WO1997040854A2 - Conjugues polypeptidiques destines au transport de substances a travers des membranes cellulaires - Google Patents

Conjugues polypeptidiques destines au transport de substances a travers des membranes cellulaires Download PDF

Info

Publication number
WO1997040854A2
WO1997040854A2 PCT/US1997/007335 US9707335W WO9740854A2 WO 1997040854 A2 WO1997040854 A2 WO 1997040854A2 US 9707335 W US9707335 W US 9707335W WO 9740854 A2 WO9740854 A2 WO 9740854A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
compound
composition
acid
cell
Prior art date
Application number
PCT/US1997/007335
Other languages
English (en)
Other versions
WO1997040854A3 (fr
Inventor
James E. Summerton
Dwight D. Weller
Original Assignee
Antivirals Inc.
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 Antivirals Inc. filed Critical Antivirals Inc.
Priority to CA002252706A priority Critical patent/CA2252706A1/fr
Priority to EP97923513A priority patent/EP0966303A2/fr
Priority to JP9539221A priority patent/JP2000509394A/ja
Priority to AU29298/97A priority patent/AU729643B2/en
Publication of WO1997040854A2 publication Critical patent/WO1997040854A2/fr
Publication of WO1997040854A3 publication Critical patent/WO1997040854A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/645Polycationic or polyanionic oligopeptides, polypeptides or polyamino acids, e.g. polylysine, polyarginine, polyglutamic acid or peptide TAT
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention relates to the use of polymeric compositions effective for delivering compounds in living organisms.
  • the compositions exhibit solubility in both hydrophilic and lipophilic environments by undergoing a reversible pH-dependent transition from a low-pH, lipophilic form to a high-pH, hydrophilic fo ⁇ n.
  • Lipid layers such as comprise cell membranes and the extracellular matrix of the stratum corneum, can constitute a daunting barrier to drug delivery.
  • a drug should freely dissolve in both the aqueous compartments of the body and the lipid layers which enclose those compartments.
  • the invention provides a composition for transporting a compound from a low-pH environment across a lipid layer to a higher-pH aqueous compartment.
  • the composition includes a polypeptide having one or more pairs of carboxyl groups, where the carboxyl groups of a pair are separated by zero, two or three amino acids, and a length of between about 8 and about 100 amino acid residues, and preferably between 10 and 50 residues.
  • the polypeptide further contains an initiator moiety at one end region of the polypeptide, to facilitate entry of said end region, and partitioning of the polypeptide, into the lipid layer.
  • Such a polypeptide is effective to undergo a reversible transition between a lipophilic form, which is effective to partition from the low-pH environment into the lipid layer, and a hydrophilic form, which is effective to partition preferentially from the lipid layer into the higher-pH aqueous compartment.
  • the polypeptide is thereby able to traverse the lipid layer from the low-pH environment to the higher-pH compartment.
  • the composition also includes, covalently attached to the polypeptide, the compound to be transported.
  • 30-100 percent of the amino acid residues forming the polypeptide, excluding the initiator moiety are glutamic acid. Pairs of glutamic acid residues in the polypeptide may be separated by other ammo acid residues, which are preferably selected from the group consisting of leucine, methionine, alanine, and 2-amino butyric acid. Alternatively, the polypeptide may be polyglutamic acid having an initiator moiety at one end region.
  • the initiator moiety may be an initiator amino acid sequence having 3-12 amino acid residues.
  • the sequence is effective to form an alpha helix at a pH higher than the pH at which a same-length polyglutamic acid forms an alpha helix.
  • Preferred amino acid residues in such a sequence are glutamic acid, leucine, methionine, alanine, 2-aminobutyric acid, norvaline, and /3-alanine, where the ratio of non-glutamic acid to glutamic acid residues is greater than 1.
  • the initiator sequence may be an end region of the polypeptide itself.
  • the may be a group linked covalently to the N or C terminus which is effective to eliminate a positive or negative charge at said terminus, and which has at least one remote polar group effective to shield polar sites near the terminus.
  • a lipophilic substance at the terminus, including a compound to be delivered, may serve as an initiator moiety.
  • a single polypeptide may also contain a combination of such initiator moieties.
  • the compound to be delivered may be attached to an end of the polypeptide component, typically the end opposite the initiator sequence.
  • the composition may have only one, or multiple compounds per polypeptide component.
  • the compound to be delivered may include a compound which is itself only sparingly soluble in free form in an injectable aqueous delivery medium, such as taxol, cyclosporin, amphotericin B.
  • compound is a sequence-specific nucleic acid binding polymer, e.g., an antisense compound.
  • the invention provides a method of facilitating the transport of a compound from a low-pH environment across a lipid layer to a higher-pH aqueous compartment.
  • the compound is covalently coupled to a polypeptide as described above.
  • the method is useful, for example, in delivering a therapeutic compound from an extracellular medium, having a given pH, to the cytosol of a cell, having a higher pH.
  • the method is used in antitumor therapy, where the cell is a tumor cell existing in an acidic extracellular medium, and the attached compound is an antineoplastic agent.
  • the method is used in the treatment of H. pylon infection, where the extracellular medium is stomach fluid, the cell is an H. pylori bacterial cell, and the compound is an antibacterial agent, or in the treatment and prevention of tooth decay, where the cell is an acid-producing cariogenic bacterial cell, and the attached compound is an agent effective against cariogenic bacteria.
  • transdermal delivery of a compound through the stratum corneum wherein the lipid layer to be traversed comprises the extracellular matrix of the stratum corneum, and in transporting a compound across the brain-blood barrier.
  • the composition is transported within a transcytotic vesicle through an endothelial cell of a capillary wall, and is taken up by a brain cell within an endocytotic vesicle.
  • Figure 1A-1B illustrate a pH-dependent transition between hydrophilic and lipophilic conformations of side chain carboxyls of a polymer
  • Figure 2A depicts the process of cytosolic entry, via endocytosis, of a polypeptide with attached compound, in accordance with one aspect of the invention
  • Figure 2B shows the polypeptide-compound complex in its high pH form, which exists in the early-stage endosome
  • Figure 2C shows the complex in its low pH form, which exists in the late-stage endosome, entering the lipid membrane, and converting back to the high pH form upon contacting the higher pH cytosol;
  • Figure 3 depicts the transdermal transport process of a polypeptide containing an attached compound, where the complex passes from the extracellular lipid matrix to the aqueous compartment beneath the stratum corneum;
  • Figure 4 shows paired, spaced glutamic carboxyls of a polypeptide in an ⁇ -helix;
  • Figures 5A-5C show representative C-terminal end structures of polypeptides, where the structures in 5B-5C have partially shielded or deleted terminal polar sites;
  • Figures 6A-6D shows representative N-terminal end structures of polypeptides, where the structures in 6B-6D have shielded terminal polar sites;
  • Figure 6E shows an "N-crown" structure which may be used to shield polar sites of N- terminal end structures in polypeptides, as shown in Fig. 6D;
  • Figure 7 shows a polypeptide functioning as a molecular engine, in accordance with an embodiment of the invention.
  • Figures 8A-8C depicts polypeptides assessed for octanol solubility and cell entry, where Fig. 8A shows a polyglutamic acid with unmodified termini, Fig. 8B illustrates the incorporation of groups which delete or shield polar sites, and Fig. 8C shows further addition of lipophilic amino acid residues at the termini;
  • Figure 9 illustrates representative sites of side-chain attachment on a polypeptide, showing, from left to right, amide and carbamate links to lysine, an ester link to glutamic acid, and disulfide, thioether, and amide links to cysteine;
  • Figure 10 shows a method of preparation of polypeptides which may be used in accordance with the invention.
  • Figures 11 A-l IB show representative polymer-compound linkages, such as may be used in attaching a compound to a side chain or terminus of a polypeptide;
  • Figure 12 illustrates enhancement of endocytosis, as shown in Figure 2A, by a lipid anchor attached to a polycationic head group;
  • Figures 13 A-C show procedures for linking a polypeptide of the invention to a Morpholino antisense oligomer
  • Figures 14A-14F show octanol/water partitioning properties of several exemplary polypeptides, as a function of pH.
  • low pH form also low pH conformation, lipophilic form, or lipophilic confor ⁇ mation
  • a polypeptide of the invention or a segment of the polypeptide, is a substantially non-ionic ⁇ -helical conformation rendered lipophilic by hydrogen bonding between paired side- chain or terminal carboxyl groups.
  • the "high pH form” (also high pH conformation, hydrophilic form, or hydrophilic conformation) of a polypeptide of the invention, or a segment of the polypeptide, is a conformation in which the side-chain or terminal carboxyl groups are wholly or predominantly in a non-hydrogen-bonded, ionic state.
  • a "reversible transition" between the lipophilic form and the hydrophilic form of a polypeptide is a transition between a nonionic, ⁇ -helical conformation, wherein side chain carboxyls are engaged in intramolecular hydrogen bonding, favored by low pH, and a form wherein side chain carboxyls are in ionic, non-hydrogen-bonded states, favored by high pH.
  • Such a transition may encompass an entire polypeptide, or it may occur at a localized region of a polypeptide, particularly when the region is in the vicinity of an aqueous/lipid interface, or has a composition that is especially lipophilic and/or especially favors the formation of an ⁇ -helix.
  • Such a localized region of a polypeptide is often effective to initiate entry of the polypeptide into a lipid phase, such as a membrane, even though regions of the polypeptide more remote from the lipid phase may be in a hydrophilic conformation.
  • an “acid amino acid” or “acid side chain acid” is one which has a free carboxyl group when incorporated into a polypeptide. Examples are aspartic acid and glutamic acid.
  • a “non-acid amino acid” or “non acid side chain acid” is one which has no free carboxyl group when incorporated into a polypeptide.
  • a "high glutamic” polypeptide is a polypeptide containing greater than about 50% glutamic acid residues.
  • an "initiator sequence” is a short (typically 3-12 amino acid residues) sequence at a terminus of a polypeptide which readily forms an ⁇ -helix at pH's attainable in endosomes of mammalian cells, generally at a pH significantly higher than that at which the same length sequence of glutamic acid residues would form an ⁇ -helix.
  • Such a sequence at die terminus of a polypeptide initiates entry of the polypeptide into a lipid layer and promotes ⁇ -helix formation in immediately adjacent segments of the polypeptide.
  • a group which "deletes” or “shields” polar groups at or near the terminus of a polypeptide is a covalently linked moiety which caps or replaces a carboxylate anion or protonated amine at the terminus. Preferred groups further mask other polar sites near the terminus by non ⁇ covalent associations, typically hydrogen bonding.
  • Endocytosis is a process by which extracellular material is taken into a cell via an invagination of the cell membrane, which closes to form a vesicle within the cell known as an endosome. Endocytosis may be receptor-mediated, where the extracellular compound binds to a specific receptor on the cell surface, or extracellular compounds may be imported nonspecifically, by virtue of their presence near the cell membrane. The latter process is also known as fluid-phase endocytosis or pinocytosis. A related process, potocytosis, takes compounds into the cell via vesicles near the cell surface known as caveolae. In all of the above processes, the vesicles enclosing the extracellular compound become increasingly acidic after formation. The compositions and methods of the invention are therefore applicable to all of these methods of transport.
  • the polymer composition of the invention includes a polypeptide component capable of undergoing a transition between a hydrophobic, ⁇ -helical form, and a hydrophilic form; an initiator moiety at one end region of the polypeptide, to facilitate partitioning of the polypeptide into a lipid layer through that end region, and the compound to be delivered, covalently attached to the polypeptide.
  • the polypeptide component contains one or more pairs, and preferably two or more pairs, of carboxyl groups, where the two carboxyls of a pair are separated by zero, two or three amino acids.
  • the composition and positioning of the amino acids of the polypeptide are such that, in the presence of both an aqueous and a lipid-like phase, the polypeptide undergoes, typically at a pH between about 4.0 and about 7.0, a reversible transition between a high-pH form, comprising a multiply-ionized hydrophilic structure, and a low-pH form, comprising a substantially non-ionic ⁇ -helical structure rendered lipophilic by hydrogen bonding between the paired carboxyls.
  • Figure IA illustrates the pH-dependent transition between hydrophilic and lipophilic forms of a polypeptide
  • Figure IB illustrates this solubility transition for a specific sequence of properly-spaced carboxyl- containing amino acid pairs.
  • the polymer composition can be used to transport an attached compound from the extracellular compartment to the cytosolic compartment within cells.
  • the process is illustrated in Fig. 2A.
  • the composition 10 is readily delivered, in an aqueous vehicle, to the extracellular compartment by virtue of the aqueous solubility imparted by the high-pH form of the polypeptide.
  • the composition is enclosed within an endosome 14.
  • the pH within the endosome decreases due to the action of ATP-driven proton pumps within the endosomal membrane (see e.g. Clague, Fuchs).
  • the composition converts, in the increasingly acidic environment of the late stage endosome, shown at 16, to its low-pH lipid-soluble form, which enters the endosomal membrane.
  • the composition Upon contacting the cytosol ic face of the endosomal membrane, the composition is actively drawn into the cytosol by virtue of progressive ionization and solvation of the polypeptide chain at the near-neutral pH of the aqueous cytosol, as shown at 18, resulting in delivery of the attached compound 20 into the desired subcellular compartment.
  • a polypeptide constructed from L-amino acids will typically be degraded in the cytosol, as shown, after the drug has been released. This unidirectional active transport process is illustrated further in Figures 2B and 2C, where only a portion of the endosomal membrane is represented.
  • entry into the membrane may be initiated by a localized lipophilic region of the polypeptide, even when other regions of the polypeptide are in a non- hydrogen-bonded, hydrophilic conformation. Such entry is especially favored when this lipophilic region is at a terminus of the polypeptide.
  • a terminus of the polypeptide may comprise amino acid sequences which readily assume a lipophilic conformation at a relatively high pH, or may be otherwise modified to render the terminus more lipophilic. Such modifications are discussed further below.
  • composition of the invention is that endosome-to-cytosol transport may be achieved without disruption of the endosomal membrane, thus avoiding leakage of lysosomal enzymes into the cytosolic compartment of the cell.
  • the polymer composition can also be used to transport an attached compound directly into the cytosol of cells in cases where the extracellular medium has a pH less than the pH of the cytosol of the cells, as described below. This allows, for example, selective delivery of compounds into eukaryotic cells, bacteria or other target cells in acidic environments.
  • the polymer composition can further be used for transdermal delivery of selecte. compounds, as described below, and as illustrated in Fig. 3.
  • the composition 10 is contacted, in its low-pH lipid-soluble form, with the surface of the epidermis, shown at 22, leading to diffusion through the extracellular lipid matrix 24 of the stratum corneum, which contains epithelial cells 26.
  • the composition Upon contact with the aqueous compartment underlying the stratum corneum, shown at 28, the composition is actively drawn into this compartment by virtue of progressive ionization and solvation of the polypeptide chain at the near-neutral pH of this compartment, thereby effecting delivery of the attached compound into the underlying tissues, with subsequent distribution throughout the body.
  • compositions are particularly useful for the delivery of compounds which are only sparingly soluble in free form in aqueous delivery media.
  • These compounds include, but are not limited to, TaxolTM, TaxolTM analogs, cyclosporin analogs, and amphotericin B.
  • Also contemplated is the delivery of sequence-specific nucleic acid binding polymers.
  • polypeptide component Two important structural features of the polypeptide component are that: a) it contains at least one properly-spaced acid pair, and b) it contains a number of amino acids sufficient to adopt a lipophilic ⁇ -helix at low pH.
  • various factors are considered, such as the properties of the compound to be delivered, the compartments from and into which the compound is to be delivered, and the desirability of including end modifications to shield exposed polar sites and increase lipophilicity at the helix termini.
  • carboxyl side chain amino acids preferably aspartic acid and glutamic acid
  • carboxyl side chain amino acids preferably aspartic acid and glutamic acid
  • the preferred number and frequency of these acids is discussed below.
  • a free carboxyl group at either polypeptide terminus may also hydrogen bond with a properly situated carboxyl side chain amino acid.
  • the carboxyl terminus may be prepared with 0-alanine, ⁇ -amino butyric acid, or another omega- amino carboxylic acid as its terminal group, by initiating the synthesis of the polypeptide using such an acid, as described in Example 1.
  • omega-carboxy alkyl amide at the C-terminus (as shown, for example, in Figs. 10 and 5C), where the carboxyl group at the end of the alkyl chain is available for hydrogen bonding.
  • the N-terminus may be capped with a diacid, such as glutaric acid (as shown in Fig. 6B) to provide an additional hydrogen-bonding carboxyl group.
  • Non-acidic amino acids The non-acid amino acids of the polymer, when present, serve principally to provide proper spacing between the two carboxyl groups of each hydrogen- bonding pair, and to adjust the pH of the partitioning transition to a selected value, as discussed below. These amino acids should not contain moieties which are cationic when the polymer is in its low-pH ⁇ -helical conformation (i.e., arginine, lysine, and histidine). (An exception is the use of low levels of amino acids such as lysine as sites for attachment of the transported compound, as described in below.) Most of the non-acid amino acids of the polymer are relatively non-polar and compatible with ⁇ -helix formation.
  • non-acid amino acids for most applications are selected from the following: tryptophan, phenylaianine, leucine, isoleucine, methionine, valine, and alanine.
  • Norvaline, ⁇ -amino butyric acid, and side chain esters of aspartic and glutamic acid are also suitable as "non-acid" amino acids.
  • leucine, methionine, alanine and ⁇ - amino butyric acid are particularly preferred.
  • polymers with a high density of acid moieties are not natural components of the interior of cells and so could prove toxic therein. However, such toxicity is much reduced or prevented if, after carrying out its drug transport function, said polymers are disassembled into natural subunits endemic in the cytosol of cells.
  • unstructured polypeptides composed of natural L-amino acids can be rapidly depolymerized in the cytosol of cells, primarily in complex multi-ring structures called proteasomes.
  • the preferred amino acids of the polymer are selected from natural L-amino acids.
  • the preferred amino acids are selected from D-amino acids.
  • other non-natural amino acids are also suitable, such as ⁇ -amino butyric acid, norvaline, and norleucine.
  • E glutamic acid
  • D aspartic acid
  • X amino acid
  • spacings of zero, two, or three amino acids between carboxyl side chain amino acids are effective. Of these, spacings of two or three amino acids between carboxyl side chain amino acids are preferred.
  • Polypeptides containing sequences wherein polar side chains alternate with nonpolar side chains are known to adopt ⁇ -sheet and related conformations.
  • Polypeptides with such alternat ⁇ ing sequences greater than about 6 amino acids in length were found to be generally insoluble in octanol at low pH, probably due to the formation of multi-chain complexes whose polar sites are not adequately shielded from the solvent. Accordingly, the polymer of the invention should be largely free of significant runs of alternating polar and nonpolar side chains.
  • polypeptides of the invention should be at least 8 amino acids in length, and preferably at least 10 amino acids in length.
  • each amino acid residue contributes approximately 1.5 A to the axial length. Since lipid bilayers of cell membranes in eukaryotic cells are typically about 33 to 36 A thick, a preferred length for the polymer composition is about 22 amino acids or more. Several additional amino acids are preferably included to assure that at least one acid side chain enters the high-pH compartment to initiate conversion to the ionic high pH form. Thus, polypeptides in the length range of about 24 to 100 amino acids, preferably 24 to 48 amino acids, are preferred for endosome-to-cytosol transport of large and/or polar compounds. For delivery of the smaller or less polar attached compounds, shorter peptides may be used.
  • the low-pH lipophilic form When the polypeptide is below, near, or even somewhat above its transition pH, the low-pH lipophilic form is present at an equilibrium concentration sufficient to effect diffusion of the polypeptide into and across the lipid membrane. Only for attached compounds which significantly interfere with this diffusion, as described above, is it necessary for the polypeptide to completely span the membrane at any given time.
  • polypeptides As discussed further below, entry into a cell membrane is most likely to be initiated at a terminus of the polypeptide, especially for more polar polypeptides. In longer polypeptides, such as those having lengths greater than about 200 amino acids, the termini are at a low concentration and are statistically less likely to contact the cell membrane. In addition, longer polypeptides, by virtue of size alone, may not be efficiently engulfed within an endosome, which is typically about 800A in diameter. Preferred polypeptides, such as described above, will thus have an average length of less than 200 amino acids, and most preferably less than about 100 amino acids. B5. Polyglutamate and polyaspartate homopolymer components.
  • High molecular weight, random-length polyglutamic acids such as are commercially available from, e.g., Aldrich Chemical Co., Milwaukee, WI, or readily polymerized from N- carboxyanhydrides (Blout), precipitate from water, but do not partition into n-pentanol, at about pH 4.
  • Such polymers also fail to cross cell membranes in direct-entry experiments, such as described below and Example 10A, at pH's attainable in endosomes of mammalian cells.
  • poly-L-glutamic acid when linked to various anticancer drugs, such as adriamycin (Hoes, Nukui), in an effort to enhance endocytotic uptake of the drug by increasing aqueous solubility, the polyglutamic acid component of the polypeptide-drug conjugate neither entered the endosomal membrane nor transported the drug across the membrane. Rather, the carrier was degraded within the fused endosome-lysosome, and the released drug then passively diffused across the lysosomal membrane into the cytosol.
  • adriamycin Hoes, Nukui
  • Polyaspartic acid is soluble in aqueous solution at acidic pH, failing to partition into n- octanol or n-pentanol even when the pH of the aqueous phase is as low as 4. Polyaspartic acid also fails to cross cell membranes in direct-entry experiments, such as described below. Such lack of lipophilicity is expected on the basis of partitioning calculations (Leo), and because molecular modeling of a polyaspartic acid ⁇ -helix shows that the carboxyl groups are unable to form the double-hydrogen-bonded pairs required to shield the polar acid sites from the environment.
  • Leo partitioning calculations
  • the first is an initiator polypeptide sequence, which may be an end-region extension of a polypeptide component containing less than about 50% acidic residues, or, in the case of a homopolymer of acidic amino acids, a more hydrophobic, alpha-helix forming region containing less than about a 50% acidic, e.g., glutamate residues.
  • the second is a hydrophobic moiety that may be, for example, the compound to be administered, or a compound effective to eliminate and/or mask end charges and polar groups.
  • Such an amino acid sequence at the polypeptide terminus is particularly useful in that it provides an initiator sequence effective to form an ⁇ -helix at a higher pH than would be required for a similar length sequence of, in the present example, glutamic acid residues, to assume this conformation.
  • Cell entry and partitioning studies have demonstrated that inclusion of such an ⁇ -helix-forming initiator sequence enhances lipid solubility and cell entry to a significantly greater extent than lipophilic modifying groups which do not in themselves initiate ⁇ -helix formation.
  • the initiator sequence in forming an ⁇ -helix at relatively high pH, also promotes subsequent ⁇ -helix propagation in the adjacent segments of the polypeptide.
  • Preferred initiator sequences are three to twelve, and preferably five to ten, amino acid residues in length, and preferably include glutamic acid, leucine, methionine, alanine, 2- aminobutyric acid, 3-aminobutyric acid, norvaline, and /3-alanine, where the ratio of non-acid side chain residues to acid side chain residues is greater than 1, and preferably greater than 2.
  • Examples of such initiator sequences are XEXX/3 and XEXXX/3 (C-terminal) and glutaric acid- XXEX and glutaric acid-XXXEX (N-terminal), where E is glutamic acid, ⁇ is / 3-alanine, and X is selected from the list above.
  • One such C-terminal sequence, leu-glu-leu-leu-/3, is shown in Fig. 8C.
  • terminal sequences provide the clearest benefit in highly polar (e.g. high glutamic) polypeptides. They are also useful in defmed-sequence polypeptides containing lower levels (i.e., less than about 50%) of acid side chain residues. Such polypeptides, however, generally have a higher propensity toward ⁇ -helix formation, and thus other initiator moieties, such as a lipophilic drug at the terminus, or a shielding group as described above, may be effective to initiate entry of such polypeptides into a membrane.
  • a polypeptide in an ⁇ -helical conformation typically contains multiple polar sites at both the C-terminus and the N-terminus which are not shielded by intramolecular hydrogen bonding. These unshielded polar and ionic sites constitute a substantial bar to initiation of polypeptides into lipid layers, due to the presence of solvated counterions and water of solvation, and the propensity of the polar termini to assume a non- ⁇ - helical conformation. Deleting the terminal charge and shielding or removing one or more of these polar sites can improve lipid solubility, particularly in the case of sho ⁇ or highly polar polypeptides, such as high-glutamic polypeptides, described above.
  • a lipophilic substance, such as a drug to be delivered, attached at a polymer terminus eliminates the charge at said terminus and provides local lipophilicity.
  • Deleting the terminal charge and shielding or removal of polar sites can also be accomplished by incorporating, at the C-terminus or the N-terminus, a group which covalently bonds to, or includes, the terminal carboxyl or amino group, and which further contains at least one remote polar group, such as a hydroxyl or carbonyl group, which is effective to shield, by hydrogen bonding, one or more additional polar groups at or near the polypeptide terminus. Specific examples follow.
  • the C-terminus of a polypeptide typically contains three carbonyl groups and a negatively-charged carboxylate ion which are not shielded by intramolecular hydrogen bonding, as illustrated in Fig. 5A.
  • One method for reducing the number of these unshielded terminal polar sites is to incorporate an ⁇ -ester, preferably a 2-hydroxyethyl ester, at the C-terminus, as illustrated in Fig. 5B.
  • Polar sites may also be conveniently eliminated from the C-terminus by initiating the synthesis of the polypeptide on a support resin with ⁇ - alanine, as shown in Fig. 5C, and as described in Example 1.
  • N-Terminus shielding The N-terminus of a polypeptide typically contains three amide protons and a positively-charged protonated amine which are not shielded by intramolecular hydrogen bonding, as illustrated in Fig. 6A.
  • Polar sites may be conveniently removed from the N-terminus by terminating the polypeptide with a diacid, as illustrated in Fig. 6B.
  • polar sites can be shielded simply by acetylating the terminal amine, as illustrated in Fig. 6C. Hydrogen bonding occurs as shown when the polypeptide is in an ⁇ -helix.
  • FIG. 6D shows one such N-crown, whose structure is given in Fig. 6E, in the H-bonded conformation it is believed to adopt when linked to the N-terminus of a polypeptide existing in an ⁇ -helical conformation.
  • FIG. 8B A polyglutamic acid having such modifications is shown in Fig. 8B.
  • a large lipophilic moiety i.e. , fluorescein in its low-pH form
  • a polypeptide having a large number of acid side chains is able to partition into the membrane in a "stepwise" manner, in which an acid side chain positioned immediately adjacent to the membrane forms a hydrogen-bonded pair with a nearby acid side chain, and the segment of the polypeptide containing this pair, having assumed a lipophilic ⁇ -helical conformation, enters the membrane.
  • the spacing between carboxylic acid side chains is of particular importance.
  • drugs are to be attached at sites along the polypeptide chain, rather than only at the termini, they must be located so as not to interfere with said spacing and pairing between side chain carboxylates. Thus, random attachment of drugs, as well as random spacing of non-acid side chain residues, including glutamic esters, should be avoided.
  • additional motive force for unidirectional transport is provided by ionization and hydration of the side-chain carboxyls once ttie polypeptide spans the membrane and encounters the higher-pH cellular cytosol.
  • a polypeptide having a high percentage of side chain carboxyls is expected to provide a high driving force to transport an attached compound across the membrane.
  • one preferred class of polypeptides for use in the present invention includes those having 30%-100%, and preferably 50%-100%, glutamic acid content, and having an initiator sequence as described herein at one terminus.
  • 100% glutamic acid content refers to the central chain of the polypeptide, excluding the initiator moiety.
  • the compound to be transported is attached at or near the other terminus of the polypeptide.
  • the pH at which the polypeptide component, or a segment thereof, converts between its hydrophilic and lipophilic forms, and is effective to partition from an aqueous to a lipid phase, or vice versa is referred to as the "transition pH".
  • this conversion should occur at a pH greater than or approximately equal to the pH of the low-pH environment, and less than the pH of the higher-pH aqueous compartment.
  • Effective transition pH varies for different applications.
  • the bacterium H. pylori a major cause of ulcers, exists in a very acidic environment in the stomach.
  • This environment may be specifically targeted, as discussed below, by the use of a polymer composition which is effective to partition into a lipid layer at a pH of about 4.5 or less.
  • the polymer composition exists in its lipophilic form at a pH safely achievable in the extracellular lipid matrix of the stratum corneum, and b) the polymer composition converts to its hydrophilic form on contacting the aqueous environment underlying the stratum corneum.
  • a transition pH in the range of about 5 to 6.5 generally satisfies these two criteria.
  • a polymer composition as described herein can function as a molecular engine, pulling the relatively polar compound into and through the endosomal membrane.
  • the motive force exerted by the engine is generated as the non-ionic lipophilic ⁇ -helical polypeptide undergoes ionization and solvation at the cytosolic face of the endosomal membrane, as illustrated in Figure 7.
  • This motive force is, in part, a function of the difference between the transition pH and the pH of the cytosol.
  • the polymer composition used for endosome-to-cytosol transport may have a fairly high transition pH, such as in the range of 6.4 to 6.8.
  • the transition pH should generally be lower.
  • transition pH is too low, endosome/lysosome fusion can occur before the polypeptide engine converts to its lipophilic form and enters the encompassing membrane, leading to enzymatic degradation of the polypeptide (assuming it comprises L-amino acids) before it can carry out its transport function.
  • the pH of the endosome is typically in the range of about 5 to 6.
  • a minimum transition pH in this range is preferred for compositions assembled from L-amino acids and intended for endocytotic entry. From the above, it is seen that the optimal transition pH can vary substantially depending on the compound to be delivered and the compartment from and into which it is to be delivered.
  • Non-Acid Amino Acids The results in Table 2C demonstrate that selection of non-acid amino acid moieties can also substantially affect the transition pH. For example, replacing leucines with phenylalanines reduces the transition pH. As shown in Table 2C, the transition pH also is reduced further by each replacement in the series: leucine -> valine -> alanine -> glycine (data for glycine not shown). Thus, one simple method of adjusting the transition pH over a broad range is by progressively replacing leucine residues (which give high transition pH values) with alanines. It is also seen that leucines give about the same transition pH values as norleucines.
  • V valine
  • Table 2 shows a selection of transition pH's attained in an n-octanol/water partitioning system by varying the structural features discussed above. By further adjusting these parameters, it is possible to prepare polypeptides with still higher or lower transition pH's.
  • a composition with a transition pH of 4 or lower may be prepared by including a high proportion of aspartic acid and alanine subunits in a relatively short polypeptide.
  • transition pH's determined from octanol/water or pentanol/water partitioning studies provide a useful guideline for selecting appropriate polypeptides for delivery of a compound to a selected environment.
  • additional factors may influence the behavior of the polypeptide in a cellular environment.
  • Studies have suggested, for example, that polypeptides containing high levels of leucine may bind to serum proteins. It has also been demonstrated herein that a polypeptide having local regions of lipophilicity at the termini may undergo a transition effective for cell entry in vitro or in vivo at a pH higher than that shown in bulk partitioning studies.
  • a candidate polypeptide may also be tested for binding to serum proteins by performing electrophoresis on agarose gels with and without added serum albumin.
  • One or more drugs or other compounds can be attached to the polypeptide through sites at the termini and/or at a limited number of selected sites within the chain. As discussed above, random attachment or high loading of compounds along the chain can impede partitioning into the membrane, especially for high acid side chain polypeptides.
  • Figure 9 shows representative linkage positions and types which are both convenient and effective for a substantial variety of compounds and polymer compositions. Shown, from left to right, are amide and carbamate links to lysine, an ester link to glutamic acid, and disulfide, thioether, and amide links to cysteine.
  • linkages selected from amide, carbamate, ester, thioether, disulfide, and hydrazone are typically easy to form and suitable for most applications. Ester and disulfide linkages are especially preferred if the linkage is to be readily cleaved in the cytosol after delivery of the compound.
  • Figure 11 illustrates representative starting materials and products of such linkages.
  • Examples 2-7 describe the linkage of representative compounds, including cyclosporin, TaxolTM (paclitaxel), and a Morpholino antisense oligomer (Summerton and Weller) to representative polymers via these linkage types.
  • Cyclosporin is linked to a polypeptide via a disulfide linkage (Example 3A), a thioether linkage (Example 4A), a carbamate linkage (Example 5), and an amide or thioether linkage (Example 6).
  • TaxolTM is conveniently linked to the polypeptide via its C-7 hydroxyl group.
  • cyclosporin A metabolite 17, which has a primary hydroxyl group (Eberle and Nuninger, 1992). In both these cases, the linkage does not involve known active sites of the molecule.
  • Other drugs that may be used in applications described below, such as metronidazole or doxorubicin, also have primary hydroxyl groups for convenient attachment. In general, numerous functional groups (hydroxyl, amino, halogen, etc.) may be used for attachment. Groups which are not known to be part of an active site of the compound are preferred, particularly if the polypeptide or any portion thereof is to remain attached to the compound after delivery.
  • Example 8 describes representative methods for purification of polymer-compound products, as well as methods for structural analysis of said products.
  • the compound/polypeptide ratio is preferably 5: 1 or less, and more preferably 1:1 for large and/or polar compounds.
  • the compound is preferably attached at or near a terminus of the polypeptide; most preferably, one terminus includes an initiator moiety and the other an attached compound.
  • transport may be enhanced by attaching multiple, e.g. 2 to 5, polypeptide carriers of the invention to a single compound molecule.
  • the carriers are preferably attached to the same region of the molecule to be transported, e.g. at or near the nucleic acid binding polymer.
  • Example 9 describes a convenient procedure for carrying out such a partitioning study and obtaining a transition pH value for a representative polymer composition. In this procedure, a polypeptide tagged with fluorescein is partitioned between n-octanol or n-pentanol and aqueous buffers of various pH, and the absorbance of each phase is measured.
  • Figures 14A-F show plots of these absorbance values as a function of pH for six representative fluorescein- tagged polypeptide compositions of the invention, where the six different polypeptides exhibit octanol/water transition pH values ranging from 5.1 to 6.9.
  • Direct Entry In screening polymer compositions for delivery of a selected compound, direct transmembrane passage can be assessed by brief stepwise reduction of the pH of the extracellular medium, which emulates the progressive pH reduction which occurs in an endosome due to the action of proton pumps embedded in its membrane. The process is also representative of direct entry in vivo when the pH of the extracellular medium is lower than the pH of the cytosol of the cells.
  • fluorescence microscopy shows a perinuclear punctate pattern in the cell.
  • a fluorescent compound enters the cytosol one sees diffuse fluorescence throughout the cell.
  • linking a fluorescent tag to the compound to be transported into the cytosol allows one to readily assess subcellular localization of the polymer-compound product after its addition to cultured cells.
  • the linked fluorescent tag should have minimal impact on the transport process. Specifically, it should be relatively small, and it should be reasonably soluble in octanol and lipid layers at low pH and reasonably soluble in aqueous solutions at neutral pH and above.
  • Two fluorescent tags suitable for most such applications are 5-carboxy-fluorescein and 7-dimethylaminocoumarin-4-acetic acid (obtained from Molecular Probes Inc., Eugene, Oregon).
  • Example 10A describes a direct-cell-entry experiment with cultured eukaryotic cells.
  • cell entry via endocytosis may be assessed by methods such as that described in Example 10.
  • FIG. 2 illustrates the entry of a polymer-compound composition of the invention into the cytosol of a eukaryotic cell by endocytotic uptake. From the figure, it is seen that entry into the cytosol of eukaryotic cells comprises multiple steps, principal of which are the initial endocytotic uptake and the subsequent transmembrane passage from the acidified endosome to the neutral cytosol.
  • cytosolic entry of such polypeptides via an endocytotic route be assessed by a functional assay for the drug component of the polypeptide- drug product, as described in Example 10.
  • initial endocytotic entry studies directed toward demonstrating and optimizing endocytotic delivery of selected polypeptides, can be carried out with polymers assembled from D-amino acids, which are not degraded by lysosomal enzymes.
  • Studies in support of the invention have shown that D- and L-polypeptides having the same sequence exhibit the same partitioning and membrane transport properties.
  • the compound linked to the polypeptide is a sequence-specific nucleic acid-binding polymer, specifically, a nonionic antisense oligomer (Summerton and Weller) targeted against a specific messenger RNA for firefly luciferase, coded by a plasmid contained in the treated cells. If the antisense oligomer gains access to the cytosolic compartment of the transfected cells, a significant reduction in luciferase activity upon dexamethasone induction, relative to untreated cells, should be observed.
  • the polymer-compound composition was contacted with cells for a period of time appropriate for endocytotic uptake, about 5 hours.
  • the polypeptide alone failed to inhibit its targeted messenger RNA, presumably because it was unable to enter the cytosol of the cells.
  • the same antisense oligo linked to the polymer of the invention inhibited luciferase activity by about 31 % relative to the control, suggesting successful delivery of the compound into the cytosolic compartment.
  • Enhancement of Endocytosis bv an Attached Affinity Moiety The polymer compositions of the invention carry a relatively high density of negative charges in the essentially neutral extracellular medium. Further, due in substantial part to the sialic acid residues on the glycocalyx, the outer surface of a eukaryotic cell also typically carries a substantial density of negative charges. Possibly because of electrostatic repulsion between the like-charged polymer and cell surface, the rate of cell entry of some polymer-compound products via fluid-phase endocytosis appears to be relatively slow. Further experimental results, described below, suggest that the rate of endocytosis can often be enhanced by using a moiety with an affinity for cell surfaces, such as a lipid anchor (i.e.
  • lipid anchors likely serve to increase the concentration of the polymer-compound at the cell surface, such that upon invagination of the cell membrane to form the endosome, a larger amount of polymer- compound is enveloped therein than would otherwise be the case.
  • the linkage be cleavable in the cytosol of cells so that the polymer-compound is released free in the cytosol rather than remaining linked to the lipid anchor embedded in the endosomal membrane.
  • a tetracationic lipid anchor (TransfectamTM, from Promega Corp., Madison, WI, USA) was added to enhance the endocytosis step by complexing with the polyanionic polymer composition. This is illustrated in Fig. 12, where the lipid anchor is shown at 30. In this case, luciferase activity was inhibited by about 68% relative to the control, compared to 31%, above, without the lipid anchor.
  • Figure 12 illustrates the likely role that a lipid anchor such as TransfectamTM plays in enhancing the initial endocytotic step, followed by its dissociation from the polymer when the polymer converts to its low-pH non-ionic form in the late-stage endosome.
  • a lipid anchor such as TransfectamTM plays in enhancing the initial endocytotic step, followed by its dissociation from the polymer when the polymer converts to its low-pH non-ionic form in the late-stage endosome.
  • lipophilic drugs e.g., TaxolTM and cyclosporin A
  • Endocytosis may also be promoted by attachment of a receptor signal, or ligand, to the polypeptide-compound composition.
  • a receptor signal or ligand
  • the ligand, attached or complexed to the composition targeted for uptake is capable of binding to a receptor on the cell surface.
  • a ligand may be used to enhance general endocytotic uptake, or to target specific cell types, as discussed further below.
  • the polymers of the invention can be used to target delivery of attached compounds to specific cells or locations within the body.
  • the specificity of targeting may be controlled by exploiting the pH differential between the cytosol and extracellular medium, or by the use of targeted cell surface receptors.
  • the very acidic environment of the stomach may be specifically exploited, as in treatment of H. pylori infection, by using a polymer composition effective to partition into a membrane at a pH less than about 4.5.
  • Other compartments in the body including the endosomal compartments of eukaryotic cells, which have a pH in the range of 4.5-6.5, do not reach this low pH and so fail to convert the polymer composition to a lipophilic state required for passage across their lipid layers.
  • a polypeptide with a higher transition pH e.g. up to 6.5 or 7.0
  • H. pylori cells would still enter H. pylori cells, it could also enter other cells via endocytosis, with a resulting decrease in selectivity.
  • a composition In order to penetrate a bacterial cell, a composition must be able to cross the outer membrane (in gram negative bacteria), the cell wall and the inner plasma membrane.
  • the bacterial cell wall generally excludes entry of globular compounds of 2000 Da or more.
  • Studies in support of the invention indicate that the polypeptide compositions described herein, though generally of higher molecular weight than 2000 Da, successfully penetrate H. pylon cells. Because the compositions, at low pH, are primarily linear rods rather than globular, it is postulated that they penetrate the cell wall via a reptation mechanism.
  • Drugs that have been used to target H. pylori include amoxicillin, bismuth salts, metronidazoiine, omeprazole, clarithromycin and tetracyclines. Therapy with a single drug has not always been effective, due to pH effects and/or inadequate concentrations of the drug at the infection site. Triple-drug therapy is therefore frequently recommended, using the combination of a bismuth salt (e.g. bismuth subsalicylate), an anti-infective (e.g. metronidazole), and either tetracycline or amoxicillin. (See e.g.
  • This application is one in which benefits could be gained by maintaining the polypeptide intact after delivery of the compound. Specifically, the intact polypeptide-compound composition is unlikely to be absorbed into the stomach or intestinal wall, thus preventing delivery of the attached compound to non-targeted body sites.
  • a polypeptide composed of D-amino acids is not degraded by proteolytic enzymes and thus would be preferred for this purpose.
  • a relatively stable compound-polypeptide linkage, such as an amide, would also be preferred in this instance.
  • the extracellular environment within such solid tumors has been shown to be more acidic than normal tissue (e.g. Kennedy, Tannock, Vaupel) due to factors such as the production of lactic acid under anaerobic conditions.
  • Measured pH's are typically in the range of 6.0 - 7.0, or, in general, about 0.3 to 1.0 units less than in the corresponding normal tissue (Vaupel).
  • the extracellular medium of tumor masses also appear to be of lower pH than the intracellular compartment of the tumor cells themselves (Newell). For example, measurements of pH by 3I P NMR, which measures primarily the intracellular pH, showed that brain tumors and sarcomas had a higher intracellular pH than the corresponding normal tissues (Vaupel).
  • hypoxic tumor cells by virtue of their low pH environment, may be targeted by an antineoplastic drug linked to a polypeptide which is effective to partition into a membrane at the pH present in this extracellular environment.
  • antineoplastic drug linked to a polypeptide which is effective to partition into a membrane at the pH present in this extracellular environment.
  • drugs include, for example, cis-platin, antimetabolites such as methotrexate and fluorouracil, topoisomerase inhibitors such as doxorubicin, alkylating agents such as cyclophosphamide and chlorambucil, and tubulin-binding plant alkaloids such as vinblastine, vincristine, docetaxel, and paclitaxel OTaxolTM).
  • direct cell entry is generally much faster than endocytosis.
  • transport occurs within a matter of minutes; see, e.g., Example 10.
  • Appreciable endocytotic entry on the other hand, generally takes many hours. Therefore, endocytotic uptake by non-targeted cells would be minimal relative to direct entry into the targeted cancer cells, driven by the existing pH differential between the extracellular medium and intracellular compartment.
  • Tooth decay (dental caries) is promoted by the production of acid (acidogenesis) by bacteria in breaking down carbohydrates.
  • acid acid
  • Igarashi Studies of pH in sucrose-induced plaque (Igarashi) showed a minimum pH of 4.6 ⁇ 0.2 in 2-day-old plaque; the pH increased to 5.7 after 21 days.
  • the dominant bacteria were Streptococci ( > 50% of total) and Actinomyces (> 10% of total).
  • bacteria taken from caries-active sites produced a final pH of 4.2 or less in sugar broth (van Houte).
  • Antibacterial agents may be targeted to the site of decay or potential decay according to the methods described above, using polypeptides effective to partition into a membrane at a pH in the range of about 4.5 to 6.5, and preferably in the range of about 4.5 to 5.5.
  • Suitable antibacterials that have been used in the treatment of dental caries include chlorhexidine, triclosan (Friedman, Bouwsma), xylitol, antibacterial enzymes, and amine fluorides such as 9- octadecen- 1-amine hydrofluoride and 1-hexadecylamine hydrofluoride.
  • Drug delivery via endocytosis may be promoted by linking or complexing the polymer- drug composition to a suitable ligand, or receptor signal.
  • a suitable ligand or receptor signal.
  • Use of such a ligand can provide cell-targeting versatility because certain receptor signals, such as mannose-6-phosphate, biotin, folic acid, and other water soluble vitamins, afford delivery to many cell types (Ludwig) and thus may be used to promote cell entry in many applications. Others may be used to focus delivery into one or a few specific cell types, as discussed further below.
  • linkage between the polymer-drug composition and the ligand be cleavable, so that after transport the polymer-drug can be released free in the cytosol.
  • Water-soluble vitamins such as riboflavin, thiamine, nicotinic acid and folic acid can be used to target cell surface receptors. These compounds are believed to be taken into the cell by potocytosis, a variation of endocytosis that is specialized for the uptake of small molecules.
  • potocytosis receptors are present in small (approx. 50 nm diameter) pits or vesicles on the cell surface, known as caveolae. These caveolae remain at or near the cell surface, going through cycles of opening and closing. Upon closing, proton pumps within the membrane produce a pH of about 6.0 within the caveola.
  • the vitamin, with attached polymer is released from the receptor (Anderson).
  • the polymer-compound composition then inserts into the lipid membrane, after undergoing a pH-dependent transition into a lipophilic conformation, and thereafter transports into the cytosol, according to the mechanism described herein.
  • this pathway does not typically involve as large a pH differential as the endocytotic pathway, it presents certain advantages.
  • the caveolae do not merge with pre- lysosomes, and thus potential exposure of the composition to degradative enzymes is avoided.
  • the water-soluble vitamin is generally released from the polymer when exposed to low pH within the caveola, allowing free polymer-drug to enter the cytosol.
  • Cells that may be specifically targeted by receptor signals include liver cells (hepatocytes), whose surfaces contain receptors that specifically recognize galactose-terminal glycoproteins. Many malignant cells overexpress certain receptors, and thus it may be possible to selectively target such cells, as has been reported using folate (Mathias) and epidermal growth factor (Deshpande). D-cycloserine has been reported to facilitate transport through the cytoplasmic membrane of bacteria (Chopra, Rapaport).
  • BBB blood-brain barrier
  • the blood-brain barrier (BBB) which regulates the exchange of materials between the bloodstream and central nervous system, presents a daunting barrier to drug transport.
  • the endothelial cells of cerebral capillaries contain "tight junctions", circumferential bands around a cell that are in close contact with adjacent cells. These junctions prevent transport between cells, and thus, for effective transport, compounds must pass through the endothelial cells themselves. Studies directed to such transport of peptides show that lipophilicity is probably the most important factor in promoting transport of a peptide across the BBB (Banga).
  • compounds may be transported across a cell by transcytosis.
  • polarized endothelial cells i.e., cells having distinct apical and basolateral membranes
  • the compound is first taken through the apical membrane in the inner capillary wall into a transcytotic vesicle.
  • a transcytotic vesicle typically attains a pH of about 6.0.
  • the vesicle transfers the compound to the basolateral membrane of the endothelial cell, on the outer capillary wall.
  • the compound is then expelled from the transcytotic vesicle, thereby releasing the compound outside of the cell and the capillary.
  • transportation of a compound across the blood- brain barrier may be effected by linking the compound to a polypeptide of the present invention which is effective to partition into a membrane at a pH within a selected range, as described below.
  • the composition is preferably further linked to a receptor signal, e.g. hypoxanthine or inosine, effective to bind to a receptor on the surface of a cerebral endothelial cell. After delivery to the cerebral bloodstream, the composition is transported across the capillary wall via transcytosis, as described above.
  • the composition is then available to be taken up by a brain cell via endocytosis and released into the cell cytosol, according to the mechanisms described herein.
  • the endosome is expected to have a pH of about 5.0.
  • the pH at which the polymer is effective to traverse a cell membrane should be between 5.0 and 6.0, or, more generally, below the pH of the transcytotic vesicle in an endothelial cell and above that of die endocytotic vesicle in the ultimate target cell. If the pH were above 6.0, in this case, the polymer would assume its lipophilic conformation within the transcytotic vesicle and penetrate its membrane, thus entering the endothelial cell instead of the targeted brain cell.
  • the principal permeability barrier of the skin consists of cornified epithelial cells surrounded by an extracellular lipid matrix.
  • the extracellular lipid matrix which constitutes the principal route for passage of compounds through the skin, consists of lipids (ceramides, cholesterol, free fatty acids, and cholesteryl sulfate) ordered in multiple sheets of lipid bilayers, with water and other polar compounds dispersed between the polar faces of the stacked bilayers.
  • This structure of multiple alternating polar and nonpolar layers presents a daunting barrier to penetration of both hydrophilic and lipophilic compounds.
  • Each of the multiple lipid bilayers acts as a substantial barrier to passage of hydrophilic compounds, while each of the layers of water and other polar compounds between the polar faces of the stacked lipid bilayers acts as a substantial barrier to passage of lipophilic compounds.
  • the stacked bilayers of the extracellular lipid matrix can be disordered by a variety of penetration enhancers, such as Azone (l-dodecyl-azacycloheptan-2-one), unsaturated long-chain alcohols, and unsaturated long-chain fatty acids, such as oleic and linoleic acid (Aungst et al, 1986; Rehfeld and Elias, 1982; Golden et al, 1986; Goodman and
  • a suitable polypeptide is contacted with the surface of the epidermis in its low-pH lipid-soluble form. Because the pH of the skin surface is typically in the range of 5.0 -5.5 (see, for example, Yosipovitch, Korting), polypeptides with transition pH's in this range or higher will exist predominantly in their low-pH form on the skin surface. The peptide, in its lipophilic form, is able to diffuse through the lipid layer of the extracellular matrix of the stratum corneum.
  • composition Upon contact with the aqueous compartment underlying the stratum corneum, the composition is actively drawn into this compartment by virtue of progressive ionization and solvation of the polypeptide chain at the near-neutral pH of this compartment. Both hydrophilic and lipophilic compounds may be transported.
  • a preferred polypeptide for this application undergoes a reversible pH-dependent transition at pH between about 5 and 6.5.
  • the polymer composition of the instant invention is preferably used in concert with one or more suitable penetration enhancers, as described above.
  • Lipophilic acids such as oleic or linoleic acid, may be added to maintain the polymer in its low-pH lipophilic form during passage across the lipid layer. These fatty acids also serve as penetration enhancers, as noted above.
  • Example 11 illustrates the use of a representative polymer composition of the invention to enhance transdermal delivery of the antirejection drug, cyclosporin A.
  • the polypeptides of the invention are likely decomposed by proteasomes after entering the cytosol of the cell, thus freeing the transported drug or other compound. This release of the drug is shown, for example, in Fig. 2.
  • One example is in delivery to the stomach, as described for the eradication of H. pylori, above.
  • Another is in the selective delivery of highly toxic drugs, e.g. to tumor cells. Because the transporting action of the polypeptide is unidirectional, proceeding from lower to higher p ⁇ , the polypeptide attached to the drug prevents the drug from diffusing back out of the target cell to which it was first delivered.
  • the intact polypeptide may also prevent or minimize exportation of the delivered drug by cellular processes. Many cells express a glycoprotein which serves as a drug-efflux pump, transporting drug compounds out of the cell. The attached polypeptide will likely interfere with this process and reduce or prevent exportation of the attached drug.
  • the polypeptide may be prepared with one or more D-amino acids adjacent to the drug, and the remainder L- amino acids.
  • the L-amino acids will be removed by proteasomes, leaving a shortened polypeptide "tail" of D-amino acids attached to the drug.
  • Formulations containing the compositions of the invention may be in solid, semi-solid, or liquid dosage forms, such as, for example, tablets, capsules, powders, sustained-release formulations, solutions, suspensions, emulsions, suppositories, ointments, lotions, or aerosols, preferably in unit dosage forms suitable for simple administration of precise dosages.
  • Such formulations typically include a conventional pharmaceutical carrier or excipient and may additionally include other medicinal agents, carriers, or adjuvants.
  • the formulation will contain about 0.5% to 75% by weight of a compound or compounds of the invention, with the remainder consisting of suitable pharmaceutical excipients.
  • excipients include pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, talcum, cellulose, glucose, gelatin, sucrose, magnesium carbonate, and the like.
  • the composition may also contain minor amounts of non-toxic auxiliary compounds such as wetting agents, emulsifying agents, or buffers.
  • Liquid compositions can be prepared by dissolving or dispersing the polypeptide-compound composition (about 0.5% to about 20%), and optional pharmaceutical adjuvants, in a carrier, such as, for example, aqueous saline, aqueous dextrose, glycerol, or ethanol, to form a solution or suspension.
  • a carrier such as, for example, aqueous saline, aqueous dextrose, glycerol, or ethanol
  • composition may be administered to a subject by a variety of known routes, e.g., orally, transdermally, as described above, or parenterally, e.g., by intravenous, subcutaneous, intraperitoneal, or intramuscular injection.
  • the composition may be prepared as a solution, suspension, emulsion, or syrup, being supplied either in liquid form or a dried form suitable for hydration in water or normal saline.
  • Transdermal delivery typically involves the use of a transdermal "patch" which allows for slow delivery of compound to a selected skin region.
  • transdermal patch delivery systems are provided by U.S. Patent 4,655,766 (fluid-imbibing osmotically driven system), and U.S. Patent 5,004,610 (rate controlled transdermal delivery system).
  • transdermal delivery it may be desirable to include permeation enhancing compounds, as described in above.
  • permeation enhancing compounds as described in above.
  • Such formulations may be provided as occluded dressings to the region of interest, or may be provided in one or more of the transdermal patch configurations described above.
  • an injectable composition for parenteral administration will typically contain the composition of the invention in a suitable IV solution, such as sterile physiological salt solution.
  • a suitable IV solution such as sterile physiological salt solution.
  • the composition may also be formulated as a suspension in a lipid or phospholipid, in a liposomal suspension, or in an aqueous emulsion.
  • a synthesis resin is prepared so that ⁇ -alanine will comprise the C-terminal residue of the polypeptide, as illustrated in Figure 10.
  • One gram of 1 % crosslinked polystyrene resin containing 0.7 mMol p-alkoxybenzyl alcohol, I (Cat. # A3039, Sigma Chem. Co., St. Louis, MO) is dissolved in 8 ml of N-methylpyrrolidinone (NMP), and 0.62 g of fluorenylmethoxy- carbonyl (FMOC) ⁇ -alanine 2 is added, followed by 316 ⁇ l of N,N'-diisopropyl carbodiimide and 41 ⁇ l N-methylimidazole. This slurry is incubated with agitation at 37°C for 100 minutes, 5
  • a suitably-protected lysine or cysteine is typically inco ⁇ orated at the selected attachment position(s).
  • the drug can be attached to the resultant amine or sulfhydryl moiety, as described in Example 2, to afford linkages illustrated in Figure 9.
  • certain drugs may be linked, suitably protected if necessary, to the gamma carboxyl of glutamic acid, or the ⁇ -carboxyl of aspartic acid, via an ester linkage.
  • the resultant amino acid is then inco ⁇ orated into the polypeptide chain at one or more selected positions.
  • Such drugs must have structures which survive the conditions used to assemble the polypeptide, to cleave the polypeptide from the synthesis resin, and to deprotect the side chains.
  • the completed polypeptide can be cleaved from the resin by washing the resin with CH 2 C1 2 , draining, and adding, per gram of resin, a solution comprising 10 ml trifluoroacetic acid (TFA), 10 ml CH 2 C1 2 , and 400 mg dithioerythritol. After 20 minutes the cleavage solution is drained into a flask and the CH 2 C1 2 removed by evaporation. Thereafter, 5 to 10 ml TFA is added and the solution held at 43 °C for 4 hrs to effect removal of protective groups. Workup of the deprotected polypeptide generally entails ether precipitation, thorough washing of the precipitate with ether, and drying. It should be appreciated that there are a variety of other peptide assembly methods known in the art which are also suitable for preparing the composition of the invention.
  • EXAMPLE 2 Attaching Compounds to Polypeptide on Column
  • drugs or other compounds e.g., a fluorescent tag for partitioning and cell entry studies
  • the compound to be attached is typically activated, by methods known in the art, to produce an active ester or active carbonate moiety effective to form an amide or carbamate linkage, respectively, to the polypeptide, such as illustrated in Figure 9.
  • EXAMPLE 3 Disulfide Linkage to Cvsteine Residue of Polypeptide
  • the deprotected polypeptide is reacted with 2,2'-dipyridyl disulfide, and the drug containing a sulfhydryl moiety is added to form the desired disulfide link, as illustrated in Figures 7 and 11.
  • a particular advantage of such a disulfide linkage is that it is relatively stable in the extracellular compartment and within endosomes, but after transport across the endosomal membrane it is readily cleaved in the cytosolic compartment. The following specific examples illustrate applications of this method. 3A.
  • the terminal FMOC group is cleaved by treatment with 20% piperidine in NMP and a terminal S-tritylated FMOC-cysteine is introduced by the method of Example IA.
  • the polypeptide is cleaved from the column, deprotected, precipitated, and washed as per Example IC.
  • the solution is diluted with an equal volume of acetonitrile and treated with sufficient dipyridyl disulfide to create a 0.4 molar solution. After stirring for 2 hours at room temperature, the mixture is diluted with an equal volume of water and washed with sufficient water-saturated ethyl acetate to remove excess dipyridyl disulfide.
  • the solution is partially evaporated to - 35 - remove residual ethyl acetate and the product isolated by passage of the solution through Amberchrome (TosoHaas) followed by elution with 0-80% acetonitrile with a 0.01% triethyl ⁇ amine buffer followed by lyophilization.
  • Amberchrome TosoHaas
  • cyclosporin A metabolite 17 which has a primary hydroxyl group (Eberle and Nuninger, 1992), is treated with succinic anhydride using the method of Chen and Tai (1995), and the acid is converted to the activated ester with N-hydroxysuccinimide and 1- (3-dimethylaminopropyl)-3-ethylcarbodiimide methiodide (Aldrich).
  • the product is dissolved in DMF and treated with 2-aminoethanethiol hydrochloride and triethylamine.
  • the reaction mixture is diluted with dichloromethane and washed with 0.1M citric acid to remove excess reagents.
  • the product is obtained by evaporation.
  • the residue is dissolved in DMF and treated with the pyridyl polypeptide disulfide from the previous paragraph.
  • the product is isolated by evaporative removal of the DMF followed by reverse phase purification on Amberchrome eluting with 0-80% acetonitrile in a 0.01 % triethylamine buffer followed by lyophilization.
  • the product is dissolved in 0. IM Na ⁇ HPO ⁇ and washed well with water-saturated ethyl acetate.
  • formic acid is added to precipitate the product, which is washed well with water containing 0.1 % formic acid, then dried thoroughly under high vacuum.
  • TaxolTM is converted into 7-glutaryl TaxolTM by the method of Gueritte-Vogelein et al. (1991).
  • the carboxyl group of this species is activated as the N- hydroxysuccinimde ester with N-hydroxysuccinimde using l-(3-dimethylaminopropyl)-3- ethylcarbodiimide methiodide (Aldrich) and 4-dimethylaminopyridine in dichloromethane.
  • Aldrich l-(3-dimethylaminopropyl)-3- ethylcarbodiimide methiodide
  • the product is dissolved in DMF and treated with 2-aminoethanethiol hydrochloride and triethylamine.
  • the reaction mixture is diluted with dichloromethane and washed with 0.1M citric acid to remove excess reagents.
  • the product is obtained by evaporation.
  • the residue is dissolved in DMF and treated with the pyridyl polypeptide disulfide from above.
  • the product is isolated as per the cyclosporin example.
  • EXAMPLE 4 Thioether Linkage to Cvsteine Residue of Polypeptide 4A.
  • Preparation of a thioether-linked polypeptide-cvclosporin conjugate Cyclosporin A metabolite 17 (see Example 3A) is treated with chloroacetic anhydride in 1:1 dichloromethane/ pyridine to form an acid chloride (see Fig. 11). The excess reagent is quenched by the addition of water, and the solvent is evaporated. The residue is dissolved in ethyl acetate and washed with 0.1M citric acid, 0.1M sodium bicarbonate, and brine, ttien evaporated. The residue is redissolved in DMF.
  • Example 3A The product is dissolved in DMF, mixed with the acrylamide species from the paragraph above, and TCEP is added. After stirring at room temperature, the product is isolated as in Example 3A.
  • the ether precipitate of a deprotected polypeptide containing one or more amine moieties, prepared as in example IC, is dissolved in aqueous 0.1 M Na ⁇ PO,,.
  • Formic acid is added sufficient to precipitate the polypeptide, and the precipitate is collected by centrifugation and washed twice with 0.1 % formic acid in water.
  • the polypeptide precipitate is then dried under high vacuum overnight.
  • the compound to be linked to the polypeptide containing at least one hydroxyl moiety, is activated by reacting with 3 equivalents of bis-nitrophenyl carbonate and 0.1 equivalent of triethylamine in NMP. After activation, the excess bis-nitrophenyl carbonate is removed and the active carbonate product (see Fig. 11) then reacted with 0.5 to 2 equivalent of the polypeptide composition, prepared as described above, in NMP. Good coupling is generally achieved by incubation for 12 to 72 hours at 43 °C.
  • FMOC derivative of glycine and l-(3-dimethylaminopropyl)-3-ethylcarbodiimide methiodide (Aldrich) in dichloromethane in the presence of 4-dimethylaminopyridine catalyst.
  • the reaction mixture is washed with acid and base to remove excess reagents and the product chromato ⁇ graphed on silica gel using 0-2% methanol in chloroform.
  • Removal of the FMOC group is accomplished by treatment of the FMOC -glycyl cyclosporin with 20% triethylamine in DMF at 50 degrees C for one hour. The triethylamine is removed by evaporation under vacuum and the free amino derivative mixed with the activated polypeptide above.
  • Example 3A The solution is evaporated in vacuo to a minimum volume and allowed to sit at 42 degrees for 24 hours.
  • the product is isolated as in Example 3A. 2.
  • a high glutamic acid content, random length peptide with C-terminal initiator region, is prepared as follows.
  • Chlorotrityl resin preloaded with N-Fmoc-/S-aIanine (Novabiochem, LaJolla CA) is reacted with N- ⁇ -Fmoc-L-glutamic acid ⁇ -t-butyl ester and N- ⁇ -Fmoc-L-leucine using HBTU
  • the resin bound initiator peptide is treated six times with the carboxyanhydride of L- glutamic acid ⁇ -(4-methoxy benzyl) ester (Hanabusa et al., 1984) in DMF, using an anhydride to initiator ratio of about 10 to 1, which is sufficient to add about 40-50 protected glutamic acid residues to the peptide.
  • the N-terminus is then reacted with excess bis-(p-nitrophenyl) ester of glutaric acid.
  • the peptide is cleaved from the resin and deprotected as described in Example IC.
  • the residue after evaporation is dissolved in dichloromethane and washed with water to remove traces of trifluoroacetic acid.
  • the isolated polyacid is dissolved in DMF and treated with TaxolTM-7-alanine (Mathew et al., 1992).
  • the reaction mixture is diluted with pH 7.0 phosphate buffer and the product purified by ion exchange chromatography on Q-Sepharose (Pharmacia, Piscataway, NJ) using a 0-1.0 M NaCl gradient.
  • the material is then isolated as the sodium salt by adso ⁇ tion to an Amberchrome SD (TosoHaas), elution with a 0-80% acetonitrile gradient, and lyophilization.
  • TaxolTM is converted into 7-glutaryl TaxolTM by the method of Gueritte-Vogelein et al. (1991).
  • the carboxyl group of this species is activated as the N- hydroxysuccinimde ester with N-hydroxysuccinimde using l-(3-dimethylaminopropyl)-3-ethyl- carbodiimide methiodide (Aldrich) and 4-dimethylaminopyridine in dichloromethane. After washing to remove excess reagents and byproducts, the ester is isolated by evaporation and mixed with the polypeptide in DMF.
  • a * Mo ⁇ holino 6-benzoyladenine
  • C * Mo ⁇ holino 6-benzoylcytosine
  • G" Mo ⁇ holino 6-phenylacetylguanine
  • U Mo ⁇ holino uracil
  • the Mo ⁇ holino antisense oligo with succinate linker 3 (2 ⁇ Mole), prepared above, is placed in a 0.75 ml vial containing a 3 mm magnetic stir bar with 31 mg (6 ⁇ Mole) of the deprotected 44-amino acid polypeptide R,-NH 2 , as defined above, shown at 4, precipitated from aqueous solution as described in Example 2B(iii), having a free amine moiety on the N- terminus.
  • DMF 150 ⁇ L
  • the reaction mixture is then incubated at 43 °C for 72 hours, diluted with 200 ⁇ L NMP, and transferred to a 2 ml screw cap vial.
  • Procedure 2 ( Figure 13B): In this procedure, the activated succinate linker is added to die polypeptide, and the adduct is reacted with the Mo ⁇ holino antisense oligo. Accordingly, an NMP suspension of 180 mg of synthesis resin/polypeptide, prepared as in Example 1, is treated with 20% piperidine in NMP, then washed repeatedly with NMP. Bis(p-nitrophenyl) succinate (150 mg) is dissolved in 0.9 ml NMP, added to a short fritted column containing the resin/polypeptide preparation and incubated 2 hours at 43°C.
  • reaction mixture is diluted with 200 ⁇ L NMP and transferred to a 2 ml screw cap vial. 600 ⁇ L of con NH 4 OH is added and the solution incubated 18 hours at 43 °C to deprotect the purine and pyrimidine bases of the Mo ⁇ holino antisense oligo.
  • product (5) is purified as described in Example 8C.
  • Procedure 3 Mo ⁇ holino antisense oligo is prepared wherein the first subunit, containing a 5'-SH, is linked to the synthesis resin via a disulfide bond, according to methods known in the art.
  • the 2vuthMo ⁇ holino oligo, with bases still protected, is cleaved from the synthesis resin using N-methyl pyrrolidinone containing 1 % wt/vol dithiothreitol and 5% v/v triethylamine.
  • the eluted oligo is precipitated with t-butylmethyl ether and the pellet washed twice with t-butylmethyl ether and then dried under high vacuum to give base-protected Mo ⁇ holino oligo with 5 * -SH and a 3 'acetyl.
  • the polypeptide is then cleaved and eluted from the resin with 49:49:2 TFA:CH 2 C1 2 :H 2 0, the CH 2 C1 2 is removed under aspirator vacuum, 98:2 TFA:H 2 0 is added, and the solution is incubated from 4 hours at 43 °C.
  • the polypeptide is then precipitated with t-butyimethyl ether and the pellet washed twice with t-butylmethyl ether and dried under high vacuum to give the chloroacetylated polypeptide shown in Figure 13C.
  • the Mo ⁇ holino oligo and polypeptide are coupled by adding 5 ⁇ mole 5'-SH Mo ⁇ holino oligo, 10 ⁇ mole chloroacetyl polypeptide, and 400 ⁇ mole N,N-dimethyl ethanolamine to 1 ml formamide, stirring until dissolution is complete, and then incubating at room temperature for 2 hours to give polypeptide-Mo ⁇ holino oligo joined by a thioether link, as shown in Procedure 3vuth, Figure 13C.
  • the bases of the Mo ⁇ holino oligo are deprotected by adding two volumes of concentration NH 4 OH to the reaction mixture and incubating 16 hours at 43 °C. After releasing excess NH 3 the product is purified by anion exchange chromatography, as described in Example 8C.
  • EXAMPLE 7A Amide Linkage to Carboxyl Moietv: This example describes the preparation of an amide-linked polypeptide-TaxolTM conjugate with a random length, high glutamic acid content peptide. A high glutamic acid content, random length peptide with C-terminal initiator region, is first prepared as follows.
  • Chlorotrityl resin preloaded with N-Fmoc-/5-alanine (Novabiochem) is reacted wim N- ⁇ - Fmoc-L-glutamic acid ⁇ -t-butyl ester and N- ⁇ -Fmoc-L-leucine using HBTU (Novabiochem) and diisopropylediylamide to produce Fmoc-Leu-Glu(OtBu)LeuLeu-NHCH 2 CH 2 COO-Resin.
  • the protected peptide is removed from the resin by treatment with 0.5% trifluoroacetic acid in dichloromethane.
  • the residue is redissolved in dichloromethane and treated with diphenyl ⁇ diazomethane (Kametani et al.) to produce the C-terminal diphenylmethyl ester, and the product is precipitated with hexane.
  • the residue is dissolved in dichloromethane and the product precipitated with ether/hexane.
  • the precipitate is dissolved in DMF containing 20% (v/v) piperidine. The solution is evaporated and the amino terminated initiator peptide is ready for elongation.
  • the initiator peptide is dissolved in DMF and reacted with the carboxyanhydride of L- glutamic acid 7-(4-methoxy benzyl) ester (Hanabusa et al).
  • the ratio of anhydride to initiator was about 60 to 1 , which is sufficient to add about 40-50 protected glutamic acid residues to the peptide.
  • the reaction was quenched by the addition of acetic anhydride, the solvent removed by evaporation, and the product purified by repetitive precipitations from dichloro ⁇ methane using ether/hexane.
  • the peptide is deprotected as described in Example IC, and prepared for conjugation by dissolution in 0.1 M Na 2 HP0 4 followed by precipitation with formic acid, as described in Example 5.
  • the polyacid is dissolved in DMF and treated with TaxolTM-7-alanine (Mathew W / 73
  • the reaction mixture is diluted with pH 7.0 phosphate buffer and the product purified by ion exchange chromatography on Q-Sepharose (Pharmacia) using a 0-1.0M NaCl gradient.
  • the material is then isolated as the sodium salt by adso ⁇ tion to an Anberchrome SD (TosoHaas), elution with a 0-80% acetonitrile gradient, and lyophilization.
  • N-terminal amine on the product thioester to form an amide linkage N-terminal amine on the product thioester to form an amide linkage.
  • Polypeptide compositions which partition well into octanol at reduced pH can often be effectively separated from undesired failure sequences generated during polypeptide assembly by partitioning between n- octanol and an aqueous buffer having a pH about 0.2 pH units below the transition pH of the polypeptide.
  • the purified polypeptide can be recovered from the octanol phase by partitioning into aqueous Na,HP0 4 and then precipitating with formic acid. Typically this purification procedure is carried out before attachment of the drug. This "purification-by-function" method has been found useful for preparations of quite long polypeptides containing a significant fraction of failure sequences.
  • a particularly versatile method for purifying polypeptide-drug products employs ion exchange chromatography followed by reverse phase chromatography, where the latter stage removes salt and provides additional purification. Use of this purification method is described for the polypeptide-Mo ⁇ holino antisense oligo products whose syntheses are described in Example 6C and whose structures are shown in Figure 13.
  • Polypeptide-Mo ⁇ holino oligo preparation is transferred to a rotovap flask, and excess ammonia is removed under aspirator vacuum. Approx. 10 ml of Tris acetate buffer (0.1 M Trizma base, acetic acid to pH 8) is added, and the solution is loaded onto a 2.5 cm by 15 cm column of Macro Prep Q anion exchange resin (BioRad Co ⁇ .). After loading, the column is washed (flow rate 5 ml/min) for 10 min. with Tris acetate buffer, followed by elution with Tris acetate buffer, increased linearly from 0 to 1.0 M in NaCl, over 40 min. In this system, the Mo ⁇ holino antisense oligo (monitored at ⁇ 54 nm) elutes in the first 15 minutes, and the polypeptide-Mo ⁇ holino product elutes at about 30 minutes.
  • Tris acetate buffer 0.1 M Trizma base, acetic acid to pH
  • polypeptide-Mo ⁇ holino peak is collected and then desalted on a 2.5 cm by 15 cm column of 50 ⁇ m polypropylene (Polysciences Co ⁇ .). This reverse phase column is washed for 15 min. with 1 % con NH 4 OH and then eluted with a 0 to 80% acetonitrile gradient, 1 % in cone. NH 4 OH, over 40 minutes at a flow rate of 5 ml/min. Fractions containing the polypeptide-Mo ⁇ holino oligo product are combined, rotovaped briefly to remove acetonitrile and ammonia, and then freeze dried. D. Mass Spectral Analysis
  • Stock buffer 1 is titrated with stock buffer 2 to form a series of buffers ranging from pH 4 to pH 8 in increments of about 0.2 pH units.
  • 0.3 ml of solvent is prepared.
  • the solvent is n-octanol for polypeptides containing less than about 50% acid amino acids and n-pentanol for polypeptides containing more than about 50% acid amino acid.
  • To each vial of this set is added 20 ⁇ l of a 0.1 mM aqueous solution of polypeptide having an attached carboxy fluorescein. Each of these vials is capped, shaken thoroughly, and then centrifuged to separate the phases.
  • This Example describes a useful method for testing for transport of a fluorescein-tagged polypeptide composition, with or without attached drug, from the extracellular medium directly into the cytosol of adherent animal cells. It should be noted that appreciable cell entry via endocytosis does not occur in the short time period (about 20 minutes) of this procedure. Because mounting medium, which is generally used in fluorescence microscopy, modifies both the osmolarity and the pH of the cells' environment, it is recommended that mounting medium not be used in these cell entry studies. Instead, cells are viewed using a water-immersion objective. Further, it is desirable mat the culture medium in which the cells are viewed does not contain phenol red, which is typically used as a pH indicator in culture medium, because this compound interferes with the fluorescent signal.
  • Medium 1 comprises serum-free medium buffered with 20 mM MES (mo ⁇ holine ethane sulfonic acid) adjusted to pH 6.5 with NaOH.
  • Medium 2 comprises serum-free medium buffered with 50 mM MES adjusted to pH 5.0.
  • a 150 ⁇ l aliquot of a 1 mM solution of fluorescein-tagged polypeptide composition is diluted into 1.9 ml of Medium 1, and 0.5 ml portions are added to 2 cm x 2 cm wells in four glass chambered slides in which Hela cells were plated 24 hours earlier. After 10 minutes 375 ⁇ l of Medium 2 is added to wells 2, 3, and 4 (giving pH 6.0).
  • the above procedure provides information on how low the pH must be in order to effect transmembrane transport of a given polypeptide composition of the invention, including, when desired, the attached compound to be transported.
  • Hela cells used in this functional assay were stably transfected with a plasmid containing a mouse mammary tumor virus promoter (inducible with dexamethasone) controlling a gene coding for the 5' untranslated region of rabbit ⁇ -globin mRNA, followed by the coding sequence for firefly luciferase (Partridge, 1996).
  • a mouse mammary tumor virus promoter inducible with dexamethasone
  • Cell-free and in-cell scrape-load translation studies have shown that the Mo ⁇ holino antisense oligomer shown in Figure 13 is highly effective in blocking the translation of luciferase from this gene construct.
  • the antisense oligomer of Figure 13 gains access to the cytosolic compartment of these transfected cells, it should effect a significant reduction in luciferase activity upon dexamethasone induction, relative to untreated cells. Such a reduction in luciferase activity, measured as relative light units in a luminometer, would be indicative of successful cytosolic delivery of the Mo ⁇ holino oligomer.
  • the polypeptide-Mo ⁇ holino antisense oligonucleotide product prepared as in Example 6C was suspended in culture medium at a concentration of 5 ⁇ M.
  • the above-described transfected Hela cells were treated with the suspension for 5 hours, and then treated for 16 hours with dexamethasone to induce luciferase synthesis.
  • cells in separate wells were treated with a) medium alone, b) Mo ⁇ holino antisense oligo, and c) polypeptide.
  • TreatmentRelative Light Units medium alone 100 polypeptide 105 polypeptide-Mo ⁇ holino product 69
  • EXAMPLE 11 Enhancement of Transdermal Delivery
  • Tritiated cyclosporin A metabolite 17 is linked to a suitable polypeptide, as described in Examples 3 and 4, and the conjugate composition, with the polypeptide moiety in its low-pH form, is suspended in a suitable penetration enhancer solution which includes a lipophilic fatty acid (e.g. , 10% linoleic acid/90% propylene glycol).
  • Tritiated cyclosporin A metabolite 17, without attached polypeptide is suspended in the same penetration enhancer solution, for comparative assessment of the rate of transdermal passage of the unmodified drug.
  • Each cyciosporin-containing solution is contacted with a defined area of skin on a nude hairless mouse, and small aliquots of blood are withdrawn periodically for assessment of the amount of tritiated drug which has passed through the skin and into the circulatory system.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Compositions polymères permettant l'apport efficace de composés dans des organismes vivants. Les compositions comprennent des polypeptides présentant une solubilité dans des environnements à la fois hydrophiles et lipophiles du fait qu'ils subissent une transition réversible dépendante du pH d'une forme lipophile à pH bas à une forme hydrophile à pH élevé.
PCT/US1997/007335 1996-05-01 1997-04-30 Conjugues polypeptidiques destines au transport de substances a travers des membranes cellulaires WO1997040854A2 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CA002252706A CA2252706A1 (fr) 1996-05-01 1997-04-30 Conjugues polypeptidiques destines au transport de substances a travers des membranes cellulaires
EP97923513A EP0966303A2 (fr) 1996-05-01 1997-04-30 Conjugues polypeptidiques destines au transport de substances a travers des membranes cellulaires
JP9539221A JP2000509394A (ja) 1996-05-01 1997-04-30 細胞膜を横切って物質を輸送するためのポリペプチド結合体
AU29298/97A AU729643B2 (en) 1996-05-01 1997-04-30 Polypeptide conjugates for transporting substances across cell membranes

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US1634796P 1996-05-01 1996-05-01
US60/016,347 1996-05-01
US2860996P 1996-10-23 1996-10-23
US60/028,609 1996-10-23

Publications (2)

Publication Number Publication Date
WO1997040854A2 true WO1997040854A2 (fr) 1997-11-06
WO1997040854A3 WO1997040854A3 (fr) 1998-03-12

Family

ID=26688485

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/007335 WO1997040854A2 (fr) 1996-05-01 1997-04-30 Conjugues polypeptidiques destines au transport de substances a travers des membranes cellulaires

Country Status (5)

Country Link
EP (1) EP0966303A2 (fr)
JP (1) JP2000509394A (fr)
AU (1) AU729643B2 (fr)
CA (1) CA2252706A1 (fr)
WO (1) WO1997040854A2 (fr)

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998032467A2 (fr) * 1997-01-24 1998-07-30 Antivirals, Inc. METHODE ET CONJUGUE DE TRAITEMENT D'INFECTIONS A $i(H. PYLORI)
WO1998035703A2 (fr) * 1997-02-14 1998-08-20 The Salk Institute For Biological Studies Procedes et compositions permettant d'apporter des agents therapeutiques a des tissus osseux a l'aide de conjugues d'oligomeres peptidiques charges negativement et d'agents therapeutiques
WO2001012235A2 (fr) * 1999-08-19 2001-02-22 University Of Southern California Apport cible de gene artificiel
US6306993B1 (en) 1997-05-21 2001-10-23 The Board Of Trustees Of The Leland Stanford, Jr. University Method and composition for enhancing transport across biological membranes
WO2002004479A1 (fr) 2000-07-06 2002-01-17 Avi Biopharma, Inc. TRANSFORMATION D'UNE COMPOSITION DE CELLULES SOUCHES TRAITEES A L'AIDE D'UN AGENT DE BLOCAGE DU FACTEUR DE CROISSANCE BETA (TGF-ss), ET PROCEDE CORRESPONDANT
US6593292B1 (en) 1999-08-24 2003-07-15 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6669951B2 (en) 1999-08-24 2003-12-30 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6730293B1 (en) 1999-08-24 2004-05-04 Cellgate, Inc. Compositions and methods for treating inflammatory diseases of the skin
JP2006516948A (ja) * 2000-11-14 2006-07-13 ニュー リバー ファーマシューティカルズ インコーポレイテッド 硫酸アバカビルを含有する新規な薬剤化合物および同化合物の製造ならびに使用方法
WO2006086667A2 (fr) 2005-02-09 2006-08-17 Avi Bio Pharma, Inc. Composition antisens et methode permettant de traiter une atrophie musculaire
WO2007009094A2 (fr) 2005-07-13 2007-01-18 Avi Biopharma, Inc. Compose et procede antibacteriens antisens
US7229961B2 (en) 1999-08-24 2007-06-12 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
EP1935434A1 (fr) * 2006-12-19 2008-06-25 Novosom AG Construction et utilisation d'éléments améliorant la transfection
WO2009117553A2 (fr) 2008-03-20 2009-09-24 University Of Florida Research Foundation, Inc. Amélioration de nostocytose de lésion de vaisseau et réparation potentielle de cellules souches
EP2143726A1 (fr) * 2008-07-11 2010-01-13 Novosom AG Acide nucléique comprenant des nucleéotides zwitterioniques
US7807780B2 (en) 2000-07-21 2010-10-05 Revance Therapeutics, Inc. Multi-component biological transport systems
US7902156B2 (en) 2005-02-18 2011-03-08 Angiochem Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
EP2351839A2 (fr) 2004-07-02 2011-08-03 AVI BioPharma, Inc. Technique et composé antibactérielle antisense
US8022179B2 (en) 2005-03-03 2011-09-20 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of an oligopeptide
EP2376633A1 (fr) * 2008-12-17 2011-10-19 AVI BioPharma, Inc. Compositions antisens et méthodes de modulation de l'hypersensibilité de contact ou de la dermatite de contact
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US8415313B2 (en) 2004-01-23 2013-04-09 Avi Biopharma, Inc. Antisense oligomers and methods for inducing immune tolerance and immunosuppression
US8450285B2 (en) 2009-06-05 2013-05-28 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
US8580748B2 (en) 2011-04-06 2013-11-12 13Therapeutics, Inc. Peptides for the treatment of hearing
WO2013186546A1 (fr) * 2012-06-12 2013-12-19 Polytherics Limited Complexes d'amphotéricine b ayant du poly(acide glutamique)
US8969310B2 (en) 2005-07-15 2015-03-03 Angiochem Inc. Potentiation of anticancer agents
US9173891B2 (en) 2009-04-20 2015-11-03 Angiochem, Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US9278987B2 (en) 2011-11-18 2016-03-08 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US9469664B2 (en) 2010-05-28 2016-10-18 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US9487786B2 (en) 2005-11-08 2016-11-08 Sarepta Therapeutics, Inc. Immunosuppression compound and treatment method
WO2017062699A1 (fr) * 2015-10-07 2017-04-13 Cormedix Inc. Formulation de pénétration cutanée à base de taurolidine
US9687561B2 (en) 2012-08-14 2017-06-27 Angiochem Inc. Peptide-dendrimer conjugates and uses thereof
US9914754B2 (en) 2008-12-05 2018-03-13 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
US9920085B2 (en) 2012-03-20 2018-03-20 Sarepta Therapeutics, Inc. Boronic acid conjugates of oligonucleotide analogues
US10017763B2 (en) 2010-09-03 2018-07-10 Sarepta Therapeutics, Inc. dsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US10080786B2 (en) 2005-03-03 2018-09-25 Revance Therapeutics, Inc. Methods for treating pain by topical application and transdermal delivery of botulinum toxin
US10106795B2 (en) 2011-10-04 2018-10-23 Royal Holloway And Bedford New College Oligomers
US10172877B2 (en) 2004-03-03 2019-01-08 Revance Therapeutics, Inc. Compositions and methods for topical diagnostic and therapeutic transport
US10980892B2 (en) 2015-06-15 2021-04-20 Angiochem Inc. Methods for the treatment of leptomeningeal carcinomatosis
US11015200B2 (en) 2015-03-18 2021-05-25 Sarepta Therapeutics, Inc. Antisense-induced exon exclusion in myostatin
US11020417B2 (en) 2015-06-04 2021-06-01 Sarepta Therapeutics, Inc Methods and compounds for treatment of lymphocyte-related diseases and conditions

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2004523480A (ja) * 2000-08-22 2004-08-05 ニュー リバー ファーマシューティカルズ, インコーポレイテッド 活性作用物質送達系ならびに活性作用物質の保護および投与方法
JP4625637B2 (ja) * 2002-02-22 2011-02-02 シャイア エルエルシー 活性物質送達系及び活性物質を保護し投与する方法
EP1583562B1 (fr) 2003-01-06 2011-06-15 Angiochem Inc. Angiopep-1, composes apparentes, et leurs utilisations
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
US8921314B2 (en) 2008-10-15 2014-12-30 Angiochem, Inc. Conjugates of GLP-1 agonists and uses thereof
IN2012DN00248A (fr) 2009-07-02 2015-05-01 Angiochem Inc

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1541436A (en) * 1976-02-02 1979-02-28 Searle & Co Immunological materials
WO1992002255A1 (fr) * 1990-08-03 1992-02-20 Farmitalia Carlo Erba S.R.L. Nouveau segment de liaison pour agents bioactifs
WO1992010212A1 (fr) * 1990-12-05 1992-06-25 Farmitalia Carlo Erba Srl Conjugues d'anthracycline
WO1997033552A1 (fr) * 1996-03-12 1997-09-18 Pg-Txl Company, L.P. Bioprecurseurs de paclitaxel solubles dans l'eau

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1541436A (en) * 1976-02-02 1979-02-28 Searle & Co Immunological materials
WO1992002255A1 (fr) * 1990-08-03 1992-02-20 Farmitalia Carlo Erba S.R.L. Nouveau segment de liaison pour agents bioactifs
WO1992010212A1 (fr) * 1990-12-05 1992-06-25 Farmitalia Carlo Erba Srl Conjugues d'anthracycline
WO1997033552A1 (fr) * 1996-03-12 1997-09-18 Pg-Txl Company, L.P. Bioprecurseurs de paclitaxel solubles dans l'eau

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
ADAMS N W ET AL: "CONTROLLED RELEASE POLYAMINO ACID-DRUG CONJUGATES FOR SUBCUTANEOUS INJECTION." 198TH ACS (AMERICAN CHEMICAL SOCIETY) NATIONAL MEETING, MIAMI BEACH, FLORIDA, USA, SEPTEMBER 10-15, 1989. IN: ABSTR PAP AM CHEM SOC, VOL. 198, ABSTRACT NO. 113, XP002048430 *
BIOLOGICAL ABSTRACTS, vol. BA88, Philadelphia, PA, US; abstract no. 110606, KIM C-K ET AL: "EFFECTS OF MOLECULAR WEIGHTS ON THE PHYSICO-PHARMACEUTICAL PROPERTIES OF POLY-L-GLUTAMIC ACID CYTARABINE CONJUGATES." XP002048437 & ARCH PHARMACAL RES (SEOUL), VOL. 12, NO. 2, PUBL. 1989, PAGE(S) 88-93., *
FABRE, ISABELLE ET AL: "Polyglutamylation, an important element in methotrexate cytotoxicity and selectivity in tumor versus murine granulocytic progenitor cells in vitro" CANCER RES., 1984, VOL. 44, NO. 8, PAGE(S) 3190-5, XP002048435 *
KATO Y ET AL: "ANTI TUMOR ACTIVITY OF 1-BETA-D ARABINOFURANOSYL CYTOSINE CONJUGATED WITH POLY GLUTAMIC-ACID AND ITS DERIVATIVE." CANCER RES, VOL. 44, NO. 1, PUBL. 1984, PAGE(S) 25-30., XP002048431 *
LI C ET AL: "Development of a highly efficacious water-soluble polymer- taxol conjugate." PROCEEDINGS OF THE EIGHTY-EIGHTH ANNUAL MEETING OF THE AMERICAN ASSOCIATION FOR CANCER RESEARCH, SAN DIEGO, CALIFORNIA, USA, APRIL 12-16, 1997., vol. 38, March 1997, page 258 XP002048436 *
MALLEY A ET AL: "ANTI TIMOTHY IMMUNO GLOBULIN E FORMATION SUPPRESSION WITH ANTIGEN D GLUTAMIC-ACID D LYSINE CO POLYMER." INT ARCH ALLERGY APPL IMMUNOL, VOL. 63, NO. 1, PUBL. 1980, PAGE(S) 113-120., XP002048428 *
MCCORMICK-THOMSON, L. ANNE ET AL: "Poly(amino acid) copolymers as a potential soluble drug delivery system. 1. Pinocytic uptake and lysosomal degradation measured in vitro" J. BIOACT. COMPAT. POLYM., 1989, VOL. 4, NO. 3, PAGE(S) 242-51, XP002048433 *
MCCORMICK-THOMSON, L. ANNE ET AL: "Poly(amino acid) copolymers as a potential soluble drug delivery system. 2. Body distribution and preliminary biocompatibility testing in vitro and in vivo" J. BIOACT. COMPAT. POLYM., 1989, VOL. 4, NO. 3, PAGE(S) 252-68, XP002048434 *
MORIMOTO Y ET AL: "ANTITUMOR AGENT POLY-AMINO-ACID CONJUGATES AS A DRUG CARRIER IN CANCER CHEMOTHERAPY." J PHARMACOBIO-DYN, VOL. 7, NO. 9, PUBL. 1984, PAGE(S) 688-698., XP002048432 *
ROSENBLATT, D. S. ET AL: "Differential effects of folinic acid and glycine, adenosine, and thymidine as rescue agents in methotrexate-treated human cells in relation to the accumulation of methotrexate polyglutamates" MOL. PHARMACOL., 1982, VOL. 21, NO. 3, PAGE(S) 718-22, XP002048427 *
ROSENBLATT, DAVID S. ET AL: "Methotrexate polyglutamates in cultured human cells" ADV. EXP. MED. BIOL., 1983, VOL. 163, NO. FOLYL ANTIFOLYL POLYGLUTAMATES, PAGE(S) 275-85, XP002048429 *

Cited By (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998032467A3 (fr) * 1997-01-24 1998-09-17 Antivirals Inc METHODE ET CONJUGUE DE TRAITEMENT D'INFECTIONS A $i(H. PYLORI)
WO1998032467A2 (fr) * 1997-01-24 1998-07-30 Antivirals, Inc. METHODE ET CONJUGUE DE TRAITEMENT D'INFECTIONS A $i(H. PYLORI)
WO1998035703A2 (fr) * 1997-02-14 1998-08-20 The Salk Institute For Biological Studies Procedes et compositions permettant d'apporter des agents therapeutiques a des tissus osseux a l'aide de conjugues d'oligomeres peptidiques charges negativement et d'agents therapeutiques
WO1998035703A3 (fr) * 1997-02-14 1999-02-11 Salk Inst For Biological Studi Procedes et compositions permettant d'apporter des agents therapeutiques a des tissus osseux a l'aide de conjugues d'oligomeres peptidiques charges negativement et d'agents therapeutiques
US6306993B1 (en) 1997-05-21 2001-10-23 The Board Of Trustees Of The Leland Stanford, Jr. University Method and composition for enhancing transport across biological membranes
US6495663B1 (en) 1997-05-21 2002-12-17 The Board Of Trustees Of The Leland Stanford Junior University Method and composition for enhancing transport across biological membranes
US20080103108A1 (en) * 1999-08-19 2008-05-01 Yanina Rozenberg Targeted artificial gene delivery
WO2001012235A2 (fr) * 1999-08-19 2001-02-22 University Of Southern California Apport cible de gene artificiel
WO2001012235A3 (fr) * 1999-08-19 2001-05-25 Univ Southern California Apport cible de gene artificiel
JP2003507348A (ja) * 1999-08-19 2003-02-25 ユニバーシティ オブ サザン カリフォルニア 標的設定人工遺伝子送達
US6593292B1 (en) 1999-08-24 2003-07-15 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6669951B2 (en) 1999-08-24 2003-12-30 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US6730293B1 (en) 1999-08-24 2004-05-04 Cellgate, Inc. Compositions and methods for treating inflammatory diseases of the skin
US6759387B2 (en) 1999-08-24 2004-07-06 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US8729010B2 (en) 1999-08-24 2014-05-20 Kai Pharmaceuticals, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
US8623833B2 (en) 1999-08-24 2014-01-07 Kai Pharmaceuticals, Inc. Compositions and methods for enhancing drug delivery across and into epithelial tissues
US7229961B2 (en) 1999-08-24 2007-06-12 Cellgate, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
EP2269654A3 (fr) * 1999-08-24 2011-04-13 Cellgate Inc. Amélioration de l'administration de médicaments à travers et dans les tissus épithéliaux en utilisant des chaînes d'oligo-arginine.
US8278264B2 (en) 1999-08-24 2012-10-02 Kai Pharmaceuticals, Inc. Compositions and methods for enhancing drug delivery across and into ocular tissues
EP3020804A1 (fr) 2000-07-06 2016-05-18 Sarepta Therapeutics, Inc. Composition et procédé de cellules souches traitées avec un agent bloquant la transformation d'un facteur de croissance transformant beta (tgf-)
WO2002004479A1 (fr) 2000-07-06 2002-01-17 Avi Biopharma, Inc. TRANSFORMATION D'UNE COMPOSITION DE CELLULES SOUCHES TRAITEES A L'AIDE D'UN AGENT DE BLOCAGE DU FACTEUR DE CROISSANCE BETA (TGF-ss), ET PROCEDE CORRESPONDANT
US7807780B2 (en) 2000-07-21 2010-10-05 Revance Therapeutics, Inc. Multi-component biological transport systems
JP2006516948A (ja) * 2000-11-14 2006-07-13 ニュー リバー ファーマシューティカルズ インコーポレイテッド 硫酸アバカビルを含有する新規な薬剤化合物および同化合物の製造ならびに使用方法
US8415313B2 (en) 2004-01-23 2013-04-09 Avi Biopharma, Inc. Antisense oligomers and methods for inducing immune tolerance and immunosuppression
US10172877B2 (en) 2004-03-03 2019-01-08 Revance Therapeutics, Inc. Compositions and methods for topical diagnostic and therapeutic transport
US9211248B2 (en) 2004-03-03 2015-12-15 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
EP2351839A2 (fr) 2004-07-02 2011-08-03 AVI BioPharma, Inc. Technique et composé antibactérielle antisense
US9534220B2 (en) 2004-07-02 2017-01-03 Sarepta Therapeutics, Inc. Antisense antibacterial method and compound
US8314072B2 (en) 2004-07-02 2012-11-20 Avi Biopharma, Inc. Antisense antibacterial method and compound
US8785410B2 (en) 2005-02-09 2014-07-22 Sarepta Therapeutics, Inc. Antisense composition and method for treating muscle atrophy
WO2006086667A2 (fr) 2005-02-09 2006-08-17 Avi Bio Pharma, Inc. Composition antisens et methode permettant de traiter une atrophie musculaire
US10626396B2 (en) 2005-02-09 2020-04-21 Sarepta Therapeutics, Inc. Antisense composition and method for treating muscle atrophy
US7888012B2 (en) 2005-02-09 2011-02-15 Avi Biopharma, Inc. Antisense composition and method for treating muscle atrophy
US10006031B2 (en) 2005-02-09 2018-06-26 Sarepta Therapeutics, Inc. Antisense composition and method for treating muscle atrophy
US7902156B2 (en) 2005-02-18 2011-03-08 Angiochem Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
US8828949B2 (en) 2005-02-18 2014-09-09 Angiochem, Inc. Aprotinin polypeptides for transporting a compound across the blood-brain barrier
US10744078B2 (en) 2005-03-03 2020-08-18 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of botulinum toxins
US10080786B2 (en) 2005-03-03 2018-09-25 Revance Therapeutics, Inc. Methods for treating pain by topical application and transdermal delivery of botulinum toxin
US8022179B2 (en) 2005-03-03 2011-09-20 Revance Therapeutics, Inc. Compositions and methods for topical application and transdermal delivery of an oligopeptide
US8536147B2 (en) 2005-07-13 2013-09-17 Sarepta Therapeutics, Inc. Antibacterial antisense oligonucleotide and method
WO2007009094A2 (fr) 2005-07-13 2007-01-18 Avi Biopharma, Inc. Compose et procede antibacteriens antisens
US8067571B2 (en) 2005-07-13 2011-11-29 Avi Biopharma, Inc. Antibacterial antisense oligonucleotide and method
US9499583B2 (en) 2005-07-13 2016-11-22 Sarepta Therapeutics, Inc. Antibacterial antisense oligonucleotide and method
US10144762B2 (en) 2005-07-13 2018-12-04 Sarepta Therapeutics, Inc. Antibacterial antisense oligonucleotide and method
US9249243B2 (en) 2005-07-13 2016-02-02 Sarepta Therapeutics, Inc. Antibacterial antisense oligonucleotide and method
US9713646B2 (en) 2005-07-15 2017-07-25 Angiochem Inc. Potentiation of anticancer agents
US8969310B2 (en) 2005-07-15 2015-03-03 Angiochem Inc. Potentiation of anticancer agents
US9487786B2 (en) 2005-11-08 2016-11-08 Sarepta Therapeutics, Inc. Immunosuppression compound and treatment method
WO2008074488A2 (fr) * 2006-12-19 2008-06-26 Novosom Ag Construction et utilisation d'éléments activateurs de transfection
EP1935434A1 (fr) * 2006-12-19 2008-06-25 Novosom AG Construction et utilisation d'éléments améliorant la transfection
WO2008074488A3 (fr) * 2006-12-19 2009-03-26 Novosom Ag Construction et utilisation d'éléments activateurs de transfection
WO2009117553A2 (fr) 2008-03-20 2009-09-24 University Of Florida Research Foundation, Inc. Amélioration de nostocytose de lésion de vaisseau et réparation potentielle de cellules souches
EP2143726A1 (fr) * 2008-07-11 2010-01-13 Novosom AG Acide nucléique comprenant des nucleéotides zwitterioniques
US9914754B2 (en) 2008-12-05 2018-03-13 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
EP2376633A1 (fr) * 2008-12-17 2011-10-19 AVI BioPharma, Inc. Compositions antisens et méthodes de modulation de l'hypersensibilité de contact ou de la dermatite de contact
US9173891B2 (en) 2009-04-20 2015-11-03 Angiochem, Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
US9187530B2 (en) 2009-06-05 2015-11-17 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
US9150617B2 (en) 2009-06-05 2015-10-06 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
US9150616B2 (en) 2009-06-05 2015-10-06 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
US9974853B2 (en) 2009-06-05 2018-05-22 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
US8450285B2 (en) 2009-06-05 2013-05-28 13Therapeutics, Inc. Immunoregulatory peptides and methods of use
US10760078B2 (en) 2010-05-28 2020-09-01 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US10202602B2 (en) 2010-05-28 2019-02-12 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US9469664B2 (en) 2010-05-28 2016-10-18 Sarepta Therapeutics, Inc. Oligonucleotide analogues having modified intersubunit linkages and/or terminal groups
US10017763B2 (en) 2010-09-03 2018-07-10 Sarepta Therapeutics, Inc. dsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US11072793B2 (en) 2010-09-03 2021-07-27 Sarepta Therapeutics, Inc. DsRNA molecules comprising oligonucleotide analogs having modified intersubunit linkages and/or terminal groups
US8580748B2 (en) 2011-04-06 2013-11-12 13Therapeutics, Inc. Peptides for the treatment of hearing
US10662431B2 (en) 2011-10-04 2020-05-26 Royal Holloway And Bedford New College Oligomers
US10421969B2 (en) 2011-10-04 2019-09-24 Royal Holloway And Bedford New College Oligomers
US10947536B2 (en) 2011-10-04 2021-03-16 Royal Holloway And Bedford New College Oligomers
US10106795B2 (en) 2011-10-04 2018-10-23 Royal Holloway And Bedford New College Oligomers
US9278987B2 (en) 2011-11-18 2016-03-08 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US10344281B2 (en) 2011-11-18 2019-07-09 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US9790499B2 (en) 2011-11-18 2017-10-17 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US11208655B2 (en) 2011-11-18 2021-12-28 Sarepta Therapeutics, Inc. Functionally-modified oligonucleotides and subunits thereof
US9920085B2 (en) 2012-03-20 2018-03-20 Sarepta Therapeutics, Inc. Boronic acid conjugates of oligonucleotide analogues
WO2013186546A1 (fr) * 2012-06-12 2013-12-19 Polytherics Limited Complexes d'amphotéricine b ayant du poly(acide glutamique)
US9687561B2 (en) 2012-08-14 2017-06-27 Angiochem Inc. Peptide-dendrimer conjugates and uses thereof
US11015200B2 (en) 2015-03-18 2021-05-25 Sarepta Therapeutics, Inc. Antisense-induced exon exclusion in myostatin
US11020417B2 (en) 2015-06-04 2021-06-01 Sarepta Therapeutics, Inc Methods and compounds for treatment of lymphocyte-related diseases and conditions
US10980892B2 (en) 2015-06-15 2021-04-20 Angiochem Inc. Methods for the treatment of leptomeningeal carcinomatosis
WO2017062699A1 (fr) * 2015-10-07 2017-04-13 Cormedix Inc. Formulation de pénétration cutanée à base de taurolidine
AU2016334086B2 (en) * 2015-10-07 2022-10-20 Cormedix Inc. Skin-penetrating formulation of taurolidine

Also Published As

Publication number Publication date
JP2000509394A (ja) 2000-07-25
WO1997040854A3 (fr) 1998-03-12
CA2252706A1 (fr) 1997-11-06
EP0966303A2 (fr) 1999-12-29
AU729643B2 (en) 2001-02-08
AU2929897A (en) 1997-11-19

Similar Documents

Publication Publication Date Title
US6030941A (en) Polymer composition for delivering substances in living organisms
AU729643B2 (en) Polypeptide conjugates for transporting substances across cell membranes
JP3814294B2 (ja) 親水性分子の脂質化のための方法および組成物
AU2002306500B2 (en) Transporters comprising spaced arginine moieties
US5583020A (en) Permeability enhancers for negatively charged polynucleotides
Jensen et al. Cytoplasmic delivery and nuclear targeting of synthetic macromolecules
AU699387B2 (en) Use of nitric oxide-releasing agents for reducing metastasis risk
Moon et al. In vitro assessment of a novel polyrotaxane-based drug delivery system integrated with a cell-penetrating peptide
US20030073640A1 (en) Novel compositions for the delivery of negatively charged molecules
US20070086981A1 (en) Delivery peptides, their constructs with active agents and use
US20110312877A1 (en) Engineered tunable nanoparticles for delivery of therapeutics, diagnostics, and experimental compounds and related compositions for therapeutic use
US20100226987A1 (en) Targeting conjugates comprising active agents encapsulated in cyclodextrin-containing polymers
EP0700295A1 (fr) Systemes de diffusion orale a mediation par la vitamine b 12? destines au gcsf et a l'epo
JP2001511113A (ja) ピペラジンベースのサイトフェクチン
KR102015524B1 (ko) 압타머-약물 콘쥬게이트 및 그 용도
Shi et al. Intelligent “Peptide-Gathering Mechanical Arm” tames wild “Trojan-Horse” peptides for the controlled delivery of cancer nanotherapeutics
Pahwa et al. Melphalan: Recent insights on synthetic, analytical and medicinal aspects
US7314956B2 (en) Multifunctional carrier for the delivery of a pharmacological agent or genetic material into a cell
US9943606B2 (en) Dendritic polypeptide-based nanocarriers for the delivery of therapeutic agents
Skwarecki et al. Synthetic strategies in construction of organic low molecular-weight carrier-drug conjugates
EP0485749A2 (fr) Modification chimique d'anticorps pour la création d'immunoconjugués
JP7068711B2 (ja) 細胞質送達ペプチド
US8772248B2 (en) Drug delivery system
Van Heeswijk et al. Synthesis, characterization and antitumor activity of macromolecular prodrugs of adriamycin
WO2024041535A1 (fr) Nanocomposition, son procédé de préparation et son utilisation

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE GH HU IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG UZ VN YU AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase in:

Ref document number: 2252706

Country of ref document: CA

Ref country code: CA

Ref document number: 2252706

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997923513

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1997923513

Country of ref document: EP