Abstract
Targeting ligands are used in drug delivery to improve drug distribution to desired cells or tissues and to facilitate cellular entry. In vivo biopanning, whereby billions of potential ligand sequences are screened in biologically-relevant and complex conditions, is a powerful method for identification of novel target ligands. This tool has impacted drug delivery technologies and expanded our arsenal of therapeutics and diagnostics. Within this review we will discuss current in vivo panning technologies and ways that these technologies can be improved to advance next-generation drug delivery strategies.
Keywords: Biopanning, drug delivery, peptides, aptamers, viral capsids, in vivo
Graphical Abstract

1. Introduction
1.1. Biopanning: A platform to identify novel target ligands
Since its inception in 1985, biopanning has proven to be a powerful method for enhancing the rapid discovery of novel biological ligands for a variety of applications including, but not limited to, drug discovery, delivery and molecular biology[1–5]. The initial biopanning principle harnessed recombinant DNA technology[6] and Sanger sequencing[7] to engineer and develop biological platforms to express and display recombinant peptide-libraries in biological systems, which allowed for high throughput screening of novel ligands to a specified target of interest[5, 8–10]. Since that time, techniques such as directed evolution and next generation sequencing have increased development of high-affinity ligands[5, 11–13]. Furthermore, recombinant libraries have expanded from peptides to include antibodies, RNA, DNA and viral capsids—all providing a fresh and comprehensive approach to identify novel ligands[4, 14–16].
1.2. Targeted Drug Delivery: A need for novel ligand development
Targeted delivery of drugs involves technologies that increase distribution of pharmacologic agents to desired locations in the body. Delivery vehicles can mitigate off-target drug actions that result in significant and dose-limited side effects[17–19]. There are two main strategies for targeted drug delivery: passive or active targeting[18, 20, 21]. Passive targeting strategies rely on the physicochemical properties (e.g. charge, size) of a carrier and the body’s physiology to affect drug distribution[20, 21]. In contrast, active targeting employs specific molecular interactions between the drug carrier and target tissues or cells for delivery[18, 20]. Despite the theoretical advantage of targeting, few FDA approved drug-formulations utilize an active targeting approach (Table 1).
Table 1:
Pharmaceutical products with active drug targeting that are either FDA approved or in late-stage clinical trials
| Biopanning method | Native Physiological Function |
Clinical Application(s) | Target(s) | Trade Names | Development |
|---|---|---|---|---|---|
| Phage Display (Antibody) | Immunoglobulin, pathogen neutralization | Cancer | CD33, CD30, HER2 | Mylotarg (Gemtuzumab ozogamicin), Adcetris (Brentuximab vedotin), Kadcyla (Trastuzumab emtansine) | Antibody Conjugated to Chemotherapeutic Drug (Pfizer/Wyeth, Seattle Genetics/Millennium/Takeda, Genentech and Roche) |
| Phage Display (Kunitz Domain: 90 AA modifications in-between beta sheets) | Protease inhibitor (e.g. APP, TFPI) | Hereditary Angioedema | Kallirkein | Kalbitor (Ecallantide) | Macromolecular active drug discovered through phage display. (Shire) |
| Phage Display (Monobody: 90 AA modifications in-between beta sheets) | Fibrinectin type III domain | Glioblastoma | VEGFR2 | Angiocept (Pegdinetanib) | Macromolecular active drug discovered through phage display. (Bristol-Myers Squibb) |
| Phage Display (Darpins: 24 ankyrin repeats) | Ankyrin, a membrane protein that anchors proteins to the actin cytoskeleton | Macular degeneration | VEGF-A | Abicipar | Macromolecular active drug discovered through phage display. (Molecular Partners AG) |
| Phage Display (Peptides, 7-12 residues) | Biologically active small molecules | Cancer | SSTR2, αν integrins, CD13 | Lutathera (Lutetium- Lu77-Dotatate), Cilengitide (RGD alone)/ Fluciclatide (RGD in combination with F18), Zafiride (NGR-TNFα) | Peptide radioisotope conjugate, peptide alone/peptide radioisotope conjugate, peptide cytokine conjugate. (Advanced Accelerator Applications/Novartis, Merck/GM Healthcare, Adis Insight) |
The most commercially successful class of targeted delivery methods are antibody-drug conjugates or peptide/protein-based systems, wherein cytotoxic drugs are conjugated to a targeting protein via cleavable linkers (Table 1)[19, 22, 23]. These therapeutics have had clinical success due to their ability to recognize target cells with high affinity, to limit off-target toxicity, and to circulate for long periods. However, many protein-based targeting platforms (e.g. antibodies) are limited by their difficulty transporting agents beyond the vasculature due to their size and high binding affinity[24–26]. Therefore, significant interest exists for the development of small protein/peptides and nucleic acid structures that have greater accessibility and tissue penetration properties. Moreover, these small peptide and nucleic acid agents can be designed for oral and transdermal delivery to increases disease state accessibility and improve delivery outcomes[27–30]. Thus, biopanning methods for identifying a variety of targeting ligands are vital for developing and designing effective active targeting-carriers for a broad range of disease applications and administration routes.
2. In vivo biopanning platforms for developing novel targeting ligands
The immune system has evolved a tremendously complex and efficient means to produce antibodies that recognize new and recurrent foreign antigens either by expanding clonal populations of B-cells or by processing and presenting new antigens for antibody production. Antibodies against the new infectious agent are then selected as part of the adaptive immune response and improved through affinity maturation[31]. Thus, the diversity of unique antibodies in humans is in the billions[31, 32]. Biopanning is a process that mimics antibody selection by screening large libraries for potential binding ligands and then engineering and refining the ligands to increase binding affinity and targeting[8, 33, 34]. Through biopanning, novel ligands can be identified for target receptors for use as molecular targeting agents in drug delivery. In this review, we will discuss the current active clinically approved biopanned agents, the literature that outlines in vivo biopanning technologies, and strategies to design novel panning platforms to improve ligand discovery and validation.
2.1. In vivo biopanning as a tool to discover novel ligands
The success of any biopanning strategy relies heavily on the design and stringency of a selection scheme to maximize stringency and eliminate non-specific binders. All biopanning paradigms utilize a ligand library that contains an inherent coding method (e.g. affiliated nucleic acid or chromophores) for moiety identification and reproduction after recovery of potential binders. Libraries are panned against a series or combination of positive- or negative-selection “schemes” to enrich the capture of specific binders over weak non-specific binders[35, 36].For example, bacteriophage libraries can be incubated initially with a non-specific cell-type to remove off-target phage-binding (i.e. negative selection strategies). After removal of non-specific binders, the recovered phage population is then incubated with the target cell-type and washed to ensure stringent selection of high-affinity binders (i.e. positive selection)[9, 37, 38]. Multiple cycles and combinations of positive- or negative-selection can be performed to enrich for binding-phage further. After recovery of the high-affinity binders, the bacteriophage DNA is sequenced to identify each binding-motif[37, 39]. Validation of specific binders from the recovered pool is generally the most challenging and time-consuming step in biopanning[11, 13, 40, 41]. In in vivo biopanning, negative selection occurs naturally and with significant stringency as off-target tissue and protein interactions eliminate non-specific ligands to enrich the recovery of target-specific ligands and cellular receptors[42–44] (Figure 1). In theory, in vivo panning harnesses the heterogeneity of an intact biological system and improves the specificity of the identified ligand[43, 44]. Over the last 20 years, several in vivo panning display technologies and strategies have been developed. This section summarizes and highlights lessons learned.
Figure 1.

In vivo phage panning schematic
2.2. In vivo Phage Display
The success of in vivo biopanning display is dependent on the library diversity and the stability of the panning platform. Increases in diversity increases overall hit rates and enhances novel outputs. Enhanced stability increases hit recovery and allows diversification of biological applications. We will outline within this review the current panning platforms each with their own current diversification and stability issues (Table 2).
Table 2:
Current in vivo Panning Platforms
| Panning platform |
Library type |
Diversity | Typical number of selection rounds |
Size | In vivo stability |
|---|---|---|---|---|---|
| Bacteriophage | Peptide | ~10e9 (M13) to ~10e11 (T7) | 3–5 rounds | 60 nm for T7, 880 nm x 9 nm for M13 | ~4 hrs for unmodified and ~20 mins for peptide displayed M13[45], ~10 mins T7[46], increases after modification to reduce immune recognition [47] |
| Antibody | ~10e8 | 2–4 rounds | |||
| Aptamer libraries | RNA and DNA | 10e10- to 10e60 (depends on length, 4^n) | > 15 rounds (without PEG modification) | ~10 kDa [48] | Low without modification |
| Somamer | N/A (added post-SELEX) | N/A (added post-SELEX) | Depends on modification. I.e. PEG --> 40 kDa, NFkB-aptamer ~110 kDa[48] | High, but depends on modification | |
| Virus | Virus | 10e6 – 10e8 | 2–5 rounds | 25 nm diam. | High |
| Displayed peptide | Theoretical 10e8 | 3–5 rounds | ~28 nm diam. | High but slightly reduced compared to unmodified[49] |
2.2.1. Bacteriophage library platforms
Bacteriophage, or phage, libraries are generated through integration of a variable peptide sequences or proteins/antibodies onto a coat protein of bacteriophages[4, 50]. The phage platform incorporates both the displayed motif and the DNA encoding information into a single compact construct, allowing for simultaneous panning and subsequent sequencing of massively diverse libraries[8, 9]. For peptide libraries, the most commonly used phage vector is the M13 phage; the foreign peptide sequence, generally 7–12 amino acids in length, can be fused to minor pIII (3–5 copies of the peptide sequence) or major pVIII (2700 copies of the peptide) coat proteins[51]. It was discovered early on in phage biopanning that fusion to the pVIII reduces specificity of the isolated peptide sequence due to the selection of low affinity peptide binders that benefit from the increased avidity that results from high copy display [51]. In contrast, libraries displayed on pIII at 3–5 copies per phage generally yield peptide binders with higher affinity. Thus, commercial libraries are generally constructed with a large diversity (~109) of peptide sequences fused to the pIII coat protein of a filamentous M13 phage. The pIII display technology has been used to fuse antibody and small protein fragments [3, 5, 9]. There are a few examples of in vivo selection phage displaying antibody or protein fragments[52–55]. Other phage platforms include T7, T4 and λ bacteriophage; however, M13 phage is most commonly used due to its single coat fusion site, and ease of purification[51]. However, it has been demonstrated that the complex purification systems with T7- and T4-lytic phage allow for increased library diversity[56]. Moreover, T7 and T4-phage are small (~60 nm diameter) compared to M13 (~900 nm, length) and have been shown to have greater tissue penetration in vivo, enhancing delivery to M13-inaccessible sites such as the liver and brain [47, 57]. Lytic phage is also thought to have increased retention and reduced degradation in lysosomes, which might increase sequence yields in in vivo panning[56].. Despite these advantages the majority of in vivo panning studies have utilized recombinant, commercially available M13 peptide phage libraries.
2.2.2. Applications of in vivo phage display in humans
Initial in vivo biopanning efforts utilized phage libraries administered intravascularly and resulted in identification of a wide array of peptide “vascular zip codes” that target vasculature-specific ligands[42, 58] (Table 3). Further, peptide zip codes were shown to target agents directly to an organ or disease-associated vasculature[27, 59–66]. Following the success of biopanning in rodents, a similar phage-display method was applied in human biopanning screens. The first in vivo human panning experiment to map the human vasculature was conducted on a patient declared dead via brain-based determinations and results published in 2002 [67]. Subsequently, similar studies were performed with cancer patients whereby phage was recovered from biopsy specimens after intravenous administration of libraries [68, 69]. These later human studies recovered peptides recognizing targets commonly associated with tumor-related vascular targets such as interleukins, annexin, cathepsins, and prostaglandins[67–69]. Considering the vast amount of data generated from these early human vascular screens, one could suggest that it may be worth revisiting the human in vivo biopanning strategies to take full advantage of next-generation sequencing technologies and bioinformatics tools, as well as other routes of administration.
Table 3.
Vascular targeting ligand zip codes discovered by in vivo phage display.
| Target | Disease Target |
Sequence | Reference |
|---|---|---|---|
| Atherosclerosis | Vascular accessible atherosclerotic plaque | CLVEAYPGLSVRSC | Houston 2001 |
| Cancer Vasculature | Lung Cancer vasculature | SVSVGMKPSPRP | Lee 2007 |
| Obesity | Adipose vascular | CKGGRAKDC | Kolonin 2004 |
| Cancer Vasculature | Gastric Cancer Vasculature | CGNSNPKSC | Zhi 2004 |
| Atherosclerosis | Vascular smooth muscle cells | CNIWGVVLSWIGVFPEC | Michon 2002 |
| Cancer Vasculature | Prostate Cancer | IAGLATPGWSHWLAL | Newton 2006 |
| Cancer Vasculature | Breast Cancer | CDCRGDCFC | Arup 1998 |
| Cancer Vasculature | Pancreatic cancer | CRGRRST | Joyce 2003 |
| Cancer Vasculature | Squamous cell carcinoma. | CSRPRRSEC | Hoffman 2003 |
2.2.3. Limitations to in vivo phage display: tissue distribution and circulation half-life
As discussed, intravenous administration of M13 phage libraries in animals and humans led to the discovery and characterization of peptide zip codes that target organ-specific vasculature and tumor-binding peptides for drug delivery applications[27, 42, 58, 67]. Bacteriophage are inherently stable in biological milieus, a useful trait for productive in vivo selection. Still, in vivo phage-display biopanning methods are limited in the reach of the application to target sites outside of the vasculature or vasculature-leaky disease sites due to limited extravasation and penetration of the M13 phage. While alternative display technologies with smaller size such as covalent display technologies such as polyosome, ribosome or RNA-peptide display, could in theory distribute more broadly into tissues, these agents have not been successfully designed to withstand the harsh conditions of an intact biological system[70, 71]. The smaller T7 phage systems might reach more potential targets in vivo through increased accessibility, but trends in biopanning strategies (away from lytic phage) would suggest other process-related limitations complicate the use of T7- and T4-pahge.
In addition to the size-based tissue exclusion, M13 biopanning is limited by a relatively short circulation half-life (4 hrs), which is reduced significantly for recombinant M13-based libraries (<20 min), likely due to a combination of reduced in vivo stability, increased biological interactions, and increased internalization rate to cells[45, 72]. Furthermore, after internalization, M13 phage are degraded within lysosomes in 30 mins[45, 72]. T7-phage have a faster clearance, with half-lives less than 20 min for both wildtype and peptide-modified phage [46, 73]. Indeed, all phage are recognized readily by the immune system and sequestered within immunological relevant organs[46]. It is worth noting that in animals with a compromised immune system, phage exhibit an increased circulation potential, like due to the reduced recognition of phage through B-cell interactions [46, 72]. Nonetheless, engineering phage or other peptide display technologies to improve the circulation half-life and to limit degradation by mutating immunogenic properties of the macromolecular structure would result in a more robust platform for in vivo selection.
2.2.4. In vivo phage panning from alternative administration routes
As intravenous administration of phage libraries resulted in rich identification of binding motifs within the vasculature, biopanning from alternative administration routes would be expected to provide access to alternative targets. Indeed, non-systemic administration of phage libraries have yielded targeting ligands for a wide variety of diseases[29, 30, 44, 74–77](Table 4). Thus, the route of administration provides access to alternative targets that would normally be restricted by intravenous routes. For example, Wan et al., biopanned for phage that could transport to the brain after applying phage to the nasal epithelium of rats[74]. In a similar fashion, Sellers et al. administered phage by peripheral injection into the gastrocnemius, of mice, to identify ligands that targeted axonal import for delivery into the spinal cord 24 hours post-injection[44]. In these studies, peripheral administration with recovery at distal sites at time that eclipse several circulation half-lives (i.e. ≥ 1 hour), provides selection pressure for recombinant phage-sequences that demonstrate strong targeting potential.
Table 4.
Alternative paradigms for non-systemic biopanning strategies.
|
In vivo Phage Panning Delivery Mechanism |
Disease Target | Citation |
|---|---|---|
| Intranasal | Neurological Disorders | Wan 2009 |
| Intramuscular | Neurological Disorders | Sellers 2016 |
| Transdermal delivery | Diabetes | Chen 2006 |
| Transdermal delivery | Obesity | Lee 2011 |
| Intratracheal | Respiratory Disorders | Wu 2003 |
| Oral | Intestinal Disorders | Duerr 2004 |
| Intraperitoneal | Gastric metastases | Akita 2006 |
2.3. In vivo viral capsid panning
2.3.1. Adeno-associated virus (AAV) panning
Viral capsids are intrinsically robust particles well-suited to in vivo library selection. Viral particles exhibit considerable stability in vivo, have diameters on the order of nanometers allowing for broad circulation, are easily cloned and produced, and have naturally evolved to encapsulate genetic information to allow for high-throughput viral clone identification[15, 78–80]. In vivo viral capsid panning has primarily been performed with adeno-associated viruses (AAVs), which are icosahedral, nonenveloped viruses belonging to the Parvoviridae family. AAVs package single-stranded DNA and are 20–25 nm in diameter allowing for effective transport and delivery. Recombinant AAVs (rAAVs) are of particular interest as vehicles for gene therapy due to their favorable safety profiles[81]. Moreover, rAAVs can be engineered to eliminate most viral DNA, as well as to target certain tissues and cell types[82]. While much of biopanning has focused on AAVs, it is worth noting that CCMV, CPMV, and TMV are attractive viral particles because they do not naturally infect humans, their surfaces may be functionalized with peptides or small molecules, and their cargo may include imaging contrast agents or small molecule drugs[79, 83, 84]. Thus, future design of panning strategies might benefit from re-engineering these vector platforms.
Recombinant AAV particles with increased tissue tropisms can be engineered by generating highly diverse capsid libraries (>106 – 108 surface protein variants) and performing directed evolution in vivo to select for variants with desired biodistribution properties. There are several different approaches to generating viral capsid libraries as discussed below. As with phage display, in vivo AAV panning involves injecting the capsid library into laboratory animals and recovering the viral DNA from harvested tissues. Library variants that are recovered at a high frequency in tissues of interest compared to off-target tissues are enriched and re-injected for another round of in vivo panning. In addition to selecting for tissue tropism, rAAV biopanning has been used to discover clones that exhibit tissue-specific transduction by utilizing a Cre-based reporter to reveal tissue-specific increases in rAAV reporter protein expression[85–87]. In this strategy, rAAV particles carrying Cre-sensitive fluorescent reporter genes are subjected to in vivo selection in transgenic mice that express Cre recombinase in specific cell-types. A higher fluorescent signal correlates to increased transduction efficiency of a particular rAAV clone in a particular cell type. After multiple rounds of in vivo selection, rAAV variants with optimized characteristics including tissue tropism and transduction efficiency may be identified and further analyzed.
2.3.2. Directed evolution of randomly, semi-randomly, and rationally designed AAV libraries
Directed evolution of viral capsids for tissue targeting and transduction potency involves imposing selective pressures on large, random or semi-random capsid libraries[88]. This tool allows for rapid affinity and selectivity improvements, enhancing AAV-mediated delivery outcomes. One successful library approach utilizing this technique, has been to randomly shuffle Cap (capsid) genes from different AAV serotypes[89]. DNA shuffling has yielded rAAV particles with up to 13-fold increased gene transfer in heart muscle cells compared to preexisting AAVs, and particles with over 400-fold increased resistance to neutralizing antibodies[89, 90]. Libraries may also be generated in a semi-random fashion by targeted mutagenesis of Cap genes. For example, a rAAV with up to 15-fold increased transduction of astrocytes was developed by mutagenizing the capsid at loop regions performing in vivo library selection[91]. Peptide insertion, whereby peptides of random lengths and sequences are incorporated into the viral capsid surface, has also been used to engineer improved AAV tropism[16, 92]. Korbelin and colleagues recently used this library approach to engineer pulmonary tissue-specific rAAVs with high gene delivery efficiency[93]. Library diversity is a critical feature in increasing likelihood of successful selection. Thus, another approach to viral capsid panning is to utilize several different types of libraries in the same experiment. By combining four different libraries, a rAAV with 100-fold increased access to projection neurons compared to preexisting AAVs was engineered[85]. Future libraries may combine libraries multiplicatively rather than additively, allowing for even higher theoretical complexity. For example, after randomly shuffling Cap genes, random peptide insertions may be introduced into the same library.
Random library generation approaches yield many rAAV particles with reduced transduction efficiency or non-functional particles, reducing the effective library diversity. To address this limitation, rational design of viral capsid libraries can be performed utilizing known viral capsid structures and functional regions. The Schaffer group recently developed the SCHEMA library approach that was successfully used for in vivo panning[87]. In this approach, the SCHEMA algorithm[94] and recombination as a shortest path (RASPP) method[95] were applied to maximize both library diversity and structural conservation. After generating and screening a library of over 1.6 million rAAV variants, a novel variant was isolated that exhibited 60% transduction efficiency in adult neural stem cells, 24-fold higher GFP expression than AAV9, and an increased ability to evade neutralizing antibodies. This example highlights how rational modifications to libraries to selectively modify diversity can improve delivery and therapeutic outcomes and should be considered in in vivo panning design.
2.4. In vivo Aptamer Panning
2.4.1. Aptamers and aptamer selection
Nucleic acid aptamers are single-stranded oligonucleotides, typically 15–100 base pairs in length, that form three-dimensional structures capable of binding to targets with high specificity and affinity. Aptamer libraries used in biopanning generally contain 1013-1015 unique sequences, consisting of a 20–60 base pair random region flanked by fixed primer regions on the 5’ and 3’ ends. From this starting pool, DNA or RNA aptamers are selected through the iterative process of Systematic Evolution of Ligands by Exponential Enrichment (SELEX), involving cycles of incubation with the target and subsequent amplification and recovery of binding sequences[2, 96]. With successive cycles, high affinity aptamers are enriched until they dominate the library population[14, 36, 97]. The population of aptamers present after selection can be characterized by next generation sequencing (NGS), and individual aptamers can then be synthesized and investigated for binding behavior.
Aptamer ligands offer several advantages over conventional targeting peptides or antibodies. Aptamers can form a variety of complex secondary structures with comparable or superior binding affinity of antibodies (micromolar to picomolar), and they can bind to a wide breadth of targets, such as ions, small molecules, and proteins. Since aptamers are nucleic-based, they exhibit reduced immunogenicity, increased tissue penetration, facile manufacturing, and high shelf-life stability compared to peptides and proteins. Thus, aptamers are an attractive class of targeting agents for drug delivery. However, aptamers are highly susceptible to nuclease degradation and kidney filtration, resulting in limited circulation time and half-life in vivo. Multiple studies have demonstrate that these challenges can be mitigated by engineering chemical modification into the aptamer backbone or side groups, non-natural nucleotide substitutions, or end capping, and reducing susceptibility to endo- and exo-nucleases[98, 99], which have aided the development of in vivo aptamer panning strategies[36].
2.4.2. Library design for in vivo SELEX
There are several considerations involved in the design aptamer libraries for in vivo SELEX, including nucleotide type, library length, chemical modification, and length. Aptamer library lengths are typically between 20–80 base pair, while aptamers short or long random regions can successfully bind, those with longer random regions exhibit greater structure complexity[100] and increased library diversity at the expense of cost for production. Both RNA and DNA aptamer libraries have yielded binding sequences from in vivo panning. RNA libraries tend to increase library diversity and improve panning outcomes but usually at the expensive of stability. Aptamers based on RNA, which is naturally single-stranded have a greater range of three dimensional structures compared with DNA, which can result in a higher binding-affinity [101]. However, RNA aptamers are also more expensive and require an extra step of reverse transcription prior to amplification.
2.4.3. Recent progress in in vivo SELEX
For drug delivery applications, in vivo biopanning has an inherent advantage over in vitro panning by providing the stringent degradation, transport, and complex competitive binding pressures present in vivo during the selection process. In vitro SELEX has been performed successfully against purified target proteins or even whole cells; however, these in vitro selected aptamers may fail to bind to the same target in vivo. This may be attributed to differences in selection conditions, as aptamer conformation and binding has been demonstrated to depend on pH, temperature and ionic conditions[102–105]. To address this shortcoming, in vivo SELEX can be performed in living animal models to identify binding constructs in the context of a representative disease environment.
Mi et al. published the first RNA aptamer identified via in vivo SELEX in 2010[106]. In this work, a random library of 2’-fluoropyrimidine modified RNA sequences was injected intravenously into hepatic colorectal cancer-bearing mice. This chemical modification increases aptamer stability through increased nuclease resistance, thus increasing potential target binding by enhancing aptamer circulation time. Fourteen cycles of SELEX were performed and an aptamer that bound to p68, an RNA helicase, with nanomolar affinity was identified[14, 106]. Since this original publication, this process has been adapted for use with human intrahepatic xenografts to identify RNA aptamers against human helicase DHX9[107].
In vivo SELEX has also been used to evolve aptamers capable of penetrating the blood brain barrier (BBB). Penetration of therapies across the BBB remains a challenge due to tight cell junctions and highly selective transport of molecules. Cheng et al. injected intravenously a 2’-fluoropyrimidine modified RNA library to identify an aptamer that could penetrate the BBB after being, one of which represented 50% of all sequences. Further characterization of the most highly enriched aptamer from round 22 revealed that it internalized by brain-vascular endothelial cells. The authors did note high levels of RNA aptamer sequences in clearance tissues (liver and kidney) which was attributed to the reticuloendothelial system. Additionally, since the in vivo SELEX strategy required over 20 rounds of selection, the authors recommended a combinatorial in vitro and in vivo selection if screening for specific disease phenotypes[108].
Library stability and circulation time can drastically affect selection efficiency. In vivo SELEX was carried out successfully using a polyethylene glycol (PEG)-modified 2’fluoropyrimidine RNA library to identify an aptamer that binds to and inhibits proliferation of non-small-cell lung cancer (NSCLC) cells[109]. Libraries were conjugated with PEG in order to increase aptamer circulation time and reduce renal clearance by increasing molecular weight. A binding aptamer was emerged at cycle 8 of selection and accounted for 90% of the enriched RNA by cycle 11, thus requiring much fewer rounds of selection compared with other in vivo SELEX reports[106, 108]. The authors attributed this early emergence to the addition of PEG within the RNA pool and the increased circulation time, which eliminated non-specific binding[109]. This aptamer was further used to facilitate delivery of a chemotherapeutic drug. In another recent example, Shen and coworkers used a DNA thiophosphate-backbone-substituted[110] library in vivo SELEX in a breast cancer bone metastasis model to identify a DNA aptamer that accumulates in the tumor microenvironment[111]. After 10 rounds of selection from tumor tissue, the top occurring aptamer was tested and shown to bind to cancer and myeloid-derived tumor suppressor cells. Furthermore, aptamer-conjugated liposomal doxorubicin formulations were effective in inhibiting tumor growth compared to non-targeted liposomes[36, 106].
For successful in vivo aptamer panning, it is critical that aptamer libraries are able to infiltrate the target area for selection, extraction, and amplification. However, unmodified aptamers are highly susceptible to nuclease degradation, resulting in a short half-life and circulation time in vivo. Additionally, their small size contributes to their rapid renal filtration. To prevent degradation, aptamer libraries have been modified chemically to replace the 2’ position with a fluoro (F), amino (NH2), or O-methyl (OCH3) group, and by capping the 3’ end with inverted thymidine[99, 112]. While chemical modifications can impart improved stability and circulation, chemical modification to the aptamer backbone or side-chains can affect aptamer folding and binding. Nonetheless, these changes can been utilized in-SELEX or post-SELEX biopanning strategies. For the in-SELEX biopanning, sequences with the chosen modifications are used in the starting DNA or RNA library and undergo subsequent rounds of selection. However, modifications are limited to 2’-aminopyrimides, 2’-fluoropyrimidines, 2’-O-methyl nucleotides, and locked nucleic acids (LNA). Mi, Cheng, and Wang all utilized in-SELEX modifications for their in vivo panning strategies[107–109, 111]. Recently, SOMAmer (slow off-rate modificed aptamer) reagents have emerged. They contain modified nucleotides that have conjugated functional moieties, creating stable structures with high nuclease resistance and binding affinities[113]. For the post-SELEX strategy, chosen modifications are implemented within pre-selected aptamers at specific locations during chemical synthesis. However, these modifications can affect aptamer structure and folding and thus impact its binding behavior.
Additionally, unmodified aptamers are within the size range for renal excretion and are thus rapidly excreted by the kidneys. To mitigate this, aptamers are often modified by the addition of a bulky side group, such as PEG or cholesterol. This strategy increased the plasma circulation half-life of Macugen, an aptamer used to treat macular degeneration, from 9 hours to 12 hours[99]. Wang utilized a combinatorial approach of 2’-fluoropyrimide and PEGylation in order to increase in vivo stability[109]. Aptamers can also be multimerized to create multivalent constructs above the renal molecular weight threshold. Borbas et al. designed a tetrameric aptamer against the MUC1 tumor marker. This construct had improved stability, tumor-retention, and pharmacokinetic properties compared to the monovalent construct[114]. As such, a library particle system displaying multiple copies of each aptamer could be another approach for in vivo aptamer panning.
3. Future of panning
In vivo panning for novel ligands has enormous potential for targeted drug delivery. The platform inherently allows for simultaneous selection of molecules that are able to withstand harsh biological environments, and home to targets in a complex and heterogeneous millieu. However, the design of successful in vivo biopanning requires consideration of multiple design principles. Success in panning is dependent on both the diversity and stability of the library. Here we will discuss ways to both enhance these parameters as well as manipulate panning procedures to artificially augment current panning systems.
3.1. Increasing in vivo library stability
Library stability is necessary to recover bound ligands. Maintaining the stability of a library in a harsh biological environment has challenges and in vivo panning requires library exposure to multiple biological milieu, potentially impacting the diversity of the final library at the target site. Novel targets might be lost as a result of degradation. Ultimately the perfect library display platform would be engineered to withstand harsh biological milieu. The conditions a display platform must withstand during biological transport include enzymatic degradation, harsh shear force, dramatic pH changes including acidic endosome/lysosomal conditions following platform internalization into cells, etc. Some groups have begun to address some these challenges in in vitro panning systems. Rangel et al. incorporated a cell penetrating peptide, penetratin, on the on the pVIII major coat protein of M13 phage, enhancing intracellular release for subcellular organelle ligand selection[39]. It’s possible strategies like these could be applied in vivo, allowing the library platform to avoid lysosomal degradation and improve ligand recovery. However, modulation to increase lysosomal stability likely does not eliminate loss of phage simply due to inherent physical transport limitations. Overcoming transport challenges is likely overcome through panning design.
3.1.1. Design of clever panning strategies to improve library recovery.
In order to increase the likelihood of completing a successful in vivo biopanning, careful consideration of the inherent biology of the target and/or disease state must be given to each strategy. The most successful targeting ligands have been discovered by in vivo panning that carefully considered the inherent disease state and a specific route of administration that would allow for increased exposure to the target. Incorporation of the disease state into the panning strategy inherently increases affinity and specificity of a ligand, through the incorporation of heterogeneity of diseased and “normal” target sites. Thoughtful routes of administration to exploit routes of delivery to disease tissue or the intended target could improve ligand selection through enhanced ligand presentation, by decreasing library degradation during transport to the target site, or by improving negative selection by passage through competitive non-specific binding targets. Such examples include Sellers et al. recent neurological panning strategy where we were able to harness biological transport within the peripheral nervous system from muscles to the spinal cord[44]. Through intramuscular injections we were able to isolate and validate a novel peptide that helps to engage this transport pathway (Figure 2). This study highlights how biological processes can be integrated into panning design strategies. While strategies like these allow for increased target exposure it does not enhance the diversity of the starting library nor does it improve ligand affinity.
Figure 2.

Improve panning strategies to take advantage of inherent biology. Here, Sellers et al. capitalized on the transport from muscles to the spinal cord to bypass the blood brain barrier [44]
3.2. Increased in vivo library diversity
In vivo panning success achieved when a ligand with high affinity and specificity to a target site is discovered. While panning strategies can be modified to improve outcomes, in general increasing the number of panned ligands will inherently increase the statistical probability of a hit. As shown in Table 2, current in vivo display platforms have significant differences in absolute diversity. Many researchers have probed these platforms in vitro to increase diversity through synthetic modification, such as artificial nucleic or amino acids, as well as, the introduction of chemical functionality. Such examples include SOMAmers, which add non-natural functional groups to aptamers to more closely mimic protein interactions, and one-bead-one-compound libraries for peptide selection which can include non-natural amino acids which can reverse chirality[113, 115]}. These platforms have yielded high affinity motifs not necessarily found in nature, and can be combined with natural ligand libraries to improve diversity. These diversification techniques have been widely applied to in vitro selection but have not been either optimized or applied for in vivo panning. Increasing the synthetic tool sets could dramatically enhance in vivo panning target identification and outcome. These days, panning products are often assessed through next generation sequencing methods where ligand hits from each panning round are screened for sequence similarities and alignment[11, 116, 117]. After even the first round of in vivo panning, clear trends generally emerge within this analysis. This pattern recognition can be reinvested into subsequent rounds of panning to enhance library affinity, specifying diversity, a technique generally termed directed evolution.
3.2.1. Directed evolution to drive improved library hits.
Directed evolution techniques have been employed in viral and phage display in an in vitro context[5, 87]. These platforms significantly enhance specificity and affinity of ligands identified. However the leap has not yet made a dramatic impact on in vivo panning[5, 38, 87, 116, 118]. In directed evolution, following rounds of selection, patterns emerge. These motifs can be incorporated into subsequent panning rounds and affinity can be honed utilizing a deconvolution approach. The libraries can be generated either through iterative single positional scanning and/or multi-site mutational sublibrary scanning[51]. In both strategies, motif modifications are generally accomplished through site-specific mutagenesis or through random generation whereby motifs are fragmented and reassembled[51]. When individuals are refining library design for subsequent panning rounds, it might also be useful to scan bioinformatic databases to see if the motif identified have sequence homology with known native ligand binders. This could be particularly useful in phage display technology as sequence specificity of clinically translated sequences have been shown through these techniques to hone to three amino acid positions [68, 119, 120]. The beauty of panning is that multiple mutations can be generated in several libraries and be explored in a parallel fashion such that non-specific binders are rapidly removed and affinity honed simultaneously (Figure 3). Unlike in vitro selection, incorporating this technique into in vivo selection allows for refinements to be made within the context of the real world application reducing time later for application optimization.
Figure 3.

Directed evolution to improve affinity and specificity. Here, high diversity libraries are injected in vivo, selected and amplified. Selected variants are used to develop re-diversified libraries via fragmentation, reassembly and mutation. These libraries are used to improve affinity in subsequent panning rounds.
3.3. Novel display platforms.
The incorporation of both diversity and stability into a single, easy to use platform would be beneficial. The reason the vast majority of in vivo panning literature has used the M13 platform is because of its experimental ease of use. New protein and viral based platform technologies are also arising on the in vivo panning horizon. Synthetic nucleocapsids that were recently developed by the Baker and King groups might also be applied to in vivo panning[121]. These synthetic protein nanocages encapsulate their own genomes and are of comparable size to AAVs, allowing high-throughput production, broad circulation in vivo, and nucleic acid sequence-based identification. Furthermore, directed evolution strategies could be easily applied to this technology. Current generation synthetic nucleocapsids possess circulation half-lives comparable to M13 bacteriophage and have been engineered to display peptides for tagging and purification. With diameters < 30 nm, synthetic nucleocapsids are therefore an attractive future platform for in vivo panning.
4. Conclusions
In vivo panning has been relatively unexplored relative to in vitro panning since its inception in 1996. However, sequences identified by in vivo panning have moved quickly toward clinical use. The most well-known recovered in vivo panned sequences RGD[27] and NGR[122] have multiple on-going clinical trials both alone and in combination with therapeutics/diagnostics (Table 1) [123–125]. Advancements in in vivo panning strategies, through clever selection design, increased in vivo stability, and innovative directed evolution strategies, will launch our discoveries and improve delivery into the next 20 years
Table 5:
In vivo SELEX
| Aptamer library | Disease Model |
Target | Affinity | Library Modification |
Random Region (bp) |
Library Circulation Time |
|---|---|---|---|---|---|---|
| RNA (Mi 2010, In vivo selection of tumor-targeting RNA Motifs) | Colorectal cancer | p68 RNA helicase | 13.8 nM | 2’-fluropyrimidine modified RNA | 40 | 20 minutes |
| RNA (Mi 2016, In vivo selection against human colorectal cancer xenografts identifies an aptamer that targets RNA helicase protein DHX9) | Colorectal cancer | DHX9 RNA helicase | 2’-fluropyrimidine modified RNA | 40 | 20–30 minutes | |
| RNA (Cheng 2010, In vivo SELEX for Identification of Brain-penetrating aptamers) | CNS delivery | Brain endothelial cells | 2’-fluropyrimidine modified RNA | 40 | 1–3 hours | |
| RNA (Wang 2018, In vivo SELEX of an inhibitory NSCLC-specific RNA aptamer from PEGylated RNA library) | NSCLC | NSCLC cells | 9 nM | 2’-fluropyrimidine modified RNA and PEG modification | 40 | 6 hours |
| DNA (Liu 2018, A novel DNA aptamer for dual targeting of polymorphonuclear myeloid-derived suppressor cells and tumor cells) | Breast cancer | Breast cancer cells + myeloid derived suppressor cells | 2.47 nM | Thiophosphate backbone substitution | 30 | 4 hours |
5. Acknowledgements.
The work was supported by NIH 1R01CA177272, 1R21NS099654, 1R21NS086500 and 1R01HL139007. HHG was supported by the Cardiovascular Pathology Training Grant (5 T32 HL 007312-37).
Abbreviations
- (AAVs)
Adeno-associated viruses
- (RASPP)
Recombination as a shortest path
- (SELEX)
Systematic Evolution of Ligands by Exponential Enrichment
- (NGS)
Next generation sequencing
- (BBB)
Blood brain barrier
- (SOMAmer)
Slow off-rate modified aptamer
6. References.
- [1].Smith GP, Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface, Science, 228 (1985) 1315–1317. [DOI] [PubMed] [Google Scholar]
- [2].Ellington AD, Szostak JW, In vitro selection of RNA molecules that bind specific ligands, Nature, 346 (1990) 818–822. [DOI] [PubMed] [Google Scholar]
- [3].McCafferty J, Griffiths AD, Winter G, Chiswell DJ, Phage antibodies: filamentous phage displaying antibody variable domains, Nature, 348 (1990) 552–554. [DOI] [PubMed] [Google Scholar]
- [4].Winter G, Griffiths AD, Hawkins RE, Hoogenboom HR, Making antibodies by phage display technology, Annual review of immunology, 12 (1994) 433–455. [DOI] [PubMed] [Google Scholar]
- [5].Esvelt KM, Carlson JC, Liu DR, A system for the continuous directed evolution of biomolecules, Nature, 472 (2011) 499–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Jackson DA, Symons RH, Berg P, Biochemical method for inserting new genetic information into DNA of Simian Virus 40: circular SV40 DNA molecules containing lambda phage genes and the galactose operon of Escherichia coli, Proceedings of the National Academy of Sciences of the United States of America, 69 (1972) 2904–2909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Sanger F, Air GM, Barrell BG, Brown NL, Coulson AR, Fiddes CA, Hutchison CA, Slocombe PM, Smith M, Nucleotide sequence of bacteriophage phi X174 DNA, Nature, 265 (1977) 687–695. [DOI] [PubMed] [Google Scholar]
- [8].Scott JK, Smith GP, Searching for peptide ligands with an epitope library, Science, 249 (1990) 386–390. [DOI] [PubMed] [Google Scholar]
- [9].Clackson T, Hoogenboom HR, Griffiths AD, Winter G, Making antibody fragments using phage display libraries, Nature, 352 (1991) 624–628. [DOI] [PubMed] [Google Scholar]
- [10].Hoogenboom HR, Griffiths AD, Johnson KS, Chiswell DJ, Hudson P, Winter G, Multi-subunit proteins on the surface of filamentous phage: methodologies for displaying antibody (Fab) heavy and light chains, Nucleic acids research, 19 (1991) 4133–4137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Liu GW, Livesay BR, Kacherovsky NA, Cieslewicz M, Lutz E, Waalkes A, Jensen MC, Salipante SJ, Pun SH, Efficient Identification of Murine M2 Macrophage Peptide Targeting Ligands by Phage Display and Next-Generation Sequencing, Bioconjugate chemistry, 26 (2015) 1811–1817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Rouet R, Jackson KJL, Langley DB, Christ D, Next-Generation Sequencing of Antibody Display Repertoires, Frontiers in immunology, 9 (2018) 118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [13].Yang W, Yoon A, Lee S, Kim S, Han J, Chung J, Next-generation sequencing enables the discovery of more diverse positive clones from a phage-displayed antibody library, Experimental & molecular medicine, 49 (2017) e308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Zhou J, Rossi J, Aptamers as targeted therapeutics: current potential and challenges, Nature reviews. Drug discovery, 16 (2017) 181–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Grimm D, Buning H, Small But Increasingly Mighty: Latest Advances in AAV Vector Research, Design, and Evolution, Human gene therapy, 28 (2017) 1075–1086. [DOI] [PubMed] [Google Scholar]
- [16].Korbelin J, Trepel M, How to Successfully Screen Random Adeno-Associated Virus Display Peptide Libraries In Vivo, Human gene therapy methods, 28 (2017) 109–123. [DOI] [PubMed] [Google Scholar]
- [17].Yoo JW, Irvine DJ, Discher DE, Mitragotri S, Bio-inspired, bioengineered and biomimetic drug delivery carriers, Nature reviews. Drug discovery, 10 (2011) 521–535. [DOI] [PubMed] [Google Scholar]
- [18].Byrne JD, Betancourt T, Brannon-Peppas L, Active targeting schemes for nanoparticle systems in cancer therapeutics, Advanced drug delivery reviews, 60 (2008) 1615–1626. [DOI] [PubMed] [Google Scholar]
- [19].Lambert JM, Morris CQ, Antibody-Drug Conjugates (ADCs) for Personalized Treatment of Solid Tumors: A Review, Advances in therapy, 34 (2017) 1015–1035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Bertrand N, Wu J, Xu X, Kamaly N, Farokhzad OC, Cancer nanotechnology: the impact of passive and active targeting in the era of modern cancer biology, Advanced drug delivery reviews, 66 (2014) 2–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Rabanel JM, Aoun V, Elkin I, Mokhtar M, Hildgen P, Drug-loaded nanocarriers: passive targeting and crossing of biological barriers, Current medicinal chemistry, 19 (2012) 3070–3102. [DOI] [PubMed] [Google Scholar]
- [22].Sievers EL, Senter PD, Antibody-drug conjugates in cancer therapy, Annual review of medicine, 64 (2013) 15–29. [DOI] [PubMed] [Google Scholar]
- [23].Ruoslahti E, Tumor penetrating peptides for improved drug delivery, Advanced drug delivery reviews, 110–111 (2017) 3–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Choi J, Credit K, Henderson K, Deverkadra R, He Z, Wiig H, Vanpelt H, Flessner MF, Intraperitoneal immunotherapy for metastatic ovarian carcinoma: Resistance of intratumoral collagen to antibody penetration, Clinical cancer research : an official journal of the American Association for Cancer Research, 12 (2006) 1906–1912. [DOI] [PubMed] [Google Scholar]
- [25].Jain RK, Physiological barriers to delivery of monoclonal antibodies and other macromolecules in tumors, Cancer research, 50 (1990) 814s–819s. [PubMed] [Google Scholar]
- [26].Thurber GM, Schmidt MM, Wittrup KD, Antibody tumor penetration: transport opposed by systemic and antigen-mediated clearance, Advanced drug delivery reviews, 60 (2008) 1421–1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Arap W, Pasqualini R, Ruoslahti E, Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model, Science, 279 (1998) 377–380. [DOI] [PubMed] [Google Scholar]
- [28].Hsu T, Mitragotri S, Delivery of siRNA and other macromolecules into skin and cells using a peptide enhancer, Proceedings of the National Academy of Sciences of the United States of America, 108 (2011) 15816–15821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Duerr DM, White SJ, Schluesener HJ, Identification of peptide sequences that induce the transport of phage across the gastrointestinal mucosal barrier, J Virol Methods, 116 (2004) 177–180. [DOI] [PubMed] [Google Scholar]
- [30].Lee NK, Kim HS, Kim KH, Kim EB, Cho CS, Kang SK, Choi YJ, Identification of a novel peptide ligand targeting visceral adipose tissue via transdermal route by in vivo phage display, J Drug Target, 19 (2011) 805–813. [DOI] [PubMed] [Google Scholar]
- [31].CAPRA JD, Janeway CA, Travers P, Walport M, Inmunobiology: the inmune system in health and disease, Garland Publishing; 1999. [Google Scholar]
- [32].Dunn-Walters DK, Ademokun AA, B cell repertoire and ageing, Current opinion in immunology, 22 (2010) 514–520. [DOI] [PubMed] [Google Scholar]
- [33].Yao VJ, D’Angelo S, Butler KS, Theron C, Smith TL, Marchio S, Gelovani JG, Sidman RL, Dobroff AS, Brinker CJ, Bradbury ARM, Arap W, Pasqualini R, Ligand-targeted theranostic nanomedicines against cancer, Journal of controlled release : official journal of the Controlled Release Society, 240 (2016) 267–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Martins IM, Reis RL, Azevedo HS, Phage Display Technology in Biomaterials Engineering: Progress and Opportunities for Applications in Regenerative Medicine, ACS Chem Biol, 11 (2016) 2962–2980. [DOI] [PubMed] [Google Scholar]
- [35].Giordano RJ, Cardó-Vila M, Lahdenranta J, Pasqualini R, Arap W, Biopanning and rapid analysis of selective interactive ligands, Nature medicine, 7 (2001) 1249. [DOI] [PubMed] [Google Scholar]
- [36].Wu YX, Kwon YJ, Aptamers: The “evolution” of SELEX, Methods, 106 (2016) 21–28. [DOI] [PubMed] [Google Scholar]
- [37].Cieslewicz M, Tang J, Yu JL, Cao H, Zavaljevski M, Motoyama K, Lieber A, Raines EW, Pun SH, Targeted delivery of proapoptotic peptides to tumor-associated macrophages improves survival, Proceedings of the National Academy of Sciences of the United States of America, 110 (2013) 15919–15924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Hawkins RE, Russell SJ, Winter G, Selection of phage antibodies by binding affinity. Mimicking affinity maturation, J Mol Biol, 226 (1992) 889–896. [DOI] [PubMed] [Google Scholar]
- [39].Rangel R, Dobroff AS, Guzman-Rojas L, Salmeron CC, Gelovani JG, Sidman RL, Pasqualini R, Arap W, Targeting mammalian organelles with internalizing phage (iPhage) libraries, Nat Protoc, 8 (2013) 1916–1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Sefah K, Shangguan D, Xiong X, O’Donoghue MB, Tan W, Development of DNA aptamers using Cell-SELEX, Nat Protoc, 5 (2010) 1169–1185. [DOI] [PubMed] [Google Scholar]
- [41].Ibsen KN, Daugherty PS, Prediction of antibody structural epitopes via random peptide library screening and next generation sequencing, Journal of immunological methods, 451 (2017) 28–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Pasqualini R, Ruoslahti E, Organ targeting in vivo using phage display peptide libraries, Nature, 380 (1996) 364–366. [DOI] [PubMed] [Google Scholar]
- [43].Pasqualini R, Koivunen E, Ruoslahti E, Alpha v integrins as receptors for tumor targeting by circulating ligands, Nature biotechnology, 15 (1997) 542–546. [DOI] [PubMed] [Google Scholar]
- [44].Sellers DL, Bergen JM, Johnson RN, Back H, Ravits JM, Horner PJ, Pun SH, Targeted axonal import (TAxI) peptide delivers functional proteins into spinal cord motor neurons after peripheral administration, Proceedings of the National Academy of Sciences of the United States of America, 113 (2016) 2514–2519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Molenaar TJ, Michon I, de Haas SA, van Berkel TJ, Kuiper J, Biessen EA, Uptake and processing of modified bacteriophage M13 in mice: implications for phage display, Virology, 293 (2002) 182–191. [DOI] [PubMed] [Google Scholar]
- [46].Srivastava AS, Kaido T, Carrier E, Immunological factors that affect the in vivo fate of T7 phage in the mouse, J Virol Methods, 115 (2004) 99–104. [DOI] [PubMed] [Google Scholar]
- [47].Ludtke JJ, Sololoff AV, Wong SC, Zhang G, Wolff JA, In vivo selection and validation of liver-specific ligands using a new T7 phage peptide display system, Drug Deliv, 14 (2007) 357–369. [DOI] [PubMed] [Google Scholar]
- [48].Rohloff JC, Gelinas AD, Jarvis TC, Ochsner UA, Schneider DJ, Gold L, Janjic N, Nucleic Acid Ligands With Protein-like Side Chains: Modified Aptamers and Their Use as Diagnostic and Therapeutic Agents, Molecular therapy. Nucleic acids, 3 (2014) e201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Ying Y, Muller OJ, Goehringer C, Leuchs B, Trepel M, Katus HA, Kleinschmidt JA, Heart-targeted adeno-associated viral vectors selected by in vivo biopanning of a random viral display peptide library, Gene therapy, 17 (2010) 980–990. [DOI] [PubMed] [Google Scholar]
- [50].Smith GP, Petrenko VA, Phage Display, Chemical reviews, 97 (1997) 391–410. [DOI] [PubMed] [Google Scholar]
- [51].Falciani C, Lozzi L, Pini A, Bracci L, Bioactive peptides from libraries, Chem Biol, 12 (2005) 417–426. [DOI] [PubMed] [Google Scholar]
- [52].Hoogenboom HR, Overview of antibody phage-display technology and its applications, Methods in molecular biology, 178 (2002) 1–37. [DOI] [PubMed] [Google Scholar]
- [53].Johns M, George AJ, Ritter MA, In vivo selection of sFv from phage display libraries, Journal of immunological methods, 239 (2000) 137–151. [DOI] [PubMed] [Google Scholar]
- [54].Deramchia K, Jacobin-Valat MJ, Vallet A, Bazin H, Santarelli X, Sanchez S, Dos Santos P, Franconi JM, Claverol S, Bonetto S, Clofent-Sanchez G, In vivo phage display to identify new human antibody fragments homing to atherosclerotic endothelial and subendothelial tissues [corrected], The American journal of pathology, 180 (2012) 2576–2589. [DOI] [PubMed] [Google Scholar]
- [55].Valadon P, Garnett JD, Testa JE, Bauerle M, Oh P, Schnitzer JE, Screening phage display libraries for organ-specific vascular immunotargeting in vivo, Proceedings of the National Academy of Sciences of the United States of America, 103 (2006) 407–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Deng X, Wang L, You X, Dai P, Zeng Y, Advances in the T7 phage display system (Review), Molecular medicine reports, 17 (2018) 714–720. [DOI] [PubMed] [Google Scholar]
- [57].Urich E, Schmucki R, Ruderisch N, Kitas E, Certa U, Jacobsen H, Schweitzer C, Bergadano A, Ebeling M, Loetscher H, Freskgard PO, Cargo Delivery into the Brain by in vivo identified Transport Peptides, Scientific reports, 5 (2015) 14104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Pasqualini R, Ruoslahti E, Tissue targeting with phage peptide libraries, Mol Psychiatry, 1 (1996) 423. [PubMed] [Google Scholar]
- [59].Houston P, Goodman J, Lewis A, Campbell CJ, Braddock M, Homing markers for atherosclerosis: applications for drug delivery, gene delivery and vascular imaging, FEBS letters, 492 (2001) 73–77. [DOI] [PubMed] [Google Scholar]
- [60].Michon IN, Hauer AD, von der Thusen JH, Molenaar TJ, van Berkel TJ, Biessen EA, Kuiper J, Targeting of peptides to restenotic vascular smooth muscle cells using phage display in vitro and in vivo, Biochimica et biophysica acta, 1591 (2002) 87–97. [DOI] [PubMed] [Google Scholar]
- [61].Kolonin MG, Saha PK, Chan L, Pasqualini R, Arap W, Reversal of obesity by targeted ablation of adipose tissue, Nature medicine, 10 (2004) 625–632. [DOI] [PubMed] [Google Scholar]
- [62].Zhi M, Wu KC, Dong L, Hao ZM, Deng TZ, Hong L, Liang SH, Zhao PT, Qiao TD, Wang Y, Xu X, Fan DM, Characterization of a specific phage-displayed Peptide binding to vasculature of human gastric cancer, Cancer biology & therapy, 3 (2004) 1232–1235. [DOI] [PubMed] [Google Scholar]
- [63].Lee TY, Lin CT, Kuo SY, Chang DK, Wu HC, Peptide-mediated targeting to tumor blood vessels of lung cancer for drug delivery, Cancer research, 67 (2007) 10958–10965. [DOI] [PubMed] [Google Scholar]
- [64].Newton JR, Kelly KA, Mahmood U, Weissleder R, Deutscher SL, In vivo selection of phage for the optical imaging of PC-3 human prostate carcinoma in mice, Neoplasia, 8 (2006) 772–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Joyce JA, Laakkonen P, Bernasconi M, Bergers G, Ruoslahti E, Hanahan D, Stage-specific vascular markers revealed by phage display in a mouse model of pancreatic islet tumorigenesis, Cancer cell, 4 (2003) 393–403. [DOI] [PubMed] [Google Scholar]
- [66].Hoffman JA, Giraudo E, Singh M, Zhang L, Inoue M, Porkka K, Hanahan D, Ruoslahti E, Progressive vascular changes in a transgenic mouse model of squamous cell carcinoma, Cancer cell, 4 (2003) 383–391. [DOI] [PubMed] [Google Scholar]
- [67].Arap W, Kolonin MG, Trepel M, Lahdenranta J, Cardo-Vila M, Giordano RJ, Mintz PJ, Ardelt PU, Yao VJ, Vidal CI, Chen L, Flamm A, Valtanen H, Weavind LM, Hicks ME, Pollock RE, Botz GH, Bucana CD, Koivunen E, Cahill D, Troncoso P, Baggerly KA, Pentz RD, Do KA, Logothetis CJ, Pasqualini R, Steps toward mapping the human vasculature by phage display, Nature medicine, 8 (2002) 121–127. [DOI] [PubMed] [Google Scholar]
- [68].Staquicini FI, Cardo-Vila M, Kolonin MG, Trepel M, Edwards JK, Nunes DN, Sergeeva A, Efstathiou E, Sun J, Almeida NF, Tu SM, Botz GH, Wallace MJ, O’Connell DJ, Krajewski S, Gershenwald JE, Molldrem JJ, Flamm AL, Koivunen E, Pentz RD, Dias-Neto E, Setubal JC, Cahill DJ, Troncoso P, Do KA, Logothetis CJ, Sidman RL, Pasqualini R, Arap W, Vascular ligand-receptor mapping by direct combinatorial selection in cancer patients, Proceedings of the National Academy of Sciences of the United States of America, 108 (2011) 18637–18642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Krag DN, Shukla GS, Shen GP, Pero S, Ashikaga T, Fuller S, Weaver DL, Burdette-Radoux S, Thomas C, Selection of tumor-binding ligands in cancer patients with phage display libraries, Cancer research, 66 (2006) 7724–7733. [DOI] [PubMed] [Google Scholar]
- [70].FitzGerald K, In vitro display technologies - new tools for drug discovery, Drug Discov Today, 5 (2000) 253–258. [DOI] [PubMed] [Google Scholar]
- [71].Zahnd C, Amstutz P, Pluckthun A, Ribosome display: selecting and evolving proteins in vitro that specifically bind to a target, Nature methods, 4 (2007) 269–279. [DOI] [PubMed] [Google Scholar]
- [72].Zou J, Dickerson MT, Owen NK, Landon LA, Deutscher SL, Biodistribution of filamentous phage peptide libraries in mice, Mol Biol Rep, 31 (2004) 121–129. [DOI] [PubMed] [Google Scholar]
- [73].Srivastava AS, Chauhan DP, Carrier E, In utero detection of T7 phage after systemic administration to pregnant mice, Biotechniques, 37 (2004) 81–83. [DOI] [PubMed] [Google Scholar]
- [74].Wan XM, Chen YP, Xu WR, Yang WJ, Wen LP, Identification of nose-to-brain homing peptide through phage display, Peptides, 30 (2009) 343–350. [DOI] [PubMed] [Google Scholar]
- [75].Chen Y, Shen Y, Guo X, Zhang C, Yang W, Ma M, Liu S, Zhang M, Wen LP, Transdermal protein delivery by a coadministered peptide identified via phage display, Nature biotechnology, 24 (2006) 455–460. [DOI] [PubMed] [Google Scholar]
- [76].Wu M, Pasula R, Smith PA, Martin WJ 2nd, Mapping alveolar binding sites in vivo using phage peptide libraries, Gene therapy, 10 (2003) 1429–1436. [DOI] [PubMed] [Google Scholar]
- [77].Akita N, Maruta F, Seymour LW, Kerr DJ, Parker AL, Asai T, Oku N, Nakayama J, Miyagawa S, Identification of oligopeptides binding to peritoneal tumors of gastric cancer, Cancer science, 97 (2006) 1075–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [78].Singh P, Gonzalez MJ, Manchester M, Viruses and their uses in nanotechnology, Drug development research, 67 (2006) 23–41. [Google Scholar]
- [79].Manchester M, Singh P, Virus-based nanoparticles (VNPs): platform technologies for diagnostic imaging, Advanced drug delivery reviews, 58 (2006) 1505–1522. [DOI] [PubMed] [Google Scholar]
- [80].Paulk NK, Pekrun K, Zhu E, Nygaard S, Li B, Xu J, Chu K, Leborgne C, Dane AP, Haft A, Zhang Y, Zhang F, Morton C, Valentine MB, Davidoff AM, Nathwani AC, Mingozzi F, Grompe M, Alexander IE, Lisowski L, Kay MA, Bioengineered AAV Capsids with Combined High Human Liver Transduction In Vivo and Unique Humoral Seroreactivity, Molecular therapy : the journal of the American Society of Gene Therapy, 26 (2018) 289–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Keeler AM, ElMallah MK, Flotte TR, Gene Therapy 2017: Progress and Future Directions, Clinical and translational science, 10 (2017) 242–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Naso MF, Tomkowicz B, Perry WL 3rd, Strohl WR, Adeno-Associated Virus (AAV) as a Vector for Gene Therapy, BioDrugs : clinical immunotherapeutics, biopharmaceuticals and gene therapy, 31 (2017) 317–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Bruckman MA, Randolph LN, VanMeter A, Hern S, Shoffstall AJ, Taurog RE, Steinmetz NF, Biodistribution, pharmacokinetics, and blood compatibility of native and PEGylated tobacco mosaic virus nano-rods and -spheres in mice, Virology, 449 (2014) 163–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Czapar AE, Zheng YR, Riddell IA, Shukla S, Awuah SG, Lippard SJ, Steinmetz NF, Tobacco Mosaic Virus Delivery of Phenanthriplatin for Cancer therapy, ACS nano, 10 (2016) 4119–4126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Tervo DG, Hwang BY, Viswanathan S, Gaj T, Lavzin M, Ritola KD, Lindo S, Michael S, Kuleshova E, Ojala D, Huang CC, Gerfen CR, Schiller J, Dudman JT, Hantman AW, Looger LL, Schaffer DV, Karpova AY, A Designer AAV Variant Permits Efficient Retrograde Access to Projection Neurons, Neuron, 92 (2016) 372–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Deverman BE, Pravdo PL, Simpson BP, Kumar SR, Chan KY, Banerjee A, Wu WL, Yang B, Huber N, Pasca SP, Gradinaru V, Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain, Nature biotechnology, 34 (2016) 204–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Ojala DS, Sun S, Santiago-Ortiz JL, Shapiro MG, Romero PA, Schaffer DV, In Vivo Selection of a Computationally Designed SCHEMA AAV Library Yields a Novel Variant for Infection of Adult Neural Stem Cells in the SVZ, Molecular therapy : the journal of the American Society of Gene Therapy, 26 (2018) 304–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Nance ME, Duan D, Perspective on Adeno-Associated Virus Capsid Modification for Duchenne Muscular Dystrophy Gene Therapy, Human gene therapy, 26 (2015) 786–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Yang L, Li J, Xiao X, Directed evolution of adeno-associated virus (AAV) as vector for muscle gene therapy, Methods in molecular biology, 709 (2011) 127–139. [DOI] [PubMed] [Google Scholar]
- [90].Koerber JT, Jang JH, Schaffer DV, DNA shuffling of adeno-associated virus yields functionally diverse viral progeny, Molecular therapy : the journal of the American Society of Gene Therapy, 16 (2008) 1703–1709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Koerber JT, Klimczak R, Jang JH, Dalkara D, Flannery JG, Schaffer DV, Molecular evolution of adeno-associated virus for enhanced glial gene delivery, Molecular therapy : the journal of the American Society of Gene Therapy, 17 (2009) 2088–2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Korbelin J, Hunger A, Alawi M, Sieber T, Binder M, Trepel M, Optimization of design and production strategies for novel adeno-associated viral display peptide libraries, Gene therapy, 24 (2017) 470–481. [DOI] [PubMed] [Google Scholar]
- [93].Korbelin J, Sieber T, Michelfelder S, Lunding L, Spies E, Hunger A, Alawi M, Rapti K, Indenbirken D, Muller OJ, Pasqualini R, Arap W, Kleinschmidt JA, Trepel M, Pulmonary Targeting of Adeno-associated Viral Vectors by Next-generation Sequencing-guided Screening of Random Capsid Displayed Peptide Libraries, Molecular therapy : the journal of the American Society of Gene Therapy, 24 (2016) 1050–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Voigt CA, Martinez C, Wang ZG, Mayo SL, Arnold FH, Protein building blocks preserved by recombination, Nature structural biology, 9 (2002) 553–558. [DOI] [PubMed] [Google Scholar]
- [95].Endelman JB, Silberg JJ, Wang ZG, Arnold FH, Site-directed protein recombination as a shortest-path problem, Protein engineering, design & selection : PEDS, 17 (2004) 589–594. [DOI] [PubMed] [Google Scholar]
- [96].Tuerk C, Gold L, Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase, Science, 249 (1990) 505–510. [DOI] [PubMed] [Google Scholar]
- [97].Wu Y, Sefah K, Liu H, Wang R, Tan W, DNA aptamer-micelle as an efficient detection/delivery vehicle toward cancer cells, Proceedings of the National Academy of Sciences of the United States of America, 107 (2010) 5–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Xiang D, Shigdar S, Qiao G, Wang T, Kouzani AZ, Zhou SF, Kong L, Li Y, Pu C, Duan W, Nucleic acid aptamer-guided cancer therapeutics and diagnostics: the next generation of cancer medicine, Theranostics, 5 (2015) 23–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Darmostuk M, Rimpelova S, Gbelcova H, Ruml T, Current approaches in SELEX: An update to aptamer selection technology, Biotechnology advances, 33 (2015) 1141–1161. [DOI] [PubMed] [Google Scholar]
- [100].Stoltenburg R, Reinemann C, Strehlitz B, SELEX--a (r)evolutionary method to generate high-affinity nucleic acid ligands, Biomolecular engineering, 24 (2007) 381–403. [DOI] [PubMed] [Google Scholar]
- [101].Germer K, Leonard M, Zhang X, RNA aptamers and their therapeutic and diagnostic applications, International journal of biochemistry and molecular biology, 4 (2013) 27–40. [PMC free article] [PubMed] [Google Scholar]
- [102].Hianik T, Ostatna V, Sonlajtnerova M, Grman I, Influence of ionic strength, pH and aptamer configuration for binding affinity to thrombin, Bioelectrochemistry, 70 (2007) 127–133. [DOI] [PubMed] [Google Scholar]
- [103].Chang AL, McKeague M, Smolke CD, Facile characterization of aptamer kinetic and equilibrium binding properties using surface plasmon resonance, Methods in enzymology, 549 (2014) 451–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Yakimovich OY, Alekseev YI, Maksimenko AV, Voronina OL, Lunin VG, Influence of DNA aptamer structure on the specificity of binding to Taq DNA polymerase, Biochemistry. Biokhimiia, 68 (2003) 228–235. [DOI] [PubMed] [Google Scholar]
- [105].Smestad J, Maher LJ 3rd, Ion-dependent conformational switching by a DNA aptamer that induces remyelination in a mouse model of multiple sclerosis, Nucleic acids research, 41 (2013) 1329–1342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Mi J, Liu Y, Rabbani ZN, Yang Z, Urban JH, Sullenger BA, Clary BM, In vivo selection of tumor-targeting RNA motifs, Nature chemical biology, 6 (2010) 22–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Mi J, Ray P, Liu J, Kuan CT, Xu J, Hsu D, Sullenger BA, White RR, Clary BM, In Vivo Selection Against Human Colorectal Cancer Xenografts Identifies an Aptamer That Targets RNA Helicase Protein DHX9, Molecular therapy. Nucleic acids, 5 (2016) e315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Cheng C, Chen YH, Lennox KA, Behlke MA, Davidson BL, In vivo SELEX for Identification of Brain-penetrating Aptamers, Molecular therapy. Nucleic acids, 2 (2013) e67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Wang H, Zhang Y, Yang H, Qin M, Ding X, Liu R, Jiang Y, In Vivo SELEX of an Inhibitory NSCLC-Specific RNA Aptamer from PEGylated RNA Library, Molecular therapy. Nucleic acids, 10 (2018) 187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].King DJ, Ventura DA, Brasier AR, Gorenstein DG, Novel combinatorial selection of phosphorothioate oligonucleotide aptamers, Biochemistry, 37 (1998) 16489–16493. [DOI] [PubMed] [Google Scholar]
- [111].Liu H, Mai J, Shen J, Wolfram J, Li Z, Zhang G, Xu R, Li Y, Mu C, Zu Y, Li X, Lokesh GL, Thiviyanathan V, Volk DE, Gorenstein DG, Ferrari M, Hu Z, Shen H, A Novel DNA Aptamer for Dual Targeting of Polymorphonuclear Myeloid-derived Suppressor Cells and Tumor Cells, Theranostics, 8 (2018) 31–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].Zhou Y, Chi H, Wu Y, Marks RS, Steele TWJ, Organic additives stabilize RNA aptamer binding of malachite green, Talanta, 160 (2016) 172–182. [DOI] [PubMed] [Google Scholar]
- [113].Wu D, Katilius E, Olivas E, Dumont Milutinovic M., Walt DR, Incorporation of Slow Off-Rate Modified Aptamers Reagents in Single Molecule Array Assays for Cytokine Detection with Ultrahigh Sensitivity, Analytical chemistry, 88 (2016) 8385–8389. [DOI] [PubMed] [Google Scholar]
- [114].Borbas KE, Ferreira CS, Perkins A, Bruce JI, Missailidis S, Design and synthesis of mono- and multimeric targeted radiopharmaceuticals based on novel cyclen ligands coupled to anti-MUC1 aptamers for the diagnostic imaging and targeted radiotherapy of cancer, Bioconjugate chemistry, 18 (2007) 1205–1212. [DOI] [PubMed] [Google Scholar]
- [115].Lam KS, Lebl M, Krchnak V, The “one-bead-one-compound” combinatorial library method, Chemical reviews, 97 (1997) 411–448. [DOI] [PubMed] [Google Scholar]
- [116].Hu D, Hu S, Wan W, Xu M, Du R, Zhao W, Gao X, Liu J, Liu H, Hong J, Effective Optimization of Antibody Affinity by Phage Display Integrated with High-Throughput DNA Synthesis and Sequencing Technologies, PloS one, 10 (2015) e0129125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [117].Dias-Neto E, Nunes DN, Giordano RJ, Sun J, Botz GH, Yang K, Setubal JC, Pasqualini R, Arap W, Next-generation phage display: integrating and comparing available molecular tools to enable cost-effective high-throughput analysis, PloS one, 4 (2009) e8338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [118].Infante YC, Pupo A, Rojas G, A combinatorial mutagenesis approach for functional epitope mapping on phage-displayed target antigen: application to antibodies against epidermal growth factor, MAbs, 6 (2014) 637–648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Pasqualini R, Vascular targeting with phage peptide libraries, Q J Nucl Med, 43 (1999) 159–162. [PubMed] [Google Scholar]
- [120].Rajotte D, Ruoslahti E, Membrane dipeptidase is the receptor for a lung-targeting peptide identified by in vivo phage display, The Journal of biological chemistry, 274 (1999) 11593–11598. [DOI] [PubMed] [Google Scholar]
- [121].Butterfield GL, Lajoie MJ, Gustafson HH, Sellers DL, Nattermann U, Ellis D, Bale JB, Ke S, Lenz GH, Yehdego A, Ravichandran R, Pun SH, King NP, Baker D, Evolution of a designed protein assembly encapsulating its own RNA genome, Nature, 552 (2017) 415–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [122].Pasqualini R, Koivunen E, Kain R, Lahdenranta J, Sakamoto M, Stryhn A, Ashmun RA, Shapiro LH, Arap W, Ruoslahti E, Aminopeptidase N is a receptor for tumor-homing peptides and a target for inhibiting angiogenesis, Cancer research, 60 (2000) 722–727. [PMC free article] [PubMed] [Google Scholar]
- [123].Joshi S, Chen L, Winter MB, Lin YL, Yang Y, Shapovalova M, Smith PM, Liu C, Li F, LeBeau AM, The Rational Design of Therapeutic Peptides for Aminopeptidase N using a Substrate-Based Approach, Scientific reports, 7 (2017) 1424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Carter A, Integrins as target: first phase III trial launches, but questions remain, Journal of the National Cancer Institute, 102 (2010) 675–677. [DOI] [PubMed] [Google Scholar]
- [125].Kapp TG, Rechenmacher F, Neubauer S, Maltsev OV, Cavalcanti-Adam EA, Zarka R, Reuning U, Notni J, Wester HJ, Mas-Moruno C, Spatz J, Geiger B, Kessler H, A Comprehensive Evaluation of the Activity and Selectivity Profile of Ligands for RGD-binding Integrins, Scientific reports, 7 (2017) 39805. [DOI] [PMC free article] [PubMed] [Google Scholar]
