Methods and compositions for delivering active agents with enhanced pharmacological properties

Abstract
Provided herein are methods of enhancing in vivo efficacy of an active agent, comprising: administering to a subject an active agent that is coupled to a bioelastic polymer or elastin-like peptide, wherein the in vivo efficacy of the active agent is enhanced as compared to the same active agent when administered to the subject not coupled to (or not associated with) a bioelastic polymer or ELP.
Description
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY

The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (filename: PHAS_003_04US_SeqList_ST25.txt, date recorded: Nov. 12, 2015, file size 4 kilobytes).


FIELD OF THE INVENTION

The present invention concerns methods and formulations for improving pharmacological properties of active agents to be delivered to a subject.


BACKGROUND OF THE INVENTION

A significant problem with many candidate drugs, or even drugs in clinical use, is insufficient or unsatisfactory in vivo efficacy. Insufficient in vivo efficacy can be manifested in a variety of ways, such as (i) low bioavailability of the active compound; (ii) undesirably short half-life of the active compound, (iii) and/or undesirably high systemic toxicity of the active compound. To avoid eliminating otherwise promising drugs from clinical use, there remains a need for new approaches to enhancing the in vivo efficacy of active compounds in their delivery to human and animal subjects.


U.S. Pat. No. 6,004,782 to Danielle et al. describes bioelastic polypeptides and the expression thereof in host cells. The use thereof as fusion proteins containing therapeutics is described in a cursory fashion at column 15, lines 43-53 therein. Enhancing the in vivo efficacy of an active agent is neither suggested nor described.


U.S. Pat. No. 6,582,926 to Chilkoti describes, among other things, methods of targeting compounds to regions of interest in a subject by administering the compound to be delivered as a conjugate with a polymer that undergoes an inverse temperature transition (such as an ELP). Compounds to be delivered include certain radionuclides, chemotherapeutic agents, cytotoxic agents, and imaging agents as set forth at column 11, lines 6-21. Enhancing the in vivo efficacy of an active agent is neither suggested nor described.


U.S. Pat. No. 6,852,834 to Chilkoti describes, among other things, fusion proteins that are isolatable by phase transition, primarily to improve the yield thereof during manufacturing. Fusion proteins of therapeutic proteins are generally described at column 11, lines 10-24. Enhancing the in vivo efficacy of an active agent is neither suggested nor described.


SUMMARY OF THE INVENTION

The present invention provides a method of enhancing in vivo efficacy of an active agent, comprising: administering to a subject an active agent that is coupled to a bioelastic polymer or elastin-like peptide, wherein the in vivo efficacy of the active agent is enhanced as compared to the same active agent when administered to the subject not coupled to (or not associated with) a bioelastic polymer or ELP. In vivo efficacy may be enhanced in one, or more, of the following ways: solubility, bioavailability, effective therapeutic dose, formulation compatibility, resistance to proteolysis, half-life of the administered peptide active therapeutic agent, persistence in the body subsequent to administration, and rate of clearance from the body subsequent to administration.


Stated otherwise, the present invention provides a method of delivering an active agent to a subject, comprising: administering to said subject a conjugate of said active agent and an elastin-like peptide; wherein the in vivo efficacy of said active agent is enhanced in said subject when said active agent is administered to said subject in conjugated form as said conjugate as compared to the same amount of said active agent administered to said subject in unconjugated form. In some embodiments, at least one of: (i) the bioavailability of said active agent is greater; (ii) the half-life of said active agent is greater, (iii) the systemic toxicity of said active agent is less, in said subject when said active agent is administered to said subject in conjugated form as said conjugate as compared to the same amount of said active agent administered to said subject in the same way (e.g., the same dosage of active agent, administered in the same vehicle or carrier composition, by the same route of administration) in unconjugated form.


The active agent may be a diagnostic agent, a therapeutic agent, an imaging agent, or a chemotherapeutical agent. In some embodiments the active agent is a (i) small molecule, (ii) radionuclide, (iii) peptide (iv) peptidomimetic, (v) protein, (vi) antisense oligonucleotide, (vii) peptide nucleic acid, (viii) siRNA, (ix) metal chelate, or (x) carbohydrate. In some embodiments the active agent is a protein or peptide. In some embodiments the active agent is an antibody such as a therapeutic or diagnostic antibody.


The conjugate is generally to the subject in a treatment-effective amount by any suitable route, such as parenteral injection.


A further aspect of the present invention is a conjugate as described herein in a pharmaceutically acceptable carrier.


A further aspect of the present invention is the use of an active agent as described herein, in conjugated form as described herein, for carrying out a method as described herein.


The foregoing and other objects and aspects of the invention are explained in greater detail in the drawings herein and the specification set forth below.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. SDS-PAGE of a library of ELPs that are polymerized at the gene level, expressed in E. coli, and purified by exploiting the phase transition of the ELPs.



FIG. 2. SDS-PAGE analysis of (A) 14C-ELP visualized by copper staining, (B) 14C-ELP autoradiography after SDS-PAGE. (C) Pharmacokinetic analysis of 14C-ELP in mice (Balb/c nu/nu) exhibits a characteristic distribution and elimination response with a terminal half-life of 8.4 hr.



FIG. 3. Uptake and localization of an ELP. All images are of squamous cell carcinoma (FaDu) cells taken with a LSM-510 laser scanning confocal fluorescence microscope. The cells are incubated with ELP-Alexa488 (green) for 1 hour prior to co-staining (A) Cells are stained with DiI-CM (red) to label the cell membrane. (B) Cells are co-stained with lysotracker red (red) which selectively stains the lysosomes. The ELP colocalizes with the lysotracker red dye (note the yellow fluorescence).



FIG. 4. (A) Synthesis of a derivative with a terminal maleimide: It shows that a derivative with a terminal maleimide is prepared by attaching a pH sensitive hydrazone linker to Doxorubicin (hereinafter as Dox), a cancer chemotherapeutic agent at the 13-keto position. Then, the terminal maleimide of the derivative is conjugated to an ELP, which presents one or more Cysteine residues. (B) It is an example of cytotoxicity of Doxorubicin conjugated to ELP2-160JM2 conjugate (hereinafter as ELP-Dox) in a MTT cell viability assay. The cytotoxicity of ELP-Doxorubicin and unconjugated Dox is a function of the equivalent Doxorubicin concentration. Compared to the free drug, ELP-Doxorubicin demonstrates almost equivalent cytotoxicity of the free drugs. (C) ELP-Dox and Dox are injected at the same concentration into mice via tail vein injections. After 1 h, no Dox can be detected from the blood samples of the mice. However, ˜20 injected gram/g serum (% ID/g) is detected from the mice injected with ELP-Dox. The result of this experiment demonstrates that the conjugated form has a greater plasma half-life of the drug. (D) It demonstrates the biodistribution of Dox and ELP-Dox injected nude mice with human tumor xenografts. Upon conjugation of Dox to ELP, a different pattern of distribution is obtained. The concentrations of Dox in the heart; liver and lung are greater than those of ELP-OPDX, however, the concentration of ELP-Dox in tumor is greater than that of Dox.



FIG. 5. Accumulation of 14C-labeled ELPs in tumors. The two ELPs reported are a thermally sensitive ELP1 and a thermally insensitive ELP2 in tumors that are either heated to 41.5° C. or not heated.



FIG. 6. Expression of different ELP fusion proteins as examples of recombinant ELP-protein conjugates. All ELP-protein conjugates are prepared by fusion of the gene of the protein, ELP and expression in a heterogeneous expression system (e.g., E. coli). The left panel shows examples of blue fluorescent protein (BFP), chloramphenicol acetyl transferase (CAT) and Kringle1-3 domains (K1-3: angiostatin). The right panel shows other examples of purified ELP-protein conjugates.



FIG. 7. SDS-PAGE of purification of ELP fusion protein in the following orientation: The preparation of the protein-ELP and ELP-protein shows that protein conjugates of ELPs can be synthesized in either orientation for CAT, BFP, and Trx. (A) Thin layer chromatography showing activity of CAT, (B) Fluorescence of BFP-ELP n ELP-BFP showing functionality of BDFP in the fusion.



FIG. 8. (I) SDS-PAGE characterization of inverse transition purifications: It shows each stage of purification for the thioredoxin/90-mer ELP fusion (49.9 kDa, lanes 1 through 5) Lane A: soluble lysate; lane B: discarded supernatant containing contaminating E. coli proteins; lane 3: resolubilized pellet fraction containing purified fusion protein, lane 4, second round supernatant; lane 5: second round pellet; lane 6: molecular weight markers (kDa). (II) Total protein (green) and thioredoxin (Trx) activity (red) for each stage of purification of the thioredoxin/90-mer ELP. Values are normalized to those determined for the soluble lysate.



FIG. 9. Examples of synthesis of ELP-peptide conjugates. All conjugates are prepared recombinantly as fusions with ELP. The two lanes in each SDS-PAGE gels from A-F show the fusion (conjugate) on left, and the peptide on right. Mass spectrometry results for each purified peptide are shown below the SDS-PAGE gels. (A) Morphine modulating neuropeptide (MMN), (B) Neuropeptide Y (NPY) (2.7 kDa) (note: although gel was overloaded, Commassie does not stain NPY) (ELP4-60-NPY 222 mg/L fusion (conjugate) on left, NPY 20 mg/L peptide on right), (C) Orexin B (3.0 kDa) (ELP4-60-Orexin B 320 mg/L fusion (conjugate) on left, Orexin B 19 mg/L peptide on right), (D) Leptin (4.0 kDa) (ELP4-60-Leptin 415 mg/L fusion (conjugate) on left, Leptin 19.5 mg/L peptide on right), (E) ACTH (4.6 kDa) (ACTH-ELP1-90 133 mg/L fusion (conjugate) on left, ACTH 19 mg/L peptide on right), (F) Pro-calcitonin (6.2 kDa) (ELP1-90-pro-CT 260 mg/L fusion (conjugate) on left, pro-CT 23 mg/L peptide on right).



FIG. 10. Examples of ELP-peptide conjugate. Recombinant fusion of antimicrobial peptide MSI-78 with ELP (ELP-peptide conjugate). Sequence of MSI-78: Sequence=GIGKFLKKAKKFGKAFVKILKK (SEQ ID NO.: 2). (A) Purification of ELP 1-90-MSI-78 and MSI-78. SDS-Page gel shows both high purity of the conjugate and the peptide produced recombinantly. (B) Purity of EP-MSI-78 conjugate determined by liquid chromatography combined with mass spectrometry. One compound was detected with a molecular weight of 2476.6 and purity is >99% by LC-ELSD (C) Bactericidal activity of MSI-78.





DETAILED DESCRIPTION OF THE INVENTION

The disclosures of all United States patent references cited herein are to be incorporated by reference herein in their entirety.


“Active agent” as used herein may be any suitable active agent, including therapeutic and diagnostic or imaging agents.


Examples of imaging agents include, but are not limited to, the following: radioisotopes (e.g., 3H, 14C, 35S, 125I, 131I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), MRI contrast agents (e.g., Gadolinum chelates (Gd)) luminescent labels such as luminol; enzymatic labels (e.g., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase, acetylcholinesterase), biotinyl groups (which can be detected by marked avidin e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or calorimetric methods), predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags). Indirect methods may also be employed in which the primary antigen-antibody reaction is amplified by the introduction of a second antibody.


“Therapeutic agent” as used herein may be any suitable therapeutic agent, including but not limited to radionuclides, chemotherapeutic agents; cytototoxic agents, parathyroid hormone related protein (parathyroid hormone related protein), growth hormone (GH) particularly human and bovine growth hormone, growth hormone-releasing hormones; interferon including α-, β-, or γ-interferons, etc, interleukin-I; interleukin-II; erythropoietin including α- and β-erythropoietin (EPO), granulocyte colony stimulating factor (GCSF), granulocyte macrophage colony stimulating factor (GM-CSF), anti-angiogenic proteins (e.g., angiostatin, endostatin) PACAP polypeptide (pituitary adenylate cyclase activating polypeptide), vasoactive intestinal peptide (VIP), thyrotrophin releasing hormone (TRH), corticotrophin releasing hormone (CRH), vasopressin, arginine vasopressin (AVP), angiotensin, calcitonin, atrial naturetic factor, somatostatin, adrenocorticotropin, gonadotropin releasing hormone, oxytocin, insulin, somatotropin, HBS antigen of hepatitis B virus, plasminogen tissue activator, coagulation factors including coagulation factors VIII and IX, glucosylceramidase, sargramostim, lenograstin, filgrastin, interleukin-2, dornase-α, molgramostim, PEG-L-asparaginase, PEG-adenosine deaminase, hirudin, eptacog-α (human blood coagulation factor VIIa) nerve growth factors, transforming growth factor, epidermal growth factor, basic fibroblast growth factor, VEGF; heparin including low molecular weight heparin, calcitonin; atrial naturetic factor; antigens; monoclonal antibodies; somatostatin; adrenocorticotropin, gonadotropin releasing hormone; oxytocin; vasopressin; cromolyn sodium; vancomycin; desferrioxamine (DFO); parathyroid hormone, anti-microbials, antifungals, an immunogen or antigen, an antibody such as a monoclonal antibody, or any combination thereof. See, e.g., U.S. Pat. Nos. 6,967,028; 6,930,090; and 6,972,300.


Example therapeutic agents include all of the therapeutic agents set forth in paragraphs 0065 through 0388 of W. Hunter, D. Gravett, et al., US Patent Application Publication No. 20050181977 (Published Aug. 18, 2005) (assigned to Angiotech International AG) the disclosure of which is incorporated by reference herein in its entirety.


“Radionuclide” as described herein may be any radionuclide suitable for delivering a therapeutic dosage of radiation to a tumor or cancer cell, including but not limited to 227Ac, 211At, 131Ba, 77Br, 109Cd, 51Cr, 67Cu, 165Dy, 155Eu, 153Gd, 198Au, 166Ho, 113mIn, 115mIn, 123I, 125I, 131I, 189Ir, 191Ir, 192Ir, 194Ir, 52Fe, 55Fe, 59Fe, 177Lu, 109Pd, 32P, 226Ra, 186Re, 188Re, 153Sm, 46Sc, 47Sc, 72Se, 75Se, 105Ag, 89Sr, 35S, 177Ta, 117mSn, 121Sn, 166Yb, 169Yb, 90Y, 212Bi, 119Sb, 197Hg, 97Ru, 100Pd, 101mRh, and 212Pb. Radionuclides may also be those useful for delivering a detectable dosage for imaging or diagnostic purposes, even where those compounds are not useful for therapeutic purposes.


“Chemotherapeutic agent” as used herein includes but is not limited to methotrexate, daunomycin, mitomycin, cisplatin (cisplatinum or cis-dianminedichloroplatinum (II)(CCDP)), vincristine, epirubicin, fluorouracil, verapamil, cyclophosphamide, cytosine arabinoside, aminopterin, bleomycin, mitomycin C, democolcine, etoposide, mithramycin, chlorambucil, melphalan, daunorubicin, doxorubicin, tamoxifen, paclitaxel, vincristine, vinblastine, camptothecin, actinomycin D, and cytarabine, combrestatin and its derivatives.


“Cytotoxic agent” as used herein includes but is not limited to ricin (or more particularly the ricin A chain), aclacinomycin, diphtheria toxin, Monensin, Verrucarin A, Abrin, Vinca alkaloids, Tricothecenes, and Pseudomonas exotoxin A.


“Immunogen” and “antigen” are used interchangeably and mean any compound to which a cellular or humoral immune response is to be directed against, and include bacterial antigens, viral antigens, and tumor antigens. Non-living immunogens (e.g., killed immunogens, subunit vaccines, recombinant proteins or peptides or the like) are currently preferred. Examples of suitable immunogens include those derived from bacterial surface polysaccharides which can be used in carbohydrate-based vaccines. Bacteria typically express carbohydrates on their cell surface as part of glycoproteins, glycolipids, O-specific side chains of lipopolysaccharides, capsular polysaccharides and the like. Exemplary bacterial strains include Streptococcus pneumonia, Neisseria meningitidis, Haemophilus influenza, Klebsiella spp., Pseudomonas spp., Salmonella spp., Shigella spp., and Group B streptococci. A number of suitable bacterial carbohydrate epitopes which may be used as the immunogen in the present invention are described in the art (e.g., Sanders, et al. Pediatr. Res. 37:812-819 (1995); Bartoloni, et al. Vaccine 13:463-470 (1995); Pirofski, et al., Infect. Immun. 63:2906-2911 (1995) and International Publication No. WO 93/21948) and are further described in U.S. Pat. No. 6,413,935. Exemplary viral antigen or immunogen includes those derived from HIV (e.g., gp120, nef, tat, pol). Exemplary fungal antigens include those derived from Candida albicans, Cryptococcus neoformans, Coccidoides spp., Histoplasma spp., and Aspergillus spp. Parasitic antigens include those derived from Plasmodium spp., Trypanosoma spp., Schistosoma spp., Leishmania spp. and the like. Exemplary carbohydrate epitopes that may be utilized as antigens or immunogens in the present invention include but are not limited to the following: Galα1,4Galβ-(for bacterial vaccines); GalNAcα-(for cancer vaccines); Manβ1,2(Manβ)nManβ-(for fungal vaccines useful against, for example, Candida albicans), where n=O→∞; GalNAcβ1,4(NeuAcα2,3)Galβ1,4Glcβ-O-ceramide (for cancer vaccines); Galα1,2(Tyvα1,3)Manα1,4Rhaα1,3Galα1,2(Tyaα1,3)Manα4Rha- and Galα1,2(Abeα1,3)Manα1,4Rhaα1,3Galα1,2(Abeα1,3)Manα1,4Rhaα1,3Galα1,2 (Abeα1,3)Manα1,4Rha-(both of which are useful against, for example, Salmonella spp.). Carbohydrate epitopes as antigens or immunogens and the synthesis thereof are described further in U.S. Pat. No. 6,413,935. In one embodiment the immunogen may be an anthrax immunogen; i.e. an immunogen that produces protective immunity to Bacillus anthracis, such as anthrax vaccine, A, (Michigan Department of Health, Lansing, Mich.; described in U.S. Pat. No. 5,728,385). Other examples of immunogens or antigens include but are not limited to those that produce an immune response or antigenic response to the following diseases and disease-causing agents: adenoviruses; Bordetella pertussus; Botulism; bovine rhinotracheitis; Branhamella catarrhalis; canine hepatitis; canine distemper; Chlamydiae; Cholera; coccidiomycosis; cowpox; cytomegalovirus; cytomegalovirus; Dengue fever; dengue toxoplasmosis; Diphtheria; encephalitis; Enterotoxigenic Escherichia coli; Epstein Barr virus; equine encephalitis; equine infectious anemia; equine influenza; equine pneumonia; equine rhinovirus; feline leukemia; flavivirus; Globulin; haemophilus influenza type b; Haemophilus influenzae; Haemophilus pertussis; Helicobacter pylori; Hemophilus; hepatitis; hepatitis A; hepatitis B; Hepatitis C; herpes viruses; HIV; HIV-1 viruses; HIV-2 viruses; HTLV; Influenza; Japanese encephalitis; Klebsiellae species; Legionella pneumophila; leishmania; leprosy; lyme disease; malaria immunogen; measles; meningitis; meningococcal; Meningococcal Polysaccharide Group A; Meningococcal Polysaccharide Group C; mumps; Mumps Virus; mycobacteria and; Mycobacterium tuberculosis; Neisseria; Neisseria gonorrhoeae; Neisseria meningitidis; ovine blue tongue; ovine encephalitis; papilloma; parainfluenza; paramyxovirus; paramyxoviruses; Pertussis; Plague; Pneumococcus; Pneumocystis carinii; Pneumonia; Poliovirus; Proteus species; Pseudomonas aeruginosa; rabies; respiratory syncytial virus; rotavirus; Rubella; Salmonellae; schistosomiasis; Shigellae; simian immunodeficiency virus; Smallpox; Staphylococcus aureus; Staphylococcus species; Streptococcus pneumoniae; Streptococcus pyogenes; Streptococcus species; swine influenza; tetanus; Treponema pallidum; Typhoid; Vaccinia; varicella-zoster virus; and Vibrio cholerae. The antigens or immunogens may, include various toxoids, viral antigens and/or bacterial antigens such as antigens commonly employed in the following vaccines: chickenpox vaccine; diphtheria, tetanus, and pertussis vaccines; haemophilus influenzae type b vaccine (Hib); hepatitis A vaccine; hepatitis B vaccine; influenza vaccine; measles, mumps, and rubella vaccines (MMR); pneumococcal vaccine; polio vaccines; rotavirus vaccine; anthrax vaccines; and tetanus and diphtheria vaccine (Td). See, e.g., U.S. Pat. No. 6,309,633. Antigens or immunogens that are used to carry out the present invention include those that are derivatized or modified in some way, such as by conjugating or coupling one or more additional groups thereto to enhance function or achieve additional functions such as targeting or enhanced delivery thereof, including but not limited to those techniques described in U.S. Pat. No. 6,493,402 to Pizzo et al. (α-2 macroglobulin complexes); U.S. Pat. No. 6,309,633; U.S. Pat. No. 6,207,157; U.S. Pat. No. 5,908,629, etc.


Interferon (IFNs) are used herein refers to natural proteins produced by the cells of the immune system of most vertebrates in response to challenges by foreign agents such as viruses, bacteria, parasites and tumor cells, and its function is to inhibit viral replication within other cells. Interferons belong to the large class of glycoproteins known as cytokines Three major classes of interferons for human have been discovered as type I, type II and type III, classified according to the type of receptor through which they signal. Human type I IFNs comprise a vast and growing group of IFN proteins, designated IFN-α, IFN-β, IFN-κ, IFN-δ, IFN-ε, IFN-τ, IFN-ω and IFN-ξ. [See Interferon-ξ/limitin: Novel type I Interferon that displays a narrow range of biological activity, Oritani Kenji and Tomiyama Yoshiaki, International Journal of hematology, 2004, 80, 325-331; Characterization of the type I interferon locus and identification of novel genes, Hardy et al., Genomics, 2004, 84, 331-345.] Homologous molecules to type I IFNs are found in many species, including most mammals, and some have been identified in birds, reptiles, amphibians and fish species. [See The interferon system of non-mammalian vertebrates, Schultz et al., Developmental and Comparative Immunology, 28, 499-508.] All type I IFNs bind to a specific cell surface receptor complex known as the IFN-α receptor (IFNAR) that consists of IFNAR1 and IFNAR2 chains. The type II IFNs only has one member called IFN-γ. Mature IFN-γ is an anti-parallel homodimer, which binds to the IFN-γ receptor (IFNGR) complex to elicit a signal within its target cell. The type III IFN group consists of three IFN-λ molecules called IFN-λ1, IFN-λ2 and IFN-λ3 (also called IL29, IL28A and IL28B respectively). [See Novel interferons, Jan Vilcek, Nature Immunology, 2003, 4, 8-9.] The IFN-λ molecules signal through a receptor complex consisting of IL10R2 (also called CRF2-4) and IFNLR1 (also called CRF2-12). [See Murine interferon lambdas (type III interferons) exhibit potent antiviral activity in vivo in a poxvirus infection model, Bartlett et al., Journal of General Virology, 2005, 86, 1589-1596.]


“Antibody” or “antibodies” as used herein refers to all types of immunoglobulins, including IgG, IgM, IgA, IgD, and IgE. The term “immunoglobulin” includes the subtypes of these immunoglobulins, such as IgG1, IgG2, IgG3, IgG4, etc. Of these immunoglobulins, IgM and IgG are preferred, and IgG is particularly preferred. The antibodies may be of any species of origin, including (for example) mouse, rat, rabbit, horse, or human, or may be humanized or chimeric antibodies. The term “antibody” as used herein includes antibody fragments which retain the capability of binding to a target antigen, for example, Fab, F(ab′)2, and Fv fragments, and the corresponding fragments obtained from antibodies other than IgG. Such fragments are also produced by known techniques. Antibodies may be for diagnostic purposes or for therapeutic purposes. Examples of therapeutic antibodies include but are not limited to herceptin, rituxan, campath (Mellinium pharma Inc.), gemtuzumab (Cell tech.), herceptin (Genentech), panorex (Centocor GSK), rituximab (Genentech), bexxar (Coraxia GSK), edrecolomab (Glaxo-wellcome), alemtuzumab (ILEX Pharmaceuticals), mylotrag (Whety-Ayerst), IMC-C225, smartin 195, and mitomomab (Imclone systems). Therapeutic antibodies include those coupled to a therapeutic compound and “cold dose” antibodies, such as for reducing non-specific binding. See, e.g., Abrams et al., U.S. Pat. No. RE38,008.


“Treat” as used herein refers to any type of treatment or prevention that imparts a benefit to a subject afflicted with a disease or at risk of developing the disease, including improvement in the condition of the subject (e.g., in one or more symptoms), delay in the progression of the disease, delay the onset of symptoms or slow the progression of symptoms, etc. As such, the term “treatment” also includes prophylactic treatment of the subject to prevent the onset of symptoms. As used herein, “treatment” and “prevention” are not necessarily meant to imply cure or complete abolition of symptoms.” to any type of treatment that imparts a benefit to a patient afflicted with a disease, including improvement in the condition of the patient (e.g., in one or more symptoms), delay in the progression of the disease, etc.


“Treatment effective amount” as used herein means an amount of the antibody sufficient to produce a desirable effect upon a patient inflicted with a condition such as cancer, diabetes, bacterial or viral infection, etc., including improvement in the condition of the patient (e.g., in one or more symptoms), delay in the progression of the disease, etc. With an immunogen a “treatment effective amount” may be an amount effective to produce an immune response or protective immunity (in whole or in part) against subsequent infection by a bacterial, viral, fungal, protozoal, or other microbial agent.


“Conjugate” as used herein refers to two or more moieties or functional groups that are covalently or noncovalently joined to one another, such that the two or more groups function together as a single structure under the conditions of the methods described herein. In one embodiment, the conjugate is a fusion protein. In some embodiments, the conjugate refers to the two moieties that are chemically or enzymatically attached to each other.


“Fusion protein” as used herein refers to a protein or peptide, produced by recombinant means (i.e., expression from a nucleic acid) that is comprised of a first protein or peptide covalently joined on expression to a second protein or peptide.


A “polymer that undergoes an inverse temperature transition” herein refers to a polymer that is soluble in an aqueous solution at a lower temperature, and is insoluble in an aqueous solution at a higher temperature.


“Transition temperature” or “Tt” as used herein, refers to the temperature above which a polymer that undergoes an inverse temperature transition is insoluble in an aqueous system (e.g., water, physiological saline solution), and below which such a polymer is soluble in an aqueous system.


A “bioelastic polymer” is, in general, a polypeptide that exhibits an inverse temperature transition. Bioelastic polymers are discussed in greater detail below. Such bioelastic polymers are typically elastin-like peptides.


While the present invention is concerned primarily with the treatment of human subjects, the invention may also be used for the treatment of animal subjects, particularly mammalian subjects such as dogs, cats, horses, cows, pigs, etc., for veterinary purposes.


Subjects in need of treatment by the methods described herein include subjects afflicted with any disorder conventionally or currently treated or diagnosed by the active agents described herein, including but not limited to subjects afflicted with solid tumors or cancers such as lung, colon, breast, brain, liver, prostate, spleen, muscle, ovary, pancreas, skin (including melanoma) etc; subjects afflicted with or at risk of developing a viral, bacterial, protozoal, or other microbial infection; etc.


Bioelastic polymers. Bioelastic polymers are known and described in, for example, U.S. Pat. No. 5,520,672 to Urry et al. In general, bioelastic polymers are polypeptides comprising elastomeric units of bioelastic pentapeptides, tetrapeptides, and/or nonapeptides (that is, “elastin-like peptides”). Thus in some embodiments the elastomeric unit is a pentapeptide, in other embodiments the elastomeric unit is a tetrapeptide, and in still other embodiments the elastomeric unit is a nonapeptide. Bioelastic polymers that may be used to carry out the present invention are set forth in U.S. Pat. No. 4,474,851, which describes a number of tetrapeptide and pentapeptide repeating units that can be used to form a bioelastic polymer. Specific bioelastic polymers that can be used to carry out the present invention are also described in U.S. Pat. Nos. 4,132,746; 4,187,852; 4,500,700; 4,589,882; and 4,870,055. Still other examples of bioelastic polymers are set forth in U.S. Pat. No. 6,699,294 to Urry, U.S. Pat. No. 6,753,311 to Fertala and Ko; and U.S. Pat. No. 6,063,061 to Wallace.


As disclosed in U.S. Pat. No. 4,474,851, elastomeric peptides may have a sequence of regularly appearing β-turns, forming an overall spiral conformation (e.g., a β-spiral, which is a series of regularly repeating β-turns). The spiral structures are more open than the more common α-helix. As a result, the atoms in the peptide backbone have a high freedom of movement (e.g., as compared to the freedom of movement for an α-helix). This is particularly true of librational motions involving peptide moieties. A libration is a torsional oscillation involving simultaneous rotational motions of the two single bonds on each side of a librating moiety. The moiety involved in a libration may be a single peptide bond or several peptide residues. For adequate freedom of motion to exist, it is important, however, that the carbonyl oxygen and the amino hydrogen of the peptide bond not be involved in hydrogen bonding to other parts of the molecule or to other molecules. Otherwise a greater energy barrier to the libration exists and motion will be restricted. Since non-hydrogen-bonded segments having freedom of motion exist in the β-spiral between the points of hydrogen bonding for the β-turns, these segments may be said to be librationally suspended. Librationally suspended segments therefore are a structural feature that exists in certain elastic peptides because of the repeating β-turns with relative infrequent hydrogen bonding. Librationally suspended segments resulting from the β-spiral structure are thought to give rise to elasticity, as will be further discussed.


Another factor leading to the high librational freedom of such molecules is the absence of significant polar interactions between the amino acid residues, either intrachain or interchain, other than a hydrogen bond within the β-turn. The amino acid residues present are mostly hydrophobic or glycine and accordingly do not exert significant forces on one another through space. If a significant number of charged or polar groups were present, electrostatic interactions might limit librational freedom and restrict the number of available states in the relaxed (non-extended) form of the molecules. Polar and charged amino acid residues are not strictly prohibited, however, if their presence does not destroy the elasticity of the elastic peptide component as a whole. For example, an occasional serine residue is present in naturally occurring tropoelastin without destroying elasticity. Accordingly, hydrophobic amino acid residues and glycines are preferred in forming elastomeric polypeptides of the present type although other amino acids may be present to a some extent.


Although not intending to be bound by theory, the elasticity of polypeptides of the β-turn structure may be caused by thermodynamic drive toward greater entropy. The relaxed state of the β-spiral has a large degree of librational freedom and thus the atoms of the peptide chain can exist in a large number of positions. When the molecules are stretched, the degree of freedom is reduced, particularly for librational motions, and when the tension is released, a thermodynamic driving force toward higher entropy results in reformation of the contracted β-spiral.


In one embodiment, the bioelastic polymers used to carry out the present invention are polypeptides of the general formula (VPGXG)m (SEQ ID NO.: 1), where X is any amino acid (e.g., Ala, Leu, Phe) and m is any suitable number such as 2, 3 or 4 up to 60, 80 or 100 or more. The frequency of the various amino acids as the fourth amino acid can be changed, as well as the frequency of X. For example, the bioelastic polymers used to carry out the present invention may be polypeptides of the general formula: [(VPGXG)m(VPGKG)n]o (SEQ ID NO.: 3), where m is 2, 3 or 4 to 20 or 30, n is 1, 2 or 3, o is at least 2, 3 or 4 up to 30, 40 or 50 or more. Any ratios of X/K can be possible, which means where m is 1, 2, or 3 up to 100, 150, or 300 or more, n is 1, 2 or 3 up to 100 or 150 or 300 or more, or is at least 1, 2, or 3 up to 100, 150 or 300 or more.


For example, bioelastic polymers used to carry out the present invention may comprise repeating elastomeric units selected from the group consisting of bioelastic pentapeptides and tetrapeptides, where the repeating units comprise amino acid residues selected from the group consisting of hydrophobic amino acid and glycine residues and where the repeating units exist in a conformation having a beta-turn of the formula:




embedded image



wherein R1-R5 represent side chains of amino acid residues 1-5, and m is 0 when the repeating unit is a tetrapeptide or 1 when the repeating unit is a pentapeptide. Nonapeptide repeating units generally consist of sequential tetra- and pentapeptides. Preferred hydrophobic amino acid residues are selected from the group consisting of alanine, valine, leucine, isoleucine, proline, phenylalanine, tryptophan, and methionine. In many cases, the first amino acid residue of the repeating unit is a residue of valine, leucine, isoleucine or phenylalanine; the second amino acid residue is a residue of proline; the third amino acid residue is a residue of glycine; and the fourth amino acid residue is glycine or a very hydrophobic residue such as tryptophan, phenylalanine or tyrosine. Particular examples include the tetrapeptide Val-Pro-Gly-Gly (SEQ ID NO.: 4), the tetrapeptide GGVP (SEQ ID NO.: 5), the tetrapeptide GGFP (SEQ ID NO.: 6), the tetrapeptide GGAP (SEQ ID NO.: 7), the pentapeptide is Val-Pro-Gly-Val-Gly (SEQ ID NO.: 8), the pentapeptide GVGVP (SEQ ID NO.: 9), the pentapeptide GKGVP (SEQ ID NO.: 10), the pentapeptide GVGFP (SEQ ID NO.: 11), the pentapeptide GFGFP (SEQ ID NO.: 12), the pentapeptide GEGVP (SEQ ID NO.: 13), the pentapeptide GFGVP (SEQ ID NO.: 14), and the pentapeptide GVGIP (SEQ ID NO.: 15). See, e.g., U.S. Pat. No. 6,699,294 to Urry.


Coupling of conjugates may be carried out by any suitable means, including chemical and recombinant means. Chemical or enzymatic coupling may be carried out by procedures known in the art. (See, e.g., U.S. Pat. Nos. 6,930,090; 6,913,903; 6,897,196; and 6,664,043). Coupling of conjugates by recombinant means (e.g., where elastin is joined to a protein or peptide such as an interleukin, by recombinant means such as by expression of a fusion protein) may also be carried out by procedures known in the art (See, e.g., U.S. Pat. Nos. 6,974,572; 6,972,322; 6,962,978; and 6,956,112).


Formulations and administration. Administering of the conjugate to the subject may be carried out by any suitable means, such as subcutaneous injection, intraperitoneal injection, intravenous injection, intramuscular injection, intratumoral, oral administration, inhalation administration, transdermal administration, etc. Preferred administration techniques are typically “systemic” in that a particular region of interest is not specifically targeted.


The conjugates (or “active compounds”) described above may be formulated for administration in a single pharmaceutical carrier or in separate pharmaceutical carriers for the treatment of a variety of conditions. In the manufacture of a pharmaceutical formulation according to the invention, the active compounds including the physiologically acceptable salts thereof, or the acid derivatives of either thereof are typically admixed with, inter alia, an acceptable carrier. The carrier must, of course, be acceptable in the sense of being compatible with any other ingredients in the formulation and must not be deleterious to the patient. The carrier may be a solid or a liquid, or both, and is preferably formulated with the compound as a unit-dose formulation, for example, a tablet, which may contain from 0.5% to 95% by weight of the active compound. One or more active compounds may be incorporated in the formulations of the invention, which may be prepared by any of the well-known techniques of pharmacy consisting essentially of admixing the components, optionally including one or more accessory ingredients.


The formulations of the invention include those suitable for oral, rectal, topical, buccal (e.g., sub-lingual), parenteral (e.g., subcutaneous, intramuscular, intradermal, or intravenous) and transdermal administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular active compound which is being used.


Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion. Such formulations may be prepared by any suitable method of pharmacy, which includes the step of bringing into association the active compound and a suitable carrier (which may contain one or more accessory ingredients as noted above). In general, the formulations of the invention are prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture. For example, a tablet may be prepared by compressing or molding a powder or granules containing the active compound, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, and/or surface active/dispersing agent(s). Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder. Formulations of the present invention suitable for parenteral administration conveniently comprise sterile aqueous preparations of the active compound, which preparations are preferably isotonic with the blood of the intended recipient. These preparations may be administered by means of subcutaneous, intravenous, intramuscular, or intradermal injection. Such preparations may conveniently be prepared by admixing the compound with water or a glycine buffer and rendering the resulting solution sterile and isotonic with the blood.


Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Formulations suitable for transdermal administration may also be delivered by iontophoresis (see, for example, Pharmaceutical Research 3 (6):318 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound. Suitable formulations comprise citrate or bis/tris buffer (pH 6) or ethanol/water and contain from 0.1 to 0.2M active ingredient. The therapeutically effective dosage of any one active agent, the use of which is in the scope of present invention, will vary somewhat from compound to compound, patient to patient, and will depend upon factors such as the condition of the patient and the route of delivery. Such dosages can be determined in accordance with routine pharmacological procedures known to those skilled in the art, particularly in light of the disclosure provided herein. In one example, the dosage is from 1 to 10 micrograms of active compound per Kilogram subject body weight.


In another example, where the therapeutic agent is 131I, the dosage to the patient is typically from 10 mCi to 100, 300 or even 500 mCi. Stated otherwise, where the therapeutic agent is 131I, the dosage to the patient is typically from 5,000 Rads to 100,000 Rads (preferably at least 13,000 Rads, or even at least 50,000 Rads). Doses for other radionuclides are typically selected so that the tumoricidal dosage is equivalent to the foregoing range for 131I.


In a preferred embodiment, the improved pharmacological properties of the invention are utilized to improve the delivery and/or dosage regime to the subject. For example, an improved half live of the active agent is utilized to reduce the frequency of dosages to the patient (e.g., one dosage or administration every three or four days; more preferably one administration per week, still more preferably one administration every two weeks; still more preferably one administration per month); an improved bioavailability is utilized to reduce the overall dosage of the active agent administered to the patient, etc.


The present invention is explained in greater detail in the following non-limiting examples.


EXAMPLES

The goal of this invention is to selectively deliver drugs or imaging agents to diseased sites in order to improve therapeutic efficacy and limit systemic toxicity.


The invention has four parts:


1. Drug or Imaging Agent carriers: The carrier is a novel macromolecular drug carrier, consisting of elastin-like polypeptides (ELPs). ELPs belong to a unique class of biopolymers that undergo an inverse temperature phase transition; they are soluble at temperatures below their transition temperature (Tt) but become insoluble and aggregate at temperatures above their Tt[1-3].


(i) The ELP may be designed with a Tt that is below the local temperature at the diseased site so that it will aggregate at the diseased site.


(ii) Alternatively the ELP may be designed to have a Tt that is above the diseased site so as to remain in soluble form.


(iii) The ELP may contain sites for the covalent or enzymatic attachment of drugs or imaging agents or targeting moieties.


(iv) The ELP may also be designed to contain genetically encodable targeting moieties (one or more) such as a peptide or protein to specifically target the ELP to the diseased site or organ.


2. Definition of


(A) Drug: Any molecule that has therapeutic value against any disease.


(B) Imaging agent: Any molecule that provides visualization of the diseased site or organ


Example of the drug or imaging agent would include, though not exclusively: (i) small molecule, (ii) radionuclide, (iii) peptide, (iv) peptidomimetic, (v) protein, (vi) antisense oligonucleotide, (vii) peptide nucleic acid, (viii) siRNA, (ix) metal chelate, (x) carbohydrates.


3. Attachment or association of drug or imaging agent. The drug can be covalently linked to the ELP through a stable or labile linkage scheme. The drug may be hydrophobically associated with the ELP. The drug may be attached to the ELP through a chelation method. The drug may be associated with the ELP through molecular recognition through secondary bonds. The drug may also be attached to the ELP through the action of an enzyme. In the case of molecules such as peptides proteins that can be produced recombinantly, the ELP and drug may be produced as a single entity in suitable host (E. coli, Pichia pastoris, mammalian cells, or baculovirus) from a synthetic or cloned gene. The “ELP-drug/imaging agent conjugate” may be synthesized so that the link between the conjugate may be stable so as to deliver the single entity as a therapeutic or imaging agent or designed to be labile under the action of pH or light, or the action of enzymes to liberate the drug from the ELP.


4. Administration: The ELP-drug conjugate or fusion protein will be: (i) injected into the subject systemically (iv, ia, ip or im) (ii) locally into the diseased site or organ, (iii) or delivered orally, or (iv) parenterally.


The injected ELP-drug/imaging agent conjugate or fusion protein will exhibit as compared to the free drug one or more of the following: (1) enhanced solubility of the drug/imaging agent in its conjugated form over free drug/imaging agent, increased circulation half-life, exhibit reduced clearance from the body, or increased bioavailability of the drug/imaging agent, resulting in reduced dose and frequency of injection, an improved therapeutic index or improved visualization of the diseased site or organ.


Synthesis and Characterization of ELPs.


ELPs are typically prepared by a recombinant synthesis in E. coli. However, other hosts may be used for recombinant synthesis as well. ELP may also be prepared by a chemical synthesis. In a typical example of a recombinant synthesis, the polymerization process is carried out at the gene level by a method called recursive directional ligation (RDL), in which a synthetic gene for a repeat sequence for the ELP (typically encoding ˜10 pentapeptides of VPGXG (SEQ ID NO.: 1)) are ligated in a head-to-tail manner recursively. After n rounds of ligation into a plasmid, this provides a library of n+1 ELP genes, all of which encode the same peptide sequence, but with MWs that are multiples of the drug.


ELP-Drug Conjugation.


An ELP containing a unique C-terminal cysteine residue is synthesized and purified by inverse transition cycling (ITC) and conjugated to Doxorubicin molecules through four different pH-sensitive, maleimide-activated, hydrazone linkers. The linkers' structures or length have little effect on the Tt of the ELP-Doxorubicin conjugates, since all conjugates' Tts are similar to those of the native ELP (data not shown). However, the ELP-Doxorubicin conjugates with longer linkers exhibits slower transition kinetics than the ELP-Doxorubicin conjugates with shorter linkers. At pH 4, the release of Doxorubicin from the ELP-Doxorubicin conjugate with the shortest linker reached almost 80% over 72 h.


Cytotoxicity of ELP-Doxorubicin Conjugates.


An acid-labile ELP-Doxorubicin conjugate is tested for cytotoxicity in an in vitro cell culture assay with FaDu cells. The unconjugated ELP, the control conjugate, does not show any inherent cytotoxicity, and thus it indicates that ELPs are non-toxic despite of substantial internalization (FIG. 4). By contrast, the ELP-Doxorubicin conjugate shows substantial cytotoxicity during either 24 or 72 h, and the level of toxicity is similar to those of an equivalent Doxorubicin concentration.


Accumulation of ELPs in Solid Tumors.


Biodistribution studies are carried out by systematically injecting 14C-labeled ELP into nude mice bearing a FaDu solid carcinoma. The accumulation of the ELPs in implanted tumors is in the range of 10-20% injected dose per gram (% ID/g). When an ELP with a Tt of ˜40° C. is systematically injected into a mice and implanted tumors are heated to 42° C., the accumulation is ˜20% ID/g. By contrast, when the same ELP is injected without heating the tumors, the accumulation was ˜10% ID/g. This data shows that a significant concentration (% ID/g) of the radiolabeled ELP localized in the tumor even when the tumor is not heated. By contrast, the injection of a small radiolabeled molecule (molecular weight <500 Da) in unconjugated form results in significantly lower accumulation in the tumor. This example demonstrates that ELPs can result in significant localization in a diseased site.









TABLE 1







List of ELP-protein conjugates synthesized recombinantly (ELP fusion


proteins), molecular weight (MW) of the target proteins, and their yield


from a 1 Liter shaker flask culture of Escherichia coli.












MW
Yield



Target Proteins
(kDa)
(mg/L)















Angiostatin (K1-3)
30.7
27



Blue fluorescent protein (BFP)
26.9
100



Calmodulin (CalM)
16.7
75



Chloramphenicol acetyltransferase (CAT)
25.7
80



Green fluorescent protein (GFP)
26.9
78-1600



Interleukin 1 receptor antagonist (IL1rRa)
17.0
50



Luciferase
60.8
10



Tissue transglutaminase (tTg)
77.0
36



Tendamistat
7.9
22



Thioredoxin (Trx)
11.7
120

















TABLE 2







Yield of peptide-ELP conjugates synthesized recombinantly in E. coli.


Both yield of the conjugate (fusion) and the target peptide is


shown, as well as purity as determined by mass spectrometry.












MW
Yield Fusion
Yield Peptide



Peptide
(kDa)
(mg/L culture)
(mg/L culture)
Purity





Morphine Modulating
2.0
224
17
99%


Neuropeptide (MMN)


Neuropeptide Y (NPY)
2.7
222
20
98%


Orexin B
3.0
320
19
91%


Leptin
4.0
415
19
97%


ACTH
4.6
133
19
99%


Calcitonin
6.2
260
23
98%









The foregoing is illustrative of the present invention, and is not to be construed as limiting thereof. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims
  • 1. A method of administering to a subject a chemotherapeutical agent conjugated to an elastin-like peptide (ELP) via a linker at the C-terminus, wherein the chemotherapeutical agent is conjugated to the ELP, wherein the subject is not subjected to hyperthermia, wherein the conjugate is administered systemically, and is formulated for weekly administration.
  • 2. The method of claim 1, wherein at least one of: (i) the bioavailability of said active agent is greater; (ii) the half-life of said active agent is greater, (iii) the systemic toxicity of said active agent is less, in said subject when said active agent is administered to said subject in conjugated form as said conjugate as compared to the same amount of said active agent administered to said subject in the same way in unconjugated form.
  • 3. The method of claim 1, wherein said chemotherapeutical agent is selected from the group consisting of: (i) doxorubicin, (ii) paclitaxel, (iii) cis-platinum, and (iv) combrestatin.
  • 4. The method of claim 1, wherein said conjugate is administered to said subject in a treatment-effective amount.
  • 5. The method of claim 1, wherein said conjugate is administered to said subject by parenteral injection.
  • 6. The method of claim 1, wherein said conjugate is administered to said subject subcutaneously.
  • 7. The method of claim 1, wherein the chemotherapeutical agent and the ELP are chemically conjugated.
  • 8. The method of claim 1, wherein the chemotherapeutical agent and the ELP are enzymatically conjugated.
  • 9. The method of claim 1, wherein the linker is a pH-sensitive linker.
  • 10. The method of claim 1, wherein the subject has cancer.
  • 11. The method of claim 1, wherein the cancer is a solid cancer.
  • 12. The method of claim 11, wherein the solid cancer is selected from the group consisting of lung cancer, colon cancer, breast cancer, brain cancer, liver cancer, prostate cancer, spleen cancer, muscle cancer, ovarian cancer, pancreatic cancer, skin cancer, and melanoma.
  • 13. The method of claim 1, wherein the in vivo efficacy of said chemotherapeutical agent is enhanced in said subject compared to the same amount of said chemotherapeutical agent administered to said subject in unconjugated form.
PRIORITY

This application is a continuation of U.S. patent application Ser. No. 13/795,955, filed Mar. 12, 2013, which is a continuation-in-part of U.S. patent application Ser. No. 13/674,285, filed Nov. 12, 2012, which is a continuation of Ser. No. 12/158,190, filed Sep. 2, 2008 (now U.S. Pat. No. 8,334,257), which is a national phase application of PCT Application No. PCT/US2006/048572, filed Dec. 20, 2006, and published in English on Jun. 28, 2007, as International Publication No. WO 2007/073486, and which claims the benefit of U.S. Provisional Patent Application Ser. No. 60/751,896, filed Dec. 20, 2005, the disclosure of each of which is incorporated by reference herein in its entirety.

GOVERNMENT FUNDING

This invention was made with Government support under grant number EB00188 and GM-061232 from the National Institutes of Health. The US Government has certain rights to this invention.

US Referenced Citations (167)
Number Name Date Kind
4132746 Urry et al. Jan 1979 A
4187852 Urry et al. Feb 1980 A
4474851 Urry Oct 1984 A
4500700 Urry Feb 1985 A
4589882 Urry May 1986 A
4605641 Bolin et al. Aug 1986 A
4749647 Thomas et al. Jun 1988 A
4752638 Nowinski et al. Jun 1988 A
4783523 Urry et al. Nov 1988 A
4870055 Urry et al. Sep 1989 A
4898926 Urry Feb 1990 A
5147855 Gozes et al. Sep 1992 A
5234907 Bolin Aug 1993 A
5235041 Cappello et al. Aug 1993 A
5236904 Gerstenberg et al. Aug 1993 A
5243038 Ferrari et al. Sep 1993 A
5288514 Ellman Feb 1994 A
5445934 Fodor et al. Aug 1995 A
5447912 Gerstenberg et al. Sep 1995 A
5496712 Cappello et al. Mar 1996 A
5506120 Yamamoto et al. Apr 1996 A
5514581 Ferrari et al. May 1996 A
5519004 Urry May 1996 A
5520672 Urry May 1996 A
5527610 Urry Jun 1996 A
5545617 Dartt et al. Aug 1996 A
5624711 Sundberg et al. Apr 1997 A
5641648 Ferrari et al. Jun 1997 A
5646016 McCoy et al. Jul 1997 A
5681816 Korman Oct 1997 A
5700914 Jorgensen et al. Dec 1997 A
5702717 Cha et al. Dec 1997 A
5747646 Hakimi et al. May 1998 A
5770570 Sudhir et al. Jun 1998 A
5770697 Ferrari et al. Jun 1998 A
5773249 Cappello et al. Jun 1998 A
5816259 Rose Oct 1998 A
5830713 Ferrari et al. Nov 1998 A
5849535 Cunningham Dec 1998 A
5854387 Urry et al. Dec 1998 A
5900405 Urry May 1999 A
5958881 Korman Sep 1999 A
5972406 Urry et al. Oct 1999 A
5972883 Gozes et al. Oct 1999 A
5998588 Hoffman et al. Dec 1999 A
6004782 Daniell et al. Dec 1999 A
6018030 Ferrari et al. Jan 2000 A
6037321 Cox et al. Mar 2000 A
6063061 Wallace et al. May 2000 A
6140072 Ferrari et al. Oct 2000 A
6153655 Martinez et al. Nov 2000 A
6184348 Ferrari et al. Feb 2001 B1
6200598 Needham Mar 2001 B1
6238916 El Halawani May 2001 B1
6258562 Salfield et al. Jul 2001 B1
6328996 Urry Dec 2001 B1
6329209 Wagner et al. Dec 2001 B1
6355776 Ferrari et al. Mar 2002 B1
6380154 Cappello et al. Apr 2002 B1
6429188 Perez et al. Aug 2002 B1
6503534 Pellet Jan 2003 B1
6537521 Uzgiris Mar 2003 B2
6541033 Shah Apr 2003 B1
6582926 Chilkoti Jun 2003 B1
6583272 Bailon Jun 2003 B1
6593394 Li et al. Jul 2003 B1
6699294 Urry Mar 2004 B2
6852834 Chilkoti Feb 2005 B2
6998387 Goke et al. Feb 2006 B1
7084243 Glaesner et al. Aug 2006 B2
7094755 Burman et al. Aug 2006 B2
7101843 Glaesner et al. Sep 2006 B2
7138486 Habener et al. Nov 2006 B2
7141547 Rosen et al. Nov 2006 B2
7144863 DeFelippis et al. Dec 2006 B2
7176278 Prior Feb 2007 B2
7226910 Wilson et al. Jun 2007 B2
7232879 Galloway et al. Jun 2007 B2
7259233 Dodd et al. Aug 2007 B2
7271149 Glaesner et al. Sep 2007 B2
7332473 Onoue et al. Feb 2008 B2
7364859 Chilkoti Apr 2008 B2
7429458 Chilkoti Sep 2008 B2
7442680 Yong et al. Oct 2008 B2
7459441 Minagawa et al. Dec 2008 B2
7468353 Bevec Dec 2008 B2
7566691 Nestor Jul 2009 B2
7582608 Bokvist et al. Sep 2009 B2
7723472 Szoka May 2010 B2
7776815 Vanderby et al. Aug 2010 B2
8178495 Chilkoti May 2012 B2
8334257 Chilkoti Dec 2012 B2
8703717 Schellenberger et al. Apr 2014 B2
8729018 Chilkoti May 2014 B2
8841255 Chilkoti Sep 2014 B2
9029505 Sadeghi et al. May 2015 B2
9127047 Chilkoti Sep 2015 B2
9200083 Chilkoti Dec 2015 B2
9328154 Chilkoti May 2016 B2
9458218 Chilkoti Oct 2016 B2
9821036 Chilkoti et al. Nov 2017 B2
20010034050 Chilkoti Oct 2001 A1
20020081309 Pettit Jun 2002 A1
20020099003 Wilson et al. Jul 2002 A1
20020142964 Nissen et al. Oct 2002 A1
20020151458 Gomariz et al. Oct 2002 A1
20030059840 Chilkoti Mar 2003 A1
20030059841 Chilkoti Mar 2003 A1
20030199445 Knudsen et al. Oct 2003 A1
20030211094 Chilkoti Nov 2003 A1
20040053370 Glaesner Mar 2004 A1
20040063631 Block Apr 2004 A1
20040110296 Vargeese et al. Jun 2004 A1
20040234609 Collier et al. Nov 2004 A1
20040266993 Evans Dec 2004 A1
20050026826 Hoenig Feb 2005 A1
20050118109 Block et al. Jun 2005 A1
20050203009 Pan et al. Sep 2005 A1
20050249730 Goetsch et al. Nov 2005 A1
20050255554 Chilkoti Nov 2005 A1
20060019347 Cho et al. Jan 2006 A1
20060247156 Vanderby et al. Nov 2006 A1
20070009602 Setton Jan 2007 A1
20070031342 Tzannis et al. Feb 2007 A1
20070041934 William Feb 2007 A1
20080032400 Dagher Feb 2008 A1
20080085860 Bokvist et al. Apr 2008 A1
20080108573 Duggan May 2008 A1
20080207492 Polt et al. Aug 2008 A1
20080221041 Block et al. Sep 2008 A1
20080261863 Whelan et al. Oct 2008 A1
20080274961 Bevec Nov 2008 A1
20080318865 Juul-Mortensen Dec 2008 A1
20090004104 Chilkoti Jan 2009 A1
20090005315 Duggan Jan 2009 A1
20090092582 Bogin et al. Apr 2009 A1
20090175821 Bridon et al. Jul 2009 A1
20090220455 Chilkoti et al. Sep 2009 A1
20090227493 Nakashima et al. Sep 2009 A1
20090270317 Chilkoti Oct 2009 A1
20100016212 Rubin et al. Jan 2010 A1
20100022455 Chilkoti Jan 2010 A1
20100184651 Maithal et al. Jul 2010 A1
20100189643 Chilkoti et al. Jul 2010 A1
20100278918 Cappola et al. Nov 2010 A1
20110039776 Chilkoti Feb 2011 A1
20110123487 Chilkoti May 2011 A1
20110178017 Sadeghi et al. Jul 2011 A1
20110236384 Setton et al. Sep 2011 A1
20130005664 Chilkoti Jan 2013 A1
20130079277 Chilkoti Mar 2013 A1
20130085099 Chilkoti Apr 2013 A1
20130085104 Chilkoti Apr 2013 A1
20130143802 Chilkoti Jun 2013 A1
20130150291 Jowett et al. Jun 2013 A1
20130172274 Chilkoti Jul 2013 A1
20130178411 Chilkoti Jul 2013 A1
20130178416 Chilkoti Jul 2013 A1
20130310538 Chilkoti Nov 2013 A1
20140024600 Chilkoti et al. Jan 2014 A1
20140171370 Arnold et al. Jun 2014 A1
20140213516 Chilkoti Jul 2014 A1
20140364371 Setton et al. Dec 2014 A1
20150080306 Chilkoti Mar 2015 A1
20150111829 Georgopoulos et al. Apr 2015 A1
20160030521 Chilkoti Feb 2016 A1
20160120952 Chilkoti May 2016 A1
Foreign Referenced Citations (48)
Number Date Country
101868476 Oct 2010 CN
102131516 Jul 2011 CN
0449592 Nov 1994 EP
0978565 Oct 2006 EP
2307038 Dec 2009 EP
S568355 Jan 1981 JP
6-206898 Jul 1994 JP
2001-514263 Sep 2001 JP
2004-528014 Sep 2004 JP
2005-508360 Mar 2005 JP
2005-508895 Apr 2005 JP
2006-503588 Feb 2006 JP
2008-521893 Jun 2006 JP
2010-502734 Jan 2010 JP
WO 198606492 Nov 1986 WO
WO 1990001024 Feb 1990 WO
WO 199632406 Oct 1996 WO
WO 199911661 Mar 1999 WO
WO 1999038984 May 1999 WO
WO 200056774 Sep 2000 WO
WO 2002066511 Aug 2002 WO
WO 2001079271 Oct 2002 WO
WO 2003041493 Dec 2003 WO
WO 2003099835 Dec 2003 WO
WO 2004020454 Mar 2004 WO
WO 2004104020 Dec 2004 WO
WO 2005000892 Jan 2005 WO
WO 2006001806 Jan 2006 WO
WO 2006059939 Jun 2006 WO
WO 2006078629 Jul 2006 WO
WO 2006110292 Oct 2006 WO
WO 2007002362 Jan 2007 WO
WO 2007024427 Mar 2007 WO
WO 2007039140 Apr 2007 WO
WO 2007073486 Jun 2007 WO
WO 2007100535 Sep 2007 WO
WO 2007103515 Sep 2007 WO
WO 2007104493 Sep 2007 WO
WO 2008028117 Mar 2008 WO
WO 2008030968 Mar 2008 WO
WO 2008141786 Nov 2008 WO
WO 2008155134 Dec 2008 WO
WO 2009059278 May 2009 WO
WO 2009158704 Dec 2009 WO
WO 2010042997 Apr 2010 WO
WO 2010080578 Jul 2010 WO
WO 2011020091 Feb 2011 WO
WO 2013028989 Feb 2013 WO
Non-Patent Literature Citations (101)
Entry
Juan et al. (“Low-dose Weekly Paclitaxel as Second-line Treatment for Advanced Non-small Cell Lung Cancer: a Phase II Study,” Jpn J Clin Oncol 2002; 32(11) 449-454).
Green et al. (“Weekly Paclitaxel Improves Pathologic Complete Remission in Operable Breast Cancer When Compared With Paclitaxel Once Every 3 Weeks,” J Clinical Oncology 2005; 23(25) 5983-5992).
Eniu et al. (“Weekly Administration of Docetaxel and Paclitaxel in Metastatic or Advanced Breast Cancer,” The Oncologist 2005;10 665-685).
Markman et al. (“Phase II Trial of Weekly Single-Agent Paclitaxel in Platinum/Paclitaxel-Refractory Ovarian Cancer,” J Clinical Oncology, 2002; 20(9) 2365-2369).
Torti et al. (“Reduced Cardiotoxicity of Doxorubicin Delivered on a Weekly Schedule: Assessment by Endomyocardial,” Biopsy Ann Intern Med. 1983; 99(6):745-749) (Year: 1983).
Furgeson et al. (“Structural optimization of a “smart” doxorubicin-polypeptide conjugate for thermally targeted delivery to solid tumors,” Journal of Controlled Release 110 (2006) 362-369; Available online Nov. 21, 2005) (Year: 2006).
Dreher et al. (“E valuation of an elastin-like polypeptide-doxorubicin conjugate for cancer therapy,” Journal of Controlled Release 91 (2003) 31-43) (Year: 2003).
Nielson et al. (“Therapeutic efficacy of anti-ErbB2 immunoliposomes targeted by a phage antibody selected for cellular endocytosis,” Biochimica et Biophysica Acta 1591 (2002) 109-118) (Year: 2002).
AntiThrombin III Deficiency < http://emedicine.medscape.com/article/954688-overview >downloaded Jan. 6, 2010, 2 pages.
Clotting Factor Definition < http://medical-dictionary.thefreedictionary.com/Ciotting+factors >—downloaded Jan. 6, 2012, 3 pages.
“What are Rare Clotting Factor Deficiencies?”, published by World Federation of Hemophilia, 2009, pp. 1-29.
Baldwin, Michael A., “Mass spectrometers for the analysis of biomolecules.” Methods in Enzymology, 402: 3-48 (2005).
Delgado et al., “The Significance of Vasoactive Intestinal Peptide in Immunomodulation,” Pharm. Rev. 56(2):249-290 (2004).
Elastin, Accession No. AAC98394, submitted to GenBank Mar. 8, 2002, 2 pages.
EP Application No. 07814724.6, Extended European Search Report dated Mar. 4, 2010, 6 pages.
EP Application No. 12826427.2, Extended European Search Report dated Dec. 9, 2014, 6 pages.
He, X., “Scandium and Albumin”—Definition http://www.springerreference.com/docs/htm1/chapterdbid/358272.html downloaded Apr. 30, 2013, 2 pages.
Holleman, Frits. Regular human insulin [internet]. Aug. 13, 2014; Diapedia 81040961114 rev. No. 9. Available from: http://dx.doi.org/1 0.14496/dia.81 040961114.9—Retrieved Aug. 24, 2015, 3 pages.
Ibrahim, Hiba, et al., “Transcriptional modulation by VIP: a rational target against inflammatory disease,” Clinical Epigenetics, 2(2) : 213 (2011).
Lichtenauer, Michael, et al., “Phosphate buffered saline containing calcium and magnesium elicits increased secretion of interleukin-1 receptor antagonist,” Laboratory Medicine, 40(5): 290-293 (2009).
PCT/US2007/077767, International Search Report and Written Opinion, dated Aug. 11, 2008, 12 pages.
PCT/US2007/077767, International Preliminary Report on Patentability, dated Mar. 10, 2009, 7 pages.
PCT/US2006/048572, International Preliminary Report on Patentability, dated Jun. 24, 2008, 4 pages.
PCT/US2009/049059, International Preliminary Report on Patentability, dated Jan. 5, 2011, 6 pages.
PCT/US2012/052304, International Search Report and Written Opinion, mailed Nov. 27, 2012, 10 pages.
PCT/US2012/052304, International Preliminary Report on Patentability, dated Feb. 25, 2014, 6 pages.
Protocols online entry for PBS, http://protocolsonline.com/recipes/phosphate-buffered-saline-pbs/, updated Jul. 16, 2012, 4 pages.
Rippe, Bengt, et al., “Plasma volume, blood volume and transcapillary escape rate (TER) of albumin in young spontaneously hypertensive rats (SHR) as compared with normotensive controls (NCR).” Clinical and Experimental Hypertension, 1(1): 39-50 (1978).
Shimazu, et al, “Thermally triggered purification and immobilization of elastin-OPH fusions”, Biotechnol Bioeng., 81(1): 74-79 (2003).
Sojikul et al., “A plant signal peptide-hepatitis B surface antigen fusion protein with enhanced stability and immunogenicity expressed in plant cells”, PNAS, vol. 100 No. 5 pp. 2209-2214 (2003).
Wilmot and Thornton, “Analysis and prediction of the different types of β-turn in proteins”, J. Mol. Biol., 203(1): 221-232 (1988).
Yasuda, H., et al., “Expression of the small peptide GLP-1 in transgenic plants”, Transgenic Res., 14(5): 677-684 (2005).
Baggio et al., “A Recombinant Human Glucagon-Like Peptide (GLP)-1-Albumin Protein (Albugon) Mimics Peptidergic Activation of GLP-1 Receptor-Dependent Pathways Coupled with Satiety, Gastrointestinal Motility, and Glucose Homeostasis,” Diabetes 53:2492-2500 (2004).
Bidwell et al., “Application of thermally responsive polypeptides directed against c-Myc transcriptional function for cancer therapy,” Mol. Cancer Ther. 4(7):1076-1085 (2005).
Chang et al., “Role of Disulfide Bonds in the Structure and Activity of Human Insulin,” Mol. Cells. 16(3):323-330 (2003).
Chen et al., “A new temperature-and pH-responsive copolymer for possible use protein conjugation,” Macromol. Chem. Phys. 196:1251-1259 (1995).
Chen, J.P., et al., “Polymer-protein conjugates, II. Affinity precipitation separation of human immunogammaglobulin by a poly(N-isopropylacrylamide)-protein A conjugate,” Biomaterials, 11:631-634 (1990).
Chilkoti et al., “Targeted drug delivery by thermally responsive polymers,” Adv. Drug Deliv. Rev. 54:613-630 (2002).
Chilkoti, A., “Biomedical Applications of Genetically Encoded Elastin Biopolymers,” Abstracts of Papers, 222nd ACS National Meeting, Chicago, IL, US, Aug. 26-30, 2001, MACR-019, see abstract.
Chilkoti, A., et al., “Site-Specific Conjugation of a Temperature-Sensitive Polymer to a Genetically-Engineered Protein,” Bioconjugate Chemistry, vol. 5, pp. 504-507, (1994).
Chow et al., “Ultra-High Expression of a Thermally Responsive Recombinant Fusion Protein in E. coli,” Biotechnol. Prog. 22:638-646 (2006).
Domschke et al., “Vasoactive intestinal peptide in man: pharmacokinetics, metabolic and circulatory effects,” Gut 19:1049-1053 (1978).
Dreher et al., “Evaluation of an elastin-like polypeptide-doxorubicin conjugate for cancer therapy,” J. Controlled Release 91:31-43 (2003).
Dreher et al., “Nitroxide conjugate of a thermally responsive elastin-like polypeptide for noninvasive thermometry,” 31(10):2755-2762 (2004).
Estall and Drucker, “Glucagon and Glucagon-Like Peptide REceptors as Drug Targets,” Curr. Pharm. Design 12(14):1731-1750 (2006).
European Search Report issued for the application serial No. EP 15163709.7 and dated Jun. 17, 2015 (8 pages).
Furgeson et al., “Structural optimization of a “smart” doxorubicin-polypeptide conjugate for thermally targeted delivery to solid tumors,” J. Control. Rel. 110(2):362-369 (2005).
Hattori et al. “Intravenous Administration of Thioredoxing Decreases Brain Damage Following transient Focal Cerebral Ischemia in Mice” Antioxidants & Redox Signaling. 6(1) (2004).
Henning and Sawmiller, “Vasoactive intestinal peptide: cardiovascular effect,” Cardiovascular Res. 49:27-37 (2001).
Hoffman, A.S., “Applications of Thermally Reversible Polymers and Hydrogels in Therapeutics and Diagnostics,” Journal of Controlled Release, 6, pp. 297-305, (1987).
Hui et al., “Structure and function studies of glucagon-like peptide-1 (GLP-1): the designing of a novel pharmacological agent for the treatment of diabetes,” Diabetes/Metab. Res. Rev. 21(4):313-331 (2005).
Hyun et al., “Capture and Release of Protein on the Nanoscale by Stimuli-Responsive Elastin-Like Polypeptide “Switches”,” J. Am. Chem. Soc. 126(23):7330-7335 (2004).
International Search Report and Written Opinion, PCT/US06/48572, dated May 9, 2008.
Kim, Jin-Soo et al., “Ribonuclease S-peptide as a carrier in fusion proteins,” Protein Science, 2:348-356, (1993).
Kobatake, Eiry et al., “Design and Gene Engineering Synthesis of an Extremely Thermostable Protein with Biological Activity,” Biomacromolecules 2000, 1:382-386.
MacArthur and Thornton, “Influence of Proline Residues on Protein Conformation,” J. Mol. Biol. 218:397-412 (1991).
McPherson et al., “Product Purification by Reversible Phase Transition Following Escherichia coli Expression of Genes Encoding up to 251 Repeats of the Elastomeric Pentapeptide GVGVP,” Protein Expression and Purification, 7, pp. 51-57, (1996).
McPherson, D. et al., “Production and purification of a recombinant elastomeric polypeptide, G-(VPGVG)19-VPGV from Eschericia coli,” Biotechnol. Prog., 8:347-352 (1992).
Meyer et al. “Purification of Recombinant Proteins by Fusion with Thermally-Responsive Polypeptides,” Nat. Biotechnol. 17:1112-1115 (1999).
Meyer et al., “Polypeptide Fusion Tag for Thermal Purification of Recombinant Proteins,” Abstracts of Papers, 217th ACS National Meeting, Anaheim, CA, US, Mar. 21-25, 1999, BIOT-078, see abstract.
Meyer et al., “Protein Purification by Fusion with an Environmentally Responsive Elastin-Like Polypeptide: Effect of Polypeptide Length on the Purification of Thioredoxin,” Biotechnol. Prog. 17:720-728 (2001).
Meyer, Dan E. et al., “Drug targeting using thermally responsive polymers and local hyperthermia,” Journal of Controlled release, Jul. 6, 2001; 74:213-224.
Meyer, Dan E. et al., “Targeting a Genetically Engineered Elastin-like Polypeptide to Solid Tumors by Local Hypothermia,” Cancer Res., Feb. 15, 2001; 61(4): 1548-1554.
Mirmira et al., “Importance of the Character and Configuration of Residues B24, B25, and B26 in Insulin-Receptor Interactions,” J. Biol. Chem. 266(3):1428-1436 (1991).
Nath and Chilkoti, “Interfacial Phase Transition of an Environmentally Responsive Elastin Biopolymer Adsorbed on Functionalized Gold Nanoparticles Studied by Colloidal Surface Plasmon Resonance,” J. Am. Chem. Soc. 123(34):8197-8202 (2001).
Ngo et al., “Computational Complexity, Protein Structure Prediction, and the Levinthal Paradox,” Chapter 14, pp. 491-495, in The Protein Folding Problem and Tertiary Structure Prediction, Marz, Jr. and Birkhauser, Boston (1994).
Nilsson, B. et al., “Fusion proteins in biotechnology and structural biology,” Curr. Opin. Struct. Biol., 2:569-575 (1992).
Nilsson, J. et al., “Affinity Fusion Strategies for Detection, Purification and Immobilization of Recombinant Proteins,” Protein Expression and Purification, 11:1-16 (1997).
Önyüksel et al., “Human VIP-α: A long-acting, biocompatible and biodegradable peptide nanomedicine for essential hypertension,” Peptides 27:2271-2275 (2006).
Önyüksel et al., “A Novel Formulation of VIP in Sterically Stabilized Micelles Amplifies Vasodilation In Vivo,” Pharmaceutical Research 16(1):155-160 (1999).
Richter et al. “The impact of Reducing Dose Frequency on Health Outcomes” Clinical Therapeutics. 25(8):2307-2335 (2003).
Rubinstein et al., “Intratracheal and subcutaneous liposomal VIP normalizes arterial pressure in spontaneously hypertensive hamsters,” International Journal of Pharmaceutics 316:144-147 (2006).
Santi et al., “Predictable and tunable half-life extension of therapeutic agents by controlled chemical release from macromolecular conjugates,” Proc. Natl. Acad. Sci. USA 109(16):6211-6216 (2012).
Sejourne et al., “Development of a Novel Bioactive Formulation of Vasoactive Intestinal Peptide in Sterically Stabilized Liposomes,” Pharmaceutical Research 14(3):362-365 (1997).
Sharma and Sharma, “Liposomes in drug delivery: progress and limitations,” Int. J. Pharm. 154:123-140 (1997).
Simoncsits et al., “Synthesis, cloning and expression in Escherichia coli of artificial genes coding for biologically active elongated precursors of the vasoactive intestinal polypeptide,” Eur. J. Biochem. 178:343-350 (1988).
Supplementary European Search Report, EP Appl. No. 06848713.1, 10 pages dated Nov. 26, 2012.
Supplementary European Search Report, EP Appl. No. 09771235.0, 6 pages dated Feb. 21, 2013.
Suzuki, et al., “Encapsulation of VIP into liposomes restores vasorelaxation in hypertension in situ,” Am. J. Physiol. 271(40):H282-H287 (1996).
Urry, D.W. et al., “Phase-structure Transitions of the Elastin Polypentapeptide-water system within the framework of composition-temperature studies,” Biopolymers, 24:2345-2346 (1985).
Urry, D.W. et al., “Temperature of Polypeptide Inverse Temperature Transition Depends on Mean Residue Hydrophobicity,” J. Am. Checm. Soc., 113:4346-4348 (1991).
Urry, D.W., “Entropic Elastic Processes in Protein Mechanisms, I. Elastic Structure Due to an Inverse Temperature Transition and Elasticity Due to Internal Chain Dynamics,” Journal of Protein Chemistry, vol. 7, No. 1, pp. 1-34 (1988).
Urry, D.W., “Free Energy Transduction in Polypeptides and Proteins Based on Inverse Temperature Transitions,” Prog. Biophys. Molec. Biol., vol. 57, pp. 23-57, (1992).
Urry, D.W., “Physical Chemistry of Biological Free Energy Transduction as Demonstrated by Elastic Protein-Based Polymers,” J. Phys. Chem. B., vol. 101, No. 51, pp. 11007-11028, (1997).
Walsh, “Therapeutic insulins and their large-scale manufacture,” Appl. Microbiol. Biotechnol. 67:151-159 (2005).
Wells, “Additivity of Mutational Effects in Proteins,” Biochem. 29(37):8509-8517 (1990).
Written Opinion of the International Searching Authority, PCT appl. No. PCT/US2006/048572 dated May 9, 2008.
Written Opinion of the International Searching Authority, PCT appl. No. PCT/US2009/049059, 5 pages dated Nov. 13, 2009.
International Search Report of the International Searching Authority, PCT appl. No. PCT/US2009/049059, 4 pages dated Nov. 13, 2009.
Chinese Patent Application No. 201280052426.0, Search Report (English translation) dated Jul. 6, 2016, 2 pages.
Derewenda, U., et al. “Molecular structure of insulin: the insulin monomer and its assembly.” British Medical Bulletin (1989); : 4-18.
Doyle et al., “The importance of the nine-amino acid C-terminal sequence of exendin-4 for binding to the GLP-1 receptor and for biological activity.” Regulatory Peptides (2003); 114(2-3): 153-158.
Gardasil (human papillomavirus-9 vaccine) package insert, downloaded Nov. 9, 2016, 23 pages.
Herceptin (trastuzmab) package insert, downloaded Nov. 9, 2016, 33 pages.
Izutsu, Ken-ichi, “Stabilization of therapeutic proteins by chemical and physical methods.” Methods Mol. Biol. (2005); 308: 287-292.
NovoSeven (eptacog alfa) package insert, downloaded Nov. 9, 2016, 11 pages.
Park, Ji-Eun and Won, Jong-In, “Thermal behviors of elastin-like polypeptides (elps) acording ot their physical proeprties and environmental conditions.” Biotech. Bioprocess. Eng. (2009); 14: 662-667.
Recombinate (antihemophilic factor, recombinant) package insert, downloaded Nov. 9, 2016, 24 pages.
The website for REACH devices, http://www.reachdevices.com/Protein/BiologicalBuffers.html, downloaded Nov. 9, 2016, 7 pages.
Xynthia (antihemophelic factor) package insert, downloaded Nov. 9, 2016, 29 pages.
Bhadra, D., et al. “Pegnology: a review of PEG-ylated systems.” Die Pharmazie (2002); 57.1: 5-29.
Related Publications (1)
Number Date Country
20160114053 A1 Apr 2016 US
Provisional Applications (1)
Number Date Country
60751896 Dec 2005 US
Continuations (2)
Number Date Country
Parent 13795955 Mar 2013 US
Child 14939225 US
Parent 12158190 US
Child 13674285 US
Continuation in Parts (1)
Number Date Country
Parent 13674285 Nov 2012 US
Child 13795955 US