EP2938626A1 - Hydrogels peptidiques ultracourts auto-assemblés pour cicatrisation de plaies, soins de la peau et applications cosmétiques - Google Patents

Hydrogels peptidiques ultracourts auto-assemblés pour cicatrisation de plaies, soins de la peau et applications cosmétiques

Info

Publication number
EP2938626A1
EP2938626A1 EP13869801.4A EP13869801A EP2938626A1 EP 2938626 A1 EP2938626 A1 EP 2938626A1 EP 13869801 A EP13869801 A EP 13869801A EP 2938626 A1 EP2938626 A1 EP 2938626A1
Authority
EP
European Patent Office
Prior art keywords
peptoid
peptide
hydrogel
amphiphilic peptide
amphiphilic
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.)
Withdrawn
Application number
EP13869801.4A
Other languages
German (de)
English (en)
Other versions
EP2938626A4 (fr
Inventor
Charlotte Hauser
Yihua LOO
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.)
Agency for Science Technology and Research Singapore
Original Assignee
Agency for Science Technology and Research Singapore
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 Agency for Science Technology and Research Singapore filed Critical Agency for Science Technology and Research Singapore
Publication of EP2938626A1 publication Critical patent/EP2938626A1/fr
Publication of EP2938626A4 publication Critical patent/EP2938626A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06026Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atom, i.e. Gly or Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0806Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/08Tripeptides
    • C07K5/0802Tripeptides with the first amino acid being neutral
    • C07K5/0804Tripeptides with the first amino acid being neutral and aliphatic
    • C07K5/0808Tripeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1008Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 0 or 1 carbon atoms, i.e. Gly, Ala
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/101Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms, e.g. Val, Ile, Leu
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention provides an amphiphilic linear peptide and/or peptoid as well as a hydrogel that includes the amphiphilic linear peptide/peptoid.
  • the amphiphilic linear peptide/peptoid is capable of self-assembling into three-dimensional macromolecular nanofibrous networks, which entrap water, and forming a hydrogel.
  • These pep- tides/peptoids include short amphiphilic sequences with a hydrophobic portion of aliphatic amino acids and at least one acidic, neutral, or basic polar amino acid.
  • the amphiphilic linear peptide/peptoid is build up of non-repetitive aliphatic amino acids, which may be in the L- or D-form.
  • a plurality of such peptides/peptoids assembles to supramolecular helical fibers and forms peptide hydrogels after assembly.
  • a corresponding hydrogel is formed in aqueous solutions at physiological pH and is thus useful for inter alia cell culture, tissue engineering, tissue regeneration, wound healing and release of bioactive moieties (including cells, nucleic acids, anti-microbials, micro-/nanoparticles, cosmetic agents and small molecule therapeutics), as well as for providing mechanical support for damaged or missing tissues.
  • Such hydrogels can also be formed in situ, wherein the gelation process occurs within the body following the injection of a peptide solution.
  • Such hydrogels which are rigid, biocompatible and entrap up to 99.9% of water, are also well suited for applications utilizing electronic devices.
  • Supramolecular structures are held together by intermolecular bondings that are responsible for the organization of polymolecular systems.
  • the non-covalent, intermolecular forces which are required for the assembly of the defined supramolecular structures are mainly electrostatic interactions, hydrogen bonds, van der Waals forces, etc.
  • Supramolecular chemistry or biology gathers a vast body of two or three dimensional complex structures and entities formed by association of chemical or biological species. These associations are governed by the principles of molecular complementarity or molecular recognition and self-assembly.
  • peptide-based biomaterials are powerful tools for potential applications in biotechnology, medicine and even technical applications. Depending on the individual properties these peptide-based hydrogels are thought to serve in the development of new materials for tissue engineering, regenerative medicine, as drug and vaccine delivery vehicles or as peptide chips for pharmaceutical research and diagnosis (E. Place et al., Nature Materials, 8, 457-470, 2009). There is also a strong interest to use peptide-based self-assembled biomaterial such as gels for the development of molecular electronic devices (A. R. Hirst et al. Angew. Chem. Int. Ed., 47, 8002-8018, 2008)
  • hydrogels contain macroscopic structures such as fibers that entangle and form meshes. Most of the peptide-based hydrogels utilize as their building blocks ⁇ -pleated sheets which assemble to fibers. Later it was shown that it is possible to design hydrogelating self- assembling fibers purely from a-helices. Besides ⁇ -sheet structure-based materials (S. Zhang et al., PNAS, 90, 3334-3338, 1993: A.
  • peptide hydrogels are in most of the cases associated with low rigidity, sometimes unfavourable physiological properties and/or complexity and the requirement of substantial processing thereof which leads to high production costs.
  • the hydrogels should also be suitable for the delivery of bioactive moieties (such as nucleic acids, small molecule therapeutics, cosmetic and anti-microbial agents) and/or for use as biomimetic scaffolds that support the in vivo and in vitro growth of cells and facilitate the regeneration of native tissue.
  • Stimuli-responsive gelation of the peptides would also be desirable, as they could then be applied as a minimally invasive injectable therapy or implanted as a biological construct to replace damaged/missing tissue.
  • biocompatible compound that is capable of forming a hydro gel that meets at least some of the above requirements to a higher extent than cur- rently available hydrogels and that is not restricted by the above mentioned limitations.
  • amphiphilic peptide and/or peptoid capable of self-assembling into three-dimensional macromolecular nanofibrous networks, which entrap water and form a hydrogel
  • the amphiphilic peptide and/or peptoid compris- ing an amphiphilic sequence consisting of: a hydrophobic sequence stretch of n aliphatic amino acids, wherein n is an integer from 2 to 15, and a liydrophilic sequence stretch linked to said hydrophobic sequence stretch and having a polar moiety which is acidic, neutral or basic, said polar moiety comprising m adjacent liydrophilic amino acids, wherein m is an integer from 1 to 5.
  • the amphiphilic peptide and/or peptoid has a C-tenninus and an N- terminus, wherein both said C-tenninus and said N-tenninus do not carry any protecting groups attached to them. In one embodiment, the amphiphilic peptide and/or peptoid has a C-tenninus and an N- temiinus, wherein the N-teni inus is protected by an N-tennmal protecting group.
  • the amphiphilic peptide and/or peptoid has a C-tenninus and an N- tenninus, wherein the C-tenninus is protected by a C-tenninal protecting group.
  • said N-tenninal protecting group has the general fonnula -C(0)-R, wherein R is selected from the group consisting of H, unsubstituted or substituted alkyls, and unsubstituted or substituted aryls.
  • R is selected from the group consisting of H, unsubstituted or substituted alkyls, and unsubstituted or substituted aryls.
  • Prefened alkyls in accordance with the present invention are methyl, ethyl, propyl, iso- propyl, butyl and isobutyl.
  • said N-tenninal protecting group is an acetyl group.
  • said N-tenninal protecting group is a peptidomimetic molecule, including natural and synthetic amino acid derivatives, wherein the N-teraiinus of said peptidomimetic molecule may be modified with a functional group selected from the group consisting of carboxylic acid, amide, alcohol, aldehyde, amine, imine, nitrile, an urea analog, thiol, phosphate, carbonate, sulfate, nitrate, maleimide, vinyl sulfone, azide, al- kyne, alkene, carbohydrate, imide, peroxide, ester, thioester, aryl, ketone, sulphite, nitrite, phosphonate and silane.
  • said C-tenninal protecting group is an amide group.
  • the C-terminus of said amphiphilic peptide and/or peptoid has the formula -CONHR or -CONRR', with R and R' being selected from the group consisting of H, unsubstituted or substituted alkyls, and unsubstituted or substituted aryls.
  • said C-terminal protecting group is an ester group.
  • the C-terminus of said amphiphilic peptide and/or peptoid has the formula -C0 2 R , with R being selected from the group consisting of H, unsubstituted or substituted alkyls, and unsubstituted or substituted aryls.
  • said C-terminal protecting group is a peptidomimetic molecule, including natural and synthetic amino acid derivatives, wherein the C-terminus of said peptidomimetic molecule may be modified with a functional group selected from the group consisting of carboxylic acid, amide, alcohol, aldehyde, amine, imine, nitrile, an urea analog, thiol, phosphate, carbonate, sulfate, nitrate, maleimide, vinyl sulfone, azide, al- kyne, alkene, carbohydrate, imide, peroxide, ester, thioester, aryl, ketone, sulphite, nitrite, phosphonate and silane.
  • the amphiphilic peptide and/or peptoid has an N-tem inal protecting group, which is an acetyl group, and a C-terminal protecting group, which is an amide group.
  • n is an integer from 2 to 6, preferably 2 to 5.
  • m is an integer from 1 to 2, preferably 1.
  • amphiphilic peptide and/or peptoid consists of o amphiphilic sequences, as defined above, which amphiphilic sequences are linked to each other, o being an integer from 1 to 50.
  • said aliphatic amino acids and said hydrophilic amino acids are either D-amino acids or L-amino acids.
  • each of the hydrophilic amino acids has a polar group which is independently selected from a hydroxyl, an ether, a carboxyl, an imido, an amido, an ester, an amino, a guanidino, a thio, a thioether, a seleno, and a telluro group.
  • said polar moiety of said hydrophilic sequence stretch comprises m adjacent hydrophilic amino acids, m being as defined above, said hydrophilic amino acids being selected from the group comprising aspartic acid, asparagine, glutamic acid, glutmnine, 5 -N-ethyl- glutamine (theanine), citrulline, thio-citrulline, cysteine, homocys- teine, methionine, ethionine, selenomethionine, telluromethionine, threonine, allo- threonine, serine, homoserine, arginine, homoarginine, ornithine (Orn), 2,4- diaminobutyric acid (Dab or Dbu), 2,3-diaminopropionic acid (Dap or Dpr), lysine and N(6)-carboxy-methyllysine, histidine, and wherein said hydrophobic sequence stretch comprises n alipha
  • m is 1 or 2. In one embodiment, m is 2 and said polar moiety comprises two identical amino acids, or m is 1 and said polar moiety comprises any one of aspartic acid, asparagine, glutamic acid, glutamine, serine, threonine, cysteine, methionine, lysine, ornithine (Orn), 2,4- diaminobutyric acid (Dab or Dbu), 2,3-diaminopropionic acid (Dap or Dpr) and histidine. In one embodiment, said polar moiety is adjacent to the hydrophobic sequence stretch of n aliphatic amino acids.
  • said polar moiety has a sequence selected from Asp, Asn, Glu, Gin, Ser, Thr, Cys, Met, Lys, Orn, Dab, Dap, His, Asn-Asn, Asp-Asp, Glu-Glu, Gln-Gln, Asn-Gln, Gln-Asn, Asp-Gin, Gin-Asp, Asn-Glu, Glu-Asn, Asp-Glu, Glu-Asp, Gln-Glu, Glu-Gln, Asp-Asn, Asn-Asp Thr-Thr, Ser-Ser, Thr-Ser, Ser-Thr, Asp-Ser, Ser-Asp, Ser- Asn, Asn-Ser, Gln-Ser, Ser-Gln, Glu-Ser, Ser-Glu, Asp-Thr, Thr-Asp, Thr-Asp, Asn, Asn-Ser, Gln-Ser, Ser-G
  • said polar moiety comprises the C-terminus of the amphiphilic peptide and/or peptoid, or said polar moiety comprises the N-terminus of the amphiphilic peptide and/or peptoid. In one embodiment, said polar moiety comprises the C-terminus of the amphiphilic peptide and/or peptoid.
  • said polar moiety consists of at least one amino acid positioned at the C-terminus of the amphiphilic peptide and/or peptoid.
  • said hydrophobic sequence stretch comprises and/or forms the N- terminus of the amphiphilic peptide and/or peptoid.
  • said aliphatic amino acids arranged in an order of decreasing amino acid size have a sequence which is a repetitive or non-repetitive sequence.
  • said aliphatic amino acids arranged in order of decreasing amino acid size have a sequence with a length of 2 to 7, preferably 2 to 6, more preferably 2 to 5 amino acids.
  • said aliphatic amino acids arranged in an order of decreasing amino acid size have a sequence selected from LIVAG, ILVAG, LIVAA, LAV AG, IV AG, LIVA, LIVG, IVA and IV, wherein, optionally, there is an A preceding such sequence at the N-terminus.
  • all or a portion of the aliphatic amino acids of the hydrophobic se- quence stretch are arranged in an order of identical amino acid size in the amphiphilic peptide and/or peptoid.
  • said aliphatic amino acids arranged in order of identical amino acid size have a sequence with a length of 2 to 4 amino acids.
  • said aliphatic amino acids arranged in an order of identical size have a sequence selected from LLLL, LLL, LL, IIII, III, II, VWV, WV, VV, AAAA, AAA, AA, GGGG, GGG, and GG.
  • the amphiphilic sequence undergoes a conformational change during self-assembly, preferably a conformational change from a random coil conformation to a helical intermediate structure to a final beta conformation.
  • the conformational change is dependent on the concentration of the amphiphilic peptide and/or peptoid, dependent on the ionic environment (e.g. the salt concentration), pH dependent and/or temperature dependent. In one embodiment, the conformational change is facilitated or triggered by changes in pH, ionic environment and/or temperature.
  • the amphiphilic linear sequence comprises a single hydrophilic and at least two aliphatic amino acids.
  • the amphiphilic sequence is one of SEQ ID NO: 1 to 86.
  • the amphiphilic peptide and/or peptoid is stable in aqueous solution at physiological conditions at ambient temperature for a period of time in the range from 1 day to at least 6 months, preferably to at least 8 months more preferably to at least 12 months.
  • the amphiphilic peptide and/or peptoid is stable in aqueous solution at physiological conditions, at a temperature up to 90 °C, for at least 1 hour.
  • the amphiphilic peptide and/or peptoid has the general formula:
  • Z is an N-terminal protecting group
  • X is, at each occurrence, independently selected from an aliphatic amino acid
  • Y is, at each occurrence, independently selected from a hydrophilic amino acid
  • Z' is a C-tenninal protecting group
  • n is an integer selected from 2 to 6, preferably 2 to 5,
  • n is selected from 1 and 2, wherein, preferably, m is 1,
  • p and q are independently selected from 0 and 1 , wherein, preferably, p is 1.
  • said aliphatic amino acid is selected from the group comprising isoleucine, norleucine, leucine, valine, alanine, glycine, homoallylglycine and homopropargylgly- cine.
  • said hydrophilic amino acid is selected from the group comprising as- partic acid, asparagine, glutamic acid, glutamine, 5-N-ethyl-glutamine (theanine), citrul- line, thio-citrulline, cysteine, homocysteine, methionine, ethionine, selenomethionine, telluromethionine, threonine, allo-threonine, serine, homoserine, arginine, homoarginine, ornithine (Oni), 2,4-diaminobutyric acid (Dab or Dbu), 2,3-diaminopropionic acid (Dap or Dpr), lysine and N(6)
  • the C-tenninal amino acid of the amphiphilic peptide and/or peptoid is a neutral or basic hydi philic (polar) amino acid.
  • m is 1, and Y is selected from neutral or basic hydrophilic amino acids.
  • Preferred neutral hydrophilic amino acids include serine and threonine.
  • Preferred basic hydrophilic amino acids include lysine (K), ornithine (Orn), 2,4-diaminobutyric acid (Dab or Dbu) and 2,3-diaminopropionic acid (Dap or Dpr).
  • the C-tenninal amino acid of the amphiphilic peptide and/or peptoid is selected from the group consisting of a lysine (K), an ornithine (Om), a 2,4- diaminobutyric acid (Dab or Dbu) and a 2,3-diaminopropionic acid (Dap or Dpr).
  • m is 1
  • Y is selected from the group consisting of a lysine (K), an ornithine (Orn), a 2,4-diaminobutyric acid (Dab or Dbu) and a 2,3-diaminopropionic acid (Dap or Dpr).
  • the amphiphilic peptide and/or peptoid has a sequence selected from the group comprising LIVAGK (SEQ ID NO: 19 or 39), LIVAG(Om) (SEQ ID NO: 43 or 44), LIVAG(Dab) (SEQ ID NO: 45 or 46), LIVAG(Dap) (SEQ ID NO: 47 or 48), ILVAGK (SEQ ID NO: 49 or 50), ILVAG(Orn) (SEQ ID NO: 51 or 52), ILVAG(Dab) (SEQ ID NO: 53 or 54), ILVAG(Dap) (SEQ ID NO: 55 or 56), AIVAGK (SEQ ID NO: 57 or 58), AIVAG(Orn) (SEQ ID NO: 59 or 60), AIVAG(Dab) (SEQ ID NO: 61 or 62), AIVAG(Dap) (SEQ ID NO: 63 or 64), IIIK (SEQ ID NO: 27 or 28), Ill(Ora)
  • the amphiphilic peptide and/or peptoid has an acetylated N-terminus.
  • the N-terminal amino acid of the amphiphilic peptide and/or peptoid is an isoleucine (I).
  • a hydrogel comprising at least one amphiphilic peptide and/or peptoid as defined above.
  • the hydrogel can comprise more than one amphiphilic peptide and/or peptoid, such as two, three, four or more amphiphilic peptides and/or pep- toids, which can differ in their amino acid sequence, N- and/or C-tenninal protecting group.
  • the hydrogel is stable in aqueous solution at ambient temperature for a period of at least 7 days, preferably at least 2 to 4 weeks, more preferably at least 1 to 6 months.
  • the hydrogel is characterized by a storage modulus G' to loss modulus G" ratio that is greater than 2.
  • the hydrogel is characterized by a storage modulus G' from 100 Pa to 80,000 Pa at a frequency in the range of from 0.02 Hz to 16 Hz.
  • the hydrogel further comprises a non-peptidic polymer.
  • the at least one non-peptidic polymer is present at a concentration of 50% (w/w) or less, preferably 40% (w/w) or less, with respect to the total weight of the hydrogel.
  • Such non- peptidic polymer may be used to modify the mechanical properties of the hydrogel (e.g. increasing its elasticity) and/or for the coupling of bioactive agents/moieties.
  • Composite hydrogels comprising an additional non-peptidic polymer are described in detail in PCT/SG2012/000421, which is hereby incorporated herein by reference in its entirety.
  • the hydrogel may comprise ultrasmall self-assembling natural peptides (dimers to hexamers) that form organogels when dissolving them in organic solvents, oils and/or mixtures of oils.
  • organogels are described in detail in SG201201239-9, which is also incorporated herein by reference in its entirety.
  • the hydrogel comprises fibers of the at least one amphiphilic peptide and/or peptoid as defined above, said fibers defining a network that is capable of entrapping at least one of a microorganism, a cell, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, an oligosaccharide, a polysaccharide, a vitamin, an inorganic mole- cule, a nano- or microparticle, a synthetic polymer, a small organic molecule, a cosmetic agent or a pharmaceutically active compound.
  • the hydrogel comprises at least one of a microorganism, a cell, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, an oligosaccharide, a polysaccharide, a vitamin, an inorganic molecule, a nano- or microparticle, a synthetic polymer, a small organic molecule, a cosmetic agent or a pharmaceutically active compound entrapped by the network of fibers of the amphiphilic peptide and/or peptoid.
  • the at least one of a microorganism, a cell, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, an oligosaccharide, a polysaccharide, a vitamin, an inorganic molecule, a nano- or microparticle, a synthetic polymer, a small organic molecule, a cosmetic agent or a pharmaceutically active compound is coupled to said non- peptidic polymer.
  • said pharmaceutically active compound is selected from the group comprising haemostatic agents, antibiotics, anti-microbial agents, anti-fungal agents, anti-inflammatory agents, analgesics, anti-coagulants, antibodies, antigens, growth factors and cytokines.
  • the hydrogel is comprised in at least one of a fuel cell, a solar cell, an electronic cell, a biosensing device, a medical device, an implant, a wound dressing, a phannaceutical composition and a cosmetic composition.
  • the phannaceutical composition or the cosmetic composition are provided in the fonn of a topical gel or cream, a spray, a powder, or a sheet, patch or membrane.
  • the pharmaceutical composition or the cosmetic composition is provided in the form of an injectable solution, which, preferably, gels in situ following injection into the body.
  • the objects are also solved by a hydrogel as defined above for use in at least one of the following: release of a pharmaceutically active compound, medical tool kit, a fuel cell, a solar cell, an electronic cell, tissue regeneration, tissue replacement, wound healing, skin care, stem cell therapy and gene therapy.
  • a method of preparing a hydrogel the method comprising dissolving at least one amphiphilic peptide and/or peptoid as defined above in an aqueous solution.
  • the dissolved at least one amphiphilic peptide and/or peptoid in aqueous solution is further exposed to temperature, wherein the temperature is in the range from 20 °C to 90 °C, preferably from 20 °C to 70 °C.
  • the dissolved at least one amphiphilic peptide and/or peptoid in aqueous solution is exposed to a temperature in the range from 35 °C to 40 °C (body temperature).
  • the at least one amphiphilic peptide and/or peptoid is dissolved at a concentration from 0.01 to 100 mg/ml, preferably at a concentration from 1 mg/ml to 50 mg ml, more preferably at a concentration from about 1 mg/ml to about 20 mg/ml.
  • the method further comprises at least one of the steps of:
  • said gelation enhancer is a salt or a solution of a salt. In one embodiment, said gelation enhancer is a cross-linking agent.
  • said adding at least one non-peptidic polymer further comprises mix- ing or cross-linking the at least one non-peptidic polymer with the at least one amphiphilic peptide and/or peptoid.
  • a surgical implant or stent comprising a peptide and/or peptoid scaffold, wherein the peptide and/or peptoid scaffold is formed by a hydrogel as defined above.
  • a pharmaceutical and/or cosmetic composition and/or a biomedical device and/or electronic device comprising at least one amphiphilic peptide and/or peptoid as defined above or a hydrogel as defined above.
  • the pharmaceutical and/or cosmetic composition is applied topically. In one embodiment, the pharmaceutical and/or cosmetic composition is injectable.
  • kits of parts comprising a first container with at least one amphiphilic peptide and/or peptoid as defined above and a second container with an aqueous solution.
  • the aqueous solution of the second container further comprises a pharmaceutically active compound.
  • the first container with at least one amphiphilic peptide and/or pep- toid further comprises a pharmaceutically active compound.
  • the kit of parts further comprises a third container with a gelation enhancer.
  • said gelation enhancer is a salt or a solution of a salt.
  • At least one of said first, second or third container is provided as a spray bottle or a syringe. In one embodiment, all of said first, second and third container are provided as spray bottles.
  • kits of parts comprising a first container with an aqueous solution of at least one amphiphilic peptide and/or peptoid as defined above and a second container with a gelation enhancer.
  • the first container further comprises a pharmaceutically active compound.
  • said gelation enhancer is a salt or a solution of a salt.
  • At least one of said first arid said second container is provided as a spray bottle or a syringe. In one embodiment, both said first and said second container are provided as spray bottles or syringes.
  • said first and said second container are provided in the form of separate compartments of a spray bottle or syringe.
  • tissue regeneration or tissue replacement comprising the steps: a) providing a hydrogel as defined above;
  • the method is performed in vitro or in vivo or ex vivo.
  • ex vivo refers, for example, to a scenario, where said cells are taken from the patient and cultured on a scaffold formed by a hydrogel as defined above, which scaffold is then implanted back into the patient.
  • the method is performed in vivo, wherein, in step a), said hydrogel is provided at a place in the body of a patient where tissue regeneration or tissue replace- ment is intended.
  • said tissue is selected from the group comprising skin tissue, nucleus pulposus in the intervertebral disc, cartilage tissue, synovial fluid and submucosal connective tissue in the bladder neck.
  • said step a) is perfom ed by injecting said hydrogel or a solution of at least one amphiphilic peptide and/or peptoid as defined above (i.e. a precursor of the hydrogel) at a place in the body of a patient where tissue regeneration or tissue replacement is intended.
  • said step a) further comprises the co-injection of a gelation enhancer, preferably of a solution of a salt.
  • said step a) further comprises the co-injection of cells.
  • the method is performed ex vivo, wherein, in step a) or b), cells from a patient or from a donor are mixed with said hydrogel, and the resulting mixture is provided at a place in the body of a patient where tissue regeneration or tissue replacement is intended.
  • said hydrogel comprises one or more bioactive therapeutics that stimulate regenerative processes and/or modulate the immune response.
  • a method of wound treatment comprising the steps: a) providing a hydrogel as defined above;
  • said wound is a bum wound.
  • said hydrogel induces autolytic debridement.
  • said hydrogel comprises at least one amphiphilic peptide and/or peptoid, wherein the C-terminal amino acid of the amphiphilic peptide and/or peptoid is selected from the group consisting of a lysine (K), an ornithine (Ora), a 2,4-diaminobutyric acid (Dab or Dbu) and a 2,3-diaminopropionic acid (Dap or Dpr).
  • said hydrogel functions as a haemostatic agent.
  • said hydrogel functions as an anti-inflammatory agent, which acts by scavenging extracellular nucleic acids that stimulate the innate immune system.
  • said hydrogel in step a), " said hydrogel is mixed with cells that repopulate the wound and expedite wound healing.
  • said hydrogel comprises one or more bioactive therapeutics that diffuse into the wound and facilitate the healing response by modulating the inflammatory response and/or stimulating regenerative processes.
  • an amphiphilic peptide and/or peptoid or of a hydrogel comprising the same or of a composi- tion comprising such peptide/peptoid/hydrogel for the manufacture of a medicament for a method of tissue regeneration or tissue replacement or for a method of wound treatment, wherein these methods are as defined further above.
  • the present invention provides an amphiphilic peptide and/or peptoid capable of self-assembling into three-dimensional macromolecular nanofibrous networks, which entrap water and form a hydro gel.
  • the amphiphilic peptide and/or peptoid includes a hydrophobic and a hydrophilic sequence.
  • This hydrophobic sequence has a length of n L- or D- amino acids, n is an integer, which may typically range from 2 to about 15.
  • the hydrophilic sequence has a polar and/or charged moiety comprising m L- or D- amino acids, m is an integer from 1 to 5.
  • Each of the m aliphatic amino acids carries an independently selected polar group.
  • the amphiphilic linear sequence has a net charge at physiological pH and, preferably, an N-terminus carrying a protecting group.
  • the protecting group can be an acetyl group.
  • the amphiphilic peptide and/or peptoid may comprise o linked amphiphilic peptide and/or peptoid sequences of n hydrophobic and m hydrophilic L- and D- amino acids, wherein o is an integer from 1 to about 50.
  • the amphiphilic peptide and/or peptoid may consist of o linked amphiphilic peptide and/or peptoid sequences of n hydrophobic and m hydrophilic L- and D- amino acids.
  • the value of n may be an integer from 2 to about 15.
  • the value of m may be 1 to 5.
  • the charged and/or polar group of each of the m hydrophilic L- and D- amino acids may be independently selected from a hydroxyl, an ether, a carboxyl, an amido, an ester, an amino, a guanidino, a thio, a thioether, a seleno, and a telluro group.
  • the charged or polar moiety of the hydrophilic sequence may comprise m L-or D-amino acids selected from the group consisting of aspartic acid, asparagine, glutamic acid, glutamine, 5-N-ethyl- glutamine (theanine), citrulline, thio-citrulline, cysteine, homocysteine, methionine, ethi- onine, selenomethionine, telluromethionine, threonine, allo-threonine, serine, homoser- ine, arginine, homoarginine, ornithine (Orn), 2,4-diaminobutyric acid (Dab or Dbu), 2,3- diaminopropionic acid (Dap or Dpr), lysine and N(6)-carboxymethyllysine.
  • aspartic acid asparagine, glutamic acid, glutamine, 5-N-ethyl- glutamine (theanine), citrulline,
  • the charged and/or polar moiety of the hydrophilic sequence may comprise two identical amino acids.
  • the two identical amino acids may be adjacent to the non-polar hydrophobic moiety.
  • the charged and/or polar moiety may consist of two amino acids with a sequence selected from Asn-Asn, Asp-Asp, Glu-Glu, Gln-Gln, Asn-Gln, Gln-Asn, Asp-Gin, Gin- Asp, Asn-Glu, Glu-Asn, Asp-Glu, Glu-Asp, Gln-Glu, Glu-Gln, Asp-Asn, Asn-Asp, Thr-Thr, Ser-Ser, Thr-Ser, Ser-Thr, Asp-Ser, Ser-Asp, Ser-Asn, Asn-Ser, Gln-Ser, Ser-Gln, Glu- Ser, Ser-Glu, Ser-Glu, Asp-Thr, Thr-Asp, Thr
  • the charged and/or polar moiety may comprise the C-terminus of the amphiphilic peptide and/or peptoid.
  • the charged and/or polar moiety may comprise (i) the C-terminus, the C-tenninus carrying an unprotected C-terminal carboxyl group or (ii) the N-terminus, the N-terminus carrying an unprotected N-terminal amino group.
  • the charged and/or polar moiety may comprise the C-terminus of the amphiphilic peptide and/or peptoid, the C-tenninus carrying an unprotected C-terminal carboxyl group and wherein the N- terminus carries a protecting group preferably the acetyl group.
  • the charged and/or polar moiety may comprise the C-terminus of the amphiphilic peptide and/or peptoid, the C- terminus carrying a protected C-terminal carboxyl group, preferably protected with an amido or ester group, and the N-terminus carrying a protecting group, preferably the acetyl group.
  • the protecting group may be an amido protecting group.
  • the charged and/or polar moiety may consist of at least one amino acid positioned at the C-tenninus of the amphiphilic peptide and/or peptoid.
  • the hydrophobic sequence may comprise at least two aliphatic amino acids that is defined by a main chain comprising 1 to about 20 carbon atoms.
  • a portion of the amino acids of the non-polar moiety may be arranged in a general sequence of decreasing size in the direction from N- to C-tenninus of the amphiphilic peptide and/or peptoid, and the size of adjacent amino acids of the non-polar moiety may be identical or smaller in the direction of the general sequence of decreasing size.
  • the general sequence of decreasing size may be preferably a non-repetitive sequence.
  • the direction of the general sequence of decreasing size in which adjacent amino acids may be of identical or smaller size may be the direction toward the charged and/or polar moiety of the sequence.
  • the portion of the amino acids an-anged in a general sequence of decreasing size may have a length of 2-7, preferably 2-6, more preferably 2, 3, 4, 5 or 6 amino acids.
  • the portion of the amino acids arranged in a general sequence of decreasing size may also have a length of n-m-1 amino acids and wherein the portion of the amino acids areanged in the general sequence of decreasing size may be positioned between the remaining non-polar amino acid of the non-polar moiety of n-m amino acids and the polar moiety.
  • the remaining non-polar amino acid of the non-polar moiety of n- m amino acids may define the N-terminus or the C-terminus of the amphiphilic peptide and/or peptoid.
  • the remaining non-polar amino acid of the non-polar moiety of n-m amino acids may be one of alanine, valine and glycine.
  • the amphiphilic linear sequence may undergo a conformational change from a random coil conformation to a helical conformation during self-assembly.
  • the conformational change may be concentration, pH, temperature and salt concentration dependent.
  • the non-polar moiety of the amphiphilic linear sequence may comprise at least one L-or D-amino acid selected from the group consisting of glycine, homoallylglycine, homopropargylglycine, alanine, valine, leucine, norleucine and isoleucine.
  • the amphiphilic linear sequence may comprise a single polar and/or charge and a single non-polar moiety.
  • the amphiphilic linear sequence may have a positive or a negative net charge.
  • the net charge may be from about -1 to about -4 or- from about +5 to about +1.
  • the net charge may be from about -1 to about -2.
  • the net charge may be -2.
  • the net charge may be +1 or +2 or +5.
  • the amphiphilic peptide and/or peptoid may be stable in aqueous solution at physiological conditions at ambient temperature for a period of time in the range from 1 day to at least 6 months, preferably at least 8 months, more preferably at least 12 months.
  • the amphiphilic peptide and/or peptoid may be stable in aqueous solution at physiological conditions at a temperature to 90 °C for at least 1 hour.
  • the C-terminal amino acid of the amphiphilic peptide and/or peptoid may be selected from the group consisting of a lysine (K), an ornithine (Orn), a 2,4- diaminobutyric acid (Dab or Dbu) and a 2,3-diaminopropionic acid (Dap or Dpr).
  • the N-terminal amino acid of the amphiphilic peptide and/or peptoid is an isoleucine (I).
  • the hydrogel may comprise more than one amphiphilic peptide and/or peptoid, such as two, three, four or more amphiphilic peptides and/or peptoids, which can differ in their amino acid sequence, N- and/or C-terminal protecting group.
  • the hydrogel may further comprise a non-peptidic polymer.
  • the invention provides a hydrogel.
  • the hydrogel includes an amphiphilic peptide and/or peptoid according to the first aspect.
  • the hydrogel may be sta- ble in aqueous solution at ambient temperature for a period of at least 7 days.
  • the hydrogel may be stable in aqueous solution at ambient temperature for a period of at least 2 to 4 weeks.
  • the hydrogel may be stable in aqueous solution at ambient temperature for a period of at least 1 to 6 months.
  • the hydrogel mechanical property may be characterized by a loss modulus G" to storage modulus G' ratio that is less thanl.
  • the hydrogel may be characterized by magnitude of storage modulus G' greater than loss modulus G" by minimum factor of 1.5.
  • the hydrogel may be characterized by a storage modulus G' of from 100 Pa to 80,000 Pa at a frequency in the range of from 0.02 Hz to 16 Hz.
  • the hy- drogel may be characterized by higher storage modulus G' with increase in the concentration of peptide.
  • the hydrogel may have a higher mechanical strength than collagen or hydrolyzed form (gelatine).
  • the hydrogel may comprise fibers of an amphophilic peptide and/or peptoid described herein.
  • the hydrogel may comprise at least one of a microorganism, a cell, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, an oligosaccharide, a polysaccharide, a vitamin, an inorganic molecule, a iiano- or microparticle, a synthetic polymer, a small organic molecule, a cosmetic agent or a pharmaceutically active compound entrapped by the network of fibers of the am- phiphilic polymer.
  • the at least one of a microorganism, a cell, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, an oligosaccharide, a polysaccharide, a vitamin, an inorganic molecule, a nano- or microparticle, a synthetic polymer, a small organic molecule, a cosmetic agent or a pharmaceutically active compound may also be coupled to the non- peptidic polymer.
  • the hydrogel may be comprised in at least one of a fuel cell, a solar cell, a electronic cell, a biosensing device, a medical device, an implant, a wound dressing, a phannaceutical composition, drug and vaccine delivery system, tissue culture medium, biosensor devices and a cosmetic composition.
  • the phannaceutical composition or the cosmetic composition may be provided in the form of a topical gel or cream, a spray, a powder, or a sheet, patch or membrane.
  • the phannaceutical composition or the cosmetic composition may also be provided in the fonn of an injectable solution, which, preferably, gels in situ following injection into the body.
  • the hydrogel may be for at least one of (controlled or sustained) release of a phannaceutically active compound, medical tool kit, a fuel cell, a solar cell, an electronic cell, tissue regeneration, tissue replacement, wound healing, skin care, stem cell therapy and gene therapy.
  • the hydrogel may be used for tissue regeneration, drug release or gene therapy.
  • the invention provides a method of preparing a hydrogel.
  • the method includes providing an amphiphilic peptide and/or peptoid according to the first aspect.
  • the method further includes dissolving and/or dispersing the amphiphilic peptide and/or peptoid in an aqueous solution.
  • the dissolved/dispersed amphiphilic peptide and/or pep- toid in aqueous solution may be further exposed to a temperature.
  • the temperature may be selected in the range from about 20 °C to about 90, preferably from 20°C to 70°C.
  • the amphiphilic peptide and/or peptoid may be dissolved at a concentration from about 0.01 ⁇ g/ml to about 100 mg ml.
  • the amphiphilic peptide and/or peptoid may be dissolved at a concentration from about 1 mg/ml to about 50 mg/ml.
  • the amphiphilic pep- tide and/or peptoid may be dissolved and/or dispersed at a concentration from about 1 mg/ml to about 30 mg/ml.
  • the method may further comprise at least one of the steps of adding at least one bioactive agent; adding at least one non-peptidic polymer; adding at least one gelation enhancer; adding at least one buffer, preferably at least one physiologically acceptable buffer.
  • the gelation enhancer may be a salt or a solution of a salt, in other embodiments, the gelation enhancer may be a cross-linking agent.
  • the invention provides a pharmaceutical and/or cosmetic composition.
  • the phamiaceutical and/or cosmetic composition includes the amphiphilic peptide and/or peptoid according to the first aspect or a hydrogel according to the second aspect.
  • the pharmaceutical and/or cosmetic composition may comprise a phannaceutically active compound.
  • the pharmaceutical and/or cosmetic composition may comprise a pharmaceutically acceptable carrier.
  • the invention provides a kit of parts.
  • the kit includes a first container and a second container.
  • the first container includes a peptide and/or peptoid according to the first aspect.
  • the second container includes an aqueous solution.
  • the aqueous solution of the second container may further comprise a pharmaceutically active compound.
  • the fist container with an amphiphilic peptide and/or peptoid may further comprise a phar- ; maceutically active compound.
  • the kit includes a first container with an aqueous solution of an amphiphilic peptide and/or peptoid according to the first aspect and a second container with a gelation enhancer.
  • the container(s) may be provided as spray bottles or syringes or compartments of syringes.
  • the invention provides for a method of tissue regeneration or tissue replacement.
  • the invention provide for a method of wound treatment.
  • Figures 1 A to 1 J represent a sorted list of some exemplary peptides of the invention capable of forming hydrogels. These peptides are embodiments in which the entire peptide consists of a single linear amphiphilic sequence. Peptides which are forming hydrogels are named with a short code, but their individual sequence is disclosed. The peptides of these examples consist of a sequence of natural amino acids containing 3 to 7 amino ac- ids. The N-terminus is acetylated which removes the charge that would otherwise restrain the amphiphilic character of the peptides.
  • Figure 2 depicts gelation pictures for peptide based hydrogels at lowest concentrations.
  • Figure 3 depicts gelation pictures for Ac-AS-6 (Ac-AIVAGS) (L) at concentrations of 5 mg/ml, 10 mg/ml, 15 mg/ml.
  • Figure 4 depicts a hypothesis of self-assembly from peptide monomers to supramolecu- lar network of condensed fibers.
  • A Assembly is believed to initiate with antiparallel pairing of two peptide monomers by changing to a-helical conformations. Subsequently, peptide pairs assemble to fibers and nano structures. Condensation of peptide fibers to fiber aggregates results in hydrogel formation.
  • Figure 5 depicts environmental scanning electron microscopy (ESEM) images of hydrogels of Ac-LD6 (Ac-LIVAGD) (L) (10 mg/ml), where Fig. 5 A, Fig. 5B and Fig.
  • FIG. 5C are images obtained at magnification of 260X, 1000X, 2000X, 2400X, 4000X at a temperature of 4 °C with HV at 10KV.
  • the images indicate the formation of fibrous structures.
  • Figure 6 shows field emission scanning electron microscopy (FESEM) images of hydrogels of Ac-LD6 (Ac-LIVAGD) (L) (15 mg/ml), where figures 6A-D are images obtained at magnifications of 6000X, 45000X, 45000X and 40000X with HV at 10 KV.
  • FESEM field emission scanning electron microscopy
  • Figure 7 depicts field emission scanning electron microscopy (FESEM) images of Ac- AD6 (Ac-AIVAGD) (D) hydrogels (20 mg/ml) at a magnification of 50 X (fig. 7A) and 20000 X (fig. 7 B) at 12 KV.
  • FESEM field emission scanning electron microscopy
  • Figure 8 shows field emission scanning electron microscopy (FESEM) images of hydrogels of Ac-AD6 (Ac-AIVAGD) (D) (20 mg/ml) obtained at 120 X (fig. 8A), and 450 X (fig. 8B).
  • FESEM field emission scanning electron microscopy
  • Figure 9A a)-f) shows the morphology and structure evaluation of the peptide scaffolds as determined by field emission scanning electron microscopy (a-f)
  • a-f A honeycomb porous structure is observed following lyophilization of 20 mg/mL Ac-AD 6 (Ac- AIVAGD) (D) hydrogel. The pores are bounded by membranes of condensed fibers as shown in close-up views of 15 mg/mL (b) and 20 mg/mL (c) Ac-ID 3 (Ac-IVD) (L) hy- drogels. Further magnification of 20 mg/mL Ac-AD 6 (L) hydrogel revealed single fibers (d, e). At lower concentrations, 0.1 mg/mL Ac-LD (Ac-LIVAGD) (L), nanostructures are observed (f).
  • Figure 9B shows an image obtained at a magnification of lOOOx, HV of 12 KV, Fig. 9C obtained at a magnification of 2500x, HV of 12 KV, Fig. 9D obtained at a magnification of 4000x, HV of 10 KV, Fig. 9E obtained at a magnification of 35000x, HV of 10 KV, Fig. 9F at a magnification of 80000x, HV of 5 KV, Fig. 9G obtained at a magnification of 120000x, HV of 10 KV, and Fig. 9H at a magnification of 200000x, HV of 10 KV.
  • Figure 10 shows (a) Far-UV CD spectra demonstrate that with increasing concentration there is the transition of Ac-LD 6 (Ac-LIVAGD) peptide conformation from random coil (below threshold concentration) to a-helical (222 and 208 nm peaks) and further ⁇ -type (negative band at 218 nm) structures.
  • Ac-LIVAGD Ac-LD 6
  • Figure 11 shows Rheology.
  • (a, b) The high mechanical strengths of different peptide hydrogels at 20 mg/mL concentration was determined by measuring storage moduli (G') as a function of angular frequency under 0.1 % strain, at 25 "C and 50°C respectively. The gels demonstrate good thermal stability compared to gelatin, which liquidified at 50°C (hence excluded in 4B).
  • (c) Mechanical strength is a function of concentration, as determined from oscillatory frequency sweep studies using Ac-LD 6 (Ac-LIVAGD) (L) under 0.1% strain at 25°C.
  • Figure 12 shows a further example of a rheology measurement for peptide based hydro- gels.
  • Figure 12A and Figure 12B depict oscillatory amplitude sweep studies at temperatures of 25°C and 50°C for Ac-AD6 (Ac-AIVAGD) (L) and Ac-AD6 (D) at a concentration of 20 mg/ml with a constant frequency of [lrad-s] and a gap of 0.8 mm.
  • the graphs indicate the plot of moduli [Pa] versus strain (%) at temperatures of 25°C and 50°C.
  • the linear viscoelastic range was observed at 0.07% to 0.2 strain % at temperatures of 25°C and 50°C.
  • FIG. 12D depict oscillatory frequency sweep Studies at temperatures of 25°C and 50°C for Ac-AD6(L) and Ac-AD6(D) at a concentration of 20 mg/ml with varying frequency ranges from 0.1 to 100 [Rad/s] with a constant strain [%] of 0.1% linear viscoelastic range and a gap of 0.8 mm.
  • Figure 13 shows a further example of a rheology measurement for peptide based hydro- gels. Depicted is a frequency sweep study of a uv cross-linked peptide at a temperature of 25 °C with 0.1 % strain.
  • Figure 14 depicts rheology measurements for gelatin- 1890 (type A, porcine skin). This figure shows moduli data obtained at 25 °C when applying different frequencies.
  • Figure 15 illustrates the biocompatibility of peptide-based hydrogels of the invention using further cell lines.
  • Fig. 15A shows a microscopy image of human primaiy renal tubule cells (HPRTC) after 72 hours after seeding on a hydro gel of Ac-LD 6 (Ac-LIVAGD) (L) in DMEM medium, grown at optimum conditions.
  • Fig. 15B shows microscopy images of human primary renal tubule cells (HPRTC) after 72 hrs after seeding on tissue culture plastic, grown at optimum conditions.
  • Fig. 15C shows microscopy images of human umbilical vein endothelial cells (HUVEC) after 72 hrs after seeding on gels of Ac-LD 6 (L) in DMEM medium, grown at optimum conditions.
  • HPRTC human primaiy renal tubule cells
  • Ac-LIVAGD Ac-LD 6
  • Fig. 15B shows microscopy images of human primary renal tubule cells (HPRTC) after 72 hrs after seeding
  • FIG. 15D shows microscopy im- ages of human umbilical vein endothelial cells (HUVEC) after 72 hrs after seeding on tissue culture plastic, grown at optimum conditions.
  • Figure 16 is a further illustration on the viability of cells in presence of a hydrogel of the invention.
  • Human fibroblast cells were cultured in the presence (Fig. 16A) and absence (Fig. 16B) of Ac-LD 6 (Ac-LIVAGD) (L) (5 mg/ml).
  • FITC Fluorescein isothiocyanate
  • Figure 17 shows the production of composite polymer-peptide hydrogels by incorporating (A) linear and (B) branched polymers with ultrashort peptides during self-assembly.
  • the resulting hydrogels have better mechanical properties (due to cross-linking and in- creased elasticity) and (C) offer opportunities to incorporate bioactive functionalities to modulate the immune and physiological response.
  • Figure 18 shows that ultrashort peptide hydrogels facilitate autolytic debridement of necrotic eschar tissue in bum wounds and enhance wound contracture.
  • Ac-LK Ac- LIVAG -NH 2
  • Ac-IK 6 Ac-ILVAGK-NH 2
  • hydrogel candidates demonstrated earlier onset and completion of autolytic debridement, compared to Mepitel®, as observed by gross examination.
  • B Gross histological analysis of skin samples (day 7) confirmed the completion of autolytic debridement for wounds dressed with peptide hydrogels.
  • Figure 19 shows the histological evaluation of bum wound healing at day 7 and 14.
  • A The tissue damage penetrated mid/deep dermis tissue, for all injuries, as examined at day 7. Necrotic epidermal tissue is observed for the Mepitel®-dressed samples, while com- plete debridement of the eschar epidermal tissues was observed for Ac-IK 6 (Ac- ILVAGK-NH 2 ) and Ac-LK 6 (Ac-LIVAGK-NH 2 ) hydrogel-dressed injuries.
  • Figure 21 shows examples of a subclass of peptides that allow stimuli-responsive gelation, e.g. gelation in the presence of salts at physiological concentrations.
  • the minimum gelation concentration decreases in the presence of salts. Interchanging the amino acids He and Leu at the N-terminus further facilitates gelation.
  • the mechanical strength of this subclass increases in the presence of higher salt concentration such as saline and phosphate-buffered saline, as compared to hydrogels solvated in pure water.
  • Figure 22 demonstrates the ability of peptides with a lysine polar head to bind nucleic acids.
  • the peptide Ac-LK 6 (Ac-LIVAGK-NHj) was extremely efficient in binding and trapping the DNA, possibly through electrostatic interactions. There was minimal release of the DNA, as observed over the course of several days. Ac-LK 6 also effectively protected the DNA against nuclease degradation (B).
  • B When hydrogels encapsulating plasmid DNA were incubated with DNAse, digested DNA fragments were not observed, compared to the naked DNA control. The composite hydrogel fragments did not migrate out of the well during electrophoresis, indicating that the peptide strongly interacts with oligonucleotides, increasing the mass and thereby hindering migration through the agarose gel.
  • Figure 23 shows that peptide hydrogels according to the present invention enhance he- mostasis.
  • Figure 24 illustrates injectable therapy for degenerative disc disease.
  • the mechanical properties of ex vivo extracted porcine nucleus pulposus (A) was determined to be slightly lower than that of our tuneable peptide hydrogels (B).
  • B The mechanical properties of ex vivo extracted porcine nucleus pulposus
  • C Using a rabbit model of degen- erative disc disease (C), 2 different treatments using peptide hydrogels encapsulating MRI contrast agent or cells were evaluated.
  • D The hydrogel persists and can still be detected in the nucleus pulposis space using MRI after 2 months, and upon dissection of the intervertebral discs, more matrix-like material was detected in the treated discs compared to untreated discs.
  • Figure 25 shows the use of peptide hydrogels as dermal and adipose fillers.
  • Peptide hydrogels were subcutaneous implanted in C57BL/6 mice. After 2 months the animals were sacrificed and the implant site was collected for histological analysis, hi all the animals, the inflammatory reaction to the implants was minimal to mild, as evident from (C)the mild foreign body type histiocytic reaction around the hydrogel implant (amorphous eosinophilic material beneath the skeletal muscle layer).
  • Figure 26 shows that ultrashort peptide hydrogels enhance wound contracture
  • (b) Quantitative evaluation of wound healing using digital planimetry revealed that the peptide hydrogels stimulated re-epithelialization and reduced granulation, compared to Mepitel. The error bars denote the standard error of the mean (n 6).
  • Figure 27 gives an overview on the profiles of various cytokines and growth factors dur- ing wound healing.
  • the complex interplay of cytokines and growth factors orchestrate the migration and proliferation of different cells to mediate the overlapping processes of inflammation, granulation, re-epithelialization, matrix formation and remodeling. Since various assays have been completed to demonstrate that our peptides are non-mutagenic, non-immunogenic and non-allergic, differences in the cytokine profile can be associated with the healing process.
  • Multiplex enzyme-linked immunosorbent assay (multiplex ELISA) was carried out on a panel of ten cytokines and growth factors for homogenized skin samples extracted at days 7 and 14.
  • the expression levels were mostly detectable, though generally low after normalizing against the total sample protein content.
  • the expression of pro-inflammatory, anti-healing interleukin-2 (IL-2) was below the detection limit for all the samples tested, including healthy skin.
  • the low expression of pro-inflammatory cytokines was partially attributed to the declining granulation and increasing re- epithelialization (particularly for wounds treated with Ac-LIVAGK-NH 2 hydrogels).
  • the p-values are obtained following AN OVA analysis of the average cytokine expression for the different treatment groups. The statistically significant (p ⁇ 0.05) variations are in bold, and subsequently subjected to post-hoc comparisons.
  • Figure 28 shows the differential expression of pro-inflammatory cytokines by bum wound injuries dressed with Mepitel and peptide hydrogels after 14 days.
  • the cytokine concentration was normalized against the total protein content. In general, the expression levels are low.
  • the groups which are statistically significant (p ⁇ 0.05) are marked with the symbol (*).
  • the line indicates the mean value while the error bars refer to standard eiTor.
  • One-way ANOVA was conducted comparing the normalized mean of different cytokines for the three treatment groups. There were no statistically significant differ- ences in normalized cytokine expression levels on day 7 (Table 1). By day 14, the cytokine expression of wounds treated with Ac-LIVAGK-NH 2 and Mepitel were comparable.
  • IL-4 interleukin-4
  • IL-6 inter- leukin-6
  • TNF-a tumour necrosis factor-alpha
  • GM-CSF granulocyte-macrophage col- ony-stimulating factor
  • IL-6 is another integral cytokine which indirectly induces leukocyte infiltration, matrix re-modeling, angiogenesis and epithelialization.
  • GM-CSF promotes re-epithelialization directly by increasing keratinocyte proliferation and indirectly by up-regulating IL-6. Since wounds treated with Ac-LIVAGK-NH 2 have almost completely re-epithelialized, the expression of these pro-inflammatory cytokines have been down-regulated; whereas for Ac-ILVAGK-NH 2 , the higher levels of IL-1 a, IL-6 and T F-O! mediate the ongoing re-epithelialization.
  • An exemplary embodiment of the invention provides a novel class of hydrogel-fomiing peptides/peptoids derived from inter alia natural amino acids. These peptides/peptoids are small amphophilic peptides with a hydrophobic portion of aliphatic amino acids and one or two polar amino acids. The peptides/peptoids (typically 3-7-mers) are typically in the L- or D-form and can self assemble into supramolecular fibers which are organized into mesh-like structures.
  • the hydrogels are generally characterized by a remarkable rigidity and are biocompatible and non-toxic. Depending on the peptide/peptoid sequence these hydro gels can show stimuli-responsive, thermoresponsive and/or thixotropic character.
  • the thickness and length of the fibers as well as the mechanical properties of the resulting hydrogel scaffold can be controlled.
  • the rigid hydrogels can be used for cultivation of a variety of primary human cells, providing peptide scaffolds that can be useful in the repair and replacement of various tissues. Also disclosed is the procedure of preparing these hydrogels. Disclosed is further the use of respective hydrogels in applications such as cell culture, tissue engineering, plastic surgery, drug and vaccine delivery, oral applications, cosmetics, packaging and the like as well as for technical applications, as for example for use in electronic devices which may include solar or fuel cells.
  • An exemplary of the present invention provides an amphiphilic peptide and/or peptoid capable of forming a hydrogel, i.e. a polymer network in which water is the dispersion medium.
  • the amphiphilic peptide and/or peptoid includes one or more linear amphiphilic sequences, each having a polar and a non-polar portion.
  • linear amphiphilic sequences each having a polar and a non-polar portion.
  • an amphiphilic peptide and/or peptoid disclosed herein includes several linear amphipliilic sequences, each of them differing from any other of the linear amphiphilic sequences.
  • an amphiphilic peptide and/or peptoid disclosed herein includes several identical linear am- phiphilic sequences.
  • an amphiphilic peptide and/or peptoid disclosed herein includes a plurality of linear amphiphilic sequences, each linear amphiphilic sequence being identical to each other linear amphiphilic sequence.
  • a peptide and/or peptoid according to an exemplary embodiment of the invention in- eludes o amphiphilic linear sequences.
  • the symbol o represents an integer selected in the range from 1 to about 25, such as from 1 to about 20, from 1 to about 18, from 1 to about 15, from 1 to about 12, from 1 to about 10, from 1 to about 8, from 1 to about 6, from 1 to about 5 from 1 to about 4 or from 1 to about 3.
  • these am- phiphilic linear sequences are linked in a consecutive manner, thereby defining a linear portion of the peptide and/or peptoid.
  • the peptide and/or peptoid has a backbone with one or more branches. In such an embodiment these amphiphilic linear sequences may be included on different branches.
  • each of the o amphiphilic linear sequences is independently selected.
  • a respective amphiphilic linear sequence has a length of n aliphatic amino acids.
  • n represents an integer selected in the range from 3 to about 18, such as from 3 to about 15, from 3 to about 14, from 3 to about 13, from 3 to about 12, from 3 to about 11, from 3 to about 10, from 3 to about 9, from 3 to about 8 or from 3 to about 7, such as 3, 4, 5, 6, 7, 8, 9 or 10 aliphatic amino acids.
  • an amphiphilic linear sequence of a peptide and/or peptoid described herein is chiral, rendering the entire amphiphilic peptide and/or peptoid chiral.
  • a corresponding linear peptide and/or peptoid i.e. an embodiment that consists of a single respective linear sequence, is accordingly a chiral peptide or peptoid.
  • a respective amphiphilic linear sequence may include any linear non-aromatic amino acid.
  • amino acid refers to an alpha-amino carboxylic acid, i.e. a carboxylic acid with an amino group in the a-position.
  • the respective amino group may be an -NH 2 group or an -NHR 1 group.
  • the moiety R 1 may be any aliphatic group, whether alkyl, alkenyl or alkynyl, with a main chain that includes 1 to 5, to 10, to 15 or to 20 carbon atoms.
  • alkenyl radicals are straight-chain or branched hydrocarbon radicals which contain one or more double bonds.
  • Alkenyl radicals generally contain about two to about twenty carbon atoms and one or more, for instance two, double bonds, such as about two to about ten carbon atoms, and one double bond.
  • Alkynyl radicals normally contain about two to about twenty carbon atoms and one or more, for example two, triple bonds, preferably such as two to ten carbon atoms, and one triple bond.
  • alkynyl radicals are straight-chain or branched hydrocarbon radicals which contain one or more triple bonds.
  • alkyl groups are methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, the n isomers of these radicals, isopropyl, isobutyl, isopentyl, sec-butyl, tert-butyl, neopentyl, 3,3 dimethylbutyl.
  • eptoid as generally used herein is meant to refer to a molecule that mimics a peptide, i.e. "peptidomimetic". Hence, these terms are used interchangeably herein.
  • a "peptidomimetic" or “peptoid” mimics the behaviour and/or structure of a peptide, in that it, just like a peptide, may have the capability to form hydrogen bonds and thus the capability to adopt a secondary structure in the same or similar manner to a peptide.
  • the "peptoid” or “peptidomimetic” may adopt a secondary structure that is typical of a peptide, such as alpha-helix, beta-sheet, random coil and/or combinations thereof.
  • a peptoid may be an oligo(N-alkyl) glycine that, similar to the side chain connected to the a carbon atom (see below) of a peptide, at the amide nitrogen carries a moiety that is in the present invention an aliphatic moiety. Accordingly, in embodiments where an -NHR 1 group (supra) is included in the amino acid and the a carbon atom is included in a -CH 2 - group, the reaction product of coupling a plurality of such amino acids may be called a peptoid.
  • a peptoid can also be taken to differ from a peptide in that it carries its side chain at the amide nitrogen rather than at the a carbon atom.
  • Peptoids are typically resistant to proteases and other modifying enzymes and can have a much higher cell permeability than peptides (see e.g. Kwon, Y.-U., and Kodadek, T., J. Am. Chem. Soc. (2007) 129, 1508-1509).
  • amino acid includes compounds in which the carboxylic acid group is shield- ed by a protecting group in the form of an ester (including an ortho ester), a silyl ester, an amide, a hydrazide, an oxazole, an 1,3-oxazoline or a 5-oxo-l,3,-oxazolidine.
  • amino acid also includes compounds in which an amino group of the form -NH 2 or - NHR 1 (supra) is shielded by a protecting group.
  • Suitable amino protecting groups include, but are not limited to, a carbamate, an amide, a sulfonamide, an imine, an imide, histidine, a N-2,5,-dimethylpyrrole, an N-l,l,4,4-tetramethyldisilylazacyclopentane ad- duct, art N-l,l,3,3-tetramethyl-l,3-disilisoindoline, an N-diphenylsilyldietbylene, an 1,3,5-dioxazine, a N-[2-(trimethylsilyl)ethoxy]methyIamine, a N-(5,5-dimethyl-3-oxo-l- cyclohexenyl)amine, a N-4,4,4-trifluoro-3-oxo-l-butenylamine, a N-9- borabicyclononane and a nitroamine.
  • a protecting group may also be present that shields both the amino and the carboxylic group such as e.g. in the form of a 2,2-dimethyl-4- alkyl-2-sila-5-oxo-l,3-oxazolidine.
  • the alpha carbon atom of the amino acid typically further carries a hydrogen atom.
  • the so called “side chain” attached to the alpha carbon atom, which is in fact the continuing main chain of the carboxylic acid, is an aliphatic moiety that may be linear or branched.
  • side chain refers to the presence of the amino acid in a peptide (supra), where a backbone is foraied by coupling a plurality of amino acids.
  • An aliphatic moiety bonded to the a carbon atom of an amino acid included in such a peptide then defines a side chain relative to the backbone.
  • an aliphatic moiety bonded to the amino group of the amino acid which likewise defines a side chain relative to the backbone of a peptoid.
  • aliphatic means, unless otherwise stated, a straight or branched hydrocarbon chain, which may be saturated or mono- or poly-unsaturated and include heteroatoms.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen.
  • An unsaturated aliphatic group contains one or more double and/or triple bonds (alkenyl or alkynyl moieties).
  • the branches of the hydrocarbon chain may include linear chains as well as non-aromatic cyclic elements.
  • the hydrocarbon chain which may, unless otherwise stated, be of any length, and contain any number of branches.
  • the hydrocarbon (main) chain includes 1 to 5, to 10, to 15 or to 20 carbon atoms.
  • alkenyl radicals are straight-chain or branched hydrocarbon radicals which contain one or more double bonds.
  • Alkenyl radicals generally contain about two to about twenty carbon atoms and one or more, for instance two, double bonds, such as about two to about ten carbon atoms, and one double bond.
  • Alkynyl radicals normally contain about two to about twenty carbon atoms and one or more, for example two, triple bonds, preferably such as two to ten carbon atoms, and one triple bond.
  • alkynyl radicals are straight-chain or branched hydrocarbon radicals which contain one or more triple bonds.
  • alkyl groups are methyl, ethyl, propyl, butyl, pentyl, liexyl, heptyl, octyl, nonyl, decyl, the n isomers of these radicals, isopropyl, isobutyl, isopentyl, sec- butyl, tert-butyl, neopentyl, 3,3 dimethylbutyl.
  • Both the main chain as well as the branches may furthermore contain heteroatoms as for instance N, O, S, Se or Si or carbon atoms may be replaced by these heteroatoms.
  • An aliphatic moiety may be substituted or unsubstituted with one or more functional groups.
  • Substituents may be any functional group, as for example, but not limited to, amino, amido, azido, carbonyl, carboxyl, keto, cyano, isocyano, dithiane, halogen, hy- droxyl, nitro, organometal, organoboron, seleno, silyl, silano, sulfonyl, thio, thiocyano, trifluoromethyl sulfonyl, p-toluenesulfonyl, broraobenzenesulfonyl, nitrobenzenesul- fonyl, and methanesulfonyl.
  • the side chain of an amino acid in a pep- tide/peptoid described herein may be of a length of 0 to about 5, to about 10, to about 15 or to about 20 carbon atoms. It may be branched and include unsaturated carbon-carbon bonds.
  • one or more natural amino acids are included in the peptide or peptoid. Such a natural amino acid may be one of the 20 building blocks of naturally occurring proteins.
  • a peptide or peptoid including a peptide/peptoid disclosed herein individual amino acids are covalently coupled via amide bonds between a carboxylic group of a first and an amino group of a second amino acid.
  • a peptide and/or peptoid disclosed herein is non-repetitive, such that two amino acids coupled to each other are always different from one another.
  • amphiphilic refers to a compound that is soluble in both polar and non-polar fluids. It also encompasses multiphase compounds.
  • the amphiphilic properties of the peptide and/or peptoid are due to the presence of both polar and non-polar moieties with- in the same peptide and/or peptoid.
  • the peptide and/or peptoid may be of surfactant nature.
  • the polar properties of a peptide and/or peptoid according to an embodiment of the invention are based on a polar moiety. Two such moiety are a - COOH side group, in particular in the form of a charged COO " group and an amino group.
  • a further such moiety is a C-terminal -COOH group if it is present in free, unpn tected form.
  • a surfactant molecule includes a polar, typically hydrophilic, head group attached to a non-polar, typically hydrocarbon, moiety.
  • Non-polar moieties of a peptide or peptoid include a hydrocarbon chain that does not carry a functional group.
  • An amphiphilic linear sequence included in a peptide and/or peptoid of an embodiment of the invention thus includes a polar moiety and a non-polar moiety.
  • the polar moiety includes an aliphatic amino acid that carries a polar group such as a hydroxyl group, a thiol group, a seleno group, an amino group, an amide group, an ether group, a thioether group or a seleno ether group.
  • the polar moiety may include an amino acid that carries a functional polar group with a proton such as hydroxyl, thiol, selenol, amine or amide.
  • the polar moiety may also include the C-terminus or the N-terminus of the peptide and/or peptoid.
  • the C-terminus or the N-terminus may in such a case be present in the form of the free carboxyl or amino group, respectively, i.e. free of a protecting group.
  • the polar moiety of a linear amphophilic sequence of an amphiphihc peptide and/or peptoid of an embodiment of the invention is defined by a single amino acid, by two consecutive amino acids or by three consecutive amino acids that is/are coupled to the non-polar moiety of the peptide/peptoid.
  • the polar moiety of the peptide/peptoid consists of two amino acids that are covalently coupled via an amide bond, both amino acids carrying a polar peptide/peptoid side chain.
  • One of these two amino acids may be a terminal amino aid of the peptide/peptoid, defining its N- or C-terminus.
  • the amphiphihc peptide/peptoid has a single amino acid with a polar side chain with the residual portion of the peptide/peptoid defining the non-polar moiety. In some embodiments the amphiphihc peptide/peptoid has two amino acids with a polar side chain while the residual portion of the peptide/peptoid defines the non-polar moiety.
  • a respective polar side chain may serve 4-methyl-4-thio-pentyl, 6-ethoxycarbonyl-4,5-dimethyl-hexyl and 6-hydroxy-4-(l- hydroxyethyl)-hexyl groups.
  • the numbering of corresponding peptide/peptoid side chains is started with "1" at the carbon atom that is covalently bonded to the a-carbon atom of the amino acid or to the amino group of the amino acid, respectively.
  • Amino acids included in the polar moiety may be or include, but are not limited to, aspartic acid, asparagine, glutamic acid, 4-fluoro-glutamic acid, 2-aminoadipic acid, ⁇ - carboxy-glutamic acid, 4-tert-butyl aspartic acid, glutamine, 5-N-ethyl-glutamine (theanine), citrulline, thio-citrulline, cysteine, homocysteine, methionine, ethionine, selenomethionine, telluromethionine, threonine, allo-threonine, serine, homoserine, arginine, homoarginine, ornithine, lysine, 5-hydroxy-lysine and N(6)-carboxymethyllysine. Any such amino acid may be present in the L-or D-form.
  • the ainphiphilic linear sequence of the amphiphihc peptide/peptoid of an embodiment of the invention can be defined as having n amino acids.
  • the non-polar moiety may then be taken to have n-1 amino acids.
  • the polar moiety consists of exactly one amino acid, such amino acid being selected from any amino acids of the foregoing paragraph.
  • the non-polar moiety may then be taken to have n-2 amino acids. In this case the polar moiety consists of exactly two amino acids.
  • the non-polar moiety may then be taken to have n-3 amino acids.
  • the polar moiety consists of exactly three amino acids.
  • the polar moiety may have a se- quence selected from Asn-Asn, Asp-Asp, Glu-Glu, Gln-Gln, Asil-Gln, Gln-Asn, Asp- Gin, Gin-Asp, Asn-Glu, Glu-Asn, Asp-Glu, Glu-Asp, Gln-Glu, Glu-Gln, Asp-Asn, Asn- Asp, Thr-Thr, Ser-Ser, Thr-Ser, Ser-Thr, Asp-Ser, Ser-Asp, Ser-Asn, Asn-Ser, Gln-Ser, Ser-Gln, Glu-Ser, Ser-Asp, Ser-Asn, Asn-Ser, Gln-Ser, Ser-Gln, Glu-Ser, Ser
  • the polar moiety may have a sequence selected from Asn- Asn-Asn, Asn-Asn-Asp, Asn-Asp- Asn, Asp-Asn-Asn, Asp-Asp-Asn, Asp-Asn-Asp, Asp-Asp-Asp, Asn-Asn-Glu, Asn-Asn- Gln, Asn-Glu-Asn, Asn-Gln-Asn, Glu-Glu-Glu, Gln-Gln-Gln, Asn-Gln-Gln, Asn-Glu- Gln, Asp- Asn-Glu, Gin- Asn- Asn, Gin- Asn- Asn, Glu- Asp-Gin, Asp-Gin- Asp, Asn-Glu- Asp, Glu-Asn-Gln, Asp-Glu-Gln, Asp-Glu-Gln, Asn-Glu-Gln,
  • the amphiphilic linear sequence of the peptide/peptoid has a net charge at physiological pH.
  • physiological pH is known to those in the art to refer to the pH value of blood, which has typically a pH value of about 7.4.
  • the respective terminus may provide the corresponding net charge.
  • the polar moiety of the amphiphilic linear sequence includes one or more amino acids that have a side chain with a functional group that is charged at physiological pH.
  • Illustrative examples of a respective functional group include an amino, a nitro-, a guanidino, a esteryl, a sulfonyl or a carboxyl group.
  • the net charge of the amphiphilic linear sequence is, as a positive or as a negative charge, equal to or smaller than the number of amino acids included in the polar moiety thereof.
  • the net charge of the amphiphilic linear sequence is one of -3, -2 or - 1.
  • the net charge of the amphiphilic linear sequence is +1, +2 or +3.
  • the respective polar side chain of an amino acid of the polar moiety, coupled to the a- carbon atom of the amino acid (supra) and/or to the amino group thereof, may typically be defined by a main chain that includes 1 to about 20, including 1 to about 15, 1 to about 10 or 1 to about 5 carbon atoms.
  • side chain is used relative to the backbone of the peptide and/or peptoid.
  • This peptide and/or peptoid side chain may be branched and thus be defined by a main chain and branches. Both the main chain and branches, if present, of the peptide and/or peptoid side chain may include one or more double or triple bonds (supra).
  • side chains include, but are not limited to, methyl, ethyl, propyl, isopropyl, propenyl, propinyl, butyl, butenyl, sec-butyl, teri-butyl, isobutyl, pentyl, neopentyl, isopentyl, pentenyl, hexyl, 3,3 dimethyl- butyl, heptyl, octyl, nonyl or decyl groups.
  • the functional polar group is bonded to this the peptide and/or peptoid side chain.
  • the amphiphilic linear sequence of the peptide/peptoid has a hydrophobic tail of aliphatic amino acids and at least one polar, including a charged, amino acid head group.
  • the non-polar moiety includes an amino acid, generally at least two amino acids, with a hydrocarbon chain that does not carry a functional group.
  • the respective side chain, coupled to the a-carbon atom of the amino acid (supra) may have a main chain that includes 0 to about 20 or 1 to about 20, including 0 to about 15, 1 to about 15, 0 to about 10, 1 to about 10, 1 to about 5 or 0 to about 5 carbon atoms.
  • the non-polar moiety may thus include an amino acid without side chain, i.e. glycine.
  • the peptide and/or peptoid side chain may be branched (supra) and include one or more double or triple bonds (supra).
  • Examples of peptide and/or peptoid side chains include, but are not limited to, methyl, ethyl, propyl, isopropyl, propenyl, propinyl, butyl, butenyl, see-butyl, terf-butyl, isobutyl, pentyl, neopentyl, isopentyl, pentenyl, hexyl, 3,3 dimethylbutyl, heptyl, octyl, nonyl or decyl groups.
  • the non-polar moiety may include an amino acid of alanine, valine, leucine, isoleucine, norleucine, norvaline, 2-(methylamino)- isobutyric acid, 2-amino-5-hexynoic acid.
  • Such an amino acid may be present in any de- sired configuration.
  • Bonded to the non-polar moiety may also be the C-terminus or the N-terminus of the pep tide/pep toid. Typically the C-terminus or the N-tenninus is in such a case shielded by a protecting group (supra).
  • the non-polar moiety includes a sequence of amino acids that is arranged in decreasing or increasing size.
  • a portion of the amino acids of the non- polar moiety may be arranged in a general sequence of decreasing or increasing size. Relative to the direction from N- to C-temiinus or from C- to N-terminus this general sequence can thus be taken to be of decreasing size.
  • general sequence of decreasing or increasing size is meant that embodiments are included in which adjacent amino acids are of about the same size as long as there is a general decrease or increase in size.
  • the size of adjacent amino acids of the non-polar moiety is accordingly identical or smaller in the direction of the general sequence of decreasing size.
  • the general sequence of decreasing or increasing size is a non-repetitive sequence.
  • the respective non-polar portion may be a sequence of three amino acids.
  • the first amino acid may have an n-nonyl side chain.
  • the second amino acid may have a 3-ethyl-2-methyl-pentyl side chain.
  • the third amino acid may have a tert-butyl side chain.
  • the non-polar moiety may be a .sequence of nine amino acids.
  • the first amino acid may have a 4-propyl -nonyl side chain.
  • the second amino acid may have an n-dodecyl side chain.
  • the third amino acid may have a 6,6- diethyl-3-octenyl side chain.
  • An n-dodecyl side chain and a 6,6-diethyl-3-octenyl side chain both have 12 carbon atoms and thus again have a comparable size, Nevertheless, the 6,6-diethyl-3-octenyl group includes an unsaturated carbon-carbon bond and is thus of slightly smaller size than the dodecyl group.
  • the fourth amino acid may have a 2- methyl -nonyl side chain.
  • the fifth amino acid may have a 3-propyl-hexyl side chain.
  • the sixth amino acid may have an n-hexyl side chain.
  • the seventh amino acid may have a 2- butynyl side chain.
  • the 8th amino acid may have an isopropyl side chain.
  • the ninth amino acid may have a methyl side chain.
  • amino acids of the non-polar moiety arranged in a general sequence of decreasing (or increasing) size only contains naturally occurring amino acids (whether in the D- or the L-form), it may for example have a length of five amino acids, such as the sequence leucine-isoleucine-valine-alanine-glycine or isoleucine-leuciner valine-alanine-glycine,
  • a general sequence of decreasing size of only natural amino acids may also have a length of four amino acids.
  • Illustrative examples include the sequences iso- leucine-valine-alanine, leucine-valine-alanine, isoleucine-valine-glycine, leucine-valine- glycine, leucine-alanine-glycine, isoleucine-alanine-glycine or isoleucine-leucine- alanine.
  • a general sequence of decreasing size of only natural amino acids may also have a length of two amino acids.
  • the next amino acid may be polar in that it carries a peptide/peptoid side chain with a polar functional group.
  • This amino acid may be positioned between the general sequence of decreasing (or increasing) size and the polar amino acid, the polar amino acid may be positioned between this additional non-polar amino acid and the gen- eral sequence of decreasing (or increasing) size or the general sequence of decreasing (or increasing) size may be positioned between the polar amino acid and this additional non- polar amino acid.
  • the general sequence of decreasing (or increasing) size is positioned between the polar amino acid and this additional non-polar amino acid.
  • the additional non-polar amino acid may for example define the N-tenninus of the pep- tide/peptoid, which may be shielded by a protecting group such as an amide, e.g. a propionic acyl or an acetyl group.
  • the general sequence of decreasing (or increasing) size may define the non-polar portion of the pep- tide/peptoid.
  • the polar amino acid may define the C-terminus of the peptide/peptoid.
  • the general sequence of decreasing (or increasing) size is thus flanked by the polar amino acid on one side and by the additional non-polar amino acid on the other side.
  • the general sequence of decreasing (or increasing) size has a length of n-m-1 amino acids
  • the remaining non-polar amino acid of the non-polar moiety of n-m amino acids is one of alanine and glycine.
  • the polar moiety of the amphiphilic linear sequence may in some embodiments be defined by two or three consecutive amino acids.
  • the polar moiety includes m aliphatic amino acids.
  • Each of the m aliphatic amino acids is independently se- lected and carries an independently selected polar group.
  • the symbol m represents an integer selected from 1, 2 and 3.
  • the at least essentially non-polar moiety accordingly has a number of n-m, i.e. n-1, n-2 or «-5amino acids.
  • n is equal to or larger than m + 2.
  • m may thus represent a number of n-2 or smaller.
  • this non-polar moiety may thus have a length of n-2 or n-3 amino acids.
  • this additional non-polar side chain may be included in an amino acid that is directly bonded to an amino acid of the general sequence of decreasing (or increasing) size.
  • the non-polar moiety may thus be defined by the non-polar moiety of decreasing (or increasing) size and the respective further amino acid with a non-polar side chain.
  • the non-polar moiety may thus have a length of n-2 amino acids, of which the non-polar moiety of decreasing (or increasing) size has a length of n-3 amino acids.
  • the general sequence of decreasing (or increasing) size may be positioned between the two polar amino acids and this additional non-polar amino acid, or the additional non- polar amino acid may be positioned between the general sequence of decreasing (or increasing) size and the two polar amino acids.
  • the general sequence of decreasing (or increasing) size is positioned between the two polar amino acids and this additional non-polar amino acid.
  • one of the two polar amino acids may define the C-terminus of the peptide/peptoid.
  • the general sequence of decreasing (or increasing) size may thus be flanked by the two consecutive polar amino acids on one side and by the additional non-polar amino acid on the other side.
  • the two consecutive polar amino acids may also be positioned between the general sequence of decreasing (or increasing) size and the additional non-polar amino acid, in which case the non-polar moiety has a first portion with a length of 71-3 amino acids and a further portion of one amino acid.
  • the inventors have observed the formation of fibers based on helical structures.
  • the fibers formed of amphiphilic linear sequences of amphiphilic peptides and/or peptoids of an embodiment of the invention typically show high mechanical strength, which renders them particularly useful in tissue regeneration applications, for instance the replacement of damaged tissue.
  • the amphophilic linear sequence of the peptide/peptoid which may represent the entire amphiphilic peptide/peptoid (supra) has been found to show remarkable stability at physiological conditions, even at elevated temperatures. It is in some embodiments stable in aqueous solution at physiological conditions at ambient temperature for a period of time in the range from 1 day to 1 month or more. It may in some embodiments be stable in aqueous solution at physiological conditions at 90 °C for at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours or at least 5 hours.
  • a hydrogel according to an embodiment of the present invention is typically character- ized by a remarkable rigidity and are generally biocompatible and non-toxic. Depending on the selected peptide/peptoid sequence these hydrogels can show thermoresponsive or fhixotropic character. Reliant on the peptide/peptoid assembling conditions the fibers differ in thickness and length. Generally rigid hydrogels are obtained that are well suited for cultivation of a variety of primary human cells, providing peptide/peptoid scaffolds that can be useful in the repair and replacement of various tissues. Disclosed is also a process of preparing these hydrogels.
  • hydrogels in applications such as cell culture, tissue engineering, orthopaedic surgery, aesthetic/plastic sur- gery, drug and vaccine delivery, oral applications, cosmetics, packaging and the like is described, as well as for technical applications, as for example for use in electronic devices which might include solar or fuel cells.
  • a hydrogel of an embodiment of the invention shows high stability at physiological conditions, even at elevated temperatures.
  • such a hydrogel is stable in aqueous solution at ambient temperature for a period of at least 7 days, at least 14 days, at least a month or more, such as at least 1 to about 6 months.
  • a hydrogel disclosed herein is coupled to a molecule or a particle, including a quantum dot, with characteristic spectral or fluorometric properties, such as a marker, including a fluorescent dye and a MRI contrast agent.
  • a respective molecule may for instance allow monitoring the fate, position and/or the integrity of the hydrogel.
  • a hydrogel disclosed herein is coupled to a molecule with binding affinity for a selected target molecule, such as a microorganism, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, a peptide, an oligosaccharide, a polysaccharide, an inorganic molecule, a synthetic polymer, a small organic molecule, a drug or a cell.
  • a target molecule such as a microorganism, a virus particle, a peptide, a peptoid, a protein, a nucleic acid, a peptide, an oligosaccharide, a polysaccharide, an inorganic molecule, a synthetic polymer, a small organic molecule, a drug or a cell.
  • Nucleic acids include for instance DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogues of the DNA or RNA generated using nucleotide analogues or using nucleic acid chemistry, locked nucleic acid molecules (LNA), and protein nucleic acids molecules (PNA).
  • DNA or RNA may be of genomic or synthetic origin and may be single or double stranded. In the present method of an embodiment of the invention typically, but not necessarily, an RNA or a DNA molecule will be used.
  • nucleic acid can be e.g. mRNA, cRNA, synthetic RNA, genomic DNA, cDNA synthetic DNA, a copolymer of DNA and RNA, oligonucleotides, etc.
  • a respec- tive nucleic acid may furthenriore contain non-natural nucleotide analogues and/or be linked to an affinity tag or a label.
  • the nucleic acid molecule may be isolated, enriched, or purified.
  • the nucleic acid molecule may for instance be isolated from a natural source by cDNA cloning or by subtractive hybridization.
  • the natural source may be mammalian, such as human, blood, semen, or tissue.
  • the nucleic acid may also be synthesized, e.g. by the triester method or by using an automated DNA synthesizer.
  • a nucleotide analogue is a nucleotide containing a modification at for instance the base, sugar, or phosphate moieties. Modifications at the base moiety include natural and synthetic modifications of A, C, G, and T/U, different purine or pyrimidine bases, such as uracil-5-yl, hy- poxanthin-9-yl, and 2-aminoadenin-9-yl, as well as non-purine or non-pyrimidine nucleo- tide bases. Other nucleotide analogues serve as universal bases.
  • Universal bases include 3-nitropyrrole and 5-nitroindole. Universal bases are able to form a base pair with any other base. Base modifications often can be combined with for example a sugar modification, such as for instance 2'-0-methoxyethyl, e.g. to achieve unique properties such as increased duplex stability.
  • a peptide may be of synthetic origin or isolated from a natural source by methods well- known in the art.
  • the natural source may be mammalian, such as human, blood, semen, or tissue.
  • a peptide, including a polypeptide may for instance be synthesized using an automated polypeptide synthesizer.
  • Illustrative examples of polypeptides are an antibody, a fragment thereof and a proteinaceous binding molecule with antibody-like functions.
  • Examples of (recombinant) antibody fragments are Fab fragments, Fv fragments, single- chain Fv fragments (scFv), diabodies, triabodies (Iliades, P., et al., FEBS Lett (1997) 409, 437-441), decabodies (Stone, E., et al., Journal of Immunological Methods (2007) 318, 88-94) and other domain antibodies (Holt, L.J., et al., Trends Biotechnol. (2003), 21, 1 1, 484-490).
  • a proteinaceous binding molecule with antibody-like functions is a mutein based on a polypeptide of the lipocalin family (WO 03/029462, Beste et al., Proc. Natl. Acad,..Sci. U.S.A. (1999) 96, 1898-1903).
  • Lipocalins such as the bilin binding protein, the human neutrophil gelatinase-associated lipocalin, human Apolipoprotein D or glycodelin, posses natural ligand-binding sites that can be modified so that they bind to selected small protein regions known as haptens.
  • glubodies see e.g.
  • Adnectins derived from a domain of hu- man fibronectin, contain three loops that can be engineered for immunoglobulin-like binding to targets (Gill, D.S. & Damle, N.K., Current Opinion in Biotechnology (2006) 17, 653-658).
  • Tetranectins derived from the respective human homotrimeric protein, likewise contain loop regions in a C-type lectin domain that can be engineered for desired binding (ibid.).
  • a modifying agent may be used that further increas- es the affinity of the respective moiety for any or a certain form, class etc. of target matter.
  • An example of a nucleic acid molecule with antibody-like functions is an aptamer.
  • An aptamer folds into a defined three-dimensional motif and shows high affinity for a given target structure.
  • an aptamer with affinity to a certain target can accordingly be formed and immobilized on a hollow particle of an embodiment of the invention.
  • a linking moiety such as an affinity tag may be used to immobilise the respective molecule.
  • a linking moiety may be a molecule, e.g. a hydrocarbon-based (including polymeric) molecule that includes nitrogen-, phosphorus-, sulphur-, carben-, halogen- or pseudohalogen groups, or a portion thereof.
  • the peptide/peptoid included in the hydrogel may include functional groups, for instance on a side chain of the peptide/peptoid, that allow for the covalent attachment of a biomolecule, for example a molecule such as a protein, a nucleic acid molecule, a polysaccharide or any combination thereof.
  • a respective functional group may be provided in shielded form, protected by a protecting group that can be released under desired conditions.
  • Examples of a respective functional group include, but are not limited to, an amino group, an aldehyde group, a thiol group, a carboxy group, an ester, an anhydride, a sulphonate, a sulphonate ester, an imido ester, a silyl halide, an epoxide, an aziridine, a phosphoramidite and a diazoalkane.
  • an affinity tag examples include, but are not limited to, biotin, dinitrophenol or di- goxigenin, oligohistidine, polyhistidine, an immunoglobulin domain, maltose-binding protein, glutathione-S-transferase (GST), calmodulin binding peptide (CBP), FLAG'- peptide, the T7 epitope (Ala-Ser-Met-Tlu--Gly-Gly-Gln-Glii-Met-Gly), maltose binding protein (MBP), the HSV epitope of the sequence Gln-Pro-Glu-Leu-Ala-Pro-Glu-Asp- Pro-Glu-Asp of herpes simplex virus glycoprotein D, the hemagglutinin (HA) epitope of the sequence Tyr-Pro-Tyr-Asp-Val-Pro-Asp-Tyr-Ala, the "myc" epitope of the transcription factor c-myc of the sequence
  • Such an oligonucleotide tag may for instance be used to hybridise to an immobilised oligonucleotide with a complementary sequence.
  • a further example of a linking moiety is an antibody, a fragment thereof or a proteinaceous binding molecule with antibody-like functions (see also above).
  • a further example of linking moiety is a cucurbituril or a moiety capable of forming a complex with a cucurbituril.
  • a cucurbituril is a macrocyclic compound that includes gly- coluril units, typically self-assembled from an acid catalyzed condensation reaction of glycoluril and formaldehyde.
  • a cucurbit[n]uril, (CB[n]) that includes n glycoluril units, typically has two portals with polar ureido carbonyl groups. Via these ureido carbonyl groups cucurbiturils can bind ions and molecules of interest.
  • cucurbit[7]uril can form a strong complex with feiTocenemethylammonium or adamantylammonium ions.
  • Either the cucurbit[7]uril or e.g. feiTocenemethylammonium may be attached to a biomolecule, while the remaining binding partner (e.g. ferroceneme- thylammonium or cucurbit[7]uril respectively) can be bound to a selected surface. Contacting the biomolecule with the surface will then lead to an immobilisation of the biomolecule.
  • Functionalised CB[7] units bound to a gold surface via alkanethiolates have for instance been shown to cause an immobilisation of a protein carrying a ferroceneme- thylammonium unit (Hwang, I., et al., J. Am. Chem. Soc. (2007) 129, 4170-4171).
  • a linking moiety include, but are not limited to an oligosaccharide, an oligopeptide, biotin, dinitrophenol, digoxigenin and a metal chelator (cf. also below).
  • a respective metal chelator such as ethylenediamine, eth- ylenediaminetetraacetic acid (EDTA), ethylene glycol tetraacetic acid (EGTA), diethy- lenetriaminepentaacetic acid (DTP A), N,N-bis(carboxymethyl)glycine (also called nihil otriacetic acid, NT A), l,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA), 2,3-dimercapto-l-propanol (dimercaprol), porphine or heme may be used in cases where the target molecule is a metal ion.
  • EDTA ethylenediamine
  • EGTA ethylene glycol tetraacetic acid
  • DTP A diethy- lenetriaminepentaacetic acid
  • N,N-bis(carboxymethyl)glycine also called ni
  • EDTA forms a complex with most monovalent, divalent, trivalent and tetravalent metal ions, such as e.g. silver (Ag + ), calcium (Ca 2+ ), manganese (Mn 2+ ), copper (Cu 2+ ), iron (Fe 2+ ), cobalt (Co 3+ ) and zirconium (Zr 4+ ), while BAPTA is specific for Ca 2+ .
  • a respective metal chelator in a complex with a respective metal ion or metal ions defines the linking moiety.
  • Such a complex is for example a receptor molecule for a peptide of a defined sequence, which may also be included in a protein.
  • a standard method used in the art is the formation of a complex between an oligohistidine tag and copper (Cu 2+ ), nickel (Ni 2+ ), cobalt (Co 2+ ), or zink (Zn 2+ ) ions, which are presented by means of the chelator nitrilotriacetic acid (NT A).
  • Avidin or streptavidin may for instance be employed to immobilise a biotinylated nucleic acid, or a biotin containing monolayer of gold may be employed (Shumaker-Parry, J.S., et al., Anal. Chem. (2004) 76, 918).
  • the biomolecule may be locally deposited, e.g. by scanning electrochemical microscopy, for instance via pyiTole-oligonucleotide patterns (e.g. Fortin, E., et al., Electroanalysis (2005) 17, 495).
  • light-directed synthesis of a biomolecule in particular of a nucleic acid molecule, including UV-linking or light dependent 5'-deprotection, may be carried out.
  • the molecule that has a binding affinity for a selected target molecule may be immobilised on the nanocrystals by any means.
  • an oligo- or polypeptide, including a respective moiety may be covalently linked to the surface of nanocrys- tals via a thio-ether-bond, for example by using co functionalized thiols.
  • Any suitable molecule that is capable of linking a nanocrystal of an embodiment of the invention to a molecule having a selected binding affinity may be used to immobilise the same on a nanocrystal.
  • a (bifunctional) linking agent such as ethyl-3- dimetliylaminocarbodiimide, N-(3-aminopropyl) 3-mercapto-benzamide, 3-aminopropyl- trimethoxysilane, 3-mercaptopropyl-trimethoxysilane, 3-(trimethoxysilyl) propyl - maleimide, or 3-(trimethoxysilyl) propyl-hydrazide may be used.
  • the surface of the nanocrystals Prior to reaction with the linking agent, the surface of the nanocrystals can be modified, for example by treatment with glacial mercaptoacetic acid, in order to generate free mercaptoacetic groups which can then employed for covalently coupling with an analyte binding partner via linking agents.
  • Embodiments of the present invention also include a hydrogel, which can be taken to be a water-swollen water-insoluble polymeric material.
  • the hydrogel includes, including contains and consists of, a peptide and/or peptoid as defined above. Since a hydrogel maintains a three-dimensional structure, a hydrogel of an embodiment of the invention may be used for a variety of applications. Since the hydrogel has a high water content and includes amino acids, it is typically of excellent biocompatibility.
  • a hydrogel according to an embodiment of the invention is typically formed by self- assembly.
  • the inventors have observed that the peptides/peptoids assemble into fibers that fonn mesh-like structures. Without being bound by theory hydrophobic interaction between non-polar portions of peptides/peptoids of an embodiment of the invention are contemplated to assist such self-assembly process.
  • the method of forming the hydrogel includes dissolving the peptide/peptoid in aqueous solution. Agitation, including mixing such as stirring, and/or sonication may be employed to facilitate dissolving the peptide/peptoid.
  • the aqueous solution with the peptide/peptoid therein is exposed to a temperature below ambient temperature, such as a temperature selected from about 2 °C to about 15 °C.
  • the aqueous solution with the peptide/peptoid therein is exposed to an elevated temperature, i.e. a temperature above ambient temperature. Typically the aqueous solution is allowed to attain the temperature to which it is exposed.
  • the aqueous solution may for example be exposed to a temperature from about 25 °C to about 85 °C or higher, such as from about 25 °C to about 75 °C, from about 25 °C to about 70 °C, from about 30 °C to about 70 °C, from about 35 °C to about 70 °C, from about 25 °C to about 60 °C, from about 30 °C to about 60 °C, from about 25 °C to about 50 °C, from about 30 °C to about 50 °C or from about 40 °C to about 65 °C, such as e.g.
  • the aqueous solution with the peptide/peptoid therein may be maintained at this temperature for a period of about 5 min to about 10 hours or more, such as about 10 min to about 6 hours, about 10 min to about 4 hours, about 10 min to about 2.5 hours, about 5 min to about 2.5 hours, about 10 min to about 1.5 hours or about 10 min to about 1 hour, such as about 15 min, about 20 min, about 25 min, about 30 min, about 35 min or about 40 min.
  • a hydrogel according to an embodiment of the invention may be included in a fuel cell, where it may for example provide a substrate between the anode and the cathode, a liquid electrolyte may be encompassed by the hydrogel.
  • a hydrogel according to an embodiment of the invention may provide a substrate between two electrodes in an elec- trophoresis apparatus.
  • the hydrogel may also be conducting.
  • the hydrogel may also serve in enhancing the efficiency of charge-separated states and/or slowing down charge recombination.
  • the hydrogel may thus be applied in any form photovoltaics, including a solar cell.
  • a hydrogel disclosed herein is a biocompatible, including a pharmaceutically acceptable hydrogel.
  • biocompatible (which also can be referred to as "tissue compatible”), as used herein, is a hydrogel that produces little if any adverse biological response when used in vivo. The term thus generally refers to the inability of a hydrogel to promote a measurably adverse biological response in a cell, including in the body of an animal, including a human.
  • a biocompatible hydrogel can have one or more of the following properties: non-toxic, non-mutagenic, non-allergenic, non-carcinogenic, and/or non-irritating.
  • a biocompatible hydrogel, in the least, can be innocuous and toler- ated by the respective cell and/or body.
  • a biocompatible hydrogel, by itself, may also improve one or more functions in the body.
  • a respective hydrogel may be biodegradable.
  • a biodegradable hydrogel gradually disintegrates or is absorbed in vivo over a period of time, e.g., within months or years. Disintegration may for instance occur via hydrolysis, may be catalysed by an enzyme and may be assisted by conditions to which the hydrogel is exposed in a human or animal body, including a tissue, a blood vessel or a cell thereof. Where a peptide is made up entirely of natural amino acids, a respective peptide can usually be degraded by enzymes of the human/animal body.
  • a hydrogel according to an embodiment of the invention may also serve as a depot for a pharmaceutically active compound such as a drug and/or micro- and nanoparticles.
  • a hydrogel according to an embodiment of the invention may be designed to mimic the natural extracellular matrix of an organism such as the human or animal body.
  • a fiber formed from the peptide/peptoid of an embodiment of the invention, including a respective hydrogel, may serve as a biological scaffold.
  • a hydrogel of an embodiment of the invention may be included in an implant, in a contact lens or may be used in tissue engi- neering.
  • the peptides consist typically of 3-7 amino acids and are able to self-assemble into complex fibrous scaffolds which are seen as hydrogels, when dissolved in water or aqueous solution. These hydrogels can retain water up to 99.9% and possess sufficiently high mechanical strength. Thus, these hydrogels can act as artificial substitutes for a variety of natural tissues without the risk of immunogenicity.
  • the hy- drogels in accordance with the present invention may be used for cultivating suitable primary cells and thus establish an injectable cell-matrix compound in order to implant or re-implant the newly formed cell-matrix in vivo. Therefore, the hydrogels in accordance with the present invention are particularly useful for tissue regeneration or tissue engineering applications.
  • a reference to an "implant” or “implantation” refers to uses and applications of/for surgical or arthroscopic implantation or injection of a hydrogel containing device (or of a peptide solution that gels in situ) into a human or animal, e.g. mammalian, body or limb.
  • Arthroscopic techniques are taken herein as a subset of surgical techniques, and any reference to surgery, surgical, etc., includes arthroscopic techniques, methods and devices.
  • a surgical implant that includes a hydrogel according to an embodiment of the invention may include a peptide and/or peptoid scaffold. This the peptide and/or peptoid scaffold may be defined by the respective hydrogel.
  • a hydrogel of an embodiment of the invention may also be included in a wound cover such as a gauze or sheet or membrane or cream or spray, serving in maintaining the wound in a moist state to promote healing.
  • the hydrogel may be temperature-sensitive. It may for instance have a lower critical solution temperature or a temperature range corresponding to such lower critical solution temperature, beyond which the gel collapses as hydrogen bonds by water molecules are released as water molecules are released from the gel.
  • the disclosed subject matter also provides improved chiral amphiphilic natural-based peptides and/or peptoids that assemble to peptide/peptoid hydrogels with very favorable material properties.
  • the advantage of these peptide/peptoid hydrogels is that they are accepted by a variety of different primary human cells, thus providing peptide scaffolds that can be useful in the repair and replacement of various tissues.
  • the character of the hydrogels can be designed to be more stable and less prone to degradation though still biocompatible.
  • a hydrogel and/ or a peptide/peptoid described herein can be administered to an organism, including a human patient per se, or in pliannaceutical compositions where it may include or be mixed with pharmaceutically active ingredients or suitable earners or ex- cipient(s).
  • Techniques for formulation and administration of respective hydrogels or pep- tides/peptoids resemble or are identical to those of low molecular weight compounds well established in the art. Exemplary routes include, but are not limited to, oral, transdermal, and parenteral delivery.
  • a hydrogel or a peptide/peptoid may be used to fill a capsule or tube, or may be provided in compressed form as a pellet.
  • the peptide/peptoid or the hydrogel may also be used in injectable or sprayable form, for instance as a suspension of a respective peptide/peptoid.
  • a hydrogel of an embodiment of the invention may for instance be applied onto the skin or onto a wound.
  • Further suitable routes of administration may, for example, include depot, oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intravenous, intramedullary injections, as well as intrathe- cal, direct intraventricular, intraperitoneal, intranasal, or intraocular injections. It is noted in this regard that for administering microparticles a surgical procedure is not required.
  • microparticles include a biodegradable polymer there is no need for device removal after release of the anti-cancer agent. Nevertheless the microparticles may be included in or on a scaffold, a coating, a patch, composite material, a gel or a plaster. In some embodiments one may administer a hydrogel and/or a peptide/peptoid in a local rather than systemic manner, for example, via injection.
  • compositions that include a hydrogel and/or a peptide/peptoid of an embodiment of the present invention may be manufactured in a manner that is itself known, e. g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with an embodiment of the present invention thus may be fonnulated in conventional manner using one or more physiologi- cally acceptable earners including excipients and auxiliaries that facilitate processing of the hydrogel and/or peptide/peptoid into preparations that can be used pharmaceutically. Proper fonnulation is dependent upon the route of administration chosen.
  • the peptide/peptoid of an embodiment of the invention may be fonnulated in aqueous solutions, for instance in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be penneated are used in the fonnulation. Such penetrants are generally known in the art.
  • the hydrogel and/or peptide/peptoid can be fonnulated readily by combining them with pharmaceutically acceptable carriers well known in the art.
  • Such earners enable the hydrogel and/or peptide/peptoid, as well as a pharmaceutically active compound, to be fonnulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slur- ries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Phannaceutical preparations for oral use can be obtained by adding a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatine, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbox- ymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Phannaceutical preparations that can be used orally include push-fit capsules made of gelatine, as well as soft, sealed capsules made of gelatine and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the peptides/peptoids may be suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All fonnulations for oral administration should be in dosages suitable for such administration.
  • the compositions may take the fomi of tablets or lozenges fonnulated in conventional manner.
  • the hydrogel and/or peptide/peptoid may be fonnulated for parenteral administration by injection, e.g., by intramuscular injections or bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage fonn, e. g., in ampules or in multi-dose containers, with an added preservative.
  • the respective compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the hydrogel and/or peptide/peptoid may be formulated for other drug delivery systems like implants, or transdermal patches or stents.
  • the ultrashort peptide hydrogels (peptide alone or polymer-peptide composite) can be applied as versatile formulations that provide sustained and controlled release of bioactive components.
  • the bioactive moieties of interest include (but are not limited to):
  • antibiotics small molecules, ions, nano- and microparticles
  • - cosmetic agents such as serum proteins, anti-ageing, whitening agents, lubricants, sunscreen agents,
  • vitamins and small molecule compounds such as vitamin E, retinoic acid, alpha- hydroxyacids
  • nano- and microparticles such as silver (for antibacterial properties), zinc oxide (antibacterial) and titanium oxide (sun-screen), as well as polymeric nano- and microparticles encapsulating above-mentioned bioactive moieties,
  • - cells including platelets, hematopoietic stem cells, adult mesenchymal stem cells, cord blood cells, adipose stem cells, induced pluripotent stem cells, keratinocytes, cartilage and fibroblasts.
  • Bioactive moieties and cells can be encapsulated into the hydrogels by mixing during the gelation process.
  • bioactive moieties can be conjugated to the peptides or non-peptidic polymers to be incorporated in composite polymer-peptide hydrogels ( Figure 17).
  • the peptides according to the present invention self-assemble into nanofibrous porous scaffolds, they form a barrier against the external environment and can protect against dehydration and infection and also arrest bleeding.
  • hydrogels are soft biomaterials, trauma to the regenerated tissue is minimized. This is particularly important if the wound requires frequent dressing changes.
  • a major disadvantage of traditional gauze dressings is the entanglement of recovering tissue with the gauze fibers. Consequently, during dressing changes, the recovering tissue is often re-injured causing significant pain and discomfort.
  • An additional advantage of the hydrogels according to the present invention is their transparency - wound recovery can be monitored without completely cleaning or undressing the wound. Furthermore, due to the propensity of the hydrogels to absorb or release water, wound exudates are effectively managed.
  • the subclass of peptides containing lysine and similar amino acids as the polar head group can potentially facilitate hemostasis.
  • Analogues and derivatives of lysine such as e-aminocaproic acid and transexamic acid, are antifvbronolytics that prevent excessive blood loss by competitively inhibiting the activation of plasminogen to plasmin.
  • the mechanism of interaction involves offering alternative enzymatic binding sites to lysine residues in fibrin, thereby preventing the breakdown of blood clots.
  • these hydrogelating hemostatic peptides can also potentially be used to seal open and surgical wounds.
  • the hydrogel according to the present invention (1) forms a barrier to prevent bacteria and fungal spores from entering the wound, (2) forms a physical barrier to prevent cells from leaking out of damaged blood vessels (hemostatic function) ⁇ (3) limits dehydration of tissues, (4) offers protection against mechanical stress and abrasion and (5) provides mechanical cues for cellular and tissue regeneration (biomimetic scaffold).
  • a subclass of peptides/peptoids with lysine and similar polar head groups with primary amines, can bind nucleic acids released by dead and damaged cells.
  • nucleic acids include ssRNA, dsRNA and unmethylated DNA, which bind nucleic acid-sensing tolllike receptors, and subsequently activate the hosts' innate immune system.
  • molecular scavengers Lee et al, PNAS 2011
  • this subclass of peptides/peptoids can potentially block the immune stimulatory effects of extracellular nucleic acids which induce pathological inflammatory responses.
  • the peptide hydrogels can potentially significantly reduce tissue trauma due to inflammatory response post-bum injury. Maintenance of hydration and induction of autolytic debridement
  • the underlying tissues are kept hydrated, which is particularly attractive for cosmetic applications and for the treatment of wounds.
  • a key advantage of using hydrogels as wound dressings is their propensity to induce autolytic debridement of the necrotic eschar tissue. Autolytic debridement is preferred over physical methods (such as scraping, surgical excision and cleaning with gauze), which damages portions of the tissue that have started to recover.
  • the ultrashort peptide hydrogels according to the present invention create room for cells to infiltrate the damage region and commence tissue regeneration.
  • the ultrashort peptide hydrogels according to the present invention can potentially be applied as a synthetic skin substitute.
  • Their nanofibrous macromolecular architecture resembles extracellular matrix, and can thus be applied as scaffolds in place of (donor and cadaveric) skin grafts.
  • Commercially available skin substitutes are typically of animal (bovine and porcine) and bacterial origin and could present with immunogenic problems and societal (religious) objections.
  • the skin substitute fonnulation could further be enhanced by the incorporation of regenerative factors and stem cells.
  • Self-assembling ultrashort peptides hydrogels can be applied as topical dressings (hydro- gels and organogels), sprays, hydrogel patches and membranes that can be re-hydrated into hydrogels (Figure 20).
  • the semi-viscous peptide solution (mixed with cells or small molecule therapeutics) can be applied as a topical gel or cream, a spray, hydrogel sheet, patch or membrane
  • sprays, topical gels and creams can physically mix with the adjacent tissue thereby allowing for uniform dispersion of encapsulated therapeutic agents.
  • As the applied peptide solution is warmed (to body temperature), gelation is en- hanced.
  • stimuli- responsive gelation is observed in the presence of increased salt concentration, such as in normal and phosphate buffered saline ( Figure 21).
  • Stimuli-responsive gelation behavior facilitates the development of sprays, as the gelation can be stimulated by a change in salt concentration.
  • sprays can be formulated, wherein the fluid peptide solution is initially separated from the gelation-trigger agent. Upon dispersion, the solutions mix and gelation occurs. This also simplifies the delivery device, as the peptide solution is significantly less viscous at lower salt concentrations. The gelation process is endother- mic and does not release any heat.
  • the nanofibrous network of the hydrogels can be used to maintain the active ingredients in suspension, preventing aggregation or precipitation over time, thereby increasing the shelf-life.
  • the high water content can also be used to maintain tissue hydration, while allowing gaseous permeability.
  • As the hydrogels (at low concentrations) are clear and transparent, they will not significantly affect physical appearance.
  • the peptide hydrogels are biocompatible in vitro and in vivo, and could potentially encourage the regeneration of native cells.
  • gelation in situ (within the body cavity) can generate nanofibrous scaffolds to replace damaged or missing tissue.
  • the biomimetic nanotopography can potentially provide cues for cell migration, regeneration and enhance the growth of co-delivered cells.
  • In situ gelation can also be applied to form hemostatic plugs and thus prevent excessive blood loss during surgical procedures.
  • the peptides are also resistant to degradation by enzymes and would thus be more durable.
  • the material can also be sterilized by UV exposure or autoclaving, without compromising the molecular structure of the dry peptide powder. Due to their small size of the ultrashort peptides (3-7 amino acids), they can potentially diffuse through intact skin and penetrate the underlying tissue to form a scaffold for tissue regeneration.
  • hydrogels can be re-constituted by adding a fixed volume of clean water to lyophilized peptide powder at the point of appli- cation, and subsequently administered as a gel, spray or patch.
  • This formulation design will greatly reduce transportation costs, thereby facilitating adoption as part of basic first aid kits particularly in war zones and third world countries.
  • This development can potentially revolutionize emergency medicine, providing a convenient, easy-to-use first-line treatment for partial thickness bum injuries, and filling a niche that is sorely neglected by commercial skin substitutes and topical hydrogel dressings currently available on the market.
  • Self-assembly behavior of the peptides in aqueous conditions allows for the development of an injectable therapy in which the peptide solution is injected and subsequently gels in situ into a hydrogel.
  • the semi-viscous peptide solution (mixed with cells or small mole- cule therapeutics) can be injected into small cavities in the body, and is hence minimally invasive.
  • the fluid may also physically mix with the adjacent tissue, thereby allowing for uniform dispersion of the cells or therapeutic agents.
  • Current treatment options for degenerative disc disease are highly invasive and require surgical intervention in the form of spinal fusions or disc replacements with a metal or ceramic implant.
  • an injecta- ble therapy is significantly less invasive and offers an alternative treatment for patients who are not eligible for surgery.
  • Stimuli -responsive gelation behavior facilitates the development of an injectable therapy, as the gelation can be stimulated by a change in temperature or salt concentration. This allows time for the clinician to position the needle and to inject the solution before the gel sets. Injected as a cold or room temperature solution, some peptides gel in situ as the aqueous mixture warms to body temperature within the body. Alternatively, co-injection with a high salt content solution (such as via a co-axial syringe, or T-junction needle) also stimulates the gelation of salt-sensitive peptides. The gelation process is endother- mic and does not release any heat.
  • the peptide hydrogels have high mechanical strength, which can provide interim me- chanical support for the damaged and recovering tissue.
  • the injectable therapy When applied to treat degenerative disc disease, the injectable therapy will gel in situ and increase the height of the intervertebral disc, thereby increasing the cushioning effect for the vertebral bones and relieving the compression on the spinal nerve.
  • the mechanical properties of the hydrogel can be further modulated to match that of native tissue by varying the peptide sequence, composition, concentration, counter-ion and salt concentration of the solution.
  • the peptide hydrogels have demonstrated good bio- compatibility, stability and resistance to enzymatic degradation, and would thus persist in the tissue for several months. As such, the therapeutic effect is expected to last for months, reducing the need for repeated treatments.
  • the peptide hydrogels are biocompatible and can support the proliferation of native cells or stem cells injected. This would facilitate regeneration of the tissue. Cell attachment, proliferation and differentiation could potentially be enhanced by conjugating or encapsulating small molecule, short peptide motifs, cytokines, growth factors and oligonucleotides (DNA, mRNA, shRNA and siRNA).
  • the peptide sequences were designed to represent an amphophilic peptide structure containing a hydrophilic head group and a hydrophobic tail.
  • the rationale for the peptides design was to create a peptide monomer of decreasing size resembling a cone shaped structure.
  • the hydrophobic tail differs by using different aliphatic amino acids. It is consisting of the following aliphatic amino acids such as glycine, alanine, valine, leucine and isoleucine and the hydrophilic head group is consisting of one or two polar or charged amino acids.
  • the sequence order of the hydrophobic tail differed by using different ali- phatic amino acids.
  • the peptides were commercially synthesized from GL Biochem, Shanghai, China.
  • peptide hydrogel-forming behavior peptides were also synthesized from other companies (Biomatik Corp., Ana- spec. Inc, American Peptide Company, USA).
  • the peptides have a purity of equal or higher than 95% verified by High-performance liquid chromatography (HPLC) and mass spectrometry.
  • HPLC High-performance liquid chromatography
  • the peptide stock solutions were dissolved in water at 5 to 10 mg/ml. Most of the peptides are acetylated at the N-terminus.
  • Gap between two plates 0.8 mm
  • Gap between two plates 0.8 mm
  • Concentrations of NaCl solutions used to prepare 10 mg/ml of Ac-LD-6 (L) Hydrogels 1 OmM, 50 mM, 100 mM, 150 mM NaCl solution.
  • HPTCs Primary human renal proximal tubule cells
  • HUVECs primary human umbilical vein endothelial cells
  • HPTCs were obtained from ScienCell Research Laboratories (Carlsbad, CA, USA).
  • HPTCs were cultivated in basal epithelial cell medium supplemented with 2% fetal bovine serum (FBS) and 1% epithelial cell growth supplement (all compo- nents obtained from ScienCell Research Laboratories).
  • FBS fetal bovine serum
  • epithelial cell growth supplement all compo- nents obtained from ScienCell Research Laboratories.
  • the culture medium for HUVECs was endothelial cell medium containing 5% FBS and 1% endothelial cell growth supplement (ScienCell Research Laboratories).
  • Peptide hydrogel wound dressings accelerate the healing of partial thickness bum wounds in a rat model.
  • Two ultrashort peptide candidates Ac- IL VAGK-NH 2 (AcIK6) and Ac-LIVAGK- NH 2 (AcLK6) were fonnulated into hydrogel patches of 25mm diameter and 1.5mm thick. Gelation occurred within minutes, producing stable hydrogels amendable to handling.
  • the two peptide candidates chosen from this study both contained lysine as a polar head group, which could potentially facilitate hemostasis of open wounds and reduce inflammation caused by extracellular nucleic acids released by dead and damaged cells.
  • Analogues and derivatives of lysine, such as e-aminocaproic acid and transexamic acid are anti-fibronolytics that prevent excessive blood loss by competitively inhibiting the activation of plasminogen to plasmin.
  • the mechanism of interaction involves offering alternative enzymatic binding sites to lysine residues in fibrin, thereby preventing the breakdown of blood clots (Figure 23).
  • the primary amine group present on the lysine can also act as a molecular scavenger to bind extracellular nucleic acids (such as ssRNA, dsRNA and unmethylated DNA) and thus block their immune stimulatory effects (Figure 22).
  • the hydrogel patches were evaluated in a rat partial thickness bum model.
  • the standard-of-care treatment Mepitel® a flexible polyamide net coated with silicone to discourage adhesion of the regenerating tissue, was used as a control.
  • Polydimethysiloxane supports were used to maintain the integrity of the soft disc-shaped hydrogels. After positioning the dressings, water-proof TegardermTM adhesive films, gauze and crepe bandages were used to secure the dressings.
  • the wound size, granulation and re-epithelialization were quantitatively assessed using digitalized planimetry every other day, for 14 days. Photographs of the wounds were taken (see figure 26a) and the images analyzed using the program PictZar® Pro to determine the area of re-epithelialization and granulation at different time points (see figure 26b). The general linear model for repeated measures was performed using the re-epithelialization and granulation areas (absolute and percentage) at days 10, 12 and 14.
  • This model enabled us to explore the time trend of re- epithelialization and granulation and the effect on this time trend caused by different treatments while adjusting for total wound size.
  • the animals were eu- thanized and skin samples were collected from the various bum injuries. Half of each sample was preserved in 10% formalin while the other half was flash frozen in liquid nitrogen. The formalin-preserved samples were subsequently embedded in paraffin, sectioned and stained with hematoxylin and eosin. These samples were then evaluated histologically using an Olympus BX51 microscope.
  • Basal cells at the epidermis-dennis interface have also begun to penetrate the dermis to form precursors to hair shaft follicles. This corroborates well with observations gleaned from gross examinations.
  • the epidermal tissue in Mepitel®-dressed wounds was thinner and more fragile.
  • Hydrogel-treated wounds also demonstrate a greater degree of matrix remodelling in the dermis.
  • Figure 19D At the boundary of the injury site ( Figure 19D), a significantly higher extent of cell replication has occurred amongst basal cells located in the hair follicles, for wounds dressed with Ac-I 6 and Ac-LK 6 hydrogels. In contrast, hair shaft follicle regeneration in Mepitel®-dressed wounds is minimal.
  • the frozen skin samples were individually homogenized using sterile metal beads in the Procarta Cell Lysis buffer. The samples were subsequently centrifuged and the superna- tant isolated for analysis.
  • the Procarta immunoassay (Affymetrix Inc. CA, USA) was performed according to the manufacturer's instructions. Briefly, the tissue lysate were incubated with antibody magnetic beads in duplicate.
  • GM-CSF granulocyte-macrophage colony- stimulating factor
  • IFNy interferon gamma
  • IL-la interleukin 1-beta
  • IL-10 interleukin 1 -alpha
  • IL-2 interleukin 1-beta
  • IL-4 interleukin 4
  • IL-6 interleukin 6
  • TGF- ⁇ tumor necrosis factor alpha
  • TNF-a tumor necrosis factor alpha
  • VEGF-A vascular endothelial growth factor A
  • the fluorescence intensity were read using a MasterPlex CT vl.0.0.2 (MiraiBio, CA, USA).
  • concentration (pg/ml) of cytokine was calculated using MasterPlex QT v2.0.0.59 (MiraiBio, CA, USA) using 4-parameter logistics curve fitting against known standards.
  • the cytokine concentration was normalized against the total protein content for a given sample, as measured using the bicinchoninic acid (BCA) assay.
  • ANOVA One-way analysis of variance
  • the peptide Ac-LK 6 (Ac-LIVAGK-NH 2 ) was used to formulate an injectable therapy to treat degenerative disc disease in a rabbit model ( Figure 24). Two treatments were evaluated, the peptide hydrogel (encapsulating an MRI contrast agent) and hydrogel encapsulating rabbit nucleus pulposus cells (labeled with iron oxide nanoparticles). All the animals responded well to treatment, and did not experience any adverse immunological or physiological reactions. After 2 months, the animals were sacrificed and their vertebral column was collected for ex vivo MRI imaging. There was minimal leakage of the MRI contrast agent from the nucleus pulposus space, indicating that the hydrogel was still present even after 2 months. In dissection of the disc, an increase in nucleus pulposus mass was observed for several samples treated with cells.
  • Ac-LIVAGK-NH 2 hydrogels encapsulating human adipose derived stem cells were implanted in SCID mice. 6 weeks post-implantation, the mice were sacrificed and fat pads were observed at the implantation site, below muscles and the native adipose tissue present in the subcutaneous space. As this was not observed for sites implanted with human mesenchymal stem cells, this result suggests that the peptide hydrogels can potentially support the proliferation of adipose precursor cells and reduce cellular necrosis and migration following adipose cell transplantation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Preparation (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne un peptide et/ou un peptoïde linéaire amphiphile ainsi qu'un hydrogel qui comprend le peptide/peptoïde linéaire amphiphile. Le peptide/peptoïde linéaire amphiphile est apte à s'auto-assembler en réseaux macromoléculaires nanofibreux tridimensionnels, qui piègent l'eau, et à former un hydrogel. Ces peptides/peptoïdes incluent des séquences amphiphiles courtes comportant une partie hydrophobe formée d'acides aminés aliphatiques et d'au moins un acide aminé polaire acide, neutre ou basique. Le peptide/peptoïde linéaire amphiphile est formé d'acides aminés aliphatiques non répétitifs qui peuvent être de la forme L ou D. Une pluralité de tels peptides/peptoïdes s'assemble en fibres hélicoïdales supramoléculaires et forme des hydrogels peptidiques après l'assemblage. Un hydrogel correspondant est formé dans des solutions aqueuses à un pH physiologique et se révèle donc utile pour la culture de cellules, l'ingénierie tissulaire, la régénération tissulaire, la cicatrisation de plaies et la libération de fractions bioactives (notamment des cellules, acides nucléiques, agents antimicrobiens, micro et nanoparticules, agents cosmétiques et agents thérapeutiques de petites molécules), entre autres, ainsi que pour fournir un soutien mécanique à des tissus lésés ou absents. De tels hydrogels peuvent également être formés in situ, le procédé de gélification se déroulant dans l'organisme après l'injection d'une solution peptidique. De tels hydrogels qui sont rigides, biocompatibles et piègent jusqu'à 99,9 % d'eau sont également bien adaptés pour des applications faisant appel à des dispositifs électroniques.
EP13869801.4A 2012-12-31 2013-12-31 Hydrogels peptidiques ultracourts auto-assemblés pour cicatrisation de plaies, soins de la peau et applications cosmétiques Withdrawn EP2938626A4 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
SG2012096699A SG2012096699A (en) 2012-12-31 2012-12-31 Amphiphilic linear peptide/peptoid and hydrogel comprising the same
PCT/SG2013/000564 WO2014104981A1 (fr) 2012-12-31 2013-12-31 Hydrogels peptidiques ultracourts auto-assemblés pour cicatrisation de plaies, soins de la peau et applications cosmétiques

Publications (2)

Publication Number Publication Date
EP2938626A1 true EP2938626A1 (fr) 2015-11-04
EP2938626A4 EP2938626A4 (fr) 2016-08-17

Family

ID=54063251

Family Applications (1)

Application Number Title Priority Date Filing Date
EP13869801.4A Withdrawn EP2938626A4 (fr) 2012-12-31 2013-12-31 Hydrogels peptidiques ultracourts auto-assemblés pour cicatrisation de plaies, soins de la peau et applications cosmétiques

Country Status (4)

Country Link
EP (1) EP2938626A4 (fr)
CN (1) CN105189532B (fr)
SG (2) SG2012096699A (fr)
WO (1) WO2014104981A1 (fr)

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2931954A1 (fr) * 2013-11-30 2015-06-04 Agency For Science, Technology And Research Nouveaux peptides hydrophobes ultracourts qui s'auto-assemblent en hydrogels nanofibreux et leurs utilisations
EP3074416A4 (fr) * 2013-11-30 2017-09-13 Agency for Science, Technology and Research Peptides auto-assemblés, peptidomimétiques et conjugués peptidiques comme blocs de construction pour biofabrication et impression
WO2016007091A1 (fr) * 2014-07-08 2016-01-14 Agency For Science, Technology And Research Peptides ultracourts en tant que sondes exogènes à deuxième harmonique pour applications en bio-imagerie
DE102015000363A1 (de) * 2015-01-20 2016-07-21 Emc Microcollections Gmbh Neue modular funktionalisierbare Peptid-Hydrogele
EP3265473A4 (fr) * 2015-03-06 2018-09-05 Agency For Science, Technology And Research Depsipeptides aliphatiques ultra-courts auto-assemblés pour applications biomédicales
WO2016149363A1 (fr) * 2015-03-16 2016-09-22 Northwestern University Nanofibres amphiphiles peptidiques marquées par un agent de contraste
CN107406486A (zh) * 2015-03-31 2017-11-28 新加坡科技研究局 用于生物医学应用的自组装超短脂族环肽
JP6908284B2 (ja) 2015-10-14 2021-07-21 ノースウエスタン ユニバーシティNorthwestern University 骨伝導性複合材料
GB201523102D0 (en) * 2015-12-30 2016-02-10 Augmented Optics Ltd Electrically active hydrophilic bio-polymers
CN106083634B (zh) * 2016-06-16 2017-12-05 淮阴师范学院 一种两亲性肽分子及其制备方法和应用
CN106432430B (zh) * 2016-08-17 2019-11-05 广州宏柯源生物科技有限公司 一种多肽、其制备方法及应用
US11021516B2 (en) * 2016-09-09 2021-06-01 Research Foundation Of The City University Of New York Self-assembling peptide polymer
KR101829051B1 (ko) * 2016-10-31 2018-02-13 주식회사 삼양사 점착력이 높고 두꺼운 하이드로겔 시트
AU2018251805B2 (en) 2017-04-12 2022-08-18 X-Therma, Inc. Novel peptoid polymers and methods of use
SG11201910173YA (en) 2017-05-11 2019-11-28 Univ King Abdullah Sci & Tech Device and method for microfluidics-based 3d bioprinting
JP7326161B2 (ja) 2017-05-11 2023-08-15 キング アブドラ ユニバーシティ オブ サイエンス アンド テクノロジー 組織エンジニアリングおよびバイオプリンティングにおける使用のためのゲルを形成し得るペプチド
US11472841B2 (en) * 2018-01-19 2022-10-18 Menicon Co., Ltd. Immunogenic composition
CN108912348B (zh) * 2018-07-28 2021-07-02 邹立人 含有蛋白的短肽水凝胶和提高蛋白储存稳定性的方法
US20210386907A1 (en) * 2018-10-16 2021-12-16 William Marsh Rice University Neutral multidomain peptide hydrogels and uses thereof
CN109456389B (zh) * 2018-12-25 2021-06-01 苏州大学 一种抗菌肽、抗菌肽水凝胶及其制备方法
JP7159861B2 (ja) * 2018-12-27 2022-10-25 株式会社Sumco 両頭研削方法
CN113396154A (zh) * 2019-02-08 2021-09-14 新加坡科技研究局 自组装短两亲性肽以及相关的方法和用途
JP6873436B2 (ja) * 2019-05-17 2021-05-19 学校法人大阪医科薬科大学 関節疾患治療用の医薬組成物及びその製造方法
CN110169950B (zh) * 2019-07-04 2023-01-24 中原工学院 一种可注射性抗菌肽水凝胶及其制备方法
CN112724823B (zh) * 2019-10-28 2022-08-02 华东理工大学 一类多肽聚合物的用途
CN111760075B (zh) * 2020-07-08 2021-08-13 西南交通大学 抗菌抗凝血涂层及其制备方法和医用材料
US11673324B2 (en) 2020-08-20 2023-06-13 King Abdullah University Of Science And Technology Nozzle for 3D bioprinting
US20230374069A1 (en) * 2020-09-02 2023-11-23 King Abdullah University Of Science And Technology Peptide-based dopa containing adhesive gels
CN113577014B (zh) * 2020-12-30 2023-03-24 广州图微科创生物科技有限公司 医疗器械、水凝胶及其制备方法与应用
CN112940077B (zh) * 2021-02-03 2023-01-03 中山大学孙逸仙纪念医院 一种装载小干扰rna的多肽水凝胶及其制备方法和应用
BR112023019020A2 (pt) * 2021-03-21 2023-11-28 Bone Sci Bio Ltd Composições dermatológicas e uso das mesmas
CN115340592A (zh) * 2021-09-09 2022-11-15 禾美生物科技(浙江)有限公司 一种活性肽及其相关用途
CN114989249B (zh) * 2022-05-27 2023-07-21 成都赛恩贝外科学研究院 纳米短肽r-life-1及其在药物、医疗美容和生物医学的应用
CN115678044B (zh) * 2022-11-08 2023-05-19 郑州大学 一种基于牛血清白蛋白的质子导电有机水凝胶及其制备和应用
CN116139248A (zh) * 2022-11-18 2023-05-23 中国科学院过程工程研究所 一种肽水凝胶、其制备方法及其在治疗中的应用

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6576239B1 (en) * 1996-09-10 2003-06-10 The Burnham Institute Angiogenic homing molecules and conjugates derived therefrom
AU2002361610B2 (en) * 2001-11-07 2007-01-11 Agensys, Inc. Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
JP3830386B2 (ja) * 2001-12-20 2006-10-04 英夫 吉田 陽極酸化法およびその処理装置
US20070048752A1 (en) * 2004-07-12 2007-03-01 Applera Corporation Mass tags for quantitative analyses
JP5244103B2 (ja) * 2006-08-09 2013-07-24 ホームステッド クリニカル コーポレイション 器官特異的蛋白質およびその使用方法
EP2331048B1 (fr) * 2008-05-29 2019-07-10 Donald Owen Agents tensioactifs biologiques oligomères
US20100221341A1 (en) * 2009-02-27 2010-09-02 Drexel University Alanine-Based Peptide Hydrogels
US9067084B2 (en) 2010-03-31 2015-06-30 Agency For Science, Technology And Research Amphiphilic linear peptide/peptoid and hydrogel comprising the same
WO2011132018A1 (fr) * 2010-04-19 2011-10-27 Universita' Degli Studi Di Milano Bicocca Nouveaux peptides autoassemblés et leur utilisation dans la formation d'hydrogels
US10449257B2 (en) 2011-11-04 2019-10-22 Agency For Science, Technology And Research Self-assembled composite ultrasmall peptide-polymer hydrogels
SG193042A1 (en) 2012-02-22 2013-09-30 Agency Science Tech & Res Organogels from ultrasmall peptides that can be used for biological and non-biological applications

Also Published As

Publication number Publication date
WO2014104981A1 (fr) 2014-07-03
SG2012096699A (en) 2014-07-30
SG11201505160PA (en) 2015-07-30
CN105189532B (zh) 2020-11-03
EP2938626A4 (fr) 2016-08-17
CN105189532A (zh) 2015-12-23

Similar Documents

Publication Publication Date Title
EP2938626A1 (fr) Hydrogels peptidiques ultracourts auto-assemblés pour cicatrisation de plaies, soins de la peau et applications cosmétiques
US9120841B2 (en) Amphiphilic linear peptidepeptoid and hydrogel comprising the same
US10071183B2 (en) Amphiphilic linear peptide/peptoid and hydrogel comprising the same
US10286111B2 (en) Crosslinked peptide hydrogels
EP2958933B1 (fr) Hydrogels de peptides réticulés
EP2885015B1 (fr) Utilisation de polypeptides à auto-assemblage en tant qu'adhésifs tissulaires
US8691944B2 (en) Fibronectin polypeptides and methods of use
US9687591B2 (en) Building stratified biomimetic tissues and organs using crosslinked ultrashort peptide hydrogel membranes
US20230226142A1 (en) Ionic self-assembling peptides
US20050282747A1 (en) Methods and compositions for wound healing
RU2458069C2 (ru) Выделенный пептид для усиления ранозаживляющей активности кератиноцитов, композиция для заживления ран у млекопитающего и лекарственное средство для применения при заживлении ран у млекопитающего
JP6548126B2 (ja) Iii型コラーゲン産生促進剤
WO2006025646A1 (fr) Peptide, fragments et derives de ce peptide favorisant l'adhesion et l'etalement des cellules
Saklani et al. Laminin mimetic angiogenic and collagen peptide hydrogel for enhance dermal wound healing
JP2010195714A (ja) 創傷治療用医薬組成物
PL237001B1 (pl) Nowy związek peptydowy jako czynnik stymulujący gojenie ran i rekonstrukcję skóry

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20150729

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20160719

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 5/103 20060101ALI20160713BHEP

Ipc: C07K 5/062 20060101ALI20160713BHEP

Ipc: C07K 5/083 20060101ALI20160713BHEP

Ipc: C07K 7/06 20060101AFI20160713BHEP

Ipc: A61K 38/07 20060101ALI20160713BHEP

Ipc: A61K 38/08 20060101ALI20160713BHEP

Ipc: A61K 47/42 20060101ALI20160713BHEP

Ipc: A61K 38/06 20060101ALI20160713BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20171219

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20230701