Next Article in Journal
Synthesis Mechanism of Low-Voltage Praseodymium Oxide Doped Zinc Oxide Varistor Ceramics Prepared Through Modified Citrate Gel Coating
Next Article in Special Issue
Identification of Late Embryogenesis Abundant (LEA) Protein Putative Interactors Using Phage Display
Previous Article in Journal
Sol-Gel-Derived Hydroxyapatite-Carbon Nanotube/Titania Coatings on Titanium Substrates
Previous Article in Special Issue
Filamentous Bacteriophage Fd as an Antigen Delivery System in Vaccination
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Phage Displayed Peptides/Antibodies Recognizing Growth Factors and Their Tyrosine Kinase Receptors as Tools for Anti-Cancer Therapeutics

1
Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, viale Europa, 11, 25123 Brescia, Italy
2
Fibroblast Reprogramming Unit, Department of Biomedical Sciences and Biotechnology, viale Europa, 11, 25123 Brescia, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2012, 13(4), 5254-5277; https://doi.org/10.3390/ijms13045254
Submission received: 9 February 2012 / Revised: 9 April 2012 / Accepted: 20 April 2012 / Published: 24 April 2012
(This article belongs to the Special Issue Phage Display)

Abstract

:
The basic idea of displaying peptides on a phage, introduced by George P. Smith in 1985, was greatly developed and improved by McCafferty and colleagues at the MRC Laboratory of Molecular Biology and, later, by Barbas and colleagues at the Scripps Research Institute. Their approach was dedicated to building a system for the production of antibodies, similar to a naïve B cell repertoire, in order to by-pass the standard hybridoma technology that requires animal immunization. Both groups merged the phage display technology with an antibody library to obtain a huge number of phage variants, each of them carrying a specific antibody ready to bind its target molecule, allowing, later on, rare phage (one in a million) to be isolated by affinity chromatography. Here, we will briefly review the basis of the technology and the therapeutic application of phage-derived bioactive molecules when addressed against key players in tumor development and progression: growth factors and their tyrosine kinase receptors.

1. Antibody Display

Antibody molecules are commonly composed of a pair of identical heavy chain (H) and a pair of identical light chain (L) polypeptides, held together by disulfide bridges and non-covalent bonds in a Y shaped form [1]. The antigen-binding site in the molecule, that is located at the two Y tips, is the variable region (V) that diversifies every single molecule and is generated by the combination of N-terminal domains of the H and L chains in the so-called fragment antigen-binding (Fab) [2].
In 1988, a functional variable region, the immunoglobulin Fv fragment, was produced in Escherichia coli [3] and one year later Orlandi et al. demonstrated the cloning by PCR of the variable regions of H and L chains, starting from hybridoma cDNA [4]. From this basis came the idea of displaying the antibody library on a phage [5,6] and since then, the genetic information of H and L domains has been transcribed from B lymphocytes or pre-B lymphocytes of several species, PCR amplified, mutated or randomized and subsequently combined by PCR assembly or sequential cloning in the phagemids [79]. The filamentous phage commonly used for peptide/antibody display purposes is the single strand DNA virus M13 that presents a rod shaped structure with a circular genome of 6407 nucleotides enclosed in approximately 2700 copies of the major coat protein P8, and capped with 5 copies of minor coat proteins (P9, P6, P3) on the ends [9,10]. The peptides/antibodies of interest are mostly exposed or “displayed” fused to the N-terminus of P3 (3–5 copies/phage) or P8 coat proteins (2700 copies/phage) [11]. Despite their abundance on the phage surface, the choice of the protein target depends on the size of the inserted peptide, since long aminoacidic sequences can interfere with physiological phage protein function. Indeed, P8 libraries can display only a six aminoacid peptide in a large number of copies [12], while P3 insertion can tolerate up to 43 amino acids without loss of phage infectivity [13]. However, several advances in engineering M13 coat proteins for improved performances have been made in recent years, thus providing a greater variety of N- and C-terminal display scaffolds, as well as artificial coat proteins to improve the stability of the genetic information and to increase the number of exogenous peptides exposed on the phage surface [14]. On the other side, several small formats of the antibody molecule that maintain the Fab site have been created and inserted within the phage genome [15]. These antibody fragments, either an entire Fab, a fragment variable (Fv) or a linker-stabilized single chain Fv (scFv), can be displayed by fusion with phage coat proteins [15]. Different antibody formats discussed in this review are shown in Figure 1. The cloning of human repertoires has led to the construction of humanized antibody libraries whose products can be used safely for human diagnostics and therapeutics.
An antibody is used by the immune system to recognize and, eventually, neutralize foreign hosts such as bacteria, viruses, etc. Behind the idea of an antibody display is the possibility that the selected molecule will recognize and possibly neutralize the function of the antigen, although the same result could be hypothetically obtained by using a peptide. Nevertheless, the sequencing of the DNA that codify for the aminoacidic sequence of the variable regions of the antibody will allow its transfer, by molecular biology techniques, to various antibody formats, as needed. Challenging the library with the desired human antigen will allow the selection of the binder phages that will be eluted, amplified, and utilized in several cycles of selection, in order to isolate the best candidate for the following purposes. At the end of the procedure, the phage DNA can be sequenced to identify the inserted variable regions, and, by changing the bacterial host, large amounts of soluble antibody fragment or peptide can be produced for further characterization [15].

2. Growth Factors and Cancer

Growth factors play a fundamental physiological role in many cell types by driving a wide variety of cellular functions, including growth, differentiation, and angiogenesis. Growth factor signaling involves their binding to high affinity sites on the cellular surface, mostly represented by tyrosine kinase receptor (RTK) molecules. All RTKs have a similar molecular architecture, with ligand-binding domains in the extracellular region, a single transmembrane helix, and a cytoplasmic region that contains the tyrosine kinase (TK) domain plus additional carboxy-terminal and juxtamembrane regulatory regions [17]. Examples of RTKs involved in cell proliferation are shown in Figure 2. Although each growth factor acts in its own way, the common strategy utilized to activate RTK signaling involves the induction of receptor dimerization that, in turn, activates the phosphorylation of specific residues on the receptor intracellular tail. Phosphorylated tyrosines become docking sites for cytoplasmic proteins, whose binding initiate the cascade of events that leads to the activation of transcription factors involved in cell growth, survival, differentiation, or angiogenesis [17].
Cancer cells have defects in regulatory circuits that govern cell proliferation and homeostasis [18]. It has been suggested that cancer is the manifestation of six essential alterations in cellular physiology: self-sufficiency in growth signals, insensitivity to growth-inhibitory signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis [18]. Tumor cells sustain their dysregulated proliferation in many ways: in an autocrine manner, by secreting the growth factors themselves and exposing the cognate receptor on cell surface or by inducing host stromal cells to produce the required growth stimulus [19,20]. On the other hand, receptor signaling can be deregulated by enhancing the number of RTKs on the cell surface, thus becoming hyper-responsive to unaltered growth factor availability or by mutating components of the signaling cascade, leading to cell proliferation independently by the presence of receptor ligand [17]. Over-expression and/or structural alteration of RTK family members are often associated to human cancers and tumor cells are known to use RTK transduction pathways to achieve tumor growth, angiogenesis and metastasis. Indeed, recent sequencing efforts in a wide variety of tumors have identified mutations in numerous RTKs, collected in the Catalogue of Somatic Mutations in Cancer (COSMIC) database [21]. A general overview of the association between tumor progression and the altered expression of growth factor/RTK axis is shown in Table 1.

2.1. EGF Family/ERBB Family

The ERBB family of RTKs includes the epidermal growth factor receptor (EGFR) ERBB1, also called HER1, and the closely related ERBB2 (HER2/Neu), ERBB3 (HER3), and ERBB4 (HER4) proteins. Aberrant activation by mutation or overexpression of ERBB receptors and their ligands has been found in many human cancers [67]. Indeed, EGFR levels are increased in many epithelial tumors, including head and neck, lung, breast, and colorectal cancers, and its over-expression usually correlates with poor clinical outcome [26,68]. Also, HER2 is frequently altered in human cancers like breast, ovarian, lung, and prostate carcinoma [69] and its up-regulation causes malignant transformation of mammary epithelial cells, breast, ovarian, lung, and prostate carcinoma. Approximately 25% of invasive primary breast cancers exhibit HER2 gene amplification, and this molecular feature correlates with reduced patient survival [70]. Targeting EGFR prevents ligand-induced receptor activation and downstream signaling, resulting in cell cycle arrest, promotion of apoptosis, and inhibition of angiogenesis [71].

2.2. HGF/MET

Hepatocyte growth factor (HGF) is expressed by stromal cells while its receptor, HGFR/c-MET, is expressed in a variety of epithelial cells that are involved in organogenesis (lung, kidney, breast) or in neuronal development, muscle development, hematopoiesis and angiogenesis [72]. The HGF-HGFR/c-MET pathway is involved in cancer as well. Indeed, the activation of HGFR/c-MET has been reported to trigger cancer cell proliferation, migration and invasion and to promote tumor vessel angiogenesis, since HGF directly stimulates endothelial cell proliferation and migration [7375]. Aberrant c-MET activation, including gene amplification, mutation, and overexpression, has been found in hematological malignancies and most solid tumors [76], including gastric, liver, and colon cancers [27,77]. In lung cancer it has been shown that, due to its cross reactivity/compensation effect with EGFR, HGFR/c-MET gene amplification can confer resistance to EGFR-targeting drugs. Thus, the targeting of HGFR/c-MET can potentially be used as a strategy for destroying refractory drug-resistant cancer cells [72].

2.3. IGFs/IGFRs

Research and clinical studies have shown that the Insulin Growth Factor 1 Receptor (IGF1R) and its ligands, IGF1 and IGF2, play an essential role, not only in normal growth and development, but also in the development, maintenance, and progression of breast, prostate, and colon cancer [78,79]. IGF mitogenic signaling pathway is an attractive therapeutic target in breast cancer per se [80], since high IGF1R levels are associated with resistance to treatment with a monoclonal antibody (mAb) that selectively recognizes the extracellular domain of HER2 and is currently used in the treatment of ERBB2-overexpressing breast cancer [81,82].

2.4. VEGFs/VEGFRs

Angiogenesis is a multistep process that results in new blood vessel formation from pre-existing vasculature whose regulation results from a dynamic balance between pro-angiogenic and anti-angiogenic factors [83]. As stated before, a pro-angiogenic switch is strictly required for tumor growth, invasion and metastatic dissemination [84]. Indeed, tumor cells produce growth factors that induce proliferation and migration of endothelial cells, such as Vascular Endothelial Growth Factors (VEGFs), Fibroblast Growth Factors (FGFs), Platelet-Derived Growth Factors (PDGFs) and angiopoietins [85].
The VEGF family of ligands and receptors play a central role in both physiological and pathological angiogenesis, and the development of VEGF antagonists is essential in anti-angiogenesis research [86]. The VEGF family is composed of seven members (VEGF (A–F), PlGF) that act through three structurally homologous tyrosine kinase receptors [VEGFR (1–3)] [87]. VEGF is a homodimeric, basic, 45 kDa glycoprotein, specific for vascular endothelial cells [88] and its binding to VEGFR2/FLK1/KDR causes endothelial cell proliferation, angiogenesis, and increased vessel permeability [89,90]. Anti-angiogenic compounds are postulated both to reduce tumor vascularization, and also to normalize vasculature within the tumor to allow the delivery of anti-tumor drugs [91]. Thus anti-angiogenic drugs specifically targeting VEGF or VEGF receptors (VEGFRs) represent a strategy for tumor control and treatment [92]. Since the introduction of the first mAb approved by the Food and Drug Administration (FDA), humanized bevacizumab (Avastin) that neutralizes VEGF, several drugs targeting VEGF-related pathways have been developed [93]. Also, recombinant antibodies, including scFv fragments, were selected against VEGF or the VEGF-VEGFR complex [9496].

2.5. FGFs/FGFRs

FGFs represent a family of at least 22 structurally homologous polypeptide growth factors that are expressed in almost all tissues. FGFs have been implicated in multiple biological processes during embryo development, wound healing, hematopoiesis, and angiogenesis [97,98]. Among them, FGF1 and FGF2 were identified as angiogenic factors [99,100], promoting the proliferation, migration, differentiation and tubulogenesis of endothelial cells in vitro and being potent stimulators of angiogenesis in vivo [101], thus playing an important role in tumorigenesis. FGFs interact with a family of four distinct, high affinity RTKs, designated FGFR1/4, whose number is greatly increased by the generation of alternative splicing isoforms of FGFR1, FGFR2 and FGFR3 [102,103]. FGF2, FGFR1, and FGFR2 have been shown to be involved in prostatic cancers [104], non-small cell lung carcinoma [39], and pancreatic cancers [57]. FGFR1 is widely expressed in a variety of tumor-derived cells and tissues and is the major Fibroblast Growth Factor Receptor (FGFR) of vascular endothelial cells [105]. It transduces pro-angiogenic and proliferative signals in human cancers, thus it may represent a target for the development of anti-angiogenic/anti-neoplastic therapies [106,107].
All these observations point to growth factors and their cognate RTK as pivotal targets in cancer therapy approaches. The aim that has been pursued in recent years with phage display libraries is the identification of an antibody or a peptide, recognizing either the growth factor or the receptor that can inhibit their interaction, thus suppressing the resulting proliferative signaling.
Several strategies to block the mitogenic signaling pathway that is activated following ligand-receptor interactions are being evaluated. There are three general classes of agents that inhibit tyrosine kinase receptors: blocking antibodies, small kinase inhibitors, and soluble ligand traps or receptor decoys. To date, agents belonging to all these classes are currently available for therapeutic intervention, and are mainly represented by mAbs directed at the ligand-binding site in the extracellular domain of the receptor and low-molecular-weight inhibitors of intracellular tyrosine kinase activity [108].

3. Preclinical Studies

Preclinical approaches using phage display technology are mainly addressed to find and characterize small molecules such as antibodies and peptides with targeting and in some cases neutralizing activity against various members of the growth factors and receptor families. In the last decade almost all the main players involved in tumor growth, angiogenesis, transition processes and all the main steps of cancer progression have been targeted. Obviously, in cancer therapy, the “anti-growth factor approach” addressed to block the ligand-receptor interaction represents a very promising strategy. As already described, growth factors mainly work through their receptors which are plasma membrane embedded proteins generally constituted of three basic parts: the extracellular, the transmembrane and the intracellular regions with functionally and spatially distinct roles. Due to their accessibility and also to their prominent role, receptors represent the largest group of drug targets and most of the therapeutic molecules and strategies are addressed against the extracellular portion/ligand-binding site. The intracellular region is less accessible to peptides or small proteins, which are often too hydrophilic to diffuse across plasma membranes. For this reason only small chemicals, like tyrosine kinase inhibitors, modulate receptor function via an interaction with its intracellular signal-transducing region.
Even though the targeting of a receptor can potentially block all the correlated growth factors, sometimes the physiological interaction with a particular ligand must be preserved. Then, specific growth factors become achievable targets, as they have secreted proteins available in the extracellular space and are often embedded in tumor stroma.

3.1. Phages Displaying Peptides

Phage display has been applied to isolate and characterize peptides endowed with anti-growth factor activities. Recently, specific EGFR binding peptides from combinatorial library selection were first screened and then narrowed by structure-function analysis. The most active motif necessary and sufficient for specific EGFR ligand binding, corresponding to EGFR 283–287 (CVRAC) was synthetized as retro-inverted derivative and became the preclinical prototype of choice [108].
Some years before, another ERBB2-avid peptide, discovered from phage display, was evaluated in human breast carcinoma cells and in breast carcinoma-xenografted mice for its potential to be used as a tumor-imaging agent and as a vehicle for specific delivery of radionuclide or cytotoxic agents for tumors overexpressing ERBB2 [109].
A random dodecapeptide library was screened against the fusion protein GST-VEGFR2 (extracellular domains I-IV of VEGFR2 fused to glutathione S-transferase). The most active peptide that competed with the VEGF binding to its receptor exerted anti-angiogenic activity both in vitro (preventing human endothelial cell proliferation) and in vivo (in the chick embryo chorioallantoic membrane) and reduced tumor growth in mice [110]. Another VEGFR-binding peptide was generated by mini peptide display technology and, similar to the previous one, was shown to effectively interfere with tumor growth and metastasis due to its anti-angiogenic effects and to block intracellular signaling pathways involved in tumor progression [111]. Again, phage library-derived heptapeptides [112114] that completely abolished VEGF binding to cell-displayed VEGFR2, showed the in vitro inhibition of VEGF-mediated proliferation of human vascular endothelial cells and a suppression of VEGF-induced angiogenesis in a rabbit corneal model [114].
Also, the FGF2/FGFR pathway has been targeted and inhibited by synthetic peptides. Peptides were selected for their binding affinity to anti-FGF2 antibodies, thus suggesting structural similarity with FGFR. This “mirror” characteristic revealed peptides able to bind and neutralize FGF2. These selected hexapeptides inhibited binding of FGF2 to its high-affinity receptor, and suppressed basal and FGF2-induced proliferation of vascular endothelial cells at submicromolar concentrations [107].
Many other peptides are in preclinical development but, unfortunately, their binding affinities are, in general, too low to support their therapeutic use [115]. This is especially true for antagonistic peptides, which need to occupy at least half of the ligand/receptor interaction site to produce an inhibitory effect. Nevertheless, these targeting peptides can be used to design peptido-mimetic molecules or further improved versions, which are more effective in targeting specific cells or tissues when conjugated with therapeutic or diagnostic agents [116,117].

3.2. Phages Displaying Antibodies

The wider class of phage display-derived molecules is certainly represented by antibody fragments, which can be easily exploited for their targeting or neutralizing properties. By the way, neutralizing antibodies do not necessarily recognize the exact receptor-binding epitope of the ligand but may block the ligand-receptor interaction by binding to sites proximal to these regions and sterically prevent their interaction.
There are many examples of anti-growth factor receptor antibodies in the preclinical field, isolated from phage display libraries. The first one, a scFv, has been selected from a library whose repertoire was derived from spleen cells of mice immunized with a soluble form of human VEGFR2/KDR [118]. This antibody fragment was shown to inhibit VEGF-induced KDR phosphorylation and VEGF-stimulated DNA synthesis in human umbilical vein endothelial cells (HUVEC) [118]. Similarly, two other scFvs significantly suppressed the mitogenic response of HUVEC to recombinant human VEGF in a dose-dependent manner and reduced VEGF-dependent cell proliferation by 60% and 40%, respectively. In vivo analysis of these recombinant antibodies in a rat cornea angiogenesis model revealed that both antibodies suppressed the development of new corneal vessels [119].
Single-domain antibodies in VHH format, specific for FGFR1, were isolated from a phage-display llama naïve library. Two populations of competitive antibodies were identified and both of them, although recognizing distinct epitopes, specifically labeled receptor-expressing cells in immunofluorescence. Antibodies from both populations effectively prevented FGF-dependent internalization and nuclear accumulation of the receptor in cultured cells [120]. Another scFv antibody (RR-C2) specifically recognized FGFR1α and FGFR1β with a K(d) value of 300 and 144 nmol/L for the two receptor isoforms, respectively [106]. The antibody fragment also recognized FGFR1 when the receptor was exposed on the cell surface, prevented the formation of the ternary complex among FGFR1, its ligand FGF2, and cell surface heparan sulfate proteoglycans, and inhibited FGF2-mediated mitogenic activity in endothelial cells of different origin in a nanomolar range of concentrations [106]. In vivo, this anti-FGFR1 scFv hampered the angiogenic activities exerted by FGF2 in the chick embryo chorioallantoic membrane assay and by an FGF-dependent breast cancer cell line in Matrigel assay [106].
Different approaches in this preclinical field include the generation and characterization of stable Ig-like bispecific antibodies (BsAb) that target two antigens at the same time. The BsAb molecule EI-04 was constructed with a scFv against IGF1R attached to the carboxyl-terminus of an IgG against EGFR. EI-04 binds to human EGFR and IGF1R with sub-nanomolar affinity, co-engages the two receptors simultaneously and blocks the binding of their respective ligands with potency similar to the parental mAbs. Because of its double inhibition, an enhanced in vivo anti-tumor efficacy over the parental mAbs has been shown in two xenograft models [121]. Indeed, in a targeting/drug delivery perspective some scFvs antibodies were also generated that specifically bind to c-MET protein, as aberrantly expressed c-MET has been implicated in human lung cancer as well as malignancy, metastasis, and drug-resistance in other human cancers. The anti-c-MET scFv showed a selective binding and internalization in several lung cancer cell lines expressing c-MET and in vivo fluorescent imaging by scFv-conjugated quantum dots showed higher tumor uptake and increased tumor to normal tissue ratios [122]. In addition, conjugation of scFv with PEGylated liposomes enabled higher delivery of doxorubicin into tumor, thus enhancing its cytotoxic activity [122]. Indeed, several other drugs have been conjugated to scFv in order to enhance their therapeutic potential such as the tubulin polymerization inhibitor monomethylauristatin [123], the potent cytotoxic agent DM1 [124], and alpha-amanitin, a toxin known to inhibit DNA transcription [125].
Regardless of the huge amount of potential candidates, only a restricted number of molecules enter clinical trial screening. In the last decade, phage display has become an interesting and increasing source of such candidates. Despite the availability of the XenoMouse model, a mouse genetically engineered with a “humanized” humoral immune system [126], humanized phage-display technology is the preferred choice of many companies for the ease of use and for the speed of isolation of the molecules of interest [127]. To date, this technology has been used to create at least 35 human mAbs that entered clinical development, mainly applied in oncology or as immunomodulatory drugs.

4. Clinical Studies

In the last decade, 45% of total mAb candidates for clinical use were derived from human repertoires and many others are actually in clinical development. So far, several human mAbs have been approved by the FDA for marketing in the United States: adalimumab (Humira; Abbott) [128] and golimumab (Simponi; Centocor) [129], for the treatment of rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn’s disease, moderate-to-severe chronic psoriasis and juvenile idiopathic arthritis; panitumumab (Vectibix; Amgen) to treat EGFR-expressing metastatic colorectal cancer [130]; canakinumab (Ilaris; Novartis), indicated for the treatment of cryopyrin-associated periodic syndromes [131]; ustekinumab (Stelara; Johnson & Johnson), used to treat moderate to severe plaque psoriasis and under investigation for multiple sclerosis, psoriatic arthritis and sarcoidosis [132]; ofatumumab (Arzerra; Genmab), for treating chronic lymphocytic leukemia, refractory to fludarabine/alemtuzumab, and also useful in treating follicular non-Hodgkin’s lymphoma, diffuse large B cell lymphoma, rheumatoid arthritis and relapsing remitting multiple sclerosis [133]; denosumab (Prolia; Amgen), approved for use in postmenopausal women with a risk of osteoporosis and for the prevention of skeletal-related events in patients with bone metastases from solid tumors [134]; and the recently approved belimumab, (Human Genome Sciences) for the treatment of systemic lupus erythematosus [135] and ipilimumab (Bristol-Myers Squibb) for the treatment of melanoma [136], that is also undergoing clinical trials for the treatment of non-small cell lung carcinoma and metastatic hormone-refractory prostate cancer [137]. Moreover, Human Genome Sciences received Biologics License Application from the FDA for raxibacumab, intended for the prophylaxis and treatment of inhaled anthrax [138].
As reported, the field of clinical application for these antibodies is predominantly autoimmunity and inflammatory pathologies followed by cancer therapy. Notably, and despite the central and pleiotropic role of growth factors in cancer biology, only a restricted number of antibodies that entered clinical trials are directed against growth factors and their receptors. For sure, the academic and preclinical effort in growth factor targeting has increased in recent years and many novel candidates are in preclinical validation, waiting for clinical development. Among them, phage-derived compounds represent an emerging category that, in terms of number of candidate identifications, is possibly overcoming other technologies such as chimerization or humanization processes, and XenoMouse platform. As companies mainly own phage display libraries, the preclinical process is often well documented on peer-reviewed original articles while less information is available on the following refinement process performed mainly by private institutions.
Based on these considerations, we tried to provide an overview on the ongoing clinical studies based on human antibodies derived from phage display and conceived as growth factors/receptors targeting agents [139]. To date, some phage derivatives against these classes of targets are in clinical evaluation, among them: IMC-A12, IMC-11F8, IMC1121b and IMC-3C5 (by ImClone), AMG-479 (by Amgen), VGX100 (by Circadian Technologies Ltd). A brief summary of their main properties is reported in Table 2.

4.1. IMC-A12 (Cixutumumab)

It is a recombinant fully human IgG1 mAb that specifically targets the human IGF1R. IMC-A12 binds with high affinity to the IGF1R, inhibits ligand-dependent receptor activation and downstream signaling. IMC-A12 also mediates robust internalization and degradation of the IGF1R. Although promising single-agent activity has been observed, the most impressive effects of targeting the IGF1R with IMC-A12 have been noted when this molecule was combined with cytotoxic agents or other targeted therapeutics [140]. In a randomized phase II study IMC-A12 was tested alone and in combination with Cetuximab in patients with metastatic colorectal cancer, refractory to anti-EGFR mAb. In both combinations, results were insufficient to warrant additional study in patients [141]. In 2010, a phase II clinical trial began and is still ongoing for the use of IMC-A12 in patients who had chemotherapy-treated mesothelioma (ClinicalTrials.gov Identifier: NCT01160458).

4.2. IMC-11F8 (Necitumumab)

It is a fully human IgG1 mAb against the EGFR with antitumor potency similar to the chimeric Cetuximab/Erbitux and might represent a safer therapeutic alternative [145]. IMC-11F8 specifically blocks the receptor/ligand interaction and is intended for the treatment of solid tumors, such as colorectal cancer, by inducing tumor-specific cell death. IMC-11F8 was evaluated in phase II clinical trial for colorectal cancer therapy and the results were well-tolerated, associated with preliminary evidence of antitumor activity, achieving biologically-relevant concentrations throughout the dosing period [146]. Also, bispecific antibodies obtained, combining IMC-A12 and IMC-11F8, were reported but have not entered the clinic yet [139].

4.3. IMC-1121b (Ramucirumab)

It is a human IgG1 mAb that specifically recognizes with high affinity (50 pM) the extracellular VEGF-binding domain of the VEGFR2 [32]. IMC-1121b prevents neo-angiogenesis in solid tumors by inhibiting and blocking VEGFR2, similarly to the FDA-approved antibody bevacizumab (Avastin) that targets VEGF itself. Nevertheless, in contrast to other agents directed against the VEGFR2/VEGF axis, ramucirumab binds a specific epitope on the extracellular domain of VEGFR2, thereby blocking all VEGF ligands from binding to this therapeutically-validated target [147]. IMC-1121b is currently in phase II clinical trial for the treatment of metastatic malignant melanoma, metastatic renal cell carcinoma, and liver cancer. Recently, in a pharmacologic and biologic study of phase I, patients with advanced solid malignancies were treated once a week with escalating doses of ramucirumab. At the end of the study, anti-angiogenic effects and anti-tumor activity were observed over a wide range of dose levels, suggesting that ramucirumab may have a favorable therapeutic index in treating malignancies amenable to VEGFR2 inhibition [32].

4.4. IMC-3C5

It is a fully human mAb that specifically binds to and hampers VEGFR3 signaling, thus inhibiting angiogenesis and decreasing tumor nutrient supply [145]. Target selectivity is guaranteed by the fact that VEGFR3 plays a critical role in the embryonic vascular system development but its expression is postnatally restricted to the endothelial cells of lymphatic vessels [148]. Recent studies have shown that VEGFR3 is expressed in many solid and hematologic malignancies, because of its involvement in the tumor-associated lymphangiogenesis process [149,150], making the molecule a strong candidate for anti-tumor therapy. Indeed, a phase I study that began in 2011 will examine the safety and tolerability of escalating doses of IMC-3C5 in patients with advanced solid tumors that are refractory to standard therapy or for which no standard therapy is available (ClinicalTrials.gov Identifier: NCT01288989).

4.5. AMG-479 (Ganitumab)

It is an IgG1 mAb that binds to and blocks IGF1R [142], initially characterized for its therapeutic activity on pancreatic carcinoma cells in vitro and in xenograft models [142]. Ganitumab successfully demonstrated good pharmacokinetic and pharmacodynamic characteristics in a phase I trial [143] and then entered a small, randomized, placebo-controlled phase II study where the combination with AMG-479/gemcitabine (a nucleoside analog used in chemotherapy) improved overall survival at six months (primary endpoint) and progression-free survival in patients with metastatic pancreatic cancer (Poster Discussion at the 2010 American Society of Clinical Oncology Annual Meeting) [144]. This promising candidate is now moving into phase III testing in this patient population.

4.6. VGX-100 (Fresolimumab)

It is a human antibody, developed by Circadian Technologies Ltd. that acts against one of the VEGFR3 ligands: the human VEGFC. As VEGFC has a specific role in causing new lymphatic vessel development, its blockade in tumors may slow cancer metastasis. Treatment for cancers, particularly glioblastoma and metastatic colorectal cancers, are the first target indications for VGX-100. Additionally, Circadian is developing VGX-100 for a number of other cancer indications, as well as an agent to treat front-of the-eye diseases [151]. In 2012 a phase I study will examine the safety and tolerability of escalating doses of VGX-100 in patients with advanced metastatic solid tumors who have no other standard treatment options both as a immunotherapy and also when used in combination with other anti-angiogenic agents such as bevacizumab (ClinicalTrials.gov Identifier: NCT01514123).
By means of phage display technology, the effortless identification of the amino acid sequence of the isolated phage has allowed the in vitro construction of several functional antibodies, carrying the identified antigen-binding site to be used as cancer targeting agents. In addition to what we described in this review, two further comments can point up the ongoing beliefs for cancer treatment: the complexity of tumor pathology needs a multitarget approach and, where possible, antibody conjugation. The established idea is that the simultaneous inhibition of angiogenesis, cancer cells and possibly the tumor supporting stroma would gain better results in fighting the disease. To this purpose several antibodies derived from phage display, targeting tumor overexpressed cytokines (e.g., GC-1008, an anti-Transforming Growth Factor β cytokine [152]), tumor stromal antigens (e.g., L19, an anti-isoform B of fibronectin [153]), or tumor-associated antigens (e.g., adecatumumab, that recognize epithelial cell-adhesion molecules [154]) are currently in clinical trials. Furthermore, once the target is really narrowed by the antigen recognition property of the antibody, the conjugation of the latter with cytotoxic or immunomodulatory drugs will strongly increase the therapeutic benefit for patients while reducing systemic drug toxicity [155].
In conclusion, antibody displayed on phage particles has become a milestone technique for the development of humanized therapeutics. Starting from laboratory bench, a lot of promising anti-cancer molecules recognizing growth factors and their TK receptors have been isolated, sequenced, and transferred to various antibody formats up to the bedside of patients. It should be pointed out that the improvements of this technology in combination with the study of the proteome of tumor/cancer cells would greatly increase the number and efficacy of these therapeutic molecules.

Acknowledgments

We are indebted with Patrizia Alessi for helpful discussion. This work was supported by grants from AIRC, MIUR, and Fondazione Berlucchi.

References

  1. Edelman, G.M. Antibody structure and molecular immunology. Science 1973, 180, 830–840. [Google Scholar]
  2. Davies, D.R.; Cohen, G.H. Interactions of protein antigens with antibodies. Proc. Natl. Acad. Sci. USA 1996, 93, 7–12. [Google Scholar]
  3. Skerra, A.; Pluckthun, A. Assembly of a functional immunoglobulin Fv fragment in Escherichia coli. Science 1988, 240, 1038–1041. [Google Scholar]
  4. Orlandi, R.; Gussow, D.H.; Jones, P.T.; Winter, G. Cloning immunoglobulin variable domains for expression by the polymerase chain reaction. Proc. Natl. Acad. Sci. USA 1989, 86, 3833–3837. [Google Scholar]
  5. McCafferty, J.; Griffiths, A.D.; Winter, G.; Chiswell, D.J. Phage antibodies: Filamentous phage displaying antibody variable domains. Nature 1990, 348, 552–554. [Google Scholar]
  6. Barbas, C.F., 3rd; Kang, A.S.; Lerner, R.A.; Benkovic, S.J. Assembly of combinatorial antibody libraries on phage surfaces: The gene III site. Proc. Natl. Acad. Sci. USA 1991, 88, 7978–7982. [Google Scholar]
  7. Watkins, N.A.; Ouwehand, W.H. Introduction to antibody engineering and phage display. Vox Sang 2000, 78, 72–79. [Google Scholar]
  8. Engberg, J.; Andersen, P.S.; Nielsen, L.K.; Dziegiel, M.; Johansen, L.K.; Albrechtsen, B. Phage-display libraries of murine and human antibody Fab fragments. Mol. Biotechnol 1996, 6, 287–310. [Google Scholar]
  9. Marks, J.D.; Hoogenboom, H.R.; Bonnert, T.P.; McCafferty, J.; Griffiths, A.D.; Winter, G. By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J. Mol. Biol 1991, 222, 581–597. [Google Scholar]
  10. Smith, G.P.; Petrenko, V.A. Phage display. Chem. Rev 1997, 97, 391–410. [Google Scholar]
  11. Georgieva, Y.; Konthur, Z. Design and screening of M13 phage display cDNA libraries. Molecules 2011, 16, 1667–1681. [Google Scholar]
  12. Iannolo, G.; Minenkova, O.; Petruzzelli, R.; Cesareni, G. Modifying filamentous phage capsid: Limits in the size of the major capsid protein. J. Mol. Biol 1995, 248, 835–844. [Google Scholar]
  13. Pande, J.; Szewczyk, M.M.; Grover, A.K. Phage display: Concept, innovations, applications and future. Biotechnol. Adv 2010, 28, 849–858. [Google Scholar]
  14. Sidhu, S.S. Engineering M13 for phage display. Biomol. Eng 2001, 18, 57–63. [Google Scholar]
  15. Azzazy, H.M.; Highsmith, W.E., Jr. Phage display technology: Clinical applications and recent innovations. Clin. Biochem 2002, 35, 425–445. [Google Scholar]
  16. Holliger, P.; Hudson, P.J. Engineered antibody fragments and the rise of single domains. Nat. Biotechnol 2005, 23, 1126–1136. [Google Scholar]
  17. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar]
  18. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar]
  19. Cheng, N.; Chytil, A.; Shyr, Y.; Joly, A.; Moses, H.L. Transforming growth factor-beta signaling-deficient fibroblasts enhance hepatocyte growth factor signaling in mammary carcinoma cells to promote scattering and invasion. Mol. Cancer Res 2008, 6, 1521–1533. [Google Scholar]
  20. Bhowmick, N.A.; Neilson, E.G.; Moses, H.L. Stromal fibroblasts in cancer initiation and progression. Nature 2004, 432, 332–337. [Google Scholar]
  21. Forbes, S.A.; Tang, G.; Bindal, N.; Bamford, S.; Dawson, E.; Cole, C.; Kok, C.Y.; Jia, M.; Ewing, R.; Menzies, A.; et al. COSMIC (the Catalogue of Somatic Mutations in Cancer): A resource to investigate acquired mutations in human cancer. Nucleic Acids Res 2010, 38, D652–D657. [Google Scholar]
  22. Takahashi, J.A.; Fukumoto, M.; Igarashi, K.; Oda, Y.; Kikuchi, H.; Hatanaka, M. Correlation of basic fibroblast growth factor expression levels with the degree of malignancy and vascularity in human gliomas. J. Neurosurg 1992, 76, 792–798. [Google Scholar]
  23. Humphrey, P.A.; Gangarosa, L.M.; Wong, A.J.; Archer, G.E.; Lund-Johansen, M.; Bjerkvig, R.; Laerum, O.D.; Friedman, H.S.; Bigner, D.D. Deletion-mutant epidermal growth factor receptor in human gliomas: Effects of type II mutation on receptor function. Biochem. Biophys. Res. Commun 1991, 178, 1413–1420. [Google Scholar]
  24. Libermann, T.A.; Nusbaum, H.R.; Razon, N.; Kris, R.; Lax, I.; Soreq, H.; Whittle, N.; Waterfield, M.D.; Ullrich, A.; Schlessinger, J. Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature 1985, 313, 144–147. [Google Scholar]
  25. Jenny, B.; Harrison, J.A.; Baetens, D.; Tille, J.C.; Burkhardt, K.; Mottaz, H.; Kiss, J.Z.; Dietrich, P.Y.; de Tribolet, N.; Pizzolato, G.P.; et al. Expression and localization of VEGF-C and VEGFR-3 in glioblastomas and haemangioblastomas. J. Pathol 2006, 209, 34–43. [Google Scholar]
  26. Baselga, J.; Arteaga, C.L. Critical update and emerging trends in epidermal growth factor receptor targeting in cancer. J. Clin. Oncol 2005, 23, 2445–2459. [Google Scholar]
  27. Seiwert, T.Y.; Jagadeeswaran, R.; Faoro, L.; Janamanchi, V.; Nallasura, V.; El Dinali, M.; Yala, S.; Kanteti, R.; Cohen, E.E.; Lingen, M.W.; et al. The MET receptor tyrosine kinase is a potential novel therapeutic target for head and neck squamous cell carcinoma. Cancer Res 2009, 69, 3021–3031. [Google Scholar]
  28. Marshall, M.E.; Hinz, T.K.; Kono, S.A.; Singleton, K.R.; Bichon, B.; Ware, K.E.; Marek, L.; Frederick, B.A.; Raben, D.; Heasley, L.E. Fibroblast growth factor receptors are components of autocrine signaling networks in head and neck squamous cell carcinoma cells. Clin. Cancer Res 2011, 17, 5016–5025. [Google Scholar]
  29. Lin, W.; Kao, H.W.; Robinson, D.; Kung, H.J.; Wu, C.W.; Chen, H.C. Tyrosine kinases and gastric cancer. Oncogene 2000, 19, 5680–5689. [Google Scholar]
  30. Zhao, Z.S.; Zhou, J.L.; Yao, G.Y.; Ru, G.Q.; Ma, J.; Ruan, J. Correlative studies on bFGF mRNA and MMP-9 mRNA expressions with microvascular density, progression, and prognosis of gastric carcinomas. World J. Gastroenterol 2005, 11, 3227–3233. [Google Scholar]
  31. Barclay, C.; Li, A.W.; Geldenhuys, L.; Baguma-Nibasheka, M.; Porter, G.A.; Veugelers, P.J.; Murphy, P.R.; Casson, A.G. Basic fibroblast growth factor (FGF-2) overexpression is a risk factor for esophageal cancer recurrence and reduced survival, which is ameliorated by coexpression of the FGF-2 antisense gene. Clin Cancer Res 2005, 11, 7683–7691. [Google Scholar]
  32. Spratlin, J.L.; Cohen, R.B.; Eadens, M.; Gore, L.; Camidge, D.R.; Diab, S.; Leong, S.; O’Bryant, C.; Chow, L.Q.; Serkova, N.J.; et al. Phase I pharmacologic and biologic study of ramucirumab (IMC-1121B), a fully human immunoglobulin G1 monoclonal antibody targeting the vascular endothelial growth factor receptor-2. J. Clin. Oncol 2010, 28, 780–787. [Google Scholar]
  33. Gao, J.; Inagaki, Y.; Song, P.; Qu, X.; Kokudo, N.; Tang, W. Targeting c-Met as a promising strategy for the treatment of hepatocellular carcinoma. Pharmacol. Res 2012, 65, 23–30. [Google Scholar]
  34. Golan, T.; Javle, M. Targeting the insulin growth factor pathway in gastrointestinal cancers. Oncology (Williston Park) 2011, 25. [Google Scholar]
  35. Reidy, D.L.; Vakiani, E.; Fakih, M.G.; Saif, M.W.; Hecht, J.R.; Goodman-Davis, N.; Hollywood, E.; Shia, J.; Schwartz, J.; Chandrawansa, K.I. Randomized, phase II study of the insulin-like growth factor-1 receptor inhibitor IMC-A12, with or without cetuximab, in patients with cetuximab- or panitumumab-refractory metastatic colorectal cancer. J. Clin. Oncol 2010, 28, 4240–4246. [Google Scholar]
  36. Essapen, S.; Thomas, H.; Green, M.; de Vries, C.; Cook, M.G.; Marks, C.; Topham, C.; Modjtahedi, H. The expression and prognostic significance of HER-2 in colorectal cancer and its relationship with clinicopathological parameters. Int. J. Oncol 2004, 24, 241–248. [Google Scholar]
  37. Kammula, U.S.; Kuntz, E.J.; Francone, T.D.; Zeng, Z.; Shia, J.; Landmann, R.G.; Paty, P.B.; Weiser, M.R. Molecular co-expression of the c-Met oncogene and hepatocyte growth factor in primary colon cancer predicts tumor stage and clinical outcome. Cancer Lett 2007, 248, 219–228. [Google Scholar]
  38. Kos, M.; Dabrowski, A. Tumour’s angiogenesis—The function of VEGF and bFGF in colorectal cancer. Ann. Univ. Mariae Curie Sklodowska Med 2002, 57, 556–561. [Google Scholar]
  39. Volm, M.; Koomagi, R.; Mattern, J.; Stammler, G. Prognostic value of basic fibroblast growth factor and its receptor (FGFR-1) in patients with non-small cell lung carcinomas. Eur. J. Cancer 1997, 33, 691–693. [Google Scholar]
  40. Katoh, M. Cancer genomics and genetics of FGFR2 (review). Int. J. Oncol 2008, 33, 233–237. [Google Scholar]
  41. Rusch, V.; Baselga, J.; Cordon-Cardo, C.; Orazem, J.; Zaman, M.; Hoda, S.; McIntosh, J.; Kurie, J.; Dmitrovsky, E. Differential expression of the epidermal growth factor receptor and its ligands in primary non-small cell lung cancers and adjacent benign lung. Cancer Res 1993, 53, 2379–2385. [Google Scholar]
  42. Horstmann, M.; Merseburger, A.S.; von der Heyde, E.; Serth, J.; Wegener, G.; Mengel, M.; Feil, G.; Hennenlotter, J.; Nagele, U.; Anastasiadis, A.; et al. Correlation of bFGF expression in renal cell cancer with clinical and histopathological features by tissue microarray analysis and measurement of serum levels. J. Cancer Res. Clin. Oncol 2005, 131, 715–722. [Google Scholar]
  43. Banumathy, G.; Cairns, P. Signaling pathways in renal cell carcinoma. Cancer Biol. Ther 2010, 10, 658–664. [Google Scholar]
  44. Yuen, J.S.; Cockman, M.E.; Sullivan, M.; Protheroe, A.; Turner, G.D.; Roberts, I.S.; Pugh, C.W.; Werner, H.; Macaulay, V.M. The VHL tumor suppressor inhibits expression of the IGF1R and its loss induces IGF1R upregulation in human clear cell renal carcinoma. Oncogene 2007, 26, 6499–6508. [Google Scholar]
  45. Kojima, S.; Inahara, M.; Suzuki, H.; Ichikawa, T.; Furuya, Y. Implications of insulin-like growth factor-I for prostate cancer therapies. Int. J. Urol 2009, 16, 161–167. [Google Scholar]
  46. Neto, A.S.; Tobias-Machado, M.; Wroclawski, M.L.; Fonseca, F.L.; Pompeo, A.C.; del Giglio, A. Molecular oncogenesis of prostate adenocarcinoma: Role of the human epidermal growth factor receptor 2 (HER-2/neu). Tumori 2010, 96, 645–649. [Google Scholar]
  47. Kwabi-Addo, B.; Ozen, M.; Ittmann, M. The role of fibroblast growth factors and their receptors in prostate cancer. Endocr. Relat. Cancer 2004, 11, 709–724. [Google Scholar]
  48. Gravdal, K.; Halvorsen, O.J.; Haukaas, S.A.; Akslen, L.A. Expression of bFGF/FGFR-1 and vascular proliferation related to clinicopathologic features and tumor progress in localized prostate cancer. Virchows Arch 2006, 448, 68–74. [Google Scholar]
  49. Knowles, M.A. Novel therapeutic targets in bladder cancer: Mutation and expression of FGF receptors. Future Oncol 2008, 4, 71–83. [Google Scholar]
  50. Black, P.C.; Dinney, C.P. Growth factors and receptors as prognostic markers in urothelial carcinoma. Curr. Urol. Rep 2008, 9, 55–61. [Google Scholar]
  51. Van Dam, P.A.; Vergote, I.B.; Lowe, D.G.; Watson, J.V.; van Damme, P.; van der Auwera, J.C.; Shepherd, J.H. Expression of c-erbB-2, c-myc, and c-ras oncoproteins, insulin-like growth factor receptor I, and epidermal growth factor receptor in ovarian carcinoma. J. Clin. Pathol 1994, 47, 914–919. [Google Scholar]
  52. Wang, S.C.; Hung, M.C. HER2 overexpression and cancer targeting. Semin. Oncol 2001, 28, 115–124. [Google Scholar]
  53. Jin, Q.; Esteva, F.J. Cross-talk between the ErbB/HER family and the type I insulin-like growth factor receptor signaling pathway in breast cancer. J. Mammary Gland Biol. Neoplasia 2008, 13, 485–498. [Google Scholar]
  54. Penault-Llorca, F.; Bertucci, F.; Adelaide, J.; Parc, P.; Coulier, F.; Jacquemier, J.; Birnbaum, D.; deLapeyriere, O. Expression of FGF and FGF receptor genes in human breast cancer. Int. J. Cancer 1995, 61, 170–176. [Google Scholar]
  55. Marsh, S.K.; Bansal, G.S.; Zammit, C.; Barnard, R.; Coope, R.; Roberts-Clarke, D.; Gomm, J.J.; Coombes, R.C.; Johnston, C.L. Increased expression of fibroblast growth factor 8 in human breast cancer. Oncogene 1999, 18, 1053–1060. [Google Scholar]
  56. Easty, D.J.; Gray, S.G.; O’Byrne, K.J.; O’Donnell, D.; Bennett, D.C. Receptor tyrosine kinases and their activation in melanoma. Pigm. Cell Melanoma Res 2011, 24, 446–461. [Google Scholar]
  57. Kornmann, M.; Beger, H.G.; Korc, M. Role of fibroblast growth factors and their receptors in pancreatic cancer and chronic pancreatitis. Pancreas 1998, 17, 169–175. [Google Scholar]
  58. Scotlandi, K.; Picci, P. Targeting insulin-like growth factor 1 receptor in sarcomas. Curr. Opin. Oncol 2008, 20, 419–427. [Google Scholar]
  59. Hughes, D.P.; Thomas, D.G.; Giordano, T.J.; Baker, L.H.; McDonagh, K.T. Cell surface expression of epidermal growth factor receptor and Her-2 with nuclear expression of Her-4 in primary osteosarcoma. Cancer Res 2004, 64, 2047–2053. [Google Scholar]
  60. Cottoni, F.; Ceccarelli, S.; Masala, M.V.; Montesu, M.A.; Satta, R.; Pirodda, C.; Rotolo, S.; Frati, L.; Marchese, C.; Angeloni, A. Overexpression of the fibroblast growth factor receptor 2-IIIc in Kaposi’s sarcoma. J. Dermatol. Sci 2009, 53, 65–68. [Google Scholar]
  61. Taylor, J.G.T.; Cheuk, A.T.; Tsang, P.S.; Chung, J.Y.; Song, Y.K.; Desai, K.; Yu, Y.; Chen, Q.R.; Shah, K.; Youngblood, V.; et al. Identification of FGFR4-activating mutations in human rhabdomyosarcomas that promote metastasis in xenotransplanted models. J. Clin. Invest 2009, 119, 3395–3407. [Google Scholar]
  62. Ribatti, D.; Vacca, A.; Rusnati, M.; Presta, M. The discovery of basic fibroblast growth factor/fibroblast growth factor-2 and its role in haematological malignancies. Cytokine Growth Factor Rev 2007, 18, 327–334. [Google Scholar]
  63. Chesi, M.; Bergsagel, P.L.; Kuehl, W.M. The enigma of ectopic expression of FGFR3 in multiple myeloma: A critical initiating event or just a target for mutational activation during tumor progression. Curr. Opin. Hematol 2002, 9, 288–293. [Google Scholar]
  64. Jose, S.; Gotway, G.; Garcia, R.; Vaziri, A.; Tirado, C.A. 8p11–12 FGFR1 rearrangements in hematological malignancies: Review of the literature. J. Assoc. Genet. Technol 2010, 36, 203–208. [Google Scholar]
  65. Ronchetti, D.; Greco, A.; Compasso, S.; Colombo, G.; Dell’Era, P.; Otsuki, T.; Lombardi, L.; Neri, A. Deregulated FGFR3 mutants in multiple myeloma cell lines with t(4;14): Comparative analysis of Y373C, K650E and the novel G384D mutations. Oncogene 2001, 20, 3553–3562. [Google Scholar]
  66. Buhring, H.J.; Sures, I.; Jallal, B.; Weiss, F.U.; Busch, F.W.; Ludwig, W.D.; Handgretinger, R.; Waller, H.D.; Ullrich, A. The receptor tyrosine kinase p185HER2 is expressed on a subset of B-lymphoid blasts from patients with acute lymphoblastic leukemia and chronic myelogenous leukemia. Blood 1995, 86, 1916–1923. [Google Scholar]
  67. Hynes, N.E.; MacDonald, G. ErbB receptors and signaling pathways in cancer. Curr. Opin. Cell Biol 2009, 21, 177–184. [Google Scholar]
  68. Voldborg, B.R.; Damstrup, L.; Spang-Thomsen, M.; Poulsen, H.S. Epidermal growth factor receptor (EGFR) and EGFR mutations, function and possible role in clinical trials. Ann. Oncol 1997, 8, 1197–1206. [Google Scholar]
  69. Hynes, N.E.; Stern, D.F. The biology of erbB-2/neu/HER-2 and its role in cancer. Biochim. Biophys. Acta 1994, 1198, 165–184. [Google Scholar]
  70. Baselga, J.; Perez, E.A.; Pienkowski, T.; Bell, R. Adjuvant trastuzumab: A milestone in the treatment of HER-2-positive early breast cancer. Oncologist 2006, 11, S4–S12. [Google Scholar]
  71. Mendelsohn, J.; Baselga, J. Epidermal growth factor receptor targeting in cancer. Semin. Oncol 2006, 33, 369–385. [Google Scholar]
  72. Karamouzis, M.V.; Konstantinopoulos, P.A.; Papavassiliou, A.G. Targeting MET as a strategy to overcome crosstalk-related resistance to EGFR inhibitors. Lancet Oncol 2009, 10, 709–717. [Google Scholar]
  73. Ding, S.; Merkulova-Rainon, T.; Han, Z.C.; Tobelem, G. HGF receptor up-regulation contributes to the angiogenic phenotype of human endothelial cells and promotes angiogenesis in vitro. Blood 2003, 101, 4816–4822. [Google Scholar]
  74. Michieli, P.; Mazzone, M.; Basilico, C.; Cavassa, S.; Sottile, A.; Naldini, L.; Comoglio, P.M. Targeting the tumor and its microenvironment by a dual-function decoy Met receptor. Cancer Cell 2004, 6, 61–73. [Google Scholar]
  75. Birchmeier, C.; Birchmeier, W.; Gherardi, E.; van de Woude, G.F. Met, metastasis, motility and more. Nat. Rev. Mol. Cell Biol 2003, 4, 915–925. [Google Scholar]
  76. Ido, A.; Tsubouchi, H. Translational research to identify clinical applications of hepatocyte growth factor. Hepatol. Res 2009, 39, 739–747. [Google Scholar]
  77. Ponzo, M.G.; Lesurf, R.; Petkiewicz, S.; O’Malley, F.P.; Pinnaduwage, D.; Andrulis, I.L.; Bull, S.B.; Chughtai, N.; Zuo, D.; Souleimanova, M.; et al. Met induces mammary tumors with diverse histologies and is associated with poor outcome and human basal breast cancer. Proc. Natl. Acad. Sci. USA 2009, 106, 12903–12908. [Google Scholar]
  78. Pacher, M.; Seewald, M.J.; Mikula, M.; Oehler, S.; Mogg, M.; Vinatzer, U.; Eger, A.; Schweifer, N.; Varecka, R.; Sommergruber, W.; et al. Impact of constitutive IGF1/IGF2 stimulation on the transcriptional program of human breast cancer cells. Carcinogenesis 2007, 28, 49–59. [Google Scholar]
  79. Pollak, M.N.; Schernhammer, E.S.; Hankinson, S.E. Insulin-like growth factors and neoplasia. Nat. Rev. Cancer 2004, 4, 505–518. [Google Scholar]
  80. Yee, D. The insulin-like growth factor system as a treatment target in breast cancer. Semin. Oncol 2002, 29, 86–95. [Google Scholar]
  81. Roberts, C.T., Jr. Control of insulin-like growth factor (IGF) action by regulation of IGF-I receptor expression. Endocr. J 1996, 43, S49–S55. [Google Scholar]
  82. Lu, Y.; Zi, X.; Zhao, Y.; Mascarenhas, D.; Pollak, M. Insulin-like growth factor-I receptor signaling and resistance to trastuzumab (Herceptin). J. Natl. Cancer Inst 2001, 93, 1852–1857. [Google Scholar]
  83. Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med 1995, 1, 27–31. [Google Scholar]
  84. Folkman, J. Tumor angiogenesis. Adv. Cancer Res 1985, 43, 175–203. [Google Scholar]
  85. Distler, J.H.; Hirth, A.; Kurowska-Stolarska, M.; Gay, R.E.; Gay, S.; Distler, O. Angiogenic and angiostatic factors in the molecular control of angiogenesis. Q. J. Nucl. Med 2003, 47, 149–161. [Google Scholar]
  86. Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med 2003, 9, 669–676. [Google Scholar]
  87. Ortega, N.; Hutchings, H.; Plouet, J. Signal relays in the VEGF system. Front. Biosci 1999, 4, D141–D152. [Google Scholar]
  88. Roy, H.; Bhardwaj, S.; Yla-Herttuala, S. Biology of vascular endothelial growth factors. FEBS Lett 2006, 580, 2879–2887. [Google Scholar]
  89. Otrock, Z.K.; Makarem, J.A.; Shamseddine, A.I. Vascular endothelial growth factor family of ligands and receptors: Review. Blood Cells Mol. Dis 2007, 38, 258–268. [Google Scholar]
  90. Holmes, K.; Roberts, O.L.; Thomas, A.M.; Cross, M.J. Vascular endothelial growth factor receptor-2: Structure, function, intracellular signalling and therapeutic inhibition. Cell Signal 2007, 19, 2003–2012. [Google Scholar]
  91. Jain, R.K. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science 2005, 307, 58–62. [Google Scholar]
  92. Tortora, G.; Melisi, D.; Ciardiello, F. Angiogenesis: A target for cancer therapy. Curr. Pharm. Des 2004, 10, 11–26. [Google Scholar]
  93. Hurwitz, H. Integrating the anti-VEGF-A humanized monoclonal antibody bevacizumab with chemotherapy in advanced colorectal cancer. Clin. Colorectal Cancer 2004, 4, S62–S68. [Google Scholar]
  94. Lu, D.; Shen, J.; Vil, M.D.; Zhang, H.; Jimenez, X.; Bohlen, P.; Witte, L.; Zhu, Z. Tailoring in vitro selection for a picomolar affinity human antibody directed against vascular endothelial growth factor receptor 2 for enhanced neutralizing activity. J. Biol. Chem 2003, 278, 43496–43507. [Google Scholar]
  95. Cooke, S.P.; Boxer, G.M.; Lawrence, L.; Pedley, R.B.; Spencer, D.I.; Begent, R.H.; Chester, K.A. A strategy for antitumor vascular therapy by targeting the vascular endothelial growth factor: Receptor complex. Cancer Res 2001, 61, 3653–3659. [Google Scholar]
  96. Vitaliti, A.; Wittmer, M.; Steiner, R.; Wyder, L.; Neri, D.; Klemenz, R. Inhibition of tumor angiogenesis by a single-chain antibody directed against vascular endothelial growth factor. Cancer Res 2000, 60, 4311–4314. [Google Scholar]
  97. Ornitz, D.M.; Itoh, N. Fibroblast growth factors. Genome Biol 2001, 2, reviews3005:1–reviews3005:12. [Google Scholar]
  98. Basilico, C.; Moscatelli, D. The FGF family of growth factors and oncogenes. Adv. Cancer Res 1992, 59, 115–165. [Google Scholar]
  99. Folkman, J.; Shing, Y. Control of angiogenesis by heparin and other sulfated polysaccharides. Adv. Exp. Med. Biol 1992, 313, 355–364. [Google Scholar]
  100. Presta, M.; Moscatelli, D.; Joseph-Silverstein, J.; Rifkin, D.B. Purification from a human hepatoma cell line of a basic fibroblast growth factor-like molecule that stimulates capillary endothelial cell plasminogen activator production, DNA synthesis, and migration. Mol. Cell. Biol 1986, 6, 4060–4066. [Google Scholar]
  101. Javerzat, S.; Auguste, P.; Bikfalvi, A. The role of fibroblast growth factors in vascular development. Trends Mol. Med 2002, 8, 483–489. [Google Scholar]
  102. Powers, C.J.; McLeskey, S.W.; Wellstein, A. Fibroblast growth factors, their receptors and signaling. Endocr. Relat. Cancer 2000, 7, 165–197. [Google Scholar]
  103. Johnson, D.E.; Williams, L.T. Structural and functional diversity in the FGF receptor multigene family. Adv. Cancer Res 1993, 60, 1–41. [Google Scholar]
  104. Giri, D.; Ropiquet, F.; Ittmann, M. Alterations in expression of basic fibroblast growth factor (FGF) 2 and its receptor FGFR-1 in human prostate cancer. Clin. Cancer Res 1999, 5, 1063–1071. [Google Scholar]
  105. Antoine, M.; Wirz, W.; Tag, C.G.; Mavituna, M.; Emans, N.; Korff, T.; Stoldt, V.; Gressner, A.M.; Kiefer, P. Expression pattern of fibroblast growth factors (FGFs), their receptors and antagonists in primary endothelial cells and vascular smooth muscle cells. Growth Factors 2005, 23, 87–95. [Google Scholar]
  106. Ronca, R.; Benzoni, P.; Leali, D.; Urbinati, C.; Belleri, M.; Corsini, M.; Alessi, P.; Coltrini, D.; Calza, S.; Presta, M.; et al. Antiangiogenic activity of a neutralizing human single-chain antibody fragment against fibroblast growth factor receptor 1. Mol. Cancer Ther 2010, 9, 3244–3253. [Google Scholar]
  107. Yayon, A.; Aviezer, D.; Safran, M.; Gross, J.L.; Heldman, Y.; Cabilly, S.; Givol, D.; Katchalski-Katzir, E. Isolation of peptides that inhibit binding of basic fibroblast growth factor to its receptor from a random phage-epitope library. Proc. Natl. Acad. Sci. USA 1993, 90, 10643–10647. [Google Scholar]
  108. Cardo-Vila, M.; Giordano, R.J.; Sidman, R.L.; Bronk, L.F.; Fan, Z.; Mendelsohn, J.; Arap, W.; Pasqualini, R. From combinatorial peptide selection to drug prototype (II): Targeting the epidermal growth factor receptor pathway. Proc. Natl. Acad. Sci. USA 2010, 107, 5118–5123. [Google Scholar]
  109. Kumar, S.R.; Quinn, T.P.; Deutscher, S.L. Evaluation of an 111In-radiolabeled peptide as a targeting and imaging agent for ErbB-2 receptor expressing breast carcinomas. Clin. Cancer Res 2007, 13, 6070–6079. [Google Scholar]
  110. Hetian, L.; Ping, A.; Shumei, S.; Xiaoying, L.; Luowen, H.; Jian, W.; Lin, M.; Meisheng, L.; Junshan, Y.; Chengchao, S. A novel peptide isolated from a phage display library inhibits tumor growth and metastasis by blocking the binding of vascular endothelial growth factor to its kinase domain receptor. J. Biol. Chem 2002, 277, 43137–43142. [Google Scholar]
  111. Rastelli, L.; Valentino, M.L.; Minderman, M.C.; Landin, J.; Malyankar, U.M.; Lescoe, M.K.; Kitson, R.; Brunson, K.; Souan, L.; Forenza, S.; et al. A KDR-binding peptide (ST100,059) can block angiogenesis, melanoma tumor growth and metastasis in vitro and in vivo. Int. J. Oncol 2011, 39, 401–408. [Google Scholar]
  112. Fairbrother, W.J.; Christinger, H.W.; Cochran, A.G.; Fuh, G.; Keenan, C.J.; Quan, C.; Shriver, S.K.; Tom, J.Y.; Wells, J.A.; Cunningham, B.C. Novel peptides selected to bind vascular endothelial growth factor target the receptor-binding site. Biochemistry 1998, 37, 17754–17764. [Google Scholar]
  113. Erdag, B.; Balcioglu, K.B.; Kumbasar, A.; Celikbicak, O.; Zeder-Lutz, G.; Altschuh, D.; Salih, B.; Baysal, K. Novel short peptides isolated from phage display library inhibit vascular endothelial growth factor activity. Mol. Biotechnol 2007, 35, 51–63. [Google Scholar]
  114. Binetruy-Tournaire, R.; Demangel, C.; Malavaud, B.; Vassy, R.; Rouyre, S.; Kraemer, M.; Plouet, J.; Derbin, C.; Perret, G.; et al. Identification of a peptide blocking vascular endothelial growth factor (VEGF)-mediated angiogenesis. EMBO J 2000, 19, 1525–1533. [Google Scholar]
  115. Nilsson, F.; Tarli, L.; Viti, F.; Neri, D. The use of phage display for the development of tumour targeting agents. Adv. Drug Deliv. Rev 2000, 43, 165–196. [Google Scholar]
  116. Yoo, M.K.; Kang, S.K.; Choi, J.H.; Park, I.K.; Na, H.S.; Lee, H.C.; Kim, E.B.; Lee, N.K.; Nah, J.W.; Choi, Y.J.; et al. Targeted delivery of chitosan nanoparticles to Peyer’s patch using M cell-homing peptide selected by phage display technique. Biomaterials 2010, 31, 7738–7747. [Google Scholar]
  117. Laakkonen, P.; Akerman, M.E.; Biliran, H.; Yang, M.; Ferrer, F.; Karpanen, T.; Hoffman, R.M.; Ruoslahti, E. Antitumor activity of a homing peptide that targets tumor lymphatics and tumor cells. Proc. Natl. Acad. Sci. USA 2004, 101, 9381–9386. [Google Scholar]
  118. Zhu, Z.; Rockwell, P.; Lu, D.; Kotanides, H.; Pytowski, B.; Hicklin, D.J.; Bohlen, P.; Witte, L. Inhibition of vascular endothelial growth factor-induced receptor activation with anti-kinase insert domain-containing receptor single-chain antibodies from a phage display library. Cancer Res 1998, 58, 3209–3214. [Google Scholar]
  119. Erdag, B.; Balcioglu, B.K.; Bahadir, A.O.; Serhatli, M.; Kacar, O.; Bahar, A.; Seker, U.O.; Akgun, E.; Ozkan, A.; Kilic, T.; et al. Identification of novel neutralizing single-chain antibodies against vascular endothelial growth factor receptor 2. Biotechnol. Appl. Biochem 2011, 58, 412–422. [Google Scholar]
  120. Veggiani, G.; Ossolengo, G.; Aliprandi, M.; Cavallaro, U.; de Marco, A. Single-domain antibodies that compete with the natural ligand fibroblast growth factor block the internalization of the fibroblast growth factor receptor 1. Biochem. Biophys. Res. Commun 2011, 408, 692–696. [Google Scholar]
  121. Dong, J.; Sereno, A.; Aivazian, D.; Langley, E.; Miller, B.R.; Snyder, W.B.; Chan, E.; Cantele, M.; Morena, R.; Joseph, I.B.; et al. A stable IgG-like bispecific antibody targeting the epidermal growth factor receptor and the type I insulin-like growth factor receptor demonstrates superior anti-tumor activity. MAbs 2011, 3, 273–288. [Google Scholar]
  122. Lu, R.M.; Chang, Y.L.; Chen, M.S.; Wu, H.C. Single chain anti-c-Met antibody conjugated nanoparticles for in vivo tumor-targeted imaging and drug delivery. Biomaterials 2011, 32, 3265–3274. [Google Scholar]
  123. Lee, J.W.; Stone, R.L.; Lee, S.J.; Nam, E.J.; Roh, J.W.; Nick, A.M.; Han, H.D.; Shahzad, M.M.; Kim, H.S.; Mangala, L.S.; et al. EphA2 targeted chemotherapy using an antibody drug conjugate in endometrial carcinoma. Clin. Cancer Res 2010, 16, 2562–2570. [Google Scholar]
  124. LoRusso, P.M.; Weiss, D.; Guardino, E.; Girish, S.; Sliwkowski, M.X. Trastuzumab emtansine: A unique antibody-drug conjugate in development for human epidermal growth factor receptor 2-positive cancer. Clin. Cancer Res 2011, 17, 6437–6447. [Google Scholar]
  125. Moldenhauer, G.; Salnikov, A.V.; Luttgau, S.; Herr, I.; Anderl, J.; Faulstich, H. Therapeutic Potential of Amanitin-Conjugated Anti-Epithelial Cell Adhesion Molecule Monoclonal Antibody Against Pancreatic Carcinoma. J. Natl. Cancer Inst 2012. [Google Scholar] [CrossRef]
  126. Green, L.L. Antibody engineering via genetic engineering of the mouse: XenoMouse strains are a vehicle for the facile generation of therapeutic human monoclonal antibodies. J. Immunol. Methods 1999, 231, 11–23. [Google Scholar]
  127. Lonberg, N. Fully human antibodies from transgenic mouse and phage display platforms. Curr. Opin. Immunol 2008, 20, 450–459. [Google Scholar]
  128. Louis, E.; Lofberg, R.; Reinisch, W.; Camez, A.; Yang, M.; Pollack, P.F.; Chen, N.; Chao, J.; Mulani, P.M. Adalimumab improves patient-reported outcomes and reduces indirect costs in patients with moderate to severe Crohn;s disease: Results from the CARE trial. J. Crohns Colitis 2012. [Google Scholar] [CrossRef]
  129. Yang, H.; Epstein, D.; Bojke, L.; Craig, D.; Light, K.; Bruce, I.; Sculpher, M.; Woolacott, N. Golimumab for the treatment of psoriatic arthritis. Health Technol. Assess 2011, 15, S87–S95. [Google Scholar]
  130. Ballestrero, A.; Garuti, A.; Cirmena, G.; Rocco, I.; Palermo, C.; Nencioni, A.; Scabini, S.; Zoppoli, G.; Parodi, S.; Patrone, F. Patient-tailored treatments with anti-EGFR monoclonal antibodies in advanced colorectal cancer: KRAS and beyond. Curr. Cancer Drug Targets 2012, 12, 316–328. [Google Scholar]
  131. Curran, M.P. Canakinumab: In patients with cryopyrin-associated periodic syndromes. BioDrugs 2012, 26, 53–59. [Google Scholar]
  132. Winchester, D.; Callis Duffin, K.; Hansen, C. Response to ustekinumab in a patient with both severe psoriasis and hypertrophic discoid lupus. Lupus 2012. [Google Scholar] [CrossRef]
  133. Hoyle, M.; Crathorne, L.; Garside, R.; Hyde, C. Ofatumumab for the treatment of chronic lymphocytic leukaemia in patients who are refractory to fludarabine and alemtuzumab: A critique of the submission from GSK. Health Technol. Assess 2011, 15, S61–S67. [Google Scholar]
  134. Waugh, N.; Royle, P.; Scotland, G.; Henderson, R.; Hollick, R.; McNamee, P. Denosumab for the prevention of osteoporotic fractures in postmenopausal women. Health Technol. Assess 2011, 15, S51–S59. [Google Scholar]
  135. Boyce, E.G.; Fusco, B.E. Belimumab: Review of use in systemic lupus erythematosus. Clin. Ther 2012. [Google Scholar] [CrossRef]
  136. Margolin, K.; Ernstoff, M.S.; Hamid, O.; Lawrence, D.; McDermott, D.; Puzanov, I.; Wolchok, J.D.; Clark, J.I.; Sznol, M.; Logan, T.F.; et al. Ipilimumab in patients with melanoma and brain metastases: An open-label, phase 2 trial. Lancet Oncol 2012, ((12)), 70090–6. [Google Scholar] [CrossRef]
  137. Cameron, F.; Whiteside, G.; Perry, C. Ipilimumab: First global approval. Drugs 2011, 71, 1093–1104. [Google Scholar]
  138. Migone, T.S.; Subramanian, G.M.; Zhong, J.; Healey, L.M.; Corey, A.; Devalaraja, M.; Lo, L.; Ullrich, S.; Zimmerman, J.; Chen, A.; et al. Raxibacumab for the treatment of inhalational anthrax. N. Engl. J. Med 2009, 361, 135–144. [Google Scholar]
  139. Thie, H.; Meyer, T.; Schirrmann, T.; Hust, M.; Dubel, S. Phage display derived therapeutic antibodies. Curr. Pharm. Biotechnol 2008, 9, 439–446. [Google Scholar]
  140. Rowinsky, E.K.; Youssoufian, H.; Tonra, J.R.; Solomon, P.; Burtrum, D.; Ludwig, D.L. IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor. Clin. Cancer Res 2007, 13, 5549s–5555s. [Google Scholar]
  141. Erinjeri, J.P.; Deodhar, A.; Thornton, R.H.; Allen, P.J.; Getrajdman, G.I.; Brown, K.T.; Sofocleous, C.T.; Reidy, D.L. Resolution of hepatic encephalopathy following hepatic artery embolization in a patient with well-differentiated neuroendocrine tumor metastatic to the liver. Cardiovasc. Intervent. Radiol 2010, 33, 610–614. [Google Scholar]
  142. Beltran, P.J.; Mitchell, P.; Chung, Y.A.; Cajulis, E.; Lu, J.; Belmontes, B.; Ho, J.; Tsai, M.M.; Zhu, M.; Vonderfecht, S.; et al. AMG 479, a fully human anti-insulin-like growth factor receptor type I monoclonal antibody, inhibits the growth and survival of pancreatic carcinoma cells. Mol. Cancer Ther 2009, 8, 1095–1105. [Google Scholar]
  143. Tolcher, A.W.; Sarantopoulos, J.; Patnaik, A.; Papadopoulos, K.; Lin, C.C.; Rodon, J.; Murphy, B.; Roth, B.; McCaffery, I.; Gorski, K.S.; et al. Phase I, pharmacokinetic, and pharmacodynamic study of AMG 479, a fully human monoclonal antibody to insulin-like growth factor receptor 1. J. Clin. Oncol 2009, 27, 5800–5807. [Google Scholar]
  144. Beltran, P.J.; Chung, Y.A.; Moody, G.; Mitchell, P.; Cajulis, E.; Vonderfecht, S.; Kendall, R.; Radinsky, R.; Calzone, F.J. Efficacy of ganitumab (AMG 479), alone and in combination with rapamycin, in Ewing’s and osteogenic sarcoma models. J. Pharmacol. Exp. Ther 2011, 337, 644–654. [Google Scholar]
  145. Tammela, T.; Zarkada, G.; Wallgard, E.; Murtomaki, A.; Suchting, S.; Wirzenius, M.; Waltari, M.; Hellstrom, M.; Schomber, T.; Peltonen, R.; et al. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature 2008, 454, 656–660. [Google Scholar]
  146. Kuenen, B.; Witteveen, P.O.; Ruijter, R.; Giaccone, G.; Dontabhaktuni, A.; Fox, F.; Katz, T.; Youssoufian, H.; Zhu, J.; Rowinsky, E.K.; et al. A phase I pharmacologic study of necitumumab (IMC-11F8), a fully human IgG1 monoclonal antibody directed against EGFR in patients with advanced solid malignancies. Clin. Cancer Res 2010, 16, 1915–1923. [Google Scholar]
  147. Lu, D.; Jimenez, X.; Zhang, H.; Bohlen, P.; Witte, L.; Zhu, Z. Selection of high affinity human neutralizing antibodies to VEGFR2 from a large antibody phage display library for antiangiogenesis therapy. Int. J. Cancer 2002, 97, 393–399. [Google Scholar]
  148. Karkkainen, M.J.; Jussila, L.; Ferrell, R.E.; Finegold, D.N.; Alitalo, K. Molecular regulation of lymphangiogenesis and targets for tissue oedema. Trends Mol. Med 2001, 7, 18–22. [Google Scholar]
  149. Achen, M.G.; Stacker, S.A. Molecular control of lymphatic metastasis. Ann. NY Acad. Sci 2008, 1131, 225–234. [Google Scholar]
  150. Grau, S.J.; Trillsch, F.; Herms, J.; Thon, N.; Nelson, P.J.; Tonn, J.C.; Goldbrunner, R. Expression of VEGFR3 in glioma endothelium correlates with tumor grade. J. Neurooncol 2007, 82, 141–150. [Google Scholar]
  151. Hajrasouliha, A.R.; Funaki, T.; Sadrai, Z.; Hattori, T.; Chauhan, S.K.; Dana, R. Vascular endothelial growth factor-C promotes alloimmunity by amplifying antigen-presenting cell maturation and lymphangiogenesis. Invest. Ophthalmol. Vis. Sci 2012, 53, 1244–1250. [Google Scholar]
  152. Grutter, C.; Wilkinson, T.; Turner, R.; Podichetty, S.; Finch, D.; McCourt, M.; Loning, S.; Jermutus, L.; Grutter, M.G. A cytokine-neutralizing antibody as a structural mimetic of 2 receptor interactions. Proc. Natl. Acad. Sci. USA 2008, 105, 20251–20256. [Google Scholar]
  153. Ronca, R.; Sozzani, S.; Presta, M.; Alessi, P. Delivering cytokines at tumor site: The immunocytokine-conjugated anti-EDB-fibronectin antibody case. Immunobiology 2009, 214, 800–810. [Google Scholar]
  154. Kurtz, J.E.; Dufour, P. Adecatumumab: An anti-EpCAM monoclonal antibody, from the bench to the bedside. Expert Opin. Biol. Ther 2010, 10, 951–958. [Google Scholar]
  155. Casi, G.; Neri, D. Antibody-drug conjugates: Basic concepts, examples and future perspectives. J. Control. Release 2012. [Google Scholar] [CrossRef]
Figure 1. Type of antibody formats, as mentioned in the text, with the relative molecular weight. Adapted from Holliger and Hudson [16].
Figure 1. Type of antibody formats, as mentioned in the text, with the relative molecular weight. Adapted from Holliger and Hudson [16].
Ijms 13 05254f1
Figure 2. Schematic example of some tyrosine kinase growth factor receptors. Adapted from Lemmon and Schlessinger [17].
Figure 2. Schematic example of some tyrosine kinase growth factor receptors. Adapted from Lemmon and Schlessinger [17].
Ijms 13 05254f2
Table 1. Growth factor and RTK altered expression in different type of tumors.
Table 1. Growth factor and RTK altered expression in different type of tumors.
Tumor typeGrowth factorRTKReferences
Glioma/GlioblastomaFGF2, VEGFCEGFR[2225]
Head-neck squamous cellMet, FGFR1, FGFR3, EGFR[2628]
Esophageal and GastricFGF2Met[2931]
Liver/HepatocarcinomaMet, VEGR2[32,33]
ColorectalFGF2, VEGFAMet, ErbB2, IGF1R[3438]
LungFGF2FGFR1, FGFR2, EGFR[3941]
RenalFGF2Met, IGF1R, VEGFR2[32,4244]
ProstateFGF2, FGF8FGFR1, FGFR3, ErbB2, IGF1R[4548]
BladderFGFR3, ErbB2[49,50]
OvarianFGFR2, ErbB2, IGF1R[40,51,52]
BreastFGF8FGFR1, FGFR2, EGFR, ErbB2, IGF1R[40,5255]
MelanomaFGFR1, FGFR2, VEGFR2[32,56]
PancreaticFGFR1[57]
SarcomasFGFR2, FGFR4, EGFR, IGF1R[5861]
Hematological malignanciesFGF2Met, ErbB2, FGFR1, FGFR3[52,6266]
Table 2. Brief summary of phage derivatives in clinical evaluation.
Table 2. Brief summary of phage derivatives in clinical evaluation.
MoleculeCompanyCommercial nameTargetRef
IMC-A12ImClone LLCCixutumumabIGF1R[140,141]
AMG-479AmgenGanitumabIGF1R[142144]
IMC-11F8ImClone LLCNecitumumabEGFR[139,145,146]
IMC-1121bImClone LLCRamucirumabVEGFR2[32,147]
IMC-3C5ImClone LLC-VEGFR3[145,148150]
VGX-100Circadian Technologies LtdFresolimumabVEGFC[151]

Share and Cite

MDPI and ACS Style

Ronca, R.; Benzoni, P.; De Luca, A.; Crescini, E.; Dell’Era, P. Phage Displayed Peptides/Antibodies Recognizing Growth Factors and Their Tyrosine Kinase Receptors as Tools for Anti-Cancer Therapeutics. Int. J. Mol. Sci. 2012, 13, 5254-5277. https://doi.org/10.3390/ijms13045254

AMA Style

Ronca R, Benzoni P, De Luca A, Crescini E, Dell’Era P. Phage Displayed Peptides/Antibodies Recognizing Growth Factors and Their Tyrosine Kinase Receptors as Tools for Anti-Cancer Therapeutics. International Journal of Molecular Sciences. 2012; 13(4):5254-5277. https://doi.org/10.3390/ijms13045254

Chicago/Turabian Style

Ronca, Roberto, Patrizia Benzoni, Angela De Luca, Elisabetta Crescini, and Patrizia Dell’Era. 2012. "Phage Displayed Peptides/Antibodies Recognizing Growth Factors and Their Tyrosine Kinase Receptors as Tools for Anti-Cancer Therapeutics" International Journal of Molecular Sciences 13, no. 4: 5254-5277. https://doi.org/10.3390/ijms13045254

Article Metrics

Back to TopTop