mi-
(2-Aminoethyl)maleimide trifluoroacetate salt (19 mg), Methoxy PEG croplate reader with excitation and emission wavelengths set at 492
Amine HCl Salt MW 5 kDa (74 mg), Lissamine RhoB sulfonyl-GK and 517 nm, respectively. One hundred percent release was achieved
amidated peptide (4 mg), and O-(7-azabenzotriazol-1-yl)-N,N,N′,N′- by adding Triton X-100 to a final concentration of 1% v/v to the microti-
tetramethyluronium hexafluorophosphate (HATU) (100 mg) were dis- ter plates. Fluorescence measurements were made initially with probe-
solved in anhydrous N,N-dimethylformamide (DMF) (5 ml). Finally, loaded liposomes, afterwards by adding peptide or conjugate solution
ethanolamine (12 μl) and 4-Methylmorpholine (NMM) (100 μl) were and, finally by adding Triton X-100 to obtain 100% leakage. The results
added. The stoichiometry of PGA:Maleimide:PEG5 kDa:RhoGK:Ethanol- were expressed as percentage of CF released relative to the positive con-
amine was 1:25:5:2:65 mol%. The reaction was left at RT for 24 h. The trol (Triton X-100).
reaction was dialyzed against distilled water using a dialysis bag
(cut-off 6–8 kDa) for 3 h. Immediately after, it was purified by SEC 2.7. Red blood cell lysis assay
(Sephadex® G-25). The lyophilized sample was analyzed by 1H-NMR
at 400 MHz (Varian Mercury) and 800 MHz (Bruker Digital Avance) Human blood was collected in 10 ml EDTA Vacutainer tubes. A small
(see Supplementary data, Figs. S2 and S3 and Table S2). aliquot was assessed for evidence of hemolysis by centrifugation at 800 g
for 10 min, and non-hemolyzed samples were carried forward into the
2.4. Formation of PGA-peptide conjugates assay. Red blood cells (RBCs) were washed three times in PBS pH 7.4 by
centrifugation at 800 g for 10 min and resuspending in the same buffer
The reaction between the modified PGA (PGAmod) and peptides with to yield a 10× dilution. RBCs were then diluted in appropriate pH buffer
cysteine at the N-terminus, took place in 1 ml of 100 mM Tris buffer at to yield approximately ± 15 × 107 cells/100 μl PBS in Costar 3797
pH 7.0 and RT for 2 h. An example of reaction would be mixing PGAmod round-bottom 96-well plates for the lysis assay. The micro titer plate
(6 mg) with Peptide 6 (1.5 mg) and Peptide 13 (0.6 mg), where the stoi- was covered with a low evaporation lid and incubated at 37 °C for 1 h
chiometry was 1:4:3 (see Table 2 for all the conjugates accomplished). to induce hemolysis. Negative controls were PBS, while positive con-
Upon completion of the reaction, an excess of cysteamine was added trols were 1% v/v solution of Triton X-100 (100% lysis). The plate was
to ensure that no free reactive maleimides were present in the final then centrifuged at 800 g for 10 min and 80 μl of supernatants were
conjugate. The PGAmod-peptide(s) conjugates were purified using a transferred to a Costar 3632 clear bottom 96-well plate. Hemoglobin ab-
Sephadex® G-25 column. sorbance was read at 560 nm using the ELx800 absorbance microplate
reader. The results were expressed as percentage of hemoglobin re-
2.5. Amino acid analysis leased relative to the positive control (Triton X-100).
See the rest of Materials and methods in the Supplementary data.
The conjugated amount of peptide along PGAmod was determined
through amino acid analysis. 0.5 mg of each sample was hydrolyzed 3. Results and discussion
with 6 N HCl at 110 °C for 18 h. Later, the samples were evaporated
and suspended in 0.5 ml of 20 mM HCl with an internal standard of 3.1. Design, synthesis and characterization of the pro-cytotoxic conjugates
AABA 2.5 mM standard (Pierce Amino Acid Standard H, Thermo Scien-
tific). Amino acid analysis was performed at the Scientific and Techno- The choice of lytic peptide was based on the cytolytic-activity/length
logical Centers of the University of Barcelona (CCiT UB), using the ratio of several cytotoxic peptides. We chose the 14-mer cytolytic pep-
AccQTag pre-column derivatization method. Reaction of amino acids tide Mitoparan, which exhibits mitochondriotoxic and apoptogenic
with 6-aminoquinolyl-N-hydroxysuccinimidyl carbamate yields deriva- properties and does not induce cancer drug resistance [10]. Given that
tives that are detected at 254 nm. 20 μl of the derivatized samples or stan- the induction of apoptosis is the best way to prevent unwanted immune
dards was injected in a column of Nova-Pak C18 4 μm, 3.9 × 150 mm responses [11], and to minimize damage and disruption of neighboring
(Waters). Data acquisition and treatment was done using the ChromPass cells, a method based on a selective activable Mitoparan (targeted pro-
Chromatography Data System (Jasco). Mitoparan) is an elegant way to efficiently kill tumor cells. Since an
enzyme-catalyzed degradation combines chemical stability and high
2.6. Leakage measurement specificity, we used peptide sequence-based linkers to achieve cytolytic
peptide release. We compared the efficacy of two enzymatic cleavage
Aliquots containing the appropriate amount of lipid in chloroform/ sites triggered by specific overexpressed tumor enzymes, namely:
methanol (1:1, v/v) were placed in a test tube, the solvents were re- MMP-2 and cathepsin B. MMP-2 is known to be overexpressed on the
moved by evaporation under a stream of O2-free nitrogen, and finally cell surface in several cancers and in tumor microvascular endothelial
traces of solvents were eliminated under vacuum in the dark for more cells. In addition, it has been strongly associated with the progression
than 3 h. After, 1 ml of buffer containing 10 mM HEPES, 100 mM NaCl, of malignancy in several types of carcinoma [12]. In cancer cells and
0.1 mM EDTA, pH 7.4 buffer and CF at a concentration of 40 mM was also tumor microvascular endothelial cells, cathepsin B is thought to
16 M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21

Table 1
Sequences of the peptides used to conjugate the polymer.
Peptides Names Sequence MW (Da) Charge[a]
1 Labeling peptide Rh-GK 742 +1
2 Labeling mitoparan CGGGIPVSLRSKGKGINLKKLAKL(Aib)KKIL 3361 +7
3 Labeling mitoparan CGGGFL(PABC)GKGINLKKLAKL(Aib)KKIL 2859 +5
4 D-Mitoparan CGinlkklakl(Aib)kkil 1766 +5
5 D-Mitoparan CGGIPVSLRSKGinlkklakl(Aib)kkil 2761 +7
6 D-Mitoparan CGPPPPPPPPPGIPVSLRSKGinlkklakl(Aib)kkil 3634 +7
7 D-Mitoparan CGGFL(PABC)GGinlkklakl(Aib)kkil 2260 +5
8 D-Mitoparan CGPPPPPPPPPGFL(PABC)GGinlkklakl(Aib)kkil 3133 +7
9 Labeling D-mitoparan CGPPPPPPPPPGIPVSLRSKGKGinlkklakl(Aib)kkil 4177 +7
10 CGPPG-penetratin CGPPGRQIKIWFQNRRMKWKK 2655 +7
11 CGY-TAT CGYGRKKRRQRRR 1718 +8
12 C-SAP(E) CVELPPPVELPPPVELPPP 2018 −3
13 Targeting peptide CGPPPPPPPPPGLTVSPWY 1954 0
[a] Global Charge at pH 7.4. All peptides are C-amided. The cleavage site is underlined: GIPVSLRSK for MMP-2 and GFL(PABC) for cathepsin B. Minor capital letters represent
D-amino acids.
Rh: Lissamine Rhodamine B sulfonyl Aib: α-aminoisobutyric acid
K: Lysine with 5(6)-carboxyfluorescein at its side chain PABC: 4-aminobenzoic acid

be overexpressed at the extra- and intracellular levels, being located only with ethanolamine and maleimide was used to assist the spectro-
mainly in the endo/lysosomal compartments [13]. scopic assignment of the totally functionalized system. All NMR signals
Here we report on an easily synthesizable peptide platform based on were assigned on the basis of chemical shift and intensity data and,
PGA polymer as a scaffold to carry cytolytic peptides. We attached a few when required, confirmed by homonuclear double resonance experi-
copies of the cytotoxic positively charged Mitoparan along with the ments (Fig. S3B). The relative number of bound groups was determined
naked PGA by selective ligation methods such as Native Chemical Liga- from the relative integration of their 1H-NMR signals in relation to those
tion (NCL) and maleimide-thiol coupling chemistry. Although the yield associated with the PGA protons (Table S2). Comparing the measured
of the peptide-polymer conjugation was significant using both strate- integral intensities of the resonance signals at 6.85 ppm (2 aromatic
gies (~50%), the final products were insoluble. The positively charged protons per maleimide unit), at 3.31 ppm (CH2-N protons of ethanol-
Mitoparan attached to the negatively charged PGA may not have been amine) and at 2.22–2.42 ppm (γ protons of the PGA backbone), we
the best combination. To gain insight into why the final product was ag- could be estimated that around 23% and 67% of the glutamates were
gregated, we conjugated PGA with a negatively charged model peptide. functionalized with maleimide and ethanolamine, respectively. Similar-
After the attachment of this peptide, the final peptide-PGA product was ly, from the measured integral of the signal at 3.70 ppm and assuming
completely soluble (data not shown). Given this result, we postulate 85 ethylene glycol moieties per PEG5 kDa group (4 methylene protons
that the strength of the ionic interactions between the glutamic acids per moiety), it estimated that PEG was incorporated in around 7%
of PGA and the lysine residues of Mitoparan is strong enough to induce of the glutamate residues. Combining the NMR and MALS data from
aggregation in the final product. Several easy and reproducible chemical the 124 Glu side-chains available in the starting 16 kDa PGA (see
modifications were performed along the naked PGA (Fig. 1a) in order to Table S3), 28 were found to be functionalized with maleimide, 8 with
improve the solubility of the final products and to prevent aggregation. PEG5 kDa and 83 capped with ethanolamine. Furthermore, we checked
Part of the negative charges on PGA was capped by ethanolamines, thus the structural conformation by circular dichroism (CD) of PGAmod at a
changing carboxylic for hydroxyl groups. Moreover, molecules of poly- several pH values. It was similar to naked PGA, having α-helical confor-
ethylene glycol of 5 kDa (PEG5 kDa) were incorporated into the polymer. mation at low pH and random coil at neutral pH (Supplementary data,
Not only does the hydrophilic character of PEG make the system more Fig. S6). Below pH ~ 6, it tended to adopt an α-helical conformation
soluble and increase the blood circulation half-life, but also it prevents (two minimum peaks around 208 and 222 nm), because of protonated
the uptake of PEGylated macromolecules by the mononuclear phago- side chains, and thus this uncharged form allowed amino acids to be
cytic system [14], although this last issue is totally speculative because brought closer together. However, at higher pH values, the main confor-
it has not been checked in this work. Furthermore, the polymer showed mation tended to be random coil, because side chains are largely ionized
red fluorescence with peptide 1 (Table 1) for cellular uptake assays. Fi- with negative charge, which repel each. Similar behavior was observed
nally, N-(2-aminoethyl) maleimide was also incorporated along the for PGAmod, although the tendency to form α-helical at low pH was
system to be ready for subsequent conjugation of Cys-peptides through hampered. At low pH, the capacity to form hydrogen bonds between
thiol-maleimide chemistry. the carboxylic groups of Glu side chains is impede by PGA modifications.
In order to characterize the molecular weight of PGAmod, we used Finally, several Cys-peptides were conjugated to the PGAmod
SEC-MALS-IR (Supplementary data, Figs. S4 and S5 and Table S3). Sig- (Table 2), and cytotoxic activity was evaluated. As an example, the con-
nals from the RI detector combined with MALS data were used to calcu- jugate PGAmod:6:13 (see Section 3.4) was characterized by SEC-MALS-
late the molecular mass of PGAmod. The cumulative mass fractions were IR to determine the MW which was 75 kDa (Table S3). This is an ideal
plotted against molar mass in Fig. S5, depicting molecular weight distri- size for increasing the blood half-life and the passive tumor targeting
butions. These measurements indicate that the PGAmod has a MW known as enhanced permeability and retention (EPR) [15].
around 60 kDa. In addition, a detailed 1H-NMR study to characterize
the PGAmod was carried out (Supplementary data, Figs. S2 and S3 and 3.2. In vitro cytotoxicity of Mitoparan and Mitoparan-conjugates
Table S2). Unequivocal assignment of all the signals was not possible
at 400 MHz (Fig. S2B) due to overlapping. In contrast, at 800 MHz the We first studied two modifications of Mitoparan peptide with differ-
system showed a better chemical shift (Fig. S3C). The PGA derivatized ent cleavage sequences at the N-terminus (Table 1, Peptides 2 and 3) in
M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21 17

Table 2
PGA, modified PGA (PGAmod), and peptide-PGAmod conjugates.

Samplesa Stoichiometry (feeding ratio) Stoichiometry (final ratio)b Conjugation yield (%) Zeta Potential (mV)

PGA – – – −30
PGAmod – – – −5
PGAmod:2 1:4 1:2.23 55.7 +1
PGAmod:3 1:4 1:2.14 53.5 +0.5
PGAmod:4 1:4 1:2.37 59.2 +1
PGAmod:5 1:4 1:2.19 54.7 −1
PGAmod:6 1:4 1:2.35 58.7 −0.5
PGAmod:7 1:4 1:2.27 56.7 −1
PGAmod:8 1:4 1:2.11 52.7 −0.5
PGAmod:10 1:4 1:2.53 63.2 +3
PGAmod:11 1:4 1:2.39 59.7 +3
PGAmod:12 1:4 1:3.2 80 −6
PGAmod:13 1:3 1:2.73 91 −5
PGAmod:6:10 1:4:4 1:2.11:1.88 52.7 (pept 6); 47 (pept 10) −0.5
PGAmod:6:13 1:4:3 1:2.23:2.34 55.7 (pept 6); 78 (pept 13) −1
PGAmod:8:10 1:4:4 1:1.98:1.89 49.5 (pept 8); 47.2 (pept 10) +1
PGAmod:8:13 1:4:3 1:2.33:2.45 58.2 (pept 8); 81.6 (pept 13) −0.5
PGAmod:9:13 1:4:3 1:1.86:2.32 46.5 (pept 9); 77.3 (pept 13) −1
a
Numbers represent peptides from Table 1.
b
The conjugated amount of peptide along PGAmod was determined through amino acid analysis.

order to compare the activity of the two peptides alone and when at- N-terminal cleavage site but also the two additional amide positions
tached to the PGAmod (Table 2, samples PGAmod:2 and PGAmod:3). A along the Mitoparan peptide per se. The peptide fragmentation along
non-cytotoxic effect was observed for both Mitoparan-PGAmod conju- the sequence thus renders Mitoparan inactive. We postulate that the
gates. However, a concentration-dependent decrease in SK-BR3 cell overexpressed cathepsin B, which is located at extra- and intracellular
viability was promoted by the free peptides (Supplementary data, levels in the tumor, would be the main protease to cleavage and
Fig. S7a). To further explore the cytotoxic effect live cell images were ex- would subsequently inactivate Mitoparan. However, MMP-2 accurately
amined by confocal laser scanning microscopy (CLSM) (Supplementary cleaved the peptide through the cleavage site, as has been previously
data, Fig. S7b), which revealed an intracellular distribution of free la- described [16,17].
beled peptides and location in the nuclei. As expected, nuclear DNA
fragmentation indicative of apoptosis was detected, as was damaged 3.3. In vitro cytotoxicity of D-Mitoparan and D-Mitoparan conjugates
the cell integrity (cell shrinkage and blebbing). However, in cells
treated with Mitoparan-PGAmod conjugates, which were not dam- An enantiomer of Mitoparan has recently been characterized [18],
aged, colocalization of Mitoparan (green) and PGAmod (red) was ob- revealing that it maintains similar cytotoxic properties with the addi-
served, indicating that they were engulfed in the same endosome, tional protease-resistant feature. Taking advantage of this property,
remaining together until lysosomal enzymatic digestion. There are two we synthesized a battery of new enantiomer Mitoparan peptides to re-
possible non-mutually exclusive explanations for the non-cytotoxic ef- place L-Mitoparan. For this purpose, the C-terminal peptide sequence,
fect observed for the Mitoparan-conjugates: 1) MMP-2 and/or cathepsin corresponding to the 14 amino acids of Mitoparan per se, was synthe-
B were inaccessible to the cleavage sites, and therefore the Mitoparan sized with D-amino acids (Table 1, peptides 4–9). Hereinafter this enan-
peptides remained attached to the polymer, which made it inactive; or tiomer will be referred to as D-Mitoparan. In addition, we synthesized
2) Mitoparan was digested at the same rate as the enzymatic cleavage D -Mitoparan peptides with 9 Pro as a spacer (Pro9) to place the
sites, thus losing cytotoxicity. To further address these two scenarios, cleavage site far from the PGA scaffold (Table 1, peptides 6, 8 and
we performed enzymatic degradation assays of the two peptides (Sup- 9), thus decreasing the possibility of the PGA polymer shielding the
plementary data, Fig. S8). N-terminal cleavage site from proteases.
Analysis of the HPLC chromatograms and the cleavage fragments by We compared the pro-apoptotic ability of the new D-Mitoparan con-
MALDI-TOF revealed that cathepsin B is capable of splitting not only the jugates with or without the Pro9 spacer (Fig. 2). For free D-Mitoparan

Fig. 2. Comparative MTT viability measurements of SK-BR3 (HER2+) cells after a 24 h incubation with free D-peptides and PGA-D-peptide conjugates (see Tables 1 and 2). Data are
expressed as the mean ± SE (n = 3). IC50 values are shown in the right table. Similar cytotoxic effects to peptides 6 and 8 were observed for peptides 4, 5, 7 and 9.
18 M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21

Fig. 3. a) Confocal laser scanning microscopy of SK-BR3 cells after 1 h of co-incubation at 37 °C in CO2 with 25 μM of PGAmod conjugates and subsequent washing with medium. The red
fluorescence (maximum intensity projection) belongs to Rhodamine-labeled PGAmod. b) Flow cytometry results of the co-incubation of SK-BR3with 25 μM of PGAmod conjugates for 1 h
and subsequent washing with medium.

peptides (6 and 8), the IC50 was similar to that of L-Mitoparan (~8 μM). the need to release D-Mitoparan to promote cellular apoptosis. A compar-
The IC50 values of D-Mitoparan-PGAmod conjugates (see Fig. 2) were 5 ison of the activity of the D-Mitoparan-PGAmod conjugates (Table 2, conju-
times higher than those of free D-Mitoparan. However, consistent with gates PGAmod:4–8) revealed a cytotoxic improvement for conjugates with
our supposition, the conjugate PGAmod:4, which did not carry an enzy- a Pro9 spacer, namely conjugates PGAmod:6 and PGAmod:8 (Fig. 2). A good
matic cleavage site, did not show any cytotoxic activity, thereby indicating correlation between activity and D-Mitoparan release was observed when

Fig. 4. Comparative MTT viability measurements of SK-BR3 (HER2+) cells after a 24 h incubation with free D-peptides and PGA-D-peptide conjugates (see Tables 1 and 2). Data are
expressed as the mean ± SE (n = 3). IC50 are shown in the right table, these values refer to peptides 6 or 8. Similar cytotoxic effects to peptides 6 and 8 were observed for peptides 4,
5, 7 and 9.
M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21 19

Fig. 5. Comparative MTT viability measurements of MDA-231 (HER2-) cells after a 24 h incubation with PGA-D-peptide conjugates (see Table 2). Data are expressed as the mean ± SE (n = 3).
IC50 values are shown in the right table, these values refer to peptides 6 or 8.

comparing the cytotoxicity of PGAmod:6 and PGAmod:8 (Fig. 2) with their system (Fig. 3, PGAmod:13). Our system attached to the cellular surface,
enzymatic digestion (Supplementary data, Fig. S9). where the HER2 receptor is located. After this binding, cells internalized
the conjugate by receptor-mediated endocytosis, thus promoting the
3.4. Improving the efficacy of pro-cytotoxic conjugates by enhancing cellular cellular uptake of our system, which showed a satisfactory rate of inter-
uptake nalization (Fig. 3b).
Having improved the cellular uptake, we designed systems with two
To continue further in the amendment of our cytotoxic conjugates, different peptides in the same carrier: D-Mitoparan and Penetratin or
we enhanced the cellular uptake of the conjugates in order to improve targeting peptide (see the combinations in Table 2). The presence of
their activity. Several studies have reported the efficiency of cell- Penetratin increases the internalization of our system, thus allowing
penetrating peptides (CPPs) in facilitating the translocation of cargoes the maximum amount taken up into lysosomes, where cathepsin B
across the plasma membrane [19]. However, although CPPs are highly could then release D-Mitoparan. The cytotoxicity observed after the con-
efficient in mediating the cell uptake of cargoes into most cell lines, jugation of Penetratin or the targeting peptide on the D-Mitopara-
the lack of cell specificity could be a serious problem in vivo. For this rea- PGAmod conjugates was doubled that with the other conjugates
son, we performed parallel studies with a targeting peptide to compare (Fig. 4). Systems with Penetratin showed a slightly higher — but not sig-
the specificity and affinity of our modified systems (Table 1, peptide 13). nificant cytotoxic effect than with targeting peptide. Surprisingly, a sim-
Peptide 13, a selected peptide (LTVSPWY) discovered by phage display ilar cytotoxic effect was detected for systems with different cleavage
technology [20], to which we introduced a Pro9 spacer and a Cysteine at sites (MMP-2 and cathepsin B). Performing a digestion of peptide 6,
its N-terminus, targets the over-expressed epidermal growth factor which holds the MMP-2 cleavage site (GIPVSLRSK), we found that
receptor 2 (HER2). This tyrosine kinase receptor promotes the growth cathepsin B was capable to cut it at two sites, thereby promoting the re-
of certain breast cancer cells, which tend to be more aggressive than lease of D-Mitoparan (Supplementary data, Table S4). This observation
other types of breast cancer [21]. For the development of this proof- implies that the sequence designed to be cut by MMP-2 can also be
of-concept we used the SK-BR3 cell line, which is HER2+ [20]. digested by cathepsin B, thus increasing the probability of D-Mitoparan
Three well-known CCPs, two positively charged (Penetratin [22] and to be released either on the cell surface by MMP-2 or in lysosomes inside
TAT [23]) and one negatively charged (SAP(E) [24]) were selected to de- the cell by cathepsin B. In addition, in order to study the specificity of the
termine the best option for our system (Table 1, peptides 10–12). Cellu- targeting peptide, we checked our pro-cytotoxic system with or without
lar uptake was quantified by fluorescence measurements using flow this targeting peptide in the presence of a non-overexpressing HER2
cytometry and CLSM (Fig. 3). Penetratin (see Table 1, peptide 10) was breast cancer cell line, namely MDA-231 cells [20]. No significant im-
significantly more efficient than the other CPPs at internalizing the sys- provement in cytotoxic activity was observed when targeting peptide
tem into the cell (Fig. 3). A closer look at the system distribution re- was present, thus validating the specificity of this peptide against the
vealed that the conjugates were within endosomes and lysosomes. HER2 receptor (Fig. 5).
Therefore, all the CPPs checked internalized PGAmod via the endosomal CLSM was used to verify the cleavage and subsequent release of
mod
pathway, the lysosome being the final fate of these conjugates. Similar D-Mitoparan from the PGA . Fig. 6 shows the localization of green la-
results were seen when the targeting peptide was conjugated to our beled D-Mitoparan (peptide 9 in Table 1) and red labeled polymer in the

Fig. 6. Confocal laser scanning microscopy (CLSM) of SK-BR3 cells after 6 h of co-incubation at 37 °C in CO2 with PGAmod:9:13 conjugate, where the concentration of peptide 9 was 25 μM.
The cells were washed with media, before taking the pictures. The fluorescence is depicted as the maximum intensity projection, where red fluorescence is due to Rhodamine-labeled
PGAmod and green fluorescence to carboxyfluorescein-labeled peptide 9 and, finally, yellow shows the colocalization. A video related to the effect of PGAmod:9:13 conjugate in SK-BR3
cells is attached in Supplementary Information (Video S1).
20 M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21

Fig. 7. Dye leakage and red blood cell hemolysis of free D-peptides and PGAmod-D-peptide conjugates. (a) Effect on membrane rupture, i.e., leakage, of Large Unilamellar Vesicles (LUVs)
containing EPC/Chol at a molar ratio of 5:1 for 1 h at RT. (b) Effect on the hemoglobin release from red blood cells for 1 h at 37 ºC. Data are expressed as the mean ± SE (n = 3). For the
conjugates, the effect on membrane rupture and hemoglobin release refers to peptide 6 or 8. Similar dye leakage and hemolysis effects to peptide 4 were observed for peptides 5, 6, 7, 8 and 9.

SK-BR3 cells after 6 h of incubation with PGAmod:9:13. We observed yel- peptide 4 (Fig. 7b), but moderate hemoglobin release was promoted by
low spots, which indicate colocalization of the two labels inside dotted the conjugates.
vesicles, corresponding to endosomes and lysosomes, but also the diffu-
sion of green-labeled D-Mitoparan in the cytoplasm and even in the cel- 4. Conclusions
lular nucleus.
A video was recorded to observe the behavior of our system (Supple- Cytolytic peptides are promising drugs for cancer treatment due to
mentary Video S1). Images were acquired every 20 min for 9 h and their high cytotoxic efficacy and because they do not induce cancer
20 min (the time in hours is shown in the top left of the video). Dur- drug resistance. However, they have the drawback that they are non-
ing this time, green (carboxyfluorescein-labeled peptide 9) and red specific and easily degradable in blood. Here we report an example of
(rhodamine-labeled PGAmod) fluorescence were observed both inside a smart, targeted, pro-cytotoxic system which carries Mitoparan in a
(dotted color) and outside the cells. Inside the cells, red dotting was safe way, inhibiting its cytotoxic effect when it is attached to the modi-
more abundant than green. However, outside the cells, green showed fied polymer. We demonstrate the innocuousness of our pro-cytotoxic
greater fluorescence than red. These observations are explained by the system in the first hours of contact with tumor cells. After only 3 h,
fact that the two probes are sensitive to changes in pH (fluorescence when a significant amount of the pro-cytotoxic system had been
of carboxyfluorescein decreases at acidic pH, whereas rhodamine does endocyted and the Mitoparan had been released by proteases, the cyto-
so at neutral-basic pH). Significant changes in cell morphology occurred toxic effects could be observed (see Video S1 attached). To overcome
after 3 h of incubation. Cells started to shrink when a cytotoxic concen- solubility issues, it was necessary to modify the PGA by incorporating
tration of peptide 9 was present in the cytoplasm. Peptide 9 was cleaved PEG and capping negative charges with ethanolamines. In addition,
from the PGAmod and released from lysosomes to reach the cytoplasm, the enantiomer form of Mitoparan was used to maximize the cytotoxic
promoting apoptosis. From 6 h of incubation, we observed a significant effect. Not only does the system include targeting peptide to favor accu-
increase in green fluorescence on the cytoplasm, presumably because of mulation in the tumor, but also a cleavage site designed to be cut specif-
the irreversible and accumulated damage of lysosomes. At this time ically by proteases overexpressed only in tumor cells, such as MMP-2
point, to facilitate monitoring of intracellular fluorescence, we added and cathepsin B. This strategy allows the cytotoxic peptide to be
fresh cellular medium, thus diluting the fluorescence outside cells. released with exquisite spatiotemporal control. From the pharmacolog-
This approach allowed us to see the spread of peptide 9 through the cy- ical viewpoint, our large targeted pro-cytotoxic system of 75 kDa, which
toplasm and even in the nucleus. carries only 2 cytotoxic peptides with a cytotoxic effect in the micromo-
lar range, could be considered insufficiently loaded. However, we con-
sider that the combined effect of targeting to HER2 + breast tumors
3.5. Lipid perturbation of liposomal membrane and in vitro via the targeting peptide and its large MW would increase its blood
hemocompatibility testing half-life and favor the well-documented EPR effect enhancing the local
concentration of the cytolytic peptide at the tumor site. Our results
The soluble systems designed in this study are neutral at physiolog- highlight the potential of a simple, targeted, pro-cytotoxic carrier for
ical pH (Table 2), having electrical charges of opposite signs (zwitter- clinical use in cancer treatment. Further studies are ongoing to test the
ionic). This neutral charge could be a positive feature because highly innocuous nature and efficacy of our system in vivo.
positive particles interact with serum proteins, thus increasing the ag- Supplementary data to this article can be found online at http://dx.
gregation of the protein and affecting its biodistribution in vivo [25], doi.org/10.1016/j.jconrel.2014.03.005.
while negatively charged ones are impeded to promote cellular uptake.
To predict the compatibility of our systems with biological environ- Acknowledgement
ments, we characterized their capacity to perturb the bilayer of lipo-
somes and to cause cell hemolysis. Dye leakage results (Fig. 7a) show Thanks go to Dr. Lidia Bardia for the technical support with cell im-
that the free D-Mitoparan (peptide 4) perturbed membrane phospho- aging and useful discussions, the IRB Advanced Digital Microscopy
lipids, inducing leakage at very low peptide/lipid ratios, reaching a maxi- (ADM) Core Facility, and Dr. Jesús García for the technical support
mum around the ratio of 0.01. Comparing this effect with the conjugates, with NMR data and useful discussions. We also acknowledge the Scien-
we observed that conjugates released only 50% of dye at the maximum tific and Technological Centres of the University of Barcelona (CCiT UB)
ratio tested (0.04), the leakage being slightly higher when Penetratin Separation Techniques Analysis Facility, the NMR Facility, and the Flow
was present. Complementary to this, robust hemolysis was observed for Cytometry Facility. We also thank Laura Nevola (IRB Barcelona) for
M. Moreno et al. / Journal of Controlled Release 182 (2014) 13–21 21

fruitful scientific discussions. We acknowledge financial support from [11] R.C. Taylor, S.P. Cullen, S.J. Martin, Apoptosis: controlled demolition at the cellular
level, Nat. Rev. Mol. Cell Biol. 9 (2008) 231–241.
MICINN-FEDER (BIO2008-799) and the Generalitat de Catalunya (XRB [12] A. Talvensaari-Mattila, P. Paakko, T. Turpeenniemi-Hujanen, Matrix metalloproteinase-
and 2009SGR-1005). 2 (MMP-2) is associated with survival in breast carcinoma, Br. J. Cancer 89 (2003)
1270–1275.
[13] K. Brix, A. Dunkhorst, K. Mayer, S. Jordans, Cysteine cathepsins: cellular roadmap to
References different functions, Biochimie 90 (2008) 194–207.
[14] D.E. Owens III, N.A. Peppas, Opsonization, biodistribution, and pharmacokinetics of
[1] C. Leuschner, W. Hansel, Membrane disrupting lytic peptides for cancer treatments, polymeric nanoparticles, Int. J. Pharm. 307 (2006) 93–102.
Curr. Pharm. Des. 10 (2004) 2299–2310. [15] H. Maeda, Macromolecular therapeutics in cancer treatment: the EPR effect and be-
[2] L. Holle, W. Song, E. Holle, Y. Wei, J. Li, T.E. Wagner, X. Yu, In vitro- and in vivo- yond, J. Control. Release 164 (2012) 138–144.
targeted tumor lysis by an MMP-2 cleavable melittin-LAP fusion protein, Int. J. [16] N. Yamamoto, N.S. Bryce, N. Metzler-Nolte, T.W. Hambley, Effects of enzymatic acti-
Oncol. 35 (2009) 829–835. vation on the distribution of fluorescently tagged MMP-2 cleavable peptides in can-
[3] L. Holle, W. Song, E. Holle, Y. Wei, T. Wagner, X. Yu, A matrix metalloproteinase 2 cer cells and spheroids, Bioconjug. Chem. 23 (2012) 1110–1118.
cleavable melittin/avidin conjugate specifically targets tumor cells in vitro and [17] B.E. Turk, L.L. Huang, E.T. Piro, L.C. Cantley, Determination of protease cleavage site
in vivo, Int. J. Oncol. 22 (2003) 93–98. motifs using mixture-based oriented peptide libraries, Nat. Biotechnol. 19 (2001)
[4] Y. Yamada, Y. Shinohara, T. Kakudo, S. Chaki, S. Futaki, H. Kamiya, H. Harashima, Mi- 661–667.
tochondrial delivery of mastoparan with transferrin liposomes equipped with a pH- [18] S. Jones, J. Howl, Enantiomer-specific bioactivities of peptidomimetic analogues of
sensitive fusogenic peptide for selective cancer therapy, Int. J. Pharm. 303 (2005) mastoparan and mitoparan: characterization of inverso mastoparan as a highly effi-
1–7. cient cell penetrating peptide, Bioconjug. Chem. 23 (2012) 47–56.
[5] N.R. Soman, G.M. Lanza, J.M. Heuser, P.H. Schlesinger, S.A. Wickline, Synthesis and [19] I. Martin, M. Teixido, E. Giralt, Intracellular fate of peptide-mediated delivered
characterization of stable fluorocarbon nanostructures as drug delivery vehicles cargoes, Curr. Pharm. Des. 19 (2013) 2924–2942.
for cytolytic peptides, Nano Lett. 8 (2008) 1131–1136. [20] M. Shadidi, M. Sioud, Identification of novel carrier peptides for the specific delivery
[6] N.R. Soman, S.L. Baldwin, G. Hu, J.N. Marsh, G.M. Lanza, J.E. Heuser, J.M. Arbeit, S.A. of therapeutics into cancer cells, FASEB J. 17 (2003) 256–258.
Wickline, P.H. Schlesinger, Molecularly targeted nanocarriers deliver the cytolytic [21] J. Kao, K. Salari, M. Bocanegra, Y.L. Choi, L. Girard, J. Gandhi, K.A. Kwei, T. Hernandez-
peptide melittin specifically to tumor cells in mice, reducing tumor growth, J. Clin. Boussard, P. Wang, A.F. Gazdar, J.D. Minna, J.R. Pollack, Molecular profiling of breast
Invest. 119 (2009) 2830–2842. cancer cell lines defines relevant tumor models and provides a resource for cancer
[7] R. Duncan, Polymer therapeutics as nanomedicines: new perspectives, Curr. Opin. gene discovery, PLoS One 4 (2009) e6146.
Biotechnol. 22 (2011) 492–501. [22] L. Theodore, D. Derossi, G. Chassaing, B. Llirbat, M. Kubes, P. Jordan, H. Chneiweiss, P.
[8] C. Li, Poly(L-glutamic acid)—anticancer drug conjugates, Adv. Drug Deliv. Rev. 54 Godement, A. Prochiantz, Intraneuronal delivery of protein kinase C pseudosubstrate
(2002) 695–713. leads to growth cone collapse, J. Neurosci. 15 (1995) 7158–7167.
[9] L. Mondragon, M. Orzaez, G. Sanclimens, A. Moure, A. Arminan, P. Sepulveda, A. [23] G. Tunnemann, R.M. Martin, S. Haupt, C. Patsch, F. Edenhofer, M.C. Cardoso, Cargo-
Messeguer, M.J. Vicent, E. Perez-Paya, Modulation of cellular apoptosis with apoptotic dependent mode of uptake and bioavailability of TAT-containing proteins and pep-
protease-activating factor 1 (Apaf-1) inhibitors, J. Med. Chem. 51 (2008) 521–529. tides in living cells, FASEB J. 20 (2006) 1775–1784.
[10] S. Jones, C. Martel, A.S. Belzacq-Casagrande, C. Brenner, J. Howl, Mitoparan and [24] I. Martin, M. Teixido, E. Giralt, Design, synthesis and characterization of a new anion-
target-selective chimeric analogues: membrane translocation and intracellular re- ic cell-penetrating peptide: SAP(E), Chembiochem 12 (2011) 896–903.
distribution induces mitochondrial apoptosis, Biochim. Biophys. Acta 1783 (2008) [25] F. Alexis, E. Pridgen, L.K. Molnar, O.C. Farokhzad, Factors affecting the clearance and
849–863. biodistribution of polymeric nanoparticles, Mol. Pharm. 5 (2008) 505–515

You might also like