US20100080850A1 - Polypeptide ligands for targeting cartilage and methods of use thereof - Google Patents

Polypeptide ligands for targeting cartilage and methods of use thereof Download PDF

Info

Publication number
US20100080850A1
US20100080850A1 US12/563,201 US56320109A US2010080850A1 US 20100080850 A1 US20100080850 A1 US 20100080850A1 US 56320109 A US56320109 A US 56320109A US 2010080850 A1 US2010080850 A1 US 2010080850A1
Authority
US
United States
Prior art keywords
polypeptide
cartilage
seq
therapeutic agent
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/563,201
Inventor
Jeffrey A. Hubbell
Dominique A. Rothenfluh
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ecole Polytechnique Federale de Lausanne EPFL
Original Assignee
Ecole Polytechnique Federale de Lausanne EPFL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ecole Polytechnique Federale de Lausanne EPFL filed Critical Ecole Polytechnique Federale de Lausanne EPFL
Priority to US12/563,201 priority Critical patent/US20100080850A1/en
Publication of US20100080850A1 publication Critical patent/US20100080850A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease

Definitions

  • the technical field of the invention is generally related to delivery of therapeutic agents to cartilage tissues using polypeptides that specifically bind cartilage.
  • Cartilage lesions are common and can pose difficulties both in diagnosis and treatment.
  • a lesion can either be a defect or a focal cartilage degradation without visible disruption of the cartilage matrix.
  • Such lesions can result from an injury as in sports, disease, or aging.
  • the prognosis of an articular cartilage defect varies according to age, mechanism of injury, site, size, associated injuries and treatment received.
  • the invention provides treatments for cartilage injury.
  • Some aspects of the inventions are substantially pure polypeptides comprising an amino acid sequence of WYRGRL (SEQ ID NO:1), DPHFHL (SEQ ID NO:2), or RVMLVR (SEQ ID NO:3), or a conservative substitution thereof, or a nucleic acid encoding the same.
  • WYRGRL SEQ ID NO:1
  • DPHFHL SEQ ID NO:2
  • RVMLVR SEQ ID NO:3
  • Such polypeptides specifically bind cartilage tissue.
  • Such polypeptides may also include a therapeutic agent.
  • Some inventive methods are related to treating cartilage of a mammal comprising administering to the mammal a pharmaceutically acceptable composition that comprises a nucleic acid encoding a polypeptide that specifically binds a cartilage tissue.
  • a pharmaceutically acceptable composition that comprises a nucleic acid encoding a polypeptide that specifically binds a cartilage tissue.
  • Such polypeptide may also encode a therapeutic agent that, for example, treats a joint or tissue.
  • a delivery system for delivering a therapeutic agent comprising: a substantially purified preparation that comprises a pharmaceutically acceptable excipient, a therapeutic agent, and a polypeptide ligand comprising an amino acid sequence in the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, and conservative substitutions thereto.
  • the polypeptide ligands may specifically bind to cartilage tissue for targeted delivery of the therapeutic agent to cartilage tissues.
  • the therapeutic agent may comprise, for example, a drug, a visualization agent, or a therapeutic polypeptide.
  • the delivery system may include, for example, a collection of nanoparticles having an average diameter of between about 10 nm and about 200 nm, wherein the nanoparticles comprise the therapeutic agent and the polypeptide ligand.
  • a biomaterial comprising a polymer and a substantially pure polypeptide comprising an amino acid sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, or a conservative substitution thereof, wherein the polypeptide specifically binds to cartilage tissue and the polymer is free of amino acids and has a molecular weight of at least 400.
  • WYRGRL SEQ ID NO:1
  • WYRGRLC SEQ ID NO:4
  • FIG. 1 a is a bar graph that shows amplification of three phage clones identified by biopanning and demonstrates that they all grow at equal rates comparable to the random library. Final selection of C1-3 was therefore not affected by differences in growth during amplification.
  • FIG. 1 b is a bar graph that demonstrates the competitive binding assay in which only phage clone C1-3 and C1-C1 could be recovered.
  • FIG. 1 c is a bar graph that shows binding specificity of C1-3 and C1-C1 to articular cartilage. Binding of these phage clones to synovial membrane resulted in a lower phage recovery by two orders of magnitude, reflecting non-specific binding. Error bars indicate mean ⁇ standard deviation from three independent experiments.
  • FIG. 2 is a graph showing inhibition of cartilage binding of C1-3 by the synthetic polypeptide WYRGRLC (SEQ ID NO:4). Data represents the percentage of maximal phage binding of clone C1-3 obtained in the absence of synthetic polypeptide. Error bars indicate mean ⁇ standard deviation from three independent experiments.
  • FIG. 3 is a bar graph showing inhibition of cartilage binding of C1-3 phage by 10 1 ⁇ M of synthetic polypeptide vs.
  • WYRGRLC SEQ ID NO:4—PPS nanoparticles and 10 ⁇ M of synthetic mismatch polypeptide.
  • WYRGRLC SEQ ID NO:4—PPS nanoparticles exhibit similar binding than the synthetic polypeptide, whereas the synthetic mismatch polypeptide does not result in a significant decrease of phage titer. Error bars indicate mean ⁇ standard deviation from three independent experiments.
  • FIG. 4 is a bar graph showing relative accumulation in articular cartilage in vivo of nanoparticles decorated with WYRGRLC (SEQ ID NO:4) compared to control nanoparticles. Error bars indicate mean ⁇ standard deviation from three independent experiments.
  • articular cartilage tissue-binding ligands have given rise to new techniques to target therapeutics towards articular cartilage tissue and new materials for treatment of articular cartilage defects.
  • the ligands are effective in vivo to target therapeutic materials to articular cartilage.
  • cartilage tissue-binding ligands are polypeptides with the amino acid sequence of WYRGRL (SEQ ID NO:1), DPHFHL (SEQ ID NO:2), or RVMLVR (SEQ ID NO:3).
  • Other ligands are polypeptides with sequences that have conservative substitutions of one of SEQ ID NOs. 1, 2, or 3.
  • Ligand is a term that refers to a chemical moiety that has specific binding to a target molecule.
  • a target refers to a predetermined molecule, tissue, or location that the user intends to bind with the ligand.
  • targeted delivery to a tissue refers to delivering a molecule to the intended target tissue; a therapeutic agent delivered to a target may be intended to act on the target itself or on some other molecule or cell, e.g., a cell or tissue that is near the target.
  • One application of the cartilage tissue-binding ligands is for targeting therapeutic agents to articular cartilage, commonly referred to as intra-articular drug delivery when drugs are delivered.
  • Intra-articular drug delivery has been termed a major challenge due to the short residence times of intra-articular injected drugs. Drugs injected by themselves tend to diffuse away rapidly or be otherwise rapidly taken up into the circulation, thus causing low bioavailability of the drug at the cartilage and unwanted systemic effects.
  • sustained-release drugs only a few reports exist on the intra-articular use of any sustained-release formulations 20-26 .
  • albumin and poly(lactic-co-glycolic acid) (PLGA) have been used as biocompatible and biodegradable polymers for this purpose either in the form of gels 22 or as microspheres 23-26 .
  • PLGA poly(lactic-co-glycolic acid)
  • Such approaches rely, however, on degradation over time to achieve sustained-release within the joint, a strategy that is limited by the properties of available biomaterials, and which makes the drugs available only when the material degrades.
  • Targeting the delivery system to articular cartilage with tissue-binding ligands makes the tissue itself a reservoir for drug release to the site of the disease process as opposed to release in the joint cavity.
  • the cartilage tissue-binding ligands are also useful to bind cartilage tissue in vitro for diagnostic, assay, or imaging purposes. For instance, sections of tissue may be exposed to cartilage tissue-binding ligands that are also bound to a fluorescent molecule or other imaging agent to visualize the location of the cartilage tissue. Or, for instance, the cartilage tissue-binding ligands may be used in affinity chromatography to isolate the tissue.
  • articular cartilage tissue-binding ligands described herein enable new techniques for controlled release by enabling sustained localization through specific binding to cartilage, and their accompanying formulations increase intra-articular bioavailability of delivered drugs or other therapeutic agents, which is beneficial for various disease processes, e.g., those involving the synovium such as rheumatoid arthritis or inflammation in clinically manifest osteoarthritis. Because convective transport of solutes into cartilage is impaired due to the inherent properties of this tissue 27,28 , the bioavailability of drugs in the cartilage matrix, which is the primary site of the disease process in osteoarthritis, can be enhanced by sustained release systems that reside in the matrix itself.
  • Targeting of the cartilage matrix as described herein is therefore useful for general intra-articular therapeutic agent delivery, such as targeting of the cartilage matrix for delivery of therapeutic agents to treat cartilage degradation in osteoarthritis and protect cartilage in conditions like rheumatoid arthritis, bacterial and reactive arthritis.
  • a therapeutic agent refers to a molecule for delivery to a target to accomplish a desirable medical or scientific function, and the term includes drugs and imaging agents.
  • therapeutic agents are matrix metalloproteinase (MMP) inhibitors, aggrecanase inhibitors, COX-inhibitors and other non-steroidal anti-inflammatory drugs (NSAIDs), glucosamin, diacerhein, methotrexate, steroids, immunosuppressing drugs (rapamycin, cyclosporine), protein therapeutics (growth factors, tissue inhibitors of matrix metalloproteinases), and oligonucleotides (e.g., siRNA, shRNA, miRNA).
  • imaging agents are fluorescent markers, radio-opaque materials, magnetic resonance imaging contrast agents, x-ray imaging agents, radiopharmaceutical imaging agents, ultrasound imaging agents, and optical imaging agents.
  • phage display of random peptides on the minor coat proteins (pIII) of bacteriophages has allowed use of affinity purification, in a process called biopanning 5 , to identify specific peptides for precise targeting of multiple tissues, both in vitro 6 and in vivo in animals and even humans 7-9 .
  • affinity purification in a process called biopanning 5
  • Arap et al. have succeeded in mapping the human vasculature and specifically targeting the microvasculature of adipose tissue in mice.
  • Another study identified a synovium-specific homing peptide by phage display.
  • phage display library fUSE5/6-mer based on filamentous phage strain fd-tet was received from the University of Missouri, Columbia.
  • Cartilage grafts, synovial fluid and synovial membrane were harvested from bovine shoulders obtained from the local slaughterhouse.
  • Cartilage grafts were stored in 0.1% sodium azide and protease inhibitors at 4° C. and used within 72 hours.
  • Buffers and solutions used for phage-display screening Blocking solution (0.1M NaHCO 3 , 1% BSA, pH 8.5), wash buffer (PBS, Tween 20 0.1-1%), elution solution (50 mM glycine-HCl, pH 2.0), neutralisation buffer (0.2M NaHPO 4 ).
  • Solvents and reagents for nanoparticle synthesis were purchased from Sigma-Aldrich (Buchs, Switzerland). All peptides (synthesis chemicals from Novabiochem, Lautelfingen, Switzerland) were synthesized on solid resin using an automated peptide synthesizer (CHEMSPEED PSW 1100, Augst, Switzerland) with standard F-moc chemistry. Purification was performed on a Waters ultrapurification system using a Waters ATLANTIS dC 18 semi-preparative column and peptides collected according to their molecular mass analyzed by time-of-flight (TOF) mass spectrometry.
  • TOF time-of-flight
  • Peptides for binding to the articular cartilage matrix were selected by exposing a fUSE5/6-mer library to bovine cartilage grafts, which provided 6.4 ⁇ 10 7 different phage clones with 6-amino acid linear-peptide inserts displayed on the minor coat protein of filamentous phage 5,18 .
  • Cartilage grafts were harvested with 8 and 4 mm biopsy cutters (two-sided surface 1 cm 2 and 0.25 cm 2 ). A slice with the intact articular surface was removed to expose the phages only to the cartilage matrix below the surface.
  • Affinity selection was preformed in polystyrene 48-well plates, which were blocked for nonspecific adhesion with a blocking solution containing 1% BSA for 2 hours prior to screening. A total of 5 screening rounds was carried out. In the first round 10 13 and in subsequent rounds 10 12 phage virions per ml were exposed to the cartilage graft, washed with PBS/Tween 20 and eluted at low pH. While the first round was supposed to give a high yield at low stringency, subsequent rounds were carried out with increasing stringency to select stronger binders. Conditions were increased from 4 hours of binding at RT and washing with PBS/0.1% Tween to 30 minutes binding at 37° C.
  • the polypeptide sequences of affinity selected phage specific to the articular cartilage matrix was determined by DNA sequencing (Microsynth AG, Balgach, Switzerland) using the sequencing primer 5′-CAT GTA CCG TAA CAC TGA G (SEQ ID NO:5). Binding specificity was determined by exposing selected phage clones (10 8 TU/ml) to articular cartilage (0.25 cm 2 ) with and without synovial fluid, to synovial membrane (3 ⁇ 4 mm, 0.24 cm 2 ) and to polystyrene after blocking. Titer counts were obtained by spot titering.
  • Competetive binding was probed by exposing a mix of selected phage clones and fUSE5/6-mer library (each 10 8 TU/ml) in the presence of synovial fluid to articular cartilage for 30 min. at 37° C., 220 RPM and washed with PBS/1% Tween 20.
  • the phage clones were identified by DNA sequencing and the corresponding titer counts calculated.
  • 10 8 TU/ml of phage and its free polypeptide in different concentrations or nanoparticles were mixed, exposed to articular cartilage and titer counts determined. All experiments have been carried out in triplicate and repeated for confirmation.
  • Nanoparticle Synthesis Poly(propylene sulfide) nanoparticles were prepared as described elsewhere 14 . Briefly, for nanoparticles between 30 and 40 nm, a monomer emulsion is prepared by dissolving 1.6% (w/v) of Pluronic F-127 (MW 12600) in 10 ml of degassed, double-distilled and filtered water. The system is continuously stirred and purged with argon for 60-90 min. Propylene sulfide is added at a Pluronics/monomer ratio of 0.4 (w/w).
  • the initiator pentaerythritol tetrathioester (TTE) (synthesized as described previously 14 ), is deprotected by mixing with a molar equivalent of 0.5M sodium methanoate and stirred under argon for 5 minutes. The deprotected initiator is then added to the emulsion. Five minutes later, 60 ⁇ l of diaza[5.4.0] bicycloundec-7-ene (DBU) is added and the reaction stirred under inert conditions for 6 hours. Exposure to air yields disulfide crosslinking of the particle core.
  • TTE pentaerythritol tetrathioester
  • Particles are subsequently purified from remaining monomers and base by 2 days of repeated dialysis against ultrapure water through a membrane with a MWCO of 6-8 kDa (Spectra/Por). Free Pluronic F-127 is removed in a second dialysis step through 300 kDa membranes. Particle size is measured after dialysis by dynamic light scattering (ZETASIZER NANO ZS, Malvern Instruments, Malvern, UK). For preparation of surface-functionalized nanoparticles, polypeptides are conjugated to Pluronics-F127 prior to nanoparticle synthesis.
  • Pluronics F-127 In order to derivatize Pluronics F-127 with vinyl sulfone, 400 ml of toluene and 15 g of Pluronic F-127 were introduced in a 3-neck round bottom flask connected to a Soxhlett filled with glass wool and dry molecular sieves and a cooling tube. Pluronic was dried azeotropically during 4 h. The dried Pluronic in toluene was cooled in an ice-bath and sodium hydride was added in a 5 equimolar excess compared with Pluronic-OH-groups.
  • the reaction was stirred for 15 minutes and divinyl sulfone was added in a 15 molar excess and the reaction was carried out in the dark for 5 days at RT under argon.
  • the reaction solution was filtrated through a celite filter cake, concentrated by rotary evaporation and then precipitated 5 times in ice-cold diethylether.
  • the polymer was dried under vacuum and stored under argon at ⁇ 20° C.
  • Nanoparticles are prepared as described above with 10% surface functionalisation corresponding to a fraction of 0.16% (w/v) of conjugated Pluronic. The presence of the polypeptide on the particle is confirmed by measuring the zeta potential by dynamic light scattering (ZETASIZER NANO ZS, Malvern Instruments, Malvern, UK) after particle synthesis.
  • the nanoparticles were fluorescently labelled with 6-iodoacetamide fluorescein (6-IAF, Molecular Probes, Eugene, Oreg.) or Alexa FLUOR 488 maleimide by adding label at 1 mg/ml of nanoparticle solution in 10 mM Tris-HCl pH 8.5 and stirred in the dark for 2 h at room temperature.
  • the knee joints were harvested and freed from surrounding muscle by microdissection. Confocal microscopy and image analysis The sections were analyzed by confocal laser scanning microscopy using a Zeiss LSM510 meta. Fluorescence was extracted by emission fingerprinting to reduce autofluorescence of cartilage tissues. The pinhole was adjusted using TETRASPECK fluorescent microspheres (Invitrogen, T14792, Carlsbad, Calif.). Images for analysis were obtained using a Zeiss 63 ⁇ APOCHROMAT objective in 10 different locations per joint with a z-stack of 10 images each. Image deconvolution was accomplished by Huygens software. Image analysis was done by ImageJ.
  • Cartilage grafts were incubated with the fUSES peptide on phage display library, which expressed linear 6-mer random peptides on minor coat proteins (pIII) with a diversity of 6.4 ⁇ 10 7 .
  • the sequences corresponding to the polypeptides displayed on the phage virions were determined by DNA sequencing after rounds 3 and 5 of affinity selection (panning). After round 3, sequencing did not reveal a consensus motif in the selected polypeptides.
  • the binding specificity of C1-3 and C1-C1 to articular cartilage was evaluated by exposing the phage clones to articular cartilage (0.25 cm 2 ) and synovial membrane (0.24 cm 2 ) and comparing to random binding of the fUSES phage library. Furthermore, the effect of the presence of synovial fluid on phage binding was probed by adding an equal volume of bovine synovial fluid to the phages, which dilutes the synovial fluid by a factor of 2.
  • FIG. 1 c shows that both C1-3 and C1-C1 exhibit specific binding to articular cartilage over synovial membrane by two orders of magnitude and that the addition of synovial fluid does not yield a significant drop in phage binding to articular cartilage.
  • Binding of the specific phage clones to synovial membrane seems to reflect background phage binding as the random phage library fUSES bound to the synovial membrane to the same extent.
  • phage titers of fUSE5 to articular cartilage were at the same level as to synovial membrane, further indicating specific binding of C1-3 and C1-C1. Phage was exposed to polystyrene wells after blocking them with BSA because it is contained in the plasticware in which affinity selection was carried out. No background phage binding to polystyrene was detectable, however.
  • phage clone C1-3 polypeptide WYRGRL, SEQ ID NO:1 which exhibits both binding specificity to articular cartilage and dominant binding compared to other phage clones.
  • Specific binding in the context of a polypeptide refers to the binding of the polypeptide specifically to the target of interest as opposed to other molecules.
  • the 6-mer polypeptide insert WYRGRL (SEQ ID NO:1) of clone C1-3 as well as its scrambled mismatch YRLGRW (SEQ ID NO:6) were synthesized on solid resin using standard Fmoc chemistry.
  • the N-terminal amino acids are acetylated and a cysteine was added to the C-terminus of the polypeptides for bioconjugation to vinyl sulfone by Michael-type addition via the free thiol.
  • a competetive binding assay against the phage clone C1-3 was performed by exposing the C1-3 and the WYRGRLC (SEQ ID NO:4) to the cartilage.
  • a dose-response curve was determined by serial dilutions of the polypeptide ranging from 50 nM to 10 ⁇ M and mixing them with 10 8 TU/ml of phage.
  • the titer counts of phage recovered gradually decreased by two orders of magnitude as the concentration of the free polypeptide in solution was increased ( FIG. 2 ).
  • An IC50 of about 200 nM can be estimated from the curve in FIG. 2 .
  • Pluronic F-127 was derivatized with vinyl sulfone.
  • the polypeptides were conjugated to Pluronic-di-vinyl sulfone via the free thiol in the C-terminal cysteine by Michael-type addition.
  • PPS nanoparticles were then prepared by inverse emulsion polymerization with Pluronic F-127 (90%) and polypeptide-conjugated Pluronic F-127 (10%) serving as the emulsifier. Because Pluronic as the emulsifier remains on the particle surface, the conjugated polypeptide is displayed on the surface of the nanoparticles, thereby adding the targeting functionality of the polypeptide to the particles.
  • Size measurements by dynamic light scattering revealed a size by volume of 38 nm for PPS particles displaying WYRGRLC (SEQ ID NO:4) (polydispersity index PDI 0.221), 31 nm for particles displaying YRLGRC (SEQ ID NO:7) PDI 0.412) and 37 nm for non-conjugated PPS particles (PDI 0.212).
  • the zeta-potential of non-conjugated PPS nanoparticles is about neutral ( ⁇ 2.64 ⁇ 8.97 mV).
  • the zeta-potential of conjugated nanoparticles shifted to +17.8 ⁇ 3.45 mV, which further confirms the presence of the polypeptide on the nanoparticle surface.
  • WYRGRLC PPS nanoparticles at 2% (w/v) were subjected to a competetive binding assay against the free polypeptides WYRGRLC (SEQ ID NO:4) and YRLGRC (SEQ ID NO:7) at concentrations of 10 ⁇ M each. Accordingly, phage clone C1-3 was exposed to the cartilage in the presence of WYRGRLC (SEQ ID NO:4)—PPS nanoparticles, WYRGRLC (SEQ ID NO:4) or YRLGRC (SEQ ID NO:7), and the amount of C1-3 phage binding to the cartilage relative to a control C1-3 phage without competitive inhibitors was measured. The results in FIG.
  • WYRGRLC PPS and PPS nanoparticles were labeled with 6-IAF and dialysed for 2 days with at least 2 buffer shifts to ensure that no free label is still in the solution.
  • a 250 Hamilton syringe Hamilton Europe, Bonaduz, Switzerland
  • Cheney reproducibility adapter was used for antero-lateral parapatellar injection with a 30 G needle.
  • binding proteins or polypeptides such as phage display s , yeast surface display 15 , mRNA display 16 or peptide-on-bead display 17 .
  • phage display using the fUSE5/6-mer library based on the filamentous phage vector fd-tet 18 was used to select short peptides which bind to the articular cartilage matrix.
  • Embodiments of the invention include using affinity selection of binding proteins or peptides against cartilage ex vivo.
  • biopanning was carried out against slices of bovine cartilage.
  • Conditions in the binding step were chosen with increasing stringency from round 1 to 5 in order to favor binding of high affinity polypeptides.
  • the selected sequences obtained from DNA sequencing of 96 clones have been evaluated in a competetive binding assay.
  • Binding specificity was conferred by the polypeptide sequence rather than the net positive charge of the polypeptide which sticks non-specifically to negatively charged proteoglycans. This is demonstrated in FIG. 3 in that 10 ⁇ M of WYRGRLC (SEQ ID NO:4) resulted in a decrease of phage titers close to 100fold. By contrast, 10 ⁇ M of YRLGRC (SEQ ID NO:7), which comprises the same amino acids just in scrambled order, did not result in a significant decrease in phage titer as compared to control phage titers without polypeptide.
  • WYRGRLC (SEQ ID NO:4) is a short polypeptide with specific binding to articular cartilage which shows a dose dependent decrease in competetive binding against phage C1-3.
  • the polypeptides were synthesized such that they contain a cysteine at the C-terminus.
  • the free thiol of cysteine is used for bioconjugation by Michael-type addition to Pluronic-di-vinyl sulfone which serves as the emulsifier in nanoparticle synthesis and therefore remains displayed on the particle surface. While this is a straightforward scheme for surface functionalisation of nanoparticles synthesized by inverse emulsion polymerization, conjugation of Pluronic requires excess of polypeptide despite the favorable kinetics of Michael-type addition of free thiols to vinyl sulfone, if conjugation close to 100% is to be achieved as evidenced by 1 H-NMR. In addition, some Pluronic is always lost during nanoparticle synthesis. Alternative synthetic schemes therefore add surface functionality to already-synthesized nanoparticles in order to limit the amount of polypeptide needed.
  • WYRGRLC(SEQ ID NO:4) PPS nanoparticles with a degree of surface functionalisation of 10% (w/w of total Pluronic used) at a concentration of 2% (w/v) against phage clone C1-3 (10 8 TU/ml) to articular cartilage resulted in a similar decrease of phage titers as 10 ⁇ M of free polypeptide WYRGRLC (SEQ ID NO:4, FIG. 3 ).
  • Targeting the extra-cellular matrix of articular cartilage essentially depends on the ability of the drug delivery system to enter the cartilage matrix and to stay there.
  • the distribution of nanoparticles in the joint is mainly governed by the capability of tissue penetration and cellular uptake. While larger particles do not enter, smaller ones are able to penetrate and reside in the cartilage ECM. It is demonstrated in herein that nanoparticles (in this case, nanoparticles with a mean volume diameter of 36 and 38 nm) are able to enter the articular cartilage ECM and meniscal tissue in addition to the synovium. It is consistent with the literature that small particles possess the ability to enter the cartilage ECM.
  • adeno-associated viruses with a mean diameter of 20-25 nm enter the articular cartilage matrix up to a depth of penetration of 450 ⁇ m in normal and 720 ⁇ m in degraded cartilage 19 .
  • WYRGRLC SEQ ID NO:4—PPS nanoparticles over non-functionalised and therefore non-targeted PPS nanoparticles 24 hours after intra-articular injection into the knee joint of mice ( FIG. 4 ).
  • Bovine cartilage Although the basic research has been performed with bovine cartilage, the polypeptide sequences described herein are expected to bind to human articular cartilage due to the general homology of these tissues. Bovine cartilage is an accepted model in cartilage research due to the very limited availability of healthy human cartilage.
  • cartilage tissue-binding ligands are polypeptides with the amino acid sequence of WYRGRL (SEQ ID NO:1), DPHFHL (SEQ ID NO:2), or RVMLVR (SEQ ID NO:3).
  • Other ligands are polypeptides or functional polypeptides with sequences that have conservative substitutions of one of SEQ ID NOs. 1, 2, or 3.
  • Certain embodiments are directed to the subset of polypeptides that have a certain percentage identity to the disclosed sequences, or a certain degree of substitution, with the subset being primarily, or only, functional polypeptides.
  • the binding activity of a polypeptide to cartilage may be determined simply by following experimental protocols as described herein.
  • a polypeptide variant of one of the polypeptide ligands may be labeled with a marker (e.g., radioactive or fluorescent) and exposed to bovine cartilage to determine its binding affinity using well-known procedures.
  • a binding assay may be performed using a simple fluorescence readout using a plate reader by labeling polypeptide variants with a fluorescent marker, e.g., 6-fluorescein iodoacetamide which reacts with the free thiol of the cysteine.
  • the binding strengths of polypeptide variants relative to e.g., WYRGRLC (SEQ ID NO:4) under given physiological conditions can be determined, e.g., sequences made using conservative substitutions, truncations of the sequences to 5 or less amino acids, addition of flanking groups, or changes or additions for adjusting sequences for solubility in aqueous solution.
  • Polypeptides of various lengths may be used as appropriate for the particular application.
  • polypeptides that contain the polypeptide ligand sequences will exhibit specific binding if the polypeptide is available for interaction with cartilage in vivo. Protein folding can affect the bioavailability of the polypeptide ligands.
  • certain embodiments are directed to polypeptides that have a polypeptide ligand but do not occur in nature, and certain other embodiments are directed to polypeptides having particular lengths, e.g., from 6 to 3000 residues, or 6-1000, or 6-100, or 6-50; artisans will immediately appreciate that every value and range within the explicitly articulated limits is contemplated. Moreover, the lower limits may be 4 or 5 instead of 6.
  • binding activity may be expected in the three residues 1-3, 2-4, 3-5, and 4-6; for four residues, binding activity may be expected in 1-4, 2-5, and 3-6; for five residues, binding activity may be expected in positions 1-5 or 2-6.
  • polypeptide sequences and/or purified polypeptides refers to a chain of amino acid residues, regardless of post-translational modification (e.g., phosphorylation or glycosylation) and/or complexation with additional polypeptides, synthesis into multisubunit complexes, with nucleic acids and/or carbohydrates, or other molecules. Proteoglycans therefore also are referred to herein as polypeptides.
  • a “functional polypeptide” is a polypeptide that is capable of promoting the indicated function. Polypeptides can be produced by a number of methods, many of which are well known in the art.
  • polypeptides can be obtained by extraction from a natural source (e.g., from isolated cells, tissues or bodily fluids), by expression of a recombinant nucleic acid encoding the polypeptide, or by chemical synthesis.
  • Polypeptides can be produced by, for example, recombinant technology, and expression vectors encoding the polypeptide introduced into host cells (e.g., by transformation or transfection) for expression of the encoded polypeptide.
  • conservative changes that can generally be made to an amino acid sequence without altering activity. These changes are termed conservative substitutions or mutations; that is, an amino acid belonging to a grouping of amino acids having a particular size or characteristic can be substituted for another amino acid. Substitutes for an amino acid sequence may be selected from other members of the class to which the amino acid belongs.
  • the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and tyrosine.
  • the polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine.
  • the positively charged (basic) amino acids include arginine, lysine and histidine.
  • the negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Such alterations are not expected to substantially affect apparent molecular weight as determined by polyacrylamide gel electrophoresis or isoelectric point. Exemplary conservative substitutions include, but are not limited to, Lys for Arg and vice versa to maintain a positive charge; Glu for Asp and vice versa to maintain a negative charge; Ser for Thr so that a free —OH is maintained; and Gln for Asn to maintain a free NH 2 .
  • amino acid residues described herein employ either the single letter amino acid designator or the three-letter abbreviation. Abbreviations used herein are in keeping with the standard polypeptide nomenclature, J. Biol. Chem., (1969), 243, 3552-3559. All amino acid residue sequences are represented herein by formulae with left and right orientation in the conventional direction of amino-terminus to carboxy-terminus.
  • a determination of the percent identity of a peptide to a sequence set forth herein may be required. In such cases, the percent identity is measured in terms of the number of residues of the peptide, or a portion of the peptide.
  • a polypeptide of, e.g., 90% identity may also be a portion of a larger peptide Variations of the disclosed polypeptide sequences include polypeptides or functional polypeptides having about 83% identity (e.g., 1 of 6 substituted) or about 67% identity (e.g., 2 of 6 substituted).
  • purified as used herein with reference to a polypeptide refers to a polypeptide that either has no naturally occurring counterpart (e.g., a peptidomimetic), or has been chemically synthesized and is thus substantially uncontaminated by other polypeptides, or has been separated or purified from other most cellular components by which it is naturally accompanied (e.g., other cellular proteins, polynucleotides, or cellular components).
  • An example of a purified polypeptide is one that is at least 70%, by dry weight, free from the proteins and naturally occurring organic molecules with which it naturally associates.
  • a preparation of the a purified polypeptide therefore can be, for example, at least 80%, at least 90%, or at least 99%, by dry weight, the polypeptide.
  • Polypeptides also can be engineered to contain a tag sequence (e.g., a polyhistidine tag, a myc tag, or a Flag® tag) that facilitates the polypeptide to be purified or marked (e.g., captured onto an affinity matrix, visualized under a microscope).
  • a tag sequence e.g., a polyhistidine tag, a myc tag, or a Flag® tag
  • a purified composition that comprises a polypeptide refers to a purified polypeptide unless otherwise indicated.
  • Polypeptides may include a chemical modification; a term that, in this context, refers to a change in the naturally-occurring chemical structure of amino acids. Such modifications may be made to a side chain or a terminus, e.g., changing the amino-terminus or carboxyl terminus. In some embodiments, the modifications are useful for creating chemical groups that may conveniently be used to link the polypeptides to other materials, or to attach a therapeutic agent.
  • the cartilage tissue-binding ligands may be used to target nanoparticles to a cartilage tissue, and such nanoparticles may include a therapeutic agent, for example, one or more of the therapeutic agents described herein. While certain polymer-therapeutics have been described elsewhere, these have mainly been designed to augment drug concentrations in tumor tissues 1 . Such alteration of the biodistribution of anticancer drugs through delivery systems aims at reducing the drug's toxicity and at improving therapeutic effects 2,3 . To be effective, a drug delivery system must escape non-specific systemic accumulation and phagocytotic clearance by the host defense immune system.
  • the nanoparticle-based therapeutic agent delivery system described herein exhibits active targeting functionality for the cartilage, specifically, cartilage extra-cellular matrix.
  • the combination of targeting functionality and a nanoparticle-based delivery system enables better control of bioavailability and biodistribution, particularly for intra-articular drug delivery to the cartilage matrix.
  • Surface functionalisation of nanoparticles with the selected peptides controls the biodistribution by specific accumulation of nanoparticles in the articular cartilage, specifically in the extra-cellular matrix.
  • the cartilage matrix itself therefore serves as a reservoir of nanoparticle encapsulated therapeutic molecules, which are delivered to the site of the disease process.
  • articular cartilage can be targeted in vivo with nanoparticles by the use of polypeptides which have been characterized to exhibit specific homing activity to articular cartilage.
  • PPS nanoparticles are used herein for demonstrative purposes, other techniques for making nanoparticles may also be adapted.
  • the targeting polypeptides were made bioavailable by exposure at a surface of the nanoparticles. Size control was achieved in this particular technique by adjusting the emulsifier to monomer ratio and yielded sizes ranging from about 20 nm to about 200 nm 14 .
  • the density of the cartilage extra-cellular matrix represents a relevant obstacle not just for the screening of combinatorial peptide libraries but potentially also for drug delivery to the cartilage
  • the use of suitably-sized nanoparticles and/or ligands with specific binding to the cartilage enhances delivery efficiency.
  • Nanoparticles are be prepared as collections of particles having an average diameter of between about 10 nm and about 200 nm, including all ranges and values between the explicitly articulated bounds, e.g., from about 20 to about 200, and from about 20 to about 40, to about 70, or to about 100 nm, depending on the polydispersity which is yielded by the preparative method.
  • Detailed methods for making and delivering nanoparticles are set forth below and in U.S. Pat. Ser. No. 60/775,132, filed Feb. 21, 2006, which is hereby incorporated by reference herein.
  • Nanoparticle systems can be utilized, such as those formed from copolymers of poly(ethylene glycol) and poly(lactic acid), those formed from copolymers of poly(ethylene oxide) and poly(beta-amino ester), and those formed from proteins such as serum albumin.
  • Other nanoparticle systems are known to those skilled in these arts. See also Devalapally et al., Cancer Chemother Pharmacol., 07-25-06; Langer et al., International Journal of Pharmaceutics, 257:169-180 (2003); and Tobio et al., Pharmaceutical Research, 15(2):270-275 (1998).
  • microparticles Larger particles of more than about 200 nm average diameter incorporating the cartilage tissue-binding ligands may also be prepared, with these particles being termed microparticles herein since they begin to approach the micron scale and fall approximately within the limit of optical resolution. For instance, certain techniques for making microparticles are set forth in U.S. Pat. Nos. 5,227,165, 6,022,564, 6,090,925, and 6,224,794.
  • Functionalization of nanoparticles to employ targeting capability requires association of the targeting polypeptide with the particle, e.g., by covalent binding using a bioconjugation technique, with choice of a particular technique being guided by the particle or nanoparticle, or other construct, that the polypeptide is to be joined to.
  • a bioconjugation technique for attaching peptides to other materials are well known and the most suitable technique may be chosen for a particular material.
  • additional amino acids may be attached to the polypeptide sequences, such as a cysteine in the case of attaching the polypeptide to thiol-reactive molecules.
  • Therapeutic agents such as therapeutic polypeptides can be furnished with targeting capability by the use of the polypeptide ligands described and advantageously exhibit longer retention times in a joint, for instance by making a fusion protein of a polypeptide ligand and a therapeutic protein.
  • the polypeptide ligands can be attached to the N- or C-terminus in normal or reverse order.
  • One of the primers, forward or reverse depending whether the polypeptide ligand is supposed to be localized at the N- or C-terminus contains a gene sequence of the appropriate therapeutic polypeptide.
  • the reverse primer corresponds to 5′-ctgatgcggccgctcTCACAGCCTGCCCCTATACCAGCCGCCGCCxxxxx-3′ (SEQ ID NO:9) which contains the codon sequence for WYRGRL in reverse complement with a N-terminal glycine (GGG) linker (capital letters), as well as a stop codon and a restriction site for NotI and an overhang.
  • the Xs correspond to the therapeutic polypeptide specific sequence and may be, e.g., around 20 base pairs long, although other lengths may be used as per conventional practice in these arts.
  • target ligands may be encoded, e.g., using thea nucleic acid sequence 5′-ctgatgeggccgctcAAGATGGAAATGAGGATCGCCGCCGCCxxxxx-3′ (SEQ ID NO: 12) that endocdes DPHFHLGGG (SEQ ID NO:13) or the nucleic acid 5′-ctgatgcggccgctcACGAACAAGCATAACACGGCCGCCGCCxxxxx-3′(SEQ ID NO:14) that encodes RVMLVRGGG (SEQ ID NO:15).
  • a DNA sequence for WYRGRL (SEQ ID NO:1) is TGGTATAGGGGCAGGCTG (SEQ ID NO: 16), and for DPHFHL (SEQ ID NO:2) is AAGATGGAAATGAGGATC (SEQ ID NO:17), and for RVMLVR (SEQ ID NO:3) is ACGAACAAGCATAACACG (SEQ ID NO:18).
  • Certain therapeutic polypeptides include proteins present in cartilage, e.g., tissue inhibitors of matrix metalloproteinase-3 (TIMP-3), growth factors (e.g., Transforming growth factor-beta (TGF- ⁇ ), growth developmental factor-5 (GDF-5), CYR61 (Cystein-rich61)/CTGF (connective tissue growth factor)/NOV (Nephroblastoma overexpressed) (CCN2), insulin-like growth factor-1 (IGF-1), and bone morphogenic proteins (BMPs).
  • TGF- ⁇ Transforming growth factor-beta
  • GDF-5 growth developmental factor-5
  • CYR61 Cystein-rich61
  • CTGF connective tissue growth factor
  • IGF-1 insulin-like growth factor-1
  • BMPs bone morphogenic proteins
  • Certain embodiments are molecules for viscosupplementation, e.g., molecules found in human cartilage that include chondroitin sulfate, keratane sulfate, hyaluronic acid, prote
  • a fusion protein is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient.
  • the fusion protein is produced using a cell, either a procaryotic or a eucaryotic cell.
  • nucleic acids encoding a fusion protein are introduced into a patient, in which case the nucleic acids may be “naked” or part of a larger construct, e.g., a vector.
  • transfected cells are introduced into a patient.
  • the site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • Tissue inhibitor of matrix metalloproteinase 3 is a relatively insoluble matricellular protein 30 which inhibits several matrix metalloproteinases (MMP-1, -2, -9) 31 in addition to aggrecanase 1 and 2, i.e. ADAMTS4 and ADAMTS5 32 .
  • MMP-1, -2, -9 matrix metalloproteinases
  • ADAMTS4 and ADAMTS5 32 matrix metalloproteinases
  • TIMP-3 is not specifically expressed in articular cartilage, however, and due to its ability of inhibiting several enzymes may have severe potential adverse effects upon systemic administration or systemic dissemination following intra-articular injection.
  • Targeted delivery of the inhibitory domain of TIMP-3 to articular cartilage can prevent potential systemic dissemination while increasing the therapeutic effect of the molecule in the cartilage matrix, i.e. at the site of the disease process in osteoarthritis.
  • the inhibitory domain of TIMP-3 also serves as a protectant against cartilage degradation in conditions such as rheumatoid arthritis, bacterial arthritis or reactive arthritis.
  • TIMP-3 fusion protein which contains one of the sequences (SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3) or conservative substitutions thereof, e.g, at the N- or C-terminus. While TIMP-3 has been selected as an embodiment, other protease inhibitors or matrix metalloproteinase inhibitors may also be made and used by following the general procedures disclosed herein.
  • TIMP-3 contains a signal domain, residues 1 to 24, the N-terminal inhibitory domain, residues 24 to 143, and an extra-cellular matrix binding domain, residues 143 to 211 (SwissProt entry P35625). It has been demonstrated that N-TIMP-3, i.e. residues 24 to 143, is sufficient for exhibiting inhibitory activities on ADAMTS4 and ADAMTS5 as well as MMP-1 and MMP-2 3 . Accordingly, it is possible to engineer a fusion protein of N-TIMP-3 (Cys24 to Asn 143) which contains the sequence WYRGRL (SEQ ID NO:1) at the C-terminus. The natural non-specific C-terminal extra-cellular matrix binding domain is substituted with a targeting polypeptide specific for articular cartilage.
  • RT-PCR for amplifying the required DNA fragments was carried out with a proof-reading DNA polymerase (Pfu Turbo polymerase, Stratagene, LaJolla, Calif.) and the fragments with a required length of 420 bp checked by agarose-gel electrophoresis.
  • the DNA fragments were gel purified with a Nucleospin II column (Macherey-Nagel, Düren, Germany), cut with the corresponding restriction enzymes BamHI and NotI (New England BioLabs Inc., Ipswich, Mass.) and ligated into the bacterial protein expression vector pGEX-4T-1 (Amersham Biosciences, GE Healthcare Europe, Otelfingen, Switzerland), which expresses trTIMP-3 as a fusion protein with glutathione-S-transferase (GST) for purification, total MW 40 kDa.
  • GST glutathione-S-transferase
  • E. coli strain BL21 was transformed with the trTIMP-3-pGEX construct by electroporation.
  • An optimal expression clone was selected by anti-GST ELISA.
  • Bacterial cultures were grown in 2 ⁇ YT until they reached an A 600 of 0.6 to 0.7. Protein expression was then induced by 0.5M IPTG for 3 hours. To collect the protein from inclusion bodies, the bacteria were centrifuged, lysed by sonication and centrifuged again. The pellet was resuspended in a denaturing buffer (0.1M Tris-HCl, 50 mM glycine, 8 mM ⁇ -mercaptoethanol, 8M urea, 0.2M PMSF, pH 8.0) and stirred overnight, centrifuged for 20 min. at 48000 ⁇ g and the supernatant collected for refolding.
  • a denaturing buffer 0.1M Tris-HCl, 50 mM glycine, 8 mM ⁇ -mercaptoethanol, 8M urea, 0.2M PMSF, pH 8.0
  • Refolding was performed by dialysing (24 kDa MWCO) the supernatant against a large volume of refolding buffer at 4° C. with decreasing amounts of urea to slowly remove the denaturing agent (0.1M Tris-HCl, 1 mM EDTA, 0.2 mM PMSF, pH 8.0 supplemented with 4M, 2M, 1M and 0M urea each for 24 hrs).
  • the last step of dialysis was carried out against PBS for 24 hours.
  • the refolded protein was freed from precipitations by centrifugation and subsequently purified by FPLC with a GST binding column (GSTrap FF, Amersham Biosciences, GE Healthcare Europe, Otelfingen, Switzerland).
  • the GST tag of purified trTIMP-3-GST fusion protein was cleaved of by incubation with thrombin for 24 hours and purified by FPLC from GST with the GSTrap FF column and from thrombin with a HiTrap Benzamidine FF column (Amersham Biosciences, GE Healthcare Europe, Otelfingen, Switzerland). Overall yield of trTIMP-3 was about 1.5 mg/l of bacterial culture.
  • the activity of purified trTIMP-3 was assessed by MMP-2 zymography in which an equimolar amount of trTIMP-3 completely inhibits MMP-2 activity indicating a nearly 100% activity of the purified protein. Further experiments could be used to demonstrate the aggrecanase inhibiting activity of trTIMP-3 and the targeting specificity to articular cartilage in a similar fashion as shown for the nanoparticles described herein. In addition, its therapeutic potential to prevent cartilage degradation could be demonstrated in a suitable animal model, e.g., in a mouse knee instability model.
  • certain embodiments are directed to vectors for expression of a therapeutic protein of interest, e.g., a therapeutic agent and a polypeptide ligand.
  • Nucleic acids encoding a polypeptide can be incorporated into vectors.
  • a vector is a replicon, such as a plasmid, phage, or cosmid, into which another nucleic acid segment may be inserted so as to bring about replication of the inserted segment.
  • Vectors of the invention typically are expression vectors containing an inserted nucleic acid segment that is operably linked to expression control sequences.
  • An expression vector is a vector that includes one or more expression control sequences
  • an expression control sequence is a DNA sequence that controls and regulates the transcription and/or translation of another DNA sequence.
  • Expression control sequences include, for example, promoter sequences, transcriptional enhancer elements, and any other nucleic acid elements required for RNA polymerase binding, initiation, or termination of transcription.
  • “operably linked” means that the expression control sequence and the inserted nucleic acid sequence of interest are positioned such that the inserted sequence is transcribed (e.g., when the vector is introduced into a host cell).
  • a DNA sequence is operably linked to an expression-control sequence, such as a promoter when the expression control sequence controls and regulates the transcription and translation of that DNA sequence.
  • operably linked includes having an appropriate start signal (e.g., ATG) in front of the DNA sequence to be expressed and maintaining the correct reading frame to permit expression of the DNA sequence under the control of the expression control sequence to yield production of the desired protein product.
  • vectors include: plasmids, adenovirus, Adeno-Associated Virus (AAV), Lentivirus (FIV), Retrovirus (MoMLV), and transposons.
  • AAV Adeno-Associated Virus
  • FMV Lentivirus
  • Retrovirus MoMLV
  • transposons transposons.
  • promoters that could be used including, but not limited to, constitutive promoters, tissue-specific promoters, inducible promoters, and the like. Promoters are regulatory signals that bind RNA polymerase in a cell to initiate transcription of a downstream (3′ direction) coding sequence.
  • Molecules for viscosupplementation may be conjugated with polypeptide ligands described herein to form a conjugate for enhanced delivery and effect in the cartilage.
  • the formation of such conjugates is within the skill of ordinary artisans and various techniques are known for accomplishing the conjugation, with the choice of the particular technique being guided by the materials to be conjugated.
  • Such conjugates may be delivered systemically or locally, e.g., orally or by injection to a joint.
  • hyaluronic acid conjugation with a polypeptide ligand disclosed herein may prolong the retention time of hyaluronic acid in the joint and therefore enhance the efficacy of intra-articular viscosupplementation with hyaluronic acid.
  • Conjugation of the polypeptide to hyaluronic acid can be performed either directly as described above, or by the use of a polymer linker.
  • polymer linkers are biocompatible hydrophilic polymers, including polymers free of amino-acids.
  • a polymer linker may be a polyethylene glycol (PEG).
  • Hyaluronic acid can be functionalized with acrylated PEG-Arg-Gly-Asp conjugates created by Michael-type addition chemistry (Park et al. Biomaterials 2003; 24:893-900).
  • polymers described herein for use with the polypeptide ligands may be free of amino acids, meaning that such polymers do not contain a natural or synthetic amino acid.
  • the conjugate is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient.
  • the conjugate is produced using a cell, either a procaryotic or a eucaryotic cell, as in the case of a biopolymer.
  • transfected cells are introduced into a patient.
  • the site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • a small gene delivery system will have advantages with respect to penetrating the dense matrix of cartilage tissue.
  • the delivery system uses significantly condensed DNA, to enter the cartilage matrix and transfect non-dividing quiescent chondrocytes or other cells.
  • polypeptide ligands are attached to polymers which contain a nuclear localisation sequence to form a conjugate and are used to condense DNA to overcome challenges to gene transfection in chondrocytes embedded in the cartilage matrix.
  • the conjugate associated with nucleic acids encoding a therapeutic polypeptide is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient.
  • a conjugate is produced using a cell, either a procaryotic or a eucaryotic cell, as in the case of a biopolymer.
  • transfected cells are introduced into a patient.
  • the site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • Polypeptides as described herein can be attached to other polymers through bioconjugation.
  • the formation of such conjugates is within the skill of ordinary artisans and various techniques are known for accomplishing the conjugation, with the choice of the particular technique being guided by the materials to be conjugated.
  • the agent is attached to a soluble polymer, and may be administer to a patient in a pharmaceutically acceptable form.
  • a drug may be encapsulated in polymerosomes or vesicles or covalently attached to polymers. In the latter case, drugs are attached to the polymer backbone with a degradable site-specific spacer or linker (Lu et al. J Control Release 2002; 78:165-73).
  • soluble hydrophilic biocompatible polymers may be used to ensure that the conjugate is soluble and will be bioavailable after introduction into the patient.
  • soluble polymers are polyvinyl alcohols, polyethylene imines, and polyethylene glycols (a term including polyethylene oxides) having a molecular weight of at least 100, 400, or between 100 and 400,000 (with all ranges and values between these explicit values being contemplated).
  • Solubility refers to a solubility in water or physiological saline of at least 1 gram per liter.
  • Biodegradable polymers may also be used, e.g., polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polycaprolactones, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, and polycyanoacylates.
  • a polypeptide-polymer association e.g., a conjugate
  • a polypeptide-polymer association is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient.
  • the site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • polypeptides ligands were discovered based on their binding affinity for the cartilage matrix but are not specifically bound to the articular surface. Therefore, besides embodiments such as targeting the cartilage matrix by therapeutic agent delivery systems or with engineered fusion proteins, the polypeptides are particularly well suited to target a defect because extracellular matrix is exposed at the defect. This feature is useful for delivering biomaterials for gene, protein and/or cell delivery to mediate cartilage defect repair. Accordingly, embodiments include treating a defect in an articular cartilage using one of the embodiments set forth herein. In fact, giving a biomaterial the capability of adhering or otherwise specifically binding to a cartilage defect does not only allow for injectable defect repair strategies but also may enhance retention of the biomaterial in the cartilage defect.
  • a variety of chemical schemes can be used to incorporate the cartilage-binding polypeptide into the biomaterial.
  • a material as described by Sawhney et al. a chemical approach for incorporation as described by Hern et al. can be employed (Sawhney et al. Macromolecules 1993; 26:581-587 and Hern et al. J. Biomed. Mater. Res. 1998; 39:266-276).
  • a material as described by Lutolf et al. a chemical approach for incorporation as described therein can be employed (Lutolf et al. Nature Biotechnol. 2003; 21:513-518).
  • the modification of such biomaterials is within the skill of ordinary artisans and various techniques are known for accomplishing the modification, with the choice of a particular technique being guided by the biomaterial and peptides to be conjugated.
  • Specific binding generally refers to a molecule that binds to a target with a relatively high affinity compared to non-target tissues, and generally involves a plurality of non-covalent interactions, such as electrostatic interactions, van der Waals interactions, hydrogen bonding, and the like.
  • Specific binding interactions characterize antibody-antigen binding, enzyme-substrate binding, and specifically binding protein-receptor interactions; while such molecules may bind tissues besides their targets from time to time, such binding is said to lack specificity and is not specific binding.
  • the peptides of SEQ ID NOs 1, 2, and 3 may bind non-cartilage tissues in some circumstances but such binding has been observed to be non-specific, as evidenced by the much greater binding of the peptides to the targeted tissue as opposed to surrounding joint tissues (data not shown).
  • embodiments include biomaterials comprising at least one of the ligands disclosed herein that are used to fill or augment a defect in a cartilage.
  • a defect refers to a void in a surface (e.g., a pit, tear, or hole) or a pathological discontinuity in a surface (e.g., a tear or eroded member).
  • Fill refers to essentially filling or covering the defect or bridging the discontinuity.
  • Augment refers to at least partial filling.
  • the biomaterial is a solid prior to placement in a patient, while in other embodiments the material is made is situ, meaning it is formed from precursors at the site of the defect.
  • biomaterials for cartilage defects may be supplemented with a ligand or other embodiment set forth herein.
  • examples of such biomaterials include U.S. Pat. Nos. 5,874,500, and 5,410,016, and which include materials formed by in-situ polymerization.
  • Biomaterials for targeting cartilage defects i.e. adhering to a cartilage defect, are suited for the delivery of therapeutic agents to mediate cartilage repair such as growth factors and for cell delivery to the repair site.
  • the use of a biomaterial for cell delivery which adheres to the defect by the use of polypeptide ligands (SEQ ID NO:1 through 3) and acts as a morphogenic guide may improve cartilage defect repair.
  • nucleic acid refers to both RNA and DNA, including siRNA, shRNA, miRNA, cDNA, genomic DNA, synthetic (e.g., chemically synthesized) DNA, as well as naturally occurring and chemically modified nucleic acids, e.g., synthetic bases or alternative backbones.
  • a nucleic acid molecule can be double-stranded or single-stranded (i.e., a sense or an antisense single strand).
  • An isolated nucleic acid refers to a nucleic acid that is separated from other nucleic acid bases that are present in a genome, including nucleic acids that normally flank one or both sides of a nucleic acid sequence in a vertebrate genome (e.g., nucleic acids that flank a gene).
  • a conservatively substituted nucleic acid refers to the substitution of a nucleic acid codon with another codon that encodes the same amino acid and also refers to nucleic acids that encode conservatively substituted amino acids, as described herein with respect to polypeptides.
  • the combination of potential codons for a polypeptide of only about six residues is manageably small.
  • nucleic acid sequences set forth herein are intended to represent both DNA and RNA sequences, according to the conventional practice of allowing the abbreviation “T” stand for “T” or for “U”, as the case may be, for DNA or RNA.
  • Polynucleotides are nucleic acid molecules of at least three nucleotide subunits.
  • Polynucleotide analogues or polynucleic acids are chemically modified polynucleotides or polynucleic acids.
  • polynucleotide analogues can be generated by replacing portions of the sugar-phosphate backbone of a polynucleotide with alternative functional groups.
  • Morpholino-modified polynucleotides referred to herein as “morpholinos,” are polynucleotide analogues in which the bases are linked by a morpholino-phosphorodiamidate backbone (see, e.g., U.S. Pat. Nos. 5,142,047 and 5,185,444).
  • polynucleotide analogues include analogues in which the bases are linked by a polyvinyl backbone, peptide nucleic acids (PNAs) in which the bases are linked by amide bonds formed by pseudopeptide 2-aminoethyl-glycine groups, analogues in which the nucleoside subunits are linked by methylphosphonate groups, analogues in which the phosphate residues linking nucleoside subunits are replaced by phosphoroamidate groups, and phosphorothioated DNAs, analogues containing sugar moieties that have 2′ O-methyl group).
  • PNAs peptide nucleic acids
  • Polynucleotides of the invention can be produced through the well-known and routinely used technique of solid phase synthesis. Alternatively, other suitable methods for such synthesis can be used (e.g., common molecular cloning and chemical nucleic acid synthesis techniques). Similar techniques also can be used to prepare polynucleotide analogues such as morpholinos or phosphorothioate derivatives. In addition, polynucleotides and polynucleotide analogues can be obtained commercially.
  • examples of pharmaceutically acceptable compositions are salts that include, e.g., (a) salts formed with cations such as sodium, potassium, ammonium, etc.; (b) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid (c) salts formed with organic acids e.g., for example, acetic acid, oxalic acid, tartaric acid; and (d) salts formed from elemental anions e.g., chlorine, bromine, and iodine.
  • salts that include, e.g., (a) salts formed with cations such as sodium, potassium, ammonium, etc.; (b) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid (c) salts formed with organic acids e.g., for example, acetic acid, oxalic acid, tartaric acid; and (d) salts formed from elemental anions e.g., chlorine, bromine
  • Pharmaceutically acceptable carriers or excipient may be used to deliver embodiments as described herein.
  • Excipient refers to an inert substance used as a diluent or vehicle for a therapeutic agent.
  • Pharmaceutically acceptable carriers are used, in general, with a compound so as to make the compound useful for a therapy or as a product.
  • a pharmaceutically acceptable carrier is a material that is combined with the substance for delivery to an animal.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • the carrier is essential for delivery, e.g., to solubilize an insoluble compound for liquid delivery; a buffer for control of the pH of the substance to preserve its activity; or a diluent to prevent loss of the substance in the storage vessel.
  • the carrier is for convenience, e.g., a liquid for more convenient administration.
  • Pharmaceutically acceptable salts of the compounds described herein may be synthesized according to methods known to those skilled in this arts.
  • a pharmaceutically acceptable composition has a carrier, salt, or excipient suited to administration to a patient.
  • inert components of such compositions are biocompatible and not toxic.
  • the compounds described herein are typically to be administered in admixture with suitable pharmaceutical diluents, excipients, extenders, or carriers (termed herein as a pharmaceutically acceptable carrier, or a carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
  • suitable pharmaceutical diluents, excipients, extenders, or carriers suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices.
  • the deliverable compound may be made in a form suitable for oral, rectal, topical, intravenous injection, intra-articular injection, or parenteral administration.
  • Carriers include solids or liquids, and the type of carrier is chosen based on the type of administration being used. Suitable binders, lubricants, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents may be included as carriers, e.g., for pills.
  • an active component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, gelatin, agar, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like.
  • the compounds can be administered orally in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions.
  • the active compounds can also be administered parentally, in sterile liquid dosage forms. Buffers for achieving a physiological pH or osmolarity may also be used.

Abstract

Ligands that specifically bind to articular cartilage tissues are disclosed, including uses for targeting therapeutics towards articular cartilage tissue and new materials for articular cartilage. The ligands are effective in vivo to target therapeutic materials to articular cartilage.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. Ser. No. 11/545,819 filed Oct. 10, 2006, which is hereby incorporated herein by reference.
  • TECHNICAL FIELD
  • The technical field of the invention is generally related to delivery of therapeutic agents to cartilage tissues using polypeptides that specifically bind cartilage.
  • BACKGROUND
  • Cartilage lesions are common and can pose difficulties both in diagnosis and treatment. A lesion can either be a defect or a focal cartilage degradation without visible disruption of the cartilage matrix. Such lesions can result from an injury as in sports, disease, or aging. The prognosis of an articular cartilage defect varies according to age, mechanism of injury, site, size, associated injuries and treatment received.
  • SUMMARY OF THE INVENTION
  • The invention, however, provides treatments for cartilage injury. Some aspects of the inventions are substantially pure polypeptides comprising an amino acid sequence of WYRGRL (SEQ ID NO:1), DPHFHL (SEQ ID NO:2), or RVMLVR (SEQ ID NO:3), or a conservative substitution thereof, or a nucleic acid encoding the same. Such polypeptides specifically bind cartilage tissue. Such polypeptides may also include a therapeutic agent.
  • Some inventive methods are related to treating cartilage of a mammal comprising administering to the mammal a pharmaceutically acceptable composition that comprises a nucleic acid encoding a polypeptide that specifically binds a cartilage tissue. Such polypeptide may also encode a therapeutic agent that, for example, treats a joint or tissue.
  • Other aspects of the invention relate to a delivery system for delivering a therapeutic agent comprising: a substantially purified preparation that comprises a pharmaceutically acceptable excipient, a therapeutic agent, and a polypeptide ligand comprising an amino acid sequence in the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, and conservative substitutions thereto. The polypeptide ligands may specifically bind to cartilage tissue for targeted delivery of the therapeutic agent to cartilage tissues. The therapeutic agent may comprise, for example, a drug, a visualization agent, or a therapeutic polypeptide. The delivery system may include, for example, a collection of nanoparticles having an average diameter of between about 10 nm and about 200 nm, wherein the nanoparticles comprise the therapeutic agent and the polypeptide ligand.
  • Other embodiments relate to a biomaterial comprising a polymer and a substantially pure polypeptide comprising an amino acid sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, or a conservative substitution thereof, wherein the polypeptide specifically binds to cartilage tissue and the polymer is free of amino acids and has a molecular weight of at least 400. A variant of WYRGRL (SEQ ID NO:1) is WYRGRLC (SEQ ID NO:4), with the C-terminal residue being used as a chemical linker.
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1 a is a bar graph that shows amplification of three phage clones identified by biopanning and demonstrates that they all grow at equal rates comparable to the random library. Final selection of C1-3 was therefore not affected by differences in growth during amplification.
  • FIG. 1 b is a bar graph that demonstrates the competitive binding assay in which only phage clone C1-3 and C1-C1 could be recovered.
  • FIG. 1 c is a bar graph that shows binding specificity of C1-3 and C1-C1 to articular cartilage. Binding of these phage clones to synovial membrane resulted in a lower phage recovery by two orders of magnitude, reflecting non-specific binding. Error bars indicate mean±standard deviation from three independent experiments.
  • FIG. 2 is a graph showing inhibition of cartilage binding of C1-3 by the synthetic polypeptide WYRGRLC (SEQ ID NO:4). Data represents the percentage of maximal phage binding of clone C1-3 obtained in the absence of synthetic polypeptide. Error bars indicate mean±standard deviation from three independent experiments.
  • FIG. 3 is a bar graph showing inhibition of cartilage binding of C1-3 phage by 101 μM of synthetic polypeptide vs. WYRGRLC (SEQ ID NO:4)—PPS nanoparticles and 10 μM of synthetic mismatch polypeptide. WYRGRLC (SEQ ID NO:4)—PPS nanoparticles exhibit similar binding than the synthetic polypeptide, whereas the synthetic mismatch polypeptide does not result in a significant decrease of phage titer. Error bars indicate mean±standard deviation from three independent experiments.
  • FIG. 4 is a bar graph showing relative accumulation in articular cartilage in vivo of nanoparticles decorated with WYRGRLC (SEQ ID NO:4) compared to control nanoparticles. Error bars indicate mean±standard deviation from three independent experiments.
  • DETAILED DESCRIPTION Introduction
  • Ligands that specifically bind to articular cartilage tissues have been discovered.
  • These articular cartilage tissue-binding ligands have given rise to new techniques to target therapeutics towards articular cartilage tissue and new materials for treatment of articular cartilage defects. The ligands are effective in vivo to target therapeutic materials to articular cartilage.
  • Three of the cartilage tissue-binding ligands are polypeptides with the amino acid sequence of WYRGRL (SEQ ID NO:1), DPHFHL (SEQ ID NO:2), or RVMLVR (SEQ ID NO:3). Other ligands are polypeptides with sequences that have conservative substitutions of one of SEQ ID NOs. 1, 2, or 3. Ligand is a term that refers to a chemical moiety that has specific binding to a target molecule. A target refers to a predetermined molecule, tissue, or location that the user intends to bind with the ligand. Thus targeted delivery to a tissue refers to delivering a molecule to the intended target tissue; a therapeutic agent delivered to a target may be intended to act on the target itself or on some other molecule or cell, e.g., a cell or tissue that is near the target.
  • One application of the cartilage tissue-binding ligands is for targeting therapeutic agents to articular cartilage, commonly referred to as intra-articular drug delivery when drugs are delivered. Intra-articular drug delivery has been termed a major challenge due to the short residence times of intra-articular injected drugs. Drugs injected by themselves tend to diffuse away rapidly or be otherwise rapidly taken up into the circulation, thus causing low bioavailability of the drug at the cartilage and unwanted systemic effects. Despite sustained-release drugs, only a few reports exist on the intra-articular use of any sustained-release formulations20-26. Mostly albumin and poly(lactic-co-glycolic acid) (PLGA) have been used as biocompatible and biodegradable polymers for this purpose either in the form of gels22 or as microspheres23-26. Such approaches rely, however, on degradation over time to achieve sustained-release within the joint, a strategy that is limited by the properties of available biomaterials, and which makes the drugs available only when the material degrades. Targeting the delivery system to articular cartilage with tissue-binding ligands makes the tissue itself a reservoir for drug release to the site of the disease process as opposed to release in the joint cavity.
  • The cartilage tissue-binding ligands are also useful to bind cartilage tissue in vitro for diagnostic, assay, or imaging purposes. For instance, sections of tissue may be exposed to cartilage tissue-binding ligands that are also bound to a fluorescent molecule or other imaging agent to visualize the location of the cartilage tissue. Or, for instance, the cartilage tissue-binding ligands may be used in affinity chromatography to isolate the tissue.
  • In some aspects, therefore, articular cartilage tissue-binding ligands described herein enable new techniques for controlled release by enabling sustained localization through specific binding to cartilage, and their accompanying formulations increase intra-articular bioavailability of delivered drugs or other therapeutic agents, which is beneficial for various disease processes, e.g., those involving the synovium such as rheumatoid arthritis or inflammation in clinically manifest osteoarthritis. Because convective transport of solutes into cartilage is impaired due to the inherent properties of this tissue27,28, the bioavailability of drugs in the cartilage matrix, which is the primary site of the disease process in osteoarthritis, can be enhanced by sustained release systems that reside in the matrix itself. Targeting of the cartilage matrix as described herein is therefore useful for general intra-articular therapeutic agent delivery, such as targeting of the cartilage matrix for delivery of therapeutic agents to treat cartilage degradation in osteoarthritis and protect cartilage in conditions like rheumatoid arthritis, bacterial and reactive arthritis.
  • The cartilage tissue-binding ligands are thus useful to direct therapeutic agents to a target. A therapeutic agent refers to a molecule for delivery to a target to accomplish a desirable medical or scientific function, and the term includes drugs and imaging agents. Examples of therapeutic agents are matrix metalloproteinase (MMP) inhibitors, aggrecanase inhibitors, COX-inhibitors and other non-steroidal anti-inflammatory drugs (NSAIDs), glucosamin, diacerhein, methotrexate, steroids, immunosuppressing drugs (rapamycin, cyclosporine), protein therapeutics (growth factors, tissue inhibitors of matrix metalloproteinases), and oligonucleotides (e.g., siRNA, shRNA, miRNA). Examples of imaging agents are fluorescent markers, radio-opaque materials, magnetic resonance imaging contrast agents, x-ray imaging agents, radiopharmaceutical imaging agents, ultrasound imaging agents, and optical imaging agents.
  • Ligand Discovery and Experimental Data
  • The development of phage display of random peptides on the minor coat proteins (pIII) of bacteriophages has allowed use of affinity purification, in a process called biopanning5, to identify specific peptides for precise targeting of multiple tissues, both in vitro6 and in vivo in animals and even humans7-9. As an example, Arap et al. have succeeded in mapping the human vasculature and specifically targeting the microvasculature of adipose tissue in mice. Another study identified a synovium-specific homing peptide by phage display. Human synovial grafts were transplanted into SCID mice and biopanning was carried out in vivo which resulted in the identification of phages with homing peptides specific for the microvascular endothelium of synovial tissue11.
  • In order to identify peptide sequences which bind to the articular cartilage extra-cellular matrix, it should first be appreciated that an in vivo approach to biopanning must address the challenges presented by the dense organization of extra-cellular matrix molecules. Indeed, the collagen II fiber network of articular cartilage has a reported mesh size of 60 nm in the superficial zone12 and largest gaps between the side chains of proteoglycan aggregates have been described to be as low as 20 nm13. Biopanning was therefore carried out not in vivo, but instead was adapted for use with ex vivo materials. Specifically, sliced bovine cartilage was used, wherein the extra-cellular matrix was exposed for affinity purification of binding phage virions.
  • Materials The phage display library fUSE5/6-mer based on filamentous phage strain fd-tet was received from the University of Missouri, Columbia. Cartilage grafts, synovial fluid and synovial membrane were harvested from bovine shoulders obtained from the local slaughterhouse. Cartilage grafts were stored in 0.1% sodium azide and protease inhibitors at 4° C. and used within 72 hours. Buffers and solutions used for phage-display screening: Blocking solution (0.1M NaHCO3, 1% BSA, pH 8.5), wash buffer (PBS, Tween 20 0.1-1%), elution solution (50 mM glycine-HCl, pH 2.0), neutralisation buffer (0.2M NaHPO4). Solvents and reagents for nanoparticle synthesis were purchased from Sigma-Aldrich (Buchs, Switzerland). All peptides (synthesis chemicals from Novabiochem, Lautelfingen, Switzerland) were synthesized on solid resin using an automated peptide synthesizer (CHEMSPEED PSW 1100, Augst, Switzerland) with standard F-moc chemistry. Purification was performed on a Waters ultrapurification system using a Waters ATLANTIS dC18 semi-preparative column and peptides collected according to their molecular mass analyzed by time-of-flight (TOF) mass spectrometry.
    Screening of phage-displayed combinatorial peptide library and binding assays Peptides for binding to the articular cartilage matrix were selected by exposing a fUSE5/6-mer library to bovine cartilage grafts, which provided 6.4×107 different phage clones with 6-amino acid linear-peptide inserts displayed on the minor coat protein of filamentous phage5,18. Cartilage grafts were harvested with 8 and 4 mm biopsy cutters (two-sided surface 1 cm2 and 0.25 cm2). A slice with the intact articular surface was removed to expose the phages only to the cartilage matrix below the surface. Affinity selection was preformed in polystyrene 48-well plates, which were blocked for nonspecific adhesion with a blocking solution containing 1% BSA for 2 hours prior to screening. A total of 5 screening rounds was carried out. In the first round 1013 and in subsequent rounds 1012 phage virions per ml were exposed to the cartilage graft, washed with PBS/Tween 20 and eluted at low pH. While the first round was supposed to give a high yield at low stringency, subsequent rounds were carried out with increasing stringency to select stronger binders. Conditions were increased from 4 hours of binding at RT and washing with PBS/0.1% Tween to 30 minutes binding at 37° C. with 220 RPM, washing with PBS/1% Tween 20 and the cartilage surface decreased from 1 cm2 to 0.25 cm2 in round five. Eluted phage was amplified overnight in E. coli strain TG1 in 2×YT medium and purified by two times PEG/NaCl (2M, 25%) precipitation. In the first round of screening, negative screens against the intact articular surface as well as synovial fluid (50% diluted in PBS to lower viscosity) were carried out in order to eliminate phages binding to these targets. Quantitative titer counts were obtained by spot titering of 15 μl of phage/bacterial culture onto LB-agar/tetracyclin plates and are given in transducing units (TU)/ml. The polypeptide sequences of affinity selected phage specific to the articular cartilage matrix was determined by DNA sequencing (Microsynth AG, Balgach, Switzerland) using the sequencing primer 5′-CAT GTA CCG TAA CAC TGA G (SEQ ID NO:5). Binding specificity was determined by exposing selected phage clones (108 TU/ml) to articular cartilage (0.25 cm2) with and without synovial fluid, to synovial membrane (3×4 mm, 0.24 cm2) and to polystyrene after blocking. Titer counts were obtained by spot titering. Competetive binding was probed by exposing a mix of selected phage clones and fUSE5/6-mer library (each 108 TU/ml) in the presence of synovial fluid to articular cartilage for 30 min. at 37° C., 220 RPM and washed with PBS/1% Tween 20. The phage clones were identified by DNA sequencing and the corresponding titer counts calculated. For competitive binding against free polypeptide or nanoparticles and to obtain a dose-response curve, 108 TU/ml of phage and its free polypeptide in different concentrations or nanoparticles were mixed, exposed to articular cartilage and titer counts determined. All experiments have been carried out in triplicate and repeated for confirmation.
    Nanoparticle Synthesis Poly(propylene sulfide) nanoparticles were prepared as described elsewhere14. Briefly, for nanoparticles between 30 and 40 nm, a monomer emulsion is prepared by dissolving 1.6% (w/v) of Pluronic F-127 (MW 12600) in 10 ml of degassed, double-distilled and filtered water. The system is continuously stirred and purged with argon for 60-90 min. Propylene sulfide is added at a Pluronics/monomer ratio of 0.4 (w/w). The initiator, pentaerythritol tetrathioester (TTE) (synthesized as described previously14), is deprotected by mixing with a molar equivalent of 0.5M sodium methanoate and stirred under argon for 5 minutes. The deprotected initiator is then added to the emulsion. Five minutes later, 60 μl of diaza[5.4.0] bicycloundec-7-ene (DBU) is added and the reaction stirred under inert conditions for 6 hours. Exposure to air yields disulfide crosslinking of the particle core. Particles are subsequently purified from remaining monomers and base by 2 days of repeated dialysis against ultrapure water through a membrane with a MWCO of 6-8 kDa (Spectra/Por). Free Pluronic F-127 is removed in a second dialysis step through 300 kDa membranes. Particle size is measured after dialysis by dynamic light scattering (ZETASIZER NANO ZS, Malvern Instruments, Malvern, UK). For preparation of surface-functionalized nanoparticles, polypeptides are conjugated to Pluronics-F127 prior to nanoparticle synthesis. In order to derivatize Pluronics F-127 with vinyl sulfone, 400 ml of toluene and 15 g of Pluronic F-127 were introduced in a 3-neck round bottom flask connected to a Soxhlett filled with glass wool and dry molecular sieves and a cooling tube. Pluronic was dried azeotropically during 4 h. The dried Pluronic in toluene was cooled in an ice-bath and sodium hydride was added in a 5 equimolar excess compared with Pluronic-OH-groups. The reaction was stirred for 15 minutes and divinyl sulfone was added in a 15 molar excess and the reaction was carried out in the dark for 5 days at RT under argon. The reaction solution was filtrated through a celite filter cake, concentrated by rotary evaporation and then precipitated 5 times in ice-cold diethylether. The polymer was dried under vacuum and stored under argon at −20° C. Derivatization was confirmed with 1H-NMR (CDCl3):=1.1 (m, PPG CH3), 3.4 (m, PPG CH), 3.5 (m, PPG CH2), 3.65 (m, PEG CH2), 6.1 and 6.4 (d, 1H each, CH2═CH—SO2—), 6.85 (q, 1H, CH2═CH—SO2—) ppm. A degree of end group derivatization of 88% was determined by 1H-NMR. All polypeptides were acetylated at the N-terminus to prevent reaction with the α-amine and synthesized with a cysteine at the C-terminus to be conjugated to Pluronics-di-vinyl sulfone by Michael-type addition via the free thiol29. For conjugation, 1.6 mM Pluronics-di-vinyl sulfone is stirred in triethanolamine buffer at pH 8.5 until it is completely dissolved and 2 mM of polypeptide is added and stirred for 3 hours at RT. Conjugation is confirmed by absent vinyl sulfone peaks by 1H-NMR in methanol. Nanoparticles are prepared as described above with 10% surface functionalisation corresponding to a fraction of 0.16% (w/v) of conjugated Pluronic. The presence of the polypeptide on the particle is confirmed by measuring the zeta potential by dynamic light scattering (ZETASIZER NANO ZS, Malvern Instruments, Malvern, UK) after particle synthesis. The nanoparticles were fluorescently labelled with 6-iodoacetamide fluorescein (6-IAF, Molecular Probes, Eugene, Oreg.) or Alexa FLUOR 488 maleimide by adding label at 1 mg/ml of nanoparticle solution in 10 mM Tris-HCl pH 8.5 and stirred in the dark for 2 h at room temperature. Unreacted label was quenched by adding 5 mg L-Cysteine. Purification was accomplished by dialysis for 24 hours against a membrane with MWCO of 24 kDa in 5 mM PBS with two buffer shifts.
    In vivo injection Labeled nanoparticles were injected into knee joints of 4-6 weeks old C57BL/6 mice at a concentration of 1% (w/v) in a volume of 5 μl using a 25 μl Hamilton syringe with Cheney reproducibility adapter and 30 G needles (animal experimentation protocol approved by the local review board, authorisation no. 1894). The mice were anesthetized with isoflurane. The animals were euthanized by CO2-asphyxiation after 24 hours. The knee joints were harvested and freed from surrounding muscle by microdissection.
    Confocal microscopy and image analysis The sections were analyzed by confocal laser scanning microscopy using a Zeiss LSM510 meta. Fluorescence was extracted by emission fingerprinting to reduce autofluorescence of cartilage tissues. The pinhole was adjusted using TETRASPECK fluorescent microspheres (Invitrogen, T14792, Carlsbad, Calif.). Images for analysis were obtained using a Zeiss 63× APOCHROMAT objective in 10 different locations per joint with a z-stack of 10 images each. Image deconvolution was accomplished by Huygens software. Image analysis was done by ImageJ.
  • Affinity Selection of Phage Display Library and Binding Assay
  • Cartilage grafts were incubated with the fUSES peptide on phage display library, which expressed linear 6-mer random peptides on minor coat proteins (pIII) with a diversity of 6.4×107. The sequences corresponding to the polypeptides displayed on the phage virions were determined by DNA sequencing after rounds 3 and 5 of affinity selection (panning). After round 3, sequencing did not reveal a consensus motif in the selected polypeptides. Sequencing of round 5 yielded three different phage clones C1-3 (having SEQ ID NO:1), C1-C1 (having SEQ ID NO:2) and C1-F1 (having SEQ ID NO:3), whereas C1-3 appeared in 94 out of 96 sequenced clones and C1-C1 and C1-F1 both only appeared once. To ensure that the selection of the three phage clones was not the result of differences in their amplification rates compared to other phages, overnight amplification of 106 particles/ml of the three selected clones and the random fUSE5 phage library was performed in a bacterial culture. As shown in FIG. 1 a, all amplification rates are equal to the rate of the random library. This suggests that the three phage clones were not selected in the panning process because of differences in their amplification speed, which would have biased the library in their favor. A competetive binding assay was carried out to assess the relative binding strength of the selected phage clones against each other. Equal amounts of the three phage clones and the random fUSES library were mixed and exposed to cartilage grafts (0.25 cm2). The corresponding titer counts of recovered phage virions were determined by DNA sequencing of 96 colonies in triplicate. It is demonstrated in FIG. 1 b that only C1-3 and C1-C1 have been recovered, thereby indicating their superior binding strength over C1-F1 and the random library. In addition, C1-3 has a higher titer count of almost one order of magnitude than C1-C1 which further demonstrates its dominant binding which was already suggested by its frequent appearance after round 5.
  • The binding specificity of C1-3 and C1-C1 to articular cartilage was evaluated by exposing the phage clones to articular cartilage (0.25 cm2) and synovial membrane (0.24 cm2) and comparing to random binding of the fUSES phage library. Furthermore, the effect of the presence of synovial fluid on phage binding was probed by adding an equal volume of bovine synovial fluid to the phages, which dilutes the synovial fluid by a factor of 2. FIG. 1 c shows that both C1-3 and C1-C1 exhibit specific binding to articular cartilage over synovial membrane by two orders of magnitude and that the addition of synovial fluid does not yield a significant drop in phage binding to articular cartilage. Binding of the specific phage clones to synovial membrane seems to reflect background phage binding as the random phage library fUSES bound to the synovial membrane to the same extent. In addition, phage titers of fUSE5 to articular cartilage were at the same level as to synovial membrane, further indicating specific binding of C1-3 and C1-C1. Phage was exposed to polystyrene wells after blocking them with BSA because it is contained in the plasticware in which affinity selection was carried out. No background phage binding to polystyrene was detectable, however.
  • Affinity selection of fUSE5/6-mer phage display library resulted in the discovery the phage clone C1-3 (polypeptide WYRGRL, SEQ ID NO:1) which exhibits both binding specificity to articular cartilage and dominant binding compared to other phage clones. Specific binding in the context of a polypeptide refers to the binding of the polypeptide specifically to the target of interest as opposed to other molecules.
  • Competetive Binding of C1-3 Against Free Polypeptides WYRGRLC and YRLGRWC
  • Based on affinity selection and the binding assays reported herein, the 6-mer polypeptide insert WYRGRL (SEQ ID NO:1) of clone C1-3 as well as its scrambled mismatch YRLGRW (SEQ ID NO:6) were synthesized on solid resin using standard Fmoc chemistry. The N-terminal amino acids are acetylated and a cysteine was added to the C-terminus of the polypeptides for bioconjugation to vinyl sulfone by Michael-type addition via the free thiol. In order to further characterize the binding properties of WYRGRLC (SEQ ID NO:4) to articular cartilage, a competetive binding assay against the phage clone C1-3 was performed by exposing the C1-3 and the WYRGRLC (SEQ ID NO:4) to the cartilage. A dose-response curve was determined by serial dilutions of the polypeptide ranging from 50 nM to 10 μM and mixing them with 108 TU/ml of phage. The titer counts of phage recovered gradually decreased by two orders of magnitude as the concentration of the free polypeptide in solution was increased (FIG. 2). An IC50 of about 200 nM can be estimated from the curve in FIG. 2.
  • Conjugation of Polypeptide to Pluronic F-127 and Nanoparticle Synthesis
  • In order to functionalize poly(propylene sulfide) (PPS) nanoparticles, Pluronic F-127 was derivatized with vinyl sulfone. The polypeptides were conjugated to Pluronic-di-vinyl sulfone via the free thiol in the C-terminal cysteine by Michael-type addition.
  • Conjugation was confirmed by 1H-NMR in methanol by the absence of peaks specific to vinyl sulfone. PPS nanoparticles were then prepared by inverse emulsion polymerization with Pluronic F-127 (90%) and polypeptide-conjugated Pluronic F-127 (10%) serving as the emulsifier. Because Pluronic as the emulsifier remains on the particle surface, the conjugated polypeptide is displayed on the surface of the nanoparticles, thereby adding the targeting functionality of the polypeptide to the particles. Size measurements by dynamic light scattering (ZETASIZER NANO ZS, Malvern Instruments, Malvern, UK) revealed a size by volume of 38 nm for PPS particles displaying WYRGRLC (SEQ ID NO:4) (polydispersity index PDI 0.221), 31 nm for particles displaying YRLGRC (SEQ ID NO:7) PDI 0.412) and 37 nm for non-conjugated PPS particles (PDI 0.212). The zeta-potential of non-conjugated PPS nanoparticles is about neutral (−2.64±8.97 mV).
  • Due to the positive charges of the polypeptide, the zeta-potential of conjugated nanoparticles shifted to +17.8±3.45 mV, which further confirms the presence of the polypeptide on the nanoparticle surface.
  • WYRGRLC (SEQ ID NO:4)—PPS nanoparticles at 2% (w/v) were subjected to a competetive binding assay against the free polypeptides WYRGRLC (SEQ ID NO:4) and YRLGRC (SEQ ID NO:7) at concentrations of 10 μM each. Accordingly, phage clone C1-3 was exposed to the cartilage in the presence of WYRGRLC (SEQ ID NO:4)—PPS nanoparticles, WYRGRLC (SEQ ID NO:4) or YRLGRC (SEQ ID NO:7), and the amount of C1-3 phage binding to the cartilage relative to a control C1-3 phage without competitive inhibitors was measured. The results in FIG. 3 show that WYRGRLC (SEQ ID NO:4)—PPS nanoparticles exhibit similar binding as the corresponding free polypeptide (WYRGRLC (SEQ ID NO:4)—PPS10.4±6% and WYRGRLC (SEQ ID NO:4)13.9±2.8% of control), whereas the YRLGRC (SEQ ID NO:7) did not bind competetively and thus did not result in a significant drop of phage titer (92±12% of control). Conjugated PPS nanoparticles at a degree of surface functionalisation of 10% at 2% (w/v) therefore seem to have similar binding to articular cartilage as the free polypeptide WYRGRLC (SEQ ID NO:4) at 10 μM.
  • Active Targeting of Articular Cartilage In Vivo
  • WYRGRLC (SEQ ID NO:4)—PPS and PPS nanoparticles were labeled with 6-IAF and dialysed for 2 days with at least 2 buffer shifts to ensure that no free label is still in the solution. A volume of 5 μl of the nanoparticles was injected into the knee joints of 4-6 weeks old C57BL/6 mice. Three mice were injected with WYRGRLC (SEQ ID NO:4)—PPS in the right and PPS particles in the left knee joint. In order to do reproducible injections a 250 Hamilton syringe (Hamilton Europe, Bonaduz, Switzerland) with a Cheney reproducibility adapter was used for antero-lateral parapatellar injection with a 30 G needle. Cryosections which were obtained after 24 hrs were analysed by confocal laser scanning microscopy. Quantification of fluorescent dots per cartilage volume as determined by sampling of z-stacks with 10 planes in 10 different locations per joint revelead an increase in particle accumulation from 29.0±1.5% for PPS particles to 83.8±4.0% for WYRGRLC (SEQ ID NO:4)—PPS particles (FIG. 4). While there is an obvious favorable accumulation of functionalized nanoparticles in the articular cartilage matrix after 24 hours, nanoparticles accumulate in the whole joint at a concentration of 2% (w/v) and enter meniscal and ligamentous tissues in addition to the synovial membrane.
  • Discussion
  • Several methods exist for affinity selection of binding proteins or polypeptides such as phage displays, yeast surface display15, mRNA display16 or peptide-on-bead display17. Herein, phage display using the fUSE5/6-mer library based on the filamentous phage vector fd-tet18 was used to select short peptides which bind to the articular cartilage matrix. Embodiments of the invention include using affinity selection of binding proteins or peptides against cartilage ex vivo.
  • In this case, biopanning was carried out against slices of bovine cartilage. Conditions in the binding step were chosen with increasing stringency from round 1 to 5 in order to favor binding of high affinity polypeptides. The selected sequences obtained from DNA sequencing of 96 clones have been evaluated in a competetive binding assay.
  • Two sequences C1-3 and C1-C1 exhibited stronger competetive binding than C1-F1 and random phages from the fUSE5 library (FIG. 1 b). Therefore, specificity of binding for C1-3 and C1-C1 to cartilage was further assessed. The phage clones were subjected to physiological conditions (37° C. and shaking) and binding specificity probed for cartilage, cartilage in the presence of synovial fluid and the synovial membrane. Phage titers of both C1-3 and C1-C1 were higher for cartilage than for synovial membrane by two orders of magnitude. More importantly, binding to the cartilage target was not impaired by the addition of synovial fluid (FIG. 1 c). This is likely to be the result of the negative screening which was carried out during the first round of biopanning. In order to eliminate phages with polypeptide sequences that potentially bind to constituents of the synovial fluid, the first screening was carried out in the presence of synovial fluid, which was discarded including the phages before the binding phages were eluted off the cartilage slice. In order assess the relative binding affinity of the polypeptide sequence, the corresponding polypeptide WYRGRLC (SEQ ID NO:4) was synthesized. In a competetive binding assay against the phage C1-3 displaying WYRGRL (SEQ ID NO:1) (108 TU/ml) an IC50 in the high nanomolar range of about 200 nM can be demonstrated (FIG. 2).
  • Binding specificity was conferred by the polypeptide sequence rather than the net positive charge of the polypeptide which sticks non-specifically to negatively charged proteoglycans. This is demonstrated in FIG. 3 in that 10 μM of WYRGRLC (SEQ ID NO:4) resulted in a decrease of phage titers close to 100fold. By contrast, 10 μM of YRLGRC (SEQ ID NO:7), which comprises the same amino acids just in scrambled order, did not result in a significant decrease in phage titer as compared to control phage titers without polypeptide. Thus WYRGRLC (SEQ ID NO:4) is a short polypeptide with specific binding to articular cartilage which shows a dose dependent decrease in competetive binding against phage C1-3. The other polypeptides, DPHFHL (SEQ ID NO:2) and RVMLVR (SEQ ID NO:3), which were discovered using the same experimental methods used for WYRGRL, could also be shown to have specific binding using these same techniques.
  • The polypeptides were synthesized such that they contain a cysteine at the C-terminus. The free thiol of cysteine is used for bioconjugation by Michael-type addition to Pluronic-di-vinyl sulfone which serves as the emulsifier in nanoparticle synthesis and therefore remains displayed on the particle surface. While this is a straightforward scheme for surface functionalisation of nanoparticles synthesized by inverse emulsion polymerization, conjugation of Pluronic requires excess of polypeptide despite the favorable kinetics of Michael-type addition of free thiols to vinyl sulfone, if conjugation close to 100% is to be achieved as evidenced by 1H-NMR. In addition, some Pluronic is always lost during nanoparticle synthesis. Alternative synthetic schemes therefore add surface functionality to already-synthesized nanoparticles in order to limit the amount of polypeptide needed.
  • Presence of the polypeptide on the nanoparticle surface as indicated by a shift in zeta-potential from neutral to positive was confirmed by competitive binding. WYRGRLC(SEQ ID NO:4)—PPS nanoparticles with a degree of surface functionalisation of 10% (w/w of total Pluronic used) at a concentration of 2% (w/v) against phage clone C1-3 (108 TU/ml) to articular cartilage resulted in a similar decrease of phage titers as 10 μM of free polypeptide WYRGRLC (SEQ ID NO:4, FIG. 3).
  • Targeting the extra-cellular matrix of articular cartilage essentially depends on the ability of the drug delivery system to enter the cartilage matrix and to stay there. In passive targeting, the distribution of nanoparticles in the joint is mainly governed by the capability of tissue penetration and cellular uptake. While larger particles do not enter, smaller ones are able to penetrate and reside in the cartilage ECM. It is demonstrated in herein that nanoparticles (in this case, nanoparticles with a mean volume diameter of 36 and 38 nm) are able to enter the articular cartilage ECM and meniscal tissue in addition to the synovium. It is consistent with the literature that small particles possess the ability to enter the cartilage ECM. It has been demonstrated that adeno-associated viruses (AAV) with a mean diameter of 20-25 nm enter the articular cartilage matrix up to a depth of penetration of 450 μm in normal and 720 μm in degraded cartilage19. While both surface-functionalised and non-functionalised PPS nanoparticles enter the articular cartilage matrix, there is a marked increase in the accumulation of WYRGRLC (SEQ ID NO:4)—PPS nanoparticles over non-functionalised and therefore non-targeted PPS nanoparticles 24 hours after intra-articular injection into the knee joint of mice (FIG. 4). WYRGRLC(SEQ ID NO:4)—PPS nanoparticles therefore exhibit specific targeting capability for the articular cartilage matrix. Although the basic research has been performed with bovine cartilage, the polypeptide sequences described herein are expected to bind to human articular cartilage due to the general homology of these tissues. Bovine cartilage is an accepted model in cartilage research due to the very limited availability of healthy human cartilage.
  • Polypeptide Ligands Specific for Articular Cartilage
  • Three of the cartilage tissue-binding ligands are polypeptides with the amino acid sequence of WYRGRL (SEQ ID NO:1), DPHFHL (SEQ ID NO:2), or RVMLVR (SEQ ID NO:3). Other ligands are polypeptides or functional polypeptides with sequences that have conservative substitutions of one of SEQ ID NOs. 1, 2, or 3.
  • Certain embodiments are directed to the subset of polypeptides that have a certain percentage identity to the disclosed sequences, or a certain degree of substitution, with the subset being primarily, or only, functional polypeptides.
  • The binding activity of a polypeptide to cartilage may be determined simply by following experimental protocols as described herein. For instance, a polypeptide variant of one of the polypeptide ligands may be labeled with a marker (e.g., radioactive or fluorescent) and exposed to bovine cartilage to determine its binding affinity using well-known procedures. A binding assay may be performed using a simple fluorescence readout using a plate reader by labeling polypeptide variants with a fluorescent marker, e.g., 6-fluorescein iodoacetamide which reacts with the free thiol of the cysteine. Using such a method, the binding strengths of polypeptide variants relative to e.g., WYRGRLC (SEQ ID NO:4) under given physiological conditions can be determined, e.g., sequences made using conservative substitutions, truncations of the sequences to 5 or less amino acids, addition of flanking groups, or changes or additions for adjusting sequences for solubility in aqueous solution.
  • Polypeptides of various lengths may be used as appropriate for the particular application. In general, polypeptides that contain the polypeptide ligand sequences will exhibit specific binding if the polypeptide is available for interaction with cartilage in vivo. Protein folding can affect the bioavailability of the polypeptide ligands. Accordingly, certain embodiments are directed to polypeptides that have a polypeptide ligand but do not occur in nature, and certain other embodiments are directed to polypeptides having particular lengths, e.g., from 6 to 3000 residues, or 6-1000, or 6-100, or 6-50; artisans will immediately appreciate that every value and range within the explicitly articulated limits is contemplated. Moreover, the lower limits may be 4 or 5 instead of 6.
  • While polypeptides of 6 residues were extensively, tested, variants that have 3, 4, or 5, residues can also be active and exhibit specific binding, as well as conservative substitutions thereof. Accordingly, every contiguous 3, 4, and 5 residues in each sequence can be rapidly screened and tested for binding using the methods set forth herein, e.g., using the sequencing and binding assays, or with competitive inhibition. Thus, in the case of SEQ ID NO:1, with W being the first residue and L the sixth residue, binding activity may be expected in the three residues 1-3, 2-4, 3-5, and 4-6; for four residues, binding activity may be expected in 1-4, 2-5, and 3-6; for five residues, binding activity may be expected in positions 1-5 or 2-6. The ordinary artisan, after reading this disclosure, will be able to quickly assay this limited number of sequences. While some decrease in binding activity might be observed when the 6-residue sequences are truncated, a core group is expected to exhibit substantial binding. This expectation is based on general observations made with binding moieties in these arts. For example, in phage display experiments, the peptide sequences often exhibit a consensus motif which usually does not involve all residues displayed on the phage virion, e.g., in Arap et al. (Nature Medicine 2002; 8:121). Less stringent conditions or sequencing of previous rounds are more likely to give a consensus motif in different peptide sequences than one very strong binding sequence.
  • Certain embodiments provide various polypeptide sequences and/or purified polypeptides. A polypeptide refers to a chain of amino acid residues, regardless of post-translational modification (e.g., phosphorylation or glycosylation) and/or complexation with additional polypeptides, synthesis into multisubunit complexes, with nucleic acids and/or carbohydrates, or other molecules. Proteoglycans therefore also are referred to herein as polypeptides. As used herein, a “functional polypeptide” is a polypeptide that is capable of promoting the indicated function. Polypeptides can be produced by a number of methods, many of which are well known in the art. For example, polypeptides can be obtained by extraction from a natural source (e.g., from isolated cells, tissues or bodily fluids), by expression of a recombinant nucleic acid encoding the polypeptide, or by chemical synthesis. Polypeptides can be produced by, for example, recombinant technology, and expression vectors encoding the polypeptide introduced into host cells (e.g., by transformation or transfection) for expression of the encoded polypeptide.
  • There are a variety of conservative changes that can generally be made to an amino acid sequence without altering activity. These changes are termed conservative substitutions or mutations; that is, an amino acid belonging to a grouping of amino acids having a particular size or characteristic can be substituted for another amino acid. Substitutes for an amino acid sequence may be selected from other members of the class to which the amino acid belongs. For example, the nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and tyrosine. The polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine and glutamine. The positively charged (basic) amino acids include arginine, lysine and histidine. The negatively charged (acidic) amino acids include aspartic acid and glutamic acid. Such alterations are not expected to substantially affect apparent molecular weight as determined by polyacrylamide gel electrophoresis or isoelectric point. Exemplary conservative substitutions include, but are not limited to, Lys for Arg and vice versa to maintain a positive charge; Glu for Asp and vice versa to maintain a negative charge; Ser for Thr so that a free —OH is maintained; and Gln for Asn to maintain a free NH2. Moreover, point mutations, deletions, and insertions of the polypeptide sequences or corresponding nucleic acid sequences may in some cases be made without a loss of function of the polypeptide or nucleic acid fragment. Substitutions may include, e.g., 1, 2, 3, or more residues. The amino acid residues described herein employ either the single letter amino acid designator or the three-letter abbreviation. Abbreviations used herein are in keeping with the standard polypeptide nomenclature, J. Biol. Chem., (1969), 243, 3552-3559. All amino acid residue sequences are represented herein by formulae with left and right orientation in the conventional direction of amino-terminus to carboxy-terminus.
  • In some cases a determination of the percent identity of a peptide to a sequence set forth herein may be required. In such cases, the percent identity is measured in terms of the number of residues of the peptide, or a portion of the peptide. A polypeptide of, e.g., 90% identity, may also be a portion of a larger peptide Variations of the disclosed polypeptide sequences include polypeptides or functional polypeptides having about 83% identity (e.g., 1 of 6 substituted) or about 67% identity (e.g., 2 of 6 substituted).
  • The term purified as used herein with reference to a polypeptide refers to a polypeptide that either has no naturally occurring counterpart (e.g., a peptidomimetic), or has been chemically synthesized and is thus substantially uncontaminated by other polypeptides, or has been separated or purified from other most cellular components by which it is naturally accompanied (e.g., other cellular proteins, polynucleotides, or cellular components). An example of a purified polypeptide is one that is at least 70%, by dry weight, free from the proteins and naturally occurring organic molecules with which it naturally associates. A preparation of the a purified polypeptide therefore can be, for example, at least 80%, at least 90%, or at least 99%, by dry weight, the polypeptide. Polypeptides also can be engineered to contain a tag sequence (e.g., a polyhistidine tag, a myc tag, or a Flag® tag) that facilitates the polypeptide to be purified or marked (e.g., captured onto an affinity matrix, visualized under a microscope). Thus a purified composition that comprises a polypeptide refers to a purified polypeptide unless otherwise indicated.
  • Polypeptides may include a chemical modification; a term that, in this context, refers to a change in the naturally-occurring chemical structure of amino acids. Such modifications may be made to a side chain or a terminus, e.g., changing the amino-terminus or carboxyl terminus. In some embodiments, the modifications are useful for creating chemical groups that may conveniently be used to link the polypeptides to other materials, or to attach a therapeutic agent.
  • Nanoparticles
  • As demonstrated by the foregoing examples, the cartilage tissue-binding ligands may be used to target nanoparticles to a cartilage tissue, and such nanoparticles may include a therapeutic agent, for example, one or more of the therapeutic agents described herein. While certain polymer-therapeutics have been described elsewhere, these have mainly been designed to augment drug concentrations in tumor tissues1. Such alteration of the biodistribution of anticancer drugs through delivery systems aims at reducing the drug's toxicity and at improving therapeutic effects2,3. To be effective, a drug delivery system must escape non-specific systemic accumulation and phagocytotic clearance by the host defense immune system. Moreover, after accumulation at the target site, penetration of the often avascular tissue must be achieved and the drugs released in active forms in order to exert the therapeutic effect. For targeting articular cartilage, non-specific systemic accumulation can be avoided by direct intra-articular injection. While this is an attractive treatment approach because it minimizes systemic effects, small compounds are prone to rapid lymphatic clearance and possess a residence time of as short as 1-5 hours4.
  • In order to minimize intra-articular injections and to increase the bioavailability of drugs in articular cartilage, the nanoparticle-based therapeutic agent delivery system described herein exhibits active targeting functionality for the cartilage, specifically, cartilage extra-cellular matrix. The combination of targeting functionality and a nanoparticle-based delivery system enables better control of bioavailability and biodistribution, particularly for intra-articular drug delivery to the cartilage matrix. Surface functionalisation of nanoparticles with the selected peptides controls the biodistribution by specific accumulation of nanoparticles in the articular cartilage, specifically in the extra-cellular matrix. The cartilage matrix itself therefore serves as a reservoir of nanoparticle encapsulated therapeutic molecules, which are delivered to the site of the disease process.
  • As explained herein, articular cartilage can be targeted in vivo with nanoparticles by the use of polypeptides which have been characterized to exhibit specific homing activity to articular cartilage. While PPS nanoparticles are used herein for demonstrative purposes, other techniques for making nanoparticles may also be adapted. As exemplified with the inverse emulsion polymerisation technique for PPS nanoparticle preparation by which the emulsifier remains displayed on the surface14, the targeting polypeptides were made bioavailable by exposure at a surface of the nanoparticles. Size control was achieved in this particular technique by adjusting the emulsifier to monomer ratio and yielded sizes ranging from about 20 nm to about 200 nm14. While the density of the cartilage extra-cellular matrix represents a relevant obstacle not just for the screening of combinatorial peptide libraries but potentially also for drug delivery to the cartilage, the use of suitably-sized nanoparticles and/or ligands with specific binding to the cartilage enhances delivery efficiency.
  • Nanoparticles are be prepared as collections of particles having an average diameter of between about 10 nm and about 200 nm, including all ranges and values between the explicitly articulated bounds, e.g., from about 20 to about 200, and from about 20 to about 40, to about 70, or to about 100 nm, depending on the polydispersity which is yielded by the preparative method. Detailed methods for making and delivering nanoparticles are set forth below and in U.S. Pat. Ser. No. 60/775,132, filed Feb. 21, 2006, which is hereby incorporated by reference herein. Numerous nanoparticle systems can be utilized, such as those formed from copolymers of poly(ethylene glycol) and poly(lactic acid), those formed from copolymers of poly(ethylene oxide) and poly(beta-amino ester), and those formed from proteins such as serum albumin. Other nanoparticle systems are known to those skilled in these arts. See also Devalapally et al., Cancer Chemother Pharmacol., 07-25-06; Langer et al., International Journal of Pharmaceutics, 257:169-180 (2003); and Tobio et al., Pharmaceutical Research, 15(2):270-275 (1998).
  • Larger particles of more than about 200 nm average diameter incorporating the cartilage tissue-binding ligands may also be prepared, with these particles being termed microparticles herein since they begin to approach the micron scale and fall approximately within the limit of optical resolution. For instance, certain techniques for making microparticles are set forth in U.S. Pat. Nos. 5,227,165, 6,022,564, 6,090,925, and 6,224,794.
  • Functionalization of nanoparticles to employ targeting capability requires association of the targeting polypeptide with the particle, e.g., by covalent binding using a bioconjugation technique, with choice of a particular technique being guided by the particle or nanoparticle, or other construct, that the polypeptide is to be joined to. In general, many bioconjugation techniques for attaching peptides to other materials are well known and the most suitable technique may be chosen for a particular material. For instance, additional amino acids may be attached to the polypeptide sequences, such as a cysteine in the case of attaching the polypeptide to thiol-reactive molecules. Herein is described an example of conjugation of the polypeptide WYRGRL (SEQ ID NO:1) with a cysteine at the C-terminus to Pluronic-F127 which was derivatized with thiol-reactive vinyl sulfone. The polypeptide was covalently bound to Pluronic by Michael-type addition and can subsequently be used for nanoparticle synthesis.
  • Proteins Targeted to Articular Cartilage Tissue
  • Therapeutic agents such as therapeutic polypeptides can be furnished with targeting capability by the use of the polypeptide ligands described and advantageously exhibit longer retention times in a joint, for instance by making a fusion protein of a polypeptide ligand and a therapeutic protein. By designing gene specific primers for the therapeutic polypeptide to be expressed, the polypeptide ligands can be attached to the N- or C-terminus in normal or reverse order. One of the primers, forward or reverse depending whether the polypeptide ligand is supposed to be localized at the N- or C-terminus, contains a gene sequence of the appropriate therapeutic polypeptide. For example, to make a fusion protein of a given therapeutic polypeptide and (GGG)WYRGRL (SEQ ID NO:8) ligand at the C-terminus, the reverse primer corresponds to 5′-ctgatgcggccgctcTCACAGCCTGCCCCTATACCAGCCGCCGCCxxxxx-3′ (SEQ ID NO:9) which contains the codon sequence for WYRGRL in reverse complement with a N-terminal glycine (GGG) linker (capital letters), as well as a stop codon and a restriction site for NotI and an overhang. The Xs correspond to the therapeutic polypeptide specific sequence and may be, e.g., around 20 base pairs long, although other lengths may be used as per conventional practice in these arts. The same example with same restriction site for a N-terminal localization of WYRGRL(GGG) (SEQ ID NO:10) corresponds to 5′-atcaggageggccgcTGGTATAGGGGCAGGCTGGGCGGCGGCxxxxx-3′ (SEQ ID NO:11) or conservative substitutions of the codon sequence. Instead of the three glycines as a linker, other linkers appropriate to the properties of the therapeutic polypeptides can be chosen. Similarly, other target ligands may be encoded, e.g., using thea nucleic acid sequence 5′-ctgatgeggccgctcAAGATGGAAATGAGGATCGCCGCCGCCxxxxx-3′ (SEQ ID NO: 12) that endocdes DPHFHLGGG (SEQ ID NO:13) or the nucleic acid 5′-ctgatgcggccgctcACGAACAAGCATAACACGGCCGCCGCCxxxxx-3′(SEQ ID NO:14) that encodes RVMLVRGGG (SEQ ID NO:15). Thus a DNA sequence for WYRGRL (SEQ ID NO:1) is TGGTATAGGGGCAGGCTG (SEQ ID NO: 16), and for DPHFHL (SEQ ID NO:2) is AAGATGGAAATGAGGATC (SEQ ID NO:17), and for RVMLVR (SEQ ID NO:3) is ACGAACAAGCATAACACG (SEQ ID NO:18).
  • Certain therapeutic polypeptides include proteins present in cartilage, e.g., tissue inhibitors of matrix metalloproteinase-3 (TIMP-3), growth factors (e.g., Transforming growth factor-beta (TGF-β), growth developmental factor-5 (GDF-5), CYR61 (Cystein-rich61)/CTGF (connective tissue growth factor)/NOV (Nephroblastoma overexpressed) (CCN2), insulin-like growth factor-1 (IGF-1), and bone morphogenic proteins (BMPs). Certain embodiments are molecules for viscosupplementation, e.g., molecules found in human cartilage that include chondroitin sulfate, keratane sulfate, hyaluronic acid, proteoglycans.
  • In some embodiments, a fusion protein is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient. In certain embodiments, the fusion protein is produced using a cell, either a procaryotic or a eucaryotic cell. In other embodiments, nucleic acids encoding a fusion protein are introduced into a patient, in which case the nucleic acids may be “naked” or part of a larger construct, e.g., a vector. In other embodiments, transfected cells are introduced into a patient. The site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • Cartilage Binding Fusion Protein: Targeted Recombinant TIMP-3
  • Tissue inhibitor of matrix metalloproteinase 3 (TIMP-3) is a relatively insoluble matricellular protein30 which inhibits several matrix metalloproteinases (MMP-1, -2, -9)31 in addition to aggrecanase 1 and 2, i.e. ADAMTS4 and ADAMTS532. As such, this molecule is beneficial in the treatment of osteoarthritis in any joint, specifically in a setting where the initiating mechanical cause is surgically corrected. Because TIMP-3 blocks these matrix degradative enzymes, the equilibrium in matrix turn-over of articular cartilage may be restored by preventing further degradation. TIMP-3 is not specifically expressed in articular cartilage, however, and due to its ability of inhibiting several enzymes may have severe potential adverse effects upon systemic administration or systemic dissemination following intra-articular injection. Targeted delivery of the inhibitory domain of TIMP-3 to articular cartilage can prevent potential systemic dissemination while increasing the therapeutic effect of the molecule in the cartilage matrix, i.e. at the site of the disease process in osteoarthritis. Moreover, the inhibitory domain of TIMP-3 also serves as a protectant against cartilage degradation in conditions such as rheumatoid arthritis, bacterial arthritis or reactive arthritis. Disclosed herein is a targeted recombinant TIMP-3 (trTIMP-3) (fusion protein which contains one of the sequences (SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3) or conservative substitutions thereof, e.g, at the N- or C-terminus. While TIMP-3 has been selected as an embodiment, other protease inhibitors or matrix metalloproteinase inhibitors may also be made and used by following the general procedures disclosed herein.
  • TIMP-3 contains a signal domain, residues 1 to 24, the N-terminal inhibitory domain, residues 24 to 143, and an extra-cellular matrix binding domain, residues 143 to 211 (SwissProt entry P35625). It has been demonstrated that N-TIMP-3, i.e. residues 24 to 143, is sufficient for exhibiting inhibitory activities on ADAMTS4 and ADAMTS5 as well as MMP-1 and MMP-23. Accordingly, it is possible to engineer a fusion protein of N-TIMP-3 (Cys24 to Asn 143) which contains the sequence WYRGRL (SEQ ID NO:1) at the C-terminus. The natural non-specific C-terminal extra-cellular matrix binding domain is substituted with a targeting polypeptide specific for articular cartilage.
  • Human (P35625) and mouse N-TIMP-3 (P39876) share 97% sequence homology differing in three amino acid residues. Two of these residues represent conservative substitutions, Thr vs. Ser at residue 74 and Asp vs. Glu at residue 110. At residue 126 His substituted for Gln, which lies in a domain linking two α-chains.
  • For cloning, the sequence in NM000362 was used for primer design. The primers were designed as explained above (see SEQ ID NO:9 for reversed primer and SEQ ID NO:11 for the forward primer). RNA was isolated using a RNeasy MinElute spin column (Qiagen, Hombrechikon, Switzerland) from immature murine articular chondrocytes which have been isolated from epiphyseal cartilage of neonatal mice using a two step digestion protocol with collagenase D (Roche, Basel, Switzerland). First-strand cDNA was generated using SuperScript III (Invitrogen, Carlsbad, Calif.) with an Oligo-dT(20) primer (Microsynth, Balgach, Switzerland). RT-PCR for amplifying the required DNA fragments was carried out with a proof-reading DNA polymerase (Pfu Turbo polymerase, Stratagene, LaJolla, Calif.) and the fragments with a required length of 420 bp checked by agarose-gel electrophoresis. The DNA fragments were gel purified with a Nucleospin II column (Macherey-Nagel, Düren, Germany), cut with the corresponding restriction enzymes BamHI and NotI (New England BioLabs Inc., Ipswich, Mass.) and ligated into the bacterial protein expression vector pGEX-4T-1 (Amersham Biosciences, GE Healthcare Europe, Otelfingen, Switzerland), which expresses trTIMP-3 as a fusion protein with glutathione-S-transferase (GST) for purification, total MW 40 kDa. For bacterial expression, E. coli strain BL21 was transformed with the trTIMP-3-pGEX construct by electroporation. An optimal expression clone was selected by anti-GST ELISA.
  • Bacterial cultures were grown in 2×YT until they reached an A600 of 0.6 to 0.7. Protein expression was then induced by 0.5M IPTG for 3 hours. To collect the protein from inclusion bodies, the bacteria were centrifuged, lysed by sonication and centrifuged again. The pellet was resuspended in a denaturing buffer (0.1M Tris-HCl, 50 mM glycine, 8 mM β-mercaptoethanol, 8M urea, 0.2M PMSF, pH 8.0) and stirred overnight, centrifuged for 20 min. at 48000×g and the supernatant collected for refolding. Refolding was performed by dialysing (24 kDa MWCO) the supernatant against a large volume of refolding buffer at 4° C. with decreasing amounts of urea to slowly remove the denaturing agent (0.1M Tris-HCl, 1 mM EDTA, 0.2 mM PMSF, pH 8.0 supplemented with 4M, 2M, 1M and 0M urea each for 24 hrs). The last step of dialysis was carried out against PBS for 24 hours. The refolded protein was freed from precipitations by centrifugation and subsequently purified by FPLC with a GST binding column (GSTrap FF, Amersham Biosciences, GE Healthcare Europe, Otelfingen, Switzerland). The GST tag of purified trTIMP-3-GST fusion protein was cleaved of by incubation with thrombin for 24 hours and purified by FPLC from GST with the GSTrap FF column and from thrombin with a HiTrap Benzamidine FF column (Amersham Biosciences, GE Healthcare Europe, Otelfingen, Switzerland). Overall yield of trTIMP-3 was about 1.5 mg/l of bacterial culture.
  • The activity of purified trTIMP-3 was assessed by MMP-2 zymography in which an equimolar amount of trTIMP-3 completely inhibits MMP-2 activity indicating a nearly 100% activity of the purified protein. Further experiments could be used to demonstrate the aggrecanase inhibiting activity of trTIMP-3 and the targeting specificity to articular cartilage in a similar fashion as shown for the nanoparticles described herein. In addition, its therapeutic potential to prevent cartilage degradation could be demonstrated in a suitable animal model, e.g., in a mouse knee instability model.
  • Vectors
  • Accordingly, certain embodiments are directed to vectors for expression of a therapeutic protein of interest, e.g., a therapeutic agent and a polypeptide ligand. Nucleic acids encoding a polypeptide can be incorporated into vectors. As used herein, a vector is a replicon, such as a plasmid, phage, or cosmid, into which another nucleic acid segment may be inserted so as to bring about replication of the inserted segment. Vectors of the invention typically are expression vectors containing an inserted nucleic acid segment that is operably linked to expression control sequences. An expression vector is a vector that includes one or more expression control sequences, and an expression control sequence is a DNA sequence that controls and regulates the transcription and/or translation of another DNA sequence. Expression control sequences include, for example, promoter sequences, transcriptional enhancer elements, and any other nucleic acid elements required for RNA polymerase binding, initiation, or termination of transcription. With respect to expression control sequences, “operably linked” means that the expression control sequence and the inserted nucleic acid sequence of interest are positioned such that the inserted sequence is transcribed (e.g., when the vector is introduced into a host cell). For example, a DNA sequence is operably linked to an expression-control sequence, such as a promoter when the expression control sequence controls and regulates the transcription and translation of that DNA sequence. The term “operably linked” includes having an appropriate start signal (e.g., ATG) in front of the DNA sequence to be expressed and maintaining the correct reading frame to permit expression of the DNA sequence under the control of the expression control sequence to yield production of the desired protein product. Examples of vectors include: plasmids, adenovirus, Adeno-Associated Virus (AAV), Lentivirus (FIV), Retrovirus (MoMLV), and transposons. There are a variety of promoters that could be used including, but not limited to, constitutive promoters, tissue-specific promoters, inducible promoters, and the like. Promoters are regulatory signals that bind RNA polymerase in a cell to initiate transcription of a downstream (3′ direction) coding sequence.
  • Targeted Deliver of Viscosupplementation
  • Molecules for viscosupplementation may be conjugated with polypeptide ligands described herein to form a conjugate for enhanced delivery and effect in the cartilage. The formation of such conjugates is within the skill of ordinary artisans and various techniques are known for accomplishing the conjugation, with the choice of the particular technique being guided by the materials to be conjugated. Such conjugates may be delivered systemically or locally, e.g., orally or by injection to a joint. Thus hyaluronic acid conjugation with a polypeptide ligand disclosed herein may prolong the retention time of hyaluronic acid in the joint and therefore enhance the efficacy of intra-articular viscosupplementation with hyaluronic acid. Conjugation of the polypeptide to hyaluronic acid can be performed either directly as described above, or by the use of a polymer linker. Examples of polymer linkers are biocompatible hydrophilic polymers, including polymers free of amino-acids. For instance, a polymer linker may be a polyethylene glycol (PEG). Hyaluronic acid can be functionalized with acrylated PEG-Arg-Gly-Asp conjugates created by Michael-type addition chemistry (Park et al. Biomaterials 2003; 24:893-900). In general, polymers described herein for use with the polypeptide ligands may be free of amino acids, meaning that such polymers do not contain a natural or synthetic amino acid.
  • In some embodiments, the conjugate is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient. In certain embodiments, the conjugate is produced using a cell, either a procaryotic or a eucaryotic cell, as in the case of a biopolymer. In other embodiments, transfected cells are introduced into a patient. The site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • Targeting Genes Condensed with Polymers with the Polypeptides Attached
  • A small gene delivery system will have advantages with respect to penetrating the dense matrix of cartilage tissue. In some embodiments, therefore, the delivery system uses significantly condensed DNA, to enter the cartilage matrix and transfect non-dividing quiescent chondrocytes or other cells. In some embodiments, therefore, polypeptide ligands are attached to polymers which contain a nuclear localisation sequence to form a conjugate and are used to condense DNA to overcome challenges to gene transfection in chondrocytes embedded in the cartilage matrix. Some aspects of these techniques have been described by in Trentin et al. PNAS 2006; 103:2506-11 and J Control Release 2005; 102:263-75. Thus in certain embodiments the conjugate associated with nucleic acids encoding a therapeutic polypeptide is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient. In certain embodiments, such a conjugate is produced using a cell, either a procaryotic or a eucaryotic cell, as in the case of a biopolymer. In other embodiments, transfected cells are introduced into a patient. The site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • Therapeutic Agents Associated with Ligands for Delivery to Cartilage
  • Polypeptides as described herein can be attached to other polymers through bioconjugation. The formation of such conjugates is within the skill of ordinary artisans and various techniques are known for accomplishing the conjugation, with the choice of the particular technique being guided by the materials to be conjugated. The addition of amino acids to the polypeptide (C- or N-terminal) which contain ionizable side chains, i.e. aspartic acid, glutamic acid, lysine, arginine, cysteine, histidine, or tyrosine, and are not contained in the active portion of the polypeptide sequence, serve in their unprotonated state as a potent nucleophile to engage in various bioconjugation reactions with reactive groups attached to polymers, i.e. homo- or hetero-bi-functional PEG (e.g., Lutolf and Hubbell, Biomacromolecules 2003; 4:713-22, Hermanson. Bioconjugate Techniques. London. Academic Press Ltd; 1996). An application where this may be useful is again for targeted delivery of a therapeutic agent. In some embodiments, the agent is attached to a soluble polymer, and may be administer to a patient in a pharmaceutically acceptable form. Or a drug may be encapsulated in polymerosomes or vesicles or covalently attached to polymers. In the latter case, drugs are attached to the polymer backbone with a degradable site-specific spacer or linker (Lu et al. J Control Release 2002; 78:165-73).
  • In general, soluble hydrophilic biocompatible polymers may be used to ensure that the conjugate is soluble and will be bioavailable after introduction into the patient. Examples of soluble polymers are polyvinyl alcohols, polyethylene imines, and polyethylene glycols (a term including polyethylene oxides) having a molecular weight of at least 100, 400, or between 100 and 400,000 (with all ranges and values between these explicit values being contemplated). Solubility refers to a solubility in water or physiological saline of at least 1 gram per liter. Domains of biodegradable polymers may also be used, e.g., polylactic acid, polyglycolic acid, copolymers of polylactic and polyglycolic acid, polycaprolactones, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, and polycyanoacylates.
  • In some embodiments, a polypeptide-polymer association, e.g., a conjugate, is prepared and introduced into the body as a purified composition in a pharmaceutically acceptable condition, or with a pharmaceutical excipient. The site of introduction may be, e.g., systemic, in a joint, or in a cartilage tissue.
  • Cartilage Defect Treatment Using Polypeptide Ligands with Specific Binding for Cartilage
  • The polypeptides ligands were discovered based on their binding affinity for the cartilage matrix but are not specifically bound to the articular surface. Therefore, besides embodiments such as targeting the cartilage matrix by therapeutic agent delivery systems or with engineered fusion proteins, the polypeptides are particularly well suited to target a defect because extracellular matrix is exposed at the defect. This feature is useful for delivering biomaterials for gene, protein and/or cell delivery to mediate cartilage defect repair. Accordingly, embodiments include treating a defect in an articular cartilage using one of the embodiments set forth herein. In fact, giving a biomaterial the capability of adhering or otherwise specifically binding to a cartilage defect does not only allow for injectable defect repair strategies but also may enhance retention of the biomaterial in the cartilage defect. A variety of chemical schemes can be used to incorporate the cartilage-binding polypeptide into the biomaterial. For example, using a material as described by Sawhney et al., a chemical approach for incorporation as described by Hern et al. can be employed (Sawhney et al. Macromolecules 1993; 26:581-587 and Hern et al. J. Biomed. Mater. Res. 1998; 39:266-276). As another example, using a material as described by Lutolf et al., a chemical approach for incorporation as described therein can be employed (Lutolf et al. Nature Biotechnol. 2003; 21:513-518). In general, the modification of such biomaterials is within the skill of ordinary artisans and various techniques are known for accomplishing the modification, with the choice of a particular technique being guided by the biomaterial and peptides to be conjugated.
  • Specific binding, as that term is commonly used in the biological arts, generally refers to a molecule that binds to a target with a relatively high affinity compared to non-target tissues, and generally involves a plurality of non-covalent interactions, such as electrostatic interactions, van der Waals interactions, hydrogen bonding, and the like. Specific binding interactions characterize antibody-antigen binding, enzyme-substrate binding, and specifically binding protein-receptor interactions; while such molecules may bind tissues besides their targets from time to time, such binding is said to lack specificity and is not specific binding. The peptides of SEQ ID NOs 1, 2, and 3 may bind non-cartilage tissues in some circumstances but such binding has been observed to be non-specific, as evidenced by the much greater binding of the peptides to the targeted tissue as opposed to surrounding joint tissues (data not shown).
  • Accordingly, embodiments include biomaterials comprising at least one of the ligands disclosed herein that are used to fill or augment a defect in a cartilage. A defect refers to a void in a surface (e.g., a pit, tear, or hole) or a pathological discontinuity in a surface (e.g., a tear or eroded member). Fill refers to essentially filling or covering the defect or bridging the discontinuity. Augment refers to at least partial filling. In some embodiments, the biomaterial is a solid prior to placement in a patient, while in other embodiments the material is made is situ, meaning it is formed from precursors at the site of the defect. Thus biomaterials for cartilage defects may be supplemented with a ligand or other embodiment set forth herein. Examples of such biomaterials include U.S. Pat. Nos. 5,874,500, and 5,410,016, and which include materials formed by in-situ polymerization. Biomaterials for targeting cartilage defects, i.e. adhering to a cartilage defect, are suited for the delivery of therapeutic agents to mediate cartilage repair such as growth factors and for cell delivery to the repair site. In accordance with techniques for autologous cartilage transplantation/implantation the use of a biomaterial for cell delivery which adheres to the defect by the use of polypeptide ligands (SEQ ID NO:1 through 3) and acts as a morphogenic guide may improve cartilage defect repair.
  • Nucleic Acids
  • Certain embodiments are directed to nucleic acids. As used herein, the term nucleic acid refers to both RNA and DNA, including siRNA, shRNA, miRNA, cDNA, genomic DNA, synthetic (e.g., chemically synthesized) DNA, as well as naturally occurring and chemically modified nucleic acids, e.g., synthetic bases or alternative backbones. A nucleic acid molecule can be double-stranded or single-stranded (i.e., a sense or an antisense single strand). An isolated nucleic acid refers to a nucleic acid that is separated from other nucleic acid bases that are present in a genome, including nucleic acids that normally flank one or both sides of a nucleic acid sequence in a vertebrate genome (e.g., nucleic acids that flank a gene). A conservatively substituted nucleic acid refers to the substitution of a nucleic acid codon with another codon that encodes the same amino acid and also refers to nucleic acids that encode conservatively substituted amino acids, as described herein with respect to polypeptides. Significantly, the combination of potential codons for a polypeptide of only about six residues is manageably small.
  • The nucleic acid sequences set forth herein are intended to represent both DNA and RNA sequences, according to the conventional practice of allowing the abbreviation “T” stand for “T” or for “U”, as the case may be, for DNA or RNA. Polynucleotides are nucleic acid molecules of at least three nucleotide subunits. Polynucleotide analogues or polynucleic acids are chemically modified polynucleotides or polynucleic acids. In some embodiments, polynucleotide analogues can be generated by replacing portions of the sugar-phosphate backbone of a polynucleotide with alternative functional groups. Morpholino-modified polynucleotides, referred to herein as “morpholinos,” are polynucleotide analogues in which the bases are linked by a morpholino-phosphorodiamidate backbone (see, e.g., U.S. Pat. Nos. 5,142,047 and 5,185,444). In addition to morpholinos, other examples of polynucleotide analogues include analogues in which the bases are linked by a polyvinyl backbone, peptide nucleic acids (PNAs) in which the bases are linked by amide bonds formed by pseudopeptide 2-aminoethyl-glycine groups, analogues in which the nucleoside subunits are linked by methylphosphonate groups, analogues in which the phosphate residues linking nucleoside subunits are replaced by phosphoroamidate groups, and phosphorothioated DNAs, analogues containing sugar moieties that have 2′ O-methyl group). Polynucleotides of the invention can be produced through the well-known and routinely used technique of solid phase synthesis. Alternatively, other suitable methods for such synthesis can be used (e.g., common molecular cloning and chemical nucleic acid synthesis techniques). Similar techniques also can be used to prepare polynucleotide analogues such as morpholinos or phosphorothioate derivatives. In addition, polynucleotides and polynucleotide analogues can be obtained commercially. For oligonucleotides, examples of pharmaceutically acceptable compositions are salts that include, e.g., (a) salts formed with cations such as sodium, potassium, ammonium, etc.; (b) acid addition salts formed with inorganic acids, for example, hydrochloric acid, hydrobromic acid (c) salts formed with organic acids e.g., for example, acetic acid, oxalic acid, tartaric acid; and (d) salts formed from elemental anions e.g., chlorine, bromine, and iodine.
  • Pharmaceutical Carriers
  • Pharmaceutically acceptable carriers or excipient may be used to deliver embodiments as described herein. Excipient refers to an inert substance used as a diluent or vehicle for a therapeutic agent. Pharmaceutically acceptable carriers are used, in general, with a compound so as to make the compound useful for a therapy or as a product. In general, for any substance, a pharmaceutically acceptable carrier is a material that is combined with the substance for delivery to an animal. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. In some cases the carrier is essential for delivery, e.g., to solubilize an insoluble compound for liquid delivery; a buffer for control of the pH of the substance to preserve its activity; or a diluent to prevent loss of the substance in the storage vessel. In other cases, however, the carrier is for convenience, e.g., a liquid for more convenient administration. Pharmaceutically acceptable salts of the compounds described herein may be synthesized according to methods known to those skilled in this arts. Thus a pharmaceutically acceptable composition has a carrier, salt, or excipient suited to administration to a patient. Moreover, inert components of such compositions are biocompatible and not toxic.
  • The compounds described herein are typically to be administered in admixture with suitable pharmaceutical diluents, excipients, extenders, or carriers (termed herein as a pharmaceutically acceptable carrier, or a carrier) suitably selected with respect to the intended form of administration and as consistent with conventional pharmaceutical practices. Thus the deliverable compound may be made in a form suitable for oral, rectal, topical, intravenous injection, intra-articular injection, or parenteral administration. Carriers include solids or liquids, and the type of carrier is chosen based on the type of administration being used. Suitable binders, lubricants, disintegrating agents, coloring agents, flavoring agents, flow-inducing agents, and melting agents may be included as carriers, e.g., for pills. For instance, an active component can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, gelatin, agar, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like. The compounds can be administered orally in solid dosage forms, such as capsules, tablets, and powders, or in liquid dosage forms, such as elixirs, syrups, and suspensions. The active compounds can also be administered parentally, in sterile liquid dosage forms. Buffers for achieving a physiological pH or osmolarity may also be used.
  • All patent applications, patents, and publications mentioned herein are hereby incorporated by reference herein to the extent they do not directly contradict the explicit disclosures set forth herein.
  • REFERENCES
    • 1. Duncan R. The dawning era of polymer therapeutics. Nat Rev Drug Discov 2003; 2-5:347-60.
    • 2. Bae Y, Nishiyama N, Fukushima S, Koyama H, Yasuhiro M, Kataoka K. Preparation and biological characterization of polymeric micelle drug carriers with intracellular pH-triggered drug release property: tumor permeability, controlled subcellular drug distribution, and enhanced in vivo antitumor efficacy. Bioconjug Chem 2005; 16-1:122-30.
    • 3. Nishiyama N, Okazaki S, Cabral H, Miyamoto M, Kato Y, Sugiyama Y, Nishio K, Matsumura Y, Kataoka K. Novel cisplatin-incorporated polymeric micelles can eradicate solid tumors in mice. Cancer Res 2003; 63-24:8977-83.
    • 4. Owen S G, Francis H W, Roberts M S. Disappearance kinetics of solutes from synovial fluid after intra-articular injection. Br J Clin Pharmacol 1994; 38-4:349-55.
    • 5. Smith G P, Petrenko V A. Phage Display. Chem. Rev 1997; 97-2:391-410.
    • 6. Kay B K, Kasanov J, Yamabhai M. Screening phage-displayed combinatorial peptide libraries. Methods 2001; 24-3:240-6.
    • 7. Arap W, Kolonin M G, Trepel M, Landenranta J, Cardo-Vila M, Giordano R J, Mintz P J, Ardelt P U, Yao V J, Vidal C I, Chen L, Flamm A, Valtanen H, Weavind L M, Hicks M E, Pollock R E, Botz G H, Bucana C D, Koivunen E, Cahill D, Troncoso P, Baggerly K A, Pentz R D, Do K A, Logothetis C J, Pasqualini R. Steps toward mapping the human vasculature by phage display. Nat Med 2002; 8-2:121-7.
    • 8. Kolonin M G, Sun J, Do K A, Vidal C I, Ji Y, Baggerly K A, Pasqualini R, Arap W. Synchronous selection of homing peptides for multiple tissues by in vivo phage display. Faseb J 2006; 20-7:979-81.
    • 9. Pasqualini R, Ruoslahti E. Organ targeting in vivo using phage display peptide libraries. Nature 1996; 380-6572:364-6.
    • 10. Kolonin M G, Saha P K, Chan L, Pasqualini R, Arap W. Reversal of obesity by targeted ablation of adipose tissue. Nat Med 2004; 10-6:625-32.
    • 11. Lee L, Buckley C, Blades M C, Panayi G, George A J, Pitzalis C. Identification of synovium-specific homing peptides by in vivo phage display selection. Arthritis Rheum 2002; 46-8:2109-20.
    • 12. Comper W D. Physicochemical aspects of cartilage extracellular matrix. In: Hall B, Newman S, eds. Cartilage: Molecular Aspects. Boston: CRC Press, 1991:59-96.
    • 13. Torzilli P A, Arduino J M, Gregory J D, Bansal M. Effect of proteoglycan removal on solute mobility in articular cartilage. J Biomech 1997; 30-9:895-902.
    • 14. Rehor A, Hubbell J A, Tirelli N. Oxidation-sensitive polymeric nanoparticles. Langmuir 2005; 21-1:411-7.
    • 15. Boder E T, Wittrup K D. Yeast surface display for screening combinatorial polypeptide libraries. Nat Biotechnol 1997; 15-6:553-7.
    • 16. Xu L, Aha P, Gu K, Kuimelis R G, Kurz M, Lam T, Lim A C, Liu H, Lohse P A, Sun L, Weng S, Wagner R W, Lipovsek D. Directed evolution of high-affinity antibody mimics using mRNA display. Chem Biol 2002; 9-8:933-42. Lam K S, Lebl M, Krchnak V. The “One-Bead-One-Compound” Combinatorial Library Method. Chem. Rev 1997; 97-2:411-48.
    • 18. Zacher A N, 3rd, Stock C A, Golden J W, 2nd, Smith G P. A new filamentous phage cloning vector: fd-tet. Gene 1980; 9-1-2:127-40.
    • 19. Madry H, Cucchiarini M, Terwilliger E F, Trippel S B. Recombinant adeno-associated virus vectors efficiently and persistently transduce chondrocytes in normal and osteoarthritic human articular cartilage. Hum Gene Ther 2003; 14-4:393-402.
    • 20. Phillips N C, Thomas D P, Knight C G, Dingle J T. Liposome-incorporated corticosteroids. II. Therapeutic activity in experimental arthritis. Ann Rheum Dis 1979; 38-6:553-7.
    • 21. Shaw I H, Knight C G, Thomas D P, Phillips N C, Dingle J T. Liposome-incorporated corticosteroids: I. The interaction of liposomal cortisol palmitate with inflammatory synovial membrane. Br J Exp Pathol 1979; 60-2:142-50.
    • 22. Ratcliffe J H, Hunneyball 1M, Smith A, Wilson C G, Davis S S. Preparation and evaluation of biodegradable polymeric systems for the intra-articular delivery of drugs. J Pharm Pharmacol 1984; 36-7:431-6.
    • 23. Ratcliffe J H, Hunneyball 1M, Wilson C G, Smith A, Davis S S. Albumin microspheres for intra-articular drug delivery: investigation of their retention in normal and arthritic knee joints of rabbits. J Pharm Pharmacol 1987; 39-4:290-5.
    • 24. Tuncay M, Calis S, Kas H S, Ercan M T, Peksoy I, Hincal A A. In vitro and in vivo evaluation of diclofenac sodium loaded albumin microspheres. J Microencapsul 2000; 17-2:145-55.
    • 25. Horisawa E, Hirota T, Kawazoe S, Yamada J, Yamamoto H, Takeuchi H, Kawashima Y. Prolonged anti-inflammatory action of DL-lactide/glycolide copolymer nanospheres containing betamethasone sodium phosphate for an intra-articular delivery system in antigen-induced arthritic rabbit. Pharm Res 2002; 19-4:403-10.
    • 26. Horisawa E, Kubota K, Tuboi I, Sato K, Yamamoto H, Takeuchi H, Kawashima Y. Size-dependency of DL-lactide/glycolide copolymer particulates for intra-articular delivery system on phagocytosis in rat synovium. Pharm Res 2002; 19-2:132-9.
    • 27. Quinn T M, Morel V, Meister J J. Static compression of articular cartilage can reduce solute diffusivity and partitioning: implications for the chondrocyte biological response. J Biomech 2001; 34-11:1463-9.
    • 28. Quinn T M, Kocian P, Meister J J. Static compression is associated with decreased diffusivity of dextrans in cartilage explants. Arch Biochem Biophys 2000; 384-2:327-34.
    • 29. Lutolf M P, Hubbell J A. Synthesis and physicochemical characterization of end-linked poly(ethylene glycol)-co-peptide hydrogels formed by Michael-type addition. Biomacromolecules 2003; 4-3:713-22.
    • 30. Staskus P W, Masiarz F R, Pallanck L J, Hawkes S P. The 21-kDa protein is a transformation-sensitive metalloproteinase inhibitor of chicken fibroblasts. J Biol Chem 1991; 266(1):449-54.
    • 31. Negro A, Onisto M, Grassato L, Caenazzo C, Garbisa S. Recombinant human TIMP-3 from Escherichia coli: synthesis, refolding, physico-chemical and functional insights. Protein Eng 1997; 10(5):593-9.
    • 32. Kashiwagi M, Tortorella M, Nagase H, Brew K. TIMP-3 is a potent inhibitor of aggrecanase 1 (ADAM-TS4) and aggrecanase 2 (ADAM-TS5). J Biol Chem 2001; 276(16):12501-4.
  • This application discloses various inventive embodiments that each have certain features. In general, these features may be mixed-and-matched with each other to create additional functional embodiments.

Claims (18)

1. A substantially pure polypeptide not found in nature comprising an amino acid sequence of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, or a conservative substitution thereof, wherein the polypeptide specifically binds a cartilage tissue.
2. The polypeptide of claim 1 wherein the sequence length is between 6 and 1000 residues.
3. The polypeptide of claim 1 comprising a therapeutic agent polypeptide.
4. The polypeptide of claim 3 wherein the therapeutic agent polypeptide is tissue inhibitor of matrix metalloproteinases-3 (TIMP-3).
5. The polypeptide of claim 1 comprising a synthetic backbone linkage.
6. A substantially pure nucleic acid not found in nature that comprises a nucleic acid sequence that encodes SEQ ID NO:1, SEQ ID NO:2 or SEQ ID NO:3, or a conservative substitution thereof, wherein a polypeptide encoded by the nucleic acid specifically binds to a cartilage tissue.
7. The nucleic acid of claim 6 wherein the polypeptide that is encoded comprises a therapeutic agent polypeptide.
8. A vector comprising the nucleic acid of claim 6.
9. A method of treating cartilage of a mammal comprising administering to the mammal a pharmaceutically acceptable composition that comprises the polypeptide of claim 1, wherein the polypeptide targets the therapeutic agent to the cartilage tissue by specifically binding cartilage tissue of the mammal.
10. A delivery system for delivering a therapeutic agent comprising: a substantially purified preparation that comprises a pharmaceutically acceptable excipient, a therapeutic agent, and a polypeptide ligand not found in nature comprising an amino acid sequence in the group consisting of SEQ ID NO:1, SEQ ID NO:2, SEQ ID NO:3, and conservative substitutions thereof, wherein the polypeptide ligand specifically binds a cartilage tissue for targeted delivery of the therapeutic agent to cartilage tissues.
11. The delivery system of claim 10 wherein the therapeutic agent comprises a drug, a visualization agent, or a therapeutic polypeptide.
12. The delivery system of claim 10 comprising a fusion polypeptide that comprises the polypeptide ligand and the therapeutic agent.
13. The delivery system of claim 10 comprising a molecule that comprises covalent bonds to the polypeptide ligand and the therapeutic agent.
14. The delivery system of claim 10 wherein the polypeptide ligand is covalently bonded to a biocompatible polymer that is associated with the therapeutic agent.
15. The delivery system of claim 14 wherein the biocompatible polymer is free of amino acids.
16. The delivery system of claim 10 comprising a collection of nanoparticles having an average diameter of between about 10 nm and about 200 nm, wherein the nanoparticles comprise the therapeutic agent and the polypeptide ligand.
17. A method of treating cartilage of a mammal comprising administering to the mammal a pharmaceutically acceptable composition that comprises the delivery system of claim 13, wherein the polypeptide targets the therapeutic agent to the cartilage tissue by specifically binding cartilage tissue of the mammal.
18. The method of claim 17 wherein the composition is administered intra-articularly.
US12/563,201 2006-10-10 2009-09-21 Polypeptide ligands for targeting cartilage and methods of use thereof Abandoned US20100080850A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/563,201 US20100080850A1 (en) 2006-10-10 2009-09-21 Polypeptide ligands for targeting cartilage and methods of use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11/545,819 US7592009B2 (en) 2006-10-10 2006-10-10 Polypeptide ligands for targeting cartilage and methods of use thereof
US12/563,201 US20100080850A1 (en) 2006-10-10 2009-09-21 Polypeptide ligands for targeting cartilage and methods of use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/545,819 Continuation US7592009B2 (en) 2006-10-10 2006-10-10 Polypeptide ligands for targeting cartilage and methods of use thereof

Publications (1)

Publication Number Publication Date
US20100080850A1 true US20100080850A1 (en) 2010-04-01

Family

ID=39417214

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/545,819 Active 2027-03-01 US7592009B2 (en) 2006-10-10 2006-10-10 Polypeptide ligands for targeting cartilage and methods of use thereof
US12/563,201 Abandoned US20100080850A1 (en) 2006-10-10 2009-09-21 Polypeptide ligands for targeting cartilage and methods of use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/545,819 Active 2027-03-01 US7592009B2 (en) 2006-10-10 2006-10-10 Polypeptide ligands for targeting cartilage and methods of use thereof

Country Status (7)

Country Link
US (2) US7592009B2 (en)
EP (1) EP2084275A4 (en)
JP (1) JP2010505444A (en)
AU (1) AU2007322346A1 (en)
CA (1) CA2665839A1 (en)
IL (1) IL198120A0 (en)
WO (1) WO2008063291A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016205004A3 (en) * 2015-06-15 2017-04-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
US10245285B2 (en) 2016-04-28 2019-04-02 Alkahest, Inc. Blood plasma and plasma fractions as therapy for tumor growth and progression
US10357513B2 (en) 2017-04-26 2019-07-23 Alkahest, Inc. Dosing regimen for treatment of cognitive and motor impairments with blood plasma and blood plasma products
US10487148B2 (en) 2010-01-28 2019-11-26 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated impairments
US10525107B2 (en) 2016-08-18 2020-01-07 Alkahest, Inc. Blood plasma fractions as a treatment for aging-associated cognitive disorders
US10626399B2 (en) 2010-01-28 2020-04-21 The Board Of Trustees Of The Leland Stanford Junior University Methods of treating cognitive symptoms of an aging-associated impairment by modulating C-C chemokine receptor type 3 (CCR3)
US10688130B2 (en) 2013-12-09 2020-06-23 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
US10688154B2 (en) 2011-04-08 2020-06-23 The Board Of Trustees Of The Leland Stanford Junior University Methods of neuroprotection involving macrophage colony stimulating factor receptor agonists
US10905779B2 (en) 2013-12-09 2021-02-02 The Board Of Trustees Of The Leland Stanford Junior University Methods for screening human blood products comprising plasma using immunocompromised rodent models
US11040068B2 (en) 2017-04-26 2021-06-22 Alkahest, Inc. Dosing regimen for treatment of cognitive and motor impairments with blood plasma and blood plasma products
US11103530B2 (en) 2018-10-26 2021-08-31 Alkahest, Inc. Methods of improving or accelerating postoperative recovery
US11236340B2 (en) 2010-01-28 2022-02-01 The Board Of Trustees Of The Leland Stanford Junior University Method of reducing the effects of aging-associated impairment of neurogenesis comprising modulating c-c chemokine receptor type 3 (CCR3)
US11773138B2 (en) 2018-10-12 2023-10-03 Theradaptive, Inc. Polypeptides including a beta-tricalcium phosphate-binding sequence and uses thereof

Families Citing this family (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2008056623A1 (en) * 2006-11-09 2010-02-25 第一三共株式会社 Composition for introduction of nucleic acid
US8846003B2 (en) 2008-03-27 2014-09-30 Symic Biomedical, Inc. Collagen-binding synthetic peptidoglycans, preparation, and methods of use
US8868149B2 (en) * 2009-07-30 2014-10-21 Oxitone Medical Ltd. Photoplethysmography device and method
EP2459213A1 (en) 2009-07-31 2012-06-06 Amgen Inc. Polypeptides that bind tissue inhibitor of metalloproteinase type three (timp-3), compositions and methods
US20130143821A1 (en) * 2010-02-04 2013-06-06 University Of Tennessee Research Foundation Hydrophobically-modified hyaluronan and methods of making and using thereof
US8524662B2 (en) 2010-12-28 2013-09-03 Depuy Mitek, Llc Compositions and methods for treating joints
US8398611B2 (en) 2010-12-28 2013-03-19 Depuy Mitek, Inc. Compositions and methods for treating joints
US8455436B2 (en) 2010-12-28 2013-06-04 Depuy Mitek, Llc Compositions and methods for treating joints
WO2012112767A2 (en) 2011-02-16 2012-08-23 Purdue Research Foundation Collagen-targeted nanoparticles
MX349693B (en) 2011-05-24 2017-08-09 Symic Ip Llc Hyaluronic acid-binding synthetic peptidoglycans, preparation, and methods of use.
US8623839B2 (en) 2011-06-30 2014-01-07 Depuy Mitek, Llc Compositions and methods for stabilized polysaccharide formulations
EP2908861A4 (en) * 2012-10-19 2016-04-20 Univ Cornell Biomimetic boundary lubricants for articular cartilage
WO2014107566A1 (en) 2013-01-04 2014-07-10 Massachusetts Institute Of Technology Surface binding of nanoparticle based drug delivery to tissue
SG11201506966RA (en) 2013-03-15 2015-10-29 Symic Biomedical Inc Extracellular matrix-binding synthetic peptidoglycans
WO2015009787A1 (en) 2013-07-19 2015-01-22 The Johns Hopkins University Biomaterials comprising hyaluronic acid binding peptides and extracellular matrix binding peptides for hyaluronic acid retention and tissue engineering applications
US11559580B1 (en) 2013-09-17 2023-01-24 Blaze Bioscience, Inc. Tissue-homing peptide conjugates and methods of use thereof
US11013814B2 (en) 2017-03-16 2021-05-25 Blaze Bioscience, Inc. Cartilage-homing peptide conjugates and methods of use thereof
WO2015075699A1 (en) * 2013-11-25 2015-05-28 Sanofi Dotam derivatives for therapeutic use
US10772931B2 (en) 2014-04-25 2020-09-15 Purdue Research Foundation Collagen binding synthetic peptidoglycans for treatment of endothelial dysfunction
EA201790470A1 (en) 2014-08-27 2017-07-31 Эмджен Инк. VARIANTS OF THE TISSUE INHIBITOR OF METALLOPROTEINAS TYPE THREE (TIPP-3), COMPOSITIONS AND METHODS
US9682099B2 (en) 2015-01-20 2017-06-20 DePuy Synthes Products, Inc. Compositions and methods for treating joints
CN108135970B (en) * 2015-09-09 2023-09-01 弗莱德哈钦森癌症中心 Cartilage homing peptides
JP7191025B2 (en) 2017-01-18 2022-12-16 フレッド ハッチンソン キャンサー センター Peptide compositions and methods of use thereof for interfering with TEAD interactions
EP3638290A4 (en) 2017-06-15 2021-04-07 Blaze Bioscience, Inc. Renal-homing peptide conjugates and methods of use thereof
WO2019010484A2 (en) 2017-07-07 2019-01-10 Symic Ip, Llc Synthetic bioconjugates
US11866466B2 (en) 2017-12-19 2024-01-09 Blaze Bioscience, Inc. Tumor homing and cell penetrating peptide-immuno-oncology agent complexes and methods of use thereof
CA3093588A1 (en) * 2018-03-16 2019-09-19 Blaze Bioscience, Inc. Truncated cartilage-homing peptides and peptide complexes and methods of use thereof

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5142047A (en) * 1985-03-15 1992-08-25 Anti-Gene Development Group Uncharged polynucleotide-binding polymers
US5185444A (en) * 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5227165A (en) * 1989-11-13 1993-07-13 Nova Pharmaceutical Corporation Liposphere delivery systems for local anesthetics
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5874500A (en) * 1995-12-18 1999-02-23 Cohesion Technologies, Inc. Crosslinked polymer compositions and methods for their use
US6022564A (en) * 1996-10-09 2000-02-08 Takeda Chemical Industries, Ltd. Method for producing a microparticle
US6090925A (en) * 1993-03-09 2000-07-18 Epic Therapeutics, Inc. Macromolecular microparticles and methods of production and use
US6224794B1 (en) * 1998-05-06 2001-05-01 Angiotech Pharmaceuticals, Inc. Methods for microsphere production
US20030233675A1 (en) * 2002-02-21 2003-12-18 Yongwei Cao Expression of microbial proteins in plants for production of plants with improved properties
US20040123343A1 (en) * 2000-04-19 2004-06-24 La Rosa Thomas J. Rice nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
US20040214272A1 (en) * 1999-05-06 2004-10-28 La Rosa Thomas J Nucleic acid molecules and other molecules associated with plants

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2090925A (en) * 1935-11-25 1937-08-24 Platt Brothers & Co Ltd Apparatus for opening and cleaning flax straw and such like fibrous materials
US6562596B1 (en) 1993-10-06 2003-05-13 Amgen Inc. Tissue inhibitor of metalloproteinase type three (TIMP-3) composition and methods
WO2001044497A2 (en) * 1999-12-02 2001-06-21 University Of Dundee Protein kinase regulation
CA2407352A1 (en) * 2000-04-21 2001-11-01 Corixa Corporation Compositions and methods for the therapy and diagnosis of acne vulgaris
US7630836B2 (en) * 2001-05-30 2009-12-08 The Kitasato Institute Polynucleotides
US20080213247A1 (en) * 2002-10-23 2008-09-04 Plowman Gregory D Mbms as Modifiers of Branching Morphogenesis and Methods of Use

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5142047A (en) * 1985-03-15 1992-08-25 Anti-Gene Development Group Uncharged polynucleotide-binding polymers
US5185444A (en) * 1985-03-15 1993-02-09 Anti-Gene Deveopment Group Uncharged morpolino-based polymers having phosphorous containing chiral intersubunit linkages
US5227165A (en) * 1989-11-13 1993-07-13 Nova Pharmaceutical Corporation Liposphere delivery systems for local anesthetics
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US6090925A (en) * 1993-03-09 2000-07-18 Epic Therapeutics, Inc. Macromolecular microparticles and methods of production and use
US5874500A (en) * 1995-12-18 1999-02-23 Cohesion Technologies, Inc. Crosslinked polymer compositions and methods for their use
US6022564A (en) * 1996-10-09 2000-02-08 Takeda Chemical Industries, Ltd. Method for producing a microparticle
US6224794B1 (en) * 1998-05-06 2001-05-01 Angiotech Pharmaceuticals, Inc. Methods for microsphere production
US20040214272A1 (en) * 1999-05-06 2004-10-28 La Rosa Thomas J Nucleic acid molecules and other molecules associated with plants
US20040123343A1 (en) * 2000-04-19 2004-06-24 La Rosa Thomas J. Rice nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
US20030233675A1 (en) * 2002-02-21 2003-12-18 Yongwei Cao Expression of microbial proteins in plants for production of plants with improved properties

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11236340B2 (en) 2010-01-28 2022-02-01 The Board Of Trustees Of The Leland Stanford Junior University Method of reducing the effects of aging-associated impairment of neurogenesis comprising modulating c-c chemokine receptor type 3 (CCR3)
US11912998B2 (en) 2010-01-28 2024-02-27 The Board Of Trustees Of The Leland Stanford Junior University Method of treating aging-associated cognitive impairment by reducing CCR3
US10626399B2 (en) 2010-01-28 2020-04-21 The Board Of Trustees Of The Leland Stanford Junior University Methods of treating cognitive symptoms of an aging-associated impairment by modulating C-C chemokine receptor type 3 (CCR3)
US10487148B2 (en) 2010-01-28 2019-11-26 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated impairments
US10688154B2 (en) 2011-04-08 2020-06-23 The Board Of Trustees Of The Leland Stanford Junior University Methods of neuroprotection involving macrophage colony stimulating factor receptor agonists
US10905779B2 (en) 2013-12-09 2021-02-02 The Board Of Trustees Of The Leland Stanford Junior University Methods for screening human blood products comprising plasma using immunocompromised rodent models
US10688130B2 (en) 2013-12-09 2020-06-23 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
US10617744B2 (en) 2015-06-15 2020-04-14 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
US11141469B2 (en) 2015-06-15 2021-10-12 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
IL278004B2 (en) * 2015-06-15 2023-04-01 Univ Leland Stanford Junior Methods and compositions for treating aging-associated conditions
AU2016279804B2 (en) * 2015-06-15 2019-03-07 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
CN107921085A (en) * 2015-06-15 2018-04-17 小利兰·斯坦福大学托管委员会 For treating the method and composition of aging-related disorders
IL278004A (en) * 2015-06-15 2020-11-30 Univ Leland Stanford Junior Methods and compositions for treating aging-associated conditions
CN107921085B (en) * 2015-06-15 2021-10-22 小利兰·斯坦福大学托管委员会 Methods and compositions for treating aging-related disorders
WO2016205004A3 (en) * 2015-06-15 2017-04-13 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for treating aging-associated conditions
KR20180025895A (en) * 2015-06-15 2018-03-09 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Methods and compositions for treating diseases associated with aging
KR102240347B1 (en) 2015-06-15 2021-04-15 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Methods and compositions for treating diseases associated with aging
US10905717B2 (en) 2016-04-28 2021-02-02 Alkahest, Inc. Blood plasma and plasma fractions as therapy for tumor growth and progression
US10245285B2 (en) 2016-04-28 2019-04-02 Alkahest, Inc. Blood plasma and plasma fractions as therapy for tumor growth and progression
US10525107B2 (en) 2016-08-18 2020-01-07 Alkahest, Inc. Blood plasma fractions as a treatment for aging-associated cognitive disorders
US11040068B2 (en) 2017-04-26 2021-06-22 Alkahest, Inc. Dosing regimen for treatment of cognitive and motor impairments with blood plasma and blood plasma products
US10874692B2 (en) 2017-04-26 2020-12-29 Alkahest, Inc. Dosing regimen for treatment of cognitive and motor impairments with blood plasma and blood plasma products
US11413308B2 (en) 2017-04-26 2022-08-16 Alkahest, Inc. Dosing regimen for treatment of cognitive and motor impairments with blood plasma and blood plasma products
US10357513B2 (en) 2017-04-26 2019-07-23 Alkahest, Inc. Dosing regimen for treatment of cognitive and motor impairments with blood plasma and blood plasma products
US11773138B2 (en) 2018-10-12 2023-10-03 Theradaptive, Inc. Polypeptides including a beta-tricalcium phosphate-binding sequence and uses thereof
US11103530B2 (en) 2018-10-26 2021-08-31 Alkahest, Inc. Methods of improving or accelerating postoperative recovery
US11298377B2 (en) 2018-10-26 2022-04-12 Alkahest, Inc. Methods of improving wound healing
US11547724B2 (en) 2018-10-26 2023-01-10 Alkahest, Inc. Methods of treating a subject for pain

Also Published As

Publication number Publication date
EP2084275A2 (en) 2009-08-05
WO2008063291A3 (en) 2008-10-02
WO2008063291A2 (en) 2008-05-29
US7592009B2 (en) 2009-09-22
AU2007322346A1 (en) 2008-05-29
EP2084275A4 (en) 2012-01-04
US20080118523A1 (en) 2008-05-22
JP2010505444A (en) 2010-02-25
CA2665839A1 (en) 2008-05-29
IL198120A0 (en) 2011-08-01

Similar Documents

Publication Publication Date Title
US7592009B2 (en) Polypeptide ligands for targeting cartilage and methods of use thereof
Kim et al. Challenge to overcome current limitations of cell-penetrating peptides
JP4129298B2 (en) Random peptides that bind to the gastrointestinal tract (GIT) transport receptor and related methods
DK2547692T5 (en) PEPTID DERIVATIVES, PREPARING thereof, AND APPLICATIONS THEREOF
JP6666613B2 (en) Malignant tumor targeting peptide
EA022976B1 (en) Peptide derivatives and use thereof as vectors for molecules in the form of conjugates
US20180155394A1 (en) Cancer cell-targeting peptide and use thereof
JP6032854B2 (en) Cyclic peptides having anti-neoplastic and anti-angiogenic activity
WO2006041205A1 (en) Angiogenesis promoter
US20170246311A1 (en) Peptides for binding alternatively activated macrophages
US20170173107A1 (en) Compstatin Analogs With Improved Potency and Pharmacokinetic Properties
US11357863B2 (en) Peptide conjugates
JP2023523262A (en) Novel cell-penetrating peptide and use thereof
EP2575848A2 (en) Caged cell penetrating peptide-polymer conjugates for diagnostic and therapeutic applications
KR102581801B1 (en) Tumor-targeting peptide variants
JP6583411B2 (en) Drug complex
Shah et al. Polyamide backbone modified cell targeting and penetrating peptides in cancer detection and treatment
US20050277575A1 (en) Therapeutic compositions and methods for treating diseases that involve angiogenesis
CN114096848A (en) Therapeutic peptides
WO2023165476A1 (en) Polypeptide compound specifically targeting sort1, and drug conjugate thereof
KR20230006405A (en) Novel cell penetrating peptides and use thereof
WO2024033929A1 (en) Peptides for the treatment of fibrosis
Cupka Development of an αᵥβ₆-Binding Peptide for In Vivo Applications: Modulation of Serum Stability and Biodistribution
AU2022251159A9 (en) Synthetic peptide shuttle agent bioconjugates for intracellular cargo delivery
Dunshee Development and characterization of extracellular matrix inspired peptides for nanoparticle self-assembly

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION