Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Mar 22.
Published in final edited form as: Trends Biotechnol. 2015 Mar 26;33(5):292–301. doi: 10.1016/j.tibtech.2015.02.008

Selection strategies for anti-cancer antibody discovery: searching off the beaten path

David Sánchez-Martín 1,*, Morten Dræby Sørensen 2,3,*, Simon Lykkemark 4, Laura Sanz 5, Peter Kristensen 6, Erkki Ruoslahti 7,8, Luis Álvarez-Vallina 5,6
PMCID: PMC5362106  NIHMSID: NIHMS852089  PMID: 25819764

Abstract

Antibody based drugs represent one of the most successful and promising therapeutic approaches in oncology. Large combinatorial phage antibody libraries are available for identification of therapeutic antibodies, and a variety of technologies exist for their further conversion into multivalent and multispecific formats optimized for the desired pharmacokinetics and the pathological context. However, there is no technology for antigen profiling of intact tumors to identify tumor markers targetable with antibodies. Such constraints have led to a relative paucity of tumor-associated antigens for antibody targeting in oncology. Here we review novel approaches aimed at the identification of antibody-targetable, accessible antigens in intact tumors. We hope that such advanced selection approaches will be useful in the development of next-generation antibody therapeutics for cancer.

Keywords: Therapeutic antibodies, tumor-associated antigens, phage display, advanced phage selections

Monoclonal antibodies in cancer therapy

One of the most promising strategies for the development of cancer therapies relies on monoclonal antibodies (mAbs). Tumor-specific mAbs can be therapeutically active as such, they can be used in targeted delivery of therapeutics to tumors, or they can serve in both capacities. The number of tumor-specific molecules available for antibody targeting is limited, and identification of new targets can expand the utility of the approach [1, 2].

According to the available information [35], there are currently 19 therapeutic mAbs for cancer indications on the market and two more were undergoing their first regulatory review as of January 2015. Seven of the 18 marketed mAbs are used against hematological malignancies, and 12 are for the treatment of solid tumors (Table 1). In the first group, only 4 different CD antigens are targeted (CD19, CD20, CD30 and CCR4), with 4 mAbs directed against anti-CD20. This paucity of targets is also found in mAbs aimed to the treatment of solid tumors. Of the 12 mAbs on the list, 6 are directed against cancer cell surface molecules. Two growth factor receptors (EGFR and HER2) are the targets of 5 mAbs. Other antibody-based therapeutics in cancer do not focus on the tumor itself, but instead aim at inhibiting tumor angiogenesis (with anti-angiogenic mAbs targeting VEGF or VEGFR-2) or promoting immune responses (immunostimulatory mAbs blocking CTLA-4 or PD1) [35]. In summary, all the therapeutic mAbs either approved or in advanced clinical trials are directed against a limited number of targets. Moreover, most of the target antigens are also expressed and accessible in normal tissues.

Table 1.

Therapeutic monoclonal antibodies for cancer indications marketed worldwide

International non-proprietary name Tradename Target/Antibody type Indication First approved First US (EU) [Japan] approval
Hematological malignancies
Rituximab MabThera, Rituxan CD20, Ch IgG1 NHL, CLL 1997 (1998)
Ibritumomab tiuxetan Zevalin CD20, Mu IgG1, 90Y-radiolabeled NHL 2002 (2004)
Ofatumumab Arzerra CD20, Hu IgG1 CLL 2009 (2010)
Obinutuzumab Gazyva CD20, Hz IgG1, glycoengineered CLL 2013 (2014)
Brentuximab vedotin Adcetris CD30, Ch IgG1, ADC HL, systemic ALCL 2011 (2012)
Mogamulizumab Poteligeo CCR4, Hz IgG1, glycoengineered T cell leukemia-lymphoma NA (NA) [2012]
Blinatumomab Blincyto CD19 × CD3 (bispecific), BiTe ALL 2014 (NA)
Solid tumors
Nimotuzumab CIMAher, BIOMAb-EGFR EGFR, Hz IgG1 SCCHN, glioma, nasopharyngeal cancer NA (NA) [NA]#
Cetuximab Erbitux EGFR, Ch IgG1 CRC 2004 (2004)
Panitumumab Vectibix EGFR, Hu IgG2 CRC 2006 (2007)
Trastuzumab Herceptin HER2, Hz IgG1 Breast cancer 1998 (2000)
Trastuzumab emtansine Kadcyla HER2, Hz IgG1, ADC Breast cancer 2013 (2013)
Pertuzumab Perjeta HER2, Hz IgG1 Breast cancer 2012 (2013)
Catumaxomab Removab EPCAM × CD3 (bispecific) Rat/Mouse IgG Malignant ascites NA (2009)
Denosumab Xgeva RANKL, Hu IgG2 Bone metastasis, bone giant cell tumors 2010 (2011)
Ipilimumab Yervoy CTLA-4, Hu IgG1 Melanoma 2011 (2011)
Pembrolizumab Keytruda PD1, Hz IgG4 Melanoma 2014 (In review)
Nivolumab Opdivo PD1, Hu IgG4 NSCLC, renal cell carcinoma, melanoma 2014 (In review) [2014]
Bevacizumab Avastin VEGF, Hz IgG1 CRC 2004 (2005)
Ramucirumab Cyramza VEGFR2, Hu IgG1 Gastric cancer 2014 (In review)

Abbreviations: ADC: antibody-drug conjugate; ALCL, anaplastic large cell lymphoma; ALL, acute lymphoblasticleukemia; BiTe, bispecific T cell engager; CD, cluster of differentiation; Ch, chimeric; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukaemia; CRC, colorectal cancer; CTLA-4, cytotoxic T lymphocyte antigen 4; EGFR, epidermal growth factor receptor; EPCAM, epithelial cell adhesion molecule; HER2, human epidermal growth factor receptor 2; HL, Hodgkin lymphoma; Hu, human; Hz, humanized; Mu, murine; NA, not approved; NHL, non-Hodgkin lymphoma; NSCLC, non-small cell lung cancer; PD1, programmed death 1; RANK-L, receptor activator of NFκb ligand; SCCHN: squamous cell carcinoma in head and neck; VEGF, vascular endothelial growth factor; VEGFR2, vascular endothelial growth factor receptor 2.

#

Argentina, China, Cuba and India, among over 20 countries.

This paucity of target antigens may be a result of current target identification methods, such as searching for differences between tumor and non-tumor cells at the DNA, RNA and protein levels [6]. Availability of biological source material such as cell lines and clinical samples is a prerequisite to target identification. Cell lines are widely used because they provide readily accessible starting material for any common target search strategy. As shown in Table 1, cancer cell membrane proteins are popular targets for antibody selection; however, the extent to which their presence and abundance in established cell lines correspond to those of primary tumors is not clear. In a recent article, a significant portion of the “membranome” (genes encoding plasma membrane proteins) lost their up-regulated state upon transition from in vivo to in vitro conditions [7]. This information has important implications for antibody selection, as one can miss potential targets. On the other hand, fresh tumor biopsy material may be difficult to obtain in sufficient amounts for conventional selection strategies, and although pieces of primary human tumors can be propagated in immunodeficient mice, changes in cell composition of the tumor stroma can impact tumor cell expression pattern [8].

Moreover, genomic and proteomic studies involve a complex sample preparation process that destroys tissue architecture. The loss of “topographical” information is a major handicap in the identification of cancer targets. In fact, tumors are complex organ-like structures containing multiple and highly interactive cell types, which include the cancer cells and a variety of non-cancer cell types (endothelial cells, pericytes, fibroblasts, immune cells, etc.), soluble factors, and extracellular matrix components. The non-cancer cell components can promote neoplastic transformation, support tumor growth and invasion, and protect the tumor from host immunity [9]. Furthermore, current protocols for target identification often ignore the tumor vasculature; which is an important site of accessible and stably targetable tumor-associated markers.

Tumor blood vessels are abnormal both structurally and functionally [9]. This creates a microenvironment characterized by elevated fluid pressure, transient hypoxia and repeated hypoxia-reperfusion injury, all factors thought to contribute to the limited diffusion of therapeutic antibodies into the tumors [10].

For many years, scientists focused their research on strategies that directly target tumor cells, and spent little time studying other components of the tumor microenvironment. However, this view is changing, and recent data show the value of targeting the tumor microenvironment in combination with therapies aimed at the tumor cells [11]. Nevertheless, the information available on antigens associated with tumor stroma that might potentially amenable to antibody targeting is limited. Here we discuss some emergent strategies that are based on antibody display technology and its adaptation to complex selection scenarios. This technology can expand the repertoire of targetable tumor molecules, while simultaneously generating new antibodies for the targeting. It can also be useful in the optimization of the structure of an antibody, and when applicable, its therapeutic payload.

New antibody phage selection strategies in oncology

A number of human antibody libraries with different formats and diversities have been generated [12]. The in vitro selection of antibodies against purified antigens is a relatively straightforward process, generally accomplished within a few rounds of affinity selection [12]. The power of this in vitro antibody selection can be further enhanced by precise control of the selection conditions. In contrast to immunization of animals, where there is little control over the nature of antibodies generated, the selection conditions can be readily manipulated in vitro. This can be accomplished for example, by presentation of specific conformations of the target antigen or by including competitors for unwanted antigens, so as to direct the selection towards targets or epitopes of interest. In vitro selection methods also overcome immunological tolerance, allowing the selection of antibodies against highly conserved targets and self-antigens [12].

However, there are more complex scenarios for antibody selection, which require design of specific selection strategies. Examples include isolation of antibodies against whole cells, where the desired target is only a fraction of the available epitopes present on the cell, and performing selection on heterogeneous cell populations or molecular preparations. Unfortunately, antibody discovery by phage display on whole cancer cells is complicated for a number of reasons, including high background of non-specifically bound phage particles, selection bias toward highly expressed epitopes, and emergence of dominant phage clones, particularly when multiple rounds of selection are used.

According to the information summarized in Table 2, only in a little more than half (29 out of 52 articles) of the selections performed on cancer cells or tissue the antigens recognized by the retrieved antibodies were identified. Most of the identified antigens were highly expressed markers, e.g. EGFR [1315], HER2 [13, 14, 1619], CD44 [13, 14, 20], and transferrin receptor [13, 14, 19, 21], which at that time were already known as cancer markers. In some cases, the phage selection was specifically designed to favor antibodies against a given antigen e.g. by using a cell line expressing high levels of a particular antigen, or by secondary screening of the clones obtained by ELISA against a purified form of the antigen.

Table 2.

Antibody selections performed by phage display on cancer cells or cancer-derived tissues.

Cancer type Cells targeted Selection system Library Type (Diversity) Identified Ag Ref.
Brain and Neural system
Glioblastoma PDT and X spheres Ex vivo + in vitro, S/I/PA NIL-h scFv (Sheets, 6.7×109) No [55]
Neuroblastoma JF, SK-N-SH, IMR-32 In vitro IL-h, PD scFv No [56]
Breast
MFC-7 In vitro NIL-h scFv (Sheets, 6.7×109) Mucin 1 [57]
8701BC In vitro, PA IL-h, PD scFv (5×108) No [58]
PDT/C Ex vivo fresh PDT, PA IL-h, PD Fab (1.7×106) Cytokeratin 10 [59]
Ex vivo, S/I NIL-h (Griffin 1, 1.2×109) GRP78 [60]
ACa SK-BR-3 In vitro, I/PA IL-h, PD scFv (5×109 and 8×108) HER2 [16]
In vitro, LCM NIL-h scFv (Tom I, 1.5×108) No [28]
In vitro, I/PA NIL-h scFv (Sheets, 6.7×109) HER2, TfR, + [19]
In vitro, spiked, S/MACS NIL-h scFv (Sheets, 6.7×109) HER2 [17]
MDA-MB-231 In vitro, S/I NIL-h scFv (1×1011) CD44, EGFR, + [13]
PDT Ex vivo, PA IL-h, PD Fab (1010) gC1q-Receptor [61]
Ca SUM159 Ex vivo X, S/PA/FACS NIL-h scFv (TomI+J; 1.5 and 1.4×108) No [62]
PM-1 In vitro, S NIL-h scFv (NBL library, 6.4×109) CD166 [63]
PDT Ex vivo, S/PA NIL-h scFv (Tom I, 1.5×108) HER2 [18]
Ex vivo, LCM/PA NIL-h scFv (Tom I, 1.5×108) No [29]
In vivo PT-BR NIL-h scFv (Tom I, 1.5×108) No [44]
Cervix
ACa HeLa In vitro ILCL-l sDAB α3β1 integrin [64]
Colorectal
ACa PDT Ex vivo, LCM NIL-h scFv (Tom I, 1.5×108) No [28]
Caco2 In vitro, PA NIL-h scFv (Nissim > 108) No [65]
SW480 In vitro, PA/S/DGC ILCL-m Fab No [66]
In vitro, S/PA/FACS NIL-h scFv (de Kruif, 3.6×108) EpCAM [67]
SW480, SW948, SW837 In vitro, S/DGC ILCL-m Fab (7.1×106 and 2.1×106) No [68]
ACa/Ca Caco2, CW-2, SW480, + In vitro, S NIL-h scFv (AIMS-5) @ [14]
Ca PDT In vivo PT-BR NIL-h scFv (Tom I, 1.5×108) No [47]
Esophagus
Squamous Ca Eca109 In vitro, PA NIL-h scFv (de Kruif, 3.6×108) No [69]
Gastric
Ca MKN45, NCI-N87, + In vitro, S NIL-h scFv (AIMS-5) @ [14]
Hematological Malignancies
AML PDC Ex vivo, S/PA NIL-h scFv (de Kruif, 3.6×108) NMNt, CD166, + [70]
PDC and HL-60 In vitro, S + ex vivo, S/FACS NIL-h scFv (de Kruif, 3.6×108) CLL-1 [71]
APML HL-60 In vitro, PA/FACS NIL-h scFv (Griffin 1, 1.2×109) No [72]
ATCL Jurkat In vitro, pathway+step-back NIL-h scFv (Metha, 1.5×1010) CD4, TfR [21]
CLL PDC Ex vivo, S IL-h, PD Fab No [73]
CML K562 In vitro, single cell in PC NIL-h scFv (TomI+J; 1.5 and 1.4×108) No [32]
Granulocytic Ly PDC In vivo PT-BR NIL-h scFv (Tom I, 1.5×108) No [47]
Multiple myeloma RPMI-8226 In vitro, S, Biotinylated PA NIL-h scFv (Griffin 1, 1.2×109) CD138 [74]
NHL Raji and Daudi In vitro, S ILCL-m Fab (1.9×107) No [75]
PDC Ex vivo, S/PA/FFPE NIL-h scFv (HuCAL, 1.6×1010) Vimentin [76]
Kidney
Renal Ca Caki-1, CFF-RC1, ACHN In vitro, S NIL-h scFv (AIMS-5) @ [14]
Liver
Hepatocellular Ca PDC and HepG2, HLF, + Ex vivo + In vitro, S NIL-h scFv (AIMS-5) @ [14]
HepG2 In vitro, PA IL-h Fab, PBMCs, (1.7×107) No [77]
Hepatocellular Ca HepG2 In vitro, PA NIL-h scFv (Griffin 1, 1.2×109) No [78]
Respiratory System
NSCLC A549 In vitro, S/PA NIL-h scFv (NBL library, 6.4×109) No [79]
In vitro, PA NIL-l sDab (5.4×108) CEA6 [80]
In vitro, PA IL-h, PD scFv (6×108) PrxI [81]
SCLC H889 In vitro, PA ILCL-m scFv (3.5×104 and 7.0×104) PDC-E2 [82]
Lung Ca A549, PC-14, Calu-3, + In vitro, S NIL-h scFv (AIMS-5) @ [14]
Mesothelium
Mesothelioma M28, VAMT-1 In vitro, I/PA NIL-h scFv (O’Connell, 5×108) No [83]
Ovarian
Ca SK-OV-3, RMG-1/-2, + In vitro, S NIL-h scFv (AIMS-5) @ [14]
SK-OV-3 In vitro, S NIL-h scFv (Nissim >108) EEF1A1 [84]
Pancreas
Ductal ACa/ACa/Ca Panc-1, BxPC-3, + In vitro, S NIL-h scFv (AIMS-5) @ [14]
Ca PDT In vivo PT-BR NIL-h scFv (Tom I, 1.5×108) No [44]
ACa SW1990, HPAF-II In vitro, I/PA NIL-h scFv (Gao) α3β1 integrin [85]
Prostate
ACa PPC-1 1st Ex vivo, 2nd in vivo, X NIL-h DAb (Garvan, 3×109) PA28αβ complex [25]
C4-2B In vitro, I/PA NIL-h scFv (5×108) ICAM-1 [86]
Ca 22Rv1 In vitro, I/PA NIL-h scFv (Gao) No [85]
Sarcomas
Chondrosarcoma PDT In vitro PT-BR NIL-h scFv (Tom I, 1.5×108) No [47]
Fibrosarcoma HT-1080 In vitro ILCL-m scFv (107) Hsp90 [87]
In vitro ILCL-m scFv (107) CD44 [20]
Osteosarcoma U2-OS In vivo PT-BR IL-h PD, (2.7×108) No [88]
Skin
Melanoma PDT In vivo PT-BR NIL-h scFv (Tom I, 1.5×108) No [47]
MeCoP In vitro, S NIL-h scFv (ETH-2-Gold 3×109) Syndecan-1 [89]
DM341-I, DM341-II, + In vitro ILCL-h, PD scFv No [90]
Squamous Ca A431 In vitro, S/I/PA NIL-h scFv (Sheets, 6.7×109) EGFR [15]

Table divided by origin of the cancer cells on which the selection is targeted. Cell targeted is the cell type on which the selection was performed. Library type is the type of phage antibody repertoire used for the selection(s). Identified antigen is the antibody cognate antigen, if identified; “No” means no antigen identified in the reference (Ref.). Table does not include selections performed on transfected cells or cells co-cultured with cancer cells or serum.

Abbreviations: ACa: Adenocarcinoma: Ag, Antigen; A(P)ML, Acute (pro)myelocytic leukaemia; ATCL, Acute T-Cell leukaemia; Ca, Carcinoma; CD, Cluster of differentiation; CL, Cell line; CLL, chronic lymphocytic leukemia; CCL-1, Chemokine (C-C motif) ligand 1; CEA6, carcinoembryonic antigen 6; CML, Chronic Myelogenous Leukemia; DAbs, single-domain antibody; DGC, density gradient centrifugation; EEF1A1, eukaryotic translation elongation factor 1 alpha 1; EGFR, epidermal growth factor receptor; EPCAM, epithelial cell adhesion molecule; gC1q-R, ; GRP78, 78 kDa glucose-regulated protein; h, Human; HER2, human epidermal growth factor receptor 2; Hsp90, heat shock protein 90; I, selection for internalization; ICAM-1, Intercellular Adhesion Molecule 1; IL, Immune library; ILCL, Immune library from immunization with cell line; l, Llama; LCM, Laser Capture Microdissection; Ly, Lymphoma; m, Mouse; NHL, non-Hodgkin lymphoma; NIL, Non-immune library; NMNt, Nicotinamide nucleotide adenylyltransferase 1; NS, Nervous system; NSCLC, Non-small cell lung cancer; PA, Library pre-absorbed on cells before selection; PC, primary cells; PDC-E2, Pyruvate Dehydrogenase Complex E2 component; PDT/C, Patient Derived Tissue/Cells; PrxI, Paired Related Homeobox 1; PT-BR, Patient Tissue, selection before removal; S, selection in suspension; SCLC, small cell lung cancer; scFv, single-chain fragment variable; Tom, Tomlinson; TfR, Transferrin Receptor; X, Xenograft; “+”, More cell lines used for cell target/more antigens identified; @, antigens identified, selection on multiple cancer cell types and cell lines, but not specific from which selections the antibody against the antigens were raised.

A few publications have identified novel tumor-associated antigens (Table 2) suggesting that new antibody selection methods should be developed to provide the means for discovering all potentially useful antigenic differences between tumors and normal tissues. Such methods should be suitable for discovery of antibodies for the targeting of low abundance antigens, sub-populations of rare cells, and antigens present in the tumor microenvironment. Phenotypic selections are being developed as alternative methods to identify novel targets. This phenotypic approach relies on the selection of antibodies based on their effect on the target, rather than previous knowledge of the target [2225]. In order to enable efficient selection of antibodies in an unmodified or minimally modified tumor environment that preserves antigenic diversity and topographic location, researchers need to develop new methods that deviate from the trodden track of target-directed selection. Such selections can be performed ex vivo on freshly isolated cancer tissue or patient derived biological fluids, or in vivo in tumor-bearing mice or in cancer patients.

Ex vivo phage selection

Tissue slide selection

It has been shown that the removal of any cell, including tumor cells, from their native environment will impact gene expression [26, 27]; thus caution has to be exercised when applying tumor cell lines to biomarker identification. Ideally, conditions as close to the natural environment as possible should be used. Another important issue affecting antigen discovery is tissue heterogeneity. To deal with both issues, novel antibody phage display selection strategies have been devised. Firstly, laser capture microdissection (LCM) has been utilized to obtain tumor specific phage antibodies because this technique allows one to isolate small clusters of target cells from tumor sections previously incubated with a phage library [28, 29]. Similarly to the commonly used subtraction methods, performing the selection before LCM creates competition for library variants recognizing antigens shared by tumor cells and surrounding tissue (pan-cell binding phage), which may increase the proportion of specific phage in the LCM-isolated cell cluster. An inherent problem with LCM is that the slide preparation conditions required for successful dissection, especially the dehydration step, drastically reduce phage infectivity [28, 3033]. Therefore, alternative procedures to retrieve the sequences of the peptides or antibodies encoded by the inserts in the phage DNA have been used, including subcloning of PCR-amplified inserts [30, 31].

Alternatively, investigators have searched for conditions that are compatible with both LCM and phage viability. For example, freeze drying the slides after selection preserves phage infectivity after LCM [33]. Most recently, Sun et al. [29] protected the phage from drying while performing LCM by using an overlying membrane that creates a moist environment capable of preserving phage viability for up to 6 hours. It is also possible to isolate clusters of cells by LCM before incubating them with the phage library [28, 34, 35]. Although LCM allows isolation of single cells from tissue sections, the general approach has been to dissect clusters of 20–50 cells. Ruan et al. [31] observed that antibody phage was recovered from single cell isolations only occasionally, and then as a single clone. Thus, the potential of antibody selection using LCM has hitherto been restricted to the use of clusters of multiple cells as the target.

Shadow-stick selection

Selecting antibodies against rare cells is often impeded by insufficient access of the phage to the target cells. To circumvent this problem, Sørensen and Kristensen [36] have presented a selection method targeting only a single cell within a heterogeneous population. The method has been dubbed ‘shadow-stick selection’ due to the use a minute disk attached to a glass pipette (resembling a hockey-stick), which is used to selectively protect the target cell, and the phage bound to it, from UV irradiation. By reducing the requirement to a single identified cell it was possible to select antibodies against an individual cultured K562 cell spiked into peripheral blood mononuclear cells (PBMC) [32] and against cells identified as fetal microchimeric cells in maternal blood [23]. This method is especially useful if the target cell is present in a very low frequency. For example, for some cancer types, circulating tumor cells (CTC) have been estimated to occur with a frequency of only one in a million of PBMCs [37]. In the shadow-stick protocol a heterogeneous cell suspension containing a few or only one target cell is applied to a glass slide (Figure 1). The cells are treated with a fixative to preserve cellular architecture and to keep the cells attached during the selection procedure and subsequent washing steps. Thereafter, the single target cell is identified and protected by a UV-impermeable disc attached to a glass capillary. By means of micromanipulation the disc is aligned above the cell of interest and the slide is exposed to UV-C light. The irradiation crosslinks the DNA of the unprotected phages bound to the surrounding non-target cells and thereby prevents their replication in E. coli. Thus, only protected phages give rise to colonies after infection, which will typically be in the range of 1–8 unique clones per single cell selection [36].

Figure 1.

Figure 1

Schematic illustration of the shadow-stick selection method. Phage affinity selection is performed on cells placed on a glass slide, including a target cell of interest (green cell). The shadow-stick is placed above the target cell before UV irradiating the slide, selectively protecting the phage bound to the target cell while “killing” all phage bound to non-target cells. Illustration by Simon Lykkemark.

The single cell selection method targets one cell in a large population of cells. For the method to be successful, the target cell should be present at a low frequency (preferably between one out of 10−4–10−6 cells). This will increase the probability of selecting antibodies that target antigens specific for the target cell. There are only 10 to 1000 copies of any given antibody present in the first round of selection, and the abundant non-target cells should capture the antibodies that bind to epitopes common to all cells [36]. This combined with gentle and controlled phage elution provides a method making it possible to select antibodies recognizing the target cell. Sufficient selectivity is achieved by using a protease sensitive helper phage [38], which can reduce the background of bound non-displaying phages. In phage antibody libraries where the antibody fusion is introduced by a phagemid only 1–10 % of the resulting phage particles will present an antibody on the surface. Thus 90–99 % of the phages binding to the single target cell are not captured through a specific antibody-antigen interaction. This background is removed by applying the protease sensitive helper phage, resulting in a low output of only1–8 unique clones per target cell [36]. Performing multiple rounds of selection generally favors enrichment of antibodies that have high affinities or target highly expressed antigens [39]. However, iterative selections, normally required when using conventional helper phages, may compromise the identification of specifically expressed antigens, as the most interesting biomarkers might not be expressed in high abundance or recognized by antibodies with high affinity; therefore only a single round of selection is preferred.

The strength of this selection method lies in the targeting of a single cell. Single cell analyses at the mRNA level have shown that phenotypically identical cells display transcriptional differences [40]. In contrast to selections on a pool of target cells, which are likely to be heterogeneous in their protein expression patterns, the single cell method can identify biomarkers uniquely expressed by individual cells.

In vivo phage selection

In vivo selection of phage peptides

Repertoires of peptides on the surface of the bacteriophage T7 have been successfully used in mouse models in vivo. Traditionally, phage display has been used in screens that are performed in vitro against a pre-defined target molecule or cell in order to identify probes recognizing the target molecule or cell. However, this strategy relies on pre-existing knowledge of the target. In the in vivo approach the phage library is screened in a live animal [41]; reviewed in ref [42] or human patient [43, 44]. The main advantage of in vivo screening is that the target is probed as it exists in situ, which eliminates the possibility that changes introduced by the removal of a tissue or cells from their natural environment. A prime example of a protein that is expressed differently in vivo and in vitro is the αvβ3 integrin; it is only expressed in highly activated cells, including tumor endothelial cells in vivo, whereas essentially all cultured cells express this integrin. Thus, screens and analyses performed with cultured cells would miss the fact that αvβ3 is an excellent marker for tumors and wounded/inflamed tissues, and is much used in targeting drugs, imaging agents, and nanoparticles to tumors [45]. In an in-vivo screen, the phage library is allowed to circulate through the organism and to interact with the molecules accessible from the blood, particularly, in the luminal surface of the vessels. Those phages harboring peptides that do not interact (or do so weakly) with any anchored molecule are washed away during the selection process. Further, the in vivo selection includes a built-in negative selection step because the whole vascular bed acts as a sink for those phages that harbor peptides capable of interacting with commonly expressed molecules. Finally, those phages displaying peptides that interact with molecules that are specific for a certain organ accumulate in that organ or tissue and can be retrieved, amplified and analyzed [41]. This in vivo selection has been very effective to map a variety of markers that distinguish the luminal surface of vessels in different organs, and that can discriminate between normal vessels and tumor vessels (vascular “zip codes”) [42].

Several peptides discovered by in vivo phage library selections have been used as targeting elements in delivering payloads to a chosen tissue, such as a tumor [46]. While peptides have their uses (for example, they are particularly well suited for the targeting of nanoparticles [45]), as we will discuss in the next subsection, antibodies have substantial advantages as targeting agents and therapeutics [2].

In vivo selection of phage antibodies

Some of the first attempts to apply the in vivo phage screening technology to antibodies have been in human patients. Krag et al. [44] combined peptide and recombinant antibody screening to run a Phase I clinical trial on 9 patients with melanoma, pancreas or breast cancer using recombinant antibody (and peptide) phage display. Two of the patients received the antibody library, and the tumor nodules were excised after 30 min of circulation to recover the bound antibodies for analysis. The study concluded that the administration of the phage was safe, and that some of the recovered antibodies were able to bind in vitro to tumor cells, but not to blood cells from the patients. However, no further characterization of the target, biodistribution, or the ability to localize the tumor in vivo was done. More recently, Shukla et al. [47] took this approach a step further by infusing a scFv library into three stage IV-patients and then evaluating 998 different phages for binding to tumor sections. They obtained 45 soluble scFv that were able to bind to tumor sections, of which only 2 did not bind to normal tissue sections. However, the binding to normal sections does not necessarily mean that the target antigen would be accessible in normal tissues in vivo, so it is possible that additional scFv would have been tumor-specific if in vivo assays had been available [47]. Interestingly, these authors identified a possible tumor-specific IL-17α mimic among the isolated phages on the basis of sequence homology.

In another in vivo phage antibody screen, Deramchia et al. [48] performed selection of human recombinant antibodies in an atherosclerotic mouse model. After three rounds of selection, with a very short circulation time, several clones were isolated from the atherosclerotic aorta. Some of the recombinant antibodies showed positive immunohistochemical staining in sections of atherosclerotic rabbit aorta and in sections of atherosclerotic lesions of human carotid and coronary arteries; however, neither the ability of the recombinant antibody to localize in the model in vivo nor the localization of the proposed target were studied.

Despite the fact that tumor blood vessels are abnormal, fragile, and hyperpermeable, and that phage can have opportunities to interact directly with cancer cells and stromal components, in vivo antibody screening in tumor-bearing mice has not been accomplished. Some of the technical limitations that may account for the lack of success include the high background with common phage selection procedures, as some organs non-specifically capture too many phages to be used as target organs for selection [41].

Sequential ex vivo and in vivo phage antibody selection

Recently, Sánchez-Martín et al. [25] reported having overcome some of the limitations of in vivo phage antibody screening and described the isolation of a tumor-homing antibody from a human single-domain antibody library. Using a prostate cancer mouse model, they combined ex vivo and in vivo rounds of selection of human recombinant antibodies from phage libraries. Initial ex vivo selections with freshly disaggregated tumors provided an enriched, but highly diversified repertoire of binders to tumor components, while minimizing the number of animals needed for the selection (Figure 2). The subsequent in vivo selection pruned the binders whose target is not available or otherwise not reachable in vivo, as well as the phages that bind to blood or tissue components, but are not tumor-specific. At that time, extensive conventional DNA sequencing was needed to minimize the rounds of selection needed and to preserve a high diversity that unveiled consensus sequences. High-throughput sequencing, which has since become available, makes these goals much easier to achieve. While helpful in all types of selection, high throughput sequencing particularly useful in the analysis in vivo results because of the substantial amount of work that goes into every round of selection. The ability of the selected antibodies to home to tumors and to deliver a payload to the tumors was examined by studying individual phage clones from the screen. Some of the phage clones stood out in that the distribution of the phage was not restricted to the endothelium but, after a 24 h circulation time, phage particles were widely distributed in perivascular areas. The proteasome activator complex PA28 was identified as the target of one of the antibodies by mass spectrometry, and verified by ELISA [25]. Further, the relevance of PA28 as a tumor marker was confirmed in studies on human primary and metastatic prostate cancer tissue [25]. These studies showed that it is possible to perform in vivo selection of human recombinant antibodies with the ability to selectively home to tumors from systemically administered injection. The target identification made possible by the availability of a tumor-homing probe can provide new tumor markers and unveil new functions for the target molecules.

Figure 2.

Figure 2

Schematic representation of the sequential selection process to obtain tumor-homing antibodies. The initial phage repertoire is pre-enriched on cell suspensions prepared from freshly excised tumors (ex vivo strategy) and the resulting enriched phage pool then systemically injected into tumor-bearing mice (in vivo strategy). Retained phage particles are recovered from the tumor. Illustration by Simon Lykkemark.

Concluding remarks and future perspectives

The generation of antibodies against cell surface markers selectively expressed by rare tumor subpopulations, but still with a crucial role in cancer progression, and the identification of antibody-targetable tumor-associated antigens in intact tumors are important bottlenecks in the development of novel immunotherapies for cancer. Hence, there is an urgent need to implement technological platforms and selection strategies for anti-cancer antibodies. Recently, considerable efforts have been devoted to optimizing phage display selection methods using antibody libraries ex vivo and in vivo. Advanced selection conditions make feasible the use of phage antibody libraries for selecting tumor-homing antibodies and the subsequent identification of their antigens for validation in primary and metastatic human cancer samples.

Among emerging techniques with high potential for applications to phage display selection against cells are the microfluidic-/lab-on-a-chip devices used for rare cell sorting. A few attempts to apply this technology for phage display selections have been made [49], but not on whole cells. This technique will be especially useful for selection against rare cells in suspension, such as circulating tumor cells.

The use of viral particles (e.g. bacteriophages) in vivo has several disadvantages. Systemically administered virus must avoid destruction by immune effector mechanism, must avoid nonspecific adhesion and sequestration, and must extravasate selectively at tumor sites. However, phages are inefficient at crossing endothelial barriers. Therefore, it would be of significant value to use other repertoire display particles that traffic to tumors more efficiently, such as macrophages, T cells, and stem cells. Although several mammalian cell surface display platforms have been developed [50], the display of antibodies on the surface of primary T lymphocytes, as a part of a chimeric antigen receptor (CAR) [51, 52], could simplify the selection process, since effective triggering would promote CAR-T cell survival and expansion. One of the biggest hurdles to overcome would be the production of large diversity repertoires. However, mammalian platforms could be used as a second round selection method using ex vivo pre-enriched phage pools.

A possible way of circumventing the limitation on the size of antibody repertoire would be to create an antibody library by inserting into the antibody framework a pool of peptide sequences obtained by screening on a desired target cell or tissue. The resulting library could then be screened on the same target to obtain antibodies for that target. The feasibility of this approach is suggested by antibody engineering work performed 20 years ago; grafting the integrin-binding RGD sequence into an antibody CDR resulted in an integrin-binding antibody [53]. Moreover, this antibody could be further evolved by introducing mutations to the CDR and selecting for additional binding properties [54]. It may be possible to adapt this strategy to screening against unknown, complex targets, including tissues targeted in vivo. All these approaches could also be easily adapted to in vivo selection processes in tumor-bearing mice, and in animal models for other diseases. Clearly, in vivo screening of large antibody libraries has great potential that has not been fully tapped.

Acknowledgments

L.A.-V. was supported by grants from the Ministerio de Economía y Competitividad (BIO2011-22738) and the Novo Nordisk Foundation (11019). E.R. was supported by grants from the National Cancer Institute. L.S. was supported by grants from the Fondo de Investigación Sanitaria/Instituto de Salud Carlos III (PI13/00090), and the Comunidad de Madrid (S2010/BMD-2312). M.D.S. was supported by a grant from the Danish Cancer Society. P.K was supported by grants from the Lundbeck foundation (R126-2012-12143), The Danish Council for Independent Research | Technology and Production Sciences (09-065063 and 0602-02377B) and the Danish Cancer Society (R40-A2115).

Footnotes

Competing financial interests

ER has a patent for in vivo screening of random libraries of compounds that Sanford-Burnham Medical Research Institute has licensed to various companies. The other authors declare no competing financial interests.

References

  • 1.Weiner LM, et al. Monoclonal antibodies: versatile platforms for cancer immunotherapy. Nature reviews. Immunology. 2010;10:317–327. doi: 10.1038/nri2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Cuesta AM, et al. Multivalent antibodies: when design surpasses evolution. Trends Biotechnol. 2010;28:355–362. doi: 10.1016/j.tibtech.2010.03.007. [DOI] [PubMed] [Google Scholar]
  • 3.Reichert JM. Antibodies to watch in 2014: Mid-year update. mAbs. 2014;6:799–802. doi: 10.4161/mabs.29282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Vacchelli E, et al. Trial Watch: Tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology. 2014;3:e27048. doi: 10.4161/onci.27048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Aranda F, et al. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology. 2014;3:e27297. doi: 10.4161/onci.27297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Carter P, et al. Identification and validation of cell surface antigens for antibody targeting in oncology. Endocrine-related cancer. 2004;11:659–687. doi: 10.1677/erc.1.00766. [DOI] [PubMed] [Google Scholar]
  • 7.Uva P, et al. Comparative Membranome expression analysis in primary tumors and derived cell lines. PloS one. 2010;5:e11742. doi: 10.1371/journal.pone.0011742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sanz L, et al. Differential transplantability of human endothelial cells in colorectal cancer and renal cell carcinoma primary xenografts. Laboratory investigation; a journal of technical methods and pathology. 2009;89:91–97. doi: 10.1038/labinvest.2008.108. [DOI] [PubMed] [Google Scholar]
  • 9.Swartz MA, et al. Tumor microenvironment complexity: emerging roles in cancer therapy. Cancer research. 2012;72:2473–2480. doi: 10.1158/0008-5472.CAN-12-0122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Heine M, et al. High interstitial fluid pressure is associated with low tumour penetration of diagnostic monoclonal antibodies applied for molecular imaging purposes. PloS one. 2012;7:e36258. doi: 10.1371/journal.pone.0036258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501:346–354. doi: 10.1038/nature12626. [DOI] [PubMed] [Google Scholar]
  • 12.Bradbury AR, et al. Beyond natural antibodies: the power of in vitro display technologies. Nature biotechnology. 2011;29:245–254. doi: 10.1038/nbt.1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rust S, et al. Combining phenotypic and proteomic approaches to identify membrane targets in a ‘triple negative’ breast cancer cell type. Molecular cancer. 2013;12:11. doi: 10.1186/1476-4598-12-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kurosawa G, et al. Comprehensive screening for antigens overexpressed on carcinomas via isolation of human mAbs that may be therapeutic. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:7287–7292. doi: 10.1073/pnas.0712202105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Heitner T, et al. Selection of cell binding and internalizing epidermal growth factor receptor antibodies from a phage display library. Journal of immunological methods. 2001;248:17–30. doi: 10.1016/s0022-1759(00)00340-9. [DOI] [PubMed] [Google Scholar]
  • 16.Ayat H, et al. Isolation of scFv antibody fragments against HER2 and CEA tumor antigens from combinatorial antibody libraries derived from cancer patients. Biologicals: journal of the International Association of Biological Standardization. 2013;41:345–354. doi: 10.1016/j.biologicals.2013.05.004. [DOI] [PubMed] [Google Scholar]
  • 17.Mazuet C, et al. Breast carcinoma specific antibody selection combining phage display and immunomagnetic cell sorting. Biochem Biophys Res Commun. 2006;348:550–559. doi: 10.1016/j.bbrc.2006.07.087. [DOI] [PubMed] [Google Scholar]
  • 18.Shukla GS, Krag DN. Phage display selection for cell-specific ligands: development of a screening procedure suitable for small tumor specimens. Journal of drug targeting. 2005;13:7–18. doi: 10.1080/10611860400020464. [DOI] [PubMed] [Google Scholar]
  • 19.Goenaga AL, et al. Identification and characterization of tumor antigens by using antibody phage display and intrabody strategies. Mol Immunol. 2007;44:3777–3788. doi: 10.1016/j.molimm.2007.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hauptschein RS, et al. Functional proteomic screen identifies a modulating role for CD44 in death receptor-mediated apoptosis. Cancer research. 2005;65:1887–1896. doi: 10.1158/0008-5472.CAN-04-3571. [DOI] [PubMed] [Google Scholar]
  • 21.Sui J, et al. Identification of CD4 and transferrin receptor antibodies by CXCR4 antibody-guided Pathfinder selection. European journal of biochemistry/FEBS. 2003;270:4497–4506. doi: 10.1046/j.1432-1033.2003.03843.x. [DOI] [PubMed] [Google Scholar]
  • 22.Xie J, et al. Prevention of cell death by antibodies selected from intracellular combinatorial libraries. Chemistry & biology. 2014;21:274–283. doi: 10.1016/j.chembiol.2013.12.006. [DOI] [PubMed] [Google Scholar]
  • 23.Sorensen MD, et al. Raising antibodies against circulating foetal cells from maternal peripheral blood. Prenatal diagnosis. 2013;33:284–291. doi: 10.1002/pd.4060. [DOI] [PubMed] [Google Scholar]
  • 24.Yea K, et al. Converting stem cells to dendritic cells by agonist antibodies from unbiased morphogenic selections. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:14966–14971. doi: 10.1073/pnas.1313671110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sanchez-Martin D, et al. Proteasome activator complex PA28 identified as an accessible target in prostate cancer by in vivo selection of human antibodies. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:13791–13796. doi: 10.1073/pnas.1300013110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Boess F, et al. Gene expression in two hepatic cell lines, cultured primary hepatocytes, and liver slices compared to the in vivo liver gene expression in rats: possible implications for toxicogenomics use of in vitro systems. Toxicological sciences: an official journal of the Society of Toxicology. 2003;73:386–402. doi: 10.1093/toxsci/kfg064. [DOI] [PubMed] [Google Scholar]
  • 27.Pan C, et al. Comparative proteomic phenotyping of cell lines and primary cells to assess preservation of cell type-specific functions. Molecular & cellular proteomics: MCP. 2009;8:443–450. doi: 10.1074/mcp.M800258-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sun Y, et al. Biopanning Phage-Display Libraries on Small Tissue Sections Captured by Laser Capture Microdissection. Journal of biotech research. 2009;1:55–63. [PMC free article] [PubMed] [Google Scholar]
  • 29.Sun Y, et al. Phage-display selection on tumor histological specimens with laser capture microdissection. Journal of immunological methods. 2009;347:46–53. doi: 10.1016/j.jim.2009.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yao VJ, et al. Targeting pancreatic islets with phage display assisted by laser pressure catapult microdissection. The American journal of pathology. 2005;166:625–636. doi: 10.1016/S0002-9440(10)62283-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ruan W, et al. Identification of clinically significant tumor antigens by selecting phage antibody library on tumor cells in situ using laser capture microdissection. Molecular & cellular proteomics: MCP. 2006;5:2364–2373. doi: 10.1074/mcp.M600246-MCP200. [DOI] [PubMed] [Google Scholar]
  • 32.Sorensen MD, et al. Microselection–affinity selecting antibodies against a single rare cell in a heterogeneous population. Journal of cellular and molecular medicine. 2010;14:1953–1961. doi: 10.1111/j.1582-4934.2010.00896.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lu H, et al. Application of laser capture microdissection to phage display peptide library screening. Oral surgery, oral medicine, oral pathology, oral radiology, and endodontics. 2004;98:692–697. doi: 10.1016/j.tripleo.2004.09.004. [DOI] [PubMed] [Google Scholar]
  • 34.Kubo N, et al. Identification of oligopeptide binding to colon cancer cells separated from patients using laser capture microdissection. Journal of drug targeting. 2008;16:396–404. doi: 10.1080/10611860802088796. [DOI] [PubMed] [Google Scholar]
  • 35.Tanaka T, et al. In situ phage screening. A method for identification of subnanogram tissue components in situ. The Journal of biological chemistry. 2002;277:30382–30387. doi: 10.1074/jbc.M203547200. [DOI] [PubMed] [Google Scholar]
  • 36.Sorensen MD, Kristensen P. Selection of antibodies against a single rare cell present in a heterogeneous population using phage display. Nature protocols. 2011;6:509–522. doi: 10.1038/nprot.2011.311. [DOI] [PubMed] [Google Scholar]
  • 37.Ross AA, et al. Detection and viability of tumor cells in peripheral blood stem cell collections from breast cancer patients using immunocytochemical and clonogenic assay techniques. Blood. 1993;82:2605–2610. [PubMed] [Google Scholar]
  • 38.Kristensen P, Winter G. Proteolytic selection for protein folding using filamentous bacteriophages. Folding & design. 1998;3:321–328. doi: 10.1016/S1359-0278(98)00044-3. [DOI] [PubMed] [Google Scholar]
  • 39.de Wildt RM, et al. Antibody arrays for high-throughput screening of antibody-antigen interactions. Nature biotechnology. 2000;18:989–994. doi: 10.1038/79494. [DOI] [PubMed] [Google Scholar]
  • 40.Subkhankulova T, et al. Modelling and measuring single cell RNA expression levels find considerable transcriptional differences among phenotypically identical cells. BMC genomics. 2008;9:268. doi: 10.1186/1471-2164-9-268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pasqualini R, Ruoslahti E. Organ targeting in vivo using phage display peptide libraries. Nature. 1996;380:364–366. doi: 10.1038/380364a0. [DOI] [PubMed] [Google Scholar]
  • 42.Teesalu T, et al. Mapping of vascular ZIP codes by phage display. Methods in enzymology. 2012;503:35–56. doi: 10.1016/B978-0-12-396962-0.00002-1. [DOI] [PubMed] [Google Scholar]
  • 43.Arap W, et al. Steps toward mapping the human vasculature by phage display. Nature medicine. 2002;8:121–127. doi: 10.1038/nm0202-121. [DOI] [PubMed] [Google Scholar]
  • 44.Krag DN, et al. Selection of tumor-binding ligands in cancer patients with phage display libraries. Cancer research. 2006;66:7724–7733. doi: 10.1158/0008-5472.CAN-05-4441. [DOI] [PubMed] [Google Scholar]
  • 45.Ruoslahti E. Peptides as targeting elements and tissue penetration devices for nanoparticles. Adv Mater. 2012;24:3747–3756. doi: 10.1002/adma.201200454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ruoslahti E, et al. Targeting of drugs and nanoparticles to tumors. J Cell Biol. 2010;188:759–768. doi: 10.1083/jcb.200910104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Shukla GS, et al. Intravenous infusion of phage-displayed antibody library in human cancer patients: enrichment and cancer-specificity of tumor-homing phage-antibodies. Cancer immunology, immunotherapy: CII. 2013;62:1397–1410. doi: 10.1007/s00262-013-1443-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Deramchia K, et al. In vivo phage display to identify new human antibody fragments homing to atherosclerotic endothelial and subendothelial tissues [corrected] The American journal of pathology. 2012;180:2576–2589. doi: 10.1016/j.ajpath.2012.02.013. [DOI] [PubMed] [Google Scholar]
  • 49.Liu Y, et al. Controlling the selection stringency of phage display using a microfluidic device. Lab on a chip. 2009;9:1033–1036. doi: 10.1039/b820985e. [DOI] [PubMed] [Google Scholar]
  • 50.Ho M, Pastan I. Mammalian cell display for antibody engineering. Methods Mol Biol. 2009;525:337–352. xiv. doi: 10.1007/978-1-59745-554-1_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Alonso-Camino V, et al. Lymphocyte display: a novel antibody selection platform based on T cell activation. PloS one. 2009;4:e7174. doi: 10.1371/journal.pone.0007174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Alonso-Camino V, et al. CARbodies: Human Antibodies Against Cell Surface Tumor Antigens Selected From Repertoires Displayed on T Cell Chimeric Antigen Receptors. Molecular therapy. Nucleic acids. 2013;2:e93. doi: 10.1038/mtna.2013.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Barbas CF, 3rd, et al. High-affinity self-reactive human antibodies by design and selection: targeting the integrin ligand binding site. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:10003–10007. doi: 10.1073/pnas.90.21.10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Smith JW, et al. Building synthetic antibodies as adhesive ligands for integrins. The Journal of biological chemistry. 1994;269:32788–32795. [PubMed] [Google Scholar]
  • 55.Zhu X, et al. Identification of internalizing human single-chain antibodies targeting brain tumor sphere cells. Mol Cancer Ther. 2010;9:2131–2141. doi: 10.1158/1535-7163.MCT-09-1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rossig C, et al. Selection of human antitumor single-chain Fv antibodies from the B-cell repertoire of patients immunized against autologous neuroblastoma. Medical and pediatric oncology. 2000;35:692–695. doi: 10.1002/1096-911x(20001201)35:6<692::aid-mpo45>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
  • 57.Wong C, et al. Human scFv antibody fragments specific for the epithelial tumour marker MUC-1, selected by phage display on living cells. Cancer immunology, immunotherapy: CII. 2001;50:93–101. doi: 10.1007/s002620100174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rothe A, et al. Construction of phage display libraries from reactive lymph nodes of breast carcinoma patients and selection for specifically binding human single chain Fv on cell lines. International journal of molecular medicine. 2004;14:729–735. doi: 10.3892/ijmm.14.4.729. [DOI] [PubMed] [Google Scholar]
  • 59.Araujo TG, et al. A novel highly reactive Fab antibody for breast cancer tissue diagnostics and staging also discriminates a subset of good prognostic triple-negative breast cancers. Cancer letters. 2014;343:275–285. doi: 10.1016/j.canlet.2013.09.029. [DOI] [PubMed] [Google Scholar]
  • 60.Jakobsen CG, et al. Phage display derived human monoclonal antibodies isolated by binding to the surface of live primary breast cancer cells recognize GRP78. Cancer research. 2007;67:9507–9517. doi: 10.1158/0008-5472.CAN-06-4686. [DOI] [PubMed] [Google Scholar]
  • 61.Rubinstein DB, et al. Receptor for the globular heads of C1q (gC1q-R, p33, hyaluronan-binding protein) is preferentially expressed by adenocarcinoma cells. International journal of cancer. Journal international du cancer. 2004;110:741–750. doi: 10.1002/ijc.20105. [DOI] [PubMed] [Google Scholar]
  • 62.Gur D, et al. Identification of single chain antibodies to breast cancer stem cells using phage display. Biotechnology progress. 2009;25:1780–1787. doi: 10.1002/btpr.285. [DOI] [PubMed] [Google Scholar]
  • 63.Wiiger MT, et al. A novel human recombinant single-chain antibody targeting CD166/ALCAM inhibits cancer cell invasion in vitro and in vivo tumour growth. Cancer immunology, immunotherapy: CII. 2010;59:1665–1674. doi: 10.1007/s00262-010-0892-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Groot AJ, et al. Reverse proteomic antibody screening identifies anti adhesive VHH targeting VLA-3. Molecular immunology. 2009;46:2022–2028. doi: 10.1016/j.molimm.2009.03.002. [DOI] [PubMed] [Google Scholar]
  • 65.Staneloudi C, et al. Development and characterization of novel photosensitizer: scFv conjugates for use in photodynamic therapy of cancer. Immunology. 2007;120:512–517. doi: 10.1111/j.1365-2567.2006.02522.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Liebman MA, et al. Generation and preliminary characterization of an antibody library with preferential reactivity to human colorectal cancer cells as compared to normal human blood cells. Immunology letters. 2004;91:179–188. doi: 10.1016/j.imlet.2003.12.002. [DOI] [PubMed] [Google Scholar]
  • 67.Huls GA, et al. A recombinant, fully human monoclonal antibody with antitumor activity constructed from phage-displayed antibody fragments. Nature biotechnology. 1999;17:276–281. doi: 10.1038/7023. [DOI] [PubMed] [Google Scholar]
  • 68.Williams BR, Sharon J. Polyclonal anti-colorectal cancer Fab phage display library selected in one round using density gradient centrifugation to separate antigen-bound and free phage. Immunology letters. 2002;81:141–148. doi: 10.1016/s0165-2478(02)00004-4. [DOI] [PubMed] [Google Scholar]
  • 69.Xu MY, et al. Production of a human single-chain variable fragment antibody against esophageal carcinoma. World journal of gastroenterology: WJG. 2004;10:2619–2623. doi: 10.3748/wjg.v10.i18.2619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Geuijen CA, et al. A proteomic approach to tumour target identification using phage display, affinity purification and mass spectrometry. Eur J Cancer. 2005;41:178–187. doi: 10.1016/j.ejca.2004.10.008. [DOI] [PubMed] [Google Scholar]
  • 71.Bakker AB, et al. C-type lectin-like molecule-1: a novel myeloid cell surface marker associated with acute myeloid leukemia. Cancer research. 2004;64:8443–8450. doi: 10.1158/0008-5472.CAN-04-1659. [DOI] [PubMed] [Google Scholar]
  • 72.Shadidi M, Sioud M. An anti-leukemic single chain Fv antibody selected from a synthetic human phage antibody library. Biochem Biophys Res Commun. 2001;280:548–552. doi: 10.1006/bbrc.2000.4158. [DOI] [PubMed] [Google Scholar]
  • 73.Baskar S, et al. A human monoclonal antibody drug and target discovery platform for B-cell chronic lymphocytic leukemia based on allogeneic hematopoietic stem cell transplantation and phage display. Blood. 2009;114:4494–4502. doi: 10.1182/blood-2009-05-222786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Delcommenne M, Klingemann HG. Detection and characterization of syndecan-1-associated heparan sulfate 6-O-sulfated motifs overexpressed in multiple myeloma cells using single chain antibody variable fragments. Human antibodies. 2012;21:29–40. doi: 10.3233/HAB-2012-0259. [DOI] [PubMed] [Google Scholar]
  • 75.Shen Y, et al. Generation and selection of immunized Fab phage display library against human B cell lymphoma. Cell research. 2007;17:650–660. doi: 10.1038/cr.2007.57. [DOI] [PubMed] [Google Scholar]
  • 76.Jarutat T, et al. Selection of vimentin-specific antibodies from the HuCAL phage display library by subtractive panning on formalin-fixed, paraffin-embedded tissue. Biological chemistry. 2007;388:651–658. doi: 10.1515/BC.2007.070. [DOI] [PubMed] [Google Scholar]
  • 77.Shui X, et al. Construction and selection of human Fab antibody phage display library of liver cancer. Hybridoma (Larchmt) 2009;28:341–347. doi: 10.1089/hyb.2009.0028. [DOI] [PubMed] [Google Scholar]
  • 78.Yu B, et al. Human scFv antibody fragments specific for hepatocellular carcinoma selected from a phage display library. World journal of gastroenterology: WJG. 2005;11:3985–3989. doi: 10.3748/wjg.v11.i26.3985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Reiersen H, et al. Screening human antibody libraries against carcinoma cells by affinity purification and polymerase chain reaction. Journal of immunological methods. 2008;330:44–56. doi: 10.1016/j.jim.2007.10.017. [DOI] [PubMed] [Google Scholar]
  • 80.Zhang J, et al. A pentavalent single-domain antibody approach to tumor antigen discovery and the development of novel proteomics reagents. Journal of molecular biology. 2004;341:161–169. doi: 10.1016/j.jmb.2004.05.069. [DOI] [PubMed] [Google Scholar]
  • 81.Luo Y, et al. Production and radioimmunoimaging of novel fully human phage display recombinant antibodies and growth inhibition of lung adenocarcinoma cell line overexpressing Prx I. Cancer biology & therapy. 2009;8:1369–1377. doi: 10.4161/cbt.8.14.8727. [DOI] [PubMed] [Google Scholar]
  • 82.Douguchi J, et al. Construction of human monoclonal single-chain Fv antibodies against small-cell lung cancer by phage display libraries derived from cell-immunized SCID mice engrafted with human peripheral blood lymphocytes. Proteomics. Clinical applications. 2009;3:1265–1272. doi: 10.1002/prca.200900060. [DOI] [PubMed] [Google Scholar]
  • 83.An F, et al. Targeted drug delivery to mesothelioma cells using functionally selected internalizing human single-chain antibodies. Mol Cancer Ther. 2008;7:569–578. doi: 10.1158/1535-7163.MCT-07-2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Sharma S, et al. Characterization of a putative ovarian oncogene, elongation factor 1alpha, isolated by panning a synthetic phage display single-chain variable fragment library with cultured human ovarian cancer cells. Clinical cancer research: an official journal of the American Association for Cancer Research. 2007;13:5889–5896. doi: 10.1158/1078-0432.CCR-07-0703. [DOI] [PubMed] [Google Scholar]
  • 85.Gao C, et al. De novo identification of tumor-specific internalizing human antibody-receptor pairs by phage-display methods. Journal of immunological methods. 2003;274:185–197. doi: 10.1016/s0022-1759(02)00522-7. [DOI] [PubMed] [Google Scholar]
  • 86.Conrad F, et al. Human antibodies targeting cell surface antigens overexpressed by the hormone refractory metastatic prostate cancer cells: ICAM-1 is a tumor antigen that mediates prostate cancer cell invasion. J Mol Med (Berl) 2009;87:507–514. doi: 10.1007/s00109-009-0446-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Eustace BK, et al. Functional proteomic screens reveal an essential extracellular role for hsp90 alpha in cancer cell invasiveness. Nature cell biology. 2004;6:507–514. doi: 10.1038/ncb1131. [DOI] [PubMed] [Google Scholar]
  • 88.Dantas-Barbosa C, et al. Isolation of osteosarcoma-associated human antibodies from a combinatorial Fab phage display library. Journal of biomedicine & biotechnology. 2009;2009:157531. doi: 10.1155/2009/157531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Orecchia P, et al. A novel human anti-syndecan-1 antibody inhibits vascular maturation and tumour growth in melanoma. Eur J Cancer. 2013;49:2022–2033. doi: 10.1016/j.ejca.2012.12.019. [DOI] [PubMed] [Google Scholar]
  • 90.Cai X, Garen A. Anti-melanoma antibodies from melanoma patients immunized with genetically modified autologous tumor cells: selection of specific antibodies from single-chain Fv fusion phage libraries. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:6537–6541. doi: 10.1073/pnas.92.14.6537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Sánchez-Martin D, et al. In vivo selection of tumor-specific antibodies. Oncotarget. 2013;4:1547. doi: 10.18632/oncotarget.1407. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES