US20090285771A1 - Methods and compositions for needleless delivery of macromolecules - Google Patents

Methods and compositions for needleless delivery of macromolecules Download PDF

Info

Publication number
US20090285771A1
US20090285771A1 US11/664,787 US66478705A US2009285771A1 US 20090285771 A1 US20090285771 A1 US 20090285771A1 US 66478705 A US66478705 A US 66478705A US 2009285771 A1 US2009285771 A1 US 2009285771A1
Authority
US
United States
Prior art keywords
macromolecule
delivery construct
human
subject
receptor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/664,787
Inventor
Randall J. Mrsny
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
TRINITY (ASSIGNMENT FOR BENEFIT OF CREDITORS) LLC
Original Assignee
Trinity Biosystems 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 Trinity Biosystems Inc filed Critical Trinity Biosystems Inc
Priority to US11/664,787 priority Critical patent/US20090285771A1/en
Assigned to SANDERLING VENTURE PARTNERS V, L.P., AS COLLATERAL AGENT reassignment SANDERLING VENTURE PARTNERS V, L.P., AS COLLATERAL AGENT SECURITY AGREEMENT Assignors: TRINITY BIOSYSTEMS, INC.
Publication of US20090285771A1 publication Critical patent/US20090285771A1/en
Assigned to TRINITY (ASSIGNMENT FOR THE BENEFIT OF CREDITORS), LLC reassignment TRINITY (ASSIGNMENT FOR THE BENEFIT OF CREDITORS), LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TRINITY BIOSYSTEMS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/554Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one sulfur as ring hetero atoms, e.g. clothiapine, diltiazem
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/28Insulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • 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
    • A61K47/6415Toxins or lectins, e.g. clostridial toxins or Pseudomonas exotoxins
    • 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
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/06Drugs for disorders of the endocrine system of the anterior pituitary hormones, e.g. TSH, ACTH, FSH, LH, PRL, GH
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates, in part, to methods and compositions for needleless delivery of macromolecules to a subject.
  • the methods and compositions involve administering to the subject a delivery construct comprising the macromolecule to be delivered, wherein the macromolecule is linked to the remainder of the construct with a linker that is cleavable at a basal-lateral membrane of a polarized epithelial cell.
  • therapeutic macromolecules are typically administered by injection. Such injections require penetration of the subject's skin and tissues and are associated with pain. Further, penetration of the skin breaches one effective nonspecific mechanism of protection against infection, and thus can lead to potentially serious infection.
  • the delivery constructs of the invention comprise a macromolecule for delivery to a subject that is linked to the remainder of the construct with a cleavable linker.
  • the linker is cleavable by an enzyme or an environmental cue that is present at the basal-lateral membrane of an epithelial cell.
  • the invention provides a delivery construct comprising a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the delivery construct.
  • the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject.
  • the cleavable linker can be cleavable by an enzyme that is present in the plasma of said subject.
  • the invention provides a polynucleotide that encodes a delivery construct comprising a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the delivery construct.
  • the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject.
  • the cleavable linker can be cleavable by an enzyme that is present in the plasma of said subject.
  • the polynucleotide that encodes a delivery construct comprises a nucleic acid sequence encoding a receptor binding domain, a nucleic acid sequence encoding a transcytosis domain, a nucleic acid sequence encoding a cleavable linker, and a nucleic acid sequence comprising a polylinker insertion site.
  • the polylinker insertion site can be oriented relative to the nucleic acid sequence encoding a cleavable linker to allow to cleavage of the cleavable linker to separate a macromolecule that is encoded by a nucleic acid inserted into the polylinker insertion site from the remainder of the encoded delivery construct.
  • the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject. In other embodiments, the cleavable linker can be cleavable by an enzyme that is present in the plasma of said subject.
  • the invention provides an expression vector comprising a polynucleotide of the invention.
  • the invention provides a cell comprising an expression vector of the invention.
  • the invention provides a composition comprising a delivery construct of the invention.
  • the composition is a pharmaceutical composition.
  • the invention provides a method for delivering a macromolecule to a subject.
  • the method comprises contacting an apical surface of a polarized epithelial cell of the subject with a delivery construct of the invention.
  • the delivery construct can comprise a receptor binding domain, a transcytosis domain, a cleavable linker, and the macromolecule to be delivered.
  • the transcytosis domain can transcytose the macromolecule to and through the basal-lateral membrane of the epithelial cell.
  • the cleavable linker can be cleaved by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject.
  • the cleavable linker can be cleavable by an enzyme that is present in the plasma of the subject. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the delivery construct, and can deliver the macromolecule to the subject free from the remainder of the construct.
  • FIGS. 1A-1D present micrographs showing adhesion to and transport across mouse tracheal epithelium of an exemplary delivery construct comprising green fluorescent protein (GFP) (Panels A-C), while GFP alone does not adhere to the epithelial cells (Panel D).
  • GFP green fluorescent protein
  • FIG. 2 presents a time course of serum nt-PE-GFP concentrations following intranasal administration of 100 ⁇ g nt-PE-GFP to anesthetized mice.
  • FIG. 3 presents the amino acid sequence of an exemplary PE.
  • FIG. 4 presents a western blot snowing transport and cleavage of an exemplary delivery construct comprising rat growth hormone (rGH) by human intestinal epithelial cell monolayers.
  • the delivery construct was incubated in contact with the apical side of the epithelial cell monolayer for 4 hours.
  • Media isolated from the basolateral side of the membrane post-incubation (lane 1) contained rGH of native apparent molecular weight, while media from the apical side of the membrane (lane 2) contained intact delivery construct.
  • Lanes 3 and 4 show intact Delivery Construct 2 and recombinant rGH, respectively.
  • FIG. 7 presents a graphical representation comparing pharmacokinetics of rGH delivered subcutaneously and with Delivery Construct 2.
  • FIG. 8 shows expression levels of IGF-1-BP3 mRNA in the liver of female BALB/c mice treated with 30 ⁇ g recombinant rGH by subcutaneous injection or with 100 ⁇ g of Delivery Construct 2 by oral gavage.
  • Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-1-BP3, as described above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • FIG. 9 shows expression levels of growth hormone (GH) receptor mRNA in the liver of female BALB/c mice treated with rGH by subcutaneous injection (30 ⁇ g) or Delivery Construct 2 by oral gavage (100 ⁇ g).
  • Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for GH receptor, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • FIG. 10 snows expression levels or insulin-like growth factor I (TGF-I); mRNA in the liver of female BALB/c mice treated with rGH by subcutaneous injection (30 ⁇ g) or Delivery Construct 2 by oral gavage (100 ⁇ g). Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-I, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • FIGS. 11A and B show serum anti-rGH IgG antibodies (diluted 1:25 in FIG. 11A and 1:200 in FIG. 11B ) of mice orally administered 3,10, or 30 ⁇ g Delivery Construct 2 (F2) or subcutaneously administered 3 or 10 ⁇ g rGH in graphs with error bars, while FIGS. 11C and D presents the same data from individual animals.
  • FIG. 12 shows a nucleotide sequence that encodes Delivery Construct 6 (SEQ ID NO:34), an exemplary Delivery Construct for delivering human growth hormone (hGH).
  • SEQ ID NO:34 an exemplary Delivery Construct for delivering human growth hormone (hGH).
  • FIGS. 13A and B show the amino acid sequence of Delivery Construct 6 (SEQ ID NO:35), an exemplary Delivery Construct for delivering hGH.
  • FIG. 14 shows a nucleotide sequence that encodes Delivery Construct 7 (SEQ ID NO:36), an exemplary Delivery Construct for delivering interferon- ⁇ (IFN- ⁇ ).
  • FIGS. 15A and B show the amino acid sequence of Delivery Construct 7 (SEQ ID NO:37), an exemplary Delivery Construct for delivering IFN- ⁇ .
  • FIGS. 17A and B show the amino acid sequence of Delivery Construct 8 (SEQ ID NO:38), an exemplary Delivery Construct for delivering proinsulin.
  • FIGS. 18 A and B shows the amino acid sequence of the two amino acid chains of Delivery Construct 9 (SEQ ID NOs:39 and 40, respectively), an exemplary Delivery Construct for delivering insulin.
  • Disulfide bonds are formed between the cysteine at position 7 of SEQ ID NO:40 (shown in FIG. 18B ) and the cysteine at position 381 of SEQ ID NO:39 (shown in FIG. 18A ) and between the cysteine at position 20 of SEQ ID NO:40 and the cysteine at position 393 of SEQ ID NO:39.
  • a “ligand” is a compound that specifically binds to a target molecule.
  • exemplary ligands include, but are not limited to, an antibody, a cytokine, a substrate, a signaling molecule, and the like.
  • a “receptor” is compound that specifically binds to a ligand.
  • a ligand or a receptor e.g., an antibody “specifically binds to” or “is specifically immunoreactive with” another molecule when the ligand or receptor functions in a binding reaction that indicates the presence of the molecule in a sample of heterogeneous compounds.
  • the ligand or receptor binds preferentially to a particular compound and does not bind in a significant amount to other compounds present in the sample.
  • a polynucleotide specifically binds under hybridization conditions to another polynucleotide comprising a complementary sequence and an antibody specifically binds under immunoassay conditions to an antigen bearing an epitope used to induce the antibody.
  • Immunoassay refers to a method of detecting an analyte in a sample involving contacting the sample with an antibody that specifically binds to the analyte and detecting binding between the antibody and the analyte.
  • a variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein.
  • solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
  • an antibody that binds a particular antigen with an affinity (K m ) of about 10 ⁇ M specifically binds the antigen.
  • Linker refers to a molecule that joins two other molecules, either covalently, or through ionic, van der Waals or hydrogen bonds, e.g., a nucleic acid molecule that hybridizes to one complementary sequence at the 5′ end and to another complementary sequence at the 3′ end, thus joining two non-complementary sequences.
  • a “cleavable linker” refers to a linker that can be degraded or otherwise severed to separate the two components connected by the cleavable linker. Cleavable linkers are generally cleaved by enzymes, typically peptidases, proteases, nucleases, lipases, and the like.
  • Cleavable linkers may also be cleaved by environmental cues, such as, for example, changes in temperature, pH, salt concentration, etc. when there is such a change in environment following transcytosis of the delivery construct across a polarized epithelial membrane.
  • “Pharmaceutical composition” refers to a composition suitable for pharmaceutical use in an animal.
  • a pharmaceutical composition comprises a pharmacologically effective amount of an active agent and a pharmaceutically acceptable carrier.
  • “Pharmacologically effective amount” refers to that amount of an agent effective to produce the intended pharmacological result.
  • “Pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, vehicles, buffers, and excipients, such as a phosphate buffered saline solution, 5% aqueous solution of dextrose, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents and/or adjuvants.
  • a “pharmaceutically acceptable salt” is a salt that can be formulated into a compound for pharmaceutical use including, e.g., metal salts (sodium, potassium, magnesium, calcium, etc.) and salts of ammonia or organic amines.
  • Preferred pharmaceutical carriers depend upon the intended mode of administration of the active agent. Typical modes of administration include enteral (e.g., oral, intranasal, rectal, Or vaginal) or parenteral (e.g., subcutaneous, intramuscular, intravenous or intraperitoneal injection; or topical (e.g., transdermal, or transmucosal administration).
  • enteral e.g., oral, intranasal, rectal, Or vaginal
  • parenteral e.g., subcutaneous, intramuscular, intravenous or intraperitoneal injection
  • topical e.g., transdermal, or transmucosal administration.
  • Small organic molecule refers to organic molecules of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes organic biopolymers (e.g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, up to about 2000 Da, or up to about 1000 Da.
  • a “subject” of diagnosis, treatment, or administration is a human or non-human animal, including a mammal or a primate, and preferably a human.
  • Treatment refers to prophylactic treatment or therapeutic treatment.
  • a “prophylactic” treatment is a treatment administered to a subject who does not exhibit signs of a disease or exhibits only early signs for the purpose of decreasing the risk of developing pathology.
  • a “therapeutic” treatment is a treatment administered to a subject who exhibits signs of pathology for the purpose of diminishing or eliminating those signs.
  • Pseudomonas exotoxin A or “PE” is secreted by Pseudomonas aeruginosa as a 67 kD protein composed of three prominent globular domains (Ia, II, and III) and one small subdomain (Ib) that connects domains II and III. See A. S. Allured et al., 1986 , Proc. Natl. Acad. Set 83:1320-1324.
  • domain Ia of PE is believed to mediate cell binding because domain Ia specifically binds to the low density lipoprotein receptor-related protein (“LRP”), also known as the ⁇ 2-macroglobulin receptor (“ ⁇ 2-MR”) and CD-91.
  • LRP low density lipoprotein receptor-related protein
  • ⁇ 2-MR ⁇ 2-macroglobulin receptor
  • Domain Ia spans amino acids 1-252.
  • Domain II of PE is believed to mediate transcytosis to the interior of a cell following binding of domain Ia to the ⁇ 2-MR. Domain II spans amino acids 253-364.
  • Domain Ib has no known function and spans amino acids 365-399.
  • Domain III mediates cytotoxicity of PE and includes an endoplasmic reticulum retention sequence. PE cytotoxicity is believed to result from ADP ribosylation of elongation factor 2, which inactivates protein synthesis. Domain III spans amino acids 400-613 of PE. Deleting amino acid E553 (“ ⁇ E553”) from domain III eliminates EF2 ADP ribosylation activity and detoxifies PE.
  • PE ⁇ E553 PE having the mutation ⁇ E553 is referred to herein as “PE ⁇ E553.”
  • Genetically modified forms of PE are described in, e.g., U.S. Pat. Nos. 5,602,095; 5,512,658 and 5,458,878 Pseudomonas exotoxin, as used herein, also includes genetically modified, allelic, and chemically inactivated forms of PE within this definition. See, e.g., Vasil et al., 1986 , Infect. Immunol. 52:538-48. Further, reference to the various domains of PE is made herein to the reference PE sequence presented as FIG. 3 .
  • one or more domain from modified PE can also be used in the chimeric immunogens of the invention so long as the domains retain functional activity.
  • modified PE e.g., genetically or chemically modified PE, or a portion of such domains
  • One of skill in the art can readily identify such domains of such modified PE based on, for example, homology to the PE sequence exemplified in FIG. 3 and test for functional activity using, for example, the assays described below.
  • Polynucleotide refers to a polymer composed of nucleotide units.
  • Polynucleotides include naturally occurring nucleic acids, such as deoxyribonucleic acid (“DNA”) and ribonucleic acid (“RNA”) as well as nucleic acid analogs.
  • Nucleic acid analogs include those which include non-naturally occurring bases, nucleotides that engage in linkages with other nucleotides other than the naturally occurring phosphodiester bond or which include bases attached through linkages other than phosphodiester bonds.
  • nucleotide analogs include, for example and without limitation, phosphorothioates, phosphorodithioates, phosphorotriesters, phosphoramidates, boranophosphates, methylphosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs), and the like.
  • PNAs peptide-nucleic acids
  • Such polynucleotides can be synthesized, for example, using an automated DNA synthesizer.
  • the term “nucleic acid” typically refers to large polynucleotides.
  • oligonucleotide typically refers to short polynucleotides, generally no greater than about 50 nucleotides.
  • nucleotide sequence is represented by a DNA sequence (i.e., A, T, G, C)
  • this also includes an RNA sequence (i.e., A, U, G, C) in which “U” replaces “T.”
  • the left-hand end of a single-stranded polynucleotide sequence is the 5′-end; the left-hand direction of a double-stranded polynucleotide sequence is referred to as the 5′-direction.
  • the direction of 5′ to 3′ addition of nucleotides to nascent RNA transcripts is referred to as the transcription direction.
  • the DNA strand having the same sequence as an mRNA is referred to as the “coding strand”; sequences on the DNA strand having the same sequence as an mRNA transcribed from that DNA and which are located 5′ to the 5′-end of the RNA transcript are referred to as “upstream sequences”; sequences on the DNA strand having the same sequence as the RNA and which are 3′ to the 3′ end of the coding RNA transcript are referred to as “downstream sequences.”
  • “Complementary” refers to the topological compatibility or matching together of interacting surfaces of two polynucleotides.
  • the two molecules can be described as complementary, and furthermore, the contact surface characteristics are complementary to each other.
  • a first polynucleotide is complementary to a second polynucleotide if the nucleotide sequence of the first polynucleotide is substantially identical to the nucleotide sequence of the polynucleotide binding partner of the second polynucleotide, or if the first polynucleotide can hybridize to the second polynucleotide under stringent hybridization conditions.
  • the polynucleotide whose sequence 5′-TATAC-3′ is complementary to a polynucleotide whose sequence is 5′-GTATA-3′.
  • % sequence identity is used interchangeably herein with the term “% identity” and refers to the level of amino acid sequence identity between two or more peptide sequences or the level of nucleotide sequence identity between two or more nucleotide sequences, when aligned using a sequence alignment program.
  • 80% identity means the same thing as 80% sequence identity determined by a defined algorithm, and means that a given sequence is at least 80% identical to another length of another sequence.
  • Exemplary levels of sequence identity include, but are not limited to, 60, 70, 80, 85, 90, 95, 98% or more sequence identity to a given sequence.
  • % sequence homology is used interchangeably herein with the term “% homology” and refers to the level of amino acid sequence homology between two or more peptide sequences or the level of nucleotide sequence homology between two or more nucleotide sequences, when aligned using a sequence alignment program.
  • 80% homology means the same thing as 80% sequence homology determined by a defined algorithm, and accordingly a homologue of a given sequence has greater than 80% sequence homology over a length of the given sequence.
  • Exemplary levels of sequence homology include, but are not limited to, 60, 70, 80, 85, 90, 95, 98% or more sequence homology to a given sequence.
  • the BLASTX program is preferred for searching nucleic acid sequences that have been translated in all reading frames against amino acid sequences in the GenBank Protein Sequences and other public databases. Both BLASTP and BLASTX are run using default parameters of an open gap penalty of 11.0, and an extended gap penalty of 1.0, and utilize the BLOSUM-62 matrix. See id.
  • a preferred alignment of selected sequences in order to determine “% identity” between two or more sequences is performed using for example, the CLUSTAL-W program in MacVector version 6.5, operated with default parameters, including an open gap penalty of 10.0, an extended gap penalty of 0.1, and a BLOSUM 30 similarity matrix.
  • Poly Amino Acid refers to a hydrophilic amino acid having a side chain that is uncharged at physiological pH, but which has at least one bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms.
  • Genetically encoded polar amino acids include Asn (N), Gln (Q) Ser (S) and Thr (T).
  • Nonpolar Amino Acid refers to a hydrophobic amino acid having a side chain that is uncharged at physiological pH and which has bonds in which the pair of electrons shared in common by two atoms is generally held equally by each of the two atoms (i.e., the side chain is not polar).
  • Genetically encoded nonpolar amino acids include Ala (A), Gly (G), He (I), Leu (L), Met (M) and Val (V).
  • Hydrophilic Amino Acid refers to an amino acid exhibiting a hydrophobicity of less than zero according to the normalized consensus hydrophobicity scale of Eisenberg et al., 1984 , J. Mol. Biol. 179:125-142. Genetically encoded hydrophilic amino acids include Arg (R), Asn (N), Asp (D), Glu (E), Gln (Q), His (H), Lys (K), Ser (S) and Thr (T).
  • Hydrophobic Amino Acid refers to an amino acid exhibiting a hydrophobicity of greater than zero according to the normalized consensus hydrophobicity scale of Eisenberg et al., 1984 , J Mol. Biol. 179:125-142. Genetically encoded hydrophobic amino acids include Ala (A), Gly (G), He (I), Leu (L), Met (M), Phe (F), Pro (P), Trp (W), Tyr (Y) and Val (V).
  • Acidic Amino Acid refers to a hydrophilic amino acid having a side chain pK value of less than 7. Acidic amino acids typically have negatively charged side chains at physiological pH due to loss of a hydrogen ion. Genetically encoded acidic amino acids include Asp (D) and Glu (E).
  • Basic Amino Acid refers to a hydrophilic amino acid having a side chain pK value of greater than 7.
  • Basic amino acids typically have positively charged side chains at physiological pH due to association with a hydrogen ion.
  • Genetically encoded basic amino acids include Arg (R), His (H) and Lys (K).
  • Encoding refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a gene encodes a protein if transcription and translation of mRNA produced by that gene produces the protein in a cell or other biological system.
  • coding strand the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings
  • non-coding strand used as the template for transcription
  • a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns.
  • Amplification refers to any means by which a polynucleotide sequence is copied and thus expanded into a larger number of polynucleotide molecules, e.g., by reverse transcription, polymerase chain reaction, ligase chain reaction, and the like.
  • Primer refers to a polynucleotide that is capable of specifically hybridizing to a designated polynucleotide template and providing a point of initiation for synthesis of a complementary polynucleotide. Such synthesis occurs when the polynucleotide primer is placed under conditions in which synthesis is induced, i.e., in the presence of nucleotides, a complementary polynucleotide template, and an agent for polymerization such as DNA polymerase.
  • a primer is typically single-stranded, but may be double-stranded. Primers are typically deoxyribonucleic acids, but a wide variety of synthetic and naturally occurring primers are useful for many applications.
  • a primer is complementary to the template to which it is designed to hybridize to serve as a site for the initiation of synthesis, but need not reflect the exact sequence of the template. In such a case, specific hybridization of the primer to the template depends on the stringency of the hybridization conditions. Primers can be labeled with, e.g., chromogenic, radioactive, or fluorescent moieties and used as detectable moieties.
  • Probe when used in reference to a polynucleotide, refers to a polynucleotide that is capable of specifically hybridizing to a designated sequence of another polynucleotide.
  • a probe specifically hybridizes to a target complementary polynucleotide, but need not reflect the exact complementary sequence of the template. In such a case, specific hybridization of the probe to the target depends on the stringency of the hybridization conditions.
  • Probes can be labeled with, e.g., chromogenic, radioactive, or fluorescent moieties and used as detectable moieties. In instances where a probe provides a point of initiation for synthesis of a complementary polynucleotide, a probe can also be a primer.
  • Hybridizing specifically to or “specific hybridization” or “selectively hybridize to”, refers to the binding, duplexing, or hybridizing of a nucleic acid molecule preferentially to a particular nucleotide sequence under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA.
  • a complex mixture e.g., total cellular DNA or RNA.
  • stringent conditions refers to conditions under which a probe will hybridize preferentially to its target subsequence, and to a lesser extent to, or not at all to, other sequences. “Stringent hybridization” and “stringent hybridization wash conditions” in the context of nucleic acid hybridization experiments such as Southern and northern hybridizations are sequence dependent, and are different under different environmental parameters.
  • highly stringent hybridization and wash conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH.
  • Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe.
  • Very stringent conditions are selected to be equal to the Tm for a particular probe.
  • stringent hybridization conditions for hybridization of complementary nucleic acids which have more than about 100 complementary residues on a filter in a Southern or northern blot is 50% formalin with 1 mg of heparin at 42° C., with the hybridization being carried out overnight.
  • An example of highly stringent wash conditions is 0.15 M NaCl at 72° C. for about 15 minutes.
  • An example of stringent wash conditions is a 0.2 ⁇ SSC wash at 65° C. for 15 minutes. See Sambrook et al. for a description of SSC buffer. A high stringency wash can be preceded by a low stringency wash to remove background probe signal.
  • An exemplary medium stringency wash for a duplex of, e.g., more than about 100 nucleotides, is 1 ⁇ SSC at 45° C. for 15 minutes.
  • An exemplary low stringency wash for a duplex of, e.g., more than about 100 nucleotides, is 4-6 ⁇ SSC at 40° C. for 15 minutes.
  • a signal to noise ratio of 2 ⁇ (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.
  • Polypeptide refers to a polymer composed of amino acid residues, related naturally occurring structural variants, and synthetic non-naturally occurring analogs thereof linked via peptide bonds, related naturally occurring structural variants, and synthetic non-naturally occurring analogs thereof. Synthetic polypeptides can be synthesized, for example, using an automated polypeptide synthesizer. Conventional notation is used herein to portray polypeptide sequences; the beginning of a polypeptide sequence is the amino-terminus, while the end of a polypeptide sequence is the carboxyl-terminus.
  • protein typically refers to large polypeptides, for example, polypeptides comprising more than about 50 amino acids.
  • protein can also refer to dimers, trimers, and multimers that comprise more than one polypeptide.
  • Constant substitution refers to the substitution in a polypeptide of an amino acid with a functionally similar amino acid.
  • the following six groups each contain amino acids that are conservative substitutions for one another:
  • I Isoleucine
  • L Leucine
  • M Methionine
  • V Valine
  • the term “about 5 ⁇ g/kg” means a range of from 4.5 ⁇ g/kg to 5.5 ⁇ g/kg.
  • “about 1 hour” means a range of from 48 minutes to 72 minutes.
  • the delivery constructs of the present invention are polypeptides that have structural domains corresponding to domains Ia and II of PE. These structural domains perform certain functions, including, but not limited to, cell recognition and transcytosis, that correspond to the functions of the domains of PE.
  • the delivery constructs of this invention can further comprise a macromolecule for delivery to a biological compartment of a subject.
  • the macromolecule can be introduced into any portion of the delivery construct that does not disrupt a cell-binding or transcytosis activity.
  • the macromolecule is connected with the remainder of the delivery construct with a cleavable linker.
  • the delivery constructs of the invention generally comprise the following structural elements, each element imparting particular functions to the delivery construct: (1) a “receptor binding domain” that functions as a ligand for a cell surface receptor and that mediates binding of the construct to a cell; (2) a “transcytosis domain” that mediates transcytosis from a lumen bordering the apical surface of a mucous membrane to the basal-lateral side of a mucous membrane; (3) the macromolecule; and (4) a cleavable linker that connects the macromolecule to the remainder of the delivery construct.
  • a “receptor binding domain” that functions as a ligand for a cell surface receptor and that mediates binding of the construct to a cell
  • a “transcytosis domain” that mediates transcytosis from a lumen bordering the apical surface of a mucous membrane to the basal-lateral side of a mucous membrane
  • the macromolecule and (4) a cleavable linker that connects
  • the delivery constructs of the invention offer several advantages over conventional techniques for local or systemic delivery of macromolecules to a subject. Foremost among such advantages is the ability to deliver the macromolecule without using a needle to puncture the skin of the subject. Many subjects require repeated, regular doses of macromolecules. For example, diabetics must inject insulin several times per day to control blood sugar concentrations. Such subjects' quality of life would be greatly improved if the delivery of a macromolecule could be accomplished without injection, by avoiding pain or potential complications associated therewith.
  • delivery constructs can be constructed and expressed in recombinant systems.
  • Recombinant technology allows one to make a delivery construct having an insertion site designed for introduction of any suitable macromolecule. Such insertion sites allow the skilled artisan to quickly and easily produce delivery constructs for delivery of new macromolecules, should the need to do so arise.
  • connection of the macromolecule to the remainder of the delivery construct with a linker that is cleaved by an enzyme present at a basal-lateral membrane of an epithelial cell allows the macromolecule to be liberated from the delivery construct and released from the remainder of the delivery construct soon after transcytosis across the epithelial membrane. Such liberation reduces the probability of induction of an immune response against the macromolecule. It also allows the macromolecule to interact with its target free from the remainder of the delivery construct.
  • the invention provides a delivery construct that comprises a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker separates the macromolecule from the remainder of the construct.
  • the cleavable linker is cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject or in the plasma of the subject.
  • the enzyme that is at a basal-lateral membrane of a polarized epithelial cell exhibits higher activity on the basal-lateral side of a polarized epithelial cell than it does on the apical side of the polarized epithelial cell. In certain embodiments, the enzyme that is in the plasma of the subject exhibits higher activity in the plasma than it does on the apical side of a polarized epithelial cell.
  • the delivery construct further comprises a second cleavable linker.
  • the first and/or the second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10).
  • the first and/or the second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10) and is cleavable by an enzyme that exhibits higher activity on the basal-lateral side of a polarized epithelial cell than it does on the apical side of the polarized epithelial cell.
  • the first and/or the second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10) and is cleavable by an enzyme that exhibits higher activity in the plasma than it does on the apical side of a polarized epithelial cell.
  • the enzyme that is present at a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
  • the receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, botulinum toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF ⁇ ; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8.
  • the receptor binding domain binds to a cell-surface receptor that is selected from the group consisting of ⁇ 2-macroglobulin receptor, epidermal growth factor receptor, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNF ⁇ receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
  • the receptor binding domain of Pseudomonas exotoxin A is Domain Ia of Pseudomonas exotoxin A.
  • the receptor binding domain of Pseudomonas exotoxin A has an amino acid sequence that is SEQ ID NO.:1.
  • the transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, botulinum toxin, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E, coli enterotoxin, shiga toxin, and shiga-like toxin.
  • the transcytosis domain is Pseudomonas exotoxin A transcytosis domain.
  • the Pseudomonas exotoxin A transcytosis domain has an amino acid sequence that is SEQ ID NO.:2.
  • the macromolecule is selected from the group of a nucleic acid, a peptide, a polypeptide, a protein, and a lipid
  • the polypeptide is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors.
  • the polypeptide is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, and glucagon-like peptide 1.
  • the polypeptide is human growth hormone.
  • the protein is human insulin.
  • the delivery constructs further comprise a second macromolecule that is selected from the group consisting of a nucleic acid, a peptide, a polypeptide, a protein, a lipid, and a small organic molecule and a second cleavable linker, wherein cleavage at said second cleavable linker separates said second macromolecule from the remainder of said construct.
  • the first macromolecule is a first polypeptide and said second macromolecule is a second polypeptide.
  • the first polypeptide and the second polypeptide associate to form a multimer.
  • the multimer is a dimer, tetramer, or octamer.
  • the dimer is an antibody.
  • the delivery constructs of the invention generally comprise a receptor binding domain.
  • the receptor binding domain can be any receptor binding domain known to one of skill in the art without limitation to bind to a cell surface receptor that is present on the apical membrane of an epithelial cell.
  • the receptor binding domain binds specifically to the cell surface receptor.
  • the receptor binding domain should bind to the cell surface receptor with sufficient affinity to allow endocytosis of the delivery construct.
  • the receptor binding domain can comprise a peptide, a polypeptide, a protein, a lipid, a carbohydrate, or a small organic molecule, or a combination thereof. Examples of each of these molecules that bind to cell surface receptors present on the apical membrane of epithelial cells are well known to those of skill in the art.
  • Suitable peptides or polypeptides include, but are not limited to, bacterial toxin receptor binding domains, such as the receptor binding domains from PE, cholera toxin, botulinum toxin, diptheria toxin, shiga toxin, shiga-like toxin, etc.; antibodies, including monoclonal, polyclonal, and single-chain antibodies, or derivatives thereof, growth factors, such as EGF, IGF-I, IGF-II, IGF-III etc.; cytokines, such as IL-1, IL-2, IL-3, IL-6, etc; chemokines, such as MIP-1a, MIP-1b, MCAF, IL-8, etc.; and other ligands, such as CD4, cell adhesion molecules from the immunoglobulin superfamily, integrins, ligands specific for the IgA receptor, etc.
  • bacterial toxin receptor binding domains such as the receptor binding domains from PE, cholera toxin, bot
  • Lipids suitable for receptor binding domains include, but are not limited to, lipids that themselves bind cell surface receptors, such as sphingosine-1-phosphate, lysophosphatidic acid, sphingosylphosphorylcholine, retinoic acid, etc.; lipoproteins such as apolipoprotein E, apolipoprotein A, etc., and glycolipids such as lipopolysaccharide, etc.; glycosphingolipids such as globotriaosylceramide and galabiosylceramide; and the like.
  • cell surface receptors such as sphingosine-1-phosphate, lysophosphatidic acid, sphingosylphosphorylcholine, retinoic acid, etc.
  • lipoproteins such as apolipoprotein E, apolipoprotein A, etc.
  • glycolipids such as lipopolysaccharide, etc.
  • glycosphingolipids such as globotri
  • Carbohydrates suitable for receptor binding domains include, but are not limited to, monosaccharides, disaccharides, and polysaccharides that comprise simple sugars such as glucose, fructose, galactose, etc.; and glycoproteins such as mucins, selectins, and the like.
  • Suitable small organic molecules for receptor binding domains include, but are not limited to, vitamins, such as vitamin A, B 1 , B 2 , B 3 , B 6 , B 9 , B 12 , C, D, E, and K, amino acids, and other small molecules that are recognized and/or taken up by receptors present on the apical surface of epithelial cells.
  • U.S. Pat. No. 5,807,832 provides an example of such small organic molecule receptor binding domains, vitamin B 12 .
  • the receptor binding domain can bind to a receptor found on an epithelial cell. In further embodiments, the receptor binding domain can bind to a receptor found on the apical membrane of an epithelial cell.
  • the receptor binding domain can bind to any receptor known to be present on the apical membrane of an epithelial cell by one of skill in the art without limitation.
  • the receptor binding domain can bind to ⁇ 2-MR, EGFR, or IGFR.
  • An example of a receptor binding domain that can bind to ⁇ 2-MR is domain Ia of PE. Accordingly, in certain embodiments, the receptor binding domain is domain Ia of PE. In other embodiments, the receptor binding domain is a portion of domain Ia of PE that can bind to ⁇ 2-MR.
  • Exemplary receptor binding domains that can bind to EGFR include, but are not limited to, EGF and TGF ⁇ .
  • Examples of receptor binding domains that can bind to IGFR include, but are not limited to, IGF-I, IGF-II, or IGF-III.
  • the receptor binding domain is EGF, IGF-I, IGF-II, or IGF-III.
  • the receptor binding domain is a portion of EGF, IGF-I, IGF-II, or IGF-III that can bind to the EGF or IGF receptor.
  • the receptor binding domain binds to a receptor that is highly expressed on the apical membrane of a polarized epithelial cell but is not expressed or expressed at low levels on antigen presenting cells, such as, for example, dendritic cells.
  • exemplary receptor binding domains that have this kind of expression pattern include, but are not limited to, TGF ⁇ , EGF, IGF-I, IGF-II, and IGF-III.
  • the delivery constructs of the invention comprise more than one domain that can function as a receptor binding domain.
  • the delivery construct can comprise PE domain Ia in addition to another receptor binding domain.
  • the receptor binding domain can be attached to the remainder of the delivery construct by any method or means known by one of skill in the art to be useful for attaching such molecules, without limitation.
  • the receptor binding domain is expressed together with the remainder of the delivery construct as a fusion protein. Such embodiments are particularly useful when the receptor binding domain and the remainder of the construct are formed from peptides or polypeptides.
  • the receptor binding domain is connected with the remainder of the delivery construct with a linker. In yet other embodiments, the receptor binding domain is connected with the remainder of the delivery construct without a linker. Either of these embodiments are useful when the receptor binding domain comprises a peptide, polypeptide, protein, lipid, carbohydrate, nucleic acid, or small organic molecule.
  • the linker can form a covalent bond between the receptor binding domain and the remainder of the delivery construct.
  • the covalent bond can be a peptide bond.
  • the linker can link the receptor binding domain to the remainder of the delivery construct with one or more non-covalent interactions of sufficient affinity.
  • linkers that interact with each other with sufficient affinity to be useful in the delivery constructs of the invention. For example, biotin can be attached to the receptor binding domain, and streptavidin can be attached to the remainder of the molecule. In certain embodiments, the linker can directly link the receptor binding domain to the remainder of the molecule.
  • the linker itself comprises two or more molecules that associate in order to link the receptor binding domain to the remainder of the molecule.
  • exemplary linkers include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, substituted carbon linkers, unsaturated carbon linkers, aromatic carbon linkers, peptide linkers, etc.
  • the linkers can be attached to the receptor binding domain and/or the remainder of the delivery construct by any means or method known by one of skill in the art without limitation.
  • the linker can be attached to the receptor binding domain and/or the remainder of the delivery construct with an ether, ester, thioether, thioester, amide, imide, disulfide, peptide, or other suitable moiety.
  • the skilled artisan can select the appropriate linker and method for attaching the linker based on the physical and chemical properties of the chosen receptor binding domain and the linker.
  • the linker can be attached to any suitable functional group on the receptor binding domain or the remainder of the molecule.
  • the linker can be attached to sulfhydryl (—S), carboxylic acid (COOH) or free amine (—NH 2 ) groups, which are available for reaction with a suitable functional group on a linker. These groups can also be used to connect the receptor binding domain directly connected with the remainder of the molecule in the absence of a linker.
  • S sulfhydryl
  • COOH carboxylic acid
  • —NH 2 free amine
  • the receptor binding domain and/or the remainder of the delivery construct can be derivatized in order to facilitate attachment of a linker to these moieties.
  • derivatization can be accomplished by attaching suitable derivative such as those available from Pierce Chemical Company, Rockford, Ill.
  • derivatization may involve chemical treatment of the receptor binding domain and/or the remainder of the molecule. For example, glycol cleavage of the sugar moiety of a carbohydrate or glycoprotein receptor binding domain with periodate generates free aldehyde groups. These free aldehyde groups may be reacted with free amine or hydrazine groups on the remainder of the molecule in order to connect these portions of the molecule. See, e.g., U.S. Pat. No.
  • any of these methods for attaching a linker to a receptor binding domain and/or the remainder of a delivery construct can also be used to connect a receptor binding domain with the remainder of the delivery construct in the absence of a linker.
  • the receptor binding domain is coupled with the remainder of the construct using a method suitable for the particular receptor binding domain.
  • any method suitable for connecting a protein, peptide, polypeptide, nucleic acid, carbohydrate, lipid, or small organic molecule to the remainder of the delivery construct known to one of skill in the art, without limitation can be used to connect the receptor binding domain to the remainder of the construct.
  • the receptor binding domain can be connected with the remainder of the construct as described, for example, in U.S. Pat. Nos. 6,673,905; 6,585,973; 6,596,475; 5,856,090; 5,663,312; 5,391,723; 6,171,614; 5,366,958; and 5,614,503.
  • the receptor binding domain can be a monoclonal antibody.
  • the chimeric immunogen is expressed as a fusion protein that comprises an immunoglobulin heavy chain from an immunoglobulin specific for a receptor on a cell to which the chimeric immunogen is intended to bind.
  • the light chain of the immunoglobulin then can be co-expressed with the chimeric immunogen, thereby forming a light chain-heavy chain dimer.
  • the antibody can be expressed and assembled separately from the remainder of the chimeric immunogen and chemically linked thereto.
  • the delivery constructs of the invention also comprise a transcytosis domain.
  • the transcytosis domain can be any transcytosis domain known by one of skill in the art to effect transcytosis of chimeric proteins that have bound to a cell surface receptor present on the apical membrane of an epithelial cell.
  • the transcytosis domain is a transcytosis domain from PE, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, or shiga-like toxin. See, for example, U.S. Pat. Nos. 5,965,406, and 6,022,950.
  • the transcytosis domain is domain II of PE.
  • the transcytosis domain need not, though it may, comprise the entire ammo acid sequence of domain II of native PE, which spans residues 253-364 of PE.
  • the transcytosis domain can comprise a portion of PE that spans residues 280-344 of domain II of PE.
  • the amino acids at positions 339 and 343 appear to be necessary for transcytosis. See Siegall et al., 1991 , Biochemistry 30:7154-59. Further, conservative or nonconservative substitutions can be made to the amino acid sequence of the transcytosis domain, as long as transcytosis activity is not substantially eliminated.
  • a representative assay that can routinely be used by one of skill in the art to determine whether a transcytosis domain has transcytosis activity is described below.
  • the transcytosis domain is believed to permit the trafficking of the delivery construct through a polarized epithelial cell after the construct binds to a receptor present on the apical surface of the polarized epithelial cell.
  • trafficking through a polarized epithelial cell is referred to herein as “transcytosis.” This trafficking permits the release of the delivery construct from the basal-lateral membrane of the polarized epithelial cell.
  • the delivery constructs of the invention can also comprise a macromolecule.
  • the macromolecule can be attached to the remainder of the delivery construct by any method known by one of skill in the art, without limitation.
  • the macromolecule is expressed together with the remainder of the delivery construct as a fusion protein.
  • the macromolecule can be inserted into or attached to any portion of the delivery construct, so long as the receptor binding domain, the transcytosis domain, and macromolecule retain their activities.
  • the macromolecule is connected with the remainder of the construct with a cleavable linker, or a combination of cleavable linkers, as described below.
  • domain Ib spans amino acids 365 to 399, and is structurally characterized by a disulfide bond between two cysteines at positions 372 and 379.
  • This portion of PE is not essential for any known activity of PE, including cell binding, transcytosis, ER retention or ADP ribosylation activity. Accordingly, domain Ib can be deleted entirely, or modified to contain a macromolecule.
  • the macromolecule can be inserted into domain Ib. If desirable, the macromolecule can be inserted into domain Ib wherein the cysteines at positions 372 and 379 are not cross-linked, this can be accomplished by reducing the disulfide linkage between the cysteines, by deleting the cysteines entirely from the Ib domain, by mutating the cysteines to other residues, such as, for example, serine, or by other similar techniques. Alternatively, the macromolecule can be inserted into the Ib loop between the cysteines at positions 372 and 379. In such embodiments, the disulfide linkage between the cysteines can be used to constrain the macromolecule if desirable.
  • the macromolecule in embodiments where the macromolecule is inserted into domain Ib of PE, or into any other portion of the delivery construct, the macromolecule should be flanked by cleavable linkers such that cleavage at the cleavable linkers liberates the macromolecule from the remainder of the construct.
  • the macromolecule can be connected with the N-terminal or C-terminal end of a polypeptide portion of the delivery construct.
  • the method of connection should be designed to avoid interference with other functions of the delivery construct, such as receptor binding or transcytosis.
  • the macromolecule can be connected with a side chain of an amino acid of the delivery construct.
  • the macromolecule is connected with the remainder of the delivery construct with a cleavable linker, as described below.
  • the macromolecule to be delivered can be connected with the remainder of the delivery construct with one or more cleavable linkers such that cleavage at the cleavable linker(s) separates the macromolecule from the remainder of the delivery construct.
  • the macromolecule of interest can also comprise a short (1-20 amino acids, preferably 1-10 amino acids, and more preferably 1-5 amino acids) leader peptide in addition to the macromolecule of interest that remains attached to the macromolecule following cleavage of the cleavable linker.
  • this leader peptide does not affect the activity or immunogenicity of the macromolecule.
  • the macromolecule can be can be inserted into the delivery construct by any method known to one of skill in the art without limitation.
  • amino acids corresponding to the macromolecule can be inserted directly into the delivery construct, with or without deletion of native amino acid sequences.
  • all or part of the Ib domain of PE can be deleted and replaced with the macromolecule.
  • the cysteine residues of the Ib loop are deleted so that the macromolecule remains unconstrained. In other embodiments, the cysteine residues of the Ib loop are linked with a disulfide bond and constrain the macromolecule.
  • the macromolecule can be any macromolecule that is desired to be introduced into a subject.
  • the macromolecule can be a peptide, a polypeptide, a protein, a nucleic acid, a carbohydrate, a lipid, a glycoprotein, synthetic organic and inorganic compounds, or any combination thereof.
  • the macromolecule can also be a detectable compound such as a radiopaque compound, including air and barium and magnetic compounds.
  • the macromolecule can be either soluble or insoluble in water.
  • the macromolecule can be a macromolecule that can perform a desirable biological activity when introduced to the bloodstream of the subject.
  • the macromolecule can have receptor binding activity, enzymatic activity, messenger activity (i.e., act as a hormone, cytokine, neurotransmitter, or other signaling molecule), luminescent or other detectable activity, or regulatory activity, or any combination thereof.
  • the macromolecule can be conjugated to or can itself be a pharmaceutically acceptable gamma-emitting moiety, including but not limited to, indium and technetium, magnetic particles, radiopaque materials such as air or barium and fluorescent compounds.
  • the macromolecule that is delivered can exert its effects in biological compartments of the subject other than the subject's blood.
  • the macromolecule can exert its effects in the lymphatic system.
  • the macromolecule can exert its effects in an organ or tissue, such as, for example, the subject's liver, heart, lungs, pancreas, kidney, brain, bone marrow, etc.
  • the macromolecule may or may not be present in the blood, lymph, or other biological fluid at detectable concentrations, yet may still accumulate at sufficient concentrations at its site of action to exert a biological effect.
  • the macromolecule can be a protein that comprises more than one polypeptide subunit.
  • the protein can be a dimer, trimer, or higher order multimer.
  • two or more subunits of the protein can be connected with a covalent bond, such as, for example, a disulfide bond.
  • the subunits of the protein can be held together with non-covalent interactions.
  • One of skill in the art can routinely identify such proteins and determine whether the subunits are properly associated using, for example, an immunoassay.
  • Exemplary proteins that comprise more than one polypeptide chain that can be delivered with a delivery construct of the invention include, but are not limited to, antibodies, insulin, IGF I, and the like.
  • the macromolecule is a peptide, polypeptide, or protein.
  • the macromolecule comprises a peptide or polypeptide that comprises about 5, about 8, about 10, about 12, about 15, about 17, about 20, about 25, about 30, about 40, about 50, or about 60, about 70, about 80, about 90, about 100, about 200, about 400, about 600, about 800, or about 1000 amino acids.
  • the macromolecule is a protein that comprises 1, 2, 3, 4, 5, 6, 7, 8, or more polypeptides.
  • the peptide, polypeptide, or protein is a molecule that is commonly administered to subjects by injection.
  • Exemplary peptides or polypeptides include, but are not limited to, IGF-I, IGF-II, IGF-III, EGF, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, clotting factors such as factor VII, vasopressin, calcitonin parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, glucagon-like peptide 1, asparaginase, and the like.
  • the macromolecule is insulin.
  • the polypeptide is growth hormone. In even more preferred embodiments, the polypeptide is human growth hormone. In an equally preferred embodiment, the polypeptide is IFN- ⁇ , more preferably IFN ⁇ -2b. In an equally preferred embodiment, the polypeptide is insulin or proinsulin. In other embodiments, the polypeptide is green fluorescent protein.
  • the sequences of all of these macromolecules are well known to those in the art, and attachment of these macromolecules to the delivery constructs is well within the skill of those in the art using standard techniques, as discussed below.
  • the macromolecule can be selected to not be cleavable by an enzyme present at the basal-lateral membrane of an epithelial cell.
  • the assays described in the examples can be used to routinely test whether such a cleaving enzyme can cleave the macromolecule to be delivered. If so, the macromolecule can be routinely altered to eliminate the offending amino acid sequence recognized by the cleaving enzyme. The altered macromolecule can then be tested to ensure that it retains activity using methods routine in the art.
  • enzymes such as hyaluronidase, streptokinase, tissue plasminogen activator, urokinase, PGE-aden
  • chemotherapeutics such as chemotherapy or anti-tumor agents which are effective against various types of human cancers, including leukemia, lymphomas, carcinomas, sarcomas, myelomas etc.
  • doxorubicin such as, for example, doxorubicin, mitomycin, cisplatin, daunorubicin, bleomycin, actinomycin D, and neocarzinostatin.
  • antiviral agents such as reverse transcriptase inhibitors and nucleoside analogs, e.g., ddI, ddC, 3TC, ddA, AZT
  • protease inhibitors e.g., Invirase, ABT-538
  • inhibitors of in RNA processing e.g., ribavirin.
  • the macromolecule can be inactive or in a less active form when administered, then be activated in the subject.
  • the macromolecule can be a peptide or polypeptide with a masked active site.
  • the peptide or polypeptide can be activated by removing the masking moiety. Such removal can be accomplished by peptidases or proteases in the cases of peptide or polypeptide masking agents.
  • the masking agent can be a chemical moiety that is removed by an enzyme present in the subject. This strategy can be used when it is desirable for the macromolecule to be active in limited circumstances. For example, it may be useful for a macromolecule to be active only in the liver of the subject.
  • the macromolecule can be selected to have a masking moiety that can be removed by an enzyme that is present in the liver, but not in other organs or tissues.
  • exemplary methods and compositions for making and using such masked macromolecules can be found in U.S. Pat. Nos. 6,080,575, 6,265,540, and 6,670,147.
  • the macromolecule can be a pro-macromolecule that is activated by a biological activity, for example by processing, present in the subject.
  • the exemplary macromolecule proinsulin can be delivered with a delivery construct of the present invention. Following delivery of the pro-macromolecule, it can be activated in the subject by appropriate processing enzymes. While it is believed that proinsulin is processed by enzymes (the endoproteases PC2 and PC3) present in highest concentration in secretory granules of pancreatic beta-cells, it is also believed that such enzyme are present in sufficient concentration in other compartments to permit activation of the pro-macromolecule into its fully active form.
  • enzymes the endoproteases PC2 and PC3
  • pro-macromolecules including, for example, proinsulin
  • many pro-macromolecules also exhibit activity similar to that of the fully active molecule. See, for example, Desbuquois et al., 2003 , Endocrinology 12:5308-5321.
  • the pro-molecule can in many cases still exert a desirable biological activity in the subject.
  • the macromolecule to be delivered to the subject is connected with the remainder of the delivery construct with one or more cleavable linkers.
  • the number of cleavable linkers present in the construct depends, at least in part, on the location of the macromolecule in relation to the remainder of the delivery construct and the nature of the macromolecule.
  • the macromolecule can be flanked by cleavable linkers, such that cleavage at both linkers separates the macromolecule.
  • the flanking cleavable linkers can be the same or different from each other.
  • the delivery constructs can comprise a single cleavable linker.
  • each subunit of the macromolecule can be separated from the remainder of the delivery construct and/or the other subunits of the macromolecule by cleavage at the cleavable linker.
  • the cleavable linkers are generally cleavable by a cleaving enzyme that is present at or near the basal-lateral membrane of an epithelial cell.
  • a cleaving enzyme that is present at or near the basal-lateral membrane of an epithelial cell.
  • cleaving enzymes could be used that are present inside the epithelial cell, such that the cleavable linker is cleaved prior to release of the delivery construct from the basal-lateral membrane, so long as the cleaving enzyme does not cleave the delivery construct before the delivery construct enters the trafficking pathway in the polarized epithelial cell that results in release of the delivery construct and macromolecule from the basal-lateral membrane of the cell.
  • the cleaving enzyme is a peptidase. In other embodiments, the cleaving enzyme is an RNAse. In yet other embodiments, the cleaving enzyme can cleave carbohydrates.
  • Preferred peptidases include, but are not limited to, Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I. Table 1 presents these enzymes together with an amino acid sequence that is recognized and cleaved by the particular peptidase.
  • the delivery construct can comprise more than one cleavable linker, wherein cleavage at either cleavable linker can separate the macromolecule to be delivered from the delivery construct.
  • the cleavable linker can be selected based on the sequence, in the case of peptide, polypeptide, or protein macromolecules for delivery, to avoid the use of cleavable linkers that comprise sequences present in the macromolecule to be delivered. For example, if the macromolecule comprises AAL, the cleavable linker can be selected to be cleaved by an enzyme that does not recognize this sequence.
  • the cleavable linker preferably exhibits a greater propensity for cleavage than the remainder of the delivery construct.
  • many peptide and polypeptide sequences can be cleaved by peptidases and proteases.
  • the cleavable linker is selected to be preferentially cleaved relative to other amino acid sequences present in the delivery construct during administration of the delivery construct.
  • the receptor binding domain is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) intact following delivery of the delivery construct to the bloodstream of the subject.
  • the translocation domain is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) intact following delivery of the delivery construct to the bloodstream of the subject.
  • the macromolecule is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) intact following delivery of the delivery construct to the bloodstream of the subject.
  • the cleavable linker is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) cleaved following delivery of the delivery construct to the bloodstream of the subject.
  • the cleavable linker is cleaved by a cleaving enzyme found in the plasma of the subject.
  • a cleaving enzyme found in the plasma of the subject. Any cleaving enzyme known by one of skill in the art to be present in the plasma of the subject can be used to cleave the cleavable linker. Use of such enzymes to cleave the cleavable linkers is less preferred than use of cleaving enzymes found near the basal-lateral membrane of a polarized epithelial cell because it is believed that more efficient cleavage will occur in near the basal-lateral membrane.
  • cleavage mediated by a plasma enzyme is sufficiently efficient to allow cleavage of a sufficient fraction of the delivery constructs to avoid adverse effects, such plasma cleaving enzymes can be used to cleave the delivery constructs.
  • the cleavable linker can be cleaved with an enzyme that is selected from the group consisting of caspase-1, caspase-3, proprotein convertase 1, proprotein convertase 2, proprotein convertase 4, proprotein convertase 4 PACE 4, prolyl oligopeptidase, endothelin cleaving enzyme, dipeptidyl-peptidase IV, signal peptidase, neprilysin, renin, and esterase.
  • an enzyme that is selected from the group consisting of caspase-1, caspase-3, proprotein convertase 1, proprotein convertase 2, proprotein convertase 4, proprotein convertase 4 PACE 4, prolyl oligopeptidase, endothelin cleaving enzyme, dipeptidyl-peptidase IV, signal peptidase, neprilysin, renin, and esterase.
  • an enzyme that is selected from the group consisting of caspase-1, caspas
  • the cleavable linker can be any cleavable linker known by one of skill in the art to be cleavable by an enzyme that is present at the basal-lateral membrane of an epithelial cell.
  • the cleavable linker comprises a peptide.
  • the cleavable linker comprises a nucleic acid, such as RNA or DNA.
  • the cleavable linker comprises a carbohydrate, such as a disaccharide or a trisaccharide.
  • the cleavable linker is a peptide that comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10).
  • the cleavable linker can be any cleavable linker known by one of skill in the art to be cleavable by an enzyme that is present in the plasma of the subject to whom the delivery construct is administered.
  • the cleavable linker comprises a peptide.
  • the cleavable linker comprises a nucleic acid, such as RNA or DNA.
  • the cleavable linker comprises a carbohydrate, such as a disaccharide or a trisaccharide.
  • the cleavable linker is a peptide that comprises an amino acid sequence that is selected from the group consisting of amino acid sequences presented in Table 2.
  • the delivery construct comprises more than one cleavable linker. In certain embodiments, cleavage at any of the cleavable linkers will separate the macromolecule to be delivered from the remainder of the delivery construct. In certain embodiments, the delivery construct comprises a cleavable linker cleavable by an enzyme present at the basal-lateral side of a polarized epithelial membrane and a cleavable linkers cleavable by an enzyme that is present in the plasma of the subject to whom the delivery construct is administered.
  • the delivery construct comprises a cleavable linker that is cleavable by an enzyme listed in Tables 4 and 5.
  • Preferred peptidases exhibit higher activity on the basolateral side of the membrane.
  • Particularly preferred peptidases exhibit much higher (e.g., 100%, 200%, or more increase in activity relative to the apical side) on the basolateral side.
  • the cleavable linker is cleavable by an enzyme that exhibits 50% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 100% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 200% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane.
  • the cleavable linker is cleavable by an enzyme that exhibits 500% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 1,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 2,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane.
  • the cleavable linker is cleavable by an enzyme that exhibits 3,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 5,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 10,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane.
  • the results in Tables 4 and 5 indicate that certain enzymes are present in higher concentration or exhibit greater activity in certain epithelial lineages as compared to other epithelial lineages.
  • the experiments described below can be used to test whether the particular epithelial cell lineage through which a macromolecule will be delivered exhibits the desired cleavage activity.
  • the cleavage activity is present in tracheal epithelial cells, but not intestinal epithelial cells.
  • the cleavage activity is present in intestinal epithelial cells out not tracheal epithelial cells.
  • the cleavage activity is present in intestinal epithelial cells and tracheal epithelial cells.
  • the cleavable linker may be cleavable by any enzyme that preferentially cleaves at the basolateral side of an epithelial membrane as compared to the apical side of the membrane.
  • Example 6.4 describes an assay that can be used to assess the activity of such enzymes, while Table 7, appended to the end of this document, provides short names and accession numbers for every known human protease or peptidase.
  • Any cleavage sequence recognized by such proteases or peptidases that preferentially cleaves a test substrate on the basolateral side of an epithelial membrane, or in the plasma, as compared to the apical side of such a membrane can also be used in the methods and compositions of the present invention.
  • one of skill in the art can readily determine the amino acid sequence recognized by such peptidases or proteases according to standard procedures known in the art or according to the known sequences, recognized by the proteases and peptidases.
  • the examples below provide methods for identifying cleaving enzymes that are present at or near the basal-lateral membrane of a polarized epithelial cell.
  • the skilled artisan can routinely use such methods to identify additional cleaving enzymes and the chemical structure(s) identified and cleaved by such cleaving enzymes.
  • Delivery constructs comprising such cleavable linkers and methods of delivering macromolecules using delivery constructs comprising such cleavable linkers are also within the scope of the present invention, whether or not such cleaving enzymes are presented in Table 7.
  • the cleavable linker can be a cleavable linker that is cleaved following a change in the environment of the delivery construct.
  • the cleavable linker can be a cleavable linker that is pH sensitive and is cleaved by a change in pH that is experienced when the delivery construct is released from the basal-lateral membrane of a polarized epithelial cell.
  • the intestinal lumen is strongly alkaline, while plasma is essentially neutral.
  • a cleavable linker can be a moiety that is cleaved upon a shift from alkaline to neutral pH.
  • the change in the environment of the delivery construct that cleaves the cleavable linker can be any environmental change that that is experienced when the delivery construct is released from the basal-lateral membrane of a polarized epithelial cell known by one of skill in the art, without limitation.
  • the invention provides methods for local or systemic delivery of a macromolecule to a subject. These methods generally comprise administering a delivery construct of the invention to a mucous membrane of the subject to whom the macromolecule is delivered.
  • the delivery construct is typically administered in the form of a pharmaceutical composition, as described below.
  • the invention provides a method for delivering a macromolecule to a subject.
  • the method comprises contacting an apical surface of a polarized epithelial cell of the subject with a delivery construct.
  • the delivery construct comprises a receptor binding domain, a transcytosis domain, a cleavable linker, and the macromolecule to be delivered.
  • the transcytosis domain can transcytose the macromolecule to and through the basal-lateral membrane of said epithelial cell.
  • the cleavable linker can be cleaved by ah enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject or in the plasma of the subject. Cleavage at the cleavable linker separates the macromolecule from the remainder of the delivery construct, thereby delivering the macromolecule to the subject.
  • the enzyme that is present at or near a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
  • the cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.:10).
  • the receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF ⁇ ; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8.
  • the receptor binding domain binds to a cell surface receptor selected from the group consisting of ⁇ 2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNF ⁇ receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
  • a cell surface receptor selected from the group consisting of ⁇ 2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNF ⁇ receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
  • the transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin.
  • the macromolecule is selected from the group consisting of a peptide, a polypeptide, a protein, a nucleic acid, and a lipid.
  • the macromolecule is growth hormone. Even more preferably, the macromolecule is human growth hormone.
  • the invention provides a method for delivering a macromolecule to the bloodstream of a subject that results in at least about 30% bioavailability of the macromolecule, comprising administering a delivery construct comprising the macromolecule to the subject, thereby delivering at least about 30% of the total macromolecule administered to the blood of the subject in a bioavailable form of the macromolecule.
  • at least about 10% of the total macromolecule administered is bioavailable to the subject.
  • at least about 15% of the total macromolecule administered is bioavailable to the subject.
  • at least about 20% of the total macromolecule administered is bioavailable to the subject.
  • at least about 25% of the total macromolecule administered is bioavailable to the subject.
  • At least about 35% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 40% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 45% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 50% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 55% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 60% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 65% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 70% of the total macromolecule administered is bioavailable to the subject.
  • the percentage of bioavailability of the macromolecule is determined by comparing the amount of macromolecule present in a subject's blood following administration of a delivery construct comprising the macromolecule to the amount of macromolecule present in a subject's blood following administration of the macromolecule through another route of administration.
  • the other route of administration is injection, e.g., subcutaneous injection, intravenous injection, intra-arterial injection, etc.
  • the percentage of bioavailability of the macromolecule is determined by comparing the amount of macromolecule present in a subject's blood following administration of a delivery construct comprising the macromolecule to the total amount of macromolecule administered as part of the delivery construct.
  • peak plasma concentrations of the delivered macromolecule in the subject are achieved about 10 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 15 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 5 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 20 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 25 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 30 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 35 minutes after administration.
  • peak plasma concentrations of the delivered macromolecule in the subject are achieved about 40 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 45 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 50 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 55 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 60 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 90 minutes alter administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 120 minutes after administration.
  • the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 10 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 1 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 0.1 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 10 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 10 ⁇ g/ml plasma.
  • the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 1 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 0.5 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 0.1 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 10 ng/ml plasma and about 1 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 10 ng/ml plasma and about 0.5 ⁇ g/ml plasma.
  • the peak plasma concentration of the delivered macromolecule is at least about 10 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 5 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 1 ⁇ g/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 500 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 250 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 100 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 50 ng/ml plasma.
  • the peak plasma concentration of the delivered macromolecule is at least about 10 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 5 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 1 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 0.1 ng/ml plasma.
  • oral administration of a delivery construct can deliver a higher effective concentration of the delivered macromolecule to the liver of the subject than is observed in the subject's plasma.
  • Effective concentration in this context, refers to the concentration experienced by targets of the macromolecule and can be determined by monitoring and/or quantifying downstream effects of macromolecule-target interactions.
  • oral administration of the delivery construct results in absorption of the delivery construct through polarized epithelial cells of the digestive mucosa, e.g., the intestinal mucosa, followed by cleavage of the construct and release of the macromolecule at the basolateral side of the mucous membrane.
  • the blood at the basolateral membrane of such digestive mucosa is carried from this location to the liver via the portal venous system.
  • the macromolecule exerts a biological activity in the liver, such as, for example, activities mediated by growth hormone, insulin, IGF-I, etc. binding to their cognate receptors, the macromolecule is believed to exert an effect in excess of what would be expected based on the plasma concentrations observed in the subject.
  • the invention provides a method of administering a macromolecule to a subject that comprises orally administering a delivery construct comprising the macromolecule to the subject, wherein the macromolecule is delivered to the subject's liver at a higher effective concentration than observed in the subject's plasma.
  • the epithelial cell is selected from the group consisting of nasal epithelial cells, oral epithelial cells, intestinal epithelial cells, rectal epithelial cells, vaginal epithelial cells, and pulmonary epithelial cells.
  • the subject is a mammal.
  • the subject is a rodent, a lagomorph, or a primate.
  • the rodent is a mouse or rat.
  • the lagomorph is a rabbit.
  • the primate is a human, monkey, or ape.
  • the subject is a human.
  • the invention provides a method for delivering a macromolecule to the bloodstream of a subject that induces a lower titer of antibodies against the macromolecule than other routes of administration.
  • entry of the macromolecule through a mucous membrane e.g., through the intestinal mucosa, causes the immune system to tolerate the macromolecule better than if the macromolecule were, for example, injected.
  • a lower titer of antibodies against the macromolecule can be produced in the subject by delivering the macromolecule with a delivery construct of the invention through the mucosa rather than injecting the macromolecule, for example, subcutaneously, intravenously, intra-arterially, intraperitoneally, or otherwise.
  • the time at which the lower titer of antibodies detected for the alternate routes of administration is detected should be roughly comparable; for example, the titer of antibodies can be determined at about 1 week, at about 2 weeks, at about 3 weeks, at about 4 weeks, at about 2 months, or at about 6 months following administration of the macromolecule with the delivery construct or by injection.
  • the invention provides a method for delivering a macromolecule to the bloodstream a subject that comprises contacting a delivery construct of the invention that comprises the macromolecule to be delivered to an apical surface of a polarized epithelial cell of the subject, such that the macromolecule is administered to the bloodstream of the subject, wherein a lower titer of antibodies specific for the macromolecule is induced in the serum of the subject than is induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct to a subject.
  • the macromolecule is selected from the group consisting of a peptide, a polypeptide, a protein, a nucleic acid, and a lipid. In certain embodiments, the macromolecule is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors.
  • the macromolecule is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, and glucagon-like peptide 1.
  • the macromolecule is human growth hormone.
  • the macromolecule is human insulin.
  • the subject is a mouse, rat, dog, goat, or human.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 95% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 90% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 85% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 80% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 75% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 70% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 65% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 60% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 55% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 55% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 50% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 45% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 40% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 35% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 30% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 25% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than 20% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 15% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 10% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 5% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 1% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • the delivery constructs of the invention can be administered to a subject by any method known to one of skill in the art.
  • the delivery constructs are contacted to a mucosal membrane of the subject.
  • the mucosal membrane can be present in the eye, nose, mouth, trachea, lungs, esophagus, stomach, small intestine, large intestine, rectum, anus, sweat glands, vulva, vagina, or penis of the subject.
  • the mucosal membrane is a mucosal membrane present in the digestive tract of the subject, such as a mucosal membrane in the mouth, esophagus, stomach, small intestine, large intestine, or rectum of the subject.
  • the delivery constructs are preferably administered to the subject orally.
  • the delivery construct can be formulated to protect the delivery construct from degradation in the acid environment of the stomach, if necessary.
  • many embodiments of the delivery constructs of the invention comprise polypeptide domains with defined activities. Unless such delivery constructs are protected from acid and/or enzymatic hydrolysis in the stomach, the constructs will generally be digested before delivery of substantial amounts of the macromolecule to be delivered. Accordingly, composition formulations that protect the delivery construct from degradation can be used in administration of these delivery constructs.
  • a pharmaceutically effective amount of the delivery construct of the invention is administered to a subject.
  • the skilled artisan can readily determine if the dosage of the delivery construct is sufficient to deliver an effective amount of the macromolecule, as described below.
  • between about 1 ⁇ g and about 1 g of delivery construct is administered.
  • between about 10 ⁇ g and about 500 mg of delivery construct is administered.
  • between about 10 ⁇ g and about 100 mg of delivery construct is administered.
  • between about 10 ⁇ g and about 1000 ⁇ g of delivery construct is administered.
  • between about 10 ⁇ g and about 250 ⁇ g of delivery construct is administered.
  • between about 10 ⁇ g and about 100 ⁇ g of delivery construct is administered.
  • the volume of a composition comprising the delivery construct that is administered will generally depend on the concentration of delivery construct and the formulation of the composition.
  • a unit dose of the delivery construct composition is between about 0.05 ml and about 1 ml, preferably about 0.5 ml.
  • the delivery construct compositions can be prepared in dosage forms containing between 1 and 50 doses (e.g., 0.5 ml to 25 ml), more usually between 1 and 10 doses (e.g., 0.5 ml to 5 ml)
  • compositions of the invention can be administered in one dose or in multiple doses.
  • a dose can be followed by one or more doses spaced by about 1 to about 6 hours, by about 6 to about 12 hours, by about 12 to about 24 hours, by about 1 day to about 3 days, by about 1 day to about 1 week, by about 1 week to about 2 weeks, by about 2 weeks to about 1 month, by about 4 to about 8 weeks, by about 1 to about 3 months, or by about 1 to about 6 months.
  • the macromolecules to be delivered are generally macromolecules for which a large amount of knowledge regarding dosage, frequency of administration, and methods for assessing effective concentrations in subjects has accumulated. Such knowledge can be used to assess efficiency of delivery, effective concentration of the macromolecule in the subject, and frequency of administration. Thus, the knowledge of those skilled in the art can be used to determine whether, for example, the amount of macromolecule delivered to the subject is an effective amount, the dosage should be increased or decreased, the subject should be administered the delivery construct more or less frequently, and the like.
  • the methods of the invention can be used to deliver, either locally or systemically, a pharmaceutically effective amount of a macromolecule to a subject.
  • a pharmaceutically effective amount of a macromolecule to a subject.
  • the skilled artisan can determine whether the methods result in delivery of such a pharmaceutically effective amount of the macromolecule.
  • the exact methods will depend on the macromolecule that is delivered, but generally will rely on either determining the concentration of the macromolecule in the blood of the subject or in the biological compartment of the subject where the macromolecule exerts its effects. Alternatively or additionally, the effects of the macromolecule on the subject can be monitored.
  • the macromolecule that is delivered is insulin, e.g., human insulin.
  • insulin e.g., human insulin.
  • the skilled artisan can determine whether a pharmaceutically effective amount of human insulin had been delivered to the subject by, for example, taking a plasma sample from the subject and determining the concentration of human insulin therein.
  • One exemplary method for determining the concentration of human insulin is by performing an ELISA assay, but any other suitable assay known to the skilled artisan can be used.
  • one of skill in the art can determine if an effective amount of human insulin had been delivered to the subject by monitoring the blood sugar concentrations of the subject.
  • human insulin among other activities, acts on hepatocytes to promote glycogen formation, thereby reducing plasma glucose concentrations. Accordingly, the subject's plasma glucose concentration can be monitored to determine whether an effective amount of insulin had been delivered.
  • any effect of a macromolecule that is administered that is known by one of skill in the art, without limitation, can be assessed in determining whether an effective amount of the macromolecule has been administered.
  • exemplary effects include, but are not limited to, receptor binding, receptor activation, downstream effects of receptor binding, downstream effects of receptor activation, coordination of compounds, effective blood clotting, bone growth, wound healing, cellular proliferation, etc. The exact effect that is assessed will depend on the macromolecule that is delivered.
  • the invention provides polynucleotides comprising a nucleotide sequence encoding the delivery constructs. These polynucleotides are useful, for example, for making the delivery constructs.
  • the invention provides an expression system that comprises a recombinant polynucleotide sequence encoding a receptor binding domain, a transcytosis domain, and a polylinker insertion site for a polynucleotide sequence encoding a macromolecule.
  • the polylinker insertion site can be anywhere in the polynucleotide sequence so long as the polylinker insertion does not disrupt the receptor binding domain or the transcytosis domain.
  • the polylinker insertion site should be oriented near a polynucleotide sequence that encodes a cleavable linker so that cleavage at the cleavable linker separates a macromolecule encoded by a nucleic acid inserted into the polylinker insertion site from the remainder of the encoded delivery construct.
  • the polynucleotide comprises one nucleotide sequence encoding a cleavable linker between the polylinker insertion site and the remainder of the polynucleotide.
  • the polylinker insertion site can be flanked by nucleotide sequences that each encode a cleavable linker.
  • the recombinant polynucleotides are based on polynucleotides encoding PE, or portions or derivatives thereof. In other embodiments, the recombinant polynucleotides are based on polynucleotides that hybridize to a polynucleotide that encodes PE under stringent hybridization conditions.
  • a nucleotide sequence encoding PE is presented as SEQ ID NO.:3. This sequence can be used to prepare PCR primers for isolating a nucleic acid that encodes any portion of this sequence that is desired. For example, PCR can be used to isolate a nucleic acid that encodes one or more of the functional domains of PE. A nucleic acid so isolated can then be joined to nucleic acids encoding other functional domains of the delivery constructs using standard recombinant techniques.
  • a polynucleotide encoding PE, PE domains, or any other functional domain useful in the delivery constructs of the invention include, but are not limited to, reverse transcription, the polymerase chain reaction (PCR), the ligase chain reaction (LCR), the transcription-based amplification system (TAS), the self-sustained sequence replication system (3SR) and the QP replicase amplification system (QB).
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • TAS transcription-based amplification system
  • 3SR self-sustained sequence replication system
  • QB QP replicase amplification system
  • Any such technique known by one of skill in the art to be useful in construction of recombinant nucleic acids can be used.
  • a polynucleotide encoding the protein or a portion thereof can be isolated by polymerase chain reaction of cDNA using primers based on the DNA sequence of PE or a nucleotide encoding a receptor binding domain.
  • Polynucleotides encoding a delivery construct or a portion thereof also can be isolated by screening genomic or cDNA libraries with probes selected from the sequences of the desired polynucleotide under stringent, moderately stringent, or highly stringent hybridization conditions.
  • nucleic acids encoding the delivery constructs of the invention can be facilitated by introducing an insertion site for a nucleic acid encoding the macromolecule into the construct.
  • an insertion site for the macromolecule can be introduced between the nucleotides encoding the cysteine residues of domain Ib.
  • the insertion site can be introduced anywhere in the nucleic acid encoding the construct so long as the insertion does not disrupt the functional domains encoded thereby.
  • the insertion site can be in the ER retention domain.
  • a nucleotide sequence encoding a portion of the Ib domain between the cysteine-encoding residues can be removed and replaced with a nucleotide sequence that includes a cloning site cleaved by a restriction enzyme.
  • the cloning site can be recognized and cleaved by PstI.
  • a polynucleotide encoding macromolecule that is flanked by PstI sequences can be inserted into the vector.
  • polynucleotides can also encode a secretory sequence at the amino terminus of the encoded delivery construct. Such constructs are useful for producing the delivery constructs in mammalian cells as they simplify isolation of the immunogen.
  • polynucleotides of the invention also encompass derivative versions of polynucleotides encoding a delivery construct.
  • derivatives can be made by any method known by one of skill in the art without limitation.
  • derivatives can be made by site-specific mutagenesis, including substitution, insertion, or deletion of one, two, three, five, ten or more nucleotides, of polynucleotides encoding the delivery construct.
  • derivatives can be made by random mutagenesis.
  • One method for randomly mutagenizing a nucleic acid comprises amplifying the nucleic acid in a PCR reaction in the presence of 0.1 mM MnCl 2 and unbalanced nucleotide concentrations. These conditions increase the misincorporation rate of the polymerase used in the PCR reaction and result in random mutagenesis of the amplified nucleic acid.
  • these recombinant polynucleotides are particularly useful for expressing delivery constructs that have a receptor binding domain that is not domain Ia of PE.
  • the recombinant polynucleotides can optionally encode an amino-terminal methionine to assist in expression of the construct.
  • the receptor binding domain can be ligated to the 5′ end of the polynucleotide encoding the transcytosis domain.
  • nucleic acids encoding mutant forms of PE that can be used as a source of nucleic acids for constructing the delivery constructs of the invention include, but are not limited to, PE ⁇ 553 and those described in U.S. Pat. Nos. 5,602,095; 5,512,658 and 5,458,878, and in Vasil et al., 1986 , Infect. Immunol 52:538-48.
  • the invention provides a polynucleotide that encodes a delivery construct.
  • the delivery construct comprises a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the construct.
  • the cleavable linker can be cleaved by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject or in the plasma of the subject.
  • the polynucleotide hybridizes under stringent hybridization conditions to any polynucleotide of this invention. In further embodiments, the polynucleotide hybridizes under stringent conditions to a nucleic acid that encodes any delivery construct of the invention.
  • the polynucleotide encodes a delivery construct that further comprises a second cleavable linker.
  • the first and/or second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10).
  • the first and/or second cleavable linker encoded by the polynucleotide is cleavable by an enzyme that is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
  • the receptor binding domain encoded by the polynucleotide is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF ⁇ ; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8.
  • the receptor binding domain encoded by the polynucleotide binds to a cell-surface receptor that is selected from the group consisting of ⁇ 2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNF ⁇ receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
  • the receptor binding domain encoded by the polynucleotide is Domain Ia of Pseudomonas exotoxin A.
  • the receptor binding domain encoded by the polynucleotide has an amino acid sequence that is SEQ ID NO.: 1.
  • the transcytosis domain encoded by the polynucleotide is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin.
  • the transcytosis domain is Pseudomonas exotoxin A transcytosis domain.
  • the Pseudomonas exotoxin A transcytosis domain has an amino acid sequence that is SEQ ID NO.:2.
  • the macromolecule encoded by the polynucleotide is selected from the group of a peptide, a polypeptide, and a protein.
  • the polypeptide is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors.
  • the polypeptide is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN- ⁇ , IFN- ⁇ , IFN- ⁇ , G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, and glucagon-like peptide 1.
  • the polypeptide is human growth hormone.
  • the protein is human insulin.
  • the invention provides a polynucleotide that encodes a delivery construct that comprises a nucleic acid sequence encoding a receptor binding domain, a nucleic acid sequence encoding a transcytosis domain, a nucleic acid sequence encoding a cleavable linker, and a nucleic acid sequence comprising a polylinker insertion site.
  • the polylinker insertion site can be oriented relative to the nucleic acid sequence encoding a cleavable linker to allow to cleavage of the cleavable linker to separate a macromolecule that is encoded by a nucleic acid inserted into the polylinker insertion site from the remainder of said delivery construct.
  • the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of said subject or in the plasma of said subject.
  • the invention provides expression vectors for expressing the delivery constructs.
  • expression vectors are recombinant polynucleotide molecules comprising expression control sequences operatively linked to a nucleotide sequence encoding a polypeptide.
  • Expression vectors can readily be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, selectable markers, etc. to result in stable transcription and translation or mRNA. Techniques for construction of expression vectors and expression of genes in cells comprising the expression vectors are well known in the art.
  • Useful promoters for use in expression vectors include, but are not limited to, a metallothionein promoter, a constitutive adenovirus major late promoter, a dexamethasone-inducible MMTV promoter, a SV40 promoter, a MRP pol III promoter, a constitutive MPSV promoter, a tetracycline-inducible CMV promoter (such as the human immediate-early CMV promoter), and a constitutive CMV promoter.
  • the expression vectors should contain expression and replication signals compatible with the cell in which the delivery constructs are expressed.
  • Expression vectors useful for expressing delivery constructs include viral vectors such as retroviruses, adenoviruses and adenoassociated viruses, plasmid vectors, cosmids, and the like. Viral and plasmid vectors are preferred for transfecting the expression vectors into mammalian cells.
  • the expression vector pcDNA1 Invitrogen, San Diego, Calif.
  • the expression control sequence comprises the CMV promoter
  • the expression vectors can be introduced into the cell for expression of the delivery constructs by any method known to one of skill in the art without limitation. Such methods include, but are not limited to, e.g., direct uptake of the molecule by a cell from solution; facilitated uptake through lipofection using, e.g., liposomes or immunoliposomes; particle-mediated transfection; etc. See, e.g., U.S. Pat. No. 5,272,065; Goeddel et al., eds, 1990 , Methods in Enzymology , vol.
  • the expression vectors can also contain a purification moiety that simplifies isolation of the delivery construct.
  • a polyhistidine moiety of, e.g., six histidine residues, can be incorporated at the amino terminal end of the protein.
  • the polyhistidine moiety allows convenient isolation of the protein in a single step by nickel-chelate chromatography.
  • the purification moiety can be cleaved from the remainder of the delivery construct following purification. In other embodiments, the moiety does not interfere with the function of the functional domains of the delivery construct and thus need not be cleaved.
  • the invention provides a cell comprising an expression vector for expression of the delivery constructs, or portions thereof.
  • the cell is preferably selected for its ability to express high concentrations of the delivery construct to facilitate purification of the protein.
  • the cell is a prokaryotic cell, for example, E. coli .
  • the delivery constructs are properly folded and comprise the appropriate disulfide linkages when expressed in E. coli.
  • the cell is a eukaryotic cell.
  • Useful eukaryotic cells include yeast and mammalian cells. Any mammalian cell known by one of skill in the art to be useful for expressing a recombinant polypeptide, without limitation, can be used to express the delivery constructs.
  • Any mammalian cell known by one of skill in the art to be useful for expressing a recombinant polypeptide can be used to express the delivery constructs.
  • Chinese hamster ovary (CHO) cells can be used to express the delivery constructs.
  • compositions Comprising Delivery Constructs
  • the delivery constructs of the invention can be formulated as compositions.
  • the compositions are generally formulated appropriately for the immediate use intended for the delivery construct.
  • the delivery construct can be formulated in a composition suitable for storage.
  • One such composition is a lyophilized preparation of the delivery construct together with a suitable stabilizer.
  • the delivery construct composition can be formulated for storage in a solution with one or more suitable stabilizers. Any such stabilizer known to one of skill in the art without limitation can be used.
  • stabilizers suitable for lyophilized preparations include, but are not limited to, sugars, salts, surfactants, proteins, chaotropic agents, lipids, and amino acids.
  • Stabilizers suitable for liquid preparations include, but are not limited to, sugars, salts, surfactants, proteins, chaotropic agents, lipids, and amino acids.
  • Specific stabilizers than can be used in the compositions include, but are not limited to, trehalose, serum albumin, phosphatidylcholine, lecithin, and arginine.
  • Other compounds, compositions, and methods for stabilizing a lyophilized or liquid preparation of the delivery constructs may be found, for example, in U.S. Pat. Nos. 6,573,237, 6,525,102, 6,391,296, 6,255,284, 6,133,229, 6,007,791, 5,997,856, and 5,917,021.
  • the delivery construct compositions of the invention can be formulated for administration to a subject.
  • Such vaccine compositions generally comprise one or more delivery constructs of the invention and a pharmaceutically acceptable excipient, diluent, carrier, or vehicle. Any such pharmaceutically acceptable excipient, diluent, carrier, or vehicle known to one of skill in the art without limitation can be used. Examples of a suitable excipient, diluent, carrier, or vehicle can be found in Remington's Pharmaceutical Sciences, 21st Ed. 2005, Mack Publishing Co., Easton.
  • the delivery construct compositions are formulated for oral administration.
  • the compositions are formulated to protect the delivery construct from acid and/or enzymatic degradation in the stomach.
  • the delivery construct Upon passage to the neutral to alkaline environment of the duodenum, the delivery construct then contacts a mucous membrane and is transported across the polarized epithelial membrane.
  • the delivery constructs may be formulated in such compositions by any method known by one of skill in the art, without limitation.
  • the oral formulation comprises a delivery construct and one or more compounds that can protect the delivery construct while it is in the stomach.
  • the protective compound should be able to prevent acid and/or enzymatic hydrolysis of the delivery construct.
  • the oral formulation comprises a delivery construct and one or more compounds that can facilitate transit of the construct from the stomach to the small intestine.
  • the one or more compounds that can protect the delivery construct from degradation in the stomach can also facilitate transit of the construct from the stomach to the small intestine.
  • the oral formulation comprises one or more compounds that can protect the delivery construct from degradation in the stomach and facilitate transit of the construct from the stomach to the small intestine.
  • inclusion of sodium bicarbonate can be useful in facilitating the rapid movement of intra-gastric delivered materials from the stomach to the duodenum as described in Mrsny et al., 1999 , Vaccine 17:1425-1433.
  • compositions so that the delivery constructs can pass through the stomach and contact polarized epithelial membranes in the small intestine include, but are not limited to, enteric-coating technologies as described in DeYoung, 1989 , Int J Pancreatol. 5 Suppl:31-6, and the methods provided in U.S. Pat. Nos. 6,613,332, 6,174,529, 6,086,918, 5,922,680, and 5,807,832.
  • the invention provides a kit that comprises a composition of the invention.
  • the kit further comprises instructions that direct administration of the composition to a mucous membrane of the subject to whom the composition is administered.
  • the kit further comprises instructions that direct oral administration of the composition to the subject to whom the composition is administered.
  • the kit comprises a composition of the invention in more or more containers.
  • the composition can be in a unit dosage form, e.g., a tablet, lozenge, capsule, etc.
  • the composition can be provided in or with a device for administering the composition, such as, for example, a device configured to administer a single-unit dose of the composition, e.g., an inhaler.
  • the delivery constructs of the invention are preferably produced recombinantly, as described below. However, the delivery constructs may also be produced by chemical synthesis using methods known to those of skill in the art.
  • Methods for expressing and purifying the delivery constructs of the invention are described extensively in the examples below. Generally, the methods rely on introduction of an expression vector encoding the delivery construct to a cell that can express the delivery construct from the vector. The delivery construct can then be purified for administration to a subject.
  • the function of these domains, and of the delivery constructs as a whole can be routinely tested to ensure that the constructs can deliver a macromolecule across mucous membranes of a subject free from the remainder of the construct,
  • the delivery constructs can be tested for cell recognition, transcytosis and cleavage using routine assays.
  • the entire chimeric protein can be tested, or, the function of various domains can be tested by substituting them for native domains of the wild-type toxin.
  • Receptor binding domain function can be tested by monitoring the delivery construct's ability to bind to the target receptor. Such testing can be accomplished using cell-based assays, with the target receptor present on a cell surface, or in cell-free assays. For example, delivery construct binding to a target can be assessed with affinity chromatography. The construct can be attached to a matrix in an affinity column, and binding of the receptor to the matrix detected, or vice versa. Alternatively, if antibodies have been identified that bind to either the receptor binding domain or its cognate receptor, the antibodies can be used, for example, to detect the receptor binding domain in the delivery construct by immunoassay, or in a competition assay for the cognate receptor.
  • An exemplary cell-based assay that detects delivery construct binding to receptors on cells comprises labeling the construct and detecting its binding to cells by, e.g., fluorescent cell sorting, autoradiography, etc.
  • transcytosis domain The function of the transcytosis domain can be tested as a function of the delivery construct's ability to pass through an epithelial membrane. Because transcytosis first requires binding to the cell, these assays can also be used to assess the function of the cell recognition domain.
  • transcytosis activity can be tested by any method known by one of skill in the art, without limitation.
  • transcytosis activity can be tested by assessing the ability of a delivery construct to enter a non-polarized cell to which it binds. Without intending to be bound to any particular theory or mechanism of action, it is believed that the same property that allows a transcytosis domain to pass through a polarized epithelial cell also allows molecules bearing the transcytosis domain to enter non-polarized cells.
  • the delivery construct's ability to enter the cell can be assessed, for example, by detecting the physical presence of the construct in the interior of the cell.
  • the delivery construct can be labeled with, for example, a fluorescent marker, and the delivery construct exposed to the cell. Then, the cells can be washed, removing any delivery construct that has not entered the cell, and the amount of label remaining determined. Detecting the label in this traction indicates that the delivery construct has entered the cell.
  • the delivery construct's transcytosis ability can be tested by assessing the delivery construct's ability to pass through a polarized epithelial cell.
  • the delivery construct can be labeled with, for example, a fluorescent marker and contacted to the apical membranes of a layer of epithelial cells. Fluorescence detected on the basal-lateral side of the membrane formed by the epithelial cells indicates that the transcytosis domain is functioning properly.
  • the function of the cleavable linker can generally be tested in a cleavage assay. Any suitable cleavage assay known by one of skill in the art, without limitation, can be used to test the cleavable linkers. Both cell-based and cell-free assays can be used to test the ability of an enzyme to cleave the cleavable linkers.
  • An exemplary cell-free assay for testing cleavage of cleavable linkers comprises preparing extracts of polarized epithelial cells and exposing a labeled delivery construct bearing a cleavable linker to the fraction of the extract that corresponds to membrane-associated enzymes.
  • the label can be attached to either the macromolecule to be delivered or to the remainder of the delivery construct.
  • these enzymes are cleavage enzymes found near the basal-lateral membrane of a polarized epithelial cell, as described above. Cleavage can be detected, for example, by binding the delivery construct with, for example, an antibody and washing off unbound molecules.
  • the binding agent used in the assay can be specific for the macromolecule, and the remainder of the construct can be labeled. In either case, cleavage can be assessed.
  • Cleavage can also be tested using cell-based assays that test cleavage by polarized epithelial cells assembled into membranes.
  • a labeled delivery construct, or portion of a delivery construct comprising the cleavable linker can be contacted to either the apical or basolateral side of a monolayer of suitable epithelial cells, such as, for example, Coco-2 cells, under conditions that permit cleavage of the linker.
  • Cleavage can be detected by detecting the presence or absence of the label using a reagent that specifically binds the delivery construct, or portion thereof.
  • an antibody specific for the delivery construct can be used to bind a delivery construct comprising a label distal to the cleavable linker in relation to the portion of the delivery construct bound by the antibody. Cleavage can then be assessed by detecting the presence of the label on molecules bound to the antibody. If cleavage has occurred, little or no label should be observed on the molecules bound to the antibody.
  • enzymes that preferentially cleave at the basolateral membrane rather than the apical membrane can be identified, and, further, the ability of such enzymes to cleave the cleavable linker in a delivery construct can be confirmed.
  • cleavage can also be tested using a fluorescence reporter assay as described in U.S. Pat. No. 6,759,207. Briefly, in such assays, the fluorescence reporter is contacted to the basolateral side of a monolayer of suitable epithelial cells under conditions that allow the cleaving enzyme to cleave the reporter. Cleavage of the reporter changes the structure of the fluorescence reporter, changing it from a non-fluorescent configuration to a fluorescent configuration. The amount of fluorescence observed indicates the activity of the cleaving enzyme present at the basolateral membrane.
  • cleavage can also be tested using an intra-molecularly quenched molecular probe, such as those described in U.S. Pat. No. 6,592,847.
  • probes generally comprise a fluorescent moiety that emits photons when excited with light of appropriate wavelength and a quencher moiety that absorbs such photons when in close proximity to the fluorescent moiety. Cleavage of the probe separates the quenching moiety from the fluorescent moiety, such that fluorescence can be detected, thereby indicating that cleavage has occurred.
  • probes can be used to identify and assess cleavage by particular cleaving enzymes by contacting the basolateral side of a monolayer of suitable epithelial cells with the probe under conditions that allow the cleaving enzyme to cleave the probe. The amount of fluorescence observed indicates the activity of the cleaving enzyme being tested.
  • exemplary delivery construct expression vectors for delivering rat growth hormone were constructed according to the following protocol.
  • the rGH gene was amplified by PCR, incorporating restriction enzymes pairs of NdeI and EcoRI, PstI and PstI, AgeI and EcoRI, or PstI and EcoRI sites at two ends of the PCR products.
  • restriction enzyme digestion the PCR products were cloned into pPE64-PstI- ⁇ 553, which was digested with the corresponding restriction enzyme pairs.
  • constructs were named as pPE-RGH(NdeI-EcoRI), pntPE-RGH(PstI), pntPE-RGH(AgeI-EcoRI), and pPE-RGH(PstI-EcoRI).
  • These constructs thus comprise sequences encoding Domains I and II of ntPE (amino acids 26-372 as shown in FIG. 3 ) and rGH (Accession No. P01244; see Seeburg et al., 1977 , Nature 270:486-494 and Page et al., 1981 , Nucleic Acids Res. 9:2087-2104), and are also tagged with a 6-His motif at the N-terminus of the polypeptide to facilitate purification.
  • the final plasmids were verified by restriction enzyme digestions and DNA sequencing.
  • Expression vectors comprising cleavable linkers were constructed by introducing sequences encoding the appropriate amino acid sequence. To do so, oligonucleotides that encode sequences complementary to appropriate restriction sites and one of the following amino acid sequences were synthesized, then ligated into an expression vector prepared as described above between the ntPE sequences and the rGH sequences.
  • the cleavable linker sequence was RQPRGGL.
  • the cleavable linker sequence was GGLRQPR.
  • the cleavable linker sequence was RQPREGR.
  • the cleavable linker sequence was RQPRVGR.
  • the cleavable linker sequence was RQPRARR.
  • a protease furin site was also inserted between the cleavable linker and rGH. To do so, constructs containing a sequence encoding the furin site with the five different cleavable linkers were made. Oligonucleotide sequences for the five cleavable linkers and a furin clip site are shown in Table 3 below. Each of the oligo duplexes was inserted into PstI site of pPE-RGH(PstI-EcoRI). The final constructs, named as pPE-RGH-F1, pPE-RGH-F2, pPE-RGH-F3, pPE-RGH-F4, pPE-RGH-F5 were confirmed by restriction enzyme digestion and DNA sequencing.
  • E. coli BL21(DE3) pLysS competent cells (Novagen, Madison, Wis.) were transformed using a standard heat-shock method in the presence of the appropriate plasmid to generate ntPE-rat Growth Hormone (rGH) expression cells, selected on ampicillin-containing media, and isolated and grown in Luria-Bertani broth (Difco; Becton Dickinson, Franklin Lakes, N.J.) with antibiotic, then induced for protein expression by the addition of 1 mM isopropyl-D-thiogalactopyranoside (IPTG) at OD 0.6. Two hours following IPTG induction, cells were harvested by centrifugation at 5,000 rpm for 10 min.
  • IPTG isopropyl-D-thiogalactopyranoside
  • the following procedures can be used to assess proper refolding of a delivery construct.
  • the protein refolding process is monitored by measuring, e.g., Delivery Construct 1 binding activity with ntPE binding receptor, CD 91 receptors, and rGH binding proteins on a Biacore SPR instrument (Biacore, Sweden) according to the manufacturer's instructions.
  • Proper refolding of other macromolecules in exemplary constructs can be tested in similar binding assays with appropriate binding agents. By testing such binding affinities, the skilled artisan can assess the proper folding of each portion of the delivery construct.
  • HTE or Caco-2 cells were seeded at a density of 5 ⁇ 10 4 cells/cm 2 onto 24-well collagen-coated polycarbonate transwell filters (Corning, Acton, Mass.) for 12-14 days.
  • Confluent monolayers achieved a transepithelial resistance (TER) of >500 ohm ⁇ cm 2 , as measured using an EVOM epithelial voltohmmeter and STX2 electrode (World Precision Instruments, Sarasota, Fla.).
  • substrates specific for the tested peptidase 500 ⁇ M or 1 mM substrate in 250 ⁇ l DMEM without FBS or antibiotics were added to either the apical (AP) or basolateral (BL) side of the monolayers.
  • Peptidase substrates were obtained from Calbiochem, Inc. (Division of EMD Biosciences, Inc., San Diego, Calif.). Cells were incubated for 2 hrs at 37° C. in a 5% CO 2 /95% air atmosphere. Both the apical and basolateral media was then measured for its specific enzyme activity according to the manufacturer's instruction. Cleavage was assessed by detecting fluorescence of the substrates, which reflects cleavage because it separates of the quenching agent from the fluorescent agent present on the substrate, which separation allows fluorescence to be detected.
  • Table 4 presents a summary of the results of these assays using HTE cells, while Table 5 presents a summary of the results of these assays using Caco-2 cells.
  • baseline control values were subtracted from substrate values before percentages were determined and tests were performed at least in duplicate.
  • the percentages presented in the tables represent the percent increase observed in assay in the apical or basolateral media, which depends on which side of the membrane exhibits higher peptidase activity. It should be noted that, even when substrate was added to the media on the apical side of the membrane, peptidase activity can be observed on the basolateral side of the membrane because of diffusion of the substrate across the membrane.
  • the following example describes experiments performed to assess the transcytosis of an exemplary delivery construct for delivering green fluorescent protein (“GFP”) across a mouse epithelial membrane. It is noted that this exemplary delivery construct does not comprise a cleavable linker; however, the presence or absence of the cleavable linker should not affect transcytosis of the delivery construct.
  • GFP green fluorescent protein
  • a nt-PE-GFP construct was applied to the trachea of anesthetized female balb/c mice which were approximately 8 weeks of age.
  • the mice were anesthetized with inhaled isoflurane and the trachea was exposed. A small hole was made on the trachea to allow application of our GFP material.
  • 100 ⁇ g of GFP alone or ntPE-GFP was used, respectively.
  • the GFP material was slowly dripped directly onto the exposed trachea in a 100 ⁇ l volume. After 15 minutes, the mice were euthanized by CO 2 asphyxiation.
  • the trachea was removed and frozen in OCT (cat#25608-930-Tissue Tek) using biopsy cryomolds (cat#4565-Tissue Tek). The samples were sectioned onto slides and visualized by fluorescence microscopy (Nikon model Eclipse E400).
  • FIGS. 1A-1C Micrographs of the epithelial sections are presented as FIGS. 1A-1C .
  • FIG. 1A shows the nt-PE-GFP construct adhering strongly to the apical surface of the trachea epithelium.
  • FIG. 1B shows transcytosis of the nt-PE-GFP construct across the trachea epithelium.
  • FIG. 1C shows release of the nt-PE-GFP construct from the basolateral side of the trachea epithelium.
  • FIG. 1D presents a micrograph of a negative control, a tracheal epithelial section from a mouse contacted with GFP alone. The tissues exposed for 15 minutes to obtain these micrographs.
  • nt-PE-GFP interacts strongly with receptors on the apical surface of mouse tracheal epithelium, transcytoses across such epithelial tissue, and releases from the basolateral surface of the mouse tracheal epithelium.
  • plasma concentrations of the nt-PE-GFP construct were determined following administration of the delivery construct using an ELISA assay as described in Example 6.6.1, below. Serum samples were taken from anesthetized mice that had received intranasal administration of 100 ⁇ g of the nt-PE-GFP delivery construct every 30 minutes following administration.
  • FIG. 2 presents the results of this experiment, demonstrating that peak plasma levels of the delivery construct reached between 500-900 ng/ml, indicating that the delivery construct displayed approximately 22% bioavailability following intranasal administration.
  • This example describes histological detection in tissues of a representative macromolecule for delivery, growth hormone.
  • animals are euthanized by CO 2 asphyxiation and exanguinated by cardiac puncture.
  • Specific tissues (lymph nodes, trachea, brain, spleen liver, GI tract) are removed, briefly rinsed in PBS to remove any residual blood and frozen in OCT. Sections (5 microns thick) are placed onto slides. Slides are fixed in acetone for 10 min and rinsed with PBS. Slides are incubated with 3% peroxidase for 5 min. Slides are then blocked with protein for an additional 5 min.
  • Primary growth hormone antibody is incubated onto slides for 30 min at a 1:100 dilution followed by PBS washes.
  • Biotin-labeled secondary antibody is then incubated for approximately 15 minutes followed by PBS washes.
  • Streptavidin HRP label is incubated onto slides for 15 min followed by PBS washes.
  • HRP Chromagen is applied for 5 min followed by several rinses in distilled H 2 0. Finally, the slides are counterstained with hematoxylin for 1 min, coverslipped, and examined for the presence of GH.
  • This example describes transport and cleavage of an exemplary delivery construct, Delivery Construct 2, comprising rat growth hormone (rGH) in an in vitro system using human tracheal epithelial cells.
  • Delivery Construct 2 comprising rat growth hormone (rGH) in an in vitro system using human tracheal epithelial cells.
  • HTE Human tracheal epithelial cells were isolated from tracheas as previously described and cultured on semi-permeable filter systems (0.45 um pore size; Corning, Acton, Mass.) coated with human placental collagen. See Yamaya et al., 1992 , Am J Physiol, 262:L713-24 and Sachs et al., 2003 , In Vitro Cell Dev Biol Anim, 39:56-62. Cell sheets were used at >10 days following plating, at which time they had a transepithelial resistance (TER) of >100 ⁇ ohms ⁇ cm 2 as measured with a “chopstick” voltohmeter (Millicell ERS, Manassas, Va.).
  • TER transepithelial resistance
  • Caco-2 cells in passage 21 were obtained from American Type Culture Collection (Manassas, Va.). Cells were routinely grown on 75 cm 2 plastic culture flasks (Becton Dickinson, Franklin Lakes, N.J.) in DMEM containing 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin at 37° C. in a 5% CO 2 /95% air atmosphere.
  • FBS fetal bovine serum
  • Caco-2 cells were seeded at a density of 5 ⁇ 10 4 cells/cm 2 onto 24-well collagen-coated polycarbonate transwell filters (Corning, Acton Mass.) for 12-14 days. Confluent monolayers achieved a transepithelial resistance (TER) of >500 ohm/cm 2 , as measured using the EVOM and STX2 electrode (World Precision Instruments).
  • TER transepithelial resistance
  • Delivery Constructs 1 and 2 proteins (10 ⁇ g in 100 ⁇ l DMEM without phenol red, FBS or antibiotics) were added to the apical side of the epithelial monolayer. Cells were incubated for 4 hrs at 37° C. in a 5% CO 2 /95% air atmosphere. Both the apical and basolateral media was then assayed for its transport and cleavage activity by testing for the presence of rGH cleaved from the delivery construct by western blot analysis, as described below. As a control, 10 ⁇ g of dextran fluorescein was also added to the apical wells in order to check for leakage.
  • TCA trichloroacetic acid
  • the Blocking Solution, Antibody Diluents, and Antibody Wash Solution from Invitrogen's WesternBreeze were used in subsequent steps.
  • the nitrocellulose membrane was placed in blocking buffer and incubated at 4° C., overnight. The membrane was washed 3 times for 3 minutes each time. The membrane was then incubated at RT with 10 ml of 1:2000 rabbit anti growth hormone (RDI RDIRtGHabr). After one hour, the membrane was again rinsed 3 times for 3 minutes each. Membrane was incubated for 1 hour at room temperature with 10 ml of goat anti rabbit IgG AP (Pierce 31340) at 1:5000. The membrane was rinsed 3 times for 3 minutes per wash.
  • RDI RDIRtGHabr 1:2000 rabbit anti growth hormone
  • results of the Western Blot analysis are presented in FIG. 4 .
  • media from the basolateral side of the epithelial cell layer contained protein consistent with cleaved rGH separated from the remainder of Delivery Construct 2.
  • media from the apical side of the epithelial cell layer contained largely intact delivery construct.
  • Application of Delivery Construct 2 to the apical side of the human epithelial cell membrane resulted in both transport to the basolateral side of the membrane and proper cleavage of the construct as shown by release of rGH detectable with anti-r(1H antibody and of proper apparent molecular weight. Similar results were also observed for Delivery Construct 1 (data not shown).
  • This example describes use of exemplary Delivery Construct 2 in a mouse model, showing effective transport and cleavage of the delivery construct in vivo and the bioactivity of the macromolecule delivered by Delivery Construct 2, rGH.
  • Delivery Construct 2 100 ⁇ g of Delivery Construct 2 (in 250 ⁇ l total volume) was orally delivered to female BALB/c mice, 5-6 weeks of age (Charles River Laboratories, Wilmington, Mass.). Delivery Construct 2 was diluted in 1 mg/ml bovine serum albumin (BSA) and phosphate buffered saline (PBS). As a positive control, control mice were subcutaneously (SC) injected on their dorsal side with 30 ⁇ g of recombinant rat growth hormone (rGH) diluted in PBS (100 ⁇ l total volume). At specific times after oral gavage and SC administration, mice were euthanized by CO 2 asphyxiation and exsanguinated. Whole blood and liver were collected and analyzed as described below. Because in the difference in molecular weights between Delivery Construct 2 and rGH, essentially the same number of rGH molecules were administered in both routes.
  • BSA bovine serum albumin
  • PBS phosphate buffered saline
  • ELISA assays were used to measure serum concentrations of rGH at defined timepoints following administration. The serum concentration data thus obtained was used to compare the pharmacokinetics of rGH administered with Delivery Construct 2 to those observed with conventional subcutaneous administration. The ELISA assays were performed as follows.
  • Costar 9018 E.I.A./R.I.A. 96-well plates were coated overnight with 200 ng/well of goat anti-rGH (Diagnostics Systems Laboratories, Cat. No. R01235) in 0.2M NaHCO 3 —Na 2 CO 3 , pH 9.4. Each 96-well plate was washed four times with PBS containing 0.05% Tween 20-0.01% thimerosal (wash buffer); blocked for 1 h with 200 ⁇ l/well of PBS/Tween 20 containing 0.5% BSA-0.01% thimerosal (assay buffer). The standard curve was prepared using recombinant rat GH (Diagnostics Systems Laboratories, Cat. No.
  • R01205 diluted in assay buffer (PBST-0.5% BSA).
  • the first point of the standard curve was prepared by adding 50 ⁇ l recombinant rat GH to 10 ml assay buffer (1:200), vortexed, and moved 200 ⁇ l to 800 ⁇ l assay buffer (1:5). For each plate, 0.5 ml was moved to 0.5 ml assay buffer by doing 1:2 serial dilutions for all the subsequent points.
  • the 10 points of the standard curve are: 100, 50, 25, 12.5, 6.25, 3.125, 1.56, 0.78, 0.39 and 0.195 ng/well. Samples were diluted at 1:10 with assay buffer, loaded 100 ⁇ l/well in triplicates onto a 96-well plate, and incubated overnight.
  • Each 96-well plate was then washed four times with wash buffer, loaded 100 ⁇ l/well of 2 nd Ab (rabbit anti-rGH, Cell Sciences, Cat. No. PAAC1) at 1:300 in assay buffer (PBST-0.5% BSA) and incubate at RT for four hours.
  • Each 96-well plate was then washed four times with wash buffer, loaded 100 ⁇ l/well of 3rd Ab (goat anti-rabbit IgG-horseradish peroxidase (HRP), Pierce, Cat. No. 31460) at 1:2000 in assay buffer (PBST-0.5% BSA) and incubated at room temperature for two hours. All incubation and coating steps were performed at room temperature on a shaker at 6 RPM.
  • ELISA results are reported as the averages of the triplicate OD (450 nm) value of each sample. Rat GH concentrations were determined by the exceeding mean value plus three times the standard error of the mean (SEM) of the appropriate control value.
  • FIG. 7 presents a graphical representation comparing pharmacokinetics of rGH delivered subcutaneously and with Delivery Construct 2.
  • the curve fitting comparison between rGH (SC) and Delivery Construct 2(Oral) was performed by evaluating ELISA data as described above using PK Solutions 2.0, Pharmacokinetics Data Analysis (Summit Research Services, Montrose, Colo.).
  • Delivery Construct 2 yields substantially higher peak serum rGH concentrations than subcutaneous administrations of rGH. Further, the peak serum concentration is achieved faster with Delivery Construct 2 relative to subcutaneous rGH.
  • the bioavailability of rGH delivered with Delivery Construct 2 observed was about 60% relative to rGH administered by subcutaneous injection.
  • This example describes analysis of the biological effects of an exemplary macromolecule, rGH, delivered with Delivery Construct 2 in an in vivo system.
  • insulin-like growth factor I-binding protein 3 IGF-1-BP3
  • GH growth hormone
  • IGF-I insulin-like growth factor I
  • IGF-1-BP3 mRNA expression is believed to be the most reliable indicator of GH receptor activation. See, e.g., Sondergaard et al., 2003 , Am J Physiol Endocrinol Metab 285:E427-32.
  • Quantitative Real Time PCR was used to detect and quantify the amount of IGF-1-BP3, GH receptor, IGF-I, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA in approximately 30 mg of mouse liver tissue prepared as described above. Collected liver tissue was stored at ⁇ 70° C. until further processing. Real-time detection of PCR was performed using the Applied Biosystems 7300 Real Time PCR system (Applied Biosystems, Foster City, Calif.). Total RNA from mouse liver was isolated according to the RNeasy Protect Mini Kit (Qiagen).
  • the primers used to amplify the cDNA were designed using Primer Express software (Applied Biosystems), synthesized by Operon (Alameda, Calif.), and are shown in Table 6:
  • IGF-I-BP3 forward: CGCAGAGAAATGGAGGACACA
  • IGF-I-BP3 reverse): GGACGCCTCTGGGACTCA
  • GH receptor GTTGACGAAATAGTGCAACCTGAT forward: GH receptor CACGAATCCCGGTCAAACTAA
  • IGF-I reverse: GCTCCGGAAGCAACACTCA
  • GAPDH forward: GCAACAGGGTGGTGGACCT
  • GAPDH reverse): GGATAGGGCCTCTCTTGCTCA
  • Equal amounts of cDNA were used in duplicate and amplified with the SYBR Green I Master Mix (Applied Biosystems).
  • the thermal cycling parameters were as follows: thermal activation for 10 min at 95° C., and 40 cycles of PCR (melting for 15 s at 95° C. and annealing/extension for 1 min at 60° C.).
  • a standard curve was constructed with a dilution curve (1:10, 1:100, 1:500, 1:1,000, 1:2,000) of total RNA from a control mouse liver sample.
  • a “no template control” was included with each PCR.
  • Amplification efficiencies were validated and normalized against GAPDH. Correct PCR product size was confirmed by electrophoresis through a 1% agarose gel stained with ethidium bromide. Purity of the amplified PCR products was determined by a heat-dissociation protocol.
  • FIG. 8 shows expression levels of IGF-1-BP3 mRNA in the liver of female BALB/c mice treated with 30 ⁇ g recombinant rGH by subcutaneous injection or with 100 ⁇ g of Delivery Construct 2 by oral gavage.
  • Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-1-BP3, as described above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • Delivery Construct 2 caused an almost 400% increase in IGF-1-BP3 mRNA expression levels in the liver 30 minutes following administration.
  • oral administration of Delivery Construct 2 effectively delivered rGH to the bloodstream of the test mice, thereby demonstrating that a delivery construct can effectively deliver an active macromolecule across a mucous membrane in an in vivo system.
  • Delivery Construct 2 delivered more active rGH to the liver than subcutaneous administration, and the effects of administration of rGH were observed substantially faster than possible with subcutaneous administration of rGH.
  • FIG. 9 shows expression levels of growth hormone (GH) receptor mRNA in the liver of female B ALB/c mice treated with recombinant rat growth hormone (rGH) by subcutaneous injection (30 ⁇ g) or Delivery Construct 2 by oral gavage (100 ⁇ g).
  • Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for GH receptor, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • Delivery Construct 2 caused such mRNA levels to decrease to about 15% of those observed prior to administration.
  • oral administration of Delivery Construct 2 is effective to deliver rGH to the bloodstream of a subject, and further, that Delivery Construct 2 delivers significantly more active rGH to mouse liver than conventional subcutaneous administration of rGH as shown by the enhanced downregulation of GH receptor mRNA expression.
  • FIG. 10 shows expression levels of insulin-like growth factor I (IGF-I) mRNA in the liver of female BALB/c mice treated with recombinant rat growth hormone (rGH) by subcutaneous injection (30 ⁇ g) or Delivery Construct 2 by oral gavage (100 ⁇ g).
  • IGF-I insulin-like growth factor I
  • rGH recombinant rat growth hormone
  • Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-I, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • GPDH glyceraldehyde-3 phosphate dehydrogenase
  • This example shows that an exemplary macromolecule, rGH, administered orally with Delivery Construct 2, is less immunogenic than rGH administered subcutaneously.
  • the serum titer of anti-rGH IgG antibodies from oral administration of 3, 10, or 30 ⁇ g Delivery Construct 2 or 3 or 10 ⁇ g subcutaneous rGH was determined in an ELISA assay. To do so, 100 ⁇ l 1 ng/ ⁇ l recombinant rGH diluted in coating buffer (0.2 M NaHCO 3 —Na 2 CO 3 , pH 9.4) was added to Costar EIA/RIA plates, then incubated at room temperature for 16-24 hours. Next, the plates were washed 4 times with 300 ⁇ l wash buffer (phosphate-buffered saline). The plates were then blocked with 200 ⁇ l blocking buffer (0.5% BSA in phosphate-buffered saline) and incubated at room temperature for 1 hour. Next, the plates were again washed four times with 300 ⁇ l wash buffer.
  • coating buffer 0.2 M NaHCO 3 —Na 2 CO 3 , pH 9.4
  • FIGS. 11A-D Representative results of the ELISAs are shown in FIGS. 11A-D .
  • the graphs presented in these figures demonstrate that 3, 10, and 30 ⁇ g Delivery Construct 2 administered orally elicited a lower titer of anti-rGH IgG antibodies than either 3 or 10 ⁇ g subcutaneous rGH.
  • subcutaneous administration of 10 ⁇ g rGH caused a substantial anti-rGH IgG response in all eight mice, while the eight mice administered 3, 10, or 30 ⁇ g Delivery Construct 2 by oral gavage had minimal anti-rGH IgG responses.
  • these observations were consistent whether the sera were diluted 1:25 ( FIGS. 11A and 11C ) or 1:200 ( FIGS. 11B and 11D ).
  • each mouse administered 3, 10, or 30 ⁇ g Delivery Construct 2 experienced a minimal immune response against rGH, as shown by the tight clustering of the data points in FIGS. 11C and D.
  • oral administration of Delivery Construct 2 not only delivers more active rGH to the liver than possible with subcutaneous injection, but further, the active rGH is less immunogenic when administered orally with Delivery Construct 2 compared to subcutaneous administration.
  • This example describes construction of an exemplary delivery construct for delivering human growth hormone, termed Delivery Construct 6. Techniques similar to those described in Example 6.1, above, were used to construct a plasmid used to express Delivery Construct 6. The nucleotide sequence of the portion of this plasmid that encodes the exemplary delivery construct is presented as FIG. 12 , while the amino acid sequence of the delivery construct is presented as FIG. 13 .
  • This example describes construction of an exemplary delivery construct for delivering IFN ⁇ (in this case, IFN ⁇ -2b), termed Delivery Construct 7.
  • IFN ⁇ IFN ⁇ -2b
  • Delivery Construct 7 Techniques similar to those described in Example 6.1, above, were used to construct a plasmid used to express Delivery Construct 7.
  • the nucleotide sequence of the portion of this plasmid that encodes the exemplary Delivery Construct 7 is presented as FIG. 14
  • the amino acid sequence of Delivery Construct 7 is presented as FIG. 15 .
  • This example demonstrates the use of Delivery Construct 7 to deliver IFN ⁇ -2b to the bloodstream of a subject in a mouse model system.
  • Deli very Construct 7 (in 250 ⁇ l total volume) was orally administered to female BALB/c mice, 5-6 weeks of age (Charles River Laboratories, Wilmington, Mass.). Delivery Construct 7 was diluted in 1 mg/ml bovine serum albumin (BSA) and phosphate buffered saline (PBS). At specific times after oral gavage and SC administration, mice were euthanized by CO 2 asphyxiation and exsanguinated. Whole blood was collected and analyzed as described below.
  • BSA bovine serum albumin
  • PBS phosphate buffered saline
  • ELISA assays were used to measure serum concentrations of IFN ⁇ -2b in one mouse immediately following administration and in three mice at 15, 30, and 75 minutes following administration. The ELISA assays were performed using R&D Systems Serum ELISA Kit No. 41110-1 according to the manufacturer's instructions.
  • ELISA results are reported as the averages of the triplicate OD (450 nm) value of each sample.
  • the results of the ELISA assays are presented in FIG. 16 .
  • IFN ⁇ -2b was detected at low (about 3 ng/ml) concentration 15 minutes after administration.
  • serum concentration of IFN ⁇ -2b was about 43 ng/ml.
  • serum concentration of IFN ⁇ -2b had fallen to about 13 ng/ml.
  • Delivery Construct 8 This example describes construction of an exemplary delivery construct for delivering proinsulin, termed Delivery Construct 8. Techniques similar to those described in Example 6.1, above, are used to construct a plasmid used to express Delivery Construct 8. The amino acid sequence of Delivery Construct 8 is presented as FIG. 17 .
  • Delivery Construct 9 This example describes construction of an exemplary delivery construct for delivering insulin, termed Delivery Construct 9. Techniques similar to those described in Example 6.1, above, are used to construct a plasmid used to express Delivery Construct 9, with certain modifications.
  • the scheme used to express Delivery Construct 9 is modified because insulin comprises two separate amino acid chains.
  • the B-chain of insulin is expressed together with the remainder of the Delivery Construct, constructed according to the general scheme presented in Example 6.1.
  • Amino acids corresponding to the A-chain are made either synthetically (e.g., chemically synthesizing the A-chain peptide from amino acids) or recombinantly (e.g., expressed in a suitable recombinant system such as, for example, E. coli , yeast, etc.)
  • the two polypeptides are then combined under conditions that permit association of the A-chain and B-chain.
  • disulfide bonds are made between the two chains of insulin as found in native insulin by application of mildly oxidizing conditions.
  • the amino acid sequence of the two amino acid chains of Delivery Construct 9 is presented as FIG. 17 .
  • the present invention provides, inter alia, delivery constructs and methods of inducing an immune response in a subject. While many specific examples have been provided, the above description is intended to illustrate rather than limit the invention. Many variations of the invention will become apparent to those skilled in the art upon review of this specification. The scope of the invention should, therefore, be determined not with reference to the above description, but instead should be determined with reference to the appended claims along with their full scope of equivalents.

Abstract

Methods and compositions for needleless delivery of macromolecules to the bloodstream of a subject are provided herein. In one aspect, the invention provides a delivery construct, comprising a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Generally, the cleavable linker is cleavable by an enzyme present in higher concentration at or near the basal-lateral membrane of a polarized epithelial cell or in the plasma than elsewhere in the body, for example, at the apical side of the polarized epithelial cell. In other aspects, the invention provides nucleic acids encoding delivery constructs of the invention, kits comprising delivery constructs of the invention, cells expressing delivery constructs of the invention, and methods of using delivery constructs of the invention.

Description

  • This application is entitled to and claims benefit of U.S. Provisional Application No. 60/615,970, filed Oct. 4, 2004, of U.S. Provisional Application No. 60/684,484, filed May 24, 2005, and of U.S. Provisional Application No. 60/718,907, filed Sep. 19, 2005, each of which is hereby incorporated by reference in its entirety.
  • 1. FIELD OF THE INVENTION
  • The present invention relates, in part, to methods and compositions for needleless delivery of macromolecules to a subject. In one aspect, the methods and compositions involve administering to the subject a delivery construct comprising the macromolecule to be delivered, wherein the macromolecule is linked to the remainder of the construct with a linker that is cleavable at a basal-lateral membrane of a polarized epithelial cell.
  • 2. BACKGROUND
  • Advances in biochemistry and molecular biology have resulted identification and characterization of many therapeutic macromolecules, including, for example, growth hormone, erythropoietin, insulin, IGF, and the like. Administration of these molecules can result in drastic improvements in quality of life for subjects afflict with a wide range of ailments.
  • However, administration of these therapeutic macromolecules remains problematic. Currently, therapeutic macromolecules are typically administered by injection. Such injections require penetration of the subject's skin and tissues and are associated with pain. Further, penetration of the skin breaches one effective nonspecific mechanism of protection against infection, and thus can lead to potentially serious infection.
  • Previous attempts have been made for needleless delivery of macromolecules to subjects. See, e.g., International Patent Publication No. WO 01/30,392. In these efforts, delivery vehicles are used to deliver macromolecules to a subject through polarized epithelial cells. However, these derivatives lack a cleavable linker that is cleavable by an enzyme at the basal-lateral membrane of a polarized epithelial cell. Without cleavage, the probability of induction of an immune response against the delivery vehicle and/or the macromolecule to be delivered is increased. Also, steric hindrance between the delivery vehicle and the macromolecule can reduce the activity of the macromolecule, reducing efficacy the treatment.
  • Accordingly, there is an unmet need for new methods and compositions that can be used to administer macromolecules to subjects without breaching the skin of the subject. This and other needs are met by the methods and compositions of the present invention.
  • 3. SUMMARY OF THE INVENTION
  • The delivery constructs of the invention comprise a macromolecule for delivery to a subject that is linked to the remainder of the construct with a cleavable linker. The linker is cleavable by an enzyme or an environmental cue that is present at the basal-lateral membrane of an epithelial cell.
  • Accordingly, in certain aspects, the invention provides a delivery construct comprising a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the delivery construct. In certain embodiments, the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject. In other embodiments, the cleavable linker can be cleavable by an enzyme that is present in the plasma of said subject.
  • In another aspect, the invention provides a polynucleotide that encodes a delivery construct comprising a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the delivery construct. In certain embodiments, the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject. In other embodiments, the cleavable linker can be cleavable by an enzyme that is present in the plasma of said subject.
  • In other embodiments, the polynucleotide that encodes a delivery construct comprises a nucleic acid sequence encoding a receptor binding domain, a nucleic acid sequence encoding a transcytosis domain, a nucleic acid sequence encoding a cleavable linker, and a nucleic acid sequence comprising a polylinker insertion site. The polylinker insertion site can be oriented relative to the nucleic acid sequence encoding a cleavable linker to allow to cleavage of the cleavable linker to separate a macromolecule that is encoded by a nucleic acid inserted into the polylinker insertion site from the remainder of the encoded delivery construct. In certain embodiments, the cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject. In other embodiments, the cleavable linker can be cleavable by an enzyme that is present in the plasma of said subject.
  • In another aspect, the invention provides an expression vector comprising a polynucleotide of the invention.
  • In still another aspect, the invention provides a cell comprising an expression vector of the invention.
  • In yet another aspect, the invention provides a composition comprising a delivery construct of the invention. In certain embodiments, the composition is a pharmaceutical composition.
  • In still another aspect, the invention provides a method for delivering a macromolecule to a subject. The method comprises contacting an apical surface of a polarized epithelial cell of the subject with a delivery construct of the invention. The delivery construct can comprise a receptor binding domain, a transcytosis domain, a cleavable linker, and the macromolecule to be delivered. The transcytosis domain can transcytose the macromolecule to and through the basal-lateral membrane of the epithelial cell. In certain embodiments, the cleavable linker can be cleaved by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject. In other embodiments, the cleavable linker can be cleavable by an enzyme that is present in the plasma of the subject. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the delivery construct, and can deliver the macromolecule to the subject free from the remainder of the construct.
  • 4. BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-1D present micrographs showing adhesion to and transport across mouse tracheal epithelium of an exemplary delivery construct comprising green fluorescent protein (GFP) (Panels A-C), while GFP alone does not adhere to the epithelial cells (Panel D).
  • FIG. 2 presents a time course of serum nt-PE-GFP concentrations following intranasal administration of 100 μg nt-PE-GFP to anesthetized mice.
  • FIG. 3 presents the amino acid sequence of an exemplary PE.
  • FIG. 4 presents a western blot snowing transport and cleavage of an exemplary delivery construct comprising rat growth hormone (rGH) by human intestinal epithelial cell monolayers. The delivery construct was incubated in contact with the apical side of the epithelial cell monolayer for 4 hours. Media isolated from the basolateral side of the membrane post-incubation (lane 1) contained rGH of native apparent molecular weight, while media from the apical side of the membrane (lane 2) contained intact delivery construct. Lanes 3 and 4 show intact Delivery Construct 2 and recombinant rGH, respectively.
  • FIG. 5 presents serum rGH concentrations in BALB/c mice which were dosed by subcutaneous (SC) injection of 30 μg of non-glycosylated recombinant rat GH. Individual mice sera were tested at a dilution of 1:10, and the group average of rGH concentration were reported (n=4 mice per time point). Standard error of the mean (SEM) was indicated by the error bars.
  • FIG. 6 presents serum rGH concentrations in BALB/c mice which were dosed orally with 100 μg of Delivery Construct 2. Individual mice sera were tested at a dilution of 1:10, and the group average of rGH concentration were reported (n=4 mice per time point). Standard error of the mean (SEM) was indicated by the error bars.
  • FIG. 7 presents a graphical representation comparing pharmacokinetics of rGH delivered subcutaneously and with Delivery Construct 2.
  • FIG. 8 shows expression levels of IGF-1-BP3 mRNA in the liver of female BALB/c mice treated with 30 μg recombinant rGH by subcutaneous injection or with 100 μg of Delivery Construct 2 by oral gavage. Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-1-BP3, as described above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • FIG. 9 shows expression levels of growth hormone (GH) receptor mRNA in the liver of female BALB/c mice treated with rGH by subcutaneous injection (30 μg) or Delivery Construct 2 by oral gavage (100 μg). Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for GH receptor, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • FIG. 10 snows expression levels or insulin-like growth factor I (TGF-I); mRNA in the liver of female BALB/c mice treated with rGH by subcutaneous injection (30 μg) or Delivery Construct 2 by oral gavage (100 μg). Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-I, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control.
  • FIGS. 11A and B show serum anti-rGH IgG antibodies (diluted 1:25 in FIG. 11A and 1:200 in FIG. 11B) of mice orally administered 3,10, or 30 μg Delivery Construct 2 (F2) or subcutaneously administered 3 or 10 μg rGH in graphs with error bars, while FIGS. 11C and D presents the same data from individual animals.
  • FIG. 12 shows a nucleotide sequence that encodes Delivery Construct 6 (SEQ ID NO:34), an exemplary Delivery Construct for delivering human growth hormone (hGH).
  • FIGS. 13A and B show the amino acid sequence of Delivery Construct 6 (SEQ ID NO:35), an exemplary Delivery Construct for delivering hGH.
  • FIG. 14 shows a nucleotide sequence that encodes Delivery Construct 7 (SEQ ID NO:36), an exemplary Delivery Construct for delivering interferon-α (IFN-α).
  • FIGS. 15A and B show the amino acid sequence of Delivery Construct 7 (SEQ ID NO:37), an exemplary Delivery Construct for delivering IFN-α.
  • FIG. 16 shows 6 presents serum IFN-α concentrations in BALB/c mice which were dosed orally with 100 μg of Delivery Construct 8. Individual mice sera were tested at a dilution of 1:10, and the group average of IFN-α concentration were reported (n=3 mice per time point).
  • FIGS. 17A and B show the amino acid sequence of Delivery Construct 8 (SEQ ID NO:38), an exemplary Delivery Construct for delivering proinsulin.
  • FIGS. 18 A and B shows the amino acid sequence of the two amino acid chains of Delivery Construct 9 (SEQ ID NOs:39 and 40, respectively), an exemplary Delivery Construct for delivering insulin. Disulfide bonds are formed between the cysteine at position 7 of SEQ ID NO:40 (shown in FIG. 18B) and the cysteine at position 381 of SEQ ID NO:39 (shown in FIG. 18A) and between the cysteine at position 20 of SEQ ID NO:40 and the cysteine at position 393 of SEQ ID NO:39.
  • 5. DETAILED DESCRIPTION OF THE INVENTION 5.1. Definitions
  • Unless defined otherwise, all technical and scientific terms used herein have the meaning commonly understood by a person skilled in the art to which this invention belongs. As used herein, the following terms have the meanings ascribed to them unless specified otherwise.
  • A “ligand” is a compound that specifically binds to a target molecule. Exemplary ligands include, but are not limited to, an antibody, a cytokine, a substrate, a signaling molecule, and the like.
  • A “receptor” is compound that specifically binds to a ligand.
  • A ligand or a receptor (e.g., an antibody) “specifically binds to” or “is specifically immunoreactive with” another molecule when the ligand or receptor functions in a binding reaction that indicates the presence of the molecule in a sample of heterogeneous compounds. Thus, under designated assay (e.g., immunoassay) conditions, the ligand or receptor binds preferentially to a particular compound and does not bind in a significant amount to other compounds present in the sample. For example, a polynucleotide specifically binds under hybridization conditions to another polynucleotide comprising a complementary sequence and an antibody specifically binds under immunoassay conditions to an antigen bearing an epitope used to induce the antibody.
  • “Immunoassay” refers to a method of detecting an analyte in a sample involving contacting the sample with an antibody that specifically binds to the analyte and detecting binding between the antibody and the analyte. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity. In one example, an antibody that binds a particular antigen with an affinity (Km) of about 10 μM specifically binds the antigen.
  • “Linker” refers to a molecule that joins two other molecules, either covalently, or through ionic, van der Waals or hydrogen bonds, e.g., a nucleic acid molecule that hybridizes to one complementary sequence at the 5′ end and to another complementary sequence at the 3′ end, thus joining two non-complementary sequences. A “cleavable linker” refers to a linker that can be degraded or otherwise severed to separate the two components connected by the cleavable linker. Cleavable linkers are generally cleaved by enzymes, typically peptidases, proteases, nucleases, lipases, and the like. Cleavable linkers may also be cleaved by environmental cues, such as, for example, changes in temperature, pH, salt concentration, etc. when there is such a change in environment following transcytosis of the delivery construct across a polarized epithelial membrane.
  • “Pharmaceutical composition” refers to a composition suitable for pharmaceutical use in an animal. A pharmaceutical composition comprises a pharmacologically effective amount of an active agent and a pharmaceutically acceptable carrier. “Pharmacologically effective amount” refers to that amount of an agent effective to produce the intended pharmacological result. “Pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, vehicles, buffers, and excipients, such as a phosphate buffered saline solution, 5% aqueous solution of dextrose, and emulsions, such as an oil/water or water/oil emulsion, and various types of wetting agents and/or adjuvants. Suitable pharmaceutical carriers and formulations are described in Remington's Pharmaceutical Sciences, 21st Ed. 2005, Mack Publishing Co., Easton. A “pharmaceutically acceptable salt” is a salt that can be formulated into a compound for pharmaceutical use including, e.g., metal salts (sodium, potassium, magnesium, calcium, etc.) and salts of ammonia or organic amines.
  • Preferred pharmaceutical carriers depend upon the intended mode of administration of the active agent. Typical modes of administration include enteral (e.g., oral, intranasal, rectal, Or vaginal) or parenteral (e.g., subcutaneous, intramuscular, intravenous or intraperitoneal injection; or topical (e.g., transdermal, or transmucosal administration).
  • “Small organic molecule” refers to organic molecules of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes organic biopolymers (e.g., proteins, nucleic acids, etc.). Preferred small organic molecules range in size up to about 5000 Da, up to about 2000 Da, or up to about 1000 Da.
  • A “subject” of diagnosis, treatment, or administration is a human or non-human animal, including a mammal or a primate, and preferably a human.
  • “Treatment” refers to prophylactic treatment or therapeutic treatment. A “prophylactic” treatment is a treatment administered to a subject who does not exhibit signs of a disease or exhibits only early signs for the purpose of decreasing the risk of developing pathology. A “therapeutic” treatment is a treatment administered to a subject who exhibits signs of pathology for the purpose of diminishing or eliminating those signs.
  • Pseudomonas exotoxin A” or “PE” is secreted by Pseudomonas aeruginosa as a 67 kD protein composed of three prominent globular domains (Ia, II, and III) and one small subdomain (Ib) that connects domains II and III. See A. S. Allured et al., 1986, Proc. Natl. Acad. Set 83:1320-1324. Without intending to be bound to any particular theory or mechanism of action, domain Ia of PE is believed to mediate cell binding because domain Ia specifically binds to the low density lipoprotein receptor-related protein (“LRP”), also known as the α2-macroglobulin receptor (“α2-MR”) and CD-91. See M. Z. Kounnas et al., 1992, J. Biol. Chem. 267:12420-23. Domain Ia spans amino acids 1-252. Domain II of PE is believed to mediate transcytosis to the interior of a cell following binding of domain Ia to the α2-MR. Domain II spans amino acids 253-364. Certain portions of this domain may be required for secretion of PE from Pseudomonas aeruginosa after its synthesis. See, e.g., Vouloux et al., 2000, J. Bacterol. 182:4051-8. Domain Ib has no known function and spans amino acids 365-399. Domain III mediates cytotoxicity of PE and includes an endoplasmic reticulum retention sequence. PE cytotoxicity is believed to result from ADP ribosylation of elongation factor 2, which inactivates protein synthesis. Domain III spans amino acids 400-613 of PE. Deleting amino acid E553 (“ΔE553”) from domain III eliminates EF2 ADP ribosylation activity and detoxifies PE. PE having the mutation ΔE553 is referred to herein as “PEΔE553.” Genetically modified forms of PE are described in, e.g., U.S. Pat. Nos. 5,602,095; 5,512,658 and 5,458,878 Pseudomonas exotoxin, as used herein, also includes genetically modified, allelic, and chemically inactivated forms of PE within this definition. See, e.g., Vasil et al., 1986, Infect. Immunol. 52:538-48. Further, reference to the various domains of PE is made herein to the reference PE sequence presented as FIG. 3. However, one or more domain from modified PE, e.g., genetically or chemically modified PE, or a portion of such domains, can also be used in the chimeric immunogens of the invention so long as the domains retain functional activity. One of skill in the art can readily identify such domains of such modified PE based on, for example, homology to the PE sequence exemplified in FIG. 3 and test for functional activity using, for example, the assays described below.
  • “Polynucleotide” refers to a polymer composed of nucleotide units. Polynucleotides include naturally occurring nucleic acids, such as deoxyribonucleic acid (“DNA”) and ribonucleic acid (“RNA”) as well as nucleic acid analogs. Nucleic acid analogs include those which include non-naturally occurring bases, nucleotides that engage in linkages with other nucleotides other than the naturally occurring phosphodiester bond or which include bases attached through linkages other than phosphodiester bonds. Thus, nucleotide analogs include, for example and without limitation, phosphorothioates, phosphorodithioates, phosphorotriesters, phosphoramidates, boranophosphates, methylphosphonates, chiral-methyl phosphonates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs), and the like. Such polynucleotides can be synthesized, for example, using an automated DNA synthesizer. The term “nucleic acid” typically refers to large polynucleotides. The term “oligonucleotide” typically refers to short polynucleotides, generally no greater than about 50 nucleotides. It will be understood that when a nucleotide sequence is represented by a DNA sequence (i.e., A, T, G, C), this also includes an RNA sequence (i.e., A, U, G, C) in which “U” replaces “T.”
  • Conventional notation is used herein to describe polynucleotide sequences: the left-hand end of a single-stranded polynucleotide sequence is the 5′-end; the left-hand direction of a double-stranded polynucleotide sequence is referred to as the 5′-direction.
  • The direction of 5′ to 3′ addition of nucleotides to nascent RNA transcripts is referred to as the transcription direction. The DNA strand having the same sequence as an mRNA is referred to as the “coding strand”; sequences on the DNA strand having the same sequence as an mRNA transcribed from that DNA and which are located 5′ to the 5′-end of the RNA transcript are referred to as “upstream sequences”; sequences on the DNA strand having the same sequence as the RNA and which are 3′ to the 3′ end of the coding RNA transcript are referred to as “downstream sequences.”
  • “Complementary” refers to the topological compatibility or matching together of interacting surfaces of two polynucleotides. Thus, the two molecules can be described as complementary, and furthermore, the contact surface characteristics are complementary to each other. A first polynucleotide is complementary to a second polynucleotide if the nucleotide sequence of the first polynucleotide is substantially identical to the nucleotide sequence of the polynucleotide binding partner of the second polynucleotide, or if the first polynucleotide can hybridize to the second polynucleotide under stringent hybridization conditions. Thus, the polynucleotide whose sequence 5′-TATAC-3′ is complementary to a polynucleotide whose sequence is 5′-GTATA-3′.
  • The term “% sequence identity” is used interchangeably herein with the term “% identity” and refers to the level of amino acid sequence identity between two or more peptide sequences or the level of nucleotide sequence identity between two or more nucleotide sequences, when aligned using a sequence alignment program. For example, as used herein, 80% identity means the same thing as 80% sequence identity determined by a defined algorithm, and means that a given sequence is at least 80% identical to another length of another sequence. Exemplary levels of sequence identity include, but are not limited to, 60, 70, 80, 85, 90, 95, 98% or more sequence identity to a given sequence.
  • The term “% sequence homology” is used interchangeably herein with the term “% homology” and refers to the level of amino acid sequence homology between two or more peptide sequences or the level of nucleotide sequence homology between two or more nucleotide sequences, when aligned using a sequence alignment program. For example, as used herein, 80% homology means the same thing as 80% sequence homology determined by a defined algorithm, and accordingly a homologue of a given sequence has greater than 80% sequence homology over a length of the given sequence. Exemplary levels of sequence homology include, but are not limited to, 60, 70, 80, 85, 90, 95, 98% or more sequence homology to a given sequence.
  • Exemplary computer programs which can be used to determine identity between two sequences include, but are not limited to, the suite of BLAST programs, e.g., BLASTN, BLASTX, and TBLASTX, BLASTP and TBLASTN, publicly available on the Internet at the NCBI website. See also Altschul et al., 1990, J. Mol. Biol. 215:403-10 (with special reference to the published default setting, i.e., parameters w=4, t=17) and Altschul et al., 1997, Nucleic Acids Res., 25:3389-3402. Sequence searches are typically carried out using the BLASTP program when evaluating a given amino acid sequence relative to amino acid sequences in the GenBank Protein Sequences and other public databases. The BLASTX program is preferred for searching nucleic acid sequences that have been translated in all reading frames against amino acid sequences in the GenBank Protein Sequences and other public databases. Both BLASTP and BLASTX are run using default parameters of an open gap penalty of 11.0, and an extended gap penalty of 1.0, and utilize the BLOSUM-62 matrix. See id.
  • A preferred alignment of selected sequences in order to determine “% identity” between two or more sequences, is performed using for example, the CLUSTAL-W program in MacVector version 6.5, operated with default parameters, including an open gap penalty of 10.0, an extended gap penalty of 0.1, and a BLOSUM 30 similarity matrix.
  • “Polar Amino Acid” refers to a hydrophilic amino acid having a side chain that is uncharged at physiological pH, but which has at least one bond in which the pair of electrons shared in common by two atoms is held more closely by one of the atoms. Genetically encoded polar amino acids include Asn (N), Gln (Q) Ser (S) and Thr (T).
  • “Nonpolar Amino Acid” refers to a hydrophobic amino acid having a side chain that is uncharged at physiological pH and which has bonds in which the pair of electrons shared in common by two atoms is generally held equally by each of the two atoms (i.e., the side chain is not polar). Genetically encoded nonpolar amino acids include Ala (A), Gly (G), He (I), Leu (L), Met (M) and Val (V).
  • “Hydrophilic Amino Acid” refers to an amino acid exhibiting a hydrophobicity of less than zero according to the normalized consensus hydrophobicity scale of Eisenberg et al., 1984, J. Mol. Biol. 179:125-142. Genetically encoded hydrophilic amino acids include Arg (R), Asn (N), Asp (D), Glu (E), Gln (Q), His (H), Lys (K), Ser (S) and Thr (T).
  • “Hydrophobic Amino Acid” refers to an amino acid exhibiting a hydrophobicity of greater than zero according to the normalized consensus hydrophobicity scale of Eisenberg et al., 1984, J Mol. Biol. 179:125-142. Genetically encoded hydrophobic amino acids include Ala (A), Gly (G), He (I), Leu (L), Met (M), Phe (F), Pro (P), Trp (W), Tyr (Y) and Val (V).
  • “Acidic Amino Acid” refers to a hydrophilic amino acid having a side chain pK value of less than 7. Acidic amino acids typically have negatively charged side chains at physiological pH due to loss of a hydrogen ion. Genetically encoded acidic amino acids include Asp (D) and Glu (E).
  • “Basic Amino Acid” refers to a hydrophilic amino acid having a side chain pK value of greater than 7. Basic amino acids typically have positively charged side chains at physiological pH due to association with a hydrogen ion. Genetically encoded basic amino acids include Arg (R), His (H) and Lys (K).
  • “Encoding” refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom. Thus, a gene encodes a protein if transcription and translation of mRNA produced by that gene produces the protein in a cell or other biological system. Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and non-coding strand, used as the template for transcription, of a gene or cDNA can be referred to as encoding the protein or other product of that gene or cDNA. Unless otherwise specified, a “nucleotide sequence encoding an amino acid sequence” includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns.
  • “Amplification” refers to any means by which a polynucleotide sequence is copied and thus expanded into a larger number of polynucleotide molecules, e.g., by reverse transcription, polymerase chain reaction, ligase chain reaction, and the like.
  • “Primer” refers to a polynucleotide that is capable of specifically hybridizing to a designated polynucleotide template and providing a point of initiation for synthesis of a complementary polynucleotide. Such synthesis occurs when the polynucleotide primer is placed under conditions in which synthesis is induced, i.e., in the presence of nucleotides, a complementary polynucleotide template, and an agent for polymerization such as DNA polymerase. A primer is typically single-stranded, but may be double-stranded. Primers are typically deoxyribonucleic acids, but a wide variety of synthetic and naturally occurring primers are useful for many applications. A primer is complementary to the template to which it is designed to hybridize to serve as a site for the initiation of synthesis, but need not reflect the exact sequence of the template. In such a case, specific hybridization of the primer to the template depends on the stringency of the hybridization conditions. Primers can be labeled with, e.g., chromogenic, radioactive, or fluorescent moieties and used as detectable moieties.
  • “Probe,” when used in reference to a polynucleotide, refers to a polynucleotide that is capable of specifically hybridizing to a designated sequence of another polynucleotide. A probe specifically hybridizes to a target complementary polynucleotide, but need not reflect the exact complementary sequence of the template. In such a case, specific hybridization of the probe to the target depends on the stringency of the hybridization conditions. Probes can be labeled with, e.g., chromogenic, radioactive, or fluorescent moieties and used as detectable moieties. In instances where a probe provides a point of initiation for synthesis of a complementary polynucleotide, a probe can also be a primer.
  • “Hybridizing specifically to” or “specific hybridization” or “selectively hybridize to”, refers to the binding, duplexing, or hybridizing of a nucleic acid molecule preferentially to a particular nucleotide sequence under stringent conditions when that sequence is present in a complex mixture (e.g., total cellular) DNA or RNA.
  • The term “stringent conditions” refers to conditions under which a probe will hybridize preferentially to its target subsequence, and to a lesser extent to, or not at all to, other sequences. “Stringent hybridization” and “stringent hybridization wash conditions” in the context of nucleic acid hybridization experiments such as Southern and northern hybridizations are sequence dependent, and are different under different environmental parameters. An extensive guide to the hybridization of nucleic acids can be found in Tijssen, 1993, Laboratory Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Acid Probes, part I, chapter 2, “Overview of principles of hybridization and the strategy of nucleic acid probe assays”, Elsevier, N.Y.; Sambrook et al., 2001, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, 3rd ed., NY; and Ausubel et al., eds., Current Edition, Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.
  • Generally, highly stringent hybridization and wash conditions are selected to be about 5° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Very stringent conditions are selected to be equal to the Tm for a particular probe.
  • One example of stringent hybridization conditions for hybridization of complementary nucleic acids which have more than about 100 complementary residues on a filter in a Southern or northern blot is 50% formalin with 1 mg of heparin at 42° C., with the hybridization being carried out overnight. An example of highly stringent wash conditions is 0.15 M NaCl at 72° C. for about 15 minutes. An example of stringent wash conditions is a 0.2×SSC wash at 65° C. for 15 minutes. See Sambrook et al. for a description of SSC buffer. A high stringency wash can be preceded by a low stringency wash to remove background probe signal. An exemplary medium stringency wash for a duplex of, e.g., more than about 100 nucleotides, is 1×SSC at 45° C. for 15 minutes. An exemplary low stringency wash for a duplex of, e.g., more than about 100 nucleotides, is 4-6×SSC at 40° C. for 15 minutes. In general, a signal to noise ratio of 2× (or higher) than that observed for an unrelated probe in the particular hybridization assay indicates detection of a specific hybridization.
  • “Polypeptide” refers to a polymer composed of amino acid residues, related naturally occurring structural variants, and synthetic non-naturally occurring analogs thereof linked via peptide bonds, related naturally occurring structural variants, and synthetic non-naturally occurring analogs thereof. Synthetic polypeptides can be synthesized, for example, using an automated polypeptide synthesizer. Conventional notation is used herein to portray polypeptide sequences; the beginning of a polypeptide sequence is the amino-terminus, while the end of a polypeptide sequence is the carboxyl-terminus.
  • The term “protein” typically refers to large polypeptides, for example, polypeptides comprising more than about 50 amino acids. The term “protein” can also refer to dimers, trimers, and multimers that comprise more than one polypeptide.
  • “Conservative substitution” refers to the substitution in a polypeptide of an amino acid with a functionally similar amino acid. The following six groups each contain amino acids that are conservative substitutions for one another:
  • Alanine (A), Serine (S), and Threonine (T)
  • Aspartic acid (D) and Glutamic acid (E)
  • Asparagine (N) and Glutamine (Q)
  • Arginine (R) and Lysine (K)
  • Isoleucine (I), Leucine (L), Methionine (M), and Valine (V)
  • Phenylalanine (F), Tyrosine (Y), and Tryptophan (W).
  • The term “about,” as used herein, unless otherwise indicated, refers to a value that is no more than 10% above or below the value being modified by the term. For example, the term “about 5 μg/kg” means a range of from 4.5 μg/kg to 5.5 μg/kg. As another example, “about 1 hour” means a range of from 48 minutes to 72 minutes.
  • 5.2. Delivery Constructs
  • Generally, the delivery constructs of the present invention are polypeptides that have structural domains corresponding to domains Ia and II of PE. These structural domains perform certain functions, including, but not limited to, cell recognition and transcytosis, that correspond to the functions of the domains of PE.
  • In addition to the portions of the molecule that correspond to PE functional domains, the delivery constructs of this invention can further comprise a macromolecule for delivery to a biological compartment of a subject. The macromolecule can be introduced into any portion of the delivery construct that does not disrupt a cell-binding or transcytosis activity. The macromolecule is connected with the remainder of the delivery construct with a cleavable linker.
  • Accordingly, the delivery constructs of the invention generally comprise the following structural elements, each element imparting particular functions to the delivery construct: (1) a “receptor binding domain” that functions as a ligand for a cell surface receptor and that mediates binding of the construct to a cell; (2) a “transcytosis domain” that mediates transcytosis from a lumen bordering the apical surface of a mucous membrane to the basal-lateral side of a mucous membrane; (3) the macromolecule; and (4) a cleavable linker that connects the macromolecule to the remainder of the delivery construct.
  • The delivery constructs of the invention offer several advantages over conventional techniques for local or systemic delivery of macromolecules to a subject. Foremost among such advantages is the ability to deliver the macromolecule without using a needle to puncture the skin of the subject. Many subjects require repeated, regular doses of macromolecules. For example, diabetics must inject insulin several times per day to control blood sugar concentrations. Such subjects' quality of life would be greatly improved if the delivery of a macromolecule could be accomplished without injection, by avoiding pain or potential complications associated therewith.
  • Furthermore, many embodiments of the delivery constructs can be constructed and expressed in recombinant systems. Recombinant technology allows one to make a delivery construct having an insertion site designed for introduction of any suitable macromolecule. Such insertion sites allow the skilled artisan to quickly and easily produce delivery constructs for delivery of new macromolecules, should the need to do so arise.
  • In addition, connection of the macromolecule to the remainder of the delivery construct with a linker that is cleaved by an enzyme present at a basal-lateral membrane of an epithelial cell allows the macromolecule to be liberated from the delivery construct and released from the remainder of the delivery construct soon after transcytosis across the epithelial membrane. Such liberation reduces the probability of induction of an immune response against the macromolecule. It also allows the macromolecule to interact with its target free from the remainder of the delivery construct.
  • Other advantages of the delivery constructs of the invention will be apparent to those of skill in the art.
  • In certain embodiments, the invention provides a delivery construct that comprises a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker separates the macromolecule from the remainder of the construct. The cleavable linker is cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject or in the plasma of the subject. In certain embodiments, the enzyme that is at a basal-lateral membrane of a polarized epithelial cell exhibits higher activity on the basal-lateral side of a polarized epithelial cell than it does on the apical side of the polarized epithelial cell. In certain embodiments, the enzyme that is in the plasma of the subject exhibits higher activity in the plasma than it does on the apical side of a polarized epithelial cell.
  • In certain embodiments, the delivery construct further comprises a second cleavable linker. In certain embodiments, the first and/or the second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10). In certain embodiments, the first and/or the second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10) and is cleavable by an enzyme that exhibits higher activity on the basal-lateral side of a polarized epithelial cell than it does on the apical side of the polarized epithelial cell. In certain embodiments, the first and/or the second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10) and is cleavable by an enzyme that exhibits higher activity in the plasma than it does on the apical side of a polarized epithelial cell.
  • In certain embodiments, the enzyme that is present at a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
  • In certain embodiments, the receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, botulinum toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF α; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8. In certain embodiments, the receptor binding domain binds to a cell-surface receptor that is selected from the group consisting of α2-macroglobulin receptor, epidermal growth factor receptor, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNFα receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor. In further embodiments, the receptor binding domain of Pseudomonas exotoxin A is Domain Ia of Pseudomonas exotoxin A. In yet further embodiments, the receptor binding domain of Pseudomonas exotoxin A has an amino acid sequence that is SEQ ID NO.:1.
  • In certain embodiments, the transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, botulinum toxin, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E, coli enterotoxin, shiga toxin, and shiga-like toxin. In further embodiments, the transcytosis domain is Pseudomonas exotoxin A transcytosis domain. In still further embodiments, the Pseudomonas exotoxin A transcytosis domain has an amino acid sequence that is SEQ ID NO.:2.
  • In certain embodiments, the macromolecule is selected from the group of a nucleic acid, a peptide, a polypeptide, a protein, and a lipid, hi further embodiments, the polypeptide is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors. In yet further embodiments, the polypeptide is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN-α, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, and glucagon-like peptide 1. In still further embodiments, the polypeptide is human growth hormone. In other embodiments, the protein is human insulin.
  • In certain embodiments, the delivery constructs further comprise a second macromolecule that is selected from the group consisting of a nucleic acid, a peptide, a polypeptide, a protein, a lipid, and a small organic molecule and a second cleavable linker, wherein cleavage at said second cleavable linker separates said second macromolecule from the remainder of said construct. In certain embodiments, the first macromolecule is a first polypeptide and said second macromolecule is a second polypeptide. In certain embodiments, the first polypeptide and the second polypeptide associate to form a multimer. In certain embodiments, the multimer is a dimer, tetramer, or octamer. In further embodiments, the dimer is an antibody.
  • 5.2.1. Receptor Binding Domain
  • The delivery constructs of the invention generally comprise a receptor binding domain. The receptor binding domain can be any receptor binding domain known to one of skill in the art without limitation to bind to a cell surface receptor that is present on the apical membrane of an epithelial cell. Preferably, the receptor binding domain binds specifically to the cell surface receptor. The receptor binding domain should bind to the cell surface receptor with sufficient affinity to allow endocytosis of the delivery construct.
  • In certain embodiments, the receptor binding domain can comprise a peptide, a polypeptide, a protein, a lipid, a carbohydrate, or a small organic molecule, or a combination thereof. Examples of each of these molecules that bind to cell surface receptors present on the apical membrane of epithelial cells are well known to those of skill in the art. Suitable peptides or polypeptides include, but are not limited to, bacterial toxin receptor binding domains, such as the receptor binding domains from PE, cholera toxin, botulinum toxin, diptheria toxin, shiga toxin, shiga-like toxin, etc.; antibodies, including monoclonal, polyclonal, and single-chain antibodies, or derivatives thereof, growth factors, such as EGF, IGF-I, IGF-II, IGF-III etc.; cytokines, such as IL-1, IL-2, IL-3, IL-6, etc; chemokines, such as MIP-1a, MIP-1b, MCAF, IL-8, etc.; and other ligands, such as CD4, cell adhesion molecules from the immunoglobulin superfamily, integrins, ligands specific for the IgA receptor, etc. See, e.g., Pastane et al., 1992, Annu. Rev. Biochem. 61:331-54; and U.S. Pat. Nos. 5,668,255, 5,696,237, 5,863,745, 5,965,406, 6,022,950, 6,051,405, 6,251,392, 6,440,419, and 6,488,926. The skilled artisan can select the appropriate receptor binding domain based upon the expression pattern of the receptor to which the receptor binding domain binds.
  • Lipids suitable for receptor binding domains include, but are not limited to, lipids that themselves bind cell surface receptors, such as sphingosine-1-phosphate, lysophosphatidic acid, sphingosylphosphorylcholine, retinoic acid, etc.; lipoproteins such as apolipoprotein E, apolipoprotein A, etc., and glycolipids such as lipopolysaccharide, etc.; glycosphingolipids such as globotriaosylceramide and galabiosylceramide; and the like. Carbohydrates suitable for receptor binding domains include, but are not limited to, monosaccharides, disaccharides, and polysaccharides that comprise simple sugars such as glucose, fructose, galactose, etc.; and glycoproteins such as mucins, selectins, and the like. Suitable small organic molecules for receptor binding domains include, but are not limited to, vitamins, such as vitamin A, B1, B2, B3, B6, B9, B12, C, D, E, and K, amino acids, and other small molecules that are recognized and/or taken up by receptors present on the apical surface of epithelial cells. U.S. Pat. No. 5,807,832 provides an example of such small organic molecule receptor binding domains, vitamin B12.
  • In certain embodiments, the receptor binding domain can bind to a receptor found on an epithelial cell. In further embodiments, the receptor binding domain can bind to a receptor found on the apical membrane of an epithelial cell. The receptor binding domain can bind to any receptor known to be present on the apical membrane of an epithelial cell by one of skill in the art without limitation. For example, the receptor binding domain can bind to α2-MR, EGFR, or IGFR. An example of a receptor binding domain that can bind to α2-MR is domain Ia of PE. Accordingly, in certain embodiments, the receptor binding domain is domain Ia of PE. In other embodiments, the receptor binding domain is a portion of domain Ia of PE that can bind to α2-MR. Exemplary receptor binding domains that can bind to EGFR include, but are not limited to, EGF and TGFα. Examples of receptor binding domains that can bind to IGFR include, but are not limited to, IGF-I, IGF-II, or IGF-III. Thus, in certain embodiments, the receptor binding domain is EGF, IGF-I, IGF-II, or IGF-III. In other embodiments, the receptor binding domain is a portion of EGF, IGF-I, IGF-II, or IGF-III that can bind to the EGF or IGF receptor.
  • In certain embodiments, the receptor binding domain binds to a receptor that is highly expressed on the apical membrane of a polarized epithelial cell but is not expressed or expressed at low levels on antigen presenting cells, such as, for example, dendritic cells. Exemplary receptor binding domains that have this kind of expression pattern include, but are not limited to, TGFα, EGF, IGF-I, IGF-II, and IGF-III.
  • In certain embodiments, the delivery constructs of the invention comprise more than one domain that can function as a receptor binding domain. For example, the delivery construct can comprise PE domain Ia in addition to another receptor binding domain.
  • The receptor binding domain can be attached to the remainder of the delivery construct by any method or means known by one of skill in the art to be useful for attaching such molecules, without limitation. In certain embodiments, the receptor binding domain is expressed together with the remainder of the delivery construct as a fusion protein. Such embodiments are particularly useful when the receptor binding domain and the remainder of the construct are formed from peptides or polypeptides.
  • In other embodiments, the receptor binding domain is connected with the remainder of the delivery construct with a linker. In yet other embodiments, the receptor binding domain is connected with the remainder of the delivery construct without a linker. Either of these embodiments are useful when the receptor binding domain comprises a peptide, polypeptide, protein, lipid, carbohydrate, nucleic acid, or small organic molecule.
  • In certain embodiments, the linker can form a covalent bond between the receptor binding domain and the remainder of the delivery construct. In certain embodiments, the covalent bond can be a peptide bond. In other embodiments, the linker can link the receptor binding domain to the remainder of the delivery construct with one or more non-covalent interactions of sufficient affinity. One of skill in the art can readily recognize linkers that interact with each other with sufficient affinity to be useful in the delivery constructs of the invention. For example, biotin can be attached to the receptor binding domain, and streptavidin can be attached to the remainder of the molecule. In certain embodiments, the linker can directly link the receptor binding domain to the remainder of the molecule. In other embodiments, the linker itself comprises two or more molecules that associate in order to link the receptor binding domain to the remainder of the molecule. Exemplary linkers include, but are not limited to, straight or branched-chain carbon linkers, heterocyclic carbon linkers, substituted carbon linkers, unsaturated carbon linkers, aromatic carbon linkers, peptide linkers, etc.
  • In embodiments where a linker is used to connect the receptor binding domain to the remainder of the delivery construct, the linkers can be attached to the receptor binding domain and/or the remainder of the delivery construct by any means or method known by one of skill in the art without limitation. For example, the linker can be attached to the receptor binding domain and/or the remainder of the delivery construct with an ether, ester, thioether, thioester, amide, imide, disulfide, peptide, or other suitable moiety. The skilled artisan can select the appropriate linker and method for attaching the linker based on the physical and chemical properties of the chosen receptor binding domain and the linker. The linker can be attached to any suitable functional group on the receptor binding domain or the remainder of the molecule. For example, the linker can be attached to sulfhydryl (—S), carboxylic acid (COOH) or free amine (—NH2) groups, which are available for reaction with a suitable functional group on a linker. These groups can also be used to connect the receptor binding domain directly connected with the remainder of the molecule in the absence of a linker.
  • Further, the receptor binding domain and/or the remainder of the delivery construct can be derivatized in order to facilitate attachment of a linker to these moieties. For example, such derivatization can be accomplished by attaching suitable derivative such as those available from Pierce Chemical Company, Rockford, Ill. Alternatively, derivatization may involve chemical treatment of the receptor binding domain and/or the remainder of the molecule. For example, glycol cleavage of the sugar moiety of a carbohydrate or glycoprotein receptor binding domain with periodate generates free aldehyde groups. These free aldehyde groups may be reacted with free amine or hydrazine groups on the remainder of the molecule in order to connect these portions of the molecule. See, e.g., U.S. Pat. No. 4,671,958. Further, the skilled artisan can generate free sulfhydryl groups on proteins to provide a reactive moiety for making a disulfide, thioether, thioester, etc. linkage. See, e.g., U.S. Pat. No. 4,659,839.
  • Any of these methods for attaching a linker to a receptor binding domain and/or the remainder of a delivery construct can also be used to connect a receptor binding domain with the remainder of the delivery construct in the absence of a linker. In such embodiments, the receptor binding domain is coupled with the remainder of the construct using a method suitable for the particular receptor binding domain. Thus, any method suitable for connecting a protein, peptide, polypeptide, nucleic acid, carbohydrate, lipid, or small organic molecule to the remainder of the delivery construct known to one of skill in the art, without limitation, can be used to connect the receptor binding domain to the remainder of the construct. In addition to the methods for attaching a linker to a receptor binding domain or the remainder of a delivery construct, as described above, the receptor binding domain can be connected with the remainder of the construct as described, for example, in U.S. Pat. Nos. 6,673,905; 6,585,973; 6,596,475; 5,856,090; 5,663,312; 5,391,723; 6,171,614; 5,366,958; and 5,614,503.
  • In certain embodiments, the receptor binding domain can be a monoclonal antibody. In some of these embodiments, the chimeric immunogen is expressed as a fusion protein that comprises an immunoglobulin heavy chain from an immunoglobulin specific for a receptor on a cell to which the chimeric immunogen is intended to bind. The light chain of the immunoglobulin then can be co-expressed with the chimeric immunogen, thereby forming a light chain-heavy chain dimer. In other embodiments, the antibody can be expressed and assembled separately from the remainder of the chimeric immunogen and chemically linked thereto.
  • 5.2.2. Transcytosis Domain
  • The delivery constructs of the invention also comprise a transcytosis domain. The transcytosis domain can be any transcytosis domain known by one of skill in the art to effect transcytosis of chimeric proteins that have bound to a cell surface receptor present on the apical membrane of an epithelial cell. In certain embodiments, the transcytosis domain is a transcytosis domain from PE, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, or shiga-like toxin. See, for example, U.S. Pat. Nos. 5,965,406, and 6,022,950. In preferred embodiments, the transcytosis domain is domain II of PE.
  • The transcytosis domain need not, though it may, comprise the entire ammo acid sequence of domain II of native PE, which spans residues 253-364 of PE. For example, the transcytosis domain can comprise a portion of PE that spans residues 280-344 of domain II of PE. The amino acids at positions 339 and 343 appear to be necessary for transcytosis. See Siegall et al., 1991, Biochemistry 30:7154-59. Further, conservative or nonconservative substitutions can be made to the amino acid sequence of the transcytosis domain, as long as transcytosis activity is not substantially eliminated. A representative assay that can routinely be used by one of skill in the art to determine whether a transcytosis domain has transcytosis activity is described below.
  • Without intending to be limited to any particular theory or mechanism of action, the transcytosis domain is believed to permit the trafficking of the delivery construct through a polarized epithelial cell after the construct binds to a receptor present on the apical surface of the polarized epithelial cell. Such trafficking through a polarized epithelial cell is referred to herein as “transcytosis.” This trafficking permits the release of the delivery construct from the basal-lateral membrane of the polarized epithelial cell.
  • 5.2.3. Macromolecules for Delivery
  • The delivery constructs of the invention can also comprise a macromolecule. The macromolecule can be attached to the remainder of the delivery construct by any method known by one of skill in the art, without limitation. In certain embodiments, the macromolecule is expressed together with the remainder of the delivery construct as a fusion protein. In such embodiments, the macromolecule can be inserted into or attached to any portion of the delivery construct, so long as the receptor binding domain, the transcytosis domain, and macromolecule retain their activities. The macromolecule is connected with the remainder of the construct with a cleavable linker, or a combination of cleavable linkers, as described below.
  • In native PE, the Ib loop (domain Ib) spans amino acids 365 to 399, and is structurally characterized by a disulfide bond between two cysteines at positions 372 and 379. This portion of PE is not essential for any known activity of PE, including cell binding, transcytosis, ER retention or ADP ribosylation activity. Accordingly, domain Ib can be deleted entirely, or modified to contain a macromolecule.
  • Thus, in certain embodiments, the macromolecule can be inserted into domain Ib. If desirable, the macromolecule can be inserted into domain Ib wherein the cysteines at positions 372 and 379 are not cross-linked, this can be accomplished by reducing the disulfide linkage between the cysteines, by deleting the cysteines entirely from the Ib domain, by mutating the cysteines to other residues, such as, for example, serine, or by other similar techniques. Alternatively, the macromolecule can be inserted into the Ib loop between the cysteines at positions 372 and 379. In such embodiments, the disulfide linkage between the cysteines can be used to constrain the macromolecule if desirable. In any event, in embodiments where the macromolecule is inserted into domain Ib of PE, or into any other portion of the delivery construct, the macromolecule should be flanked by cleavable linkers such that cleavage at the cleavable linkers liberates the macromolecule from the remainder of the construct.
  • In other embodiments, the macromolecule can be connected with the N-terminal or C-terminal end of a polypeptide portion of the delivery construct. In such embodiments, the method of connection should be designed to avoid interference with other functions of the delivery construct, such as receptor binding or transcytosis. In yet other embodiments, the macromolecule can be connected with a side chain of an amino acid of the delivery construct. The macromolecule is connected with the remainder of the delivery construct with a cleavable linker, as described below. In such embodiments, the macromolecule to be delivered can be connected with the remainder of the delivery construct with one or more cleavable linkers such that cleavage at the cleavable linker(s) separates the macromolecule from the remainder of the delivery construct. It should be noted that, in certain embodiments, the macromolecule of interest can also comprise a short (1-20 amino acids, preferably 1-10 amino acids, and more preferably 1-5 amino acids) leader peptide in addition to the macromolecule of interest that remains attached to the macromolecule following cleavage of the cleavable linker. Preferably, this leader peptide does not affect the activity or immunogenicity of the macromolecule.
  • In embodiments where the macromolecule is expressed together with another portion of the delivery construct as a fusion protein, the macromolecule can be can be inserted into the delivery construct by any method known to one of skill in the art without limitation. For example, amino acids corresponding to the macromolecule can be inserted directly into the delivery construct, with or without deletion of native amino acid sequences. In certain embodiments, all or part of the Ib domain of PE can be deleted and replaced with the macromolecule. In certain embodiments, the cysteine residues of the Ib loop are deleted so that the macromolecule remains unconstrained. In other embodiments, the cysteine residues of the Ib loop are linked with a disulfide bond and constrain the macromolecule.
  • The macromolecule can be any macromolecule that is desired to be introduced into a subject. Thus, the macromolecule can be a peptide, a polypeptide, a protein, a nucleic acid, a carbohydrate, a lipid, a glycoprotein, synthetic organic and inorganic compounds, or any combination thereof. The macromolecule can also be a detectable compound such as a radiopaque compound, including air and barium and magnetic compounds. In certain embodiments, the macromolecule can be either soluble or insoluble in water. In certain embodiments, the macromolecule can be a macromolecule that can perform a desirable biological activity when introduced to the bloodstream of the subject. For example, the macromolecule can have receptor binding activity, enzymatic activity, messenger activity (i.e., act as a hormone, cytokine, neurotransmitter, or other signaling molecule), luminescent or other detectable activity, or regulatory activity, or any combination thereof. In, for example, diagnostic embodiments, the macromolecule can be conjugated to or can itself be a pharmaceutically acceptable gamma-emitting moiety, including but not limited to, indium and technetium, magnetic particles, radiopaque materials such as air or barium and fluorescent compounds.
  • In other embodiments, the macromolecule that is delivered can exert its effects in biological compartments of the subject other than the subject's blood. For example, in certain embodiments, the macromolecule can exert its effects in the lymphatic system. In other embodiments, the macromolecule can exert its effects in an organ or tissue, such as, for example, the subject's liver, heart, lungs, pancreas, kidney, brain, bone marrow, etc. In such embodiments, the macromolecule may or may not be present in the blood, lymph, or other biological fluid at detectable concentrations, yet may still accumulate at sufficient concentrations at its site of action to exert a biological effect.
  • Further, the macromolecule can be a protein that comprises more than one polypeptide subunit. For example, the protein can be a dimer, trimer, or higher order multimer. In certain embodiments, two or more subunits of the protein can be connected with a covalent bond, such as, for example, a disulfide bond. In other embodiments, the subunits of the protein can be held together with non-covalent interactions. One of skill in the art can routinely identify such proteins and determine whether the subunits are properly associated using, for example, an immunoassay. Exemplary proteins that comprise more than one polypeptide chain that can be delivered with a delivery construct of the invention include, but are not limited to, antibodies, insulin, IGF I, and the like.
  • Accordingly, in certain embodiments, the macromolecule is a peptide, polypeptide, or protein. In certain embodiments, the macromolecule comprises a peptide or polypeptide that comprises about 5, about 8, about 10, about 12, about 15, about 17, about 20, about 25, about 30, about 40, about 50, or about 60, about 70, about 80, about 90, about 100, about 200, about 400, about 600, about 800, or about 1000 amino acids. In certain embodiments, the macromolecule is a protein that comprises 1, 2, 3, 4, 5, 6, 7, 8, or more polypeptides. In certain embodiments, the peptide, polypeptide, or protein is a molecule that is commonly administered to subjects by injection. Exemplary peptides or polypeptides include, but are not limited to, IGF-I, IGF-II, IGF-III, EGF, IFN-α, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, clotting factors such as factor VII, vasopressin, calcitonin parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, glucagon-like peptide 1, asparaginase, and the like. In a preferred embodiment, the macromolecule is insulin. In certain preferred embodiments, the polypeptide is growth hormone. In even more preferred embodiments, the polypeptide is human growth hormone. In an equally preferred embodiment, the polypeptide is IFN-α, more preferably IFNα-2b. In an equally preferred embodiment, the polypeptide is insulin or proinsulin. In other embodiments, the polypeptide is green fluorescent protein. The sequences of all of these macromolecules are well known to those in the art, and attachment of these macromolecules to the delivery constructs is well within the skill of those in the art using standard techniques, as discussed below.
  • In certain embodiments, the macromolecule can be selected to not be cleavable by an enzyme present at the basal-lateral membrane of an epithelial cell. For example, the assays described in the examples can be used to routinely test whether such a cleaving enzyme can cleave the macromolecule to be delivered. If so, the macromolecule can be routinely altered to eliminate the offending amino acid sequence recognized by the cleaving enzyme. The altered macromolecule can then be tested to ensure that it retains activity using methods routine in the art.
  • Other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, antineoplastic compounds, such as nitrosoureas, e.g., carmustine, lomustine, semustine, strepzotocin; methylhydrazines, e.g., procarbazine, dacarbazine; steroid hormones, e.g., glucocorticoids, estrogens, progestins, androgens, tetrahydrodesoxycaricosterone; immunoactive compounds such as immunosuppressives, e.g., pyrimemamine, trimethopterin, pemcillamine, cyclosporine, azathioprine; and immunostimulants, e.g., levamisole, diethyl dithiocarbamate, enkephalins, endorphins; antimicrobial compounds such as antibiotics, e.g., β-lactam, penicillin, cephalosporins, carbapenims and monobactams, β-lactamase inhibitors, aminoglycosides, macrolides, tetracyclins, spectinomycin; antimalarials, amebicides; antiprotazoals; antifungals, e.g., amphotericin β, antivirals, e.g., acyclovir, idoxuridine, ribavirin, trifluridine, vidarbine, gancyclovir; parasiticides; antihalmintics; radiopharmaceutics; gastrointestinal drugs; hematologic compounds; immunoglobulins; blood clotting proteins, e.g., antihemophilic factor, factor IX complex; anticoagulants, e.g., dicumarol, heparinNa; fibrolysin inhibitors, e.g., tranexamic acid; cardiovascular drugs; peripheral anti-adrenergic drugs; centrally acting antihypertensive drugs, e.g., methyldopa, methyldopa HCl; antihypertensive direct vasodilators, e.g., diazoxide, hydralazine HCl; drugs affecting renin-angiotensin system; peripheral vasodilators, e.g., phentolamine; anti-anginal drugs; cardiac glycosides; inodilators, e.g., amrinone, milrinone, enoximone, fenoximone, imazodan, sulmazole; antidysrhythmics; calcium entry blockers; drugs affecting blood lipids, e.g., ranitidine, bosentan, rezulin; respiratory drugs; sypamomimetic drugs, e.g., albuterol, bitolterol mesylate, dobutamine HCl, dopamine HCl, ephedrine So, epinephrine, fenfluramine HCl, isoproterenol HCl, methoxamine HCl, norepinephrine bitartrate, phenylephrine HCl, ritodrine HCl; cholmomimetic drugs, e.g., acetylcholine Cl; anticholinesterases, e.g., edrophonium Cl; cholinesterase reactivators; adrenergic blocking drugs, e.g., acebutolol HCl, atenolol, esmolol HCl, labetalol HCl, metoprolol, nadolol, phentolamine mesylate, propanolol HCl; antimuscarinic drugs, e.g., anisotropine methylbromide, atropine SO4, clinidium Br, glycopyrrolate, ipratropium Br, scopolamine HBr; neuromuscular blocking drugs; depolarizing drugs, e.g., atracurium besylate, hexafluorenium Br, metocurine iodide, succinylcholine Cl, tubocurarine Cl, vecuronium Br; centrally acting muscle relaxants, e.g., baclofen; neurotransmitters and neurotransmitter agents, e.g., acetylcholine, adenosine, adenosine triphosphate; amino acid neurotransmitters, e.g., excitatory amino acids, GABA, glycine; biogenic amine neurotransmitters, e.g., dopamine, epinephrine, histamine, norepinephrine, octopamine, serotonin, tyramine; neuropeptides, nitric oxide, K+ channel toxins; antiparkinson drugs, e.g., amaltidine HCl, benztropine mesylate, carbidopa; diuretic drugs, e.g., dichlorphenamide, methazolamide, bendroflumethiazide, polythiazide; antimigraine drugs, e.g, carboprost tromethamine mesylate, methysergide maleate.
  • Still other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, hormones such as pituitary hormones, e.g., chorionic gonadotropin, cosyntropin, menotropins, somatotropin, iorticotropin, protirelin, thyrotropin, vasopressin, lypressin; adrenal hormones, e.g., beclomethasone dipropionate, betamethasone, dexarnethasone, triamcinolone; pancreatic hormones, e.g., glucagon, insulin; parathyroid hormone, e.g., dihydrochysterol; thyroid hormones, e.g., calcitonin etidronate disodium, levothyroxine Na, liothyronine Na, liotrix, thyroglobulin, teriparatide acetate; antithyroid drugs; estrogenic hormones; progestins and antagonists; hormonal contraceptives; testicular hormones; gastrointestinal hormones, e.g., cholecystokinin, enteroglycan, galanin, gastric inhibitory polypeptide, epidermal growth factor-urogastrone, gastric inhibitory polypeptide, gastrin-releasing peptide, gastrins, pentagastrin, tetragastrin, motilin, peptide YY, secretin, vasoactive intestinal peptide, sincalide.
  • Still other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, enzymes such as hyaluronidase, streptokinase, tissue plasminogen activator, urokinase, PGE-adenosine deaminase; intravenous anesthetics such as droperidol, etomidate, fetanyl citrate/droperidol, hexobarbital, ketamine HCl, methohexital Na, thiamylal Na, thiopental Na; antiepileptics, e.g., carbamazepine, clonazepam, divalproex Na, ethosuximide, mephenyloin, paramethadione, phenyloin, primidone.
  • Still other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, peptides and proteins such as ankyrins, arrestins, bacterial membrane proteins, clathrin, connexins, dystrophin, endothelin receptor, spectrin, selectin, cytokines; chemokines; growth factors, insulin, erythropoietin (EPO), tumor necrosis factor (TNF), neuropeptides, neuropeptide Y, neurotensin, transforming growth factor α, transforming growth factor β, interferon (IFN); hormones, growth inhibitors, e.g., genistein, steroids etc; glycoproteins, e.g., ABC transporters, platelet glycoproteins, GPIb-IX complex, GPIIb-IIIa complex, vitronectin, thrombomodulin, CD4, CD55, CD58, CD59, CD44, lymphocye function-associated antigen, intercellular adhesion molecule, vascular cell adhesion molecule, Thy-1, antiporters, CA-15-3 antigen, fibronectins, laminin, myelin-associated glycoprotein, GAP, GAP-43.
  • Yet other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, cytokines and cytokine receptors such as Interleukin-1 (IL-1), IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-1 receptor, IL-2 receptor, IL-3 receptor, IL-4 receptor, IL-5 receptor, IL-6 receptor, IL-7 receptor, IL-8 receptor, IL-9 receptor, IL-10 receptor, IL-11 receptor, IL-12 receptor, IL-13 receptor, IL-14 receptor, IL-15 receptor, IL-16 receptor, IL-17 receptor, IL-18 receptor, lymphokine inhibitory factor, macrophage colony stimulating factor, platelet derived growth factor, stem cell factor, tumor growth factor β, tumor necrosis factor, lymphotoxin, Fas, granulocyte colony stimulating factor, granulocyte macrophage colony stimulating factor, interferon α, interferon β, and interferon γ.
  • Still other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, growth factors and protein hormones such as erythropoietin, angiogenin, hepatocyte growth factor, fibroblast growth factor, keratinocyte growth factor, nerve growth factor, tumor growth factor α, thrombopoietin, thyroid stimulating factor, thyroid releasing hormone, neurotrophin, epidermal growth factor, VEGF, ciliary neurotrophic factor, LDL, somatomedin, insulin growth factor, insulin-like growth factor I and II; chemokines such as ENA-78, ELC, GRO-α, GRO-β, GRO-γ, HRG, LEF, IP-10, MCP-1, MCP-2, MCP-3, MCP-4, MIP-1α, MIP-1β, MG, MDC, NT-3, NT-4, SCF, LIF, leptin, RANTES, lymphotactin, eotaxin-1, eotaxin-2, TARC, TECK, WAP-1, WAP-2, GCP-1, GCP-2; α-chemokine receptors, e.g., CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, CXCR6, CXCR7; and β-chemokine receptors, e.g., CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7.
  • Yet other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, chemotherapeutics, such as chemotherapy or anti-tumor agents which are effective against various types of human cancers, including leukemia, lymphomas, carcinomas, sarcomas, myelomas etc., such as, for example, doxorubicin, mitomycin, cisplatin, daunorubicin, bleomycin, actinomycin D, and neocarzinostatin.
  • Still other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, antibodies such as anti-cluster of differentiation antigen CD-1 through CD-166 and the ligands or counter receptors for these molecules; anti-cytokine antibodies, e.g., anti-IL-1 through anti-IL-18 and the receptors for these molecules; anti-immune receptor antibodies; antibodies against T cell receptors, major histocompatibility complexes I and II, B cell receptors, selectin killer inhibitory receptors, killer activating receptors, OX-40, MadCAM-1, Gly-CAM2, integrins, cadherens, sialoadherens, Fas, CTLA-4, Fc γ-receptors, Fc α-receptors, Fc ε-receptors, Fc μ-receptors, and their ligands; anti-metalloproteinase antibodies, e.g., antibodies specific for collagenase, MMP-1 through MMP-8, TIMP-1, TIMP-2; anti-cell lysis/proinflammatory molecules, e.g., perforin, complement components, prostanoids, nitron oxide, thromboxanes; and anti-adhesion molecules, e.g., carcioembryonic antigens, lamins, fibronectins.
  • Yet other examples of macromolecules that can be delivered according to the present invention include, but are not limited to, antiviral agents such as reverse transcriptase inhibitors and nucleoside analogs, e.g., ddI, ddC, 3TC, ddA, AZT; protease inhibitors, e.g., Invirase, ABT-538; and inhibitors of in RNA processing, e.g., ribavirin.
  • Further, specific examples of macromolecules that can be delivered with the delivery constructs of the present invention Capoten, Monopril, Pravachol, Avapro, Plavix, Cefzil, Duricef/Ultracef, Azactam, Videx, Zerit, Maxipime, VePesid, Paraplatin, Platinol, Taxol, UFT, Buspar, Serzone, Stadol NS, Estrace, Glucophage (Bristol-Myers Squibb); Ceclor, Lorabid, Dynabac, Prozac, Darvon, Permax, Zyprexa, Humalog, Axid, Gemzar, Evista (Eli Lily); Vasotec/Vaseretic, Mevacor, Zocor, Prinivil/Prinizide, Plendil, Cozaar/Hyzaar, Pepcid, Prilosec, Primaxin, Noroxin, Recombivax HB, Varivax, Timoptic/XE, Trusopt, Proscar, Fosamax, Sinemet, Crixivan, Propecia, Vioxx, Singulair, Maxalt, Ivermectin (Merck & Co.); Diflucan, Unasyn, Sulperazon, Zithromax, Trovan, Procardia XL, Cardura, Norvasc, Dofetilide, Feldene, Zoloft, Zeldox, Glucotrol XL, Zyrtec, Eletriptan, Viagra, Droloxifene, Aricept, Lipitor (Pfizer); Vantin, Rescriptor, Vistide, Genotropin, Micronase/Glyn./Glyb., Fragmin, Total Medrol, Xanax/alprazolam, Sermion, Halcion/triazolam, Freedox, Dostinex, Edronax, Mirapex, Pharmorubicin, Adriamycin, Camptosar, Remisar, Depo-Provera, Caverject, Detrusitol, Estring, Healon, Xalatan, Rogaine (Pharmacia & Upjohn); Lopid, Accrupil, Dilantin, Cognex, Neurontin, Loestrin, Dilzem, Fempatch, Estrostep, Rezulin, Lipitor, Omnicef, FemHRT, Suramin, and Clinafloxacin (Warner Lambert).
  • Yet further examples of macromolecules which may be delivered by the delivery constructs of the present invention may be found in: Goodman and Gilman's The Pharmacological Basis of Therapeutics, 9th ed. McGraw-Hill 1996, incorporated herein by reference in its entirety.
  • In certain embodiments, the macromolecule can be inactive or in a less active form when administered, then be activated in the subject. For example, the macromolecule can be a peptide or polypeptide with a masked active site. The peptide or polypeptide can be activated by removing the masking moiety. Such removal can be accomplished by peptidases or proteases in the cases of peptide or polypeptide masking agents. Alternatively, the masking agent can be a chemical moiety that is removed by an enzyme present in the subject. This strategy can be used when it is desirable for the macromolecule to be active in limited circumstances. For example, it may be useful for a macromolecule to be active only in the liver of the subject. In such cases, the macromolecule can be selected to have a masking moiety that can be removed by an enzyme that is present in the liver, but not in other organs or tissues. Exemplary methods and compositions for making and using such masked macromolecules can be found in U.S. Pat. Nos. 6,080,575, 6,265,540, and 6,670,147.
  • In another example of such embodiments, the macromolecule can be a pro-macromolecule that is activated by a biological activity, for example by processing, present in the subject. For example, the exemplary macromolecule proinsulin can be delivered with a delivery construct of the present invention. Following delivery of the pro-macromolecule, it can be activated in the subject by appropriate processing enzymes. While it is believed that proinsulin is processed by enzymes (the endoproteases PC2 and PC3) present in highest concentration in secretory granules of pancreatic beta-cells, it is also believed that such enzyme are present in sufficient concentration in other compartments to permit activation of the pro-macromolecule into its fully active form. Further, it should be noted that many pro-macromolecules, including, for example, proinsulin, also exhibit activity similar to that of the fully active molecule. See, for example, Desbuquois et al., 2003, Endocrinology 12:5308-5321. Thus, even if not all of the pro-macromolecule is converted to the fully active form, the pro-molecule can in many cases still exert a desirable biological activity in the subject.
  • 5.2.4. Cleavable Linkers
  • In the delivery constructs of the invention, the macromolecule to be delivered to the subject is connected with the remainder of the delivery construct with one or more cleavable linkers. The number of cleavable linkers present in the construct depends, at least in part, on the location of the macromolecule in relation to the remainder of the delivery construct and the nature of the macromolecule. When the macromolecule is inserted into the delivery construct, the macromolecule can be flanked by cleavable linkers, such that cleavage at both linkers separates the macromolecule. The flanking cleavable linkers can be the same or different from each other. When the macromolecule can be separated from the remainder of the delivery construct with cleavage at a single linker, the delivery constructs can comprise a single cleavable linker. Further, where the macromolecule is, e.g., a dimer or other multimer, each subunit of the macromolecule can be separated from the remainder of the delivery construct and/or the other subunits of the macromolecule by cleavage at the cleavable linker.
  • The cleavable linkers are generally cleavable by a cleaving enzyme that is present at or near the basal-lateral membrane of an epithelial cell. By selecting the cleavable linker to be cleaved by such enzymes, the macromolecule can be liberated from the remainder of the construct following transcytosis across the mucous membrane and release from the epithelial cell into the cellular matrix on the basal-lateral side of the membrane. Further, cleaving enzymes could be used that are present inside the epithelial cell, such that the cleavable linker is cleaved prior to release of the delivery construct from the basal-lateral membrane, so long as the cleaving enzyme does not cleave the delivery construct before the delivery construct enters the trafficking pathway in the polarized epithelial cell that results in release of the delivery construct and macromolecule from the basal-lateral membrane of the cell.
  • In certain embodiments, the cleaving enzyme is a peptidase. In other embodiments, the cleaving enzyme is an RNAse. In yet other embodiments, the cleaving enzyme can cleave carbohydrates. Preferred peptidases include, but are not limited to, Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I. Table 1 presents these enzymes together with an amino acid sequence that is recognized and cleaved by the particular peptidase.
  • TABLE 1
    Peptidases Present Near Basal-Lateral
    Mucous Membranes
    Amino Acid Sequence
    Peptidase Recognized and Cleaved
    Cathepsin GI Ala-Ala-Pro-Phe
    (SEQ ID NO.: 4)
    Chymotrypsin I Gly-Gly-Phe
    (SEQ ID NO.: 5)
    Elastase I Ala-Ala-Pro-Val
    (SEQ ID NO.: 6)
    Subtilisin AI Gly-Gly-Leu
    (SEQ ID NO.: 7)
    Subtilisin AII Ala-Ala-Leu
    (SEQ ID NO.: 8)
    Thrombin I Phe-Val-Arg
    (SEQ ID NO.: 9)
    Urokinase I Val-Gly-Arg
    (SEQ ID NO.: 10)
  • In certain embodiments, the delivery construct can comprise more than one cleavable linker, wherein cleavage at either cleavable linker can separate the macromolecule to be delivered from the delivery construct. In certain embodiments, the cleavable linker can be selected based on the sequence, in the case of peptide, polypeptide, or protein macromolecules for delivery, to avoid the use of cleavable linkers that comprise sequences present in the macromolecule to be delivered. For example, if the macromolecule comprises AAL, the cleavable linker can be selected to be cleaved by an enzyme that does not recognize this sequence.
  • Further, the cleavable linker preferably exhibits a greater propensity for cleavage than the remainder of the delivery construct. As one skilled in the art is aware, many peptide and polypeptide sequences can be cleaved by peptidases and proteases. In certain embodiments, the cleavable linker is selected to be preferentially cleaved relative to other amino acid sequences present in the delivery construct during administration of the delivery construct. In certain embodiments, the receptor binding domain is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) intact following delivery of the delivery construct to the bloodstream of the subject. In certain embodiments, the translocation domain is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) intact following delivery of the delivery construct to the bloodstream of the subject. In certain embodiments, the macromolecule is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) intact following delivery of the delivery construct to the bloodstream of the subject. In certain embodiments, the cleavable linker is substantially (e.g., about 99%, about 95%, about 90%, about 85%, about 80, or about 75%) cleaved following delivery of the delivery construct to the bloodstream of the subject.
  • In other embodiments, the cleavable linker is cleaved by a cleaving enzyme found in the plasma of the subject. Any cleaving enzyme known by one of skill in the art to be present in the plasma of the subject can be used to cleave the cleavable linker. Use of such enzymes to cleave the cleavable linkers is less preferred than use of cleaving enzymes found near the basal-lateral membrane of a polarized epithelial cell because it is believed that more efficient cleavage will occur in near the basal-lateral membrane. However, if the skilled artisan determines that cleavage mediated by a plasma enzyme is sufficiently efficient to allow cleavage of a sufficient fraction of the delivery constructs to avoid adverse effects, such plasma cleaving enzymes can be used to cleave the delivery constructs. Accordingly, in certain embodiments, the cleavable linker can be cleaved with an enzyme that is selected from the group consisting of caspase-1, caspase-3, proprotein convertase 1, proprotein convertase 2, proprotein convertase 4, proprotein convertase 4 PACE 4, prolyl oligopeptidase, endothelin cleaving enzyme, dipeptidyl-peptidase IV, signal peptidase, neprilysin, renin, and esterase. See, e.g., U.S. Pat. No. 6,673,574. Table 2 presents these enzymes together with an amino acid sequence(s) recognized by the particular peptidase. The peptidase cleaves a peptide comprising these sequences at the N-terminal side of the amino acid identified with an asterisk.
  • TABLE 2
    Plasma Peptidases
    Amino Acid Sequence
    Peptidase Recognized and Cleaved
    Caspase-1 Tyr-Val-Ala-Asp-Xaa*
    (SEQ ID NO.: 11)
    Caspase-3 Asp-Xaa-Xaa-Asp-Xaa*
    (SEQ ID NO.: 12)
    Proprotein convertase Arg-(Xaa)n-Arg-Xaa*;
    1 n = 0, 2, 4 or 6
    (SEQ ID NO.: 13)
    Proprotein convertase Lys-(Xaa)n-Arg-Xaa*;
    2 n = 0, 2, 4, or 6
    (SEQ ID NO.: 14)
    Proprotein convertase Glp-Arg-Thr-Lys-Arg-Xaa*
    4 (SEQ ID NO.: 15)
    Proprotein convertase Arg-Val-Arg-Arg-Xaa*
    4 PACE 4 (SEQ ID NO.: 16)
    Decanoyl-Arg-Val-Arg-Arg-Xaa*
    (SEQ ID NO.: 17)
    Prolyloligopeptidase Pro-Xaa*-Trp-Val-Pro-Xaa
    Endothelin cleaving (SEQ ID NO.: 18)
    enzyme in combination
    with dipeptidyl-
    peptidase IV
    Signal peptidase Trp-Val*-Ala-Xaa
    (SEQ ID NO.: 19)
    Neprilysin in com- Xaa-Phe*-Xaa-Xaa
    bination with (SEQ ID NO.: 20)
    dipeptidyl-peptidase Xaa-Tyr*-Xaa-Xaa
    IV (SEQ ID NO.: 21)
    Xaa-Trp*-Xaa-Xaa
    (SEQ ID NO.: 22)
    Renin in combination Asp-Arg-Tyr-Ile-Pro-Phe-His-
    with dipeptidyl- Leu*-Leu-(Val, Ala or Pro)-
    peptidase IV Tyr-(Ser, Pro, or Ala)
    (SEQ ID NO.: 23)
  • Thus, in certain more preferred embodiments, the cleavable linker can be any cleavable linker known by one of skill in the art to be cleavable by an enzyme that is present at the basal-lateral membrane of an epithelial cell. In certain embodiments, the cleavable linker comprises a peptide. In other embodiments, the cleavable linker comprises a nucleic acid, such as RNA or DNA. In still other embodiments, the cleavable linker comprises a carbohydrate, such as a disaccharide or a trisaccharide. In certain embodiments, the cleavable linker is a peptide that comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10).
  • Alternatively, in less preferred embodiments, the cleavable linker can be any cleavable linker known by one of skill in the art to be cleavable by an enzyme that is present in the plasma of the subject to whom the delivery construct is administered. In certain embodiments, the cleavable linker comprises a peptide. In other embodiments, the cleavable linker comprises a nucleic acid, such as RNA or DNA. In still other embodiments, the cleavable linker comprises a carbohydrate, such as a disaccharide or a trisaccharide. In certain embodiments, the cleavable linker is a peptide that comprises an amino acid sequence that is selected from the group consisting of amino acid sequences presented in Table 2.
  • In certain embodiments, the delivery construct comprises more than one cleavable linker. In certain embodiments, cleavage at any of the cleavable linkers will separate the macromolecule to be delivered from the remainder of the delivery construct. In certain embodiments, the delivery construct comprises a cleavable linker cleavable by an enzyme present at the basal-lateral side of a polarized epithelial membrane and a cleavable linkers cleavable by an enzyme that is present in the plasma of the subject to whom the delivery construct is administered.
  • Further, Tables 4 and 5, below, present results of experiments testing the ability of peptidases to cleave substrates when applied to the basal-lateral or apical surface of a polarized epithelial membrane. The sequences recognized by these enzymes are well-known in the art. Thus, in certain embodiments, the delivery construct comprises a cleavable linker that is cleavable by an enzyme listed in Tables 4 and 5. Preferred peptidases exhibit higher activity on the basolateral side of the membrane. Particularly preferred peptidases exhibit much higher (e.g., 100%, 200%, or more increase in activity relative to the apical side) on the basolateral side. Thus, in certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 50% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 100% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 200% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 500% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 1,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 2,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 3,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 5,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane. In certain embodiments, the cleavable linker is cleavable by an enzyme that exhibits 10,000% higher activity on the basal-lateral side of the membrane than on the apical side of the membrane.
  • Still further, the results in Tables 4 and 5 indicate that certain enzymes are present in higher concentration or exhibit greater activity in certain epithelial lineages as compared to other epithelial lineages. Thus, the experiments described below can be used to test whether the particular epithelial cell lineage through which a macromolecule will be delivered exhibits the desired cleavage activity. In certain embodiments, the cleavage activity is present in tracheal epithelial cells, but not intestinal epithelial cells. In other embodiments, the cleavage activity is present in intestinal epithelial cells out not tracheal epithelial cells. In certain embodiments, the cleavage activity is present in intestinal epithelial cells and tracheal epithelial cells.
  • In certain embodiments, the cleavable linker may be cleavable by any enzyme that preferentially cleaves at the basolateral side of an epithelial membrane as compared to the apical side of the membrane. Example 6.4, below, describes an assay that can be used to assess the activity of such enzymes, while Table 7, appended to the end of this document, provides short names and accession numbers for every known human protease or peptidase. Any cleavage sequence recognized by such proteases or peptidases that preferentially cleaves a test substrate on the basolateral side of an epithelial membrane, or in the plasma, as compared to the apical side of such a membrane can also be used in the methods and compositions of the present invention. In such embodiments, one of skill in the art can readily determine the amino acid sequence recognized by such peptidases or proteases according to standard procedures known in the art or according to the known sequences, recognized by the proteases and peptidases.
  • The examples below provide methods for identifying cleaving enzymes that are present at or near the basal-lateral membrane of a polarized epithelial cell. The skilled artisan can routinely use such methods to identify additional cleaving enzymes and the chemical structure(s) identified and cleaved by such cleaving enzymes. Delivery constructs comprising such cleavable linkers and methods of delivering macromolecules using delivery constructs comprising such cleavable linkers are also within the scope of the present invention, whether or not such cleaving enzymes are presented in Table 7.
  • In other embodiments, the cleavable linker can be a cleavable linker that is cleaved following a change in the environment of the delivery construct. For example, the cleavable linker can be a cleavable linker that is pH sensitive and is cleaved by a change in pH that is experienced when the delivery construct is released from the basal-lateral membrane of a polarized epithelial cell. For instance, the intestinal lumen is strongly alkaline, while plasma is essentially neutral. Thus, a cleavable linker can be a moiety that is cleaved upon a shift from alkaline to neutral pH. The change in the environment of the delivery construct that cleaves the cleavable linker can be any environmental change that that is experienced when the delivery construct is released from the basal-lateral membrane of a polarized epithelial cell known by one of skill in the art, without limitation.
  • 5.3. Methods for Delivering a Macromolecule
  • In another aspect, the invention provides methods for local or systemic delivery of a macromolecule to a subject. These methods generally comprise administering a delivery construct of the invention to a mucous membrane of the subject to whom the macromolecule is delivered. The delivery construct is typically administered in the form of a pharmaceutical composition, as described below.
  • Thus, in certain aspects, the invention provides a method for delivering a macromolecule to a subject. The method comprises contacting an apical surface of a polarized epithelial cell of the subject with a delivery construct. In certain embodiments, the delivery construct comprises a receptor binding domain, a transcytosis domain, a cleavable linker, and the macromolecule to be delivered. The transcytosis domain can transcytose the macromolecule to and through the basal-lateral membrane of said epithelial cell. The cleavable linker can be cleaved by ah enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject or in the plasma of the subject. Cleavage at the cleavable linker separates the macromolecule from the remainder of the delivery construct, thereby delivering the macromolecule to the subject.
  • In certain embodiments, the enzyme that is present at or near a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I. In certain embodiments, the cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.:10).
  • In certain embodiments, the receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF α; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8. In certain embodiments, the receptor binding domain binds to a cell surface receptor selected from the group consisting of α2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNFα receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
  • In certain embodiments, the transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin.
  • In certain embodiments, the macromolecule is selected from the group consisting of a peptide, a polypeptide, a protein, a nucleic acid, and a lipid. In a preferred embodiment, the macromolecule is growth hormone. Even more preferably, the macromolecule is human growth hormone.
  • In certain embodiments, the invention provides a method for delivering a macromolecule to the bloodstream of a subject that results in at least about 30% bioavailability of the macromolecule, comprising administering a delivery construct comprising the macromolecule to the subject, thereby delivering at least about 30% of the total macromolecule administered to the blood of the subject in a bioavailable form of the macromolecule. In certain embodiments, at least about 10% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 15% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 20% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 25% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 35% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 40% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 45% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 50% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 55% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 60% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 65% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 70% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 75% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 80% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 85% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, at least about 90% of the total macromolecule administered is bioavailable to the subject, in certain embodiments, at least about 95% of the total macromolecule administered is bioavailable to the subject. In certain embodiments, the percentage of bioavailability of the macromolecule is determined by comparing the amount of macromolecule present in a subject's blood following administration of a delivery construct comprising the macromolecule to the amount of macromolecule present in a subject's blood following administration of the macromolecule through another route of administration. In certain embodiments, the other route of administration is injection, e.g., subcutaneous injection, intravenous injection, intra-arterial injection, etc. In other embodiments, the percentage of bioavailability of the macromolecule is determined by comparing the amount of macromolecule present in a subject's blood following administration of a delivery construct comprising the macromolecule to the total amount of macromolecule administered as part of the delivery construct.
  • In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 10 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 15 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 5 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 20 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 25 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 30 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 35 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 40 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 45 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 50 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 55 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 60 minutes after administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 90 minutes alter administration. In certain embodiments, peak plasma concentrations of the delivered macromolecule in the subject are achieved about 120 minutes after administration.
  • In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 10 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 1 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 0.1 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 0.01 ng/ml plasma and about 10 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 10 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 1 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 0.5 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 1 ng/ml plasma and about 0.1 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 10 ng/ml plasma and about 1 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is between about 10 ng/ml plasma and about 0.5 μg/ml plasma.
  • In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 10 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 5 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 1 μg/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 500 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 250 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 100 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 50 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 10 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 5 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 1 ng/ml plasma. In certain embodiments, the peak plasma concentration of the delivered macromolecule is at least about 0.1 ng/ml plasma.
  • Moreover, without intending to be bound to any particular theory or mechanism of action, it is believed that oral administration of a delivery construct can deliver a higher effective concentration of the delivered macromolecule to the liver of the subject than is observed in the subject's plasma. “Effective concentration,” in this context, refers to the concentration experienced by targets of the macromolecule and can be determined by monitoring and/or quantifying downstream effects of macromolecule-target interactions. While still not bound to any particular theory, it is believed that oral administration of the delivery construct results in absorption of the delivery construct through polarized epithelial cells of the digestive mucosa, e.g., the intestinal mucosa, followed by cleavage of the construct and release of the macromolecule at the basolateral side of the mucous membrane. As one of skill in the art will recognize, the blood at the basolateral membrane of such digestive mucosa is carried from this location to the liver via the portal venous system. Thus, when the macromolecule exerts a biological activity in the liver, such as, for example, activities mediated by growth hormone, insulin, IGF-I, etc. binding to their cognate receptors, the macromolecule is believed to exert an effect in excess of what would be expected based on the plasma concentrations observed in the subject. Accordingly, in certain embodiments, the invention provides a method of administering a macromolecule to a subject that comprises orally administering a delivery construct comprising the macromolecule to the subject, wherein the macromolecule is delivered to the subject's liver at a higher effective concentration than observed in the subject's plasma.
  • In certain embodiments, the epithelial cell is selected from the group consisting of nasal epithelial cells, oral epithelial cells, intestinal epithelial cells, rectal epithelial cells, vaginal epithelial cells, and pulmonary epithelial cells.
  • In certain embodiments, the subject is a mammal. In further embodiments, the subject is a rodent, a lagomorph, or a primate. In yet further embodiments, the rodent is a mouse or rat. In other embodiments, the lagomorph is a rabbit. In still other embodiments, the primate is a human, monkey, or ape. In a preferred embodiment, the subject is a human.
  • In another aspect, the invention provides a method for delivering a macromolecule to the bloodstream of a subject that induces a lower titer of antibodies against the macromolecule than other routes of administration. Without intending to be bound by any particular theory or mechanism of action, it is believed that entry of the macromolecule through a mucous membrane, e.g., through the intestinal mucosa, causes the immune system to tolerate the macromolecule better than if the macromolecule were, for example, injected. Thus, a lower titer of antibodies against the macromolecule can be produced in the subject by delivering the macromolecule with a delivery construct of the invention through the mucosa rather than injecting the macromolecule, for example, subcutaneously, intravenously, intra-arterially, intraperitoneally, or otherwise. Generally, the time at which the lower titer of antibodies detected for the alternate routes of administration is detected should be roughly comparable; for example, the titer of antibodies can be determined at about 1 week, at about 2 weeks, at about 3 weeks, at about 4 weeks, at about 2 months, or at about 6 months following administration of the macromolecule with the delivery construct or by injection.
  • Accordingly, in certain embodiments, the invention provides a method for delivering a macromolecule to the bloodstream a subject that comprises contacting a delivery construct of the invention that comprises the macromolecule to be delivered to an apical surface of a polarized epithelial cell of the subject, such that the macromolecule is administered to the bloodstream of the subject, wherein a lower titer of antibodies specific for the macromolecule is induced in the serum of the subject than is induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct to a subject.
  • In certain embodiments, the macromolecule is selected from the group consisting of a peptide, a polypeptide, a protein, a nucleic acid, and a lipid. In certain embodiments, the macromolecule is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors. In certain embodiments, the macromolecule is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN-α, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, and glucagon-like peptide 1. In certain embodiments, the macromolecule is human growth hormone. In certain embodiments, the macromolecule is human insulin. In certain embodiments, the subject is a mouse, rat, dog, goat, or human.
  • In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 95% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 90% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 85% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 80% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 75% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 70% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 65% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 60% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 55% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 55% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 50% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 45% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 40% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 35% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 30% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 25% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than 20% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 15% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 10% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 5% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct. In certain embodiments, the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 1% of the titer of antibodies induced by subcutaneously administering the macromolecule separately from the remainder of the delivery construct.
  • 5.3.1. Methods of Administration
  • The delivery constructs of the invention can be administered to a subject by any method known to one of skill in the art. In certain embodiments, the delivery constructs are contacted to a mucosal membrane of the subject. For example, the mucosal membrane can be present in the eye, nose, mouth, trachea, lungs, esophagus, stomach, small intestine, large intestine, rectum, anus, sweat glands, vulva, vagina, or penis of the subject. Preferably, the mucosal membrane is a mucosal membrane present in the digestive tract of the subject, such as a mucosal membrane in the mouth, esophagus, stomach, small intestine, large intestine, or rectum of the subject.
  • In such embodiments, the delivery constructs are preferably administered to the subject orally. Thus, the delivery construct can be formulated to protect the delivery construct from degradation in the acid environment of the stomach, if necessary. For example, many embodiments of the delivery constructs of the invention comprise polypeptide domains with defined activities. Unless such delivery constructs are protected from acid and/or enzymatic hydrolysis in the stomach, the constructs will generally be digested before delivery of substantial amounts of the macromolecule to be delivered. Accordingly, composition formulations that protect the delivery construct from degradation can be used in administration of these delivery constructs.
  • 5.3.2. Dosage
  • Generally, a pharmaceutically effective amount of the delivery construct of the invention is administered to a subject. The skilled artisan can readily determine if the dosage of the delivery construct is sufficient to deliver an effective amount of the macromolecule, as described below. In certain embodiments, between about 1 μg and about 1 g of delivery construct is administered. In other embodiments, between about 10 μg and about 500 mg of delivery construct is administered. In still other embodiments, between about 10 μg and about 100 mg of delivery construct is administered. In yet other embodiments, between about 10 μg and about 1000 μg of delivery construct is administered. In still other embodiments, between about 10 μg and about 250 μg of delivery construct is administered. In yet other embodiments, between about 10 μg and about 100 μg of delivery construct is administered. Preferably, between about 10 μg and about 50 μg of delivery construct is administered.
  • The volume of a composition comprising the delivery construct that is administered will generally depend on the concentration of delivery construct and the formulation of the composition. In certain embodiments, a unit dose of the delivery construct composition is between about 0.05 ml and about 1 ml, preferably about 0.5 ml. The delivery construct compositions can be prepared in dosage forms containing between 1 and 50 doses (e.g., 0.5 ml to 25 ml), more usually between 1 and 10 doses (e.g., 0.5 ml to 5 ml)
  • The delivery construct compositions of the invention can be administered in one dose or in multiple doses. A dose can be followed by one or more doses spaced by about 1 to about 6 hours, by about 6 to about 12 hours, by about 12 to about 24 hours, by about 1 day to about 3 days, by about 1 day to about 1 week, by about 1 week to about 2 weeks, by about 2 weeks to about 1 month, by about 4 to about 8 weeks, by about 1 to about 3 months, or by about 1 to about 6 months.
  • The macromolecules to be delivered are generally macromolecules for which a large amount of knowledge regarding dosage, frequency of administration, and methods for assessing effective concentrations in subjects has accumulated. Such knowledge can be used to assess efficiency of delivery, effective concentration of the macromolecule in the subject, and frequency of administration. Thus, the knowledge of those skilled in the art can be used to determine whether, for example, the amount of macromolecule delivered to the subject is an effective amount, the dosage should be increased or decreased, the subject should be administered the delivery construct more or less frequently, and the like.
  • 5.3.3. Determining Amounts of Macromolecule Delivered
  • The methods of the invention can be used to deliver, either locally or systemically, a pharmaceutically effective amount of a macromolecule to a subject. The skilled artisan can determine whether the methods result in delivery of such a pharmaceutically effective amount of the macromolecule. The exact methods will depend on the macromolecule that is delivered, but generally will rely on either determining the concentration of the macromolecule in the blood of the subject or in the biological compartment of the subject where the macromolecule exerts its effects. Alternatively or additionally, the effects of the macromolecule on the subject can be monitored.
  • For example, in certain embodiments of the present invention, the macromolecule that is delivered is insulin, e.g., human insulin. In such embodiments, the skilled artisan can determine whether a pharmaceutically effective amount of human insulin had been delivered to the subject by, for example, taking a plasma sample from the subject and determining the concentration of human insulin therein. One exemplary method for determining the concentration of human insulin is by performing an ELISA assay, but any other suitable assay known to the skilled artisan can be used.
  • Alternatively, one of skill in the art can determine if an effective amount of human insulin had been delivered to the subject by monitoring the blood sugar concentrations of the subject. As is well-known in the art, human insulin, among other activities, acts on hepatocytes to promote glycogen formation, thereby reducing plasma glucose concentrations. Accordingly, the subject's plasma glucose concentration can be monitored to determine whether an effective amount of insulin had been delivered.
  • Any effect of a macromolecule that is administered that is known by one of skill in the art, without limitation, can be assessed in determining whether an effective amount of the macromolecule has been administered. Exemplary effects include, but are not limited to, receptor binding, receptor activation, downstream effects of receptor binding, downstream effects of receptor activation, coordination of compounds, effective blood clotting, bone growth, wound healing, cellular proliferation, etc. The exact effect that is assessed will depend on the macromolecule that is delivered.
  • 5.4. Polynucleotides Encoding Delivery Constructs
  • In another aspect, the invention provides polynucleotides comprising a nucleotide sequence encoding the delivery constructs. These polynucleotides are useful, for example, for making the delivery constructs. In yet another aspect, the invention provides an expression system that comprises a recombinant polynucleotide sequence encoding a receptor binding domain, a transcytosis domain, and a polylinker insertion site for a polynucleotide sequence encoding a macromolecule. The polylinker insertion site can be anywhere in the polynucleotide sequence so long as the polylinker insertion does not disrupt the receptor binding domain or the transcytosis domain. The polylinker insertion site should be oriented near a polynucleotide sequence that encodes a cleavable linker so that cleavage at the cleavable linker separates a macromolecule encoded by a nucleic acid inserted into the polylinker insertion site from the remainder of the encoded delivery construct. Thus, in embodiments where the polylinker insertion site is at an end of the encoded construct, the polynucleotide comprises one nucleotide sequence encoding a cleavable linker between the polylinker insertion site and the remainder of the polynucleotide. In embodiments where the polylinker insertion site is not at the end of the encoded construct, the polylinker insertion site can be flanked by nucleotide sequences that each encode a cleavable linker.
  • In certain embodiments, the recombinant polynucleotides are based on polynucleotides encoding PE, or portions or derivatives thereof. In other embodiments, the recombinant polynucleotides are based on polynucleotides that hybridize to a polynucleotide that encodes PE under stringent hybridization conditions. A nucleotide sequence encoding PE is presented as SEQ ID NO.:3. This sequence can be used to prepare PCR primers for isolating a nucleic acid that encodes any portion of this sequence that is desired. For example, PCR can be used to isolate a nucleic acid that encodes one or more of the functional domains of PE. A nucleic acid so isolated can then be joined to nucleic acids encoding other functional domains of the delivery constructs using standard recombinant techniques.
  • Other in vitro methods that can be used to prepare a polynucleotide encoding PE, PE domains, or any other functional domain useful in the delivery constructs of the invention include, but are not limited to, reverse transcription, the polymerase chain reaction (PCR), the ligase chain reaction (LCR), the transcription-based amplification system (TAS), the self-sustained sequence replication system (3SR) and the QP replicase amplification system (QB). Any such technique known by one of skill in the art to be useful in construction of recombinant nucleic acids can be used. For example, a polynucleotide encoding the protein or a portion thereof can be isolated by polymerase chain reaction of cDNA using primers based on the DNA sequence of PE or a nucleotide encoding a receptor binding domain.
  • Guidance for using these cloning and in vitro amplification methodologies are described in, for example, U.S. Pat. No. 4,683,195; Mullis et al., 1987, Cold Spring Harbor Symp. Quant. Biol. 51:263; and Erlich, ed., 1989, PCR Technology, Stockton Press, NY. Polynucleotides encoding a delivery construct or a portion thereof also can be isolated by screening genomic or cDNA libraries with probes selected from the sequences of the desired polynucleotide under stringent, moderately stringent, or highly stringent hybridization conditions.
  • Construction of nucleic acids encoding the delivery constructs of the invention can be facilitated by introducing an insertion site for a nucleic acid encoding the macromolecule into the construct. In certain embodiments, an insertion site for the macromolecule can be introduced between the nucleotides encoding the cysteine residues of domain Ib. In other embodiments, the insertion site can be introduced anywhere in the nucleic acid encoding the construct so long as the insertion does not disrupt the functional domains encoded thereby. In certain embodiments, the insertion site can be in the ER retention domain.
  • In more specific embodiments, a nucleotide sequence encoding a portion of the Ib domain between the cysteine-encoding residues can be removed and replaced with a nucleotide sequence that includes a cloning site cleaved by a restriction enzyme. For example, the cloning site can be recognized and cleaved by PstI. In such examples, a polynucleotide encoding macromolecule that is flanked by PstI sequences can be inserted into the vector.
  • Further, the polynucleotides can also encode a secretory sequence at the amino terminus of the encoded delivery construct. Such constructs are useful for producing the delivery constructs in mammalian cells as they simplify isolation of the immunogen.
  • Furthermore, the polynucleotides of the invention also encompass derivative versions of polynucleotides encoding a delivery construct. Such derivatives can be made by any method known by one of skill in the art without limitation. For example, derivatives can be made by site-specific mutagenesis, including substitution, insertion, or deletion of one, two, three, five, ten or more nucleotides, of polynucleotides encoding the delivery construct. Alternatively, derivatives can be made by random mutagenesis. One method for randomly mutagenizing a nucleic acid comprises amplifying the nucleic acid in a PCR reaction in the presence of 0.1 mM MnCl2 and unbalanced nucleotide concentrations. These conditions increase the misincorporation rate of the polymerase used in the PCR reaction and result in random mutagenesis of the amplified nucleic acid.
  • Several site-specific mutations and deletions in chimeric molecules derived from PE have been made and characterized. For example, deletion of nucleotides encoding amino acids 1-252 of PE yields a construct referred to as “PE40.” Deleting nucleotides encoding amino acids 1-279 of PE yields a construct referred to as “PE37.” See U.S. Pat. No. 5,602,095. In both of these constructs, the receptor binding domain of PE, i.e., domain Ia, has been deleted. Nucleic acids encoding a receptor binding domain can be ligated to these constructs to produce delivery constructs that are targeted to the cell surface receptor recognized by the receptor binding domain. Of course, these recombinant polynucleotides are particularly useful for expressing delivery constructs that have a receptor binding domain that is not domain Ia of PE. The recombinant polynucleotides can optionally encode an amino-terminal methionine to assist in expression of the construct. In certain embodiments, the receptor binding domain can be ligated to the 5′ end of the polynucleotide encoding the transcytosis domain.
  • Other nucleic acids encoding mutant forms of PE that can be used as a source of nucleic acids for constructing the delivery constructs of the invention include, but are not limited to, PEΔ553 and those described in U.S. Pat. Nos. 5,602,095; 5,512,658 and 5,458,878, and in Vasil et al., 1986, Infect. Immunol 52:538-48.
  • Accordingly, in certain embodiments, the invention provides a polynucleotide that encodes a delivery construct. The delivery construct comprises a receptor binding domain, a transcytosis domain, a macromolecule to be delivered to a subject, and a cleavable linker. Cleavage at the cleavable linker can separate the macromolecule from the remainder of the construct. The cleavable linker can be cleaved by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of the subject or in the plasma of the subject.
  • In certain embodiments, the polynucleotide hybridizes under stringent hybridization conditions to any polynucleotide of this invention. In further embodiments, the polynucleotide hybridizes under stringent conditions to a nucleic acid that encodes any delivery construct of the invention.
  • In certain embodiments, the polynucleotide encodes a delivery construct that further comprises a second cleavable linker. In certain embodiments, the first and/or second cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10). In certain embodiments, the first and/or second cleavable linker encoded by the polynucleotide is cleavable by an enzyme that is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
  • In certain embodiments, the receptor binding domain encoded by the polynucleotide is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF α; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8. In certain embodiments, the receptor binding domain encoded by the polynucleotide binds to a cell-surface receptor that is selected from the group consisting of α2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNFα receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor. In further embodiments, the receptor binding domain encoded by the polynucleotide is Domain Ia of Pseudomonas exotoxin A. In yet further embodiments, the receptor binding domain encoded by the polynucleotide has an amino acid sequence that is SEQ ID NO.: 1.
  • In certain embodiments, the transcytosis domain encoded by the polynucleotide is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin. In further embodiments, the transcytosis domain is Pseudomonas exotoxin A transcytosis domain. In still further embodiments, the Pseudomonas exotoxin A transcytosis domain has an amino acid sequence that is SEQ ID NO.:2.
  • In certain embodiments, the macromolecule encoded by the polynucleotide is selected from the group of a peptide, a polypeptide, and a protein. In certain embodiments, the polypeptide is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors. In further embodiments, the polypeptide is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN-α, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, adrenocorticototropin, enkephalin, and glucagon-like peptide 1. In still further embodiments, the polypeptide is human growth hormone. In other embodiments, the protein is human insulin.
  • In other embodiments, the invention provides a polynucleotide that encodes a delivery construct that comprises a nucleic acid sequence encoding a receptor binding domain, a nucleic acid sequence encoding a transcytosis domain, a nucleic acid sequence encoding a cleavable linker, and a nucleic acid sequence comprising a polylinker insertion site. The polylinker insertion site can be oriented relative to the nucleic acid sequence encoding a cleavable linker to allow to cleavage of the cleavable linker to separate a macromolecule that is encoded by a nucleic acid inserted into the polylinker insertion site from the remainder of said delivery construct. The cleavable linker can be cleavable by an enzyme that is present at a basal-lateral membrane of a polarized epithelial cell of said subject or in the plasma of said subject.
  • 5.5. Expression Vectors
  • In still another aspect, the invention provides expression vectors for expressing the delivery constructs. Generally, expression vectors are recombinant polynucleotide molecules comprising expression control sequences operatively linked to a nucleotide sequence encoding a polypeptide. Expression vectors can readily be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, selectable markers, etc. to result in stable transcription and translation or mRNA. Techniques for construction of expression vectors and expression of genes in cells comprising the expression vectors are well known in the art. See, e.g., Sambrook et al., 2001, Molecular Cloning—A Laboratory Manual, 3rd edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., and Ausubel et al., eds., Current Edition, Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.
  • Useful promoters for use in expression vectors include, but are not limited to, a metallothionein promoter, a constitutive adenovirus major late promoter, a dexamethasone-inducible MMTV promoter, a SV40 promoter, a MRP pol III promoter, a constitutive MPSV promoter, a tetracycline-inducible CMV promoter (such as the human immediate-early CMV promoter), and a constitutive CMV promoter.
  • The expression vectors should contain expression and replication signals compatible with the cell in which the delivery constructs are expressed. Expression vectors useful for expressing delivery constructs include viral vectors such as retroviruses, adenoviruses and adenoassociated viruses, plasmid vectors, cosmids, and the like. Viral and plasmid vectors are preferred for transfecting the expression vectors into mammalian cells. For example, the expression vector pcDNA1 (Invitrogen, San Diego, Calif.), in which the expression control sequence comprises the CMV promoter, provides good rates of transfection and expression into such cells.
  • The expression vectors can be introduced into the cell for expression of the delivery constructs by any method known to one of skill in the art without limitation. Such methods include, but are not limited to, e.g., direct uptake of the molecule by a cell from solution; facilitated uptake through lipofection using, e.g., liposomes or immunoliposomes; particle-mediated transfection; etc. See, e.g., U.S. Pat. No. 5,272,065; Goeddel et al., eds, 1990, Methods in Enzymology, vol. 185, Academic Press, Inc., CA; Krieger, 1990, Gene Transfer and Expression—A Laboratory Manual, Stockton Press, NY; Sambrook et al., 1989, Molecular Cloning—A Laboratory Manual, Cold Spring Harbor Laboratory, NY; and Ausubel et al., eds., Current Edition, Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, NY.
  • The expression vectors can also contain a purification moiety that simplifies isolation of the delivery construct. For example, a polyhistidine moiety of, e.g., six histidine residues, can be incorporated at the amino terminal end of the protein. The polyhistidine moiety allows convenient isolation of the protein in a single step by nickel-chelate chromatography. In certain embodiments, the purification moiety can be cleaved from the remainder of the delivery construct following purification. In other embodiments, the moiety does not interfere with the function of the functional domains of the delivery construct and thus need not be cleaved.
  • 5.6. Cell for Expressing a Delivery Construct
  • In yet another aspect, the invention provides a cell comprising an expression vector for expression of the delivery constructs, or portions thereof. The cell is preferably selected for its ability to express high concentrations of the delivery construct to facilitate purification of the protein. In certain embodiments, the cell is a prokaryotic cell, for example, E. coli. As described in the examples, the delivery constructs are properly folded and comprise the appropriate disulfide linkages when expressed in E. coli.
  • In other embodiments, the cell is a eukaryotic cell. Useful eukaryotic cells include yeast and mammalian cells. Any mammalian cell known by one of skill in the art to be useful for expressing a recombinant polypeptide, without limitation, can be used to express the delivery constructs. For example, Chinese hamster ovary (CHO) cells can be used to express the delivery constructs.
  • 5.7. Compositions Comprising Delivery Constructs
  • The delivery constructs of the invention can be formulated as compositions. The compositions are generally formulated appropriately for the immediate use intended for the delivery construct. For example, if the delivery construct is not to be administered immediately, the delivery construct can be formulated in a composition suitable for storage. One such composition is a lyophilized preparation of the delivery construct together with a suitable stabilizer. Alternatively, the delivery construct composition can be formulated for storage in a solution with one or more suitable stabilizers. Any such stabilizer known to one of skill in the art without limitation can be used. For example, stabilizers suitable for lyophilized preparations include, but are not limited to, sugars, salts, surfactants, proteins, chaotropic agents, lipids, and amino acids. Stabilizers suitable for liquid preparations include, but are not limited to, sugars, salts, surfactants, proteins, chaotropic agents, lipids, and amino acids. Specific stabilizers than can be used in the compositions include, but are not limited to, trehalose, serum albumin, phosphatidylcholine, lecithin, and arginine. Other compounds, compositions, and methods for stabilizing a lyophilized or liquid preparation of the delivery constructs may be found, for example, in U.S. Pat. Nos. 6,573,237, 6,525,102, 6,391,296, 6,255,284, 6,133,229, 6,007,791, 5,997,856, and 5,917,021.
  • Further, the delivery construct compositions of the invention can be formulated for administration to a subject. Such vaccine compositions generally comprise one or more delivery constructs of the invention and a pharmaceutically acceptable excipient, diluent, carrier, or vehicle. Any such pharmaceutically acceptable excipient, diluent, carrier, or vehicle known to one of skill in the art without limitation can be used. Examples of a suitable excipient, diluent, carrier, or vehicle can be found in Remington's Pharmaceutical Sciences, 21st Ed. 2005, Mack Publishing Co., Easton.
  • In certain embodiments, the delivery construct compositions are formulated for oral administration. In such embodiments, the compositions are formulated to protect the delivery construct from acid and/or enzymatic degradation in the stomach. Upon passage to the neutral to alkaline environment of the duodenum, the delivery construct then contacts a mucous membrane and is transported across the polarized epithelial membrane. The delivery constructs may be formulated in such compositions by any method known by one of skill in the art, without limitation.
  • In certain embodiments, the oral formulation comprises a delivery construct and one or more compounds that can protect the delivery construct while it is in the stomach. For example, the protective compound should be able to prevent acid and/or enzymatic hydrolysis of the delivery construct. In certain embodiments, the oral formulation comprises a delivery construct and one or more compounds that can facilitate transit of the construct from the stomach to the small intestine. In certain embodiments, the one or more compounds that can protect the delivery construct from degradation in the stomach can also facilitate transit of the construct from the stomach to the small intestine. Preferably, the oral formulation comprises one or more compounds that can protect the delivery construct from degradation in the stomach and facilitate transit of the construct from the stomach to the small intestine. For example, inclusion of sodium bicarbonate can be useful in facilitating the rapid movement of intra-gastric delivered materials from the stomach to the duodenum as described in Mrsny et al., 1999, Vaccine 17:1425-1433.
  • Other methods for formulating compositions so that the delivery constructs can pass through the stomach and contact polarized epithelial membranes in the small intestine include, but are not limited to, enteric-coating technologies as described in DeYoung, 1989, Int J Pancreatol. 5 Suppl:31-6, and the methods provided in U.S. Pat. Nos. 6,613,332, 6,174,529, 6,086,918, 5,922,680, and 5,807,832.
  • 5.7.1. Kits Comprising Compositions
  • In yet another aspect, the invention provides a kit that comprises a composition of the invention. In certain embodiments, the kit further comprises instructions that direct administration of the composition to a mucous membrane of the subject to whom the composition is administered. In certain embodiments, the kit further comprises instructions that direct oral administration of the composition to the subject to whom the composition is administered.
  • In certain embodiments, the kit comprises a composition of the invention in more or more containers. In certain embodiments, the composition can be in a unit dosage form, e.g., a tablet, lozenge, capsule, etc. In certain embodiments, the composition can be provided in or with a device for administering the composition, such as, for example, a device configured to administer a single-unit dose of the composition, e.g., an inhaler.
  • 5.8. Making and Testing Delivery Constructs
  • The delivery constructs of the invention are preferably produced recombinantly, as described below. However, the delivery constructs may also be produced by chemical synthesis using methods known to those of skill in the art.
  • 5.8.1. Manufacture of Delivery Constructs
  • Methods for expressing and purifying the delivery constructs of the invention are described extensively in the examples below. Generally, the methods rely on introduction of an expression vector encoding the delivery construct to a cell that can express the delivery construct from the vector. The delivery construct can then be purified for administration to a subject.
  • 5.8.2. Testing Delivery Constructs
  • Having selected the domains of the delivery construct, the function of these domains, and of the delivery constructs as a whole, can be routinely tested to ensure that the constructs can deliver a macromolecule across mucous membranes of a subject free from the remainder of the construct, For example, the delivery constructs can be tested for cell recognition, transcytosis and cleavage using routine assays. The entire chimeric protein can be tested, or, the function of various domains can be tested by substituting them for native domains of the wild-type toxin.
  • 5.8.2.1. Receptor binding/Cell recognition
  • Receptor binding domain function can be tested by monitoring the delivery construct's ability to bind to the target receptor. Such testing can be accomplished using cell-based assays, with the target receptor present on a cell surface, or in cell-free assays. For example, delivery construct binding to a target can be assessed with affinity chromatography. The construct can be attached to a matrix in an affinity column, and binding of the receptor to the matrix detected, or vice versa. Alternatively, if antibodies have been identified that bind to either the receptor binding domain or its cognate receptor, the antibodies can be used, for example, to detect the receptor binding domain in the delivery construct by immunoassay, or in a competition assay for the cognate receptor. An exemplary cell-based assay that detects delivery construct binding to receptors on cells comprises labeling the construct and detecting its binding to cells by, e.g., fluorescent cell sorting, autoradiography, etc.
  • 5.8.2.2. Transcytosis
  • The function of the transcytosis domain can be tested as a function of the delivery construct's ability to pass through an epithelial membrane. Because transcytosis first requires binding to the cell, these assays can also be used to assess the function of the cell recognition domain.
  • The delivery construct's transcytosis activity can be tested by any method known by one of skill in the art, without limitation. In certain embodiments, transcytosis activity can be tested by assessing the ability of a delivery construct to enter a non-polarized cell to which it binds. Without intending to be bound to any particular theory or mechanism of action, it is believed that the same property that allows a transcytosis domain to pass through a polarized epithelial cell also allows molecules bearing the transcytosis domain to enter non-polarized cells. Thus, the delivery construct's ability to enter the cell can be assessed, for example, by detecting the physical presence of the construct in the interior of the cell. For example, the delivery construct can be labeled with, for example, a fluorescent marker, and the delivery construct exposed to the cell. Then, the cells can be washed, removing any delivery construct that has not entered the cell, and the amount of label remaining determined. Detecting the label in this traction indicates that the delivery construct has entered the cell.
  • In other embodiments, the delivery construct's transcytosis ability can be tested by assessing the delivery construct's ability to pass through a polarized epithelial cell. For example, the delivery construct can be labeled with, for example, a fluorescent marker and contacted to the apical membranes of a layer of epithelial cells. Fluorescence detected on the basal-lateral side of the membrane formed by the epithelial cells indicates that the transcytosis domain is functioning properly.
  • 5.8.2.3. Cleavable Linker Cleavage
  • The function of the cleavable linker can generally be tested in a cleavage assay. Any suitable cleavage assay known by one of skill in the art, without limitation, can be used to test the cleavable linkers. Both cell-based and cell-free assays can be used to test the ability of an enzyme to cleave the cleavable linkers.
  • An exemplary cell-free assay for testing cleavage of cleavable linkers comprises preparing extracts of polarized epithelial cells and exposing a labeled delivery construct bearing a cleavable linker to the fraction of the extract that corresponds to membrane-associated enzymes. In such assays, the label can be attached to either the macromolecule to be delivered or to the remainder of the delivery construct. Among these enzymes are cleavage enzymes found near the basal-lateral membrane of a polarized epithelial cell, as described above. Cleavage can be detected, for example, by binding the delivery construct with, for example, an antibody and washing off unbound molecules. If label is attached to the macromolecule to be delivered, then little or no label should be observed on the molecule bound to the antibodies. Alternatively, the binding agent used in the assay can be specific for the macromolecule, and the remainder of the construct can be labeled. In either case, cleavage can be assessed.
  • Cleavage can also be tested using cell-based assays that test cleavage by polarized epithelial cells assembled into membranes. For example, a labeled delivery construct, or portion of a delivery construct comprising the cleavable linker, can be contacted to either the apical or basolateral side of a monolayer of suitable epithelial cells, such as, for example, Coco-2 cells, under conditions that permit cleavage of the linker. Cleavage can be detected by detecting the presence or absence of the label using a reagent that specifically binds the delivery construct, or portion thereof. For example, an antibody specific for the delivery construct can be used to bind a delivery construct comprising a label distal to the cleavable linker in relation to the portion of the delivery construct bound by the antibody. Cleavage can then be assessed by detecting the presence of the label on molecules bound to the antibody. If cleavage has occurred, little or no label should be observed on the molecules bound to the antibody. By performing such experiments, enzymes that preferentially cleave at the basolateral membrane rather than the apical membrane can be identified, and, further, the ability of such enzymes to cleave the cleavable linker in a delivery construct can be confirmed.
  • Further, cleavage can also be tested using a fluorescence reporter assay as described in U.S. Pat. No. 6,759,207. Briefly, in such assays, the fluorescence reporter is contacted to the basolateral side of a monolayer of suitable epithelial cells under conditions that allow the cleaving enzyme to cleave the reporter. Cleavage of the reporter changes the structure of the fluorescence reporter, changing it from a non-fluorescent configuration to a fluorescent configuration. The amount of fluorescence observed indicates the activity of the cleaving enzyme present at the basolateral membrane.
  • Further, cleavage can also be tested using an intra-molecularly quenched molecular probe, such as those described in U.S. Pat. No. 6,592,847. Such probes generally comprise a fluorescent moiety that emits photons when excited with light of appropriate wavelength and a quencher moiety that absorbs such photons when in close proximity to the fluorescent moiety. Cleavage of the probe separates the quenching moiety from the fluorescent moiety, such that fluorescence can be detected, thereby indicating that cleavage has occurred. Thus, such probes can be used to identify and assess cleavage by particular cleaving enzymes by contacting the basolateral side of a monolayer of suitable epithelial cells with the probe under conditions that allow the cleaving enzyme to cleave the probe. The amount of fluorescence observed indicates the activity of the cleaving enzyme being tested.
  • 6. EXAMPLES
  • The following examples merely illustrate the invention, and are not intended to limit the invention in any way.
  • 6.1. Construction of a Delivery Construct
  • Five exemplary delivery construct expression vectors for delivering rat growth hormone (rGH) were constructed according to the following protocol. First, the rGH gene was amplified by PCR, incorporating restriction enzymes pairs of NdeI and EcoRI, PstI and PstI, AgeI and EcoRI, or PstI and EcoRI sites at two ends of the PCR products. After restriction enzyme digestion, the PCR products were cloned into pPE64-PstI-Δ553, which was digested with the corresponding restriction enzyme pairs. The resulting constructs were named as pPE-RGH(NdeI-EcoRI), pntPE-RGH(PstI), pntPE-RGH(AgeI-EcoRI), and pPE-RGH(PstI-EcoRI). These constructs thus comprise sequences encoding Domains I and II of ntPE (amino acids 26-372 as shown in FIG. 3) and rGH (Accession No. P01244; see Seeburg et al., 1977, Nature 270:486-494 and Page et al., 1981, Nucleic Acids Res. 9:2087-2104), and are also tagged with a 6-His motif at the N-terminus of the polypeptide to facilitate purification. The final plasmids were verified by restriction enzyme digestions and DNA sequencing.
  • Expression vectors comprising cleavable linkers were constructed by introducing sequences encoding the appropriate amino acid sequence. To do so, oligonucleotides that encode sequences complementary to appropriate restriction sites and one of the following amino acid sequences were synthesized, then ligated into an expression vector prepared as described above between the ntPE sequences and the rGH sequences. For Delivery Construct 1, the cleavable linker sequence was RQPRGGL. For Delivery Construct 2, the cleavable linker sequence was GGLRQPR. For Delivery Construct 3, the cleavable linker sequence was RQPREGR. For Delivery Construct 4, the cleavable linker sequence was RQPRVGR. For Delivery Construct 5, the cleavable linker sequence was RQPRARR.
  • To separate rGH from PE protein in the event that the fusion protein is taken up by antigen presenting cells, a protease furin site was also inserted between the cleavable linker and rGH. To do so, constructs containing a sequence encoding the furin site with the five different cleavable linkers were made. Oligonucleotide sequences for the five cleavable linkers and a furin clip site are shown in Table 3 below. Each of the oligo duplexes was inserted into PstI site of pPE-RGH(PstI-EcoRI). The final constructs, named as pPE-RGH-F1, pPE-RGH-F2, pPE-RGH-F3, pPE-RGH-F4, pPE-RGH-F5 were confirmed by restriction enzyme digestion and DNA sequencing.
  • TABLE 3
    Oligonucleotide pairs for introducing a furin
    cleavage site and protease cleavage site
    pPE-RGH-1 1 F:
    AACTGCAGCGCCAGCCTCGAGGAGGATTACTGCAGAA
    (SEQ ID NO: 24)
    1 R:
    TTCTGCAGTAATCCTCCTCGAGGCTGGCGCTGCAGTT
    (SEQ ID NO: 25)
    pPE-RGH-2 2 F:
    AACTGCAGGGAGGCTTACGCCAGCCTCGACTGCAGAA
    (SEQ ID NO: 26)
    2 R:
    TTCTGCAGTCGAGGCTGGCGTAAGCCTCCCTGCAGTT
    (SEQ ID NO: 27)
    pPE-RGH-3 3 F:
    AACTGCAGCGCCAGCCTCGAGAGGGCCGTCTGCAGAA
    (SEQ ID NO: 28)
    3 R:
    TTCTGCAGACGGCCCTCTCGAGGCTGGCGCTGCAGTT
    (SEQ ID NO: 29)
    pPE-RGH-4 4 F:
    AACTGCAGCGCCAGCCTCGAGTCGGCCGTCTGCAGAA
    (SEQ ID NO: 30)
    4 R:
    TTCTGCAGACGGCCGACTCGAGGCTGGCGCTGCAGTT
    (SEQ ID NO: 31)
    pPE-RGH-5 5 F:
    AACTGCAGCGCCAGCCTCGAGCACGTCGTCTGCAGAA
    (SEQ ID NO: 32)
    5 R:
    TTCTGCAGACGACGTGCTCGAGGCTGGCGCTGCAGTT
    (SEQ ID NO: 33)
  • 6.2. Expression of Delivery Constructs
  • E. coli BL21(DE3) pLysS competent cells (Novagen, Madison, Wis.) were transformed using a standard heat-shock method in the presence of the appropriate plasmid to generate ntPE-rat Growth Hormone (rGH) expression cells, selected on ampicillin-containing media, and isolated and grown in Luria-Bertani broth (Difco; Becton Dickinson, Franklin Lakes, N.J.) with antibiotic, then induced for protein expression by the addition of 1 mM isopropyl-D-thiogalactopyranoside (IPTG) at OD 0.6. Two hours following IPTG induction, cells were harvested by centrifugation at 5,000 rpm for 10 min. Inclusion bodies were isolated following cell lysis and proteins were solubilized in the buffer containing 100 mM Tris_HCl (pH 8.0), 2 mM EDTA, 6 M guanidine HCl, and 65 mM dithiothreitol. Solubilized His ntPE-rGH is refolded in the presence of 0.1 M Tris, pH=7.4, 500 mM L-arginine, 0.9 mM GSSG, 2 mM EDTA. The refolded proteins were purified by Q sepharose Ion Exchange and Superdex 200 Gel Filtration chromatography (Amersham Biosciences, Inc., Sweden). The purity of proteins was assessed by SDS-PAGE and analytic HPLC (Agilent, Inc. Palo Alto, Calif.).
  • 6.3. Characterization of a Delivery Construct
  • The following procedures can be used to assess proper refolding of a delivery construct. The protein refolding process is monitored by measuring, e.g., Delivery Construct 1 binding activity with ntPE binding receptor, CD 91 receptors, and rGH binding proteins on a Biacore SPR instrument (Biacore, Sweden) according to the manufacturer's instructions. Proper refolding of other macromolecules in exemplary constructs can be tested in similar binding assays with appropriate binding agents. By testing such binding affinities, the skilled artisan can assess the proper folding of each portion of the delivery construct.
  • 6.4. Delivery Construct Cleavage Assays
  • This example describes experiments performed to identify and verify enzymes that can be used to cleave the cleavable linkers of the delivery constructs described herein. First, Caco-2 (ATCC Accession No. HTB-37) cells in passage 21 were obtained from American Type Culture Collection (Manassas, Va.). Human tracheal epithelial (HTE) cells were obtained from J. Whiddecombe of the Department of Physiology at the University of California, Davis Medical School. Caco-2 cells were routinely grown on 75 cm2 plastic culture flasks (Becton Dickinson, Franklin Lakes, N.J.) in DMEM containing 10% fetal bovine serum and 1% penicillin-streptomycin at 37° C. in a 5% CO2/95% air atmosphere. HTE cells were grown as described in Yamaya et al., 1992, Am J Physiol 262(6 Pt 1):L713-24.
  • To identify suitable cleavable linkers, HTE or Caco-2 cells were seeded at a density of 5×104 cells/cm2 onto 24-well collagen-coated polycarbonate transwell filters (Corning, Acton, Mass.) for 12-14 days. Confluent monolayers achieved a transepithelial resistance (TER) of >500 ohm·cm2, as measured using an EVOM epithelial voltohmmeter and STX2 electrode (World Precision Instruments, Sarasota, Fla.). To determine specific enzyme activity, substrates specific for the tested peptidase (500 μM or 1 mM substrate in 250 μl DMEM without FBS or antibiotics) were added to either the apical (AP) or basolateral (BL) side of the monolayers. Peptidase substrates were obtained from Calbiochem, Inc. (Division of EMD Biosciences, Inc., San Diego, Calif.). Cells were incubated for 2 hrs at 37° C. in a 5% CO2/95% air atmosphere. Both the apical and basolateral media was then measured for its specific enzyme activity according to the manufacturer's instruction. Cleavage was assessed by detecting fluorescence of the substrates, which reflects cleavage because it separates of the quenching agent from the fluorescent agent present on the substrate, which separation allows fluorescence to be detected.
  • Table 4 presents a summary of the results of these assays using HTE cells, while Table 5 presents a summary of the results of these assays using Caco-2 cells. For all results, baseline control values were subtracted from substrate values before percentages were determined and tests were performed at least in duplicate. The percentages presented in the tables represent the percent increase observed in assay in the apical or basolateral media, which depends on which side of the membrane exhibits higher peptidase activity. It should be noted that, even when substrate was added to the media on the apical side of the membrane, peptidase activity can be observed on the basolateral side of the membrane because of diffusion of the substrate across the membrane.
  • TABLE 4
    AP BL
    Peptidase tested in % % A405 A405
    HTE cells AP > BL BL > AP nm nm
    AP-500 uM Cathepsin B I 86% 0.37 0.20
    BL-500 uM Cathepsin B I 64% 0.35 0.21
    AP-500 uM Cathepsin G I 882%  0.04 0.004
    BL-500 uM Cathepsin G I  6% 0.02 0.02
    AP-500 uM Cathepsin G II 371%  0.02 0.01
    BL-500 uM Cathepsin G II  0%  0% 0.02 0.02
    AP-500 uM Cathepsin G III 11% 0.02 0.02
    BL-500 uM Cathepsin G III 400%  0.01 0.03
    AP-500 uM Chymotrypsin I 74% 0.04 0.02
    BL-500 uM Chymotrypsin I 36% 0.03 0.04
    AP-500 uM Elastase I 49% 0.05 0.03
    BL-500 uM Elastase I 23% 0.02 0.03
    AP-500 uM Elastase II 43% 0.29 0.20
    BL-500 uM Elastase II 31% 0.18 0.13
    AP-500 uM Elastase III 89% 0.04 0.02
    BL-500 uM Elastase III 967%  0.03 0.003
    AP-500 uM Elastase IV 84% 0.35 0.19
    BL-500 uM Elastase IV 65% 0.23 0.14
    AP-500 uM Elastase VIII 529%  0.16 0.03
    BL-500 uM Elastase VIII 181%  0.09 0.03
    AP-500 uM Papain 57% 0.02 0.02
    BL-500 uM Papain  5% 0.03 0.03
    AP-500 uM Subtilisin A I  9% 0.02 0.02
    BL-500 uM Subtilisin A I 3000%  0.001 0.03
    AP-500 uM Subtilisin A II 21% 0.02 0.02
    BL-500 uM Subtilisin A II 55% 0.01 0.02
    AP-500 uM Thrombin I 42% 0.15 0.11
    BL-500 uM Thrombin I 15% 0.09 0.10
    AP-500 uM Thrombin II 445%  0.40 0.07
    BL-500 uM Thrombin II 741%  0.41 0.05
    AP-500 uM Urokinase I  8% 0.11 0.10
    BL-500 uM Urokinase I  4% 0.13 0.13
    AP-1 mM Cathepsin B I 17% 0.18 0.15
    BL-1 mM Cathepsin B I 42% 0.24 0.17
    AP-1 mM Cathepsin G I 114%  0.05 0.02
    BL-1 mM Cathepsin G I 47% 0.02 0.03
    AP-1 mM Cathepsin G II 138%  0.05 0.02
    BL-1 mM Cathepsin G II 19% 0.03 0.03
    AP-1 mM Cathepsin G III 225%  0.07 0.02
    BL-1 mM Cathepsin G III 54% 0.02 0.03
    AP-1 mM Chymotrypsin I 35% 0.06 0.04
    BL-1 mM Chymotrypsin I 90% 0.04 0.07
    AP-1 mM Elastase I 108%  0.02 0.03
    BL-1 mM Elastase I 864%  0.01 0.09
    AP-1 mM Elastase II 62% 0.28 0.17
    BL-1 mM Elastase II 42% 0.33 0.23
    AP-1 mM Elastase III 318%  0.02 0.01
    BL-1 mM Elastase III 131%  0.04 0.02
    AP-1 mM Elastase IV 94% 0.41 0.21
    BL-1 mM Elastase IV 61% 0.30 0.19
    AP-1 mM Elastase VIII 233%  0.14 0.04
    BL-1 mM Elastase VIII 41% 0.06 0.05
    AP-1 mM Papain 424%  0.02 0.004
    BL-1 mM Papain 141%  0.02 0.01
    AP-1 mM Subtilisin A I 18% 0.03 0.03
    BL-1 mM Subtilisin A I 290%  0.01 0.04
    AP-1 mM Subtilisin A II 17% 0.03 0.02
    BL-1 mM Subtilisin A II 318%  0.01 0.04
    AP-1 mM Thrombin I 27% 0.17 0.14
    BL-1 mM Thrombin I 28% 0.17 0.13
    AP-1 mM Thrombin II 20% 0.12 0.10
    BL-1 mM Thrombin II 13% 0.05 0.06
    AP-1 mM Urokinase I 14% 0.19 0.21
    BL-1 mM Urokinase I  4% 0.21 0.20
  • TABLE 5
    AP BL
    Peptidase tested in % % A405 A405
    Caco-2 cells AP > BL BL > AP nm nm
    AP-500 uM Cathepsin B I 150% 0.14 0.06
    BL-500 uM Cathepsin B I  34% 0.17 0.12
    AP-10 mM Cathepsin G I 195% 0.31 0.10
    BL-10 mM Cathepsin G I 145% 0.42 1.03
    AP-500 uM Cathepsin G III  35% 0.014 0.01
    BL-500 uM Cathepsin G III 185% 0.03 0.01
    AP-500 uM Cathepsin G I 232% 0.05 0.01
    BL-500 uM Cathepsin G I 1709%  0.01 0.15
    AP-500 uM Cathepsin G II 3900%  0.01 0.0003
    BL-500 uM Cathepsin G II 1330%  0.003 0.04
    AP-500 uM Chymotrypsin I 342% 0.01 0.04
    BL-500 uM Chymotrypsin I 403% 0.03 0.16
    AP-500 uM Elastase I  73% 0.01 0.01
    BL-500 uM Elastase I 295% 0.04 0.01
    AP-500 uM Elastase II  59% 0.12 0.07
    BL-500 uM Elastase II  89% 0.01 0.02
    AP-500 uM Elastase III  85% 0.01 0.07
    BL-500 uM Elastase III 320% 0.003 0.01
    AP-500 uM Elastase IV  32% 0.11 0.08
    BL-500 uM Elastase IV  12% 0.02 0.02
    AP-500 uM Elastase VIII  16% 0.02 0.02
    BL-500 uM Elastase VIII 115% 0.01 0.02
    AP-500 uM Papain  19% 0.018 0.02
    BL-500 uM Papain 339% 0.07 0.02
    AP-500 uM Subtilisin A I ***23%  0.05
    BL-500 uM Subtilisin A I ***94%  0.20
    AP-500 uM Subtilisin A II N/A
    BL-500 uM Subtilisin A II ***11%  0.02
    AP-500 uM Thrombin I  81% 0.04 0.02
    BL-500 uM Thrombin I 254% 0.01 0.04
    AP-Thrombin II 500 uM  42% 0.08 0.06
    BL-Thrombin II 500 uM  62% 0.09 0.06
    AP-500 uM Urokinase I 111% 0.12 0.06
    BL-500 uM Urokinase I 1044%  0.005 0.05
    AP-1 mM Cathepsin B I 109% 0.27 0.13
    BL-1 mM Cathepsin B I  58% 0.12 0.2
    AP-20 mM Cathepsin G I 129% 0.10 0.23
    BL-20 mM Cathepsin G I 540% 0.11 0.70
    AP-1 mM Cathepsin G III  37% 0.01 0.01
    BL-1 mM Cathepsin G III 103% 0.07 0.03
    AP-1 mM Cathepsin G I 107% 0.08 0.04
    BL-1 mM Cathepsin G I 144% 0.12 0.05
    AP-1 mM Cathepsin G II  11% 0.05 0.06
    BL-1 mM Cathepsin G II 7850%  0.00 0.04
    AP-1 mM Chymotrypsin I 107% 0.08 0.04
    BL-1 mM Chymotrypsin I 288% 0.02 0.07
    AP-1 mM Elastase I 217% 0.03 0.001
    BL-1 mM Elastase I 880% 0.003 0.02
    AP-1 mM Elastase II  27% 0.17 0.14
    BL-1 mM Elastase II  34% 0.02 0.03
    AP-1 mM Elastase III 192% 0.02 0.01
    BL-1 mM Elastase III  77% 0.02 0.01
    AP-1 mM Elastase IV  42% 0.16 0.11
    BL-1 mM Elastase IV  10% 0.04 0.05
    AP-1 mM Elastase VIII  70% 0.05 0.03
    BL-1 mM Elastase VIII 332% 0.11 0.03
    AP-1 mM Papain  61% 0.02 0.01
    BL-1 mM Papain  0% 0.005 0.005
    AP-1 mM Subtilisin A I ***61%  0.13
    BL-1 mM Subtilisin A I ***44%  0.09
    AP-1 mM Subtilisin A II N/A
    BL-1 mM Subtilisin A II N/A
    AP-1 mM Thrombin I 420% 0.11 0.02
    BL-1 mM Thrombin I 3400%  0.005 0.16
    AP-Thrombin II 1 mM 163% 0.14 0.05
    BL-Thrombin II 1 mM  29% 0.11 0.09
    AP-1 mM Urokinase I  57% 0.17 0.11
    BL-1 mM Urokinase I 230% 0.05 0.15
    ***denotes % over baseline control only
    “—” denotes values below baseline control
  • 6.5. Delivery of an Exemplary Macromolecule—Green Fluorescent Protein
  • The following example describes experiments performed to assess the transcytosis of an exemplary delivery construct for delivering green fluorescent protein (“GFP”) across a mouse epithelial membrane. It is noted that this exemplary delivery construct does not comprise a cleavable linker; however, the presence or absence of the cleavable linker should not affect transcytosis of the delivery construct.
  • Briefly, a nt-PE-GFP construct was applied to the trachea of anesthetized female balb/c mice which were approximately 8 weeks of age. The mice were anesthetized with inhaled isoflurane and the trachea was exposed. A small hole was made on the trachea to allow application of our GFP material. In our experiments, 100 μg of GFP alone or ntPE-GFP was used, respectively. The GFP material was slowly dripped directly onto the exposed trachea in a 100 μl volume. After 15 minutes, the mice were euthanized by CO2 asphyxiation. The trachea was removed and frozen in OCT (cat#25608-930-Tissue Tek) using biopsy cryomolds (cat#4565-Tissue Tek). The samples were sectioned onto slides and visualized by fluorescence microscopy (Nikon model Eclipse E400).
  • Micrographs of the epithelial sections are presented as FIGS. 1A-1C. FIG. 1A shows the nt-PE-GFP construct adhering strongly to the apical surface of the trachea epithelium. FIG. 1B shows transcytosis of the nt-PE-GFP construct across the trachea epithelium. FIG. 1C shows release of the nt-PE-GFP construct from the basolateral side of the trachea epithelium. FIG. 1D presents a micrograph of a negative control, a tracheal epithelial section from a mouse contacted with GFP alone. The tissues exposed for 15 minutes to obtain these micrographs.
  • The micrographs demonstrate that nt-PE-GFP interacts strongly with receptors on the apical surface of mouse tracheal epithelium, transcytoses across such epithelial tissue, and releases from the basolateral surface of the mouse tracheal epithelium.
  • In addition, plasma concentrations of the nt-PE-GFP construct were determined following administration of the delivery construct using an ELISA assay as described in Example 6.6.1, below. Serum samples were taken from anesthetized mice that had received intranasal administration of 100 μg of the nt-PE-GFP delivery construct every 30 minutes following administration. FIG. 2 presents the results of this experiment, demonstrating that peak plasma levels of the delivery construct reached between 500-900 ng/ml, indicating that the delivery construct displayed approximately 22% bioavailability following intranasal administration.
  • 6.6. Detection of Growth Hormone Protein in Tissue by Histological Examination
  • This example describes histological detection in tissues of a representative macromolecule for delivery, growth hormone. Following administration of a delivery construct, animals are euthanized by CO2 asphyxiation and exanguinated by cardiac puncture. Specific tissues (lymph nodes, trachea, brain, spleen liver, GI tract) are removed, briefly rinsed in PBS to remove any residual blood and frozen in OCT. Sections (5 microns thick) are placed onto slides. Slides are fixed in acetone for 10 min and rinsed with PBS. Slides are incubated with 3% peroxidase for 5 min. Slides are then blocked with protein for an additional 5 min. Primary growth hormone antibody is incubated onto slides for 30 min at a 1:100 dilution followed by PBS washes. Biotin-labeled secondary antibody is then incubated for approximately 15 minutes followed by PBS washes. Streptavidin HRP label is incubated onto slides for 15 min followed by PBS washes. HRP Chromagen is applied for 5 min followed by several rinses in distilled H 20. Finally, the slides are counterstained with hematoxylin for 1 min, coverslipped, and examined for the presence of GH.
  • 6.7. Transport and Cleavage of an Exemplary Delivery Construct in an in Vitro System
  • This example describes transport and cleavage of an exemplary delivery construct, Delivery Construct 2, comprising rat growth hormone (rGH) in an in vitro system using human tracheal epithelial cells.
  • 6.7.1. Growth of Human Tracheal Epithelial Cells
  • Human tracheal epithelial (HTE) cells were isolated from tracheas as previously described and cultured on semi-permeable filter systems (0.45 um pore size; Corning, Acton, Mass.) coated with human placental collagen. See Yamaya et al., 1992, Am J Physiol, 262:L713-24 and Sachs et al., 2003, In Vitro Cell Dev Biol Anim, 39:56-62. Cell sheets were used at >10 days following plating, at which time they had a transepithelial resistance (TER) of >100 Ω·ohms·cm2 as measured with a “chopstick” voltohmeter (Millicell ERS, Manassas, Va.).
  • Caco-2 cells in passage 21 were obtained from American Type Culture Collection (Manassas, Va.). Cells were routinely grown on 75 cm2 plastic culture flasks (Becton Dickinson, Franklin Lakes, N.J.) in DMEM containing 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin at 37° C. in a 5% CO2/95% air atmosphere. For the transport and cleavage studies, Caco-2 cells were seeded at a density of 5×104 cells/cm2 onto 24-well collagen-coated polycarbonate transwell filters (Corning, Acton Mass.) for 12-14 days. Confluent monolayers achieved a transepithelial resistance (TER) of >500 ohm/cm2, as measured using the EVOM and STX2 electrode (World Precision Instruments).
  • 6.7.2. Transport and Cleavage Assays
  • To determine transport and cleavage activity, Delivery Constructs 1 and 2 proteins (10 μg in 100 μl DMEM without phenol red, FBS or antibiotics) were added to the apical side of the epithelial monolayer. Cells were incubated for 4 hrs at 37° C. in a 5% CO2/95% air atmosphere. Both the apical and basolateral media was then assayed for its transport and cleavage activity by testing for the presence of rGH cleaved from the delivery construct by western blot analysis, as described below. As a control, 10 μg of dextran fluorescein was also added to the apical wells in order to check for leakage.
  • Following the 4 hour incubation described above, apical and basolateral media samples were precipitated by the addition of trichloroacetic acid (TCA). The amount of TCA added to each sample was twenty percent relative to the sample volume. Each sample was vortexed and placed on ice for 30 minutes. After samples this incubation on ice, samples were centrifuged at 14,000 RPM for 10 minutes. Next, the supernatants from each sample were aspirated and tubes containing the remaining pellet were left open to air dry. 4 μl 0.2M NaOH was added to each pellet. Five minutes after the addition of NaOH, pellets were resuspended in 36 μl 8M Urea.
  • Next, 10 μl Sample Buffer containing DTT (Invitrogen NP0007, NP0009) was added to each sample. Samples were then placed on a 100° C. heating block for 5 minutes. Half (19 μl) of each sample was then loaded on to a 4%-12% Tris Bis Gel (Invitrogen NP3022BOX). For controls, recombinant rat GH (RDI R0125) and Delivery Construct 1 or Delivery Construct 2 proteins were also loaded directly into the gel. Electrophoresis was at 150V for 30 minutes. From the gel, samples were transferred onto nitrocellulose at 30V for 1 hour.
  • The Blocking Solution, Antibody Diluents, and Antibody Wash Solution from Invitrogen's WesternBreeze were used in subsequent steps. The nitrocellulose membrane was placed in blocking buffer and incubated at 4° C., overnight. The membrane was washed 3 times for 3 minutes each time. The membrane was then incubated at RT with 10 ml of 1:2000 rabbit anti growth hormone (RDI RDIRtGHabr). After one hour, the membrane was again rinsed 3 times for 3 minutes each. Membrane was incubated for 1 hour at room temperature with 10 ml of goat anti rabbit IgG AP (Pierce 31340) at 1:5000. The membrane was rinsed 3 times for 3 minutes per wash. 5 μl of substrate (Pierce 34042) was added to the membrane. After color development reached desired intensity, reaction was halted by the removal of substrate and the addition of purified water. Finally, the membrane was washed in purified water for 30 minutes and air dried.
  • Results of the Western Blot analysis are presented in FIG. 4. As seen in FIG. 4, media from the basolateral side of the epithelial cell layer contained protein consistent with cleaved rGH separated from the remainder of Delivery Construct 2. In contrast, media from the apical side of the epithelial cell layer contained largely intact delivery construct. Thus, application of Delivery Construct 2 to the apical side of the human epithelial cell membrane resulted in both transport to the basolateral side of the membrane and proper cleavage of the construct as shown by release of rGH detectable with anti-r(1H antibody and of proper apparent molecular weight. Similar results were also observed for Delivery Construct 1 (data not shown).
  • 6.8. Delivery of an Exemplary Macromolecule in an In Vivo System
  • This example describes use of exemplary Delivery Construct 2 in a mouse model, showing effective transport and cleavage of the delivery construct in vivo and the bioactivity of the macromolecule delivered by Delivery Construct 2, rGH.
  • 6.8.1. Administration of a Delivery Construct Comprising Rat Growth Hormone
  • Using an animal feeding needle, 100 μg of Delivery Construct 2 (in 250 μl total volume) was orally delivered to female BALB/c mice, 5-6 weeks of age (Charles River Laboratories, Wilmington, Mass.). Delivery Construct 2 was diluted in 1 mg/ml bovine serum albumin (BSA) and phosphate buffered saline (PBS). As a positive control, control mice were subcutaneously (SC) injected on their dorsal side with 30 μg of recombinant rat growth hormone (rGH) diluted in PBS (100 μl total volume). At specific times after oral gavage and SC administration, mice were euthanized by CO2 asphyxiation and exsanguinated. Whole blood and liver were collected and analyzed as described below. Because in the difference in molecular weights between Delivery Construct 2 and rGH, essentially the same number of rGH molecules were administered in both routes.
  • 6.8.2. Pharmacokinetics of an Exemplary Macromolecule Administered with a Delivery Construct in an In Vivo System
  • To assess the pharmacokinetics of an exemplary macromolecule delivered with a delivery construct, ELISA assays were used to measure serum concentrations of rGH at defined timepoints following administration. The serum concentration data thus obtained was used to compare the pharmacokinetics of rGH administered with Delivery Construct 2 to those observed with conventional subcutaneous administration. The ELISA assays were performed as follows.
  • Costar 9018 E.I.A./R.I.A. 96-well plates were coated overnight with 200 ng/well of goat anti-rGH (Diagnostics Systems Laboratories, Cat. No. R01235) in 0.2M NaHCO3—Na2CO3, pH 9.4. Each 96-well plate was washed four times with PBS containing 0.05% Tween 20-0.01% thimerosal (wash buffer); blocked for 1 h with 200 μl/well of PBS/Tween 20 containing 0.5% BSA-0.01% thimerosal (assay buffer). The standard curve was prepared using recombinant rat GH (Diagnostics Systems Laboratories, Cat. No. R01205) diluted in assay buffer (PBST-0.5% BSA). The first point of the standard curve was prepared by adding 50 μl recombinant rat GH to 10 ml assay buffer (1:200), vortexed, and moved 200 μl to 800 μl assay buffer (1:5). For each plate, 0.5 ml was moved to 0.5 ml assay buffer by doing 1:2 serial dilutions for all the subsequent points. The 10 points of the standard curve are: 100, 50, 25, 12.5, 6.25, 3.125, 1.56, 0.78, 0.39 and 0.195 ng/well. Samples were diluted at 1:10 with assay buffer, loaded 100 μl/well in triplicates onto a 96-well plate, and incubated overnight. Each 96-well plate was then washed four times with wash buffer, loaded 100 μl/well of 2nd Ab (rabbit anti-rGH, Cell Sciences, Cat. No. PAAC1) at 1:300 in assay buffer (PBST-0.5% BSA) and incubate at RT for four hours. Each 96-well plate was then washed four times with wash buffer, loaded 100 μl/well of 3rd Ab (goat anti-rabbit IgG-horseradish peroxidase (HRP), Pierce, Cat. No. 31460) at 1:2000 in assay buffer (PBST-0.5% BSA) and incubated at room temperature for two hours. All incubation and coating steps were performed at room temperature on a shaker at 6 RPM. The HRP substrate, TMB (3,3′,5,5″tetramethylbenzidine), used to quantify bound antibody, was measured at 450 nm.
  • ELISA results are reported as the averages of the triplicate OD (450 nm) value of each sample. Rat GH concentrations were determined by the exceeding mean value plus three times the standard error of the mean (SEM) of the appropriate control value.
  • The results of the ELISA assays are presented in FIGS. 5-7. FIG. 5 presents serum rGH concentrations in BALB/c mice which were dosed by subcutaneous (SC) injection of 30 μg of non-glycosylated recombinant rat GH. Individual mice sera were tested at a dilution of 1:10, and the group average of rGH concentration were reported (n=4 mice per time point). Standard error of the mean (SEM) was indicated by the error bars. As shown in FIG. 5, peak serum concentration of about 240 ng/ml rGH was observed 30 minutes after administration of subcutaneous rGH.
  • FIG. 6 presents serum rGH concentrations in BALB/c mice which were dosed orally with 100 μg of Delivery Construct 2. Individual mice sera were tested at a dilution of 1:10, and the group average of rGH concentration were reported (n=4 mice per time point). Standard error of the mean (SEM) was indicated by the error bars. As shown in FIG. 5, peak serum concentration of about 280 ng/ml rGH was observed 20 minutes after administration of Delivery Construct 2.
  • FIG. 7 presents a graphical representation comparing pharmacokinetics of rGH delivered subcutaneously and with Delivery Construct 2. The curve fitting comparison between rGH (SC) and Delivery Construct 2(Oral) was performed by evaluating ELISA data as described above using PK Solutions 2.0, Pharmacokinetics Data Analysis (Summit Research Services, Montrose, Colo.). As shown in FIG. 7, Delivery Construct 2 yields substantially higher peak serum rGH concentrations than subcutaneous administrations of rGH. Further, the peak serum concentration is achieved faster with Delivery Construct 2 relative to subcutaneous rGH. The bioavailability of rGH delivered with Delivery Construct 2 observed was about 60% relative to rGH administered by subcutaneous injection.
  • 6.8.3. Assays Demonstrating Activity of a Macromolecule Following Delivery with a Delivery Construct
  • This example describes analysis of the biological effects of an exemplary macromolecule, rGH, delivered with Delivery Construct 2 in an in vivo system. In brief, insulin-like growth factor I-binding protein 3 (IGF-1-BP3), growth hormone (GH) receptor and insulin-like growth factor I (IGF-I) expression levels were assessed in liver tissue from mice administered either Delivery Construct 2 or subcutaneous rGH as described above. These transcripts were analyzed because of the well-characterized effects of GH binding to its receptor on IGF-1-BP3 and GH receptor levels. In particular, functional activation of the GH receptor following binding by GH is known to result in upregulation of IGF-1-BP3 and downregulation of GH receptor expression. Of these, upregulation of IGF-1-BP3 mRNA expression is believed to be the most reliable indicator of GH receptor activation. See, e.g., Sondergaard et al., 2003, Am J Physiol Endocrinol Metab 285:E427-32.
  • Thus, Quantitative Real Time PCR was used to detect and quantify the amount of IGF-1-BP3, GH receptor, IGF-I, and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) mRNA in approximately 30 mg of mouse liver tissue prepared as described above. Collected liver tissue was stored at −70° C. until further processing. Real-time detection of PCR was performed using the Applied Biosystems 7300 Real Time PCR system (Applied Biosystems, Foster City, Calif.). Total RNA from mouse liver was isolated according to the RNeasy Protect Mini Kit (Qiagen). Total RNA was used to generate cDNA for oligo dT oligodeoxynucleotide primer (T12-18) following the protocol for Omniscript Reverse Transcriptase (Qiagen). The primers used to amplify the cDNA were designed using Primer Express software (Applied Biosystems), synthesized by Operon (Alameda, Calif.), and are shown in Table 6:
  • TABLE 6
    RT-PCR Primers
    IGF-I-BP3 (forward): CGCAGAGAAATGGAGGACACA;
    IGF-I-BP3 (reverse): GGACGCCTCTGGGACTCA;
    GH receptor GTTGACGAAATAGTGCAACCTGAT;
    (forward):
    GH receptor CACGAATCCCGGTCAAACTAA;
    (reverse):
    IGF-I (forward): GCTATGGCTCCAGCATTCG;
    IGF-I (reverse): GCTCCGGAAGCAACACTCA
    GAPDH (forward): GCAACAGGGTGGTGGACCT
    GAPDH (reverse): GGATAGGGCCTCTCTTGCTCA
  • Equal amounts of cDNA were used in duplicate and amplified with the SYBR Green I Master Mix (Applied Biosystems). The thermal cycling parameters were as follows: thermal activation for 10 min at 95° C., and 40 cycles of PCR (melting for 15 s at 95° C. and annealing/extension for 1 min at 60° C.). A standard curve was constructed with a dilution curve (1:10, 1:100, 1:500, 1:1,000, 1:2,000) of total RNA from a control mouse liver sample. A “no template control” was included with each PCR. Amplification efficiencies were validated and normalized against GAPDH. Correct PCR product size was confirmed by electrophoresis through a 1% agarose gel stained with ethidium bromide. Purity of the amplified PCR products was determined by a heat-dissociation protocol.
  • The results of this analysis are shown in FIGS. 8-10. FIG. 8 shows expression levels of IGF-1-BP3 mRNA in the liver of female BALB/c mice treated with 30 μg recombinant rGH by subcutaneous injection or with 100 μg of Delivery Construct 2 by oral gavage. Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-1-BP3, as described above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control. As shown in FIG. 8, administration of subcutaneous rGH resulted in an about 250% increase in IGF-1-BP3 mRNA expression levels in the liver at 60 minutes following administration. In contrast, Delivery Construct 2 caused an almost 400% increase in IGF-1-BP3 mRNA expression levels in the liver 30 minutes following administration. Thus, oral administration of Delivery Construct 2 effectively delivered rGH to the bloodstream of the test mice, thereby demonstrating that a delivery construct can effectively deliver an active macromolecule across a mucous membrane in an in vivo system. Moreover, Delivery Construct 2 delivered more active rGH to the liver than subcutaneous administration, and the effects of administration of rGH were observed substantially faster than possible with subcutaneous administration of rGH.
  • FIG. 9 shows expression levels of growth hormone (GH) receptor mRNA in the liver of female B ALB/c mice treated with recombinant rat growth hormone (rGH) by subcutaneous injection (30 μg) or Delivery Construct 2 by oral gavage (100 μg). Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for GH receptor, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control. As shown in FIG. 9, administration of subcutaneous rGH resulted in a reduction in GH receptor mRNA expression levels in the liver at 60 minutes following administration to about 65% of those observed prior to administration. In contrast, Delivery Construct 2 caused such mRNA levels to decrease to about 15% of those observed prior to administration. Thus, these results confirm that oral administration of Delivery Construct 2 is effective to deliver rGH to the bloodstream of a subject, and further, that Delivery Construct 2 delivers significantly more active rGH to mouse liver than conventional subcutaneous administration of rGH as shown by the enhanced downregulation of GH receptor mRNA expression.
  • FIG. 10 shows expression levels of insulin-like growth factor I (IGF-I) mRNA in the liver of female BALB/c mice treated with recombinant rat growth hormone (rGH) by subcutaneous injection (30 μg) or Delivery Construct 2 by oral gavage (100 μg). Total RNA extracted from the liver was subjected to quantitative RT-PCR using primers specific for IGF-I, shown above. Values were normalized to glyceraldehyde-3 phosphate dehydrogenase (GAPDH) and expressed as % of control. As shown in FIG. 10, administration of either subcutaneous rGH or Delivery Construct 2 resulted in a reduction in IGF-1 mRNA expression levels in the liver at 30 minutes following administration to about 20% of those observed prior to administration. Thus, both subcutaneous rGH and orally-administered Delivery Construct 2 yielded the same effects, demonstrating that Delivery Construct 2 can deliver active rGH to the bloodstream of a
  • 6.9. Reduced Immunogenicity of Macromolecules Administered with a Delivery Construct
  • This example shows that an exemplary macromolecule, rGH, administered orally with Delivery Construct 2, is less immunogenic than rGH administered subcutaneously.
  • To assess the relative immunogenicity in mice for rGH administered a delivery construct relative to subcutaneous administration, the serum titer of anti-rGH IgG antibodies from oral administration of 3, 10, or 30 μg Delivery Construct 2 or 3 or 10 μg subcutaneous rGH was determined in an ELISA assay. To do so, 100 μl 1 ng/μl recombinant rGH diluted in coating buffer (0.2 M NaHCO3—Na2CO3, pH 9.4) was added to Costar EIA/RIA plates, then incubated at room temperature for 16-24 hours. Next, the plates were washed 4 times with 300 μl wash buffer (phosphate-buffered saline). The plates were then blocked with 200 μl blocking buffer (0.5% BSA in phosphate-buffered saline) and incubated at room temperature for 1 hour. Next, the plates were again washed four times with 300 μl wash buffer.
  • After preparation of the plates, 100 μl diluted samples, standard positive control, or assay buffer as negative control was added to the appropriate well and incubated for one hour. Mouse serum samples and positive control (anti-rGH IgG) were diluted 1:20 in assay buffer (0.5% BSA in phosphate-buffered saline). The plates were then washed four times with 300 μl wash buffer. Next, 100 μl secondary antibody (goat anti-mouse IgG conjugated to horseradish peroxidase, 0.4 mg/ml, Pierce #31430, diluted at 1:6000 in assay buffer and incubated at room temperature for one hour. Next, the plates were again washed four times with 300 μl wash buffer. Next, 100 μl 3,3′,5,5′-Tetramethylbenzidine substrate (Sigma) was added to each well and incubated for 2-10 minutes, depending on color development. 100 μl/well 1M H2SO4 was then added to stop the reaction and absorbance read at 450 nm. All assays were performed in triplicate and the results averaged.
  • Representative results of the ELISAs are shown in FIGS. 11A-D. The graphs presented in these figures demonstrate that 3, 10, and 30 μg Delivery Construct 2 administered orally elicited a lower titer of anti-rGH IgG antibodies than either 3 or 10 μg subcutaneous rGH. In particular, subcutaneous administration of 10 μg rGH caused a substantial anti-rGH IgG response in all eight mice, while the eight mice administered 3, 10, or 30 μg Delivery Construct 2 by oral gavage had minimal anti-rGH IgG responses. Further, these observations were consistent whether the sera were diluted 1:25 (FIGS. 11A and 11C) or 1:200 (FIGS. 11B and 11D). Finally, it should be noted that each mouse administered 3, 10, or 30 μg Delivery Construct 2 experienced a minimal immune response against rGH, as shown by the tight clustering of the data points in FIGS. 11C and D. Thus, these results demonstrate that oral administration of Delivery Construct 2 not only delivers more active rGH to the liver than possible with subcutaneous injection, but further, the active rGH is less immunogenic when administered orally with Delivery Construct 2 compared to subcutaneous administration.
  • 6.10. Exemplary Delivery Construct for Delivery of Human Growth Hormone
  • This example describes construction of an exemplary delivery construct for delivering human growth hormone, termed Delivery Construct 6. Techniques similar to those described in Example 6.1, above, were used to construct a plasmid used to express Delivery Construct 6. The nucleotide sequence of the portion of this plasmid that encodes the exemplary delivery construct is presented as FIG. 12, while the amino acid sequence of the delivery construct is presented as FIG. 13.
  • 6.11. Exemplary Delivery Construct for Delivering Inferferon Alpha
  • This example describes construction of an exemplary delivery construct for delivering IFNα (in this case, IFNα-2b), termed Delivery Construct 7. Techniques similar to those described in Example 6.1, above, were used to construct a plasmid used to express Delivery Construct 7. The nucleotide sequence of the portion of this plasmid that encodes the exemplary Delivery Construct 7 is presented as FIG. 14, while the amino acid sequence of Delivery Construct 7 is presented as FIG. 15.
  • 6.12. Delivery of Interferon Alpha in an In Vivo System
  • This example demonstrates the use of Delivery Construct 7 to deliver IFNα-2b to the bloodstream of a subject in a mouse model system.
  • Using an animal feeding needle, 100 μg of Deli very Construct 7 (in 250 μl total volume) was orally administered to female BALB/c mice, 5-6 weeks of age (Charles River Laboratories, Wilmington, Mass.). Delivery Construct 7 was diluted in 1 mg/ml bovine serum albumin (BSA) and phosphate buffered saline (PBS). At specific times after oral gavage and SC administration, mice were euthanized by CO2 asphyxiation and exsanguinated. Whole blood was collected and analyzed as described below.
  • ELISA assays were used to measure serum concentrations of IFNα-2b in one mouse immediately following administration and in three mice at 15, 30, and 75 minutes following administration. The ELISA assays were performed using R&D Systems Serum ELISA Kit No. 41110-1 according to the manufacturer's instructions.
  • ELISA results are reported as the averages of the triplicate OD (450 nm) value of each sample. The results of the ELISA assays are presented in FIG. 16. As shown in FIG. 16, IFNα-2b was detected at low (about 3 ng/ml) concentration 15 minutes after administration. 30 minutes following administration, serum concentration of IFNα-2b was about 43 ng/ml. 45 minutes following administration, serum concentration of IFNα-2b had fallen to about 13 ng/ml.
  • 6.13. Exemplary Delivery Construct for Delivering Proinsulin
  • This example describes construction of an exemplary delivery construct for delivering proinsulin, termed Delivery Construct 8. Techniques similar to those described in Example 6.1, above, are used to construct a plasmid used to express Delivery Construct 8. The amino acid sequence of Delivery Construct 8 is presented as FIG. 17.
  • 6.14. Exemplary Delivery Construct for Delivering Insulin
  • This example describes construction of an exemplary delivery construct for delivering insulin, termed Delivery Construct 9. Techniques similar to those described in Example 6.1, above, are used to construct a plasmid used to express Delivery Construct 9, with certain modifications.
  • In particular, the scheme used to express Delivery Construct 9 is modified because insulin comprises two separate amino acid chains. In this example, the B-chain of insulin is expressed together with the remainder of the Delivery Construct, constructed according to the general scheme presented in Example 6.1. Amino acids corresponding to the A-chain are made either synthetically (e.g., chemically synthesizing the A-chain peptide from amino acids) or recombinantly (e.g., expressed in a suitable recombinant system such as, for example, E. coli, yeast, etc.) The two polypeptides are then combined under conditions that permit association of the A-chain and B-chain. Then, disulfide bonds are made between the two chains of insulin as found in native insulin by application of mildly oxidizing conditions. The amino acid sequence of the two amino acid chains of Delivery Construct 9 is presented as FIG. 17.
  • The present invention provides, inter alia, delivery constructs and methods of inducing an immune response in a subject. While many specific examples have been provided, the above description is intended to illustrate rather than limit the invention. Many variations of the invention will become apparent to those skilled in the art upon review of this specification. The scope of the invention should, therefore, be determined not with reference to the above description, but instead should be determined with reference to the appended claims along with their full scope of equivalents.
  • All publications and patent documents cited in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication or patent document were so individually denoted. Citation of these documents is not an admission that any particular reference is “prior art” to this invention.
  • TABLE 7
    Human Peptidases by Class
    Aspartic-type peptidases
    BAE1_HUMAN (P56817)
    BAE2_HUMAN (Q9Y5Z0)
    CATD_HUMAN (P07339)
    CATE_HUMAN (P14091)
    NAP1_HUMAN (O96009)
    PEPA_HUMAN (P00790
    PEPC_HUMAN (P20142)
    RENI_HUMAN (P00797)
    VPRT_HUMAN (P10265)
    Other Peptidases
    FAC2_HUMAN (Q9Y256)
    Cysteine-type peptidases
    BLMH_HUMAN (Q13867)
    CATB_HUMAN (P07858)
    CATC_HUMAN (P53634)
    CATF_HUMAN (Q9UBX1)
    CATH_HUMAN (P09668)
    CATK_HUMAN (P43235)
    CATL_HUMAN (P07711)
    CATO_HUMAN (P43234)
    CATS_HUMAN (P25774)
    CATW_HUMAN (P56202)
    CATZ_HUMAN (Q9UBR2)
    CSL2_HUMAN (O60911)
    TNAG_HUMAN (Q9UJW2)
    CAN1_HUMAN (P07384)
    CAN2_HUMAN (P17655)
    CAN3_HUMAN (P20807)
    CAN5_HUMAN (O15484)
    CAN6_HUMAN (Q9Y6Q1)
    CAN7_HUMAN (Q9Y6W3)
    CAN9_HUMAN (O14815)
    CANA_HUMAN (Q9HC96)
    CANB_HUMAN (Q9UMQ6)
    UBL1_HUMAN (P09936)
    UBL3_HUMAN (P15374)
    UBL5_HUMAN (Q9Y5K5)
    GPI8_HUMAN (Q92643)
    LGMN_HUMAN (Q99538)
    CFLA_HUMAN (O15519)
    I1BC_HUMAN (P29466)
    ICE2_HUMAN (P42575)
    ICE3_HUMAN (P42574)
    ICE4_HUMAN (P49662)
    ICE5_HUMAN (P51878)
    ICE6_HUMAN (P55212)
    ICE7_HUMAN (P55210)
    ICE8_HUMAN (Q14790)
    ICE9_HUMAN (P55211)
    ICEA_HUMAN (Q92851)
    ICEE_HUMAN (P31944)
    MLT1_HUMAN (Q9UDY8)
    PGPI_HUMAN (Q9NXJ5)
    FAFX_HUMAN (Q93008)
    FAFY_HUMAN (O00507)
    UB10_HUMAN (Q14694)
    UB11_HUMAN (P51784)
    UB12_HUMAN (O75317)
    UB13_HUMAN (Q92995)
    UB14_HUMAN (P54578)
    UB15_HUMAN (Q9Y4E8)
    UB16_HUMAN (Q9Y5T5)
    UB18_HUMAN (Q9UMW8)
    UB19_HUMAN (O94966)
    UB20_HUMAN (Q9Y2K6)
    UB21_HUMAN (Q9UK80)
    UB22_HUMAN (Q9UPT9)
    UB24_HUMAN (Q9UPU5)
    UB25_HUMAN (Q9UHP3)
    UB26_HUMAN (Q9BXU7)
    UB28_HUMAN (Q96RU2)
    UB29_HUMAN (Q9HBJ7)
    UB32_HUMAN (Q8NFA0)
    UB33_HUMAN (Q8TEY7)
    UB35_HUMAN (Q9P2H5)
    UB36_HUMAN (Q9P275)
    UB37_HUMAN (Q86T82)
    UB38_HUMAN (Q8NB14)
    UB40_HUMAN (Q9NVE5)
    UB42_HUMAN (Q9H9J4)
    UB44_HUMAN (Q9H0E7)
    UB46_HUMAN (P62068)
    UBP1_HUMAN (O94782)
    UBP2_HUMAN (O75604)
    UBP3_HUMAN (Q9Y6I4)
    UBP4_HUMAN (Q13107)
    UBP5_HUMAN (P45974)
    UBP6_HUMAN (P35125)
    UBP7_HUMAN (Q93009)
    UBP8_HUMAN (P40818)
    GGH_HUMAN (Q92820)
    SEN1_HUMAN (Q9P0U3)
    SEN3_HUMAN (Q9H4L4)
    SEN5_HUMAN (Q96HI0)
    SEN6_HUMAN (Q9GZR1)
    SEN7_HUMAN (Q9BQF6)
    SEN8_HUMAN (Q96LD8)
    SNP2_HUMAN (Q9HC62)
    ESP1_HUMAN (Q14674)
    Metallopeptidases
    AMPB_HUMAN (Q9H4A4)
    AMPE_HUMAN (Q07075)
    AMPN_HUMAN (P15144)
    ART1_HUMAN (Q9NZ08)
    LCAP_HUMAN (Q9UIQ6)
    LKHA_HUMAN (P09960)
    PSA_HUMAN (P55786)
    RNPL_HUMAN (Q9HAU8)
    THDE_HUMAN (Q9UKU6)
    ACET_HUMAN (P22966)
    ACE_HUMAN (P12821)
    MEPD_HUMAN (P52888)
    NEUL_HUMAN (Q9BYT8)
    PMIP_HUMAN (Q99797)
    MM01_HUMAN (P03956)
    MM02_HUMAN (P08253)
    MM03_HUMAN (P08254)
    MM07_HUMAN (P09237)
    MM08_HUMAN (P22894)
    MM09_HUMAN (P14780)
    MM10_HUMAN (P09238)
    MM11_HUMAN (P24347)
    MM12_HUMAN (P39900)
    MM13_HUMAN (P45452)
    MM14_HUMAN (P50281)
    MM15_HUMAN (P51511)
    MM16_HUMAN (P51512)
    MM17_HUMAN (Q9ULZ9)
    MM19_HUMAN (Q99542)
    MM20_HUMAN (O60882)
    MM21_HUMAN (Q8N119)
    MM24_HUMAN (Q9Y5R2)
    MM25_HUMAN (Q9NPA2)
    MM26_HUMAN (Q9NRE1)
    MM28_HUMAN (Q9H239)
    BMP1_HUMAN (P13497)
    MEPA_HUMAN (Q16819)
    MEPB_HUMAN (Q16820)
    AD02_HUMAN (Q99965)
    AD07_HUMAN (Q9H2U9)
    AD08_HUMAN (P78325)
    AD09_HUMAN (Q13443)
    AD10_HUMAN (O14672)
    AD11_HUMAN (O75078)
    AD12_HUMAN (O43184)
    AD15_HUMAN (Q13444)
    AD17_HUMAN (P78536)
    AD18_HUMAN (Q9Y3Q7)
    AD19_HUMAN (Q9H013)
    AD20_HUMAN (O43506)
    AD21_HUMAN (Q9UKJ8)
    AD22_HUMAN (Q9P0K1)
    AD28_HUMAN (Q9UKQ2)
    AD29_HUMAN (Q9UKF5)
    AD30_HUMAN (Q9UKF2)
    AD33_HUMAN (Q9BZ11)
    AT10_HUMAN (Q9H324)
    AT12_HUMAN (P58397)
    AT14_HUMAN (Q8WXS8)
    AT15_HUMAN (Q8TE58)
    AT16_HUMAN (Q8TE57)
    AT17_HUMAN (Q8TE56)
    AT18_HUMAN (Q8TE60)
    AT19_HUMAN (Q8TE59)
    AT20_HUMAN (P59510)
    ATS1_HUMAN (Q9UHI8)
    ATS2_HUMAN (O95450)
    ATS3_HUMAN (O15072)
    ATS4_HUMAN (O75173)
    ATS5_HUMAN (Q9UNA0)
    ATS6_HUMAN (Q9UKP5)
    ATS7_HUMAN (Q9UKP4)
    ATS8_HUMAN (Q9UP79)
    ATS9_HUMAN (Q9P2N4)
    ECE1_HUMAN (P42892)
    ECE2_HUMAN (O60344)
    ECEL_HUMAN (O95672)
    KELL_HUMAN (P23276)
    NEP_HUMAN (P08473)
    PEX_HUMAN (P78562)
    CBP1_HUMAN (P15085)
    CBP2_HUMAN (P48052)
    CBP4_HUMAN (Q9UI42)
    CBP5_HUMAN (Q8WXQ8)
    CBP6_HUMAN (Q8N4T0)
    CBPB_HUMAN (P15086)
    CBPC_HUMAN (P15088)
    CBPD_HUMAN (O75976)
    CBPE_HUMAN (P16870)
    CBPM_HUMAN (P14384)
    CBPN_HUMAN (P15169)
    CPX2_HUMAN (Q8N436)
    CPXM_HUMAN (Q96SM3)
    IDE_HUMAN (P14735)
    MPPA_HUMAN (Q10713)
    MPPB_HUMAN (O75439)
    NRDC_HUMAN (O43847)
    UCR1_HUMAN (P31930)
    UCR2_HUMAN (P22695)
    AMPL_HUMAN (P28838)
    PEL1_HUMAN (Q8NDH3)
    DNPE_HUMAN (Q9ULA0)
    MDP1_HUMAN (P16444)
    CGL1_HUMAN (Q96KP4)
    CGL2_HUMAN (Q96KN2)
    ACY1_HUMAN (Q03154)
    GCP_HUMAN (Q9NPF4)
    AMP1_HUMAN (P53582)
    PEPD_HUMAN (P12955)
    XPP2_HUMAN (O43895)
    AMP2_HUMAN (P50579)
    P2G4_HUMAN (Q9UQ80)
    FOH1_HUMAN (Q04609)
    NLD2_HUMAN (Q9Y3Q0)
    NLDL_HUMAN (Q9UQQ1)
    TFR1_HUMAN (P02786)
    TFR2_HUMAN (Q9UP52)
    AF31_HUMAN (O43931)
    AF32_HUMAN (Q9Y4W6)
    SPG7_HUMAN (Q9UQ90)
    YME1_HUMAN (Q96TA2)
    PAPA_HUMAN (Q13219)
    FAC1_HUMAN (O75844)
    DPP3_HUMAN (Q9NY33)
    MS2P_HUMAN (O43462)
    Serine-type peptidases
    ACRL_HUMAN (P58840)
    ACRO_HUMAN (P10323)
    APOA_HUMAN (P08519)
    BSS4_HUMAN (Q9GZN4)
    C1R_HUMAN (P00736)
    C1S_HUMAN (P09871)
    CAP7_HUMAN (P20160)
    CATG_HUMAN (P08311)
    CFAB_HUMAN (P00751)
    CFAD_HUMAN (P00746)
    CFAI_HUMAN (P05156)
    CLCR_HUMAN (Q99895)
    CO2_HUMAN (P06681)
    CORI_HUMAN (Q9Y5Q5)
    CRAR_HUMAN (P48740)
    CTRB_HUMAN (P17538)
    CTRL_HUMAN (P40313)
    DES1_HUMAN (Q9UL52)
    EL1_HUMAN (Q9UNI1)
    EL2A_HUMAN (P08217)
    EL2B_HUMAN (P08218)
    EL3A_HUMAN (P09093)
    EL3B_HUMAN (P08861)
    ELNE_HUMAN (P08246)
    ENTK_HUMAN (P98073)
    FA10_HUMAN (P00742)
    FA11_HUMAN (P03951)
    FA12_HUMAN (P00748)
    FA7_HUMAN (P08709)
    FA9_HUMAN (P00740)
    GRAA_HUMAN (P12544)
    GRAB_HUMAN (P10144)
    GRAH_HUMAN (P20718)
    GRAK_HUMAN (P49863)
    GRAM_HUMAN (P51124)
    HATT_HUMAN (O60235)
    HEPS_HUMAN (P05981)
    HGFA_HUMAN (Q04756)
    HGFL_HUMAN (P26927)
    HGF_HUMAN (P14210)
    HPTR_HUMAN (P00739)
    HPT_HUMAN (P00738)
    KAL_HUMAN (P03952)
    KLK1_HUMAN (P06870)
    KLK2_HUMAN (P20151)
    KLK3_HUMAN (P07288)
    KLK4_HUMAN (Q9Y5K2)
    KLK5_HUMAN (Q9Y337)
    KLK6_HUMAN (Q92876)
    KLK7_HUMAN (P49862)
    KLK8_HUMAN (O60259)
    KLK9_HUMAN (Q9UKQ9)
    KLKA_HUMAN (O43240)
    KLKB_HUMAN (Q9UBX7)
    KLKC_HUMAN (Q9UKR0)
    KLKD_HUMAN (Q9UKR3)
    KLKE_HUMAN (Q9P0G3)
    KLKF_HUMAN (Q9H2R5)
    LCLP_HUMAN (P34168)
    MAS2_HUMAN (O00187)
    MCT1_HUMAN (P23946)
    NETR_HUMAN (P56730)
    PLMN_HUMAN (P00747)
    PR27_HUMAN (Q9BQR3)
    PRN3_HUMAN (P24158)
    PRTC_HUMAN (P04070)
    PRTZ_HUMAN (P22891)
    PS23_HUMAN (O95084)
    PSS8_HUMAN (Q16651)
    ST14_HUMAN (Q9Y5Y6)
    TEST_HUMAN (Q9Y6M0)
    THRB_HUMAN (P00734)
    TMS2_HUMAN (O15393)
    TMS3_HUMAN (P57727)
    TMS4_HUMAN (Q9NRS4)
    TMS5_HUMAN (Q9H3S3)
    TMS6_HUMAN (Q8IU80)
    TPA_HUMAN (P00750)
    TRB1_HUMAN (Q15661)
    TRB2_HUMAN (P20231)
    TRY1_HUMAN (P07477)
    TRY2_HUMAN (P07478)
    TRY3_HUMAN (P35030)
    TRYA_HUMAN (P15157)
    TRYD_HUMAN (Q9BZJ3)
    TRYG_HUMAN (Q9NRR2)
    TS50_HUMAN (Q9UI38)
    UROK_HUMAN (P00749)
    HRA1_HUMAN (Q92743)
    HRA2_HUMAN (O43464)
    HRA3_HUMAN (P83110)
    HRA4_HUMAN (P83105)
    FURI_HUMAN (P09958)
    MS1P_HUMAN (Q14703)
    NEC1_HUMAN (P29120)
    NEC2_HUMAN (P16519)
    PCK5_HUMAN (Q92824)
    PCK6_HUMAN (P29122)
    PCK7_HUMAN (Q16549)
    PCK9_HUMAN (Q8NBP7)
    TPP2_HUMAN (P29144)
    PPCE_HUMAN (P48147)
    DPP4_HUMAN (P27487)
    DPP6_HUMAN (P42658)
    SEPR_HUMAN (Q12884)
    ACPH_HUMAN (P13798)
    CPVL_HUMAN (Q9H3G5)
    PRTP_HUMAN (P10619)
    RISC_HUMAN (Q9HB40)
    CLPP_HUMAN (Q16740)
    LONM_HUMAN (P36776)
    SPC3_HUMAN (Q9BY50)
    SPC4_HUMAN (P21378)
    DPP2_HUMAN (Q9UHL4)
    PCP_HUMAN (P42785)
    TSSP_HUMAN (Q9NQE7)
    HYEP_HUMAN (P07099)
    TPP1_HUMAN (O14773)
    RHB1_HUMAN (O75783)
    RHB2_HUMAN (Q9NX52)
    RHB4_HUMAN (P58872)
    Threonine-type peptidases
    PS7L_HUMAN (Q8TAA3)
    PSA1_HUMAN (P25786)
    PSA2_HUMAN (P25787)
    PSA3_HUMAN (P25788)
    PSA4_HUMAN (P25789)
    PSA5_HUMAN (P28066)
    PSA6_HUMAN (P60900)
    PSA7_HUMAN (O14818)
    PSB1_HUMAN (P20618)
    PSB2_HUMAN (P49721)
    PSB3_HUMAN (P49720)
    PSB4_HUMAN (P28070)
    PSB5_HUMAN (P28074)
    PSB6_HUMAN (P28072)
    PSB7_HUMAN (Q99436)
    PSB8_HUMAN (P28062)
    PSB9_HUMAN (P28065)
    PSBA_HUMAN (P40306)

Claims (68)

1. A delivery construct, comprising:
a)- a receptor binding domain,
b)- a transcytosis domain,
c)- a macromolecule to be delivered to a subject, and
d)- a cleavable linker,
wherein cleavage at said cleavable linker separates said macromolecule from the remainder of said construct, and wherein said cleavable linker is cleavable by an enzyme that i) exhibits greater activity at a basal-lateral membrane of a polarized epithelial cell of said subject than at an apical membrane of the polarized epithelial cell, or ii) exhibits greater activity in the plasma of said subject than at an apical membrane of the polarized epithelial cell of the subject.
2. The delivery construct of claim 1, further comprising a second cleavable linker.
3. The delivery construct of claim 1, wherein said cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.:10).
4. The delivery construct of claim 1, wherein said enzyme that is present at a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
5. The delivery construct of claim 1, wherein said receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, botulinum toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF α; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8.
6. The delivery construct of claim 1, wherein said receptor binding domain binds to a cell-surface receptor that is selected from the group consisting of α2-macroglobulin receptor, epidermal growth factor receptor, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNFα receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
7. The delivery construct of claim 5, wherein said receptor binding domain of Pseudomonas exotoxin A is Domain Ia of Pseudomonas exotoxin A.
8. The delivery construct of claim 7, wherein said receptor binding domain of Pseudomonas exotoxin A has an amino acid sequence that is SEQ ID NO.:1.
9. The delivery construct of claim 1, wherein said transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, botulinum toxin, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin.
10. The delivery construct of claim 9, wherein said transcytosis domain is Pseudomonas exotoxin A transcytosis domain.
11. The delivery construct of claim 10, wherein said Pseudomonas exotoxin A transcytosis domain has an amino acid sequence that is SEQ ID NO.:2.
12. The delivery construct of claim 1, wherein said macromolecule is selected from the group of a nucleic acid, a peptide, a polypeptide, a protein, and a lipid.
13. The delivery construct of claim 12, wherein said polypeptide is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors.
14. The delivery construct of claim 13, wherein said polypeptide is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN-α, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, proinsulin, insulin, glucocorticoid, amylin, adrenocorticototropin, enkephalin, and glucagon-like peptide 1.
15. The delivery construct of claim 14, wherein said polypeptide is human growth hormone.
16. The delivery construct of claim 12, wherein said protein is human insulin.
17. The delivery construct of claim 12, wherein said protein is human IFN-α.
18. The delivery construct of claim 12, wherein said protein is human IFN-α2b.
19. The delivery construct of claim 12, wherein said protein is human proinsulin.
20. The delivery construct of claim 12, further comprising a second macromolecule that is selected from the group of a nucleic acid, a peptide, a polypeptide, a protein, a lipid, or a small organic molecule and a second cleavable linker, wherein cleavage at said second cleavable linker separates said second macromolecule from the remainder of said construct.
21. The delivery construct of claim 17, wherein said macromolecule is a first polypeptide and said second macromolecule is a second polypeptide.
22. The delivery construct of claim 21, wherein said first polypeptide and said second polypeptide associate to form a multimer.
23. The delivery construct of claim 22, wherein said multimer is a dimer, tetramer, or octamer.
24. The delivery construct of claim 23, wherein said dimer is an antibody.
25. A polynucleotide that encodes a delivery construct, said delivery construct comprising:
a)- a receptor binding domain,
b)- a transcytosis domain,
c)- a macromolecule to be delivered to a subject, and
d)- a cleavable linker,
wherein cleavage at said cleavable linker separates said macromolecule from the remainder of said construct, and wherein said cleavable linker is cleavable by an enzyme that i) exhibits greater activity at a basal-lateral membrane of a polarized epithelial cell of said subject than at an apical membrane of the polarized epithelial cell, or ii) exhibits greater activity in the plasma of said subject than at an apical membrane of the polarized epithelial cell of the subject.
26. A polynucleotide that hybridizes under stringent hybridization conditions to the polynucleotide of claim 25.
27. The polynucleotide of claim 25, wherein said delivery construct further comprises a second cleavable linker.
28. The polynucleotide of claim 25, wherein said cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ED NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.:10).
29. The polynucleotide of claim 25, wherein said enzyme that is present at a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
30. The polynucleotide of claim 25, wherein said receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, botulinum toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF α; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8.
31. The polynucleotide of claim 25, wherein said receptor binding domain binds to a cell-surface receptor that is selected from the group consisting of α2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNFα receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
32. The polynucleotide of claim 30, wherein said receptor binding domain of Pseudomonas exotoxin A is Domain Ia of Pseudomonas exotoxin A.
33. The polynucleotide of claim 31, wherein said receptor binding domain of Pseudomonas exotoxin A has an amino acid sequence that is SEQ ID NO.:1.
34. The polynucleotide of claim 25, wherein said transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, botulinum toxin, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin.
35. The polynucleotide of claim 34, wherein said transcytosis domain is Pseudomonas exotoxin A transcytosis domain.
36. The polynucleotide of claim 35, wherein said Pseudomonas exotoxin A transcytosis domain has an amino acid sequence that is SEQ ID NO.: 2.
37. The polynucleotide of claim 25, wherein said macromolecule is selected from the group of a peptide, a polypeptide, and a protein.
38. The polynucleotide of claim 37, wherein said polypeptide is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors.
39. The polynucleotide of claim 38, wherein said polypeptide is selected from the group consisting of IGF-I, IGF-II, IGF-1H, EGF, IFN-α, IFN-α, 2b, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, proinsulin, insulin, glucocorticoid, amylin, adrenocorticototropin, enkephalin, and glucagon-like peptide 1.
40. The polynucleotide of claim 39, wherein said polypeptide is human growth hormone.
41. The polynucleotide of claim 39, wherein said protein is human insulin.
42. A polynucleotide that encodes a delivery construct, said polynucleotide comprising:
a)- a nucleic acid sequence encoding a receptor binding domain,
b)- a nucleic acid sequence encoding a transcytosis domain,
c)- a nucleic acid sequence encoding a cleavable linker, and
d)- a nucleic acid sequence comprising a polylinker insertion site,
wherein cleavage at said cleavable linker separates said macromolecule from the remainder of said construct, and wherein said cleavable linker is cleavable by an enzyme that i) exhibits greater activity at a basal-lateral membrane of a polarized epithelial cell of said subject than at an apical membrane of the polarized epithelial cell, or ii) exhibits greater activity in the plasma of said subject than at an apical membrane of the polarized epithelial cell of the subject.
43. An expression vector comprising the polynucleotide of claim 25.
44. A cell comprising the expression vector of claim 43.
45. A composition comprising a delivery construct, said delivery construct comprising:
a)- a receptor binding domain,
b)- a transcytosis domain,
c)- a macromolecule to be delivered to a subject, and
d)- a cleavable linker,
wherein cleavage at said cleavable linker separates said macromolecule from the remainder of said construct, and wherein said cleavable linker is cleavable by an enzyme that i) exhibits greater activity at a basal-lateral membrane of a polarized epithelial cell of said subject than at an apical membrane of the polarized epithelial cell, or ii) exhibits greater activity in the plasma of said subject than at an apical membrane of the polarized epithelial cell of the subject.
46. The composition of claim 45, wherein said composition further comprises a pharmaceutically acceptable diluent, excipient, vehicle, or carrier.
47. The composition of claim 45, wherein said composition is formulated for nasal or oral administration.
48. A method for delivering a macromolecule to a subject, comprising contacting an apical surface of a polarized epithelial cell of the subject with a delivery construct, wherein said delivery construct comprises a receptor binding domain, a transcytosis domain, a cleavable linker, and the macromolecule, wherein the transcytosis domain transcytosis the macromolecule to and through the basal-lateral membrane of said epithelial cell, wherein cleavage at said cleavable linker separates said macromolecule from the remainder of said construct, and wherein said cleavable linker is cleavable by an enzyme that i) exhibits greater activity at a basal-lateral membrane of a polarized epithelial cell of said subject than at an apical membrane of the polarized epithelial cell, or ii) exhibits greater activity in the plasma of said subject than at an apical membrane of the polarized epithelial cell of the subject, and wherein cleavage at the cleavable linker separates the macromolecule from the remainder of the delivery construct, thereby delivering the macromolecule to the subject.
49. The method of claim 48, wherein said receptor binding domain is selected from the group consisting of receptor binding domains from Pseudomonas exotoxin A, cholera toxin, diptheria toxin, shiga toxin, or shiga-like toxin; monoclonal antibodies; polyclonal antibodies; single-chain antibodies; TGF α; EGF; IGF-I; IGF-II; IGF-III; IL-1; IL-2; IL-3; IL-6; MIP-1a; MIP-1b; MCAF; and IL-8.
50. The method of claim 48, wherein said receptor binding domain binds to a cell surface receptor selected from the group consisting of α2-macroglobulin receptor, EGFR, IGFR, transferrin receptor, chemokine receptor, CD25, CD11B, CD11C, CD80, CD86, TNFα receptor, TOLL receptor, M-CSF receptor, GM-CSF receptor, scavenger receptor, and VEGF receptor.
51. The method of claim 48, wherein said transcytosis domain is selected from the group consisting of transcytosis domains from Pseudomonas exotoxin A, botulinum toxin, diptheria toxin, pertussis toxin, cholera toxin, heat-labile E. coli enterotoxin, shiga toxin, and shiga-like toxin.
52. The method of claim 48, wherein said macromolecule is selected from the group consisting of a peptide, a polypeptide, a protein, a nucleic acid, and a lipid.
53. The method of claim 48, wherein said enzyme that is present at a basal-lateral membrane of a polarized epithelial cell is selected from the group consisting of Cathepsin GI, Chymotrypsin I, Elastase I, Subtilisin AI, Subtilisin AII, Thrombin I, and Urokinase I.
54. The method of claim 48, wherein said cleavable linker comprises an amino acid sequence that is selected from the group consisting of Ala-Ala-Pro-Phe (SEQ ID NO.:4), Gly-Gly-Phe (SEQ ID NO.:5), Ala-Ala-Pro-Val (SEQ ID NO.:6), Gly-Gly-Leu (SEQ ID NO.:7), Ala-Ala-Leu (SEQ ID NO.:8), Phe-Val-Arg (SEQ ID NO.:9), Val-Gly-Arg (SEQ ID NO.: 10).
55. The method of claim 48, wherein the epithelial cell is selected from the group consisting of nasal epithelial cells, oral epithelial cells, intestinal epithelial cells, rectal epithelial cells, vaginal epithelial cells, and pulmonary epithelial cells.
56. The method of claim 48, wherein said mammal is a human.
57. The method of claim 48, wherein said delivery construct contacts the apical membrane of the epithelial cell.
58. A method for delivering a macromolecule to the bloodstream of a subject, comprising contacting the delivery construct of claim 1 to an apical surface of a polarized epithelial cell of the subject, such that the macromolecule is delivered to the bloodstream of the subject, wherein a lower titer of antibodies specific for the macromolecule is induced in the serum of the subject than is induced by subcutaneously administering the macromolecule to a subject separately from the remainder of the delivery construct.
59. The method of claim 58, wherein the macromolecule is selected from the group consisting of a peptide, a polypeptide, a protein, a nucleic acid, and a lipid.
60. The method of claim 58, wherein the macromolecule is selected from the group consisting of polypeptide hormones, cytokines, chemokines, growth factors, and clotting factors.
61. The method of claim 58, wherein the macromolecule is selected from the group consisting of IGF-I, IGF-II, IGF-III, EGF, IFN-α, IFN-β, IFN-γ, G-CSF, GM-CSF, IL-1, IL-2, IL-3, IL-6, IL-8, IL-12, EPO, growth hormone, factor VII, vasopressin, calcitonin, parathyroid hormone, luteinizing hormone-releasing factor, tissue plasminogen activators, proinsulin, insulin, glucocorticoid, amylin, adrenocorticototropin, enkephalin, and glucagon-like peptide 1.
62. The method of claim 58, wherein the macromolecule is human growth hormone.
63. The method of claim 58, wherein the macromolecule is human insulin.
64. The method of claim 58, wherein the subject is a mouse, dog, goat, or human.
65. The method of claim 58, wherein the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by with the delivery construct is less than about 75% of the titer of antibodies induced by subcutaneously administering the macromolecule to a subject separately from the remainder of the delivery construct.
66. The method of claim 58, wherein the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 50% of the titer of antibodies induced by subcutaneously administering the macromolecule to a subject separately from the remainder of the delivery construct.
67. The method of claim 58, wherein the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 25% of the titer of antibodies induced by subcutaneously administering the macromolecule to a subject separately from the remainder of the delivery construct.
68. The method of claim 58, wherein the titer of antibodies specific for the macromolecule induced in the serum of the subject by the macromolecule delivered by the delivery construct is less than about 10% of the titer of antibodies induced by subcutaneously administering the macromolecule to a subject separately from the remainder of the delivery construct.
US11/664,787 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules Abandoned US20090285771A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/664,787 US20090285771A1 (en) 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US61597004P 2004-10-04 2004-10-04
US68448405P 2005-05-24 2005-05-24
US71890705P 2005-09-19 2005-09-19
US11/664,787 US20090285771A1 (en) 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules
PCT/US2005/035803 WO2006044205A2 (en) 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules

Publications (1)

Publication Number Publication Date
US20090285771A1 true US20090285771A1 (en) 2009-11-19

Family

ID=36203399

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/664,787 Abandoned US20090285771A1 (en) 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules
US11/244,349 Expired - Fee Related US7713737B2 (en) 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/244,349 Expired - Fee Related US7713737B2 (en) 2004-10-04 2005-10-04 Methods and compositions for needleless delivery of macromolecules

Country Status (7)

Country Link
US (2) US20090285771A1 (en)
EP (1) EP1804832A4 (en)
JP (1) JP2008515808A (en)
KR (1) KR20070073863A (en)
AU (1) AU2005296064A1 (en)
CA (1) CA2583202A1 (en)
WO (1) WO2006044205A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11324833B2 (en) 2018-11-07 2022-05-10 Applied Molecular Transport Inc. Cholix-derived carriers for oral delivery of heterologous payload
US11426466B2 (en) 2018-03-08 2022-08-30 Applied Molecular Transport Inc. Toxin-derived delivery constructs for pulmonary delivery

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003080646A2 (en) * 2002-03-04 2003-10-02 Medical College Of Ohio Modified plasminogen activator inhibitor type-1 and methods based thereon
JP2008515808A (en) * 2004-10-04 2008-05-15 トリニティ バイオシステムズ インコーポレーテッド Methods and compositions for needle-free polymer delivery
US20090155297A1 (en) * 2004-10-04 2009-06-18 Trinity Biosystems, Inc. Methods and Compositions for Inducing an Immune Response Against Multiple Antigens
EP1812054A4 (en) * 2004-10-04 2010-01-13 Trinity Biosystems Inc Methods and compositions for immunizing against pseudomonas infection
DE102005019302A1 (en) * 2005-04-26 2006-11-16 Toxogen Gmbh Carrier for targeting nerve cells
US20090304684A1 (en) * 2005-12-05 2009-12-10 Trinity Biosystems, Inc. Methods and compositions for needleless delivery of antibodies
EP1971367A4 (en) * 2005-12-05 2010-04-07 Trinity Biosystems Inc Methods and compositions for needleless delivery of binding partners
EP2010205A2 (en) * 2006-03-16 2009-01-07 Trinity Biosystems, Inc. Methods for increasing the size of animals using needleless delivery constructs
US20090092660A1 (en) * 2006-08-09 2009-04-09 Trinity Biosystems, Inc. Methods and compositions for needleless delivery of particles
CA2722754A1 (en) * 2007-04-27 2009-01-29 The University Of Toledo Modified plasminogen activator inhibitor type-1 molecule and methods based thereon
PL2197903T3 (en) * 2007-09-04 2015-03-31 Us Gov Health & Human Services Deletions in domain ii of pseudomonas exotoxin a that reduce non-specific toxicity
WO2009045543A1 (en) * 2007-10-04 2009-04-09 The Regents Of The University Of California Treatment of conditions related to shock
WO2009131435A1 (en) * 2008-04-23 2009-10-29 Erasmus University Medical Center Rotterdam Linker containing bungarotoxin and a binding peptide
US11246915B2 (en) 2010-09-15 2022-02-15 Applied Molecular Transport Inc. Cholix toxin-derived fusion molecules for oral delivery of biologically active cargo
CN103249401B (en) * 2010-09-15 2016-01-20 兰德尔·J·米斯尼 The transit sequence using bacteriotoxin to derive sends the system and method for bioactivator
JP2013538858A (en) 2010-10-02 2013-10-17 ザ レジェンツ オブ ザ ユニヴァースティ オブ カリフォルニア Minimizing bowel dysfunction
LT2646470T (en) 2010-11-30 2017-06-12 F. Hoffmann-La Roche Ag Low affinity anti-transferrin receptor antibodies and their use to transfer therapeutic scfv across the blood brain barrier
DK3139965T3 (en) 2014-05-07 2021-11-22 Applied Molecular Transport Inc FUSION MOLECULES DERIVED FROM CHOLIX TOXIN FOR ORAL ADMINISTRATION OF BIOLOGICALLY ACTIVE SUBSTANCES
MX2021005346A (en) 2018-11-07 2021-10-13 Applied Molecular Transport Inc Delivery constructs for transcytosis and related methods.
KR20220066056A (en) 2019-08-16 2022-05-23 어플라이드 몰레큘라 트랜스포트 인크. Compositions, Formulations, and Interleukin Preparation and Tablets
WO2022241167A1 (en) * 2021-05-12 2022-11-17 Applied Molecular Transport Inc. Delivery constructs derived from bacterial toxins and uses thereof

Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5328984A (en) * 1991-03-04 1994-07-12 The United States As Represented By The Department Of Health & Human Services Recombinant chimeric proteins deliverable across cellular membranes into cytosol of target cells
US5374620A (en) * 1990-06-07 1994-12-20 Genentech, Inc. Growth-promoting composition and its use
US5458878A (en) * 1990-01-02 1995-10-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services P. exotoxin fusio proteins have COOHG220101al alterations which increase cytotoxicity
US5512658A (en) * 1990-05-11 1996-04-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pseudomonas exotoxins (PE) and conjugates thereof having lower animal toxicity with high cytocidal activity through substitution of positively charged amino acids
US5541287A (en) * 1992-06-09 1996-07-30 Neorx Corporation Pretargeting methods and compounds
US5602095A (en) * 1992-06-18 1997-02-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Recombinant pseudomonas exotoxin with increased activity
US5668255A (en) * 1984-06-07 1997-09-16 Seragen, Inc. Hybrid molecules having translocation region and cell-binding region
US5696077A (en) * 1992-06-23 1997-12-09 Associated Synapse Biologics Pharmaceutical composition containing botulinum B complex
US5696237A (en) * 1986-09-24 1997-12-09 The United States Of America As Represented By The Department Of Health And Human Services Recombinant antibody-toxin fusion protein
US5863745A (en) * 1986-09-24 1999-01-26 The United States Of America As Represented By The Department Of Health And Human Services Recombinant antibody-toxin fusion protein
US6022950A (en) * 1984-06-07 2000-02-08 Seragen, Inc. Hybrid molecules having translocation region and cell-binding region
US6022966A (en) * 1993-11-22 2000-02-08 Neorx Corporation Pretargeting methods and compounds
US6086900A (en) * 1997-03-26 2000-07-11 Board Of Regents, The University Of Texas Systems Methods and compositions for using membrane-penetrating proteins to carry materials across cell membranes
US6251392B1 (en) * 1997-10-20 2001-06-26 Epicyte Pharmaceuticals, Inc. Epithelial cell targeting agent
US6391280B1 (en) * 1997-01-10 2002-05-21 Epicyte Pharmaceutical, Inc. J chain polypeptide targeting molecule linked to an imaging agent
US20020106370A1 (en) * 1994-05-13 2002-08-08 Donald Leonard Nicholas Cardy Improvements in or relating to peptide delivery
US6485726B1 (en) * 1995-01-17 2002-11-26 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US6488926B1 (en) * 1993-07-30 2002-12-03 Medeva Holdings B.V. Vaccine compositions
US20030054012A1 (en) * 2000-05-12 2003-03-20 Fitzgerald David J. Pseudomonas exotoxin a-like chimeric immunogens for eliciting a secretory iga-mediated immune response
US20040001801A1 (en) * 2002-05-23 2004-01-01 Corvas International, Inc. Conjugates activated by cell surface proteases and therapeutic uses thereof
US6673574B2 (en) * 2000-11-30 2004-01-06 Unigene Laboratories Inc. Oral delivery of peptides using enzyme-cleavable membrane translocators
US20040071731A1 (en) * 2000-12-21 2004-04-15 Fitzgerald David J. Chimeric protein comprising non-toxic pseudomonas exotoxin a and type iv pilin sequences
US20040209330A1 (en) * 2003-03-24 2004-10-21 Wenfeng Xu Anti-IL-22RA antibodies and binding partners and methods of using in inflammation
US20040228831A1 (en) * 2003-05-15 2004-11-18 Belinka Benjamin A. Polymeric conjugates for tissue activated drug delivery
US20060104993A1 (en) * 2004-10-04 2006-05-18 Mrsny Randall J Methods and compositions for immunizing against Pseudomonas infection
US20060110409A1 (en) * 2002-07-19 2006-05-25 Shone Clifford C Targeted agents for nerve regeneration
US20060153798A1 (en) * 2004-10-04 2006-07-13 Mrsny Randall J Methods and compositions for needleless delivery of macromolecules
US20070141070A1 (en) * 2005-12-05 2007-06-21 Trinity Biosystems, Inc. Methods and compositions for needleless delivery of antibodies
US20070148131A1 (en) * 2005-12-05 2007-06-28 Trinity Biosystems, Inc. Methods and compositions for needleless delivery of binding partners
US7314632B1 (en) * 1997-07-11 2008-01-01 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pseudomonas exotoxin A-like chimeric immunogens

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL78775A (en) 1985-05-15 1992-06-21 Biotech Australia Pty Ltd Oral vaccines
CA2171192A1 (en) 1993-09-06 1995-03-16 Fiorenzo Stirpe Ribosome inactivating proteins extracted from seeds of saponaria ocymoides and vaccaria pyramidata, their preparation and immunotoxins containing them
US6072041A (en) 1996-06-03 2000-06-06 Case Western Reserve University Fusion proteins for protein delivery
DE69729389D1 (en) 1996-11-06 2004-07-08 Nasa PROTEASE ACTIVATED PSEUDOMONAS EXOTOXIN-A-LIKE PROPROTEINS
JPH10204455A (en) * 1997-01-27 1998-08-04 Chiyoda Corp Liquefaction of natural gas
US6472140B1 (en) 1997-09-05 2002-10-29 The General Hospital Corporation α-2- macroglobulin therapies and drug screening methods for Alzheimer's disease.
AU784657B2 (en) * 1999-10-22 2006-05-18 Genentech Inc. Delivery of proteins across polar epithelial cell layers
EP1224290B1 (en) 1999-10-22 2006-06-28 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, as represented by THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES Disulfide conjugated cell toxins and methods of making and using them

Patent Citations (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5668255A (en) * 1984-06-07 1997-09-16 Seragen, Inc. Hybrid molecules having translocation region and cell-binding region
US6022950A (en) * 1984-06-07 2000-02-08 Seragen, Inc. Hybrid molecules having translocation region and cell-binding region
US5965406A (en) * 1984-06-07 1999-10-12 Seragen, Inc. Recombinant DNAS encoding three-part hybrid proteins
US5696237A (en) * 1986-09-24 1997-12-09 The United States Of America As Represented By The Department Of Health And Human Services Recombinant antibody-toxin fusion protein
US5863745A (en) * 1986-09-24 1999-01-26 The United States Of America As Represented By The Department Of Health And Human Services Recombinant antibody-toxin fusion protein
US6051405A (en) * 1986-09-24 2000-04-18 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Constructs encoding recombinant antibody-toxin fusion proteins
US5458878A (en) * 1990-01-02 1995-10-17 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services P. exotoxin fusio proteins have COOHG220101al alterations which increase cytotoxicity
US5512658A (en) * 1990-05-11 1996-04-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pseudomonas exotoxins (PE) and conjugates thereof having lower animal toxicity with high cytocidal activity through substitution of positively charged amino acids
US5374620A (en) * 1990-06-07 1994-12-20 Genentech, Inc. Growth-promoting composition and its use
US5328984A (en) * 1991-03-04 1994-07-12 The United States As Represented By The Department Of Health & Human Services Recombinant chimeric proteins deliverable across cellular membranes into cytosol of target cells
US5541287A (en) * 1992-06-09 1996-07-30 Neorx Corporation Pretargeting methods and compounds
US5602095A (en) * 1992-06-18 1997-02-11 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Recombinant pseudomonas exotoxin with increased activity
US5854044A (en) * 1992-06-18 1998-12-29 National Institutes Of Health Recombinant pseudomonas exotoxin with increased activity
US5696077A (en) * 1992-06-23 1997-12-09 Associated Synapse Biologics Pharmaceutical composition containing botulinum B complex
US6488926B1 (en) * 1993-07-30 2002-12-03 Medeva Holdings B.V. Vaccine compositions
US6022966A (en) * 1993-11-22 2000-02-08 Neorx Corporation Pretargeting methods and compounds
US20020106370A1 (en) * 1994-05-13 2002-08-08 Donald Leonard Nicholas Cardy Improvements in or relating to peptide delivery
US6485726B1 (en) * 1995-01-17 2002-11-26 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US6391280B1 (en) * 1997-01-10 2002-05-21 Epicyte Pharmaceutical, Inc. J chain polypeptide targeting molecule linked to an imaging agent
US6086900A (en) * 1997-03-26 2000-07-11 Board Of Regents, The University Of Texas Systems Methods and compositions for using membrane-penetrating proteins to carry materials across cell membranes
US20050079171A1 (en) * 1997-07-11 2005-04-14 The Government Of The Usa As Represented By The Secretary Of The Dept. Of Health & Human Services Pseudomonas exotoxin A-like chimeric immunogens for eliciting a secretory IgA-mediated immune response
US7314632B1 (en) * 1997-07-11 2008-01-01 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Pseudomonas exotoxin A-like chimeric immunogens
US6251392B1 (en) * 1997-10-20 2001-06-26 Epicyte Pharmaceuticals, Inc. Epithelial cell targeting agent
US6440419B1 (en) * 1997-10-20 2002-08-27 Epicyte Pharmaceutical, Inc. Epithelial cell targeting agent
US20030054012A1 (en) * 2000-05-12 2003-03-20 Fitzgerald David J. Pseudomonas exotoxin a-like chimeric immunogens for eliciting a secretory iga-mediated immune response
US6673574B2 (en) * 2000-11-30 2004-01-06 Unigene Laboratories Inc. Oral delivery of peptides using enzyme-cleavable membrane translocators
US20040071731A1 (en) * 2000-12-21 2004-04-15 Fitzgerald David J. Chimeric protein comprising non-toxic pseudomonas exotoxin a and type iv pilin sequences
US20070003578A1 (en) * 2000-12-21 2007-01-04 The Gov. Of The U.S.A As Represented By The Sec. Of The Dep. Of Health And Huamn Services Chimeric protein comprising non-toxic pseudomonas exotoxin and type IV pilin sequences
US20040001801A1 (en) * 2002-05-23 2004-01-01 Corvas International, Inc. Conjugates activated by cell surface proteases and therapeutic uses thereof
US20060110409A1 (en) * 2002-07-19 2006-05-25 Shone Clifford C Targeted agents for nerve regeneration
US20040209330A1 (en) * 2003-03-24 2004-10-21 Wenfeng Xu Anti-IL-22RA antibodies and binding partners and methods of using in inflammation
US20040228831A1 (en) * 2003-05-15 2004-11-18 Belinka Benjamin A. Polymeric conjugates for tissue activated drug delivery
US20060104993A1 (en) * 2004-10-04 2006-05-18 Mrsny Randall J Methods and compositions for immunizing against Pseudomonas infection
US20060153798A1 (en) * 2004-10-04 2006-07-13 Mrsny Randall J Methods and compositions for needleless delivery of macromolecules
US20070141070A1 (en) * 2005-12-05 2007-06-21 Trinity Biosystems, Inc. Methods and compositions for needleless delivery of antibodies
US20070148131A1 (en) * 2005-12-05 2007-06-28 Trinity Biosystems, Inc. Methods and compositions for needleless delivery of binding partners

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11426466B2 (en) 2018-03-08 2022-08-30 Applied Molecular Transport Inc. Toxin-derived delivery constructs for pulmonary delivery
US11324833B2 (en) 2018-11-07 2022-05-10 Applied Molecular Transport Inc. Cholix-derived carriers for oral delivery of heterologous payload
US11504433B2 (en) 2018-11-07 2022-11-22 Applied Molecular Transport Inc. Cholix-derived carriers for oral delivery of heterologous payload

Also Published As

Publication number Publication date
EP1804832A4 (en) 2008-12-24
AU2005296064A1 (en) 2006-04-27
WO2006044205A3 (en) 2006-09-08
JP2008515808A (en) 2008-05-15
US7713737B2 (en) 2010-05-11
EP1804832A2 (en) 2007-07-11
US20060153798A1 (en) 2006-07-13
WO2006044205A2 (en) 2006-04-27
CA2583202A1 (en) 2006-04-27
KR20070073863A (en) 2007-07-10

Similar Documents

Publication Publication Date Title
US7713737B2 (en) Methods and compositions for needleless delivery of macromolecules
US20090148401A1 (en) Methods and compositions for needleless delivery of binding partners
US20090092660A1 (en) Methods and compositions for needleless delivery of particles
US10799565B2 (en) Cholix toxin-derived fusion molecules for oral delivery of biologically active cargo
US10617741B2 (en) Compositions and methods for oral delivery of therapeutic cargo
US20220275029A1 (en) Compositions and particles for payload delivery
US20220226445A1 (en) Cholix toxin-derived fusion molecules for oral delivery of biologically active cargo
US20090305978A1 (en) Methods for increasing the size of animals using needleless delivery constructs
CN101072584A (en) Methods and compositions for needleless delivery of macromolecules

Legal Events

Date Code Title Description
AS Assignment

Owner name: SANDERLING VENTURE PARTNERS V, L.P., AS COLLATERAL

Free format text: SECURITY AGREEMENT;ASSIGNOR:TRINITY BIOSYSTEMS, INC.;REEL/FRAME:022693/0286

Effective date: 20090515

AS Assignment

Owner name: TRINITY (ASSIGNMENT FOR THE BENEFIT OF CREDITORS),

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TRINITY BIOSYSTEMS, INC.;REEL/FRAME:024924/0182

Effective date: 20100603

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION