Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
RATIONAL MODIFICATIONS OF CELLPENETRATING PEPTIDES FOR DRUG DELIVERY Applications in tumor targeting and oligonucleotide delivery Maarja Mäe Rational modifications of cell-penetrating peptides for drug delivery Applications in tumor targeting and oligonucleotide delivery Maarja Mäe Cover picture: Stefanie Leuker ©Maarja Mäe, Stockholm 2009 ISBN 978-91-7155-792-6 Printed in Sweden by Universitetsservice, Stockholm 2009 Distributor: Department of Neurochemistry, Stockholm University To my beloved mother List of Publications This thesis is based on the following publications, referred to in the text by the corresponding Roman numerals: I Myrberg H, Zhang L, Mäe M and Langel Ü Design of a tumor-homing cell-penetrating peptide Bioconjugate chemistry, 2008; 19(1): 70-75. II Mäe M, Myrberg H, EL Andaloussi S and Langel Ü Design of a tumor-homing cell-penetrating peptide for drug delivery International Journal of Peptide Research and Therapeutics, 2009. In press. III Mäe M, Enbäck J, Laakkonen P, Rosenthal Aizman K, Langel Ü and Lindgren M Target-activated cell-penetrating peptide Manuscript. IV Mäe M, EL Andaloussi S, Lundin P, Johansson HJ, Guterstam P and Langel Ü A stearylated CPP for delivery of splice-correcting oligonucleotides using a non-covalent co-incubation strategy Accepted in Journal of Controlled Release. Additional publications I Mäe M,* Myrberg H,* Jiang Y, Paves H, Valkna A and Langel Ü Internalisation of cell-penetrating peptides into tobacco protoplasts Biochimica et Biophysica Acta, 2005; 1669(2), 101-107. II Mäe M and Langel Ü Cell-penetrating peptides as vectors for peptide, protein and oligonucleotide delivery Current Opinion in Pharmacology, 2006; 6(5): 509-14. III Johansson HJ, EL Andaloussi S, Holm T, Mäe M, Jänes J, Maimets T and Langel Ü Characterization of a novel cytotoxic cell-penetrating peptide derived from p14ARF protein Molecular Therapy, 2008; 16(1): 115-23. *These authors contributed equally to this work. Abstract High molecular weight biomolecules are becoming increasingly important in the development of new therapeutics. However, the hydrophilic nature and size of such molecules creates a major limitation for their application – poor penetration through biological membranes. Over a decade ago, a new class of peptides, cell-penetrating peptides (CPPs), with membrane translocating ability, was discovered. They have shown remarkable capability to transport various hydrophilic macromolecules inside the cells in a relatively non-toxic fashion. However, there are at least two limiting factors for successful application of CPPs: the lack of cell-type specificity when applied in vivo and restricted bioavailability resulting from endocytic uptake of CPPs and subsequent entrapment in endosomal compartments. This thesis aims at designing delivery vehicles for therapeutic substances. In papers I-III, the CPPs have been rationally modified in order to achieve in vivo selectivity towards cancer cells. The first two papers employ tumor homing peptides as targeting moieties coupled to the N-termini of CPPs and evaluated for cargo delivery. In the third paper, a CPP is Cterminally prolonged with a matrix metalloproteinase 2 (MMP-2) specific cleavage site followed by an inactivating amino acid sequence. In tissues overexpressing MMP-2, i. e. in proximity to cancer, the CPP is activated after proteolytic removal of the inactivating sequence, thus the cargo can be effectively transported inside the nearby cells. In paper IV, an amphipathic CPP, transportan 10 (TP10), has been N-terminally prolonged with a stearic acid tail and applied for the delivery of splice-correcting oligonucleotides. We show that stearyl-TP10 is as effective in oligonucleotide cargo delivery as commercial transfection agent Lipofectamine™ 2000. Moreover, stearylTP10 displays superior characteristics, preserved efficacy in serum and insignificant toxicity to cells, to the cationic lipid. In conclusion, we believe that these results give new insights for improving drug delivery. More specifically, the rational modifications of CPPs greatly potentiate their application in cargo delivery both in vitro and in vivo. Contents Introduction ..................................................................................................1 Cancer .........................................................................................................................1 Tumor angiogenesis..................................................................................................3 Matrix metalloproteinases........................................................................................4 MMPs in the hallmarks of cancer.......................................................................5 The function of MMP-2 in tumor development................................................6 Targeted tumor therapy...........................................................................................7 Tumor homing peptides ......................................................................................7 Target-activated prodrugs..................................................................................9 Antisense therapy......................................................................................................9 Alternative splicing ............................................................................................10 Aberrant splicing in diseases ...........................................................................11 Cell-penetrating peptides.......................................................................................13 Uptake mechanism of CPPs..............................................................................14 CPPs as delivery vectors ...................................................................................15 Aims of the study ......................................................................................20 Methodological considerations................................................................21 Design and choice of CPPs.....................................................................................21 Peptide and ON synthesis ......................................................................................22 Purification and characterization of synthesized peptides and ONs...............23 Coupling of cargos to the peptides (Paper I-IV)................................................23 Selection of cargos..................................................................................................24 Cell cultures..............................................................................................................24 Breast cancer cells (Paper I-III)......................................................................24 Endothelial cells (Paper III) .............................................................................25 HeLa cells (Paper IV).........................................................................................25 Evaluation of CPP uptake and cargo delivery.....................................................26 Fluorescence based methods...........................................................................26 Functional assays to evaluate CPPs as delivery vectors .............................27 Toxicity studies ........................................................................................................28 In vivo studies .........................................................................................................28 MDA-MB-435 tumor bearing mice (Paper I and II) .....................................28 MDA-MB-231 tumor bearing mice (Paper III) ..............................................28 Determination of MMP levels (Paper III).............................................................29 In vitro MMP-2 cleavage assay (Paper III).........................................................29 Serum stability.........................................................................................................29 Results and discussion .............................................................................30 Design and evaluation of tumor homing CPPs (Paper I and II)......................30 Target-activated CPP (Paper III)..........................................................................32 Stearylated CPPs for PS 2’-OMe RNA delivery using a non-covalent coincubation strategy (Paper IV)..............................................................................33 Conclusions .................................................................................................35 Populärvetenskaplig sammanfattning på svenska .............................36 Acknowledgements ...................................................................................38 References ..................................................................................................40 Abbreviations 2’-OMe RNA 2’-MOE RNA AO ATP BMD c Cbl CK2 CPP Cys DMD EMMPRIN Fmoc i. v. LDH MALDI-TOF MBHA MMP mRNA MT MTX OA-Hy OCT ON PMO PNA PTD PS RP-HPLC siRNA snRNP SPPS t-Boc TFA TIMP 2’-O-methyl RNA 2’-O-methoxy-ethyl RNA Antisense oligonucleotide Adenosine triphosphate Becker muscular dystrophy Cyclic Chlorambucil Casein kinase 2 Cell-penetrating peptide Cysteine Duchenne muscular dystrophy Extracellular matrix metalloproteinase inducers Fluorenylmethyloxycarbonyl Intravenous Lactate dehydrogenase Matrix assisted laser desorption ionization time-of-flight 4-Methylbenzhydrylamine Matrix metalloproteinase Messenger RNA Membrane type Methotrexate Hydroxamate derivative of oleic acid Optimum cutting temperature compound Oligonucleotide Phosphorodiamidate morpholino oligomer Peptide nucleic acid Protein transduction domain Phosphorothioate Reversed phase high performance liquid chromatography Short interfering RNA Small nuclear ribonucleoprotein Solid phase peptide synthesis tert-Butyloxycarbonyl Trifluoroacetic acid Tissue inhibitor of metalloproteinase TGF TP10 WST-1 Transforming growth factor Transportan 10 (4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]1,3-benzene disulfonate Introduction Drug delivery remains a challenge in modern medicine. A drug has to enter the target cells in order to exert its biological activity. However, most bioactive molecules, like some anticancer drugs or antisense oligonucleotides, have restricted access into the cell interior due to the highly selective membrane barrier. Therefore, the development of “carriers” for such therapeutics capable of surmounting the greatest obstacle in drug delivery, the plasma membrane, is highly desired. There are several viral and non-viral vehicles developed for drug delivery. A class of peptides, cell-penetrating peptides (CPPs), has recently gained growing attention as non-viral vectors. CPPs have the ability to translocate various cargos, ranging from small drug molecules and oligonucleotides (ONs) to large plasmids and proteins, through the plasma membrane in a relatively non-toxic fashion. However, CPPs suffer from substantial entrapment in endosomal compartments as well as lack of cell-type specificity. This thesis aims at improving CPPs as therapeutic vehicles using cancer as a model system. First, we concentrate on rational modifications of already known CPPs with targeting moieties for application in tumor cellspecific drug delivery. The tumor microenvironment, more specifically angiogenesis and the overexpression of specific matrix metalloproteinases, is taken into account when designing the peptides. Second, we modify wellknown CPPs with a hydrophobic moiety in order to improve the noncovalent cellular delivery of splice-correcting ONs. Cancer Cancer is one of the most frequent causes of death worldwide. It is easy to neglect that cancer is not a single disease, but many diseases characterized by uncontrolled cell proliferation. Tumors are classified into four major groups according to their origin - carcinomas (epithelial), sarcomas (mesenchymal), hematopoietic cancers (blood and immune system) and neuroectodermal tumors (originate from nervous system). However, tumor tissue comprises not only cancer cells, but also stromal cells including fibroblasts, adipocytes, endothelial-, immune and inflammatory cells, which all have a significant impact on tumor progression (Fig. 1) (Mueller and Fusenig 2004). 1 Six essential alterations have been proposed as the hallmarks of cancer, that together dictate the malignant growth of all tumors (Hanahan and Weinberg 2000): 1. 2. 3. 4. 5. 6. self-sufficiency in growth signals, insensitivity to growth-inhibitory signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis, tissue invasion and metastasis. Genomic instability, i. e. mutations in oncogenes and tumor-supressor genes that control normal cell cycle processes result in the abovementioned six alterations. The process when genetically unstable cancer cells escape from the primary tumor and find their way through the blood or lymphatic stream to a distant tissue and adapt to the tissue is called metastasis (Gupta and Massagué 2006). The complex multistep nature of this process is the reason why actually only a relatively small fraction of primary cancer cells are capable of developing metastasis. Cancer metastasis makes it especially complicated to treat this disease since it is difficult to identify cancer cells in distant organs and moreover problematic to address with systemic therapies. Figure 1. Tumor microenvironment. Tumor tissue comprises not only cancer cells, but also stromal cells including fibroblasts, adipocytes, endothelial-, immune and inflammatory cells. Different matrix metalloproteinases (MMPs) are synthesized by both stromal and cancer cells. Adapted from Egeblad and Werb 2002. Breast cancer is by far the most frequent cancer in women worldwide, comprising 23% of all cancers, with an estimated 1.15 million new cases per year 2 (Parkin, et al. 2005). Not the primary tumors, but distant metastasis are the cause of mortality in breast cancer (Weigelt, et al. 2005). Most common metastatic sites of this malignancy are lung, bone, liver and pleural cavity (Weigelt, et al. 2005). There are countless of molecules and a number of processes that results in genomic instability required for tumor development and progression; however in the next sections only an overview of angiogenesis and matrix metalloproteinases (MMPs) as well as their role in cancer progression will be discussed in more detail. Tumor angiogenesis Angiogenesis, the formation of new blood vessels from pre-existing ones, is essential during growth and development, wound healing, female reproductive cycle and pregnancy, but is otherwise quiescent in adult organisms. Furthermore, angiogenesis plays an important role also in many pathological processes such as cancer (Bergers and Benjamin 2003), stroke, Alzheimer’s disease, and in eye morbidity and blindness. The hypothesis that tumor growth is angiogenesis-dependent was first postulated by Judah Folkman in 1971 (Folkman 1971) and afterwards has been confirmed by a large number of research groups (reviewed in Bergers and Benjamin 2003). It is now established that tumors cannot grow beyond a size of 1-2 mm3 without blood supply. Angiogenesis is not merely a prerequisite for tumor growth but also for the development and progression into metastasis. Malignant tumor cells recruit blood as well as lymphatic vessels to spread to distant organs. Tumor blood vessels are morphologically different from their normal counterparts: they are tortuous, irregularly shaped, leaky, dilated and often have blind ends (Dewhirst, et al. 1989). Additionally, blood flows irregularly and more slowly (McDonald and Choyke 2003). This all results in hypoxic (oxygen deficit) tumor tissue, which in turn induces the sprouting of new blood vessels, so called “angiogenic switch” (Folkman and Hanahan 1991, Yancopoulos, et al. 2000). The onset of angiogenesis is determined by the type of tumor as well as its microenvironment. Tumors express various proangiogenic factors that induce the formation of new blood vessels, for instance vascular endothelial growth factor A, B, C (Ferrara 2002), fibroblast growth factor, platelet derived growth factor B, angiopoietin-1 (Yancopoulos, et al. 2000), placental growth factor (Carmeliet, et al. 2001), as well as various MMPs (Genís, et al. 2006, Kim and Kim 1999, van Hinsbergh and Koolwijk 2008). Moreover, certain proteins, i. e. tumor vascular markers, are expressed on the tumor endothelial cells but not on the vasculature of normal tissues. Some of these well-characterized proteins are roundabout-4, endothelial3 specific protein disulphide isomerase, fibronectin, DELTA4 and tumor endothelial markers (reviewed in Neri and Bicknell 2005). Such tumor-specific molecules, already known and yet to be discovered, create an excellent possibility for tumor targeting. Matrix metalloproteinases MMPs are proteolytic enzymes capable of degrading various proteins. They play an important role not only in angiogenesis, bone remodeling, and normal wound healing, but also in pathological processes like arthritis, atherosclerosis and cancer. The primary function of MMPs in normal physiological processes is the maintenance of extracellular matrix. MMPs are zinc-dependent endopeptidases first discovered by Gross and Lapiere in 1962 when they demonstrated that cultivating bullfrog tadpole tissues on collagen gels resulted in degradation of the substrate into collagen peptides (Gross and Lapiere 1962). To date more than 20 different MMPs are recognized and they are commonly divided into 2 main groups, secreted or membrane-bound MMPs. These groups are in turn divided into six subgroups according to their structure and putative substrate specificity: collagenases, gelatinases, stromeolysins, matrilysins, membrane type (MT) MMPs and other types that do not fit into the abovementioned classification (Vihinen and Kähäri 2002). Most of the MMPs are not constitutively expressed in the adult body; instead their expression is rapidly induced when needed by external stimuli like cytokines, growth factors, hormones, cellmatrix and cell-cell interactions (Westermarck and Kähäri 1999). Additionally, the activity of already secreted MMPs is tightly controlled by endogenous inhibitors like tissue inhibitors of MMPs (TIMPs) (reviewed in Baker, et al. 2002), α2-macroglobulin (Sottrup-Jensen 1989) and thrombospondin-2 (Yang, et al. 2001). In cancer, for instance, tumor cells produce factors, extracellular MMP inducers (EMMPRINs) that enhance the production of MMPs from stromal fibroblasts (Biswas, et al. 1995). All MMPs are synthesized as latent proenzymes that can be activated by other MMPs or various other proteins (reviewed in Chakraborti, et al. 2003). Latency of these enzymes is maintained through the so called “cysteine switch” mechanism proposed by Springman, et al. 1990. The pro-domain of MMPs contains a conserved cysteine (Cys) that interacts with an active site zinc atom (bound to three histidines) and maintains the enzyme inactive (Fig. 3). Hence the dissociation of zinc atom from the Cys residue leads to activation of the enzyme, and thus this process is called “cysteine switch”. 4 MMPs in the hallmarks of cancer Initially, MMPs were thought to be exclusively important in tumor invasion and metastasis, however, later studies have shown that MMPs are involved in various steps of cancer development (Hanahan and Weinberg 2000). MMPs have shown to play a crucial role in both cancer-promoting and cancer-inhibiting events (Fig. 2). MMPs are able to promote tumor cell growth by releasing the precursors of growth factors bound to the cell membrane, for example transforming growth factor α (TGF-α) (Peschon, et al. 1998). On the other hand, activation of TGF-β by MMPs can negatively regulate cancer-cell growth (Derynck, et al. 2001). Additionally, MMPs are capable of controlling whether a tumor cell undergoes apoptosis or not, possibly by indirectly influencing the integrin signaling; and moreover they have shown to be able to induce angiogenesis, that is highly essential for tumor growth beyond the microscopic size (Egeblad and Werb 2002). One interesting aspect is the functional role that MMPs have in the immune response to cancer. Indeed, MMPs may be involved in cancer cell escape from the immune surveillance (Coussens and Werb 2002). However, the most well-characterized function of MMPs is their role in tumor invasion and metastasis. Overexpression of certain MMPs promote tumor progression while TIMPs have an inhibitory effect (thoroughly reviewed in Egeblad and Werb 2002). More specifically, MMP-2 is widely reported to play a pivotal role in tumor associated events. Figure 2. The role of MMPs in cancer development. 5 The function of MMP-2 in tumor development The MMPs are a large and diverse group of enzymes, therefore only MMP-2 and related MT1-MMP (MMP-14) that play important roles in this thesis will be depicted in more detail. MMP-2 is a 72 kDa gelatinase A (and type IV collagenase) particularly important in tumor development and progression (Fig. 3). Since MMP2 and MMP-9 degrade type IV collagen, the core component of the basement membrane and the main barrier separating in situ and invasive tumor, it is believed to play a crucial role in tumor metastasis. There have been studies suggesting that MMP-2 overexpression is involved in breast cancer metastasis for example to brain (Mendes, et al. 2007) and lungs (Gupta, et al. 2007). Moreover, it has been shown that active MMP-2 is detected more frequently in malignant than benign breast carcinomas (Hanemaaijer, et al. 2000). Figure 3. Schematic diagram of MMP-2 (a) and MT1-MMP (b). Signal, N-terminal signal sequence; Pro, pro-peptide; Cys, cysteine thiol group interacting with zinc; Catalytic; catalytic domain; Fi, gelatin- or collagen-binding type II repeats of fibronectin; Zn, zinc binding site; H, hinge; Hemopexin; determines the substrate specificity and has interaction site for tissue inhibitors of MMPs; Fu, recognition site for intracellular furin-like serin proteases; TM, single span transmembrane domain; Cy, short cytoplasmic domain. As mentioned above, like all MMPs, MMP-2 is expressed as a zymogen and its activation is a well-studied event where MT1-MMP (Fig. 3) and TIMP-2 play important roles (Kinoshita, et al. 1998, Strongin, et al. 1995, Butler, et al. 1998, Sato, et al. 2005, Zucker, et al. 1998). First, active MT1-MMP binds to the N-terminus of TIMP-2 and attaches soluble TIMP-2 to the cell surface. This MT1-MMP–TIMP-2 complex acts as a receptor for pro-MMP2, in such that pro-MMP-2 binds to the C-terminus of TIMP-2. Consequently, the prodomain of latent MMP-2 is cleaved by an adjacent MT1MMP (TIMP-2 free) creating an activated intermediate which is fully activated through intermolecular autocleavage (Atkinson, et al. 1995). In case TIMP-2 is expressed in excess to MT1-MMP, the first step of MMP-2 acti- 6 vation process is blocked and no MMP-2 activation occurs (Kinoshita, et al. 1996, Sato, et al. 1996, Strongin, et al. 1995). In tumors, cancer cell interaction with fibroblasts, endothelial cells and other tumor stromal cells determine the expression of different MMPs. In breast cancer, both MMP-2 and MT1-MMP have been shown to be expressed by vascular endothelial cells (Chun, et al. 2004, Jia, et al. 2000) and MT1-MMP additionally by myofibroblasts adjacent to breast cancer cells (Bisson, et al. 2003). In conclusion, it is difficult to discriminate which cells in the tumor tissue that are expressing specific type of MMPs, but it is sure that cellular interplay determines the level of particular MMP expression. Targeted tumor therapy Although there are number of treatments available today to fight cancer, the need for improved therapeutics is still desired (Clarke, et al. 2005a, Clarke, et al. 2005b). The development of new compounds for the systemic treatment of cancer is a challenging task mainly due to the heterogeneic nature of tumors. Moreover, a successful therapeutic must first be delivered to the target site and then enter the target cell to exert its biological effect while having little or no effect on non-targeted cells for minimal the side-effects. In order to fulfill those criteria, a drug has to first overcome the physiological barriers like rapid degradation in the blood stream, renal clearance, and the immune system, and then cellular barriers like cellular membranes, entrapment in endosomes, and intracellular degradative enzymes. This simplified description gives an idea of how complicated it is to develop promising cancer therapeutics. Novel strategies are required to improve the targeting of anticancer agents to tumor tissue and to minimize the adverse effects on healthy organs. Next, two approaches, that take advantage of tumor vasculature and the molecules expressed on the tumor blood vessels as well as tumor stroma, are described as possible vehicles for targeted tumor therapies. Tumor homing peptides Tumor homing peptides are a class of peptides that bind to tumor vasculature after intravenous (i. v.) injection. These peptides are discovered by screening of phage display libraries first introduced by Smith in 1985 (Smith 1985). Phage libraries are created by incorporating random DNA sequences into the phage genome; in turn each phage expresses only one type of peptide on its surface. The phage libraries can then be applied for in vivo screening by injecting to the tail vein of mice and allowing them to distribute throughout the body (Ruoslahti 2002). Phages that display a tumor-specific peptide on their surface become concentrated in the tumor tissue, tumor is isolated, 7 phages extracted and amplified and injected for repeated rounds until one type of phage is in majority. Finally, the DNA that encodes the expressed peptide is sequenced and this peptide can be used for in vivo tumor targeting. Table 1. Selection of tumor homing peptides applied in cargo delivery. Peptide Cargo a a NGR , RGD Dox F3 b 2’OMe/PS RNA Fl/− LyP-1a RGDa AdLuc b Cancer Analyses/Effect Reference Breast Inhibited tumor growth Arap, et al. 1998 Breast Inhibited tumor growth and metastasis Breast Tumor imaging/Inhibited tumor growth Breast/Brain Tumor imaging Henke, et al. 2008 Laakkonen, et al. 2004 Niu, et al. 2007 CREKA Nanoparticle Breast Self-amplifying homing GX1a 99 Gastric Tumor imaging Lipo-Dox Lung, oral Inhibited tumor growth Simberg, et al. 2007 Zhi, et al. 2004, Hui, et al. 2008 Lee, et al. 2007 Fl Metastasis Metastasis imaging Yang, et al. 2008 SP5-52b TMTP1 b TcmO4- −, no cargo; acyclic; blinear; 2’OMe, 2’-O-methyl RNA; AdLuc, adenovirus carrying firefly luciferase reporter gene; Dox, doxorubicin; Fl, fluorescein; Lipo-Dox, liposome carrying doxorubicin; PS, phosphorpthioate; 99Tc, technetium 99. A number of tumor homing peptides targeting different tumors have been identified by screening such aforementioned libraries. Many of these peptides have been successfully applied in tumor imaging and treatment (Table 1). One excellent study published recently by Henke et al. utilized the tumor homing peptide F3 (Porkka, et al. 2002) which is believed to bind to cell surface nucleolin (Christian, et al. 2003). They utilized F3 for antisense ON delivery to breast tumor xenografts (Henke, et al. 2008). F3-conjugated to 2’OMe/PS RNA antisense was applied to downregulate Id1, a transcription factor required for tumor invasiveness, metastasis and angiogenesis which led to significant inhibition of tumor growth and metastasis. The majority of tumor homing peptides bind to markers on tumor blood vessels, however there are also peptides homing to tumor lymphatics. Laakkonen et al. discovered a cyclic breast tumor lymphatic’s specific peptide, LyP-1, which is not only homing to primary tumors but also to their metastasis (Laakkonen, et al. 2004). Recently, a mitochondrial/cell-surface protein p32 was shown to be a receptor for LyP-1 (Fogal, et al. 2008). Moreover, there is no need for conjugating LyP-1 to an anticancer drug because the systemic treatment with only the peptide inhibited tumor growth. All the given reports in Table 1 illustrate the vast potential of tumor homing peptides in cancer treatment and imaging. 8 Target-activated prodrugs The second strategy for tumor targeting involves so called prodrugs, designed to be non-toxic until cleaved by enzymes that are overexpressed in tumor environment. There are several proteases shown to be abundantly expressed in proximity to tumors but not in normal tissues. For example, several cancers have reported to have increased levels of MMP-2, -9, -14 (Duffy, et al. 2000, Fingleton 2006, Garzetti, et al. 1996, Yamamura, et al. 2002), plasmin (Dano, et al. 2005), cathepsin B (Jedeszko and Sloane 2004), urokinase-type plasminogen activator (Dano, et al. 2005) and prostatespecific antigen (Partin, et al. 2002). The prodrug idea has been employed to improve the anticancer drugs’ therapeutic index, i. e. the ratio between the toxic dose and the therapeutic dose of a drug. For instance, Albright et al. utilized a MMP-selective prodrug where doxorubicin was conjugated to a heptapeptide, which prevented the prodrug from entering cells. Upon MMP cleavage doxorubicin is “activated” and taken up by nearby cells. Moreover, they showed that this prodrug cured 8 out of 10 mice with HT1080 xenografts at doses below the maximum tolerated dose, while doxorubicin alone cured only 2 out of 20 mice at its maximum tolerated dose (Albright, et al. 2005). Additionally, two other groups developed anticancer drug-peptide derivatives that have the ability to bind to circulating albumin (Mansour, et al. 2003, Warnecke, et al. 2007). Interestingly, the albumin-bound drugs were found to be specifically and efficiently cleaved by target enzymes. Moreover, such prodrugs demonstrated superior antitumor efficacy in vivo compared to free drug. Table 2 provides an overview of several studies that have taken advantage of various tumor-specific enzymes in prodrug design and applied in tumor targeting. Table 2. Selection of target-activated prodrugs applied in tumor treatment. Carrier Enzyme specificity Cancer Cargo Reference Mansour, et al. 2003 Albumin MMP-2 Melanoma Dox Avidin MMP-2 Melanoma Biotinyl-melittin Holle, et al. 2003 Tripeptide Plasmin Breast Dox Heptapeptide MMP-2, -9, -14 Fibrosrcoma Dox Devy, et al. 2004 Albright, et al. 2005 Hexapeptide MMP-2, -9 Fibrosrcoma MTX Chau, et al. 2006 Albumin Cathepsin B Ovarian Warnecke, et al. 2007 MTX Dox, doxorubicin; MTX, methotrexate. Antisense therapy Antisense technology holds a great potential as a therapeutic tool in gene therapy. In 1978 Zamecnik and Stephenson reported, what is now considered 9 as a proof-of-principle for antisense technology, that an ON complementary to the 3’ end of Rous sarcoma virus could block viral replication in chicken fibroblasts (Zamecnik and Stephenson 1978). Antisense technology exploits 15-22 nucleotide long oligonucleotide (AO) analogues designed to be complementary to a selected gene (transcriptional level) or mRNA (posttranscriptional level) and have the ability to bind to target sequence through Watson-Crick hybridization (Crooke 2004). These interactions can lead to inhibition of transcription of chromosomal DNA (Cutrona, et al. 2000, Janowski, et al. 2005); or translational arrest of mRNA, through RNase H cleavage of mRNA-ON duplex or through steric hindrance of the ribosomal assembly on mRNA (Gleave and Monia 2005). There is a growing repertoire of antisense mechanisms that can be exploited for post-transcriptional gene silencing. The most recognized are RNA interference first published by Fire et al. where the double stranded short interfering RNA (siRNA) duplexes suppressed gene activity in Caenorhabditis elegans (Fire, et al. 1998); micro RNAs, naturally occurring post-transcriptional gene regulatory elements (Ambros 2001), and splicecorrecting AOs, selectively manipulating alternative pre-mRNA splicing. The latter will be discussed below in more detail. Alternative splicing In constitutive splicing, introns (non-coding sequences) are removed from newly transcribed pre-mRNAs and all exons (coding sequences) are joined together in the order of which they occur in the pre-mRNAs. In alternative splicing, different mRNAs are produced from the same gene (for example through exon skipping); it is a key post-transcriptional modification that contributes to protein diversity through highly regulated splicing events. Pre-mRNA splicing takes place in the cell nucleolus and is carried out by a splicing machinery called the spliceosome. The spliceosome consists of small nuclear ribonucleoproteins (snRNP) and a large number of nonsnRNPs. The spliceosome recognizes intron/exon boundaries, the so-called 5’ and 3’ splice sites (invariant GU and AG intronic nucleotides, respectively), branch point and polypyrimidin tract and catalyses accurate cleavage and rejoining of exons (Srebrow and Kornblihtt 2006). Additionally, lessconserved consensus motifs (intronic and exonic splicing enhancers and silencers) participate in the splicing process. Mutations in cis- and trans-acting elements of splicing might result in disruption of normal splicing which can lead to various serious diseases. In the following section the role of aberrant splicing in disease, as a direct cause or contribution for disease development like cancer, will be described. 10 Aberrant splicing in diseases As already mentioned above, the disruption of normal alternative splicing patterns can lead to various diseases, such as cystic fibrosis, muscular dystrophies, β-thalassaemia, and several neurological disorders (Garcia-Blanco, et al. 2004, Pajares, et al. 2007). For example, mutations in the dystrophin gene can lead to Duchenne muscular dystrophy (DMD) which is a severe disease characterized by progressive degenerative myopathy or to its milder allelic disorder called Becker muscular dystrophy (BMD) (Koenig, et al. 1989). Most nonsense mutations within this gene lead to a shift in the translational reading frame and thus complete absence of functional dystrophin protein resulting in development of DMD. In BMD, the nonsense mutations occur in exonic splicing enhancer sequences in the dystrophin gene, which leads to partial in-frame skipping of an exon and production of partly functional dystrophin protein. Aberrations in alternative splicing have also been observed in many cancer-related genes (thoroughly reviewed in (Pajares, et al. 2007, Srebrow and Kornblihtt 2006). Cis-acting mutations that affect the splicing of oncogenes and tumor supressors can have contributory roles in cancer development. However, most-cancer associated splicing abnormalities are not caused by mutations in the affected gene suggesting there are mutations in trans-acting splicing elements (Wang and Cooper 2007). Application of ONs for splice correction As already mentioned, several types of AOs have been utilized to downregulate gene expression through RNase-H mediated degradation of mRNA or by steric blockage of gene promoter elements. However, growing knowledge about the importance of aberrant splicing in human diseases has resulted in development of various strategies for abrogating splicing defects. For instance, small molecule inhibitors are attractive in targeting different protein isoforms (Garcia-Blanco, et al. 2004) or trans-splicing approach repairing genetic defects in the mature mRNA (reviewed in Wood, et al. 2007). Additionally, splice-correcting AOs can be employed for altering aberrant splicing patterns on the pre-mRNA level. Since unmodified RNA or DNA have poor stability in biological systems, several ON analogues have been developed to improve their stability and even specificity and affinity for target sequences. Most commonly used RNA analogues include phosphorothioate (PS) (Campbell, et al. 1990), 2’-O-methyl (2’OMe) (Morvan, et al. 1993), 2’-O-methoxy-ethyl (MOE) (Baker, et al. 1997) modified RNAs, as well as phosphorodiamidate morpholino oligomers (PMO) (Summerton 1999) and peptide nucleic acids (PNAs) (Egholm, et al. 1993) (Fig. 4). These AOs are reported to block cryptic splice sites; induce exon skipping or inclusion to alter expression and; induce exon removal to eliminate a non11 sense mutation to restore the reading frame around a genomic deletion. Some examples of such manipulations are given in Table 3. Additionally, it is possible to direct natural alternative splicing towards a desired protein isoform. For example, the bcl-x gene is alternatively spliced to two proteins with antagonistic functions, the pro-apoptotic Bcl-xS and the anti-apoptotic Bcl-xL. The latter has been shown to be overexpressed in various cancers (Kirkin, et al. 2004). By blocking the alternative 5’ splice site in the bcl-x intron 2 with AOs, the splicing can be shifted from the bcl-xL to the bcl-xS isoform and thereby make cells susceptible to apoptotic stimuli (Mercatante, et al. 2001, Taylor, et al. 1999). Figure 4. A selection of widely utilized ON analogues. 1st generation: PS-RNA (phosphorothioate RNA) has an improved stability in human serum. 2nd generation: 2’-OMe RNA (2’-O-methyl RNA) and 2’-MOE (2’-O-methoxy-ethyl) have reduced toxicity and slightly increased affinity for RNA and is not recognized by RNase H. 3rd generation: PMO (phosphorodiamidate morpholino oligomer) and PNA (peptide nucleic acid), uncharged ON analogues with improved target specificity. All examples given in Table 3 illustrate the potency of such AOs to be applied in clinical trials. However, the efficacy of these RNA-based AOs still remains poor due to delivery-related issues. Commonly used transfection systems including cationic liposomes, cationic polymers, viruses, micro in12 jection and electroporation are all facing several drawbacks. For example, cationic liposomes are restricted to in vitro applications due to their sensitivity to serum proteins (Liu, et al. 2003) and are generally toxic. Polyethylene imine (Boussif, et al. 1995), the most commonly used cationic polymer, is highly toxic probably due to its non-degradable nature. Viruses are limited to plasmid delivery and are possibly immunogenic in vivo and moreover carry a risk for viral recombination. All the listed problems with current transfection methods for ONs clearly indicate that new better agents are needed for successful ON delivery. A relatively new class of peptides, cell-penetrating peptides (CPPs), is especially promising for this application. Table 3. Modulation of aberrant splicing in various diseases. Disease β-thalassemiaa β-thalassemiab β-thalassemiac Hutchinson-Gilford progeria syndromea Cystic fibrosisa Ataxia-telangiectasiaa Duchenne muscular dystrophyc Duchenne muscular dystrophyc Frontotemporal dementia and parkinsonisma Target gene AO Blockage of cryptic splice site β-globin 2’OMe RNA β-globin PMO β-globin MOE Lamin PMO CFTR ATM 2’OMe RNA PMO Exon skipping Dystrophin 2’OMe RNA Dystrophin PMO Tau 2’OMe RNA Reference Sierakowska, et al. 1996 Lacerra, et al. 2000 Sazani, et al. 2002 Scaffidi and Misteli 2005 Friedman, et al. 1999 Du, et al. 2007 Lu, et al. 2005 Alter, et al. 2006, Fletcher, et al. 2006 Kalbfuss, et al. 2001 Exon inclusion Spinal muscular atrophya SMN MOE Hua, et al. 2007 Glioblastomaa FGFR-1 PMO Bruno, et al. 2004 a in vitro; bex vivo; cin vivo; ATM, ataxia-telangiectasia mutated; CFTR, cystic fibrosis transmembrane conductance regulator; FGFR, fibroblast growth factor receptor; SMN, survival motor neuron. Cell-penetrating peptides The cellular plasma membrane possesses an effective barrier for most hydrophilic macromolecules used for therapeutic purposes. The need to deliver biologically active agents into cells has encouraged researchers to develop various delivery vehicles. Unfortunately, most delivery systems used today suffer from different limitations that need to be overcome to be applicable in vivo. 14 years ago a new class of peptides, CPPs, also referred to as protein transduction domains (PTDs), with membrane translocation ability was dis13 covered. The first presented CPP, penetratin (also named Antp), was derived from the third helix of the Drosophila melanogaster antennapedia transcription factor homeodomain (amino acids 43-58) (Derossi, et al. 1994). Penetratin, pVEC (Elmquist, et al. 2001) as well as Tat (48-60) (Vivés, et al. 1997) peptides represent CPPs derived from naturally occurring proteins. There are also chimeric CPPs like transportan, which has 12 amino acids derived from the neuropeptide galanin fused together with 14 amino acids derived from the wasp venom mastoparan (Pooga, et al. 1998). Additionally, two widely used chimeric CPPs are MPG (Morris, et al. 1997) and the deletion analogue of transportan, TP10 (Soomets, et al. 2000). A third group of CPPs is the synthetic peptide family, where polyarginines are the best studied members (Wender, et al. 2000). Generally, CPPs are relatively short (5-40 amino acids), cationic and/or amphipathic peptides that are capable of carrying diverse biological cargos across cellular membranes and possess relatively low cellular toxicity. Uptake mechanism of CPPs Although the uptake mechanism of CPPs is still debated, the new findings in this research field during the past few years have contributed to the understanding of how these short peptides make their way into the cell interior. It is important to gain information about their uptake mechanism in order to be able to rationally design new CPPs or to modify the “old” ones for cargo transport into specific cellular compartments. Based on a number of investigations, four main events for CPP uptake have been suggested: first, interactions with cell surface; second, translocation through cell membrane either via different types of endocytosis or direct translocation through cell membrane; third, destination to different intracellular compartments; and fourth, in the case of endocytic uptake, the endosomal release of the CPPs. Independent of the subsequent cellular uptake route, it is believed that internalization of CPPs or CPP-cargo complexes begins with interactions with components on the surface of the plasma membrane. It has been shown that this occurs through binding to cell surface proteoglycans, mostly through heparane sulfate proteoglycans (Foerg, et al. 2007, Gerbal-Chaloin, et al. 2007, Poon and Gariepy 2007, Pujals, et al. 2006, Lundin, et al. 2008, Richard, et al. 2005). This binding is followed by activation of small Rho GTPases which in turn results in remodeling of the actin network and thus contributes to the cell membrane fluidity and following translocation processes. Additionally, hydrophobic interactions between CPP sequences and cell membrane lipids have been proposed to be important in creating the first contact (Pujals, et al. 2006). 14 As mentioned above after CPP-cell surface interactions either direct translocation or endocytosis can occur (Table 4). Energy-dependent fluid phase endocytosis comprises at least macropinocytosis, clathrin-mediated endocytosis, caveolae-mediated endocytosis and clathrin- and caveolaeindependent endocytosis (thoroughly reviewed in Conner and Schmid 2003, Futaki, et al. 2007, Henriques, et al. 2007, Herce and Garcia 2007). There is evidence that CPPs can utilize several different endocytosis pathways simultaneously, thus when blocking one pathway the other one/ones become active (Duchardt, et al. 2007, Padari, et al. 2005, Lundin, et al. 2008). Moreover, the choice of endocytic pathway seems to be dependent on the size and nature of attached cargo. Additionally, energy independent direct translocation through cell membrane has also been supported by several studies (Duchardt, et al. 2007, Palm-Apergi, et al. 2008). Table 4. Defined uptake mechanisms for Tat, transportan, TP10, penetratin and polyarginine. Cargo Mechanism Fluorescein Tat Clathrin-mediated endocytosis Macropinocytosis EGFP Phage Cre-recombinase Avidin-gold PNA PNA Fluorescein Fluorescein/biotin Direct translocation Caveolae-mediated endocytosis Caveolae-mediated endocytosis Macropinocytosis Transportan Mainly macropinocytosis Clathrin-mediated endocytosis TP10 Clathrin-mediated endocytosis Polyarginine Clathrin-mediated endocytosis Macropinocytosis Penetratin Direct translocation Fluorescein Macropinocytosis EGFP, enhanced green fluorescent protein. Reference Richard, et al. 2005, Kawamura, et al. 2006 Nakase, et al. 2004, Nakase, et al. 2007 Tünnemann, et al. 2006 Fittipaldi, et al. 2003 Eguchi, et al. 2001 Wadia, et al. 2004 Padari, et al. 2005 EL Andaloussi, et al. 2006 Lundin, et al. 2008 Kawamura, et al. 2006 Nakase, et al. 2004, Nakase, et al. 2007 Dupont, et al. 2007, Christiaens, et al. 2004 Sugita, et al. 2008 The intracellular fate of CPPs is determined by the internalization pathway. For example, if the uptake occurs through clathrin-mediated endocytosis, the peptides are destined to the degradative route. On the other hand micropinocytosis has been proposed to bypass the lysosomal pathway (Hewlett, et al. 1994). 15 The results from the plethora of experiments are not fully conclusive, as it seems that the translocation mechanism utilized is dependent on the nature of cargo, cell-type, CPP and the concentration of CPP. Recent studies have shown that at higher concentrations, uptake occurs via direct translocations, and at lower concentrations via the endocytosis pathway (Duchardt, et al. 2007, Fretz, et al. 2007). Suggested uptake mechanisms for some most utilized CPPs – Tat, transportan, TP10, penetratin and polyarginine - are given in Table 4. CPPs as delivery vectors CPPs are versatile tools and their ability to enter a wide variety of cell types in a non-discriminating manner forms the favorable basis for their practical use. Cargos that have been transported by CPPs are peptides, proteins, drugs, nanoparticles, lipsomes, ONs, phages, plasmids and imaging agents (thoroughly described in recent reviews: Dietz and Bähr 2004, EL Andaloussi, et al. 2005, Stewart, et al. 2008). However, the application of CPPs in systemic tumor targeting and for splice correction in vitro will be discussed below in more detail. CPPs in tumor targeting In the case of systemic delivery for treatment of cancer, the nondiscriminating mode of cell translocation is an undesirable characteristic. Therefore, strategies to regulate the selective uptake of CPPs are highly required for in vivo applications, such as in cancer treatment. In order to achieve selectivity of CPPs in vivo, there are two main possibilities; coupling of a targeting moiety to the CPP (Table 5) or by modifying the CPP in such that it becomes inactivated and can be activated locally, e. g. in proximity to tumors, by enzymes expressed at the site of action (Table 6). Table 5. Selection of studies describing the application of CPPs in systemic tumor targeting in vivo. CPP Tat Penetratin Cargo P15 peptide p53 peptide TLM Ovalbumin Tat Penetratin β-galactosidase scFv antibody Penetratin, SynB Doxorubicin Cellular response Reduction of tumor mass Induction of apoptosis in glioma cells Improvement of protein vaccination Monitoring of tissue distribution Delivery and prolonged retention of scFv antibody into solid tumors Accumulation of doxorubicin into mouse brain parenchyma Reference Perera, et al. 2008 Senatus, et al. 2006 Bleifuss, et al. 2006 Cai, et al. 2006 Jain, et al. 2005 Rousselle, et al. 2000 16 Perea et al. screened a random phage display library and identified a cyclic 9 amino acids long peptide, P15, which abrogated casein kinase 2 (CK2) phosphorylation (Perera, et al. 2008). CK2, a serine-threonine kinase, is frequently dysregulated in many human tumors. By fusing Tat to P15 and injecting it into TC-1 tumor bearing mice, the tumor mass was substantially decreased. Recently, two interesting studies were carried out by the same group with a p53 derived C-terminal peptide linked to the N-terminus of penetratin (p53p-Ant) (Li, et al. 2005, Senatus, et al. 2006). In the first study, Li et al. showed that p53p-Ant induced selective cell death only in pre-malignant or malignant cells, without affecting normal cells or null p53 cells (Li, et al. 2005). In the second study, Senatus et al. showed that the same chimeric peptide induced apoptosis in human and rat glioma cells both in vitro and in vivo (Senatus, et al. 2006). The possibility to deliver proteins inside cells in order to elucidate the protein function in complex biological processes has been challenged by difficulties in traditional delivery approaches. Plasmid-based genetic recombination techniques have several disadvantages such as limited transfection efficiency and cellular toxicity. Recently, several studies have been published utilizing CPPs for delivery of biologically active full-length proteins into animals. In study by Jain et al. penetratin was shown to improve tumor retention of single-chain Fv antibodies (Jain, et al. 2005). Monoclonal antibody-based radiopharmaceuticals have two major impediments, long circulation times and extremely poor penetration. Thus, they showed that coadministration of penetratin and the scFv antibody resulted in improved retention and distribution of single-chain Fvs. A novel prodrug design of tumor targeting CPPs based on enzyme activation of precursor peptides has recently been developed by Jiang et al. 2004. They showed that the uptake of nona-arginine was effectively blocked when fused to polyanionic domain. They introduced MMP-2 and MMP-9 cleavable linkers between the CPP and the inactivating sequence, hence in the presence of these enzymes the CPP could be activated and cargo could be transported into nearby cells. This selective targeting strategy was reported to be applicable as a tumor imaging approach in mice bearing human fibrosarcoma xenografts (Jiang, et al. 2004). At least two other studies with similar setup have been reported, however, both these studies were carried out only in vitro (Table 6). Table 6. Target-activated CPPs. CPP Inactivating Cargo Target enzyme sequence All D-Arg9 Glu6 Fl, Cy5 MMP-2/-9 All L-Penetratin NLS Fl Cathepsin B All-D-Arg8 Poly-Asp Fl PSA Asp, aspartic acid; Fl, fluorescein; PSA, prostate specific antigen. Reference Jiang, et al. 2004 Pipkorn, et al. 2006 Goun, et al. 2006 17 Chemical modification of CPPs One main consideration when utilizing CPPs for cargo delivery is their cellular fate and, hence, the effects on the bioactivity of the cargo. As mentioned in the previous section, CPPs are taken up primarily via endocytic pathways, and consequently the cargo is commonly retained in endosomal compartments without reaching either the cytoplasm or other cellular compartments such as the nucleus and mitochondria. In vitro this obstacle can be overcome with the use of different lysosomotropic agents, e. g. chloroquine, but this approach is not optimal for in vivo applications. In order to promote endosomal escape and increase the transfection efficiency of CPPs, a number of different strategies have been employed. For example the fusion of the Tat peptide with the influenza virus HA2 domain (Wadia, et al. 2004) and the development of a histidine-containing endosomolytic alpha-helical penetratin analogue (Lundberg, et al. 2007). Stearylation of CPPs has proven to be another successful methodology to increase the transfection efficiency of both plasmids (Futaki, et al. 2001) and siRNA (Nakamura, et al. 2007, Tönges, et al. 2006), through a non-covalent approach resulting in the formation of nanoparticle complexes. In addition to N-terminal stearylation, the presence of a C-terminal cysteamide appears to be crucial for CPP-mediated siRNA delivery using the MPG peptide (Simeoni, et al. 2003). This modification has been believed to increase membrane association (Moulton, et al. 2003, Weller, et al. 2005) and stabilize complex formation (Gros, et al. 2006). CPPs in splice-correcting ON delivery Splice-correcting ONs, negatively charged or uncharged ON-analogues, have shown promising therapeutic potential in redirecting splicing in various diseases. However, the commonly used delivery methods for such ONs suffer, as already mentioned, from number of drawbacks. Therefore, as CPPs are increasingly utilized for transport of a variety of cargos, they have also been applied for the delivery of splice-correcting ON analogues such as PNAs, PMOs and 2’-OMe RNAs (Abes, et al. 2007, EL Andaloussi, et al. 2006, Moulton, et al. 2007). Kole and co-workers have developed a functional splice correction assay for evaluating the cellular delivery efficiency of antisense oligonucleotides (Kang, et al. 1998). This assay is based on the HeLa pLuc 705 cell-line that is stably transfected with a luciferase coding gene interrupted by a mutated β-globin intron 2. This mutation causes aberrant splicing of luciferase pre-mRNA resulting in synthesis of non-functional luciferase. Masking the aberrant splicing site with an antisense ON redirects splicing towards the correct mRNA and consequently restores luciferase activity. This positive read-out assay has been extensively used for evaluating the delivery efficacy of various CPPs (Table 7). Early studies utilized mainly classical CPPs like Tat and penetratin (Astriab-Fisher, et al. 2002, Moulton, et al. 2003), while 18 later several other CPPs like M918 (EL Andaloussi, et al. 2007) and (RXR)4 (McClorey, et al. 2006) have been found to be more potent for these applications. Table 7. Application of CPPs in delivery of splice-correcting oligonucleotides. CPP Tat, Pen Tat Tat, TP, Pen M918 Tat Cargo 2’-OMe RNA PMO PNA PNA PNA Targeted pre-mRNA Luciferase Luciferase Luciferase Luciferase Luciferase Reference Astriab-Fisher, et al. 2002 Moulton, et al. 2003 EL Andaloussi, et al. 2006 EL Andaloussi, et al. 2007 Moulton, et al. 2007, Shiraishi and Nielsen 2006 MAP PNA Luciferase Wolf, et al. 2006 (RXR)4 PMO Dystrophin Moulton, et al. 2007 McClorey, et al. 2006 Pip2 PNA Luciferase, dystrophin Ivanova, et al. 2008 B peptide, P007 PMO Dystrophin Wu, et al. 2008, Yin, et al. 2008a, Yin, et al. 2008b R6-penetratin PNA Luciferase Abes, et al. 2007 MAP, model amphipathic peptide; Pen, penetratin; PMO, phosphorodiamidate morpholino oligomers; PNA, peptide nucleic acid; TP, transportan. Moreover, a number of research groups have successfully applied CPPconjugated antisense ONs to restore dystrophin expression in vivo (Table 7) (McClorey, et al. 2006, Wu, et al. 2008, Yin, et al. 2008a, Yin, et al. 2008b). They utilized murine models with muscular dystrophy and demonstrated, after i. v. injection of CPP-morpholino conjugates, widespread correction of dystrophin expression in skeletal as well as in cardiac muscle. Despite great progress with CPPs in delivery of splice-correcting ONs, it is important to mention that all the studies so far have been carried out with CPPs covalently conjugated to splice-correcting ONs. However, the covalent conjugation of ON cargo is considerably more laborious, thus a simple co-incubation approach would be highly desirable. 19 Aims of the study The main goal of this thesis was to improve CPPs as cargo delivery vehicles for tumor targeting in vivo and ON delivery in vitro. Specific aims for each paper are presented below: Paper I & II Design of tumor homing CPPs by conjugating pVEC to two different homing peptides and evaluation of these chimeric peptides in cargo delivery. Paper III Rational design of MMP-2 activatable CPP, NoPe, and application in tumor targeting. Paper IV Evaluation of stearylated CPPs in delivery of splice-correcting PS 2’-OMe RNA utilizing a simple non-covalent co-incubation strategy. 20 Methodological considerations The materials and methods used in the thesis are described in detail in each paper. Therefore, mainly theoretical and practical aspects of selecting methods will be discussed here and the protocols will only be briefly explained. Design and choice of CPPs Several CPPs have been exploited in this thesis for modification and hence facilitated targeted intracellular delivery of various cargos (Table 8). In paper I, the cyclic homing sequence PEGA was selected for its ability to accumulate in breast tumors (Essler and Ruoslahti 2002). In order to make PEGA a cell-penetrating peptide it was conjugated to the Nterminus of pVEC (Elmquist, et al. 2001). The resulting PEGA-pVEC was studied regarding homing and cell-penetrating capacity. Paper II is a follow-up study to paper I. Here, the breast tumor homing peptide CREKA (Simberg, et al. 2007) was selected to be coupled to pVEC. The CREKA peptide contains only 5 amino acids and is not cyclic, thus, making it easier to synthesize than the PEGA peptide. CREKA was conjugated to the N-terminus of pVEC and the conjugate was assessed for homing ability and cell-penetrating capacity. CREKA conjugated to scrambled pVEC was used as a negative control. In paper III, a previously reported CPP, YTA4 (Lindgren, et al. 2006) was selected to be modified in order to achieve selectivity in vivo. The novel peptide, NoPe, was designed with the help of a CPP prediction algorithm published by Hällbrink et al. (Hällbrink, et al. 2005). The YTA4 peptide was prolonged C-terminally with a sequence consisting of mainly negatively charged amino acids spaced with glycines. In addition, an MMP-2 specific cleavage sequence (Kratz, et al. 2001, Mansour, et al. 2003) was introduced between the CPP and the negatively charged inactivating sequences. The applicability of NoPe in tumor targeting was assessed in a pre-clinical model for breast cancer. In paper IV, several well-known CPPs were evaluated as vectors for splice-correcting PS 2’-OMe RNA delivery. Since CPPs are known to be primarily taken up by endocytic pathway, the bioactive cargo will most likely be entrapped in the endosomes. In order to facilitate the release of the 21 cargo inside the cells we introduced two modifications to the peptides − Nterminal stearylation or C-terminal cysteamidation. Modified as well as unmodified peptides were utilized for transport of splice-correcting ONs inside the nucleolus and the transfection efficiencies were compared with Lipofectamine™ 2000. Table 8. Peptides utilized in the thesis. Peptide Sequence Reference PEGA a,b pVEC a,b Essler and Ruoslahti 2002 Elmquist, et al. 2001 PEGA-pVEC a,b CREKA a,b Simberg, et al. 2007 CREKA-pVEC a,b Mäe, et al. 2008 Scr-pVEC a,b YTA4 a,c NoPe a,c Penetratin d TP10 d AGYLLGKINLKALAALAKKILf-NH2 Arg9 d RRRRRRRRR-NH2 MPG e cCPGPEGAGC-NH2 LLIILRRRIRKQAHAHSK-NH2 cCPGPEGAGCLLIILRRRIRKQAHAHSK-NH2 Myrberg, et al. 2008 CREKA-NH2 CREKALLIILRRRIRKQAHAHSK-NH2 AARIKLRSRQHIKLRHL-NH2 Elmquist and Langel 2003 Lindgren, et al. 2006 IAWVKAFIRKLRKGPLG-NH2 IAWVKAFIRKLRKGPLG-IAGEDGDEGF-NH2 Paper III RQIKIWFQNRRMKWKK-NH2 Derossi, et al. 1994 GALFLGFLGAAGSTMGAWSQPKKKRKV Soomets, et al. 2000 Futaki, et al. 2001 f Morris, et al. 1997 Peptides were N-terminally labeled with fluoresceinyl carboxylic acida, chlorambucilb, methotrexatec or had either N-terminal stearyl-d or acetyl-e modification or C-terminal cysteamide modificationf. c, cyclilized between the cysteines; Scr, scrambled. Peptide and ON synthesis All peptides were synthesized using solid phase peptide synthesis (SPPS), first introduced by Bruce Merrifield in 1963 (Merrifield 1963). The principle of SPPS is that the amino acids are anchored to an insoluble support, resin, which is stable to the chemical reactions carried out during the synthesis. An excess of pre-activated amino acids with either tert-Butyloxycarbonyl (tBoc) or 9-fluorenylmethyloxycarbonyl (Fmoc) protected Nα-amino groups are assembled in a step-wise manner on a resin forming a peptide bond between the amino group and the Nα-protected amino acid. After each coupling, excess of coupling reagents are easily washed away and temporary Nα-protecting group is removed in order to be able to couple the next amino acid. t-Boc is removed with trifluoroacetic acid (TFA) and Fmoc with piperidine. Before coupling a new amino acid, the resin is washed once again. This cycle is repeated until the last amino acid has been coupled. In order to generate C-terminally amidated peptides, t-Boc chemistry with 4-methylbenzhydrylamine (MBHA) resin or Fmoc chemistry with Rink 22 amide resin was applied. The choice of chemistry was determined by the compatibility of the cargo with final cleavage conditions. The final cleavage from the solid support was carried out with hydrofluoric acid (in t-Boc chemistry) or TFA (in Fmoc chemistry). In order to get C-terminally cysteamide modified peptides cysteamine-2 chlorotrityl resin was used (Fmoc chemistry). Synthesis of PS 2’-OMe RNA was performed using disposable columns packed with polystyrene-based solid support, functionalized for synthesis of ON sequences with 2’-OMe RNA monomers at the 3’-end. 5’labelling was carried out using Cy5 amidite. Purification and characterization of synthesized peptides and ONs The synthesized peptides were purified on preparative or semi-preparative reversed phase high performance liquid chromatography (RP-HPLC) on a C18 column using acetonitrile/water gradient containing 0.1% trifluoracetic acid. The identity of the purified products was verified by analytical RPHPLC and by matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass-spectrometry. The mass-spectra were acquired in positive ion reflector mode using α-cyano-4-hydroxycinnamic acid as a matrix. Crude 2’-OMe RNAs were purified by anion exchange chromatography, desalted, and freeze dried. Coupling of cargos to the peptides (Paper I-IV) In general, two main strategies have been employed to couple CPPs to different cargos: covalent coupling, where the cargo and the CPP are conjugated via a covalent bond, or non-covalent complex formation, where the cargo is simply co-incubated with the CPP. The advantage of using the first strategy is that a defined molecule is obtained, which is desirable in therapeutic applications. Also, the amount of peptide needed in this setting can be less as compared to the complex formation strategy, which could be beneficial as some CPPs display toxicity at higher concentrations. However, this strategy is rather laborious and expensive, requiring pre-activation of peptides as well as extensive purification both before and after conjugation. The advantage of using the co-incubation strategy is the simplicity; as only mixing of CPP and cargo is necessary, followed by addition of the mixture to cells and, thus, complicated chemical conjugation is not required. 23 Additionally, these complexes are highly stable to serum degradation, an attractive feature for in vivo applications. Selection of cargos 5(6)-carboxyfluorescein (further referred to as fluorescein), chlorambucil (Cbl) and methotrexate (MTX) were covalently coupled to the N-terminus of the peptides and phosphorothioate 2’-O-methyl RNA (PS 2’-OMe RNA) was non-covalently complexed with CPPs. Peptides used for MTX coupling were synthesized by Fmoc chemistry since MTX is not compatible with hydrogen fluoride treatment. Fluorescein is a commonly used fluorophore with an excitation wavelength of 494 nm and an emission wavelength of 518 nm. Fluorescein is pHsensitive and for optimal results when quantifying fluorescence, the pH should be kept between 6 and 10. The nitrogen mustard analog, Cbl, is a well characterized cytotoxic agent, which acts by alkylating and crosslinking the DNA, resulting in inhibition of further cell proliferation. The effect is mediated through alkylation of guanine N7 in the major groove of DNA and subsequent interstrand crosslinking (Faguet 1994, Mattes, et al. 1986). Moreover, this interstrand crosslink may induce apoptosis (Mattes, et al. 1986, Whiteside, et al. 2003). MTX is a folate antagonist clinically used in anti-cancer therapy (McGuire 2003). Briefly, MTX inhibits the metabolism of folates which are important in synthesis of nucleotides (especially thymine), thus inhibiting DNA replication and concomitantly decreasing cell proliferation (Genestier, et al. 2000). Cell cultures Breast cancer cells (Paper I-III) All breast cancer cell-lines used in this thesis are derived from tumor metastasis pleural effusions. MCF-7 is an invasive human ductal carcinoma and it is the most commonly used breast cancer cell-line in cancer research (Soule, et al. 1973). MDA-MB-231, a human breast adenocarcinoma cell-line, was originally obtained from a patient who had acquired MTX resistance during chemotherapy (Worm, et al. 2001). This cell-line was selected because they lack the reduced folate carrier, i. e. the MTX transport protein. MDA-MB435 is likewise one of the most commonly used breast cancer cell-lines and is, similar to MCF-7, derived from invasive ductal carcinoma (Cailleau, et al. 1978). 24 Endothelial cells (Paper III) The EA.hy 926 cell-line is a hybrid cell-line derived by fusing human umbilical vein endothelial cells with permanent human lung carcinoma cells A549 (Edgell, et al. 1983). They have been shown to express and secrete a number of molecules characteristic to endothelial cells (Edgell, et al. 1983, Emeis and Edgell 1988, Nelimarkka, et al. 1997, Nelimarkka, et al. 1998), among others MMPs and their tissue inhibitors (Nelimarkka, et al. 1998). These cells were chosen in paper III to represent the vascular endothelial cells in a co-culture assay with MDA-MB-231 to mimic tumor environment in vitro. HeLa cells (Paper IV) HeLa cells are an immortalized cell line derived from cervical cancer cells taken from Henrietta Lacks after her death in 1951. Since then, this has been the most widely used cell line within various research areas and, in particular, in the field of cancer. The main beneficial characteristics with these cells are that they grow rapidly and are very robust. In paper IV, a stably transfected HeLa cell line, HeLa pLuc 705 cells (Fig. 5), was used (Kang, et al. 1998). Here, a plasmid carrying the luciferase coding sequence interrupted by an insertion of intron 2 from βglobin pre-mRNA carrying a cryptic splice site is stably transfected into cells. Unless the aberrant splice site is masked by antisense ONs, the premRNA of luciferase will be improperly processed. Thus, by using these cells, various vectors can be evaluated by measuring luciferase activity. Figure 5. HeLa pLuc 705 reporter system. Luciferase pre-mRNA was modified with insertion of β-globin intron 2 carrying a point mutation at nucleotide 705. Blockage of this site with antisense ON redirects splicing towards the fuctional mRNA. 25 Evaluation of CPP uptake and cargo delivery In this thesis, the internalization efficiency and cargo delivery by CPPs have been studied with two different methods; quantification of fluoresceinyllabeled CPPs by spectrofluorometry and confocal microscopy. One general problem with peptides is that they are rapidly degraded both intracellularly and extracellularly (Palm, et al. 2007, Tréhin, et al. 2004). The risk for measuring fluorescent degradation products should therefore always be considered. Each method has advantages as well as disadvantages and to understand the cell-penetrating ability of peptides several different methods to study the translocation over the plasma membrane should be combined. Fluorescence based methods Quantitative uptake The quantified CPP is usually N-terminally labeled with fluorescein. The quantification method is described in detail by Holm, et al. 2006. In short, fluoresceinyl-CPP was incubated with the cells. After incubation, cells were washed and peptides bound outside of the cell membrane were removed by trypsination. The cells were lysed in sodium hydroxide solution, thus the fluorescence was not measured from intact cells, but from cell lysates. Before measuring the fluorescence of internalized peptide or peptide-cargo complexes by spectrofluorometry, the lysed cells were centrifuged in order to eliminate the risk of measuring peptides on the cell membrane. The amount of internalized cargo was normalized against the total protein content in the cell lysate. This normalization helps to correct for small variances in the amount of cells from different experiment. Even though the method is straightforward and gives a quantitative number of the amount of internalized cargo, one should always consider the possible false interpretations of the results. For instance, it is impossible to differ between cytosolic, nuclear, or endosomal localization of the peptide. Confocal microscopy (Paper I-III) Microscopy is a useful method for studying localization of fluorophorelabeled peptide inside the cell or in tissue sections. Early microscopy studies of CPPs were performed on fixed cells, using methanol or paraformaldehyde fixation. However, the fixation step appeared to cause artifacts on the CPP uptake pattern and a reevaluation of the uptake mechanism led the field to use unfixed, live cells. In paper I-IV confocal microscopy was used were only light from a specific focal plane is detected. By scanning a focal plane crossing the nucleus of a cell, there will be reduced risk of detecting fluoresceinyl-labeled peptide bound to the outside of the cell membrane. 26 Functional assays to evaluate CPPs as delivery vectors As a result of the previously mentioned flaws associated with using fluorescence-based methods to assess CPP internalization, introduction of functional assays are highly desirable. In this thesis two of those have been employed: splice correction and cell proliferation. Splice correction assay (paper IV) The splice correction assay developed by Kole and co-workers 1998, was previously described in the thesis (Kang, et al. 1998). In addition to obtaining information of the delivery efficacy, it also provides information concerning the sub-cellular localization of conjugates. In contrary to siRNA assays that targets mRNA for degradation and other methods that exert their effect by down regulation, this assay generates a positive read-out. Therefore, this assay was used to evaluate uptake of CPPs complexed with 2´OMe RNA targeting the aberrant splice site. Briefly, cells were treated with 2´-OMe RNA complexed with CPP for 4 h, after which media was replaced and cells were allowed to grow for additionally 20 h. After cell lysis, luciferase activity was measured and normalized to protein content. Cell proliferation assays In order to evaluate whether CPPs are successful at transporting drug molecules inside cells, two cell viability assays were used. To make sure that the toxic effects were mediated by the cargo and not from the CPP, peptides without cargos were used as controls. In paper I the CellTiter-Glo™ Luminescent Cell Viability Assay from Promega was used. This assay measures the amount of viable cells based on the quantity of ATP that is produced by metabolically active cells. The luciferase from the assay produces oxyluciferin in a reaction together with luciferin, oxygen and ATP. Oxyluciferin releases light and the amount of light produced is proportional to the number of viable cells. In papers II-IV cell proliferation reagent WST-1 from Roche Molecular Biochemicals was utilized. This method has been used for quantification of cell proliferation and cell viability by monitoring the mitochondrial activity in cells. Mitochondrial dehydrogenases in viable cells cleaves the tetrazolium salt WST-1 (4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]1,3-benzene disulfonate) to formazan. The produced formazan can be quantified by measuring absorbance at 420 nm. The amount of formazan formed is proportional to the number of metabolically active, viable cells. 27 Toxicity studies The lactate dehydrogenase (LDH) leakage assay is a simple and commonly applied in vitro assay to evaluate the integrity of the plasma membrane. LDH is a cytosolic enzyme that catalyzes the conversion of lactate to pyruvate with concomitant exchange of NAD+ to NADH. In the CytoTox-ONE™ assay used in this thesis, released LDH is measured via an enzymatic reaction in which nicotinamide adenine dinucleotide (NADH) reacts with resazurin that is converted to the end product resorufin which can be quantified by measuring fluorescence at 560/590 (ex/em) nm. The main advantage with this assay is that it is fast. However, LDH is quite large molecule, with a molecular weight of 132 kDa, thus it should be kept in mind that small pores or early membrane disruption may not be detected with this method. On then other hand, Johansson et al. demonstrated that LDH assay correlated quite well with a substantially smaller molecule, deoxyglucose, leakage (Johansson, et al. 2008). In vivo studies Mice bearing two commonly used tumor types were utilized as models for in vivo studies. MDA-MB-435 tumor bearing mice (Paper I and II) To produce tumors, nude BALB/c mice were orthotopically injected with MDA-MB-435 tumor cells in the mammary fat pad. The tumor bearing mice were used for homing analysis of peptides when the tumor diameter reached about 10 mm. Tissue distribution of fluoresceinyl-labeled peptides was studied by i. v. injecting the peptides into the tumor-bearing mice. The injected peptides were allowed to circulate wherafter the mice were perfused with 4% parafolmaldehyde through the left ventricle of the heart. Tumors and control tissues were dissected and frozen in optimum cutting temperature compound (OCT) embedding medium after which the frozen sections were prepared for immunohistological analysis and observed under a confocal microscope to determine the accumulation of peptides in vivo. MDA-MB-231 tumor bearing mice (Paper III) Athymic, 4-6-weeks old female BALB/c nu/nu mice from Taconic Europe were maintained under specific pathogen free-conditions in a temperature and humidity controlled environment. Mice under anesthesia (Ketamin 90 mg/kg, Xylazin 10 mg/kg) were injected subcutaneously with 2.5 x 106 MDA-MB-231 human tumor cells to induce tumors. Animals were used for 28 3 independent in vivo biodistribution experiments at 8-12 weeks after tumor implantation. All animal studies were conducted according to the guidelines of the Provincial Governement of Southern Finland and the protocol was approved by the Experimental Animal Committee at University of Helsinki, Finland. Determination of MMP levels (Paper III) The Amersham Biotrak Activity Assay System (GE Healthcare, Uppsala, Sweden) was utilized to determine the levels of MMP-2 and MMP-14 in the cell-lines according to the manufacture’s description. Briefly, 150,000 cells/well were seeded in complete medium two days prior the experiment on a 12-well plate. One day prior the measurement of protein levels, cell medium was changed to serum free complete medium in order to exclude MMPs found in serum. In vitro MMP-2 cleavage assay (Paper III) In order to study the MMP-2-specific cleavage of NoPe, the fluoresceinylpeptide was incubated with active human MMP-2 enzyme for 30 min atroom temperature. The cleavage was monitored at 494/518 (ex/em) nm with spectrofluorometry. The increase in fluorescence by MMP-2 cleavage was inhibited by the MMP-2 selective inhibitor hydroxamate derivative of oleic acid (OA-Hy) (Emonard, et al. 1999). Additionally, the cleavage of MTX-NoPe with recombinant MMP-2 was confirmed with analytical RP-HPLC. The identity of the cleavage products was verified by MALDI-TOF massspectrometry. Serum stability MTX-NoPe was incubated in 100% fetal bovine serum and frozen after 5, 10, 15, 30 and 60 min to stop the activity of serum enzymes. Thereafter the degradation products were separated from the serum proteins using Millipore’C18 ZipTips and analyzed by MALDI-TOF mass-spectrometer. 29 Results and discussion The articles included in this thesis describe the possibilities to modify CPPs in order to be applied in tumor targeting in vivo and improving ON delivery in vitro. In paper I and II, a well-known CPP pVEC has been coupled to two different cancer homing peptides to obtain tumor targeting and cellular uptake in vivo. In paper III, tumor cell specificity has been achieved through inactivation of a CPP, YTA4, with mainly negatively charged amino acids. An MMP-2 specific amino acid sequence was placed between the CPP and the inactivation sequence, so that the peptide could be activated in the presence of MMP-2, i. e. in close proximity to tumors. In paper IV, several CPPs have been N-terminally stearylated in order to improve the delivery of splice-correcting 2’-OMe RNA and compared with commercially available transfection agent Lipofectamine™ 2000. Design and evaluation of tumor homing CPPs (Paper I and II) Chemotherapy used for treatment of cancer has severe side effects, which are mainly caused by the toxicity of the drugs to normal healthy cells and additionally, in many cases tumor cells have developed resistance to the anticancer drugs. Clearly tumor cell-specific delivery vehicles are urgently required to overcome these impediments. As CPPs have gained extensive attention as promising vehicles for therapeutics we designed two tumor homing CPPs by conjugating homing peptide to the pVEC peptide. In paper I, we conjugated a cyclic homing peptide PEGA to the Nterminus of pVEC. PEGA has been isolated by screening in vivo phage display library and it has been shown to target normal breast vasculature as well as premalignant breast tissue and primary breast tumors in vivo (Essler and Ruoslahti 2002). However, the PEGA-peptide is not able to translocate the plasma membrane. Therefore, in order to overcome this barrier PEGA was coupled to pVEC (Elmquist, et al. 2001). The cell-penetrating ability of fluoresceinyl-PEGA-pVEC was first demonstrated in vitro and compared with fluoresceinyl-PEGA and -pVEC. Both, confocal microscopy and quantitative uptake experiments showed that fluoresceinyl-PEGA-pVEC was able to translocate into MCF-7, MDA-MB231 and MDA-MB-435 (only with microscopy) cell lines. The uptake pat30 tern was similar in both MCF-7 and MDA-MB-231 cells. PEGA alone did not display any cell-penetrating properties, whereas when coupled to pVEC, the uptake was greatly improved, although not reaching the same levels as for pVEC itself. There are two possible explanations for decreased uptake of PEGA-pVEC as compared to pVEC. As mentioned in the introduction, CPPs are rich in positively charged amino acids, thus when coupling PEGA peptide, that has one glutamic acid in the sequence, the net positive charge is reduced, most likely resulting in reduced uptake. Another explanation is that the hydrophobic amino acids in the N-terminus of pVEC have been shown previously to be important for its uptake (Elmquist, et al. 2006). Coupling the homing peptide to the N-terminus of CPP can restrict the full interaction of the peptide with plasma membrane and cause reduced uptake. Second, we wanted to demonstrate the drug delivery ability of PEGApVEC in vitro. The DNA alkylating agent Cbl was chosen to exert its anticancer properties. Cbl is believed to enter cells by passive diffusion (Adair and McElnay 1986), which is an extremely inefficient process. In order to improve the uptake of Cbl, we coupled it to the N-terminus of PEGA-pVEC via an amide bond leaving the alkylating moiety of Cbl available for DNA cross-linking. After 48 h exposure of Cbl or Cbl-peptide conjugates the cytotoxicity of the drug was improved with the factor of 4 when conjugated to PEGA-pVEC. Third, the tumor homing properties of the fluorescein-labeled chimeric peptide were evaluated. Fluorecseinyl-peptides were injected into the tail vein of mice bearing MDA-MB-435 tumors, allowed to circulate for 2 h whereafter the mice were sacrificed, tumor and control organs were isolated, and the accumulation of fluorescence was assessed with fluorescence microscopy. Fluoresceinyl-PEGA, -pVEC and -PEGA-pVEC were all found in tumor tissue where they partially co-localized with the blood vessel marker MECA-32. The co-localization was particularly prominent with fluoresceinyl-PEGA-pVEC. In some areas, fluoresceinyl-PEGA-pVEC appeared to be also in tumor cells, although it is not fully accurate to draw such conclusion from that experiment, since the tissue sections are fixed in paraformaldehyde and it is know that even mild fixation can lead to artifactual redistribution of CPPs (Richard, et al. 2003). Apparently, the accumulation of fluoresceinyl-PEGA-pVEC in control organs was substantially lower than that of control peptides. In paper II, another linear homing peptide CREKA was chosen to be coupled to pVEC. CREKA, a 5 amino acid long peptide has been previously shown to accumulate in various types of tumors in vivo (Simberg, et al. 2007). CREKA selectively homes to tumor blood vessels and stroma where it recognizes clotted plasma proteins. We showed that CREKA alone is not taken up by cells, but when coupled to pVEC it becomes a CPP. The advantages of CREKA-pVEC over PEGA-pVEC are the shorter length, easier to synthesize and does not include an extra cyclization step. Moreover, Cbl alone did not have any effect on MDA-MB-231 cell proliferation at concen31 trations studied, while Cbl-pVEC and Cbl-CREKA-pVEC reduced the percentage of proliferating cells from 100% to approximately 40% at 10 µM and 20 µM concentration, respectively. In conclusion, the resultant homing cell-penetrating peptides demonstrated the conserved desirable properties of the parent peptides, making them promising drug delivery vectors. Target-activated CPP (Paper III) Bearing in mind the abovementioned drawbacks associated with CPPs as well as with small molecule chemotherapeutics, we designed an inactivated CPP, activatable in the vicinity of tumors overexpressing MMPs. This peptide was named NoPe as in no non-specific cellular penetration. We chose the previously published CPP, YTA4 (Lindgren, et al. 2006), and prolonged it C-terminally with mainly negatively charged amino acids spaced with glycines and exploited its drug (MTX) delivery properties. Figure 6. Schematic diagram of NoPe. In order to mimic tumor environment in vitro, we set up a co-culture experiment comprising of breast cancer and endothelial cells overexpressing MMP-2 and MMP-14, respectively. We first tested the uptake of fluoresceinyl-NoPe and -YTA4 in both these cell-lines separately and could observe diminished uptake of fluoresceinyl-NoPe as compared to YTA4. Thereafter we tested whether the NoPe peptide could be taken up by cells in the co-culture assay. Indeed, fluoresceinyl-NoPe was detected inside the cells and moreover this uptake could be significantly decreased when MMP2 specific inhibitor was added prior to treatment. Additionally, the correct cleavage of NoPe was demonstrated using recombinant active MMP-2. The cleavage was monitored spectrofluorometrically and with analytical HPLC and the expected cleavage products were determined with mass-spectrometry. Moreover, the MTX-NoPe was found intact after 1 h incubation in 100% fetal bovine serum, which could be compared to the suggested half-life of penetratin, 5 min (Palm, et al. 2007). 32 Next, to validate the applicability of our target activated CPP in vivo, we injected fluoresceinyl-NoPe i. v. to MDA-MB-231 tumor-bearing mice. After 1 h circulation the peptide was mainly found in tumor tissue and substantially lower amounts were also detected in the spleen and liver. In contrast to NoPe, fluoresceinyl-YTA4, displayed negligible levels in tumor tissue while high levels were found in control organs. MDA-MB-231 cells are known to be resistant to MTX treatment (Ref), thus our goal was not only to target MTX specifically to tumor tissue, but upon activation also transport the drug inside cancer cells. We first tested MTX-NoPe in vitro in a co-culture assay, but we could not detect any significant effect of the conjugate on cell proliferation. This indicates that in cell culture the level of MMPs is too low to activate enough of the MTXpeptide to have an effect on cell proliferation. Therefore, further in vivo experiments are required for evaluating the potency of this prodrug. Stearylated CPPs for PS 2’-OMe RNA delivery using a non-covalent co-incubation strategy (Paper IV) Utilizing CPPs to facilitate cellular uptake and to improve the general bioavailability of various types of ONs holds great promise for future therapeutic purposes. CPPs have mainly been covalently attached to ONs; however this adds additional synthesis steps that can be, on occasion, somewhat laborious. Therefore, many groups have recently employed a simple coincubation approach, where peptides and the negatively charged ONs are mixed at certain molar ratios in order to create nanoparticle complexes that are simply added to the cells (Meade and Dowdy 2008). Previously, two chemical modifications, N-terminal stearylation and C-terminal cysteamidation, have been shown to be critical to improve the non-covalent cellular delivery of ONs. Therefore in paper IV, we modified three peptides- TP10, penetratin and nona-arginine N-terminally with stearyl moiety and compared their transfection efficiency with the commercially available transfection agent Lipofectamine™ 2000 and previously published MPG peptide. First, the transfection efficiency of Cy5-labelled PS 2’-OMe RNA with unmodified CPPs TP10, penetratin, and nonaarginine were evaluated. In spite of considerable quantitative uptake of ONs no significant splice correction was detected. This contradiction can be explained with the fact that the complexes were probably entrapped inside endosomes. This was further confirmed in the case of TP10 and Pen; when the lysosomotrophic agent, chloroquine was added to the cells simultaneously with complexes, the splice correction was significantly increased. 33 Due to the low transfection efficiency observed for unmodified peptides, we stearylated all three peptides N-terminally and evaluated their transfection efficiency. No significant difference between the peptides in quantitative uptake could be detected, however stearyl-TP10 proved to be exceptionally effective for the delivery of splice-correcting ONs. Furthermore, it is worth mentioning that correctly spliced luciferase intensity reached levels comparable with results achieved with Lipofectamine™ 2000. However, no significant increase in splice correction for stearyl-Pen and -Arg9 in complex with 2’-OMe ON was detected, thus, apparently the stearyl moiety does not potentiate all CPPs. Since the cationic lipids are associated with quite severe cellular toxicity we evaluated the effect of stearylated TP10 on cell proliferation. Stearyl-TP10 displayed no long-term toxicity even at the highest CPP:ON ratios, on the other hand Lipofectamine™ 2000 induced significant impairment of metabolic activity at the concentration suggested by the manufacturer. In conclusion, we have shown that stearylation of TP10 results in a non-toxic and highly efficient ON delivery vector reaching transfection efficiencies compareable with commercial transfection agent Lipofectamine™ 2000. Moreover, the splice correction mediated by stearyl-TP10 was not affected by the presence of serum. Taken together, these results are particularly encouraging for future in vivo applications of TP10. 34 Conclusions The major findings of this thesis are listed and described below: Paper I and II Conjugation of tumor homing peptides to CPPs results in chimeric peptides with both homing and cell-penetrating properties. Moreover, by coupling Cbl to the N-terminus of these peptides and the drug, efficacy is improved several-fold in vitro. These results are encouraging for future applications in targeted drug delivery. Paper III Rationally designed MMP-2 activated CPP, fluoresceinyl-NoPe, prove to be applicable in tumor imaging, since it selectively accumulates in breast tumor xenografts in vivo. Paper IV Introduction of an N-terminal stearyl-moiety to an amphipathic CPP, TP10, significantly improves the non-covalent cellular delivery of splice-correcting oligonucleotides. Futhermore, the efficacy is preserved in serum and the complex do not display any significant cytotoxicity − characteristics being of great importance for future in vivo applications. This thesis has proved that the rational modification of CPPs greatly potentiate their application in cargo delivery both in vitro and in vivo. 35 Populärvetenskaplig sammanfattning på svenska Cancer är ett tillstånd där celler delar sig okontrollerat och förstör omkringliggande vävnad, något som idag är en av västvärldens vanligaste dödsorsaker. Behandlingen av cancer har utvecklats över tid och idag existerar en rad olika behandlingsmetoder. Strålbehandling, kirurgi, immunterapi och cytostatikabehandling är exempel på metoder som används. Vilken behandlingsmetod som är effektiv är avhängig på typen av cancer och tumörens stadium. Cytostatikabehandling innefattar användandet av läkemedel vars syfte är att döda cancercellerna eller stoppa tumörers tillväxt. Huvudproblemet med många av dagens cytostatikabehandlingar är att de är toxiska. Detta innebär att även normala celler påverkas av behandlingen, en effekt som resulterar i biverkningar såsom till exempel håravfall och magbesvär. En särskild problematik uppstår då mängden läkemedel måste begränsas för att inte orsaka alltför svåra bieffekter trots att tumören egentligen kräver en högre dos. Forskningen för att göra cytostatikabehandlingen mer effektiv är utbredd och fokus ligger på att utveckla metoder som gör att läkemedlet endast angriper cancercellerna. En metod att transportera olika ämnen och läkemedel in i en cell är med hjälp av cell-penetrerande peptider. En peptid, som består av aminosyror, är en beståndsdel i ett protein. Vanligtvis upptas cellpenetrerande peptider av alla typer av celler, men genom att modifiera dessa peptider är det möjligt göra upptagningen specifik för cancerceller. Genom att använda dessa modifierade cell-penetrerande peptider och koppla cytotoxiska läkemedel till dem, skapas cytostatika som bara angriper cancercellerna och inte normala celler. I förlängningen medför detta en mer effektiv cancerterapi, samtidigt som biverkningarna minskar. I den första och andra artikeln har en cell-penetrerande peptid kopplats till målsökande peptider och en cancerdrog. Den målsökande peptiden urskiljer cancercellerna och med hjälp av den cell-penetrerande peptiden ökar cancerdrogens effektivitet genom en bättre upptagning i cellen. Den tredje artikeln behandlar en cell-pentrerande peptid med en inaktiverande aminosyrasekvens, vilken aktiveras vid förekomsten av ett specifikt enzym, vilket frigör den cell-penetrerade peptiden. Även här är 36 peptiden kopplad till en cancerdrog. Enzymet förekommer rikligt i närheten av tumörer. I den fjärde och sista artikeln modifieras en cell-penetrerande peptid med en fettmolekyl som bidrar till att upptagningen av splicing korrigerande oligonukleotider förbättras. Dessa oligonukleotider kan korrigera en muterad gen så att ett funktionellt protein bildas. Ett flertal genetiska sjukdomar och olika former av cancer är i många fall ett resultat av mutationer som gör att proteiner överuttrycks eller att inte korrekta proteiner bildas. Genom att använda ovannämnda oligonukleotider med cell-penetrerande peptider som transportörer skulle flera av dessa sjukdomar möjligen kunna botas. Sammantaget påvisar modifieringarna av de cell-penetrerande peptider som beskrivs i avhandlingen, en förbättrad upptagning av oligonukleotider och droger i cancerceller. 37 Acknowledgements Finally, this long journey is coming to an end. There are so many mixed feelings when I am looking back at the past four years. There have been tough times, but there have also been good times and I must say, these last years have probably been the best time of my life. First and foremost, I appreciate all the amazing people I have met during my time here. First I would like to thank my supervisor Ülo, for giving me the opportunity to do my PhD in your group. Thank you for always having time for me when I wanted to discuss my projects or when I had something on my mind, even when I was so called “unstable”. Now I actually appreciate the way you have pushed me even though I never imagined saying this out loud... I would also like to thank other teachers from the department, Anders, for your interesting lectures in neurochemistry; Mattias, for your constructive comments after each seminar; Anna, Kerstin, and Tiit for everything you have done at the department. And of course Birgitta, MarieLouise, Siv and Ulla for all your administrative help which made my life a lot easier. Then I would like to thank all my collaborators and colleagues, especially the ones from my group: Helena, for being such a good friend and for all the support in the lab, and most of all I have really missed our daily “confessions” since you defended your thesis; Maria, for being always so positive in life and in research, and at all times listening to my ideas as well as my complaints; Katri, for your help with the peptide synthesis and for sharing your expert knowledge in chemistry with me; Per, it was a pleasure to work with you on the fourth paper, you are an impressive person and I am really glad I met you; and last but definitely not least Samir, for being such an inspiring person, I have learned so much from you. But most of all, thank you for being the best killkompis, we have had so much fun! I would also like to thank all the other people in Ülo’s group: Emelia, for being a great friend and for teaching me to understand chemistry and irony (now I can laugh at all your jokes); Ulla, I am so happy that you moved into our office, I hope I can support you as much when you are writing your thesis; Tina, your energy is making me jealous and I have really missed your company in the lab; Henrik, for always helping me when I needed it and for being so calm and reasonable, what would we have done without you in Australia?! Peter G, for synthesizing valuable oligonucleotides. Indrek and Imre, for 38 being so positive and fun guys. All the “old” members in Ülo’s group, Mats, I could not have managed without your computer knowledge! Linda, Pontus, Peter J, Yang, Kalle, Külliki, Riina and Ursel you all have a special place in my heart. I would also like to thank all the other PhD students at the department for creating a great atmosphere. Especially, Johanna, for being who you are! We have come a long way together and our friendship means a lot to me.Thank you for our great conversations and parties, and for influencing me to become more tough and not to be afraid to say what I think; Karolina, for being such a wonderful person and for helping me with confusing chemistry questions; Jessica, for being so thoughtful, always trying to make people glad; Caroline, good luck in States, you have really worked hard for it! Thank you again Helena, Maria, Per and Samir for proof-reading my thesis; Farid and Imre for helping me with the popular scientific overviews. All my foreign friends: Anne B, Agatha, Ganesh and Nomchit. You have thought me so much about other cultures and I have enjoyed our delicious special-food dinners and talks. I would also like to thank all my estonian friends outside the lab. Anneli, for being such a good friend, who is there for me whenever I need it; Tõnu, for always making me laugh; Jaana, for wonderful parties and I hope you will continue visiting me in Sweden; Anne M, I am so glad that we have been keeping in touch; Kairi, I have really missed you here in Stockholm; Erki, for your never ending interest in my “blossoming” proteins; Margret, Ave, Peeter and Raul, for our great dinners and get-togethers, Leili, Ago, Martin and Madis for being my extra family; and of course Evelin, no one will ever understand the unique bond between us, thank you for everything!!! Among other things I really enjoy your ability to make everything sound so glamorous and interesting! Thank you Mimmi, Gerd and Dan for always making me feel like home in Visserfjärda. My beloved family – without your support I would never have been where I am now in the first place. Heli and Andres and your wonderful sons Franck and Patrick, thank you for making me feel so welcomed and loved every single time I come home. My little brother and sisters, thanks for always making me smile. Piret, thank you for your never-ending kindness and for making my father’s life so wonderful! My dear father, you have been the biggest and most important support through all my study years!!! Thank you for inspiring me and providing me everything I have ever needed. Farid, no words are enough to describe how much you mean to me!!! Thank you for your never-ending support and for making me believe in myself. I cannot imagine my life without you anymore! I love you so much! 39 References Abes, S., Turner, J. J., Ivanova, G. D., Owen, D., Williams, D., Arzumanov, A., Clair, P., Gait, M. J. and Lebleu, B. Efficient splicing correction by PNA conjugation to an R6-Penetratin delivery peptide. Nucleic Acids Res. 2007; 35: 4495-4502. Adair, C. G. and McElnay, J. C. Studies on the mechanism of gastrointestinal absorption of melphalan and chlorambucil. Cancer Chemother Pharmacol. 1986; 17: 95-98. Ambros, V. microRNAs: tiny regulators with great potential. Cell. 2001; 107: 823-826. Baker, A. H., Edwards, D. R. and Murphy, G. Metalloproteinase inhibitors: biological actions and therapeutic opportunities. J Cell Sci. 2002; 115: 37193727. Baker, B. F., Lot, S. S., Condon, T. P., Cheng-Flournoy, S., Lesnik, E. A., Sasmor, H. M. and Bennett, C. F. 2'-O-(2-Methoxy)ethyl-modified anti-intercellular adhesion molecule 1 (ICAM-1) oligonucleotides selectively increase the ICAM-1 mRNA level and inhibit formation of the ICAM-1 translation initiation complex in human umbilical vein endothelial cells. J Biol Chem. 1997; 272: 11994-12000. Bergers, G. and Benjamin, L. E. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003; 3: 401-410. Bleifuss, E., Kammertoens, T., Hutloff, A., Quarcoo, D., Dorner, M., Straub, P., Uckert, W. and Hildt, E. The translocation motif of hepatitis B virus improves protein vaccination. Cell Mol Life Sci. 2006; 63: 627-635. Boussif, O., Lezoualc'h, F., Zanta, M. A., Mergny, M. D., Scherman, D., Demeneix, B. and Behr, J. P. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. Proc Natl Acad Sci U S A. 1995; 92: 7297-7301. Bruno, I. G., Jin, W. and Cote, G. J. Correction of aberrant FGFR1 alternative RNA splicing through targeting of intronic regulatory elements. Hum Mol Genet. 2004; 13: 2409-2420. Cailleau, R., Olive, M. and Cruciger, Q. V. Long-term human breast carcinoma cell lines of metastatic origin: preliminary characterization. In Vitro. 1978; 14: 911-915. Campbell, J. M., Bacon, T. A. and Wickstrom, E. Oligodeoxynucleoside phosphorothioate stability in subcellular extracts, culture media, sera and cerebrospinal fluid. J Biochem Biophys Methods. 1990; 20: 259-267. Chakraborti, S., Mandal, M., Das, S., Mandal, A. and Chakraborti, T. Regulation of matrix metalloproteinases: an overview. Mol Cell Biochem. 2003; 253: 269-285. 40 Christian, S., Pilch, J., Åkerman, M. E., Porkka, K., Laakkonen, P. and Ruoslahti, E. Nucleolin expressed at the cell surface is a marker of endothelial cells in angiogenic blood vessels. J Cell Biol. 2003; 163: 871-878. Clarke, M., Collins, R., Darby, S., Davies, C., Elphinstone, P., Evans, E., Godwin, J., Gray, R., Hicks, C., James, S., MacKinnon, E., McGale, P., McHugh, T., Peto, R., Taylor, C. and Wang, Y. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005a; 366: 2087-2106. Clarke, M., Collins, R., Darby, S., Davies, C., Evans, V., Godwin, J., Gray, R., McGale, P., Peto, R. and Wang, Y. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005b; 365: 1687-1717. Coussens, L. M. and Werb, Z. Inflammation and cancer. Nature. 2002; 420: 860-867. Crooke, S. T. Progress in antisense technology. Annu Rev Med. 2004; 55: 61-95. Dano, K., Behrendt, N., Hoyer-Hansen, G., Johnsen, M., Lund, L. R., Ploug, M. and Romer, J. Plasminogen activation and cancer. Thromb Haemost. 2005; 93: 676-681. Derossi, D., Joliot, A. H., Chassaing, G. and Prochiantz, A. The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem. 1994; 269: 10444-10450. Derynck, R., Akhurst, R. J. and Balmain, A. TGF-beta signaling in tumor suppression and cancer progression. Nat Genet. 2001; 29: 117-129. Dewhirst, M. W., Tso, C. Y., Oliver, R., Gustafson, C. S., Secomb, T. W. and Gross, J. F. Morphologic and hemodynamic comparison of tumor and healing normal tissue microvasculature. Int J Radiat Oncol Biol Phys. 1989; 17: 91-99. Dietz, G. P. and Bähr, M. Delivery of bioactive molecules into the cell: the Trojan horse approach. Mol Cell Neurosci. 2004; 27: 85-131. Du, L., Pollard, J. M. and Gatti, R. A. Correction of prototypic ATM splicing mutations and aberrant ATM function with antisense morpholino oligonucleotides. Proc Natl Acad Sci U S A. 2007; 104: 6007-6012. Duffy, M. J., Maguire, T. M., Hill, A., McDermott, E. and O'Higgins, N. Metalloproteinases: role in breast carcinogenesis, invasion and metastasis. Breast Cancer Res. 2000; 2: 252-257. Dupont, E., Prochiantz, A. and Joliot, A. Identification of a signal peptide for unconventional secretion. J Biol Chem. 2007; 282: 8994-9000. Edgell, C. J., McDonald, C. C. and Graham, J. B. Permanent cell line expressing human factor VIII-related antigen established by hybridization. Proc Natl Acad Sci U S A. 1983; 80: 3734-3737. Egeblad, M. and Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002; 2: 161-174. Egholm, M., Buchardt, O., Christensen, L., Behrens, C., Freier, S. M., Driver, D. A., Berg, R. H., Kim, S. K., Norden, B. and Nielsen, P. E. PNA hybridizes to complementary oligonucleotides obeying the Watson-Crick hydrogenbonding rules. Nature. 1993; 365: 566-568. 41 Eguchi, A., Akuta, T., Okuyama, H., Senda, T., Yokoi, H., Inokuchi, H., Fujita, S., Hayakawa, T., Takeda, K., Hasegawa, M. and Nakanishi, M. Protein transduction domain of HIV-1 Tat protein promotes efficient delivery of DNA into mammalian cells. J Biol Chem. 2001; 276: 26204-26210. EL Andaloussi, S., Holm, T. and Langel, Ü. Cell-penetrating peptides: mechanisms and applications. Curr Pharm Des. 2005; 11: 3597-3611. EL Andaloussi, S., Johansson, H. J., Holm, T. and Langel, Ü. A novel cell-penetrating peptide, M918, for efficient delivery of proteins and peptide nucleic acids. Mol Ther. 2007; 15: 1820-1826. Elmquist, A., Hansen, M. and Langel, Ü. Structure-activity relationship study of the cell-penetrating peptide pVEC. Biochim Biophys Acta. 2006; 1758: 721729. Elmquist, A. and Langel, Ü. In vitro uptake and stability study of pVEC and its all-D analog. Biol Chem. 2003; 384: 387-393. Elmquist, A., Lindgren, M., Bartfai, T. and Langel, U. VE-cadherinderived cell-penetrating peptide, pVEC, with carrier functions. Exp Cell Res. 2001; 269: 237-244. Emonard, H., Marcq, V., Mirand, C. and Hornebeck, W. Inhibition of gelatinase A by oleic acid. Ann N Y Acad Sci. 1999; 878: 647-649. Essler, M. and Ruoslahti, E. Molecular specialization of breast vasculature: a breast-homing phage-displayed peptide binds to aminopeptidase P in breast vasculature. Proc Natl Acad Sci U S A. 2002; 99: 2252-2257. Faguet, G. B. Chronic lymphocytic leukemia: an updated review. J Clin Oncol. 1994; 12: 1974-1990. Ferrara, N. VEGF and the quest for tumour angiogenesis factors. Nat Rev Cancer. 2002; 2: 795-803. Fingleton, B. Matrix metalloproteinases: roles in cancer and metastasis. Front Biosci. 2006; 11: 479-491. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A., Driver, S. E. and Mello, C. C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998; 391: 806-811. Fogal, V., Zhang, L., Krajewski, S. and Ruoslahti, E. Mitochondrial/cellsurface protein p32/gC1qR as a molecular target in tumor cells and tumor stroma. Cancer Res. 2008; 68: 7210-7218. Folkman, J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971; 285: 1182-1186. Friedman, K. J., Kole, J., Cohn, J. A., Knowles, M. R., Silverman, L. M. and Kole, R. Correction of aberrant splicing of the cystic fibrosis transmembrane conductance regulator (CFTR) gene by antisense oligonucleotides. J Biol Chem. 1999; 274: 36193-36199. Futaki, S., Suzuki, T., Ohashi, W., Yagami, T., Tanaka, S., Ueda, K. and Sugiura, Y. Arginine-rich peptides. An abundant source of membrane-permeable peptides having potential as carriers for intracellular protein delivery. J Biol Chem. 2001; 276: 5836-5840. Garcia-Blanco, M. A., Baraniak, A. P. and Lasda, E. L. Alternative splicing in disease and therapy. Nat Biotechnol. 2004; 22: 535-546. 42 Garzetti, G. G., Ciavattini, A., Lucarini, G., Goteri, G., Romanini, C. and Biagini, G. Increased serum 72 KDa metalloproteinase in serous ovarian tumors: comparison with CA 125. Anticancer Res. 1996; 16: 2123-2127. Genestier, L., Paillot, R., Quemeneur, L., Izeradjene, K. and Revillard, J. P. Mechanisms of action of methotrexate. Immunopharmacology. 2000; 47: 247257. Gleave, M. E. and Monia, B. P. Antisense therapy for cancer. Nat Rev Cancer. 2005; 5: 468-479. Gross, J. and Lapiere, C. M. Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci U S A. 1962; 48: 1014-1022. Gupta, G. P. and Massagué, J. Cancer metastasis: building a framework. Cell. 2006; 127: 679-695. Hällbrink, M., Kilk, K., Elmquist, A., Lundberg, P., Lindgren, M., Jiang, Y., Pooga, M., Soomets, U. and Langel, Ü. Prediction of cell-penetrating peptides. Int J Pep Res Ther. 2005; 11: 249-259. Hanahan, D. and Weinberg, R. A. The hallmarks of cancer. Cell. 2000; 100: 57-70. Hanemaaijer, R., Verheijen, J. H., Maguire, T. M., Visser, H., Toet, K., McDermott, E., O'Higgins, N. and Duffy, M. J. Increased gelatinase-A and gelatinase-B activities in malignant vs. benign breast tumors. Int J Cancer. 2000; 86: 204207. Hewlett, L. J., Prescott, A. R. and Watts, C. The coated pit and macropinocytic pathways serve distinct endosome populations. J Cell Biol. 1994; 124: 689703. Holle, L., Song, W., Holle, E., Wei, Y., Wagner, T. and Yu, X. A matrix metalloproteinase 2 cleavable melittin/avidin conjugate specifically targets tumor cells in vitro and in vivo. Int J Oncol. 2003; 22: 93-98. Holm, T., Johansson, H., Lundberg, P., Pooga, M., Lindgren, M. and Langel, Ü. Studying the uptake of cell-penetrating peptides. Nat Protoc. 2006; 1: 1001-1005. Hua, Y., Vickers, T. A., Baker, B. F., Bennett, C. F. and Krainer, A. R. Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol. 2007; 5: e73. Hui, X., Han, Y., Liang, S., Liu, Z., Liu, J., Hong, L., Zhao, L., He, L., Cao, S., Chen, B., Yan, K., Jin, B., Chai, N., Wang, J., Wu, K. and Fan, D. Specific targeting of the vasculature of gastric cancer by a new tumor-homing peptide CGNSNPKSC. J Control Release. 2008; 131: 86-93. Ivanova, G. D., Arzumanov, A., Abes, R., Yin, H., Wood, M. J., Lebleu, B. and Gait, M. J. Improved cell-penetrating peptide-PNA conjugates for splicing redirection in HeLa cells and exon skipping in mdx mouse muscle. Nucleic Acids Res. 2008; Jedeszko, C. and Sloane, B. F. Cysteine cathepsins in human cancer. Biol Chem. 2004; 385: 1017-1027. Johansson, H. J., EL Andaloussi, S., Holm, T., Mäe, M., Jänes, J., Maimets, T. and Langel, Ü. Characterization of a novel cytotoxic cell-penetrating peptide derived from p14ARF protein. Mol Ther. 2008; 16: 115-123. 43 Kang, S. H., Cho, M. J. and Kole, R. Up-regulation of luciferase gene expression with antisense oligonucleotides: implications and applications in functional assay development. Biochemistry. 1998; 37: 6235-6239. Kirkin, V., Joos, S. and Zörnig, M. The role of Bcl-2 family members in tumorigenesis. Biochim Biophys Acta. 2004; 1644: 229-249. Lee, T. Y., Lin, C. T., Kuo, S. Y., Chang, D. K. and Wu, H. C. Peptidemediated targeting to tumor blood vessels of lung cancer for drug delivery. Cancer Res. 2007; 67: 10958-10965. Lindgren, M., Rosenthal-Aizman, K., Saar, K., Eiriksdottir, E., Jiang, Y., Sassian, M., Östlund, P., Hällbrink, M. and Langel, Ü. Overcoming methotrexate resistance in breast cancer tumour cells by the use of a new cell-penetrating peptide. Biochem Pharmacol. 2006; 71: 416-425. Liu, D., Ren, T. and Gao, X. Cationic transfection lipids. Curr Med Chem. 2003; 10: 1307-1315. Lundberg, P., EL Andaloussi, S., Sütlü, T., Johansson, H. and Langel, Ü. Delivery of short interfering RNA using endosomolytic cell-penetrating peptides. Faseb J. 2007; 21: 2664-2671. Lundin, P., Johansson, H., Guterstam, P., Holm, T., Hansen, M., Langel, Ü. and EL Andaloussi, S. Distinct Uptake Routes of Cell-Penetrating Peptide Conjugates. Bioconjug Chem. 2008; Mäe, M., Myrberg, H., EL Andaloussi, S. and Langel, Ü. Design of a tumor homing cell-penetrating peptide for drug delivery. Int J Pept Res Ther. 2008; Mattes, W. B., Hartley, J. A. and Kohn, K. W. DNA sequence selectivity of guanine-N7 alkylation by nitrogen mustards. Nucleic Acids Res. 1986; 14: 29712987. McDonald, D. M. and Choyke, P. L. Imaging of angiogenesis: from microscope to clinic. Nat Med. 2003; 9: 713-725. McGuire, J. J. Anticancer antifolates: current status and future directions. Curr Pharm Des. 2003; 9: 2593-2613. Meade, B. R. and Dowdy, S. F. Enhancing the cellular uptake of siRNA duplexes following noncovalent packaging with protein transduction domain peptides. Adv Drug Deliv Rev. 2008; 60: 530-536. Merrifield, R. B. Solid phase peptide synthesis. I. The synthesis of a tetrapeptide. J Am Chem Soc. 1963; 85: 2149-2154. Morris, M. C., Vidal, P., Chaloin, L., Heitz, F. and Divita, G. A new peptide vector for efficient delivery of oligonucleotides into mammalian cells. Nucleic Acids Res. 1997; 25: 2730-2736. Morvan, F., Porumb, H., Degols, G., Lefebvre, I., Pompon, A., Sproat, B. S., Rayner, B., Malvy, C., Lebleu, B. and Imbach, J. L. Comparative evaluation of seven oligonucleotide analogues as potential antisense agents. J Med Chem. 1993; 36: 280-287. Moulton, H. M., Fletcher, S., Neuman, B. W., McClorey, G., Stein, D. A., Abes, S., Wilton, S. D., Buchmeier, M. J., Lebleu, B. and Iversen, P. L. Cellpenetrating peptide-morpholino conjugates alter pre-mRNA splicing of DMD (Duchenne muscular dystrophy) and inhibit murine coronavirus replication in vivo. Biochem Soc Trans. 2007; 35: 826-828. 44 Moulton, H. M., Hase, M. C., Smith, K. M. and Iversen, P. L. HIV Tat peptide enhances cellular delivery of antisense morpholino oligomers. Antisense Nucleic Acid Drug Dev. 2003; 13: 31-43. Mueller, M. M. and Fusenig, N. E. Friends or foes - bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 2004; 4: 839-849. Myrberg, H., Zhang, L., Mäe, M. and Langel, Ü. Design of a tumorhoming cell-penetrating peptide. Bioconjug Chem. 2008; 19: 70-75. Nelimarkka, L. O., Nikkari, S. T., Ravanti, L. S., Kahari, V. M. and Jarvelainen, H. T. Collagenase-1, stromelysin-1 and 92 kDa gelatinase are associated with tumor necrosis factor-alpha induced morphological change of human endothelial cells in vitro. Matrix Biol. 1998; 17: 293-304. Neri, D. and Bicknell, R. Tumour vascular targeting. Nat Rev Cancer. 2005; 5: 436-446. Padari, K., Säälik, P., Hansen, M., Koppel, K., Raid, R., Langel, Ü. and Pooga, M. Cell transduction pathways of transportans. Bioconjug Chem. 2005; 16: 1399-1410. Palm, C., Jayamanne, M., Kjellander, M. and Hällbrink, M. Peptide degradation is a critical determinant for cell-penetrating peptide uptake. Biochim Biophys Acta. 2007; 1768: 1769-1776. Parkin, D. M., Bray, F., Ferlay, J. and Pisani, P. Global cancer statistics, 2002. CA Cancer J Clin. 2005; 55: 74-108. Partin, A. W., Hanks, G. E., Klein, E. A., Moul, J. W., Nelson, W. G. and Scher, H. I. Prostate-specific antigen as a marker of disease activity in prostate cancer. Oncology (Williston Park). 2002; 16: 1024-1038, 1042; discussion 1042, 10471028, 1051. Perera, Y., Farina, H. G., Hernandez, I., Mendoza, O., Serrano, J. M., Reyes, O., Gomez, D. E., Gomez, R. E., Acevedo, B. E., Alonso, D. F. and Perea, S. E. Systemic administration of a peptide that impairs the protein kinase (CK2) phosphorylation reduces solid tumor growth in mice. Int J Cancer. 2008; 122: 57-62. Pipkorn, R., Waldeck, W., Spring, H., Jenne, J. W. and Braun, K. Delivery of substances and their target-specific topical activation. Biochim Biophys Acta. 2006; 1758: 606-610. Pooga, M., Hallbrink, M., Zorko, M. and Langel, U. Cell penetration by transportan. Faseb J. 1998; 12: 67-77. Pujals, S., Fernandez-Carneado, J., Lopez-Iglesias, C., Kogan, M. J. and Giralt, E. Mechanistic aspects of CPP-mediated intracellular drug delivery: relevance of CPP self-assembly. Biochim Biophys Acta. 2006; 1758: 264-279. Richard, J. P., Melikov, K., Brooks, H., Prevot, P., Lebleu, B. and Chernomordik, L. V. Cellular uptake of unconjugated TAT peptide involves clathrindependent endocytosis and heparan sulfate receptors. J Biol Chem. 2005; 280: 15300-15306. Richard, J. P., Melikov, K., Vivés, E., Ramos, C., Verbeure, B., Gait, M. J., Chernomordik, L. V. and Lebleu, B. Cell-penetrating peptides. A reevaluation of the mechanism of cellular uptake. J Biol Chem. 2003; 278: 585-590. Rousselle, C., Clair, P., Lefauconnier, J. M., Kaczorek, M., Scherrmann, J. M. and Temsamani, J. New advances in the transport of doxorubicin through the 45 blood-brain barrier by a peptide vector-mediated strategy. Mol Pharmacol. 2000; 57: 679-686. Ruoslahti, E. Specialization of tumour vasculature. Nat Rev Cancer. 2002; 2: 83-90. Scaffidi, P. and Misteli, T. Reversal of the cellular phenotype in the premature aging disease Hutchinson-Gilford progeria syndrome. Nat Med. 2005; 11: 440-445. Shiraishi, T. and Nielsen, P. E. Enhanced delivery of cell-penetrating peptide-peptide nucleic acid conjugates by endosomal disruption. Nat Protoc. 2006; 1: 633-636. Simberg, D., Duza, T., Park, J. H., Essler, M., Pilch, J., Zhang, L., Derfus, A. M., Yang, M., Hoffman, R. M., Bhatia, S., Sailor, M. J. and Ruoslahti, E. Biomimetic amplification of nanoparticle homing to tumors. Proc Natl Acad Sci U S A. 2007; 104: 932-936. Smith, G. P. Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science. 1985; 228: 1315-1317. Sottrup-Jensen, L. Alpha-macroglobulins: structure, shape, and mechanism of proteinase complex formation. J Biol Chem. 1989; 264: 11539-11542. Soule, H. D., Vazguez, J., Long, A., Albert, S. and Brennan, M. A human cell line from a pleural effusion derived from a breast carcinoma. J Natl Cancer Inst. 1973; 51: 1409-1416. Srebrow, A. and Kornblihtt, A. R. The connection between splicing and cancer. J Cell Sci. 2006; 119: 2635-2641. Stewart, K. M., Horton, K. L. and Kelley, S. O. Cell-penetrating peptides as delivery vehicles for biology and medicine. Org Biomol Chem. 2008; 6: 22422255. Sugita, T., Yoshikawa, T., Mukai, Y., Yamanada, N., Imai, S., Nagano, K., Yoshida, Y., Shibata, H., Yoshioka, Y., Nakagawa, S., Kamada, H., Tsunoda, S. I. and Tsutsumi, Y. Comparative study on transduction and toxicity of protein transduction domains. Br J Pharmacol. 2008; 153: 1143-1152. Summerton, J. Morpholino antisense oligomers: the case for an RNase Hindependent structural type. Biochim Biophys Acta. 1999; 1489: 141-158. Tünnemann, G., Martin, R. M., Haupt, S., Patsch, C., Edenhofer, F. and Cardoso, M. C. Cargo-dependent mode of uptake and bioavailability of TATcontaining proteins and peptides in living cells. Faseb J. 2006; 20: 1775-1784. Vihinen, P. and Kähäri, V. M. Matrix metalloproteinases in cancer: prognostic markers and therapeutic targets. Int J Cancer. 2002; 99: 157-166. Wadia, J. S., Stan, R. V. and Dowdy, S. F. Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med. 2004; 10: 310-315. Wang, G. S. and Cooper, T. A. Splicing in disease: disruption of the splicing code and the decoding machinery. Nat Rev Genet. 2007; 8: 749-761. Warnecke, A., Fichtner, I., Sass, G. and Kratz, F. Synthesis, cleavage profile, and antitumor efficacy of an albumin-binding prodrug of methotrexate that is cleaved by plasmin and cathepsin B. Arch Pharm (Weinheim). 2007; 340: 389395. 46 Weigelt, B., Peterse, J. L. and van 't Veer, L. J. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005; 5: 591-602. Weller, K., Lauber, S., Lerch, M., Renaud, A., Merkle, H. P. and Zerbe, O. Biophysical and biological studies of end-group-modified derivatives of Pep-1. Biochemistry. 2005; 44: 15799-15811. Westermarck, J. and Kähäri, V. M. Regulation of matrix metalloproteinase expression in tumor invasion. Faseb J. 1999; 13: 781-792. Wood, M., Yin, H. and McClorey, G. Modulating the expression of disease genes with RNA-based therapy. PLoS Genet. 2007; 3: e109. Yamamura, T., Nakanishi, K., Hiroi, S., Kumaki, F., Sato, H., Aida, S. and Kawai, T. Expression of membrane-type-1-matrix metalloproteinase and metalloproteinase-2 in nonsmall cell lung carcinomas. Lung Cancer. 2002; 35: 249-255. Yang, W., Luo, D., Wang, S., Wang, R., Chen, R., Liu, Y., Zhu, T., Ma, X., Liu, R., Xu, G., Meng, L., Lu, Y., Zhou, J. and Ma, D. TMTP1, a novel tumorhoming peptide specifically targeting metastasis. Clin Cancer Res. 2008; 14: 54945502. Yang, Z., Strickland, D. K. and Bornstein, P. Extracellular matrix metalloproteinase 2 levels are regulated by the low density lipoprotein-related scavenger receptor and thrombospondin 2. J Biol Chem. 2001; 276: 8403-8408. Zamecnik, P. C. and Stephenson, M. L. Inhibition of Rous sarcoma virus replication and cell transformation by a specific oligodeoxynucleotide. Proc Natl Acad Sci U S A. 1978; 75: 280-284. Zhi, M., Wu, K. C., Dong, L., Hao, Z. M., Deng, T. Z., Hong, L., Liang, S. H., Zhao, P. T., Qiao, T. D., Wang, Y., Xu, X. and Fan, D. M. Characterization of a specific phage-displayed Peptide binding to vasculature of human gastric cancer. Cancer Biol Ther. 2004; 3: 1232-1235. 47