WO2009015345A1 - Pharmaceutical compositions comprising fc fusion proteins - Google Patents

Pharmaceutical compositions comprising fc fusion proteins Download PDF

Info

Publication number
WO2009015345A1
WO2009015345A1 PCT/US2008/071209 US2008071209W WO2009015345A1 WO 2009015345 A1 WO2009015345 A1 WO 2009015345A1 US 2008071209 W US2008071209 W US 2008071209W WO 2009015345 A1 WO2009015345 A1 WO 2009015345A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
formulation
proteins
active peptide
antibody
Prior art date
Application number
PCT/US2008/071209
Other languages
French (fr)
Inventor
Michael Treuheit
Gayathri Ratnaswamy
Original Assignee
Amgen 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 Amgen Inc. filed Critical Amgen Inc.
Publication of WO2009015345A1 publication Critical patent/WO2009015345A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • 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
    • A61K47/68Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • Embodiments of the invention provide, among other things, protein formulations, comprising a peptibody comprised of an Fc region and an active peptide, wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
  • the protein is a pharmaceutically active protein.
  • the formulation is a stable pharmaceutical formulation.
  • the formulation further comprises one or more pharmaceutically acceptable: osmotic balancing agents; anti-oxidants; antibiotics; antimycotics; bulking agents; lyoprotectants; anti-foaming agents; chelating agents; preservatives; colorants; analgesics; or additional pharmaceutical agents.
  • a formulation comprising a protein comprised of an active peptide, wherein the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
  • Disorder(s) a malediction, a condition that deleteriously alters health.
  • “Dysfunction” means, as used herein, a disorder, disease, or deleterious effect of an otherwise normal process.
  • Ligand-binding moiety(s) herein means a molecular entity that binds a ligand, typically, a part of a larger molecular entity that binds the ligand, or a molecular entity derived therefrom.
  • Modified protein(s),” "modified polypeptide(s),” or “modified fragment(s)” herein means a protein or a polypeptide or a fragment of a protein or polypeptide comprising a chemical moiety (structure) other than those of the twenty naturally occurring amino acids that form naturally occurring proteins. Modifications most often are covalently attached, but can also be attached non-covalently to a protein or other polypeptide, such as a fragment of a protein.
  • “Moiety(s)” herein means a molecular entity that embodies a specific structure and/or function, without extraneous components. For instance, in most cases, only a small part of a ligand-binding protein is responsible for ligand binding. This part of the protein, whether continuously encoded or discontinuously, is an example of a ligand-binding moiety.
  • Peptibody refers to a molecule comprising an antibody Fc domain (i.e., C H 2 and C H 3 antibody domains) that excludes antibody CHI , CL, VH, and VL domains as well as Fab and F(ab)2, wherein the Fc domain is attached to one or more peptides, preferably a pharmacologically active peptide, particularly preferably a randomly generated pharmacologically active peptide.
  • Fc domain i.e., C H 2 and C H 3 antibody domains
  • “Pharmaceutical formulation(s)” means the same as “pharmaceutical composition(s)” as the term is defined herein above. "Pharmaceutically acceptable” is used herein in accordance with its well-known meaning in the art to denote that which is acceptable for medical or veterinary use, preferably for medical use in humans, particularly approved for such use by the U.S. Food and Drug Administration or other authority as described above regarding the meaning of "pharmaceutical.”
  • OX40 receptor expressed on T cells and other ligands listed herein;
  • illustrative proteins are certain antibody and antibody-related proteins, including Fc fusion protein and peptibodies, such as, for instance, those listed immediately below and elsewhere herein and other fusion proteins comprising an Fc region or a fragment or derivative thereof: OPGL specific antibodies, peptibodies, and related proteins, and the like (also referred to as RANKL specific antibodies, peptibodies and the like), including fully humanized and human OPGL specific antibodies, particularly fully humanized monoclonal antibodies, including but not limited to the antibodies described in International Publication Number WO 03/002713, which is incorporated herein in its entirety as to OPGL specific antibodies and antibody related proteins, particularly those having the sequences set forth therein, particularly, but not limited to, those denoted therein: 9H7; 18B2; 2D8; 2El 1; 16El; and 22B3, including the OPGL specific antibodies having either the light chain of SEQ ID NO: 2 as set forth therein in Figure 2 and/or the heavy chain of S
  • Betaseron® Interferon-beta
  • Campath® Almtuzumab, anti-CD52 monoclonal antibody
  • Agents that may be used in formulating protein compositions include, but are not limited to, acetate, citrate, succinate, phosphate, certain amino acids, "Tris,” Good's buffers (MES, ADA, PIPES, ACES, Cholamine chloride, BES, TES, HEPES, Acetamidoglycine, Tricine, Glycinamide and Bicine), certain other organic acids, and certain polyanionic carboxylate buffers (including the aforementioned citrate and succinate).
  • Equally important is to ensure that the buffer is compatible with the protein drug, other formulation excipients, and does not catalyze any degradation reactions. It is also important in pharmaceutical compositions to consider the possibility that a given buffering agent will be unacceptable for administration for ancillary reasons, such as deleterious effects on patient comfort. Some buffering agents are unsuitable for this purpose because they cause stinging or irritation at the point of administration. For example, citrate is known to cause stinging upon injection. Such effects are more pronounced for SC and IM administration, because the formulation remains at the administration site for some time, than for IV administration, where the formulation is diluted immediately. For IV administration, however, the total load of buffering agent administered must be assessed, because the buffering agent itself can be harmful to the patient.
  • formulations in accordance with the invention may be administered by any suitable route, including those well-known to those skilled in the art of administering therapeutics to a subject.
  • routes in a variety of embodiments include but are not limited to administration of the compositions orally, ocularly, mucosally, topically, rectally, pulmonarily, such as by inhalation spray, and epicutaneously.
  • Parenteral routes of administration are useful in embodiments of the invention, including administration by intravenous, intraarterial, intracardiac, intraspinal, intrathecal, intraosseous, intraarticular, intrasynovial, intracutaneous, intradermal, subcutaneous, peritoneal, and/or intramuscular injection.
  • intravenous, intraarterial, intracutaneous, intradermal, subcutaneous and/or intramuscular injection are used.
  • intravenous, intraarterial, intracutaneous, subcutaneous, and/or intramuscular injection are used.

Abstract

The invention herein described provides, among other things: methods for making protein formulations based on isoelectric points; protein formulations made by the methods; and uses of the methods and of the protein formulations.

Description

PHARMACEUTICAL. COMPOSITIONS COMPRISING FC FUSION PROTEINS
This application claims the benefit of U.S. Provisional Application No. 60/935,073 filed July 25, 2007, which is herein incorporated by reference in its entirety. Field of the Invention
The invention relates to the preparation and use of proteins, particularly for therapeutic applications. It relates to systematic methods for formulating such proteins based on pi, to the formulations thus obtained, and to the uses thereof. Among such proteins are antibodies and Fc fusion proteins, including, for instance, peptibodies. Background of the Invention
Recombinant DNA based methods developed over the past two decades have made it economically possible to produce peptides and proteins with the quality and in the quantities required for therapeutic use. The capability engendered a still on-going revolution in the treatment of disease using biopharmaceutical agents. The increasing use of peptide and protein pharmaceutical agents poses a challenge to formulations scientists, however. Peptides and proteins generally are more fragile than the small organic molecules that, historically, have been the predominant concern of pharmaceutical scientists formulating agents for therapeutic use. They are also more complicated, chemically, typically less soluble, and often are difficult to formulate at concentrations necessary for therapeutic efficacy. They are subject to processes that result in aggregation, agglomeration, adhesion, and denaturation that do not typically affect small organic molecule therapeutic agents, particularly during storage. As yet, there are few systematic methods for predicting effective formulations for peptide and protein pharmaceuticals, and it can take considerable ingenuity and effort to determine a pharmaceutically effective formulation for a given peptide or protein therapeutic agent. Many aspects of pharmaceutical production and formulation processes are pH sensitive. The conformation and activity of proteins are critically dependent upon pH. Proteins are susceptible to a variety of pH sensitive reactions that are deleterious to their efficacy, typically many more than affect small molecule drugs. For instance, to mention just a few salient examples, the side chain amides of glutamine are deamidated at low pH (less than 4.0) and the side chains of asparagine are deamidated at high pH (greater than 6.0). Aspartic acid residues promote the hydrolysis of adjacent peptide bonds at low pH. The stability and disposition of disulfide bonds is highly dependent on pH, particularly in the presence of thiols. Solubility, flocculation, aggregation, precipitation, and fibrillation of proteins are critically dependent on pH. And pH is an important factor in surface adsorption of many pharmaceutical peptides and proteins. Determining the correct pH for a pharmaceutical product is critical to its stability, effectiveness, and shelf life. pH is an important consideration in designing formulations for administration that will be acceptable, as well as safe and effective.
To establish and maintain pH, pharmaceutical processes and formulations use one or more buffering agents. A variety of buffering agents are available for pharmaceutical use. The buffer or buffers for a given application must be effective at the desired pH. They must also provide sufficient buffer capacity to maintain the desired pH for as long as necessary. A good buffer for a pharmaceutical composition must satisfy numerous other requirements as well. It must be appropriately soluble. It must not form deleterious complexes with metal ions, be toxic, or unduly penetrate, solubilize, or adsorb on membranes or other surfaces. It should not interact with other components of the composition in any manner which decreases their availability or effectiveness. It must be stable and effective at maintaining pH over the range of conditions to which it will be exposed during formulation and during storage of the product. It must not be deleteriously affected by oxidation or other reactions occurring in its environment, such as those that occur in the processing of the composition in which it is providing the buffering action. If carried over or incorporated into a final product, a buffering agent must be safe for administration, compatible with other components of the composition over the shelf-life of the product, and acceptable for administration to the end user.
Knowing the proper pH thus is an important aspect of formulating proteins. Although it would be useful to know the proper pH for a given formulation at the beginning of the formulation process, there has not been, as yet, a reliable method for predicting the appropriate range of pH or the optimum pH for formulating proteins. As a result, formulating proteins has involved a considerable amount of work on a trial and error, hit and miss basis. The prior art thus does not provide methods for predicting the best pH range for a given protein formulation, particularly Fc fusion protein formulations, especially, for instance, peptibody formulations.
Summary Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an active peptide such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an active peptide such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 4.0 or less. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an Fc region and an active peptide such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an Fc region and an active peptide such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, wherein the protein is a peptibody comprising an Fc region and an active peptide such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, wherein the protein is a peptibody comprising an Fc region and an active peptide such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an active peptide based on the pi of the active peptide, such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an active peptide based on the pi of the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of protein is 6.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition. Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an Fc region and an active peptide based on the pi of the active peptide, such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an Fc region and an active peptide based on the pi of the protein, such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, wherein the protein is a peptibody comprising an Fc region and an active peptide, based on the pi of the active peptide, such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, wherein the protein is a peptibody comprising an Fc region and an active peptide, based on the pi of the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. In certain aspects and embodiments the formulation is a pharmaceutically acceptable protein composition. Embodiments of the invention provide, among other things, methods for formulating a protein, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the protein is determined by a predictive method.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising: (A) predicting the pi of the active peptide, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an active peptide based on the pi of the protein, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an Fc region and an active peptide, comprising: (A) predicting the pi of the active peptide, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method. Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a protein comprising an Fc region and active peptide based on the pi of the protein, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a peptibody comprising an active peptide based, comprising: (A) predicting the pi of the active peptide, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method.
Embodiments of the invention provide, among other things, methods for producing a protein formulation, comprising formulating a peptibody comprising an active peptide based on the pi of the protein, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the invention provides methods for formulating stable pharmaceutically acceptable protein compositions. In certain aspects and embodiments the pi of the active peptide is determined by a predictive method.
Embodiments of the invention provide, among other things, protein formulations comprising a protein, wherein the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active agent. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation. Embodiments of the invention provide, among other things, protein formulations comprising a protein comprised of an active peptide, wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation.
Embodiments of the invention provide, among other things, protein formulations comprising a protein comprised of an active peptide, wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation.
Embodiments of the invention provide, among other things, protein formulations, comprising a protein comprised of an Fc region and an active peptide, wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation.
Embodiments of the invention provide, among other things, protein formulations comprising a protein comprised of an Fc region and an active peptide, wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation.
Embodiments of the invention provide, among other things, protein formulations, comprising a peptibody comprised of an Fc region and an active peptide, wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation. Embodiments of the invention provide, among other things, protein formulations, wherein the protein is a peptibody comprising an Fc region and an active peptide, and wherein the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In certain aspects and embodiments the protein is a pharmaceutically active protein. In certain aspects and embodiments the formulation is a stable pharmaceutical formulation.
The following numbered paragraphs are illustrative of various aspects and embodiments of the invention herein; but, taken alone or in any combination they are in no way exhaustive or otherwise limitative of the many and diverse aspects, features and embodiments of the invention. The phrase used below, "any of the foregoing or the following" refers to any of the foregoing or the following numbered paragraphs. The subject matter is set out in the paragraphs in this manner to indicate that some or all of the subject matter of any one or more paragraphs may be combined with some or all of the subject matter of any one or more other paragraphs. These paragraphs are set out particularly to provide a written description of all the subject matter that may be recited not only by the paragraphs taken alone but by any combination of any part or all of any one or more of them. Applicant specifically reserves the right to set forth such claims as may thereby be composed, and to set out in full the subject matter described by reference to any such combination should the same be desirable or advantageous in prosecuting any patent or other intellectual property rights on the subject matter of this application.
1. A method for formulating a protein comprising an active peptide, comprising: formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
2. A method for formulating a protein comprising an active peptide, comprising: formulating the protein such that the pH of the formulation is (i) 4.5 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less.
3. A method for formulating a protein comprising an Fc region and an active peptide, comprising: formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
4. A method for formulating a protein comprising an Fc region and an active peptide, comprising: formulating the protein such that the pH of the formulation is (i) 4.5 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. 5. A method for formulating a protein comprising an active peptide, comprising: formulating the protein based on its pi such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less. 6. A method for formulating a protein comprising an active peptide, comprising: formulating the protein based on its pi such that the pH of the formulation is (i) 4.5 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less.
7. A method for formulating a protein comprising an Fc region and an active peptide, comprising: formulating the protein based on its pi such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
8. A method for formulating a protein comprising an Fc region and an active peptide, comprising: formulating the protein based on its pi such that the pH of the formulation is (i) 4.5 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less.
9. A method for formulating a protein, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less
10. A method for formulating a protein comprising an active peptide, comprising: (A) predicting the pi of the active peptide, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
11. A method for formulating a protein comprising an active peptide, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less.
12. A method for formulating a protein comprising an Fc region and an active peptide, comprising: (A) predicting the pi of the active peptide, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
13. A method for formulating a protein comprising an Fc region and an active peptide, comprising: (A) predicting the pi of the protein, and (B) formulating the protein such that the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less.
14. A method according to any of the foregoing or the following, wherein the pi of the protein is 6.0 or more and the pH of the formulation is 4.6 to 5.6. 15. A method according to any of the foregoing or the following, wherein the pi of the protein is 6.0 or less and the pH of the formulation is 7.0 to 8.0.
16. A method according to any of the foregoing or the following, wherein the pi of the active peptide is 4.0 or more and the pH of the formulation is 4.6 to 5.6.
17. A method according to any of the foregoing of the following, wherein the pi of the active peptide is 4.0 or less and the pH of the formulation is 7.0 to 8.0.
18. A method according to any of the foregoing or the following, wherein the pi of the active peptide substantially determines the pi of the protein.
19. A method according to any of the foregoing or the following, wherein the Fc region is derived from an IgG antibody or is derivative or a modification of an Fc region derived from an IgG antibody.
20. A method according to any of the foregoing or the following, wherein the IgG is an IgGl, IgG2, or IgG4.
21. A method according to any of the foregoing or the following, wherein the protein is a peptibody. 22. A method according to any of the foregoing or the following, wherein the protein is an antibody.
23. A method according to any of the foregoing or the following, wherein the protein is an antibody-derived protein.
24. A method according to any of the foregoing or the following, wherein the protein comprises one or more of a VH domain or a VL domain of an antibody.
25. A method according to any of the foregoing or the following, where the protein is a monoclonal or polyclonal antibody.
26. A method according to any of the foregoing or the following, wherein the protein is a humanized antibody. 27 '. A method according to any of the foregoing or the following, wherein the protein is selected from the group consisting of single chain antibodies, scFv(s), intrabodies, maxibodies, minibodies and diabodies. 28. A method according to any of the foregoing or the following, wherein the protein is a receptibody.
29. A method according to any of the foregoing or the following, wherein the protein comprises an Fab region of an antibody. 30. A method according to any of the foregoing or the following, wherein the active peptide binds specifically to a target ligand.
31. A method according to any of the foregoing or the following, wherein the protein is selected from the group consisting of proteins that bind specifically to one or more of CD proteins, HER receptor family proteins, cell adhesion molecules, growth factors, nerve growth factors, fibroblast growth factors, colony stimulating factors, transforming growth factors (TGF), insulin-like growth factors, osteoinductive factors, insulins, insulin-related proteins, coagulation proteins, coagulation-related proteins, colony stimulating factors (CSFs), blood proteins, serum proteins, blood group antigens, receptors, receptor-associated proteins, growth hormone receptors, T-cell receptors, neurotrophic factors, neurotrophins, relaxins, interferons, interleukins, viral antigens, lipoproteins, integrins, rheumatoid factors, immunotoxins, surface membrane proteins, transport proteins, homing receptors, addressins, regulatory proteins, immunoadhesins, and receptor ligands.
32. A method according to any of the foregoing or the following, wherein the protein is selected from the group consisting of: OPGL specific binding proteins, myostatin specific binding proteins, IL-4 receptor specific binding proteins, ILl-Rl specific binding proteins, Ang2 specific binding proteins, NGF-specifϊc binding proteins, CD22 specific binding proteins, IGF-I receptor specific binding proteins, B7RP-1 specific binding proteins, IFN gamma specific binding proteins, TALL-I specific binding proteins, stem cell factors, Flt-3 ligands, and IL- 17 receptors. 33. A method according to any of the foregoing or the following, wherein the protein is selected from the group consisting of proteins that bind specifically to one or more of: CD3, CD4, CD8, CD19, CD20, CD22, CD30, CD34; HER2, HER3, HER4, the EGF receptor; LFA-I, MoI, pl50,95, VLA-4, ICAM-I, VCAM, alpha v/beta 3 integrin, OPGL, Ang2, Angl, stem cell factors, vascular endothelial growth factor ("VEGF"); growth hormone, thyroid stimulating hormone, follicle stimulating hormone, luteinizing hormone, growth hormone releasing factor, parathyroid hormone, mullerian-inhibiting substance, human macrophage inflammatory protein (MIP-I -alpha), erythropoietin (EPO), NGF, NGF-beta, platelet-derived growth factor (PDGF), aFGF, bFGF, epidermal growth factor (EGF), TGF- alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-beta4, TGF-beta5, IGF-I, IGF-II, des(l-3)- IGF-I (brain IGF-I), insulin, insulin A-chain, insulin B-chain, proinsulin, insulin-like growth factor binding proteins, factor VIII, tissue factor, von Willebrands factor, protein C, alpha- 1- antitrypsin, plasminogen activators, such as urokinase and tissue plasminogen activator ("t- PA"), bombazine, thrombin, thrombopoietin, M-CSF, GM-CSF, G-CSF, albumin, IgE, flk2/flt3 receptor, obesity (OB) receptor, bone-derived neurotrophic factor (BDNF), NT-3, NT-4, NT-5, NT-6; relaxin A-chain, relaxin B-chain, prorelaxin; interferon-alpha, -beta, and - gamma; IL-I to IL- 15; IL-I to IL- 15 receptors, AIDS envelope viral antigen; calcitonin, glucagon, atrial natriuretic factor, lung surfactant, tumor necrosis factor-alpha and -beta, enkephalinase, RANTES, mouse gonadotropin-associated peptide, DNAse, inhibin, and activin; protein A or D, bone morphogenetic protein (BMP), superoxide dismutase, decay accelerating factor (DAF), thrombopoietin receptor, TALL-I /B AFF/ AGP-3, myostatin, amyloid beta, thymic stromal lyphopoietin, RANKL (RANK ligand), c-kit, OX40L, glucagon receptor, IGF-I, B7RP-1, TNF, TNF receptor type I (p55), TRAIL-R2, activin A, CSF-I receptor, c-Kit and OX40L .
34. A method according to any of the foregoing or the following, wherein the formulation is a liquid.
35. A method according to any of the foregoing or the following, wherein the formulation is a lyophilate that, upon reconstitution, provides a liquid with properties as set forth in the foregoing or the following.
36. A method according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is at least 20 mg/ml.
37. A method according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is 20 to 500 mg/ml. 38. A method according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is 0.1 to 400 mg/ml.
39. A method according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is between any of 0.1 to 1.0, 0.2 to 2.0, 0.5 to 5.0, 1.0 to 10, 2.0 to 20, 5.0 to 25, 10 to 50, 25 to 75, 50 to 100, 75 to 150, 100 to 200, 150 to 250, 200 to 300, 250 to 350, 300 to 500, 150 to 300, 300 to 500 or more mg/ml.
40. A method according to any of the foregoing or the following, wherein the protein is stable in the formulation for any of at least 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50 or more months. 41. A method in accordance with any of the foregoing or the following, wherein the concentration of the protein in the formulation is any of between 0.1 and 10, 0.1 and 25, 0.1 and 50, 0.1 and 100, 0.1 and 150, 0.1 and 200, 0.1 and 250, 0.1 and 300, 0.1 and 400, 0.1 and 400 or higher mg/ml; 0.2 and 10, 0.2 and 25, 0.2 and 50, 0.2 and 100, 0.2 and 150, 0.2 and 200, 0.2 and 250, 0.2 and 300, 0.2 and 400, 0.2 and 400 or higher mg/ml; 0.5 and 10, 0.5 and 25, 0.5 and 50, 0.5 and 100, 0.5 and 150, 0.5 and 200, 0.5 and 250, 0.5 and 300, 0.5 and 400, 0.5 and 400 or higher mg/ml; 1.0 and 10, 1.0 and 25, 1.0 and 50, 1.0 and 100, 1.0 and 150, 1.0 and 200, 1.0 and 250, 1.0 and 300, 1.0 and 400, 1.0 and 400 or higher mg/ml; 5.0 and 10, 5.0 and 25, 5.0 and 50, 5.0 and 100, 5.0 and 150, 5.0 and 200, 5.0 and 250, 5.0 and 300, 5.0 and 400, 5.0 and 400 or higher mg/ml; 10 and 25, 10 and 50, 10 and 100, 10 and 150, 10 and 200, 10 and 250, 10 and 300, 10 and 400, 10 and 400 or higher mg/ml; 25 and 50, 25 and 100, 25 and 150, 25 and 200, 25 and 250, 25 and 300, 25 and 400, 25 and 400 or higher mg/ml; 50 and 100, 50 and 150, 50 and 200, 50 and 250, 50 and 300, 50 and 400, 50 and 400 or higher mg/ml; 150 and 300, 200 and 400, 200 and 400 or higher mg/ml. 42. A method in accordance with any of the foregoing or the following, wherein the formulation further comprises a buffering agent, and wherein the concentration of the buffering agent is any of 0.5 to 5.0, 1.0 to 10, 2.0 to 20, 5.0 to 50, 10 to 100, 50 to 150, 100 to 200, 100 to 500, 150 to 300, 200 to 500, 250 to 500 mM or more.
43. A method in accordance with any of the foregoing of the following, wherein the formulation further comprises a buffering agent, wherein the buffering agent is one or more of acetate, phosphate, or histidine.
44. A method in accordance with any of the foregoing or the following, wherein at the pH of the formulation, the protein in the formulation is self-buffering and provides at least approximately 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 99.5% of the buffer capacity of the composition.
45. A method in accordance with any of the foregoing or the following, wherein the formulation further comprises a salt, wherein the concentration of the salt is any of about 0.5, 1, 2, 5, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500 or more than 500 mM. 46. A method in accordance with any of the foregoing or the following, wherein the formulation further comprises a salt, wherein the concentration of the salt is in any of 0.5 to 5, 1 to 10, 5 to 25, 15 to 35, 25 to 50, 35 to 75, 50 to 100, 75 to 150, 100 to 200, 150 to 250, 200 to 300, 250 to 400, 300 to 500, 400 to 600, or 500 to more than 600 mM. 47. A method according to any of the foregoing or the following, wherein the formulation further comprises a surfactant.
48. A method according to any of the foregoing or the following, wherein the formulation further comprises a pharmaceutically acceptable surfactant. 49. A method according to any of the foregoing or the following, wherein the formulation further comprises a surfactant, wherein the surfactant is one or more of polysorbate 20, polysorbate 80, other fatty acid esters of sorbitan, polyethoxylates, and poloxamer 188.
50. A method according to any of the foregoing or the following, wherein the formulation further comprises a surfactant, wherein the surfactant is one or more of polysorbate 20 and polysorbate 80.
51. A method in accordance with any of the foregoing or the following, wherein the formulation further comprises a surfactant, wherein the surfactant is any one or more of Tween 20, including but not limited to from about 0.0005% or about 0.01% Tween 20; sodium cholate, including but not limited to from about 0.001% to about 0.01% sodium cholate; sodium glycholate, including but not limited to from about 0.001% to about 0.01% sodium glycholate; sodium deoxy cholate, including but not limited to from about 0.001% to 0.01% sodium deoxycholate; sodium glycodeoxycholate, including but not limited to from about 0.001% to about 0.01% sodium glycodeoxycholate; CHAPS, including but not limited to from about 0.001 % to about 0.01 % CHAPS; CHAPSO, including but not limited to from about 0.001% to about 0.01% CHAPSO; Emphigen BB, including but not limited to from about 0.001% to about 0.01% Emphigen BB; SDS, including but not limited to from about 0.001% to about 0.01% SDS; Mega-8, including but not limited to from about 0.001% to about 0.01% Mega-8; Genepol C-100, including but not limited to from about 0.001% to about 0.01% Genepol C-100; Brij 35, including but not limited to from about 0.001% to about 0.01% Brij 35; Pluronic F-68, including but not limited to from about 0.001% to about 0.01% Pluronic F-68; Pluronic F-127, including but not limited to from about 0.001% to about 0.01% Pluronic F-127; Zwittergent 3-12, including but not limited to from about 0.001% to about 0.01% Zwittergent 3-12; PEG-8000, including but not limited to from about 0.001% to about 0.01 % PEG-8000; PEG-4000, including but not limited to from about 0.001 % to about 0.01 % PEG-4000; HPCD, including but not limited to from about 0.001% to about 0.1% HPCD; and Triton X-100, including but not limited to from about 0.001% to about 0.01% Triton X-100. 52. A method according to any of the foregoing or the following, wherein the formulation further comprises a polyol.
53. A method according to any of the foregoing or the following, wherein the formulation further comprises a polyol in a hypotonic, isotonic, or hypertonic amount. 54. A method according to any of the foregoing or the following, wherein the formulation further comprises a polyol, wherein the polyol is one or more of sorbitol, mannitol, sucrose, trehalose, or glycerol.
55. A method according to any of the foregoing or the following, wherein the formulation further comprises a polyol, wherein the polyol is any one of approximately 5% sorbitol, 5% mannitol, 9% sucrose, 9% trehalose, or 2.5% glycerol, very especially in this regard 5% sorbitol, 5% mannitol, 9% sucrose, 9% trehalose, or 2.5% glycerol
56. A method according to any of the foregoing or the following, wherein the formulation further comprises an amino acid.
57. A method according to any of the foregoing or the following, wherein the formulation further comprises an amino acid, wherein the amino acid is any of arginine or histidine.
58. A method according to any of the foregoing or the following, wherein the formulation further comprises a non-ionic excipient.
59. A method according to any of the foregoing or the following, wherein the formulation further comprises an anionic excipient.
60. A method according to any of the foregoing or the following, wherein the formulation further comprises in addition to said protein one or more additional pharmaceutical agents.
61. A method according to any of the foregoing or the following, wherein the formulation further comprises one or more pharmaceutically acceptable: osmotic balancing agents; anti-oxidants; antibiotics; antimycotics; bulking agents; lyoprotectants; anti-foaming agents; chelating agents; preservatives; colorants; analgesics; or additional pharmaceutical agents.
62. A formulation produced by a method according to any of the foregoing or the following.
63. A lyophilate formulation produced by a method according to any of the foregoing or the following. 64. A method for treating a subject, comprising administering to a subject in an amount and by a route effective for said treatment, a formulation produced by a method according to any of the foregoing or the following.
65. A formulation comprising a protein comprised of an active peptide, wherein the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
66. A formulation comprising a protein comprised of an active peptide, wherein the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
67. A formulation comprising a protein comprised of an active peptide and an Fc region, wherein the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
68. A formulation comprising a protein comprised of an active peptide and an Fc region, wherein the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation. 69. A formulation comprising a peptibody comprised of an Fc region and an active peptide, wherein the pH of the formulation is (i) 4.5 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
70. A formulation comprising a peptibody comprised of an Fc region and an active peptide, wherein the pH of the formulation is (i) 4.5 to 5.6 if the pi of the peptibody is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the peptibody is 6.0 or less, and wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
71. A formulation according to any of the foregoing or the following, wherein the pi of the active peptide substantially determines the pi of the protein. 12. A formulation according to any of the foregoing or the following, wherein the
Fc region is derived from an IgG antibody or is derivative or a modification of an Fc region derived from an IgG antibody. 73. A formulation according to any of the foregoing or the following, wherein the Fc region is derived from an IgGl, IgG2 or IgG4 antibody.
74. A formulation according to any of the foregoing or the following, wherein the protein is a peptibody. 75. A formulation according to any of the foregoing or the following, wherein the protein is an antibody.
76. A formulation according to any of the foregoing or the following, wherein the protein is an antibody-derived protein.
77. A formulation according to any of the foregoing or the following, wherein the protein comprises one or more of a VH domain or a VL domain of an antibody.
78. A formulation according to any of the foregoing or the following, where the protein is a monoclonal or polyclonal antibody.
79. A formulation according to any of the foregoing or the following, wherein the protein is a humanized antibody. 80. A formulation according to any of the foregoing or the following, wherein the protein is selected from the group consisting of single chain antibodies, scFv(s), intrabodies, maxibodies, minibodies and diabodies.
81. A formulation according to any of the foregoing or the following, wherein the protein is a receptibody. 82. A formulation according to any of the foregoing or the following, wherein the protein comprises an Fab region of an antibody.
83. A formulation according to any of the foregoing or the following, wherein the active peptide binds specifically to a target ligand.
84. A formulation according to any of the foregoing or the following, wherein the formulation is a liquid.
85. A formulation according to any of the foregoing or the following, wherein the formulation is a lyophilate and upon reconstitution is a liquid with properties as set forth in the foregoing or the following.
86. A formulation according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is at least 20 mg/ml.
87. A formulation according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is 20 to 500 mg/ml. 88. A formulation according to any of the foregoing or the following, wherein the concentration of the protein in the formulation is 0.1 to 400 mg/ml.
89. A formulation in accordance with any of the foregoing or the following, wherein the concentration of the protein is between any of 0.1 to 1.0, 0.2 to 2.0, 0.5 to 5.0, 1.0 to 10, 2.0 to 20, 5.0 to 25, 10 to 50, 25 to 75, 50 to 100, 75 to 150, 100 to 200, 150 to 250, 200 to 300, 250 to 350, 300 to 500, 150 to 300, 300 to 500 or more mg/ml.
90. A formulation in accordance with any of the foregoing or the following, wherein the protein is stable in the formulation for any of at least 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50 or more months. 91. A formulation in accordance with any of the foregoing or the following, wherein the concentration of the protein is any of between 0.1 and 10, 0.1 and 25, 0.1 and 50, 0.1 and 100, 0.1 and 150, 0.1 and 200, 0.1 and 250, 0.1 and 300, 0.1 and 400, 0.1 and 400 or higher mg/ml; 0.2 and 10, 0.2 and 25, 0.2 and 50, 0.2 and 100, 0.2 and 150, 0.2 and 200, 0.2 and 250, 0.2 and 300, 0.2 and 400, 0.2and 400 or higher mg/ml; 0.5 and 10, 0.5 and 25, 0.5 and 50, 0.5 and 100, 0.5 and 150, 0.5 and 200, 0.5 and 250, 0.5 and 300, 0.5 and 400, 0.5 and 400 or higher mg/ml; 1.0 and 10, 1.0 and 25, 1.0 and 50, 1.0 and 100, 1.0 and 150, 1.0 and 200, 1.0 and 250, 1.0 and 300, 1.0 and 400, 1.0 and 400 or higher mg/ml; 5.0 and 10, 5.0 and 25, 5.0 and 50, 5.0 and 100, 5.0 and 150, 5.0 and 200, 5.0 and 250, 5.0 and 300, 5.0 and 400, 5.0 and 400 or higher mg/ml; 10 and 25, 10 and 50, 10 and 100, 10 and 150, 10 and 200, 10 and 250, 10 and 300, 10 and 400, 10 and 400 or higher mg/ml; 25 and 50, 25 and 100, 25 and 150, 25 and 200, 25 and 250, 25 and 300, 25 and 400, 25 and 400 or higher mg/ml; 50 and 100, 50 and 150, 50 and 200, 50 and 250, 50 and 300, 50 and 400, 50 and 400 or higher mg/ml; 150 and 300, 200 and 400, 200 and 400 or higher mg/ml.
92. A formulation in accordance with any of the foregoing or the following, further comprising a buffering agent, and wherein the concentration of the buffering agent is any of
0.5 to 5.0, 1.0 to 10, 2.0 to 20, 5.0 to 50, 10 to 100, 50 to 150, 100 to 200, 100 to 500, 150 to 300, 200 to 500, 250 to 500 mM or more.
93. A formulation in accordance with any of the foregoing of the following, further comprising a buffering agent, wherein the buffering agent is one or more of acetate, phosphate, or histidine.
94. A formulation in accordance with any of the foregoing or the following, wherein at the pH of the formulation, the protein is self-buffering and provides at least approximately 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 99.5% of the buffer capacity of the composition.
95. A formulation in accordance with any of the foregoing or the following, further comprising a salt, wherein the concentration of the salt is any of about 0.5, 1, 2, 5, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500 or more mM and may be in the range of any of 0.5 to 5, 1 to 10, 5 to 25, 15 to 35, 25 to 50, 35 to 75, 50 to 100, 75 to 150, 100 to 200, 150 to 250, 200 to 300, 250 to 400, 300 to 500, 400 to 600 mM or higher.
96. A formulation according to any of the foregoing or the following, wherein the formulation further comprises a surfactant. 97. A formulation according to any of the foregoing or the following, wherein the formulation further comprises one or more pharmaceutically acceptable surfactants.
98. A formulation according to any of the foregoing or the following, wherein the surfactant is one or more of polysorbate 20, polysorbate 80, other fatty acid esters of sorbitan, polyethoxylates, and poloxamer 188. 99. A formulation according to any of the foregoing or the following, wherein the surfactant is one or more of polysorbate 20 and polysorbate 80.
100. A formulation in accordance with any of the foregoing or the following, further comprising a surfactant, wherein the surfactant is any one or more of Tween 20, including but not limited to from about 0.0005% or about 0.01% Tween 20; sodium cholate, including but not limited to from about 0.001% to about 0.01% sodium cholate; sodium glycholate, including but not limited to from about 0.001% to about 0.01% sodium glycholate; sodium deoxycholate, including but not limited to from about 0.001% to 0.01% sodium deoxycholate; sodium glycodeoxycholate, including but not limited to from about 0.001% to about 0.01% sodium glycodeoxycholate; CHAPS, including but not limited to from about 0.001% to about 0.01 % CHAPS; CHAPSO, including but not limited to from about 0.001 % to about 0.01 % CHAPSO; Emphigen BB, including but not limited to from about 0.001% to about 0.01% Emphigen BB; SDS, including but not limited to from about 0.001% to about 0.01% SDS; Mega-8, including but not limited to from about 0.001% to about 0.01% Mega-8; Genepol C- 100, including but not limited to from about 0.001% to about 0.01% Genepol C-100; Brij 35, including but not limited to from about 0.001% to about 0.01% Brij 35; Pluronic F-68, including but not limited to from about 0.001% to about 0.01% Pluronic F-68; Pluronic F-127, including but not limited to from about 0.001% to about 0.01% Pluronic F-127; Zwittergent 3- 12, including but not limited to from about 0.001% to about 0.01% Zwittergent 3-12; PEG- 8000, including but not limited to from about 0.001% to about 0.01% PEG-8000; PEG-4000, including but not limited to from about 0.001% to about 0.01% PEG-4000; HPCD, including but not limited to from about 0.001% to about 0.1% HPCD; and Triton X-100, including but not limited to from about 0.001% to about 0.01% Triton X-100. 101. A formulation according to any of the foregoing or the following, wherein the formulation further comprises a polyol.
102. A formulation according to any of the foregoing or the following, wherein the polyol is one or more of sorbitol, mannitol, sucrose, trehalose, or glycerol.
103. A formulation according to any of the foregoing or the following, further comprising a polyol in a hypotonic, isotonic, or hypertonic amount.
104. A formulation according to any of the foregoing or the following, further comprising a polyol in an isotonic amount.
105. A formulation according to any of the foregoing or the following, further comprising a polyol, wherein the polyol is any one or more of sorbitol, mannitol, sucrose, trehalose, or glycerol.
106. A formulation according to any of the foregoing or the following, further comprising a polyol, wherein the polyol is any one of approximately 5% sorbitol, 5% mannitol, 9% sucrose, 9% trehalose, or 2.5% glycerol, very especially in this regard 5% sorbitol, 5% mannitol, 9% sucrose, 9% trehalose, or 2.5% glycerol 107. A formulation according to any of the foregoing or the following, wherein the formulation further comprises an amino acid.
108. A formulation according to any of the foregoing or the following, further comprising an amino acid, wherein the amino acid is any of arginine or histidine.
109. A formulation according to any of the foregoing or the following, wherein the formulation further comprises a non-ionic excipient.
110. A formulation according to any of the foregoing or the following, wherein the formulation further comprises an anionic excipient.
111. A formulation according to any of the foregoing or the following, wherein the formulation further comprises in addition to said protein one or more additional pharmaceutical agents.
112. A formulation according to any of the foregoing or the following, wherein the formulation further comprises one or more pharmaceutically acceptable: osmotic balancing agents; anti-oxidants; antibiotics; antimycotics; bulking agents; lyoprotectants; anti-foaming agents; chelating agents; preservatives; colorants; analgesics; or additional pharmaceutical agents.
113. A method according to any of the foregoing or the following, wherein the protein is selected from the group consisting of proteins that bind specifically to one or more of CD proteins, HER receptor family proteins, cell adhesion molecules, growth factors, nerve growth factors, fibroblast growth factors, colony stimulating factors, transforming growth factors (TGF), insulin-like growth factors, osteoinductive factors, insulins, insulin-related proteins, coagulation proteins, coagulation-related proteins, colony stimulating factors (CSFs), blood proteins, serum proteins, blood group antigens, receptors, receptor-associated proteins, growth hormone receptors, T-cell receptors, neurotrophic factors, neurotrophins, relaxins, interferons, interleukins, viral antigens, lipoproteins, integrins, rheumatoid factors, immunotoxins, surface membrane proteins, transport proteins, homing receptors, addressins, regulatory proteins, immunoadhesins, and receptor ligands.
114. A formulation according to any of the foregoing or the following, wherein the protein is selected from the group consisting of: OPGL specific binding proteins, myostatin specific binding proteins, IL-4 receptor specific binding proteins, ILl-Rl specific binding proteins, Ang2 specific binding proteins, NGF-specifϊc binding proteins, CD22 specific binding proteins, IGF-I receptor specific binding proteins, B7RP-1 specific binding proteins, IFN gamma specific binding proteins, TALL-I specific binding proteins, stem cell factors, Flt-3 ligands, and IL- 17 receptors.
115. A method according to any of the foregoing or the following, wherein the protein is selected from the group consisting of proteins that bind specifically to one or more of: CD3, CD4, CD8, CD19, CD20, CD22, CD30, CD34; HER2, HER3, HER4, the EGF receptor; LFA-I, MoI, pl50,95, VLA-4, ICAM-I, VCAM, alpha v/beta 3 integrin, OPGL, Ang2, Angl, stem cell factors, vascular endothelial growth factor ("VEGF"); growth hormone, thyroid stimulating hormone, follicle stimulating hormone, luteinizing hormone, growth hormone releasing factor, parathyroid hormone, mullerian-inhibiting substance, human macrophage inflammatory protein (MIP-I -alpha), erythropoietin (EPO), NGF, NGF-beta, platelet-derived growth factor (PDGF), aFGF, bFGF, epidermal growth factor (EGF), TGF- alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-beta4, TGF-beta5, IGF-I, IGF-II, des(l-3)- IGF-I (brain IGF-I), insulin, insulin A-chain, insulin B-chain, proinsulin, insulin-like growth factor binding proteins, factor VIII, tissue factor, von Willebrands factor, protein C, alpha- 1- antitrypsin, plasminogen activators, such as urokinase and tissue plasminogen activator ("t- PA"), bombazine, thrombin, thrombopoietin, M-CSF, GM-CSF, G-CSF, albumin, IgE, flk2/flt3 receptor, obesity (OB) receptor, bone-derived neurotrophic factor (BDNF), NT-3, NT-4, NT-5, NT-6; relaxin A-chain, relaxin B-chain, prorelaxin; interferon-alpha, -beta, and - gamma; IL-I to IL- 15; IL-I to IL- 15 receptors, AIDS envelope viral antigen; calcitonin, glucagon, atrial natriuretic factor, lung surfactant, tumor necrosis factor-alpha and -beta, enkephalinase, RANTES, mouse gonadotropin-associated peptide, DNAse, inhibin, and activin; protein A or D, bone morphogenetic protein (BMP), superoxide dismutase, decay accelerating factor (DAF), thrombopoietin receptor, TALL-I /B AFF/ AGP-3, myostatin, amyloid beta, thymic stromal lyphopoietin, RANKL (RANK ligand), c-kit, OX40L, glucagon receptor, IGF-I, B7RP-1, TNF, TNF receptor type I (p55), TRAIL-R2, activin A, CSF-I receptor, c-Kit and OX40L.
116. A method for treating a subject, comprising administering to a subject in an amount and by a route effective for said treatment, a formulation according to any of the foregoing or the following.
Glossary
The meanings ascribed to various terms and phrases as used herein are illustratively explained below.
"A" or "an" herein means "at least one;" "one or more than one." "About," unless otherwise stated explicitly herein, means +/- 20%. For instance about
100 herein means 80 to 120, about 5 means 4 to 6, about 0.3 means 0.24 to 0.36, and about 60% means 48% to 72% (not 40% to 80%).
"Active peptide" as used herein means a portion of a protein, such as an Fc fusion protein that has a measurable function. In embodiments the function is specific binding to a target. In this regard specific binds means selective but not necessarily exclusive, much the same as antibodies generally bind specifically to antigens, but not exclusively. Generally, an active peptide comprises all of the protein necessary for the function. An active peptide generally will contain those portions of the protein directly involved in carrying out the function, portions indirectly involved and some portions of the protein that interconnect or terminate these portions.
For instance, for a protein wherein the function is specific binding, the active peptide generally will include the portions of the protein directly involved in binding, such as those that make contact with the target and chemically bond to it. It also will include portions of that facilitate contact and bonding, such as by providing flexibility, but do not themselves contact the target directly. Where two or more discontinuous portions of the protein are involved directly and/or indirectly in binding a target, the active peptide also will include those portions of the protein that connect the discontinuous portions. The active peptide also may include portions of the protein adjacent to the portions directly or indirectly involved in contacting the target and the portions that lie between them.
In embodiments, active peptides include some or all of a region that spans the interstice between the binding portions and the other parts of the protein. In Fc fusion proteins comprising an active peptide and an Fc region, for instance, the active may include some or all a linker within the protein that connects the portions of the active peptide described above to the Fc region of the protein. In embodiments in this regard, in Fc fusion proteins comprising an active peptide and an Fc region, the active peptide comprises all of the protein outside the Fc region.
An active peptide may comprise one or more polypeptide chains. In embodiments it comprises one chain. In embodiments it comprises two chains.
"Agonist(s)" means herein a molecular entity that is different from a corresponding stimulatory ligand but has a similar stimulatory effect. For instance (although agonists work through other mechanisms), for a hormone that stimulates an activity by binding to a corresponding hormone receptor, an agonist is a chemically different entity that binds the hormone receptor and similarly stimulates its activity.
"Antagonist(s)" means herein a molecular entity that is different from a corresponding ligand and has an opposite effect. For instance (although antagonists work through other mechanisms), one type of antagonist of a hormone that stimulates an activity by binding to a corresponding hormone receptor is a chemical entity that is different from the hormone and binds the hormone receptor but does not stimulate the activity engendered by hormone binding, and by this action inhibits the effector activity of the hormone.
"Antibody(s)" is used herein in accordance with its ordinary meaning in the biochemical and biotechno logical arts.
Among antibodies within the meaning of the term as it is used herein, are those isolated from biological sources, including monoclonal and polyclonal antibodies, antibodies made by recombinant DNA techniques (also referred to at times herein as recombinant antibodies), including those made by processes that involve activating an endogenous gene and those that involve expression of an exogenous expression construct, including antibodies made in cell culture and those made in transgenic plants and animals, and antibodies made by methods involving chemical synthesis, including peptide synthesis and semi-synthesis. Also within the scope of the term as it is used herein, except as otherwise explicitly set forth, are chimeric antibodies and hybrid antibodies, among others. The prototypical antibody is a tetrameric glycoprotein comprised of two identical light chain-heavy chain dimers joined together by disulfide bonds. There are two types of vertebrate light chains, kappa and lambda. Each light chain is comprised of a constant region and a variable region. The two light chains are distinguished by constant region sequences. There are five types of vertebrate heavy chains: alpha, delta, epsilon, gamma, and mu. Each heavy chain is comprised of a variable region and three constant regions. The five heavy chain types define five classes of vertebrate antibodies (isotypes): IgA, IgD, IgE, IgG, and IgM. Each isotype is made up of, respectively, (a) two alpha, delta, epsilon, gamma, or mu heavy chains, and (b) two kappa or two lambda light chains. The heavy chains in each class associate with both types of light chains; but, the two light chains in a given molecule are both kappa or both lambda. IgD, IgE, and IgG generally occur as "free" heterotetrameric glycoproteins. IgA and IgM generally occur in complexes comprising several IgA or several IgM heterotetramers associated with a "J" chain polypeptide. Some vertebrate isotypes are classified into subclasses, distinguished from one another by differences in constant region sequences. There are four human IgG subclasses, IgGl, IgG2, IgG3, and IgG4, and two IgA subclasses, IgAl and IgA2, for example. All of these and others not specifically described above are included in the meaning of the term "antibody(s)" as used herein.
The term "antibody(s)" further includes amino acid sequence variants of any of the foregoing as described further elsewhere herein.
"Antibody-derived" as used herein means any protein produced from an antibody, and any protein of a design based on an antibody. The term includes in its meaning proteins produced using all or part of an antibody, those comprising all or part of an antibody, and those designed in whole or in part on the basis of all or part of an antibody. "Antibody- derived" proteins include, but are not limited to, Fc, Fab, and Fab2 fragments and proteins comprising the same, VH domain and VL domain fragments and proteins comprising the same, other proteins that comprise a variable and/or a constant region of an antibody, in whole or in part, scFv(s) intrabodies, maxibodies, minibodies, diabodies, amino acid sequence variants of the foregoing, and a variety of other such molecules, including but not limited to others described elsewhere herein. "Antibody polypeptide(s)" as used herein, except as otherwise noted, means a polypeptide that is part of an antibody, such as a light chain polypeptide, a heavy chain polypeptide and a J chain polypeptide, to mention a few examples, including among others fragments, derivatives, and variants thereof, and related polypeptides. "Antibody-related" as used herein means any protein or mimetic resembling in its structure, function, or design an antibody or any part of an antibody. Among "antibody- related" proteins as the term is used herein are "antibody-derived" proteins as described above. It is to be noted that the terms "antibody-derived" and "antibody-related" substantially overlap; both terms apply to many such proteins. Examples of "antibody-related" proteins, without implying limitation in this respect, are peptibodies and receptibodies. Other examples of "antibody-related" proteins are described elsewhere herein.
"Approximately" unless otherwise noted means the same as "about."
"Binding moiety(s)" means a part of a molecule or a complex of molecules that binds specifically to part of another molecule or complex of molecules. The binding moiety may be the same or different from the part of the molecule or complex of molecules to which it binds. The binding moiety may be all of a molecule or complex of molecules as well.
"Binds specifically" is used herein in accordance with its ordinary meaning in the art and means, except as otherwise noted, that binding is stronger with certain specific moieties than it is to other moieties in general, that it is stronger than non-specific binding that may occur with a wide variety of moieties, and that binding is selective for certain moieties and does not occur to as strong an extent with others. In the extreme case of specific binding, very strong binding occurs with a single type of moiety, and there is no non-specific binding with any other moiety.
"Co-administer" means an administration of two or more agents in conjunction with one another, including simultaneous and/or sequential administration.
"Cognate(s)" herein means complementary, fitting together, matching, such as, for instance, two jigsaw puzzles that fit one another, the cylinder mechanism of a lock and the key that opens it, the substrate binding site of an enzyme and the substrate of the enzyme, and a target and target binding protein that binds specifically thereto. "Cognate binding moieties" herein means binding moieties that bind specifically to one another. Typically, but not always, it means a pair of binding moieties that bind specifically to one another. The moieties responsible for highly selective binding of a specific ligand and ligand receptor provide an illustrative example of cognate binding moieties. Another example is provided by the moieties that bind an antigen and an antibody.
"Composition" means any composition of matter comprising one or more constituents, such as a formulation. "Comprised of is a synonym of "comprising" (see below).
"Comprising" means including, without further qualification, limitation, or exclusion as to what else may or may not be included. For example, "a composition comprising x and y" means any composition that contains x and y, no matter what else it may contain. Likewise, "a method comprising x" is any method in which x is carried out, no matter what else may occur.
"Concentration" is used herein in accordance with its well-known meaning in the art to mean the amount of an item in a given amount of a mixture containing the item, typically expressed as a ratio. For example, concentration of a solute, such as a protein in a solution, can be expressed in many ways, such as (but not limited to): (A) Weight Percent (i) = weight of solute per 100 units of solvent volume; (B) Weight Percent (ii) = weight of solute per 100 units of total weight; (C) Weight Percent (iii) = weight of solute per 100 units of solvent by weight; (D) Mass Percent = mass of solute per 100 mass units of solution; (E) Mole Fraction = moles of solute per total moles of all components; (F) Molarity = moles of solute per liter of solution (i.e., solute plus solvent); (G) Molality = moles of solute per Kg of solvent; and (H) Volume Molality = moles of solute per liter of solvent.
"De novo" is used herein in accordance with its well-known meaning in the art, to denote something newly made. For instance, a de novo amino acid sequence is one not derived from a naturally occurring amino acid sequence, although, such a de novo sequence may have similarities with a naturally occurring sequence. De novo amino acid sequences can be generated, for instance, by a priori design, by combinatorial methods, and by selection methods. They can be made, for example, by chemical synthesis, by semi-synthesis, and by a variety of recombinant DNA techniques, all of which are well know to those skilled in the art.
"Deleterious" means, as used herein, harmful. By way of illustration, "deleterious" processes include, for example, harmful effects of disease processes and harmful side effects of treatments.
"Derivative(s)" is used herein to mean derived from, in substance, form, or design, such as, for instance, a polypeptide that is based on but differs from a reference polypeptide, for instance, by alterations to its amino acid sequence, by fusion to another polypeptide, or by covalent modification.
"Disease(s)" a pathology, a condition that deleteriously affects health of a subject.
"Disorder(s)" a malediction, a condition that deleteriously alters health. "Dysfunction" means, as used herein, a disorder, disease, or deleterious effect of an otherwise normal process.
"Effective amount" generally means an amount which provides the desired local or systemic effect. For example, an effective amount is an amount sufficient to effectuate a beneficial or desired clinical result. The effective amount can be provided all at once in a single administration or in fractional amounts that provide the effective amount in several administrations. The precise determination of what would be considered an effective amount may be based on factors individual to each subject, including their size, age, injury, and/or disease or injury being treated, and amount of time since the injury occurred or the disease began. One skilled in the art will be able to determine the effective amount for a given subject based on these considerations which are routine in the art. As used herein, "effective dose" means the same as "effective amount."
"Effective route" generally means a route which provides for delivery of an agent to a desired compartment, system, location, or the like. For example, without limitation, an effective route is one through which an agent can be administered to provide at the desired site of action an amount of the agent sufficient to effectuate a beneficial or desired clinical result.
"Formulation(s)" means "composition(s)" as defined herein above, such as a combination of at least one active ingredient with one or more other ingredients for one or more particular uses, such as storage, further processing, sale, and/or administration to a subject, such as, for example, administration to a subject of a specific agent in a specific amount, by a specific route, or to treat a specific disease.
"Fragment(s)" herein means part of a larger entity, such as a part of a protein; for instance, a polypeptide consisting of less than the entire amino acid sequence of a larger polypeptide. As used herein, the term includes fragments formed by terminal deletion and fragments formed by internal deletion, including those in which two or more non-contiguous portions of a polypeptide are joined together to form a smaller polypeptide, which is a fragment of the original.
"Fusion protein(s)" herein means a protein formed by fusing all or part of two polypeptides, which may be either the same or different. Typical fusion proteins are made by recombinant DNA techniques, by end to end joining of nucleotides encoding the two (or more) polypeptides.
"Genetically engineered" herein means produced using a deliberate process of genetic alteration, such as by recombinant DNA technology, classical methods of genetic manipulation, chemical methods, a combination of all three, or other methods.
"Homolog(s)" herein means having homology to another entity, such as a protein that is homologous to another protein. Homologous means resembling in function or in structure, including, without limitation, primary, secondary, and/or tertiary structure.
"Isoelectric pH" as used herein means the same as "isoelectric point." "Isoelectric point" (also called the isoelectric pH) is used herein with reference to any given peptide or protein to denote the pH at which the net charge thereon is zero; that is, at the isoelectric pH the negative charge of the given peptide or protein is equal to its positive charge.
"Kit" means a collection of items used together for a given purpose or purposes. "Ligand(s)" herein means a molecular entity that binds selectively and stoichiometrically to one or more specific sites on one or more other molecular entities. Binding typically is non-covalent, but can be covalent as well. A very few examples, among many others, are (a) antigens, which typically bind non-covalently to the binding sites on cognate antibodies; (b) hormones, which typically bind hormone receptors, non-covalently; (c) lectins, which bind specific sugars, non-covalently; (d) biotins, which bind multiple sites on avidin and other avidin-like proteins, non-covalently; (e) hormone antagonists, which bind hormone receptors and inhibit their activity and/or that of the corresponding hormone; and (f) hormone agonists, which similarly bind hormone receptors but stimulate their activity.
"Ligand-binding moiety(s)" herein means a molecular entity that binds a ligand, typically, a part of a larger molecular entity that binds the ligand, or a molecular entity derived therefrom.
"Ligand-binding protein(s)" herein means a protein that binds a ligand.
"Ligand moiety(s)" herein means a molecular entity that binds to a ligand-binding molecular entity in much the same way as does the corresponding ligand. A ligand moiety can be all of a ligand, or part of it, derived from a ligand, or generated de novo. Typically, however, the ligand moiety is more or less exclusively the aspect thereof that binds corresponding ligand-binding entities. The ligand moiety need not comprise, and the term generally does not denote, structural features other than those required for ligand binding. "mAb" as used herein means monoclonal antibody(s), including but not limited to humanized monoclonal antibodies, chimeric monoclonal antibodies, derivatives of monoclonal antibodies, and modified forms of all of the forgoing, to name just a few.
"Mimetic(s)" herein means a chemical entity with structural or functional characteristics of another, generally unrelated chemical entity. For instance, one kind of hormone mimetic is a non-peptide organic molecule that binds to the corresponding receptor in the same way as the corresponding hormone.
"mM" means millimolar; 10" moles per liter.
"Modified protein(s)," "modified polypeptide(s)," or "modified fragment(s)" herein means a protein or a polypeptide or a fragment of a protein or polypeptide comprising a chemical moiety (structure) other than those of the twenty naturally occurring amino acids that form naturally occurring proteins. Modifications most often are covalently attached, but can also be attached non-covalently to a protein or other polypeptide, such as a fragment of a protein. "Moiety(s)" herein means a molecular entity that embodies a specific structure and/or function, without extraneous components. For instance, in most cases, only a small part of a ligand-binding protein is responsible for ligand binding. This part of the protein, whether continuously encoded or discontinuously, is an example of a ligand-binding moiety.
"Naturally occurring" means occurs in nature, without human intervention. "Non-naturally occurring" means does not occur in nature or, if it occurs in nature, is not in its naturally occurring state, environment, circumstances, or the like; altered by the hand of man.
"PBS" generally stands for phosphate buffered saline. The exact composition of PBS varies. A composition useful in the context of the present invention is 10 mM sodium phosphate, 100 mM sodium chloride, pH 7.4. This composition is merely illustrative and not limitative.
"Peptibody" refers to a molecule comprising an antibody Fc domain (i.e., CH2 and CH3 antibody domains) that excludes antibody CHI , CL, VH, and VL domains as well as Fab and F(ab)2, wherein the Fc domain is attached to one or more peptides, preferably a pharmacologically active peptide, particularly preferably a randomly generated pharmacologically active peptide. The production of peptibodies is generally described in PCT publication WO00/24782, published May 4, 2000, which is herein incorporated by reference in its entirety, particularly as to the structure, synthesis, properties, and uses of peptibodies.
"Peptide(s)" herein means the same as polypeptide; often, but not necessarily, it is used in reference to a relatively short polypeptide. "pH" is used in accordance with its well-known and universal definition as follows: pH = - log [H3O+].
"Pharmaceutical" as used herein means is acceptable for use in a human or non-human subject for the treatment thereof, particularly for use in humans, and approved therefor by a regulatory authority empowered to regulate the use thereof such as, for example, the Food and Drug Administration in the United States, European Agency for the Evaluation of Medicinal Products, Japan's Ministry of Health, Labor and Welfare, or other regulatory agency such as those listed in R. Ng, DRUGS: FROM DISCOVERY TO APPROVAL, Wiley-Liss (Hoboken, NJ) (2004), which is herein incorporated by reference in its entirety, particularly as to regulatory authorities concerned with drug approval, especially as listed in Chapter 7 therein. As used herein the phrase "wherein the composition has been approved for pharmaceutical use by an authority legally empowered to grant such approval" means an entity or institution or the like, established by law and by law charged with the responsibility and power to regulate and approve the use of drugs for use in humans, and in some cases, in non-humans. Approval by any one such agency anywhere meets this qualification. It is not necessary for the approving agency to be that of the state in which, for instance, a patent has issued or an infringement of a patent is occurring. Examples of such entities include the U. S. Food and Drug Administration and the other agencies listed herein above.
As used herein, "pharmaceutical" also may refer to a product produced in accordance with good manufacturing practices, such as those described in, among others, Chapter 9 and Chapter 10, of R. Ng, DRUGS: FROM DISCOVERY TO APPROVAL, Wiley-Liss
(Hoboken, NJ) (2004), which is herein incorporated by reference in its entirety, particularly in parts pertinent to good manufacturing practices for pharmaceutical protein formulations, in particular, as set forth in Chapters 9 and 10.
"Pharmaceutical composition(s)" means a compositions suitable for "pharmaceutical" use in accordance with the definition of "pharmaceutical" set forth herein above.
"Pharmaceutical formulation(s)" means the same as "pharmaceutical composition(s)" as the term is defined herein above. "Pharmaceutically acceptable" is used herein in accordance with its well-known meaning in the art to denote that which is acceptable for medical or veterinary use, preferably for medical use in humans, particularly approved for such use by the U.S. Food and Drug Administration or other authority as described above regarding the meaning of "pharmaceutical."
"Pharmaceutically active agent" is used herein to mean any substance that has a pharmaceutical effect, such as substances useful for treating a disorder or disease in a subject.
"pi" as used herein means (stands for) isoelectric point, also referred to as isoelectric pH. "Polypeptide(s)" see "Protein(s)."
"Precursor(s)" is used herein in accordance with its well-known meaning in the art to denote an entity from which another entity is derived. For instance, a precursor protein is a protein that undergoes processing, such as proteolytic cleavage or modification, thereby giving rise to another precursor protein (which will undergo further processing) or a mature protein. "Protein(s)" herein means a polypeptide or a complex of polypeptides, in accordance with its well-known meaning in the art. As used herein, "protein(s)" includes both straight chain and branched polypeptides. It includes unmodified and modified polypeptides, including naturally occurring modifications and those that do not occur naturally. Such modifications include chemical modifications of the termini, the peptide backbone, and the amino acid side chains; amino acid substitutions, deletions and additions; and incorporation of unusual amino acids and other moieties, to name just a few such modifications. It also includes "engineered" polypeptides and complexes thereof, such as, but not limited to, any polypeptide or complex of polypeptides that has been deliberative Iy altered in its structure by, for instance, recombinant DNA techniques, chemical synthesis, and/or covalent modification, including deliberate alteration of amino acid sequence and/or posttranslational modifications.
"Self-buffering" means the capacity of a substance, such as a pharmaceutical protein, to resist change in pH sufficient for a given application, in the absence of other buffers.
"Semi-ύfe novo " herein means (a) partly designed in accordance with a particular reference and or produced from a precursor, and (b) partly designed without reference to a particular reference (such as designed solely by general principles and not based on any particular reference), for example, a polypeptide made by producing a first peptide in a bacterial expression system, producing a second peptide by chemical synthesis, and then joining the two peptides together to form the polypeptide. "Semi-synthesis" means as used herein a combination of chemical and non-chemical methods of synthesis.
"Stable" as to pharmaceutical compositions is a term of art and is used herein in accordance therewith. The meaning of the term in this regard is illustrated by the following two documents.
(1) "Quality of Biotechno logical Products: Stability Testing of
Biotechnological/Biological Products, ICH Q5C" published July 1996 by the International Conference on Harmonisation of Technical Requirements of Pharmaceuticals for Human Use and available from the U.S. Food and Drug Administration, which is herein incorporated by reference in its entirety, particularly in parts pertinent to stability of pharmaceutical compositions. A copy of this document is attached at Appendix A to U.S. Provisional Application No. 60/935,073, of which priority is claimed herein and which is herein incorporated by reference in its entirety.
(2) "Guidance for Industry Q1A(R2) Stability Testing of New Drug Substances and Products" published by the U.S. Department of Health and Human Services, Food and Drug
Administration, Center for Drug Evaluation and Research, Center for Biologies Evaluation and Research, and the International Conference on Harmonisation of Technical Requirements of Pharmaceuticals for Human Use, November 2003, Revision 2, available from the U.S. Food and Drug Administration, which is herein incorporated by reference in its entirety, particularly in parts pertinent to the stability of pharmaceutical compositions. A copy of this document is attached at Appendix B to U.S. Provisional Application No. 60/935,073, of which priority is claimed herein and which is herein incorporated by reference in its entirety.
It is to be appreciated that stability in this regard, as discussed in the foregoing documents, may be determined in a variety of ways and its exact determination may vary from one pharmaceutical agent to another. In general, however, stable means that, for instance, for a typical pharmaceutical product intended for use by doctors to treat patients, a "stable" pharmaceutical composition is a formulation that, between the time the composition is made and the time it is used (or it reaches the end of its intended shelf life; i.e., its expiration date), when it is properly handled and stored, any changes that occur in its physical, chemical, or biological properties do not render it unsafe or ineffective for its intended use. Thus, in general as the term is used herein with reference to pharmaceutical compositions is meant that a composition approved for pharmaceutical use by an appropriate agency having jurisdiction over the approval of drugs for human use, such as the Federal Food and Drug Administration in the United States (and the like, as discussed above regarding the term "pharmaceutical"), retains the properties required for such approval over the time during which is may be said to be stable.
"Subject" means a vertebrate, such as a mammal, such as a human. Mammals include, but are not limited to, humans, farm animals, sport animals, and pets. Subjects in need of treatment by methods and/or compositions of the present invention include those suffering from a disorder, dysfunction, or disease, or a side effect thereof, or from a side effect of a treatment thereof.
"Substantially" is used herein in accordance with its plain and ordinary definition to mean to a great extent or degree. For example, substantially complete means complete to a great extent, complete to a great degree. By way of further illustration, substantially free of residue means to a great extent free of residue, free of residue to a great degree. Should numerical accuracy be required, depending on context, "substantially," as used herein means, at least, 80% or more, particularly 90% or more, very particularly 95% or more. "Therapeutically effective" is used herein in accordance with its well-known meaning in the art to denote that which achieves an improvement in the prognosis or condition of a subject or that otherwise achieves a therapeutic objective, including, for instance, a reduction in the rate of progress of a disease even if a subject's condition, nonetheless, continues to deteriorate. "Therapeutically effective amount" generally is used to qualify the amount of an agent to encompass those amounts that achieve an improvement in disorder severity. For example, effective neoplastic therapeutic agents prolong the survivability of the subject, inhibit the rapidly-proliferating cell growth associated with the neoplasm, or effect a regression of the neoplasm. Treatments that are therapeutically effective within the meaning of the term as used herein, include treatments that improve a subject's quality of life even if they do not improve the disease outcome per se.
"Treat," "treating," or "treatment" are used broadly in relation to the invention and each such term encompasses, among others, preventing, ameliorating, inhibiting, or curing a deficiency, dysfunction, disease, or other deleterious process, including those that interfere with and/or result from a therapy.
"Variant(s)" herein means a naturally occurring or synthetic version of, for instance, a protein that is structurally different from the original but related in structure and/or function, such as an allelic variant, a paralog, or a homo log of a protein. Description of the Invention
Many aspects of protein formulations are critically dependent on pH. The solubility of proteins generally is highly dependent on pH for instance, and their stability typically is maximal in a narrow range of pH. Thus, the proper pH for solubility and stability, among other things, should always be identified as early as possible during pre-formulation studies. Given its importance to protein formulation, especially for pharmaceutical protein formulations, a number of approaches have been developed for predicting or rapidly determining the best pH for a given formulation. For instance, accelerated stability studies and calorimetric screening studies have been demonstrated to be useful for this purpose. (See, for instance, Remmele et ah, "Minimization of recombinant human Flt3 ligand aggregation at the Tm plateau: a matter of thermal reversibility," Biochemistry 3^(16): 5241-7 (1999), which is incorporated by reference herein in its entirety particularly in parts pertinent to determining the proper pH for protein formulations as may be useful in the invention herein described.) However, these techniques generally are experimentally demanding and can require considerable material and time. There is a need for a simple predictive method in this regard that preferably does not require, or at least reduces, the burden of experimental determinations.
Various aspects and embodiments of the invention provide systematic methods for determining the appropriate and/or the optimal pH for protein formulations. In embodiments the proteins comprise an active peptide. In embodiments the proteins are antibodies. In embodiments the proteins are IgGl, IgG2, or IgG4 antibodies. In embodiments the proteins are fusion proteins. In embodiments the proteins are fusion proteins comprising an active peptide. In embodiments the proteins are Fc fusion proteins. In embodiments the proteins are Fc fusion proteins comprising an active peptide. In embodiments the proteins are Fc fusion proteins in which the Fc region is derived from an IgGl, IgG2, or IgG4 antibody. In embodiments the proteins are peptibodies. In embodiments the proteins are peptibodies comprising an active peptide and an Fc region derived from an IgGl, IgG2, or IgG4 antibody. In embodiments in these and in other respects the proteins are pharmaceutical proteins.
In accordance with various aspects and embodiments of the invention, the pi of the protein or of its active peptide is determined, preferably by a predictive method, or by empirical measurement, and the pH of the formulation is based on the thus determined pi.
In embodiments the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less. In embodiments the pi of the protein is determined by a predictive method. In embodiments the pi of the protein is determined by an empirical method. In general, predictive methods are preferred in this regard. In various aspects and embodiments of the invention in this regard, the protein is a pharmaceutically active agent. In various aspects and embodiments the formulation is a stable pharmaceutically acceptable formulation. In various aspects and embodiments the protein is a pharmaceutically active agent and the formulation is a stable, pharmaceutically acceptable formulation.
In certain embodiments of the invention the protein comprises an active peptide and the pH of the formulation is: (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less, wherein the resulting formulation is a stable pharmaceutical protein composition. In embodiments the pi of the active peptide is determined by a predictive method. In embodiments the pi of the active peptide is determined by an empirical method. In general, predictive methods are preferred in this regard. In various aspects and embodiments of the invention in this regard, the protein is a pharmaceutically active agent. In various aspects and embodiments the formulation is a stable pharmaceutically acceptable formulation. In various aspects and embodiments the protein is a pharmaceutically active agent and the formulation is a stable, pharmaceutically acceptable formulation.
In certain embodiments of the invention the protein comprises an active peptide and the pi of the active peptide substantially determines the pi of the protein.
In embodiments the protein is an Fc fusion protein comprising an Fc region and an active peptide, wherein the pi of the active peptide substantially determines the pi of the protein.
In certain embodiments the protein is a peptibody comprising an active peptide and an Fc region and the pi of the active peptide substantially determines the pi of the peptibody.
In certain embodiments, the protein is an antibody and the active peptide is a portion thereof comprising an antigen combining site of the protein.
In certain embodiments of the invention the protein is a heterotetrameric antibody, the active peptide is a portion thereof comprising an antigen binding site, and the pi of the intact heterotetrameric antibody is determined substantially by two active peptides therein each comprising one of the antibody's two antigen binding sites.
In certain aspects and embodiments the protein is an antibody and the active peptide is a peptide comprising the antigen combining side thereof. In certain aspects and embodiments in this regard the antibody is an IgG antibody, or a fragment, derivative, or modification thereof. In certain aspects and embodiments in this regard the antibody is an IgGl or IgG2 antibody, or a fragment, derivative, or modification thereof. In certain aspects and embodiments in this regard the antibody is an IgGl antibody, or a fragment, derivative, or modification thereof. In certain aspects and embodiments in this regard the antibody is an IgG2 antibody, or a fragment, derivative, or modification thereof.
In certain aspects and embodiments of the invention the protein is an Fc fusion protein comprising an Fc region and an active peptide. In certain aspects and embodiments in this regard the active peptide of the Fc fusion protein binds specifically to a target ligand. In certain aspects and embodiments in this regard the Fc region is the Fc region of an IgG antibody, or a fragment, derivative, or modification thereof. In certain aspects and embodiments in this regard the Fc region is the Fc region of an IgGl or IgG2 antibody or a fragment, derivative, or modification thereof. In certain aspects and embodiments in this regard the Fc region is the Fc region of an IgGl antibody, or a fragment, derivative, or modification thereof. In certain aspects and embodiments in this regard the Fc region is the Fc region of an IgG2 antibody, or a fragment, derivative, or modification thereof.
Fc Resions and Active Peptides
As to certain aspects and embodiments of the invention herein described pertaining to antibodies, by active peptide is meant a peptide comprising the antigen combining site of the antibody, generally other than peptides comprising intact Fc regions of the antibody. Similarly, as to certain aspects and embodiments of the invention pertaining to Fc fusion proteins that comprise in addition to an Fc region a region that binds a target, by active peptide is meant a peptide comprising the target binding site of the fusion protein, generally other than peptides comprising intact Fc regions of the Fc fusion protein.
It is to be appreciated that naturally occurring antibodies, and many Fc fusion proteins are multimeric, and that the antigen and target binding portions, respectively, thereof, are comprised of regions of more than one polypeptide. Accordingly, the use of the term active peptide herein includes not only moieties formed of a single polypeptide but also moieties formed by more than one polypeptide chain. In the latter case, therefore, an active peptide, as the term is used herein, can include two or more peptides not covalently attached to one another. Unless otherwise noted, use of the term is inclusive. Similarly, the term Fc region and/or Fc peptide are used herein explicitly to include not only the Fc region of a single polypeptide but the associate Fc regions of, for instance, the two heavy chains of a naturally occurring IgG antibody. It is to be appreciated that the term is used to refer to the intact Fc region, or to fragments, derivatives, or modifications thereof as the case may be, and may refer, inclusively of the foregoing, to either a single chain's Fc region or to the type of Fc region formed by association of two (or more) polypeptides. Unless otherwise noted, use of the term is inclusive.
1. Determining and Predicting the Isoelectric Point of a Peptide or a Protein A standard definition of isoelectric point is given by Tanford: "A protein is said to be isoelectric if its average net charge "Z" is zero." (See C. Tanford, PHYSICAL CHEMISTRY OF MACROMOLECULES, John Wiley & Sons, New York (1961), page 562.) As noted therein, an isoionic protein solution (in pure water) generally will be close to isoelectric, provided the isoelectric pH of the protein is between pH 3 and pH 11 (which is the case for most proteins). (An isoionic protein solution is one that contains only the protein and the solvent and specifically does not contain any ions other than those arising from dissociation of the solvent.)
Methods have been developed for predicting the isoelectric points of polypeptides, both small peptides, in which conformation effects on ionization often do not substantially affect pi, and for larger proteins where conformational effects may play an important role. A variety of such methods may be used in accordance with certain aspects and preferred embodiments of the invention, in which the pi or a protein or, more particularly, of an active peptide is to be determined by a predictive method.
Among suitable programs in this regard are: Sednterp vl .09 (AKA Sedimentation Interpretation Program vl .09 and Ex-PASY (Compute pI/MW)). Sednterp is a well known and readily available program for predicting polypeptide and protein pis as describe by Laue et ah, "Computer-aided interpretation of analytical sedimentation data for proteins," pp 90- 125 in Analytical Ultracentrifugation in Biochemistry and Polymer Science, Harding et ah, Eds., The Royal Society of Chemistry, Cambridge, United Kingdom (1992), which is herein incorporated by reference in its entirety, particularly in parts pertinent to predicting the pis of proteins. In addition to the foregoing, a description of the program also is available from, among other sources, the following web site: \^\[lvQ^^oJ^ήjov2jl^^oJy^άoy^^άn\£r^2 philo/descript.txt. The program itself is well known and widely available. For instance, the program is available as "freeware" that can be downloaded from a variety of web sites that can be easily identified using any standard web search program to search for the term "Sednterp." Additional information pertaining thereto, including instructions and its use for, in particular, predicting peptide and protein pis, suitable for use in accordance with the present invention is available at, among other sources: h ttp ://rasmb , bbri .or g/rasmb/windo w s /sedn terp- ph i Io . Ex-PASY (Compute pI/MW) is a well known and widely available product of the Expert Protein Analysis System ("Ex-PASY") of the Swiss Institute of Bioinformatics (SIB), which is dedicated to the analysis of protein sequences and structure (among other things). Ex-PASY in general and the Compute pI/MW tool in particular are described in Gasteiger et ah, "Protein Identification and Analysis Tools on the ExPASy Server," pp 571-607 in The
Proteomics Protocols Handbook, John M. Walker, Ed., Humana Press (2005), which is herein incorporated by reference in its entirety, particular in parts pertinent to the prediction of protein pis using Compute pI/MW. Ex-PASY is accessible via the web at http : // cxpasy.org/ . The Compute pI/Mw tool, available at the Ex-PASY site, computes the theoretical pi (isoelectric point) and Mw (molecular weight) for user entered polypeptide sequences as well as for a list of UniProt Knowledgebase (Swiss-Prot or TrEMBL) entries. Documentation on the features and use of the tool is available at http://exρasy.org/tools''ρi__tool-doc.html. Access to the tool is available at: ^^2l^SM^!Sl^!^!-^--^^M^-
While predictive methods of determining pi's are preferred in accordance with the invention herein described, especially as to determining the pi's of active peptides (as discussed elsewhere herein, methods for the empirical determination of the isoelectric points of peptides and proteins also may be used to determine pi's. The empirical determination of pi's using isoelectric focusing gel electrophoresis, for instance, is a well-known and widely available method that can be used in this regard, and there is a wide variety of readily available reagents, kits, and devices for carrying out isoelectric focusing determinations that may be useful in some aspects of the invention.
Standard techniques for isoelectric focusing are described in, among a great many other references in this regard, R. K. Scopes, PROTEIN PURIFICATION, Principles and Practice, 3rd Ed., Springer, New York (1994), particularly pages 299-300, and PROTEIN PURIFICATION METHODS, A PRACTICAL APPROACH, Harris et al, Eds., IRL Press, Oxford (1989), particularly pages 29-33, each of which is incorporated herein by reference in its entirety particularly in parts pertinent to the determining the isoelectric points of peptides and proteins as may be useful in accordance with various aspects and embodiments of the invention herein described.
Methods suitable for use in various aspects and embodiments of the invention for predicting the isoelectric points of peptides and proteins are well known to those skilled in the art, such as, for example, those described in Skoog et al., "Calculation of the isoelectric points of polypeptides from the amino acid composition," Trends Anal. Chem. 5_: 82-83 (1986) and Patrickios et al., "Polypeptide amino acid composition and isoelectric point," Anal. Biochem. 231 : 82-91 (1995), each of which is herein incorporated by reference in its entirety particularly in parts pertinent to the prediction of isoelectric points of peptides and proteins as may be useful in carrying out the invention herein described.
Additional resources for predicting isoelectric points of peptides and proteins in accordance with various aspects and embodiments of the invention herein described include, for instance, those described in Hiller et al., "JVirGel: calculation of virtual two-dimensional protein gels," Nuc. Acids Res. 3_1: 3862-65 (2003), "PHEPS: web-based pH-dependent Protein Electrostatics Server," Nuc. Acids Res. 34: Web server issue, W43 - W47 (2006), and Antosiewicz et al., "Prediction of pH-dependent Properties of Proteins," J. MoI. Biol. 238: 415-36 (1994), each of which is incorporated herein by reference in its entirety in parts pertinent to the prediction or determination of the isoelectric points of peptides and proteins as may be useful in accordance with various aspects and embodiments of the invention herein described.
2. Proteins
The invention herein disclosed may be practiced with a variety of proteins as herein described. Among preferred proteins in this regard are pharmaceutical proteins for veterinary and/or human therapeutic use, particularly proteins for human therapeutic use. Also among preferred proteins are proteins that are soluble in aqueous solutions, particularly those that are soluble at relatively high concentrations and those that are stable for long periods of time.
Further among preferred proteins of the invention are proteins for pharmaceutical formulations that do not induce a highly deleterious antigenic response following administration to a subject. Preferred in this regard are proteins for veterinary and/or human medical use, particularly, regarding the latter, humanized and human proteins.
Further among preferred proteins of the invention are proteins that bind selectively to specific targets, including ligand-binding proteins and protein ligands. Antigen-binding proteins, proteins derived therefrom, and proteins related thereto are among the particularly preferred embodiments of the invention in this regard. Highly preferred proteins of the invention in this regard are antibodies and proteins derived from antibodies or incorporating antibodies, in whole or part, including, to name just a few such entities: monoclonal antibodies, polyclonal antibodies, genetically engineered antibodies, hybrid antibodies, bi- specific antibodies, single chain antibodies, genetically altered antibodies, including antibodies with one or more amino acid substitutions, additions, and/or deletions (antibody muteins), chimeric antibodies, antibody derivatives, antibody fragments, particularly Fc fragments and fragments comprising or derived from an Fc fragment, which may be from any of the foregoing and also may be similarly engineered or modified derivatives thereof, fusion proteins comprising an antibody or a moiety derived from an antibody or from an antibody fragment, which may be any of the foregoing or a modification or derivative thereof, conjugates comprising an antibody or a moiety derived from an antibody, including any of the foregoing, or modifications or derivatives thereof, and chemically modified antibodies, antibody fragments, antibody fusion proteins, and the like, including all of the foregoing, particularly Fc fragments and fragments comprising or derived from an Fc fragment.
a. Antibodies, Antibody-Derived, and Antibody-Related Proteins and the Like Among particularly preferred proteins in accordance with the invention are antibody polypeptides, such as heavy and light chain polypeptides that have the same amino acid sequence as those that occur in and make up naturally-occurring antibodies, such as those that occur in sera and antisera, including such polypeptides and proteins isolated from natural sources, as well as those that are made by hybridoma technologies, by activation of an endogenous gene (by homologous or non-homologous recombination, for instance), by expression of an exogenous gene under the control of an endogenous transcription control region, by expression of an exogenous expression construct, by semi-synthesis and by de novo synthesis, to name some techniques commonly employed for making antibodies and antibody- related polypeptides and proteins that can be used to produce antibody polypeptides and proteins in accordance with the invention. Included among these antibody-related polypeptides and proteins are those in whole or part having a de novo amino acid sequence, those that comprise all or one or more parts of an antibody (that is: a continuous chain of amino acids having the same sequence as any four or more residues in the amino acid sequence of a naturally occurring antibody polypeptide), those having an amino acid sequence that matches in some way that of a naturally occurring antibody, but differs from it in other ways, those that have the same but different amino acid sequences as a naturally occurring counterpart or sequence relating thereto, but differ from the counterpart in one or more post-translational modifications, and those comprised in part of any of the foregoing (in part or in whole) fused to one or more polypeptide regions that can be of or derived from or related to a second, different antibody polypeptide, and can be of or derived from any other polypeptide or protein, whether naturally occurring, resembling but differing therefrom, having a semi-<ie novo amino acid sequence and/or a de novo sequence, among others. Such hybrids are generally referred to herein as fusion polypeptides and/or fusion proteins.
Further among preferred proteins in accordance with the invention herein described are modified proteins in accordance with all of the foregoing. Included among such modified proteins are proteins modified chemically by a non-covalent bond, covalent bond, or both a covalent and non-covalent bond. Also included are all of the foregoing further comprising one or more post-translational modifications which may be made by cellular modification systems or modifications introduced ex vivo by enzymatic and/or chemical methods, or introduced in other ways.
Also among preferred embodiments in accordance with various aspects and preferred embodiments of the invention in these and other regards are proteins comprising one or more CDR and/or CDR-derived and/or CDR-related regions of an antibody or one or more FR and/or FR-derived and/or FR-related regions of an antibody. In this regard CDR means complementary determining region; that is, a hypervariable region of a light or heavy chain of an antibody, typically about 9 to 12 amino acids in length that usually is an important part of an antigen specific binding moiety of an antibody. FR in this regard means a framework region of an antibody; that is, a region of about 15 to 20 amino acids that separates CDRs in the antigen specific binding moiety of an antibody. The terms CDR-derived and CDR-related, and the terms FR-derived and FR-related have the same meanings as to CDR and FR, respectively, as set forth in the above Glossary for the terms antibody-derived and antibody- related as to the term antibody. As to all of the foregoing, particularly preferred in the invention are human, humanized, and other proteins that do not engender a significantly deleterious immune response when administered to a human. Also preferred in the invention are proteins in accordance with all the foregoing that similarly do not cause a significantly deleterious immune response on administration to non-humans.
Among very particularly preferred proteins in accordance with the invention in these regards are fusion proteins comprising antibodies and/or antibody-derived proteins, polypeptides, or fragments or the like, including all of those described above. Among very particularly preferred fusion proteins of the invention in this regard are fusion proteins comprising an antibody or antibody-derived protein or fragment such as those described above and a ligand-binding moiety, such as those illustratively described herein. Among especially preferred proteins in accordance with various aspects and embodiments of the invention in this regard are peptibodies .
Included among such proteins are proteins that comprises all or part of one or more than one antibodies, such as but not limited to those that include one or more of an Fc, Fab, and Fab2 fragments or regions of antibodies and those that comprise one or more of a VH and/or a VL domain or fragment of an antibody. Among such proteins are, without limitations, single chain antibodies, scFv(s), intrabodies, maxibodies, minibodies and diabodies, to name just a few.
Included among, but without limitation, antibodies, antibody derived and antibody- related proteins in accordance with the invention are polyclonal and monoclonal antibodies, proteins derived there from , and proteins related thereto. Also included among, but without limitation, antibodies, antibody derived and antibody related protein in accordance with the invention are humanized antibodies.
Regarding antibodies, antibody-derived, and antibody-related proteins in accordance with the foregoing and with other aspects of the invention herein disclosed, see, for instance, Protein Engineering: Principles and Practice, Jeffrey L. Cleland and Chares S. Craik, eds. Wiley-Liss, Inc., New York (1996), particularly therein Kelley, Robert F., "Engineering Therapeutic Antibodies," Chapter 15, pp. 399-434 and Hollinger, P. & Hudson, P., "Engineered antibody fragments and the rise of single domains," Nature Biotechnology, September 2005, 1126-1136, each of which is herein incorporated by reference in its entirety particularly in parts pertinent to the structure and engineering of antibodies, particularly biopharmaceutical antibodies, and antibody-derived and antibody-related proteins, particularly antibody-derived and antibody-related pharmaceutical proteins in accordance with the invention herein described. b. Target Binding Proteins
Also among preferred proteins of the invention in this regard are antibodies and other types of target binding proteins, and proteins relating thereto or derived therefrom, and protein ligands, and proteins derived therefrom or relating thereto particularly those comprising an Fc region of an antibody or a region derived from an Fc region. Among especially preferred ligand-binding proteins in this regard are proteins that bind signal and effector proteins, and proteins relating thereto or derived therefrom.
Among such binding proteins, including antibodies, and Fc fusion proteins, including proteins derived therefrom and proteins related thereto, are those that bind to one or more of the following, alone or in any combination:
(i) CD proteins including but not limited to CD3, CD4, CD8, CD19, CD20, CD22, CD30, and CD34; including those that interfere with receptor binding.
(ii) HER receptor family proteins, including, for instance, HER2, HER3, HER4, and the EGF receptor; (iii) cell adhesion molecules, for example, LFA-I, MoI, pl50,95, VLA-4,
ICAM-I, VCAM, and alpha v/beta 3 integrin;
(iv) growth factors, including but not limited to, for example, vascular endothelial growth factor ("VEGF"), growth hormone, thyroid stimulating hormone, follicle stimulating hormone, luteinizing hormone, growth hormone releasing factor, parathyroid hormone, mullerian-inhibiting substance, human macrophage inflammatory protein (MIP-I- alpha), erythropoietin (EPO), nerve growth factor, such as NGF-beta, platelet-derived growth factor (PDGF), fibroblast growth factors, including, for instance, aFGF and bFGF, epidermal growth factor (EGF), transforming growth factors (TGF), including, among others, TGF-alpha and TGF-beta, including TGF-betal, TGF-beta2, TGF-beta3, TGF-beta4, or TGF-beta5, insulin-like growth factors-I and -II (IGF-I and IGF-II), des(l-3)-IGF-I (brain IGF-I), and osteoinductive factors;
(v) insulins and insulin-related proteins, including but not limited to insulin, insulin A-chain, insulin B-chain, proinsulin, and insulin-like growth factor binding proteins; (vi) coagulation and coagulation-related proteins, such as, among others, factor VIII, tissue factor, von Willebrands factor, protein C, alpha- 1 -antitrypsin, plasminogen activators, such as urokinase and tissue plasminogen activator ("t-PA"), bombazine, thrombin, and thrombopoietin; (vii) other blood and serum proteins, including but not limited to albumin, IgE, and blood group antigens;
(viii) colony stimulating factors (CSFs) and receptors thereof, including the following, among others, M-CSF, GM-CSF, and G-CSF, and receptors thereof, such as CSF-I receptor (c-fms);
(ix) receptors and receptor-associated proteins, including, for example, flk2/flt3 receptor, obesity (OB) receptor, growth hormone receptors, thrombopoietin receptors ("TPO-R," "c-mpl"), glucagon receptors, interleukin receptors, interferon receptors, T-cell receptors, stem cell factor receptors (scfr's), such as c-Kit, and other receptors listed herein; (x) receptor ligands, including, for example, OX40L, the ligand for the
OX40 receptor expressed on T cells, and other ligands listed herein;
(xi) neurotrophic factors, including but not limited to, bone-derived neurotrophic factor (BDNF) and neurotrophin-3, -4, -5, or -6 (NT-3, NT-4, NT-5, or NT-6);
(xii) relaxin A-chain, relaxin B-chain, and prorelaxin; (χϋi) interferons and interferon receptors, including for example, interferon- alpha, -beta, and -gamma, and interferon-alpha, -beta, and -gamma receptors;
(xiv) interleukins (ILs) and interleukin receptors, including but not limited to IL-I to IL- 15 and IL-I to IL- 15 receptors, such as the IL-8 receptor, among others;
(xv) viral antigens, including but not limited to, an AIDS envelope viral antigen;
(xvi) lipoproteins, calcitonin, glucagon, atrial natriuretic factor, lung surfactant, tumor necrosis factor-alpha and -beta, enkephalinase, RANTES (regulated on activation normally T-cell expressed and secreted), mouse gonadotropin-associated peptide, DNAse, inhibin, and activin; (xvii) integrin, protein A or D, rheumatoid factors, immunotoxins, bone morphogenetic protein (BMP), superoxide dismutase, surface membrane proteins, decay accelerating factor (DAF), AIDS envelope, transport proteins, homing receptors, addressins, regulatory proteins, immunoadhesins, antibodies;
(xviii) myostatins, TALL proteins, including TALL-I, amyloid proteins, including but not limited to amyloid-beta proteins, thymic stromal lymphopoietins ("TSLP"), RANK ligand ("OPGL"), c-kit, TNF receptors, including TNF Receptor Type 1, TRAIL-R2, angiopoietins, and
(xix) biologically active fragments or variants of any of the foregoing. As to all of the foregoing, particularly preferred are those that are effective therapeutic agents, particularly those that exert a therapeutic effect by binding a target, particularly a target among those listed above, including targets derived therefrom, targets related thereto, and modifications thereof. c. Particular Illustrative Proteins
Among particular illustrative proteins are certain antibody and antibody-related proteins, including Fc fusion protein and peptibodies, such as, for instance, those listed immediately below and elsewhere herein and other fusion proteins comprising an Fc region or a fragment or derivative thereof: OPGL specific antibodies, peptibodies, and related proteins, and the like (also referred to as RANKL specific antibodies, peptibodies and the like), including fully humanized and human OPGL specific antibodies, particularly fully humanized monoclonal antibodies, including but not limited to the antibodies described in International Publication Number WO 03/002713, which is incorporated herein in its entirety as to OPGL specific antibodies and antibody related proteins, particularly those having the sequences set forth therein, particularly, but not limited to, those denoted therein: 9H7; 18B2; 2D8; 2El 1; 16El; and 22B3, including the OPGL specific antibodies having either the light chain of SEQ ID NO: 2 as set forth therein in Figure 2 and/or the heavy chain of SEQ ID NO:4, as set forth therein in Figure 4, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication.
Myostatin binding proteins, peptibodies, and related proteins, and the like, including myostatin specific peptibodies, particularly those described in US Application Publication Number 2004/0181033 and International Publication Number WO2004/058988 which are incorporated by reference herein in their entirety particularly in parts pertinent to myostatin specific peptibodies, including but not limited to peptibodies of the mTN8-19 family, including those of SEQ ID NOS: 305-351, including TN8-19-1 through TN8-19-40, TN8-19 conl and TN8-19 con2; peptibodies of the mL2 family of SEQ ID NOS: 357-383; the mL15 family of SEQ ID NOS: 384-409; the mL17 family of SEQ ID NOS: 410-438; the mL20 family of SEQ ID NOS: 439-446; the mL21 family of SEQ ID NOS: 447-452; the mL24 family of SEQ ID NOS: 453-454; and those of SEQ ID NOS: 615-631, each of which is individually and specifically incorporated by reference herein in their entirety fully as disclosed in the foregoing publication. IL-4 receptor specific antibodies, peptibodies, and related proteins, and the like, particularly those that inhibit activities mediated by binding of IL-4 and/or IL- 13 to the receptor, including those described in International Publication No. WO 2005/047331 of International Application Number PCT/US2004/03742 and in US patent application publication number 2005/112694, which are incorporated herein by reference in there entirety particularly in parts pertinent to IL-4 receptor specific antibodies, particularly such antibodies as are described therein, particularly, and without limitation, those designated therein: LlHl; L1H2; L1H3; L1H4; L1H5; L1H6; L1H7; L1H8; L1H9; LlHlO; LlHI l; L2H1; L2H2; L2H3; L2H4; L2H5; L2H6; L2H7; L2H8; L2H9; L2H10; L2H11; L2H12; L2H13; L2H14; L3H1; L4H 1 ; L5H 1 ; L6H 1 , each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication.
Interleukin 1 -receptor 1 ("ILl-Rl") specific antibodies, peptibodies, and related proteins, and the like, including but not limited to those described in U.S. Application Publication Number US2004/097712A1 which is incorporated herein by reference in its entirety in parts pertinent to ILl-Rl specific binding proteins, monoclonal antibodies in particular, especially, without limitation, those designated therein: 15CA, 26F5, 27F2, 24E12, and 10H7, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the aforementioned U.S. application publication.
Ang2 specific antibodies, peptibodies, and related proteins, and the like, including but not limited to those described in International Publication Number WO 03/057134 and U.S. Application Publication Number US2003/0229023, each of which is incorporated herein by reference in its entirety particularly in parts pertinent to Ang2 specific antibodies and peptibodies and the like, especially those of sequences described therein and including but not limited to: Ll(N); Ll(N) WT; Ll(N) IK WT; 2xLl(N); 2xLl(N) WT; Con4 (N), Con4 (N) IK WT, 2xCon4 (N) IK; LlC; LlC IK; 2xLlC; Con4C; Con4C IK; 2xCon4C IK; Con4-Ll (N); Con4-LlC; TN- 12-9 (N); C17 (N); TN8-8(N); TN8-14 (N); Con 1 (N), also including anti-Ang 2 antibodies and formulations such as those described in International Publication Number WO 2003/030833 which is incorporated herein by reference in its entirety as to the same, particularly Ab526; Ab528; Ab531; Ab533; Ab535; Ab536; Ab537; Ab540; Ab543; Ab544; Ab545; Ab546; A551; Ab553; Ab555; Ab558; Ab559; Ab565; AbFlAbFD; AbFE; AbFJ; AbFK; AbGlD4; AbGClE8; AbHlC12; AbIAl; AbIF; AbIK, AbIP; and AbIP, in their various permutations as described therein, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication. NGF specific antibodies, peptibodies, and related proteins, and the like including, in particular, but not limited to those described in US Application Publication Number US2005/0074821 and US Patent Number 6,919,426 which are incorporated herein by reference in their entirety particularly as to NGF-specific antibodies and related proteins in this regard, including in particular, but not limited to, the NGF-specific antibodies therein designated 4D4, 4G6, 6H9, 7H2, 14D10 and 14Dl 1, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing publication.
CD22 specific antibodies, peptibodies, and related proteins, and the like, such as those described in US 5,789,554 which is incorporated herein by reference in its entirety as to CD22 specific antibodies and related proteins, particularly human CD22 specific antibodies, such as but not limited to humanized and fully human antibodies, including but not limited to humanized and fully human monoclonal antibodies, particularly including but not limited to human CD22 specific IgG antibodies, such as, for instance, a dimer of a human-mouse monoclonal hLL2 gamma-chain disulfide linked to a human-mouse monoclonal hLL2 kappa- chain, including, but limited to, for example, the human CD22 specific fully humanized antibody in Epratuzumab, CAS registry number 501423-23-0.
IGF-I receptor specific antibodies, peptibodies, and related proteins, and the like, such as those described in International Patent Application Number PCT/US2005/046493, which is incorporated herein by reference in its entirety as to IGF-I receptor specific antibodies and related proteins, including but not limited to the IGF-I specific antibodies therein designated LlHl, L2H2, L3H3, L4H4, L5H5, L6H6, L7H7, L8H8, L9H9, LlOHlO, Ll IHl 1, L12H12, L13H13, L14H14, L15H15, L16H16, L17H17, L18H18, L19H19, L20H20, L21H21, L22H22, L23H23, L24H24, L25H25, L26H26, L27H27, L28H28, L29H29, L30H30, L31H31, L32H32, L33H33, L34H34, L35H35, L36H36, L37H37, L38H38, L39H39, L40H40, L41H41, L42H42, L43H43, L44H44, L45H45, L46H46, L47H47, L48H48, L49H49, L50H50, L51H51, L52H52, and IGF-lR-binding fragments and derivatives thereof, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing International Application. Also among non- limiting examples of anti-IGF-lR antibodies for use in the methods and compositions of the present invention are each and all of those described in:
US Pat. App. Pub. No. 06/0040358 (published February 23, 2006), 05/0008642 (published January 13, 2005), 04/0228859 (published November 18, 2004), including but not limited to, for instance, antibody IA (DSMZ Deposit No. DSM ACC 2586), antibody 8 (DSMZ Deposit No. DSM ACC 2589), antibody 23 (DSMZ Deposit No. DSM ACC 2588) and antibody 18 as described therein;
PCT Pub. No. WO 06/138729 (published December 28, 2006), WO 05/016970 (published February 24, 2005), and Lu et al, 2004, J Biol Chem. 279:2856-65, including but not limited to antibodies 2F8, A12, and IMC-A12 as described therein;
PCT Pub. No. WO 07/012614 (published February 1, 2007), WO 07/000328 (published January 4, 2007), WO 06/013472 (published February 9, 2006), and 05/058967 (published June 30, 2005), 03/059951 (published July 24, 2003); US Pat. App. Pub. No. 05/0084906 (published April 21, 2005), including but not limited to antibody 7C10, chimaeric antibody C7C10, antibody h7C10, antibody 7H2M, chimaeric antibody *7C10, antibody GM 607, humanized antibody 7C10 version 1, humanized antibody 7C10 version 2, humanized antibody 7C10 version 3, and antibody 7H2HM, as described therein; US Pat. App. Pub. No. 05/0249728 (published November 10, 2005), 05/0186203
(published August 25, 2005), 04/0265307 (published December 30, 2004), 03/0235582 (published December 25, 2003) and Maloney et al., 2003, Cancer Res. 63:5073-83, including but not limited to antibody EM164, resurfaced EM164, humanized EM164, huEM164 vl .0, huEM164 vl.l, huEM164 vl.2, and huEM164 vl.3 as described therein; US Pat. No. 7,037,498 (issued May 2, 2006), US Pat. App. No. 05/0244408 (published
November 30, 2005), 04/0086503 (published May 6, 2004), Cohen, et al., 2005, Clinical Cancer Res. 11 :2063-73, e.g., antibody CP-751,871, including but not limited to each of the antibodies produced by the hybridomas having the ATCC accession numbers PTA-2792, PTA-2788, PTA-2790, PTA-2791, PTA-2789, PTA-2793, and antibodies 2.12.1, 2.13.2, 2.14.3, 3.1.1, 4.9.2, and 4.17.3, as described therein;
US Pat. App. No. 05/0136063 (published June 23, 2005), 04/0018191 (published January 29, 2004), including but not limited to antibody 19Dl 2 and an antibody comprising a heavy chain encoded by a polynucleotide in plasmid 15H12/19D12 HCA (γ4), deposited at the ATCC under number PTA-5214, and a light chain encoded by a polynucleotide in plasmid 15H12/19D12 LCF (K), deposited at the ATCC under number PTA-5220, as described therein; US Pat. App. No. 04/0202655 (published October 14, 2004), including but not limited to antibodies PINT-6A1, PINT-7A2, PINT-7A4, PINT-7A5, PINT-7A6, PINT-8A1, PINT- 9A2, PINT-I IAl, PINT-11A2, PINT-11A3, PINT-11A4, PINT-11A5, PINT-11A7, PINT- 11A12, PINT-12A1, PINT-12A2, PINT-12A3, PINT-12A4, and PINT-12A5, as described therein;
Each and all of which are herein incorporated by reference in their entireties, particularly as to the aforementioned antibodies, peptibodies, and related proteins and the like that target IGF-I receptors.
B-7 related protein 1 specific antibodies, peptibodies, related proteins and the like ("B7RP-1," also is referred to in the literature as B7H2, ICOSL, B7h, and CD275), particularly B7RP-specifϊc fully human monoclonal IgG2 antibodies, particularly fully human IgG2 monoclonal antibody that binds an epitope in the first immunoglobulin-like domain of B7RP- 1 , especially those that inhibit the interaction of B7RP- 1 with its natural receptor, ICOS, on activated T cells in particular, especially, in all of the foregoing regards, those disclosed in U.S. Provisional Application Number 60/700,265, filed 18 July 2005 and International Publication Number WO07/011941, which are incorporated herein by reference in their entireties as to such antibodies and related proteins, including but not limited to antibodies designated therein as follow: 16H (having light chain variable and heavy chain variable sequences SEQ ID NO:1 and SEQ ID NO: 7 respectively therein); 5 D (having light chain variable and heavy chain variable sequences SEQ ID NO:2 and SEQ ID NO:9 respectively therein); 2H (having light chain variable and heavy chain variable sequences SEQ ID NO:3 and SEQ ID NO: 10 respectively therein); 43H (having light chain variable and heavy chain variable sequences SEQ ID NO:6 and SEQ ID NO: 14 respectively therein); 41H (having light chain variable and heavy chain variable sequences SEQ ID NO:5 and SEQ ID NO: 13 respectively therein); and 15H (having light chain variable and heavy chain variable sequences SEQ ID NO:4 and SEQ ID NO: 12 respectively therein), each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing U.S. Provisional Application.
IL- 15 specific antibodies, peptibodies, and related proteins, and the like, such as, in particular, humanized monoclonal antibodies, particularly antibodies such as those disclosed in U.S. Application Publication Numbers: US2003/0138421; US2003/023586; US2004/0071702; and US Patent Number 7,153,507 each of which is incorporated herein by reference in its entirety as to IL- 15 specific antibodies and related proteins, including peptibodies, including particularly, for instance, but not limited to, HuMax IL- 15 antibodies and related proteins, such as, for instance, 146B7. IFN gamma specific antibodies, peptibodies, and related proteins and the like, especially human IFN gamma specific antibodies, particularly fully human anti-IFN gamma antibodies, such as, for instance, those described in US Application Publication Number US2005/0004353, which is incorporated herein by reference in its entirety as to IFN gamma specific antibodies, particularly, for example, the antibodies therein designated 1118; 1118*; 1119; 1121; and 1121* each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing US Application Publication.
TALL-I specific antibodies, peptibodies, and the related proteins, and the like, and other TALL specific binding proteins, such as those described in U.S. Application Publication Numbers 2003/0195156 and 2006/135431 each of which is incorporated herein by reference in its entirety as to TALL-I binding proteins, particularly the molecules of Tables 4 and 5B, each of which is individually and specifically incorporated by reference herein in its entirety fully as disclosed in the foregoing US Application Publication.
Parathyroid hormone ("PTH") specific antibodies, peptibodies, and related proteins, and the like, such as those described in US Patent Number 6,756,480, which is incorporated herein by reference in its entirety, particularly in parts pertinent to proteins that bind PTH.
Thrombopoietin receptor ("TPO-R") specific antibodies, peptibodies, and related proteins, and the like, such as those described in US Patent Number 6,835,809, which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind TPO-R.
Hepatocyte growth factor ("HGF") specific antibodies, peptibodies, and related proteins, and the like, including those that target the HGF/SF:cMet axis (HGF/SF:c-Met), such as the fully human monoclonal antibodies that neutralize hepatocyte growth factor/scatter (HGF/SF) described in US Patent Application Publication Number US2005/0118643 and International Publication Number WO2005/017107, huL2G7 described in US Patent Number 7,220,410 and OA-5d5 described in US Patent Numbers 5,686,292, 6,468,529, and in International Publication Number WO 96/38557, each of which is incorporated herein by reference in its entirety, particularly in parts pertinent to proteins that bind HGF.
TRIAL-R2 specific antibodies, peptibodies, related proteins and the like, such as those described in US Provisional Applications 60/713,433 filed 31 August 2005 and 60/713,478 filed 31 August 2005, each of which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind TRAIL-R2. Activin A specific antibodies, peptibodies, related proteins, and the like, including but not limited to those described in US Provisional Patent Application Number 60/843,430 filed
8 September 2006, which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind Activin A. TGF-beta specific antibodies, peptibodies, related proteins, and the like, including but not limited to those described in US Patent Number 6,803,453 and US Patent Application
Publication Number 2007/110747, each of which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind TGF-beta.
Amyloid-beta protein specific antibodies, peptibodies, related proteins, and the like, including but not limited to those described in International Publication Number
WO2006/081171, which is herein incorporated by reference in its entirety, particularly in parts pertinent to proteins that bind amyloid-beta proteins. c-Kit specific antibodies, peptibodies, related proteins, and the like, including but not limited to those described in US Provisional Patent Application Number 60/794,771, which is incorporated herein by reference in its entirety, particularly in parts pertinent to proteins that bind c-Kit and/or other stem cell factor receptors.
OX40L specific antibodies, peptibodies, related proteins, and the like, including but not limited to those described in US Patent Application Number 11/068,289, which is incorporated herein by reference in its entirety, particularly in parts pertinent to proteins that bind OX40L and/or other ligands of the OXO40 receptor.
Activase® (Alteplase, tPA); Aranesp® (Darbapoetin-alfa, Erythropoietin, rHuEpo);
Avonex® (Interferon beta- Ia); Bexxar® (Tositumomab, anti-CD22 monoclonal antibody);
Betaseron® (Interferon-beta); Campath® (Alemtuzumab, anti-CD52 monoclonal antibody);
Enbre®l (etanercept, TNF-receptor /Fc fusion protein, TNF blocker); Eprex® (Erythropoietin, rHuEpo); Erbitux® (Cetuximab, anti-EGFR / HERl / c-ErbB-1); Genotropin® (Somatropin,
Human Growth Hormone); Herceptin(r) (Trastuzumab, anti-HER2/neu (erbB2) receptor mAb); Humatrope® (Somatropin, Human Growth Hormone); Humira® (Adalimumab);
Insulin in Solution; Infergen® (Interferon Alfacon-1); Kineret® (Anakinra), Leukine®
(Sargamostim, rhuGM-CSF); LymphoCide® (Epratuzumab, anti-CD22 mAb); Lymphostat B® (Belimumab, anti-BlyS mAb); Metalyse® (Tenecteplase, t-PA analog); Mylotarg®
(Gemtuzumab ozogamicin); NeoRecormon® (Epoetin-beta, Erythropoietin, rHuEpo);
Neumega® (Oprelvekin, Human Interleukin-11); Neulasta® (Pegylated filgastrim, pegylated
G-CSF, pegylated hu-Met-G-CSF); Neupogen® (Filgrastim , G-CSF, hu-MetG-CSF); Orthoclone OKT3® (Muromonab-CD3, anti-CD3 monoclonal antibody), Remicade® (Infliximab, anti-TNF-alpha monoclonal antibody), Reopro® (Abciximab, anti-GP lib/Ilia receptor monoclonal antibody), Rituxan® (Rituximab, anti-CD20 mAb); Roferon-A®- (Interferon alfa-2a); Simulect® (Basiliximab); Synagis® (Palivizumab); Tysabri® (Natalizumab, anti-alpha4integrin mAb); Valortim® (MDX-1303, anti-B. anthracis Protective Antigen mAb); Vectibix® (Panitumumab); Xolair® (Omalizumab), Zenapx® (Daclizumab); Zenapax® (Daclizumab, anti-IL-2Rα mAb) and Zevalin® (Ibritumomab tiuxetan).
BR2-Fc (huBR3 / huFc fusion protein, soluble BAFF antagonist); CNTO 148 (Golimumab, anti-TNFα mAb); HGS-ETRl (Mapatumumab; human anti-TRAIL Receptor- 1 mAb); HuMax-CD20 (Ocrelizumab, anti-CD20 human mAb); HuMax-EGFR (zalutumumab); HuMax-EGFr (Zalutumumab); M200 (Volociximab, anti -α5βl integrin mAb); MDX-OlO (Ipilimumab, anti-CTLA-4 mAb and VEGFR-I (IMC- 18Fl););
Anti-BR3 mAb; anti-C. difficile Toxin A and Toxin B C mAbs MDX-066 (CDA-I) and MDX-1388); anti-CD22 dsFv-PE38 conjugates (CAT-3888 and CAT-8015); anti-CD25 mAb (HuMax-TAC); anti-CD3 mAb (NI-0401); anti-CD30 mAb (MDX-060); anti-CD38 mAb (HuMax CD38); anti-CD40L mAb; anti-Cripto mAb; anti-CTGF Idiopathic Pulmonary Fibrosis Phase I Fibrogen (FG-3019); anti-CTLA4 mAb; anti-eotaxinl mAb (CAT-213); anti- FGF8 mAb; anti-ganglioside GD2 mAb; anti-ganglioside GM2 mAb; anti-GDF-8 human mAb (MYO-029); anti-GM-CSF Receptor mAb (CAM-3001); anti-HepC mAb (HuMax HepC); anti-IFN-alpha mAb (MEDI-545, MDX-1103); anti-IGFIR mAb; anti-IGF-lR mAb (HuMax-Inflam); anti-IL12 mAb (ABT-874); anti-IL12/IL23 mAb (CNTO 1275); anti-IL13 mAb (CAT-354); anti-IL2Ra mAb (HuMax-TAC); anti-IL5 Receptor mAb; anti-integrin receptors mAb (MDX-018, CNTO 95); anti-IP 10 Ulcerative Colitis mAb (MDX-1100); anti- LLY antibody; anti-Mannose Receptor/hCGβ mAb (MDX-1307); anti-mesothelin dsFv-PE38 conjugate (CAT-5001); anti-PDlmAb (MDX-1106 (ONO-4538)); anti-PDGFRα antibody (IMC-3G3); anti-TGF-beta mAb (C- 1008); anti-TRAIL Receptor-2 human mAb (HGS- ETR2); anti-TWEAK mAb; anti-VEGFR/Flt-1 mAb; anti-ZP3 mAb (HuMax-ZP3); NVS Antibody #1; and NVS Antibody #2.
d. Sequence Variation
Particularly preferred proteins in regard to all of the foregoing and the following, include those that comprise a region that is 70% or more, especially 80% or more, more especially 90% or more, yet more especially 95% or more, particularly 97% or more, more particularly 98% or more, yet more particularly 99% or more identical in amino acid sequence to a reference amino acid sequence of a binding protein, as illustrated above, particularly a pharmaceutical binding protein, such as a GenBank or other reference sequence of a reference protein. Identity in this regard can be determined using a variety of well-known and readily available amino acid sequence analysis software. Preferred software includes those that implement the Smith- Waterman algorithms, considered a satisfactory solution to the problem of searching and aligning sequences. Other algorithms also may be employed, particularly where speed is an important consideration. Commonly employed programs for alignment and homology matching of DNAs, RNAs, and polypeptides that can be used in this regard include FASTA, TFASTA, BLASTN, BLASTP, BLASTX, TBLASTN, PROSRCH, BLAZE, and MPSRCH, the latter being an implementation of the Smith-Waterman algorithm for execution on massively parallel processors made by MasPar.
The BLASTN, BLASTX, and BLASTP programs are among preferred programs for such determinations, the former for polynucleotide sequence comparisons and the latter two for polypeptide sequence comparisons; particularly BLASTX for comparison of the polypeptide sequences from all three reading frames of polynucleotide sequence and BLASTP for a single polypeptide sequence.
BLAST provides a variety of user definable parameters that are set before implementing a comparison. Some of them are more readily apparent than others on graphical user interfaces, such as those provided by NCBI BLAST and other sequence alignment programs that can be accessed on the internet. The settings and their values are set out and explained on the service web sites and are explained and set out in particular detail in a variety of readily available texts, including but not limited to BIOINFORMATICS: SEQUENCE AND GENOME ANALYSIS, 2nd Ed., David W. Mount, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (2004), especially Chapters 3, 4, 5, and 6 as to comparison of protein and nucleic acid sequences in general and as to BLAST comparisons and searches in particular; SEQUENCE ANALYSIS IN A NUTSHELL: A GUIDE TO COMMON TOOLS AND DATABASES, Scott Markel and Darryl Leon, O'Reilly & Associates, Sebastopol, California (2003), especially Chapter 7 as to BLAST in particular, each of which is herein incorporated by reference in its entirety particularly in parts pertinent to comparison of nucleotide and polypeptide sequences and to determining their degree of identity, similarity, homology and/or the like, especially as to comparison of a test sequence and a reference sequence to calculate a degree (percent) of identity between them.
In preferred embodiments of the invention in this regard, relatedness of sequences is defined as the identity score in percent returned by any one or another of the aforementioned BLAST comparison searches with e =10 and all other parameters set to their default values on the NCBI web server as set forth in SEQUENCE ANALYSIS IN A NUTSHELL: A GUIDE TO COMMON TOOLS AND DATABASES, Scott Markel and Darryl Leon, O'Reilly & Associates, Sebastopol, California (2003), pages 47-51 which are incorporated herein by reference in their entireties and in all particulars of the preferred settings for parameters of the present invention for comparing sequences using BLAST, such as those on NCBI BLAST.
The following references provide additional information on sequence comparisons in this regard, and in others. GUIDE TO HUMAN GENOME COMPUTING, Ed. Martin J. Bishop, Academic Press, Harcourt Brace & Company Publishers, New York (1994), which is incorporated herein by reference in its entirety with regard to the foregoing, particularly in parts pertinent to determining identity and or homology of amino acid or polynucleotide sequences, especially Chapter 7. The BLAST programs are described in Altschul et al., "Basic Local Alignment Research Tool," J MoI Biol 215: 403-410 (1990), which is incorporated by reference herein in its entirety. Additional information concerning sequence analysis and homology and identity determinations are provided in, among many other references well-known and readily available to those skilled in the art: NUCLEIC ACID
AND PROTEIN SEQUENCE ANALYSIS: A PRACTICAL APPROACH, Eds. M. J. Bishop and C. J. Rawings, IRL Press, Oxford, UK (1987); PROTEIN STRUCTURE: A PRACTICAL APPROACH, Ed. T. E. Creighton, IRL Press, Oxford, UK (1989); Doolittle, R. F.: "Searching through sequence databases," Met Enz. 183: 99-110 (1990); Meyers and Miller: "Optimal alignments in linear space" Comput. Applica. in Biosci 4: 11-17 (1988);
Needleman and Wunsch: "A general method applicable to the search for similarities in amino acid sequence of two proteins," J MoI Biol 48: 443-453 (1970) and Smith and Waterman "Identification of common molecular subsequences," J MoI Biol 147: 1950 et seq. (1981), each of which is incorporated herein by reference in its entirety with reference to the foregoing, particularly in parts pertinent to sequence comparison and identity and homology determinations.
Particularly preferred embodiments in this regard have 50% to 150% of the activity of the aforementioned reference protein, particularly highly preferred embodiments in this regard have 60% to 125% of the activity of the reference protein, yet more highly preferred embodiments have 75% to 110% of the activity of the reference protein, still more highly preferred embodiments have 85% to 125% the activity of the reference, still more highly preferred embodiments have 90% to 110% of the activity of the reference. 3. Formulations a. Some General Aspects of Formulations
Many reagents and methods conventionally employed for the formulation of protein pharmaceuticals can be used for the formulation of compositions in accordance with various aspects and preferred embodiments of the invention. However, embodiments of the invention are based on the pi of the protein and/or on that of an active peptide of a protein.
In many other respects, compositions in accordance with various aspects and embodiments of the invention can be formulated using reagents and methods conventionally employed for the formulation of proteins, in particular, reagents and methods employed for the formulation of pharmaceuticals, including pharmaceuticals for veterinary and human use, especially those reagents and methods suitable for formulating protein pharmaceuticals for veterinary and especially for human use.
In accordance therewith, many methods and ingredients for formulating and using pharmaceuticals that are well-known and routine in the pertinent arts can be used in designing, making, and using protein formulations in accordance with various aspects and preferred embodiments of the invention relating thereto. Such methods and ingredients are described in, to name just a few readily available references in this regard, REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY, 21st Ed.; Beringer et al. Editors, Lippincott, Williams & Wilkins, Philadelphia, PA (2005); ANSEL'S PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 8th Ed., Allen et al, Editors, Lippincott, Williams & Wilkins, Philadelphia, PA (2005); and PHARMACEUTICAL FORMULATION OF PEPTIDES AND PROTEINS, Sven Frokjaer and Lars Hovgaard, Editors, CRC Press, Boca Raton, Florida (2000), each of which is herein incorporated in its entirety particularly in parts pertinent to conventional ingredients and methods that may be used in formulations of proteins in accordance with various aspects and embodiments of the invention relating thereto. Additional methods and ingredients that can be useful in this regard are disclosed in, among others, US 6,171,586; WO 2005/044854; US 6,288,030; US 6,267,958; WO 2004/055164; US 4,597,966; US 2003/0138417; US 6,252,055; US 5,608,038; US 6,875,432; US 2004/0197324; WO 02/096457; US 5,945,098; US 5,237,054; US 6,485,932; US 6,821,515; US 5,792,838; US 5,654,403; US 5,908,826; EP 0 804 163; and WO 2005/063291, each of which is incorporated herein by reference in its entirety particularly in parts pertinent to pharmaceutically acceptable protein formulations in accordance with the invention.
Various specific aspects of the ingredients and specific types of formulations are further described below, by way of illustration. The description thus provided is not exhaustive of the methods and compositions possible for protein formulations in accordance with the various aspects and embodiments of the invention, nor is it in any way exclusive.
In preferred embodiments of a variety of aspects of the invention, formulations of proteins comprise a protein and a carrier, which also may be referred to herein variously, as the case may be, as one or more of: a vehicle, a primary vehicle, a diluent, a primary diluent, a primary carrier, a solvent and/or a primary solvent. In the broadest sense the carrier may be a gas, a liquid, or a solid, as suits the phase of the composition and/or its use(s). In some embodiments of the invention in this regard, the carrier is a solid, such as a powder in which a protein may be dispersed. In preferred embodiments in this regard, the carrier is a liquid, particularly a liquid in which the protein is highly soluble, particularly at concentrations that provide the desired buffer capacity. Liquid carriers may be organic or non-organic. Preferably they are aqueous, most preferably they are largely or entirely comprised of pure water.
It will be appreciated that formulations for pharmaceutical use in accordance with various aspects and embodiments of the invention must be compatible with the processes and conditions to which they will be subjected, such as, for instance, sterilization procedures (generally applied before mixing with an active agent), and conditions during storage.
Almost invariably, formulations in accordance with numerous aspects and embodiments of the invention will contain additional ingredients including but not limited in any way to excipients and other pharmaceutical agents.
Formulations in accordance with various aspects and embodiments of the invention may contain, among others, excipients, as described below, including but not limited to ingredients for modifying, maintaining, or preserving, for example, osmolality, osmolarity, viscosity, clarity, color, tonicity, odor, sterility, stability, rate of dissolution or release, adsorption or penetration of the formulations and/or primary polypeptide and/or protein.
Formulations will, of course, depend upon, for example, the particular protein being formulated, the other active agents, such as other pharmaceuticals, that will be comprised in the formulation, the intended route of administration, the method of administration to be employed, the dosage, the dosing frequency, and the delivery format, among others.
Formulations in accordance with embodiments of the invention provide protein compositions comprising a buffering agent, wherein the concentration of the buffering agent is any of 0.5 to 5.0, 1.0 to 10, 2.0 to 20, 5.0 to 50, 10 to 100, 50 to 150, 100 to 200, 100 to 500, 150 to 300, 200 to 500, 250 to 500 mM or more. Among buffering agents and buffers useful in this regard, for example without limitation as further described elsewhere herein are: acetate, phosphate, histidine, and succinate.
In embodiments the proteins are self-buffering. In embodiments, the formulations comprise a protein and a solvent, wherein at a desired pH, the protein is self-buffering and thereby provides at least approximately 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 99.5% of the buffer capacity of the composition.
Formulations in accordance with certain of the preferred embodiments in various aspects of the invention provide protein compositions, particularly pharmaceutical protein compositions, comprising a protein and a solvent, A, wherein the concentration of the protein is between approximately: 0.1 and 10, 0.1 and 25, 0.1 and 50, 0.1 and 100, 0.1 and 150, 0.1 and 200, 0.1 and 250, 0.1 and 300, 0.1 and 400, 0.1 and 400 or higher mg/ml; 0.2 and 10, 0.2 and 25, 0.2 and 50, 0.2 and 100, 0.2 and 150, 0.2 and 200, 0.2 and 250, 0.2 and 300, 0.2 and 400, 0.2 and 400 or higher mg/ml; 0.5 and 10, 0.5 and 25, 0.5 and 50, 0.5 and 100, 0.5 and 150, 0.5 and 200, 0.5 and 250, 0.5 and 300, 0.5 and 400, 0.5 and 400 or higher mg/ml; 1.0 and 10, 1.0 and 25, 1.0 and 50, 1.0 and 100, 1.0 and 150, 1.0 and 200, 1.0 and 250, 1.0 and 300, 1.0 and 400, 1.0 and 400 or higher mg/ml; 5.0 and 10, 5.0 and 25, 5.0 and 50, 5.0 and 100, 5.0 and 150, 5.0 and 200, 5.0 and 250, 5.0 and 300, 5.0 and 400, 5.0 and 400 or higher mg/ml; 10 and 25, 10 and 50, 10 and 100, 10 and 150, 10 and 200, 10 and 250, 10 and 300, 10 and 400, 10 and 400 or higher mg/ml; 25 and 50, 25 and 100, 25 and 150, 25 and 200, 25 and 250, 25 and 300, 25 and 400, 25 and 400 or higher mg/ml; 50 and 100, 50 and 150, 50 and 200, 50 and 250, 50 and 300, 50 and 400, 50 and 400 or higher mg/ml; 150 and 300, 200 and 400, 200 and 400 or higher mg/ml.
Formulations in accordance with certain of the preferred embodiments in various aspects of the invention provide protein compositions, particularly pharmaceutically acceptable protein compositions, comprising a protein and a solvent, further comprising a salt, wherein the salt may be any of about 0.5, 1, 2, 5, 10, 15, 20, 25, 50, 75, 100, 125, 150, 175, 200, 250, 300, 350, 400, 500 or more mM and may be in the range of any of 0.5 to 5, 1 to 10, 5 to 25, 15 to 35, 25 to 50, 35 to 75, 50 to 100, 75 to 150, 100 to 200, 150 to 250, 200 to 300, 250 to 400, 300 to 500, 400 to 600 niM or higher. In certain of the embodiments in this regard the foregoing, concentrations refer to the concentration of the salt in the solvent prior to addition of the protein, which may be associated with ions that then may contribute to the salt concentration. In this regard the concentrations are those of the solvent with which the protein is formulated. In other embodiments the foregoing salt concentrations refer to the total salt concentration of the formulation including the contribution of the solvent and any contribution due to protein associated salts.
Formulations in accordance with certain of the preferred embodiments in various aspects of the invention provide protein compositions, particularly pharmaceutical protein compositions, comprising a protein and a solvent, and further comprising one or more pharmaceutically acceptable salts; osmotic balancing agents (tonicity agents); anti-oxidants; antibiotics; antimycotics; bulking agents; lyoprotectants; anti-foaming agents; chelating agents; preservatives; colorants; analgesics; or additional pharmaceutical agents. Formulations in accordance with certain of the preferred embodiments in various aspects of the invention provide protein compositions, particularly pharmaceutical protein compositions, comprising a protein and a solvent, and further comprising one or more pharmaceutically acceptable polyols in an amount that is hypotonic, isotonic, or hypertonic, preferably approximately isotonic, particularly preferably isotonic, especially preferably any one or more of sorbitol, mannitol, sucrose, trehalose, or glycerol, particularly especially preferably approximately 5% sorbitol, 5% mannitol, 9% sucrose, 9% trehalose, or 2.5% glycerol, very especially in this regard 5% sorbitol, 5% mannitol, 9% sucrose, 9% trehalose, or 2.5% glycerol. The foregoing polyols and concentrations thereof are provided solely by way of illustration and are in no way exclusive or exhaustive of polyols and their use in accordance with embodiments of the invention.
Formulations in accordance with certain of the preferred embodiments in various aspects of the invention provide protein compositions, particularly pharmaceutical protein compositions, comprising a protein and a solvent, and further comprising one or more pharmaceutically acceptable surfactants, preferably one or more of polysorbate 20, polysorbate 80, other fatty acid esters of sorbitan, polyethoxylates, and poloxamer 188, particularly preferably polysorbate 20 or polysorbate 80, preferably approximately 0.001 to 0.1% polysorbate 20 or polysorbate 80, very preferably approximately 0.002 to 0.02% polysorbate 20 or polysorbate 80, especially 0.002 to 0.02% polysorbate 20 or polysorbate 80. Many other such surfactants may be employed in embodiments of the invention. Included among such others are the following: Tween 20, including but not limited to from about 0.0005% or about 0.01% Tween 20; sodium cholate, including but not limited to from about 0.001% to about 0.01% sodium cholate; sodium glycholate, including but not limited to from about 0.001% to about 0.01% sodium glycholate; sodium deoxy cholate, including but not limited to from about 0.001% to 0.01% sodium deoxycholate; sodium glycodeoxycholate, including but not limited to from about 0.001% to about 0.01% sodium glycodeoxycholate; CHAPS, including but not limited to from about 0.001% to about 0.01% CHAPS; CHAPSO, including but not limited to from about 0.001% to about 0.01% CHAPSO; Emphigen BB, including but not limited to from about 0.001% to about 0.01% Emphigen BB; SDS, including but not limited to from about 0.001% to about 0.01% SDS; Mega-8, including but not limited to from about 0.001% to about 0.01% Mega-8; Genepol C-100, including but not limited to from about 0.001% to about 0.01% Genepol C-100; Brij 35, including but not limited to from about 0.001% to about 0.01% Brij 35; Pluronic F-68, including but not limited to from about 0.001% to about 0.01% Pluronic F-68; Pluronic F-127, including but not limited to from about 0.001% to about 0.01% Pluronic F-127; Zwittergent 3-12, including but not limited to from about 0.001% to about 0.01% Zwittergent 3-12; PEG-8000, including but not limited to from about 0.001% to about 0.01% PEG-8000; PEG-4000, including but not limited to from about 0.001% to about 0.01% PEG-4000; HPCD, including but not limited to from about 0.001% to about 0.1% HPCD; and Triton X- 100, including but not limited to from about 0.001 % to about 0.01% Triton X-100.
Formulations in accordance with various aspects and embodiments of the invention are stable for at least any of 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50 or more months. Formulations in accordance with certain of the preferred embodiments in various aspects of the invention provide protein compositions, particularly pharmaceutical protein compositions, comprising a protein and a solvent, wherein the protein is a pharmaceutical agent and the composition is a sterile formulation thereof suitable for treatment of a veterinary or a human medical subject. Also among formulations in accordance with various aspects and embodiments of the invention herein described are lyophilized compositions in accordance with the foregoing, particularly lyophilized compositions that when reconstituted provide a formulation as described above and elsewhere herein. b. Excipients and Other Additional Ingredients
As discussed above, certain embodiments in accordance with aspects of the invention provide protein compositions, particularly pharmaceutical protein compositions, that comprise, in addition to the protein, one or more excipients such as those illustratively described in this section and elsewhere herein. Excipients can be used in the invention in this regard for a wide variety of purposes, such as adjusting physical, chemical, or biological properties of formulations, such as adjustment of viscosity, and or processes of the invention to improve effectiveness and or to stabilize such formulations and processes against degradation and spoilage due to, for instance, stresses that occur during manufacturing, shipping, storage, pre-use preparation, administration, and thereafter.
A variety of expositions are available on protein stabilization and formulation materials and methods useful in this regard, such as Arakawa et al., "Solvent interactions in pharmaceutical formulations," Pharm Res. 8(3): 285-91 (1991); Kendrick et al, "Physical stabilization of proteins in aqueous solution," in: RATIONAL DESIGN OF STABLE PROTEIN FORMULATIONS : THEORY AND PRACTICE, Carpenter and Manning, eds. Pharmaceutical Biotechnology. 13: 61-84 (2002), and Randolph et al., "Surfactant-protein interactions," Pharm Biotechnol. 13: 159-75 (2002), each of which is herein incorporated by reference in its entirety, particularly in parts pertinent to excipients and processes of the same for formulations in accordance with the current invention, especially as to protein pharmaceutical products and processes for veterinary and/or human medical uses.
Various excipients useful in the invention are listed in Table 1 and further described below.
Table 1: Types of Excipients and Their Functions
Figure imgf000062_0001
i. Buffering agents
Buffering agents are used almost universally to establish and maintain the pH of protein formulations.
A wide variety of buffering agents suitable for use in protein formulations are well known. Each of them works over a relatively narrow range of pH. Moreover, each has well known advantages and drawbacks. As is well known to those skilled in the art of formulating proteins, particularly for formulating pharmaceutical proteins, care must be exercised to choose a buffering agent with the needed capacity in the desired pH range in which the protein will be stable for the desired period of time and that does not present ancillary proteins. Agents that may be used in formulating protein compositions include, but are not limited to, acetate, citrate, succinate, phosphate, certain amino acids, "Tris," Good's buffers (MES, ADA, PIPES, ACES, Cholamine chloride, BES, TES, HEPES, Acetamidoglycine, Tricine, Glycinamide and Bicine), certain other organic acids, and certain polyanionic carboxylate buffers (including the aforementioned citrate and succinate). Several factors need to be considered when choosing a buffer. First and foremost, the buffer species and its concentration need to be defined based on its pKa and the desired formulation pH. Equally important is to ensure that the buffer is compatible with the protein drug, other formulation excipients, and does not catalyze any degradation reactions. It is also important in pharmaceutical compositions to consider the possibility that a given buffering agent will be unacceptable for administration for ancillary reasons, such as deleterious effects on patient comfort. Some buffering agents are unsuitable for this purpose because they cause stinging or irritation at the point of administration. For example, citrate is known to cause stinging upon injection. Such effects are more pronounced for SC and IM administration, because the formulation remains at the administration site for some time, than for IV administration, where the formulation is diluted immediately. For IV administration, however, the total load of buffering agent administered must be assessed, because the buffering agent itself can be harmful to the patient. Potassium ions, for instance, administered in potassium phosphate buffer, can cause cardiovascular complications. See Hollander et al. for instance, Am. Fam. Physician. 73(2): 283-90 (2006). Additional considerations apply to the choice of a buffer for a lyophilized formulation.
For instance, some buffers, sodium phosphate for example, crystallize out of the protein amorphous phase during freezing, with consequent, large and undesirable shifts in pH. Other common buffering agents, acetate and imidazole for example, should not be used to prepare lyophilates because they sublime or evaporate during lyophilization, with resultant, undesirable shifts in pH, both during lyophilization and/or upon or after reconstitution. ii. Osmolality (tonicity) adjusting agents
Non-ionic and ionic agents may be used to adjust the osmolality (tonicity) of compositions in accordance with the invention, including many well known and other lesser known compounds useful for this purpose. Salts are useful in this regard, for instance. In embodiments, NaCl is used as a tonicifying agent. In embodiments KCl, LiCl or another salt is used as a tonicifying agent, alone or in combination with other tonicifying agents. In embodiments, a non-ionic substance is used to adjust tonicity. In embodiments glycerine is used. In embodiments a polyol is used to adjust tonicity, such as but not limited to the polyols described specifically elsewhere herein. iii. Salts
Salts may be used in accordance with embodiments of the invention to, for example, adjust the ionic strength and/or the isotonicity of a formulation and/or to improve the physical stability of a protein or other ingredient(s) of a composition. In embodiments salts prevent or reduce protein insolubility and/or aggregation. In embodiments salts also are effective for reducing the viscosity of protein formulations.
Among salts useful in embodiments of the invention are NaCl and KCl to name just two. In embodiments the concentration of salt is between 10 and 250 mM. In embodiments is it between 20 and 200 mM. In embodiments it is between 50 and 200 mM. In embodiments it is between 50 and 150 mM.
As is well known, ions can stabilize the native state of proteins by binding to charged residues on the protein's surface and by shielding charged and polar groups in the protein and reducing the strength of their electrostatic interactions, attractive, and repulsive interactions. Ions also can stabilize the denatured state of a protein by binding to, in particular, the denatured peptide linkages (-CONH) of the protein. Furthermore, ionic interaction with charged and polar groups in a protein also can reduce intermolecular electrostatic interactions and, thereby, prevent or reduce protein aggregation and insolubility.
Ionic species differ significantly in their effects on proteins. A number of categorical rankings of ions and their effects on proteins have been developed that can be used in formulating compositions in accordance with the invention. One example is the Hofmeister series, which ranks ionic and polar non-ionic solutes by their effect on the conformational stability of proteins in solution. As to a given protein and conditions, stabilizing solutes may be referred to as "kosmotropic," and destabilizing solutes may be referred to as chaotropic. Kosmotropes commonly are used at high concentrations (e.g., >1 molar ammonium sulfate) to precipitate proteins from solution ("salting-out"). Chaotropes commonly are used to denture and/or to solubilize proteins ("salting-in"). The Hofmeister series has been used to depict the relative effectiveness of ions to "salt-in" and "salt-out" proteins from solutions. iv. Amino acids
Free amino acids can be used in protein formulations in accordance with various embodiments of the invention such as, to name a few, bulking agents, stabilizers and antioxidants. Lysine, proline, serine, and alanine, for instance, can be used for stabilizing proteins in a formulation. Methionine is useful as an antioxidant. Glycine is useful in lyophilization to ensure correct cake structure and properties. As a result, it is a common ingredient in lyophilized formulations and reconstituted lyophilates, such as Neumega® (Oprelvekin, Human Interleukin-11), Genotropin® (Somatropin, Human Growth Hormone), and Humatrope® (Somatropin, Human Growth Hormone). Arginine may be useful to inhibit protein aggregation, in both liquid and lyophilized formulations, as it is in, for example, Activase® (Alteplase, tPA), Avonex® (Interferon beta-la), and Enbrel® (Etanercept).. v. Polyols
Polyols include sugars, e.g., mannitol, sucrose, and sorbitol and polyhydric alcohols such as, for instance, glycerol and propylene glycol, and, for purposes of discussion herein, polyethylene glycol (PEG) and related substances. Polyols generally are kosmotropic. In embodiments they are useful stabilizing agents in both liquid and lyophilized formulations to protect proteins from physical and chemical degradation processes. In embodiments polyols are useful for adjusting the tonicity of formulations.
Sorbitol and sucrose are among agents useful for adjusting tonicity . For lyophilized compositions they are useful as stabilizers to protect against freeze-thaw stresses during transport or the preparation of bulks during the manufacturing process.
Also among polyols useful in embodiments is mannitol, which is commonly used to ensure structural stability of the cake in lyophilized formulations, such as, for example Leukine® (Sargamostim), Enbrel® (Etanercept) and Betaseron® (Interferon-beta). It ensures structural stability to the cake. It is generally used with a lyoprotectant, e.g., sucrose. Reducing sugars (which contain free aldehyde or ketone groups), such as glucose and lactose, can glycate surface lysine and arginine residues and, therefore, are used cautiously or avoided. In addition, sugars that form reactive species, such as sucrose (which is hydrolyzed to fructose and glucose under acidic conditions, and consequently engenders glycation), also must be used with caution or avoided completely.
PEG is useful to stabilize proteins and as a cryoprotectant and can be used in the invention in this regard. It is used in Recombinate®, in this regard, for instance. vi. Surfactants
Proteins are susceptible to adsorption on surfaces and to denaturation and consequent aggregation at air-liquid, solid-liquid, and liquid-liquid interfaces. These effects generally scale inversely with protein concentration, and typically are exacerbated by physical agitation, such as that generated during the shipping and handling of a product. In embodiments surfactants are used to prevent, minimize, or reduce surface adsorption. Useful surfactants in the invention in this regard include polysorbate 20, polysorbate 80, other fatty acid esters of sorbitan polyethoxylates, and poloxamer 188.
In embodiments surfactants are used to maintain protein conformational stability. The use of surfactants in this regard is protein-specific, since any given surfactant typically will stabilize some proteins and destabilize others.
Polysorbates are susceptible to oxidative degradation and often, as supplied, contain sufficient quantities of peroxides to cause oxidation of protein residue side-chains, especially methionine. Consequently, polysorbates should be used carefully, and when used, should be employed at their lowest effective concentration. In this regard, polysorbates exemplify the general rule that excipients should be used in their lowest effective concentrations. vii. Antioxidants
A variety of processes can result in harmful oxidation of proteins in pharmaceutical formulations. To some extent deleterious oxidation of proteins can be prevented in pharmaceutical formulations by maintaining proper levels of ambient oxygen and temperature and by avoiding exposure to light. In addition, antioxidant excipients can be used to prevent oxidative degradation. Among useful antioxidants in this regard are reducing agents, oxygen/free-radical scavengers, and chelating agents. Antioxidants for use in therapeutic protein formulations in accordance with the invention preferably are water-soluble and maintain their activity throughout the shelf life of a product. EDTA is an antioxidant that can be used in accordance with the invention in this regard, in much the same way it has been used in formulations of acidic fibroblast growth factor and in products such as Kineret® (Anakinra), and Ontak® (Denileukin, Difϊtox). Antioxidants also can damage proteins and must be used carefully in formulations of active intact proteins. For instance, reducing agents, such as glutathione can disrupt intramolecular disulfide linkages. viii. Metal Ions
Formulations in accordance with the invention may include metal ions that are protein co-factors and that are necessary to form protein coordination complexes, such as zinc necessary to form certain insulin suspensions. Metal ions also can inhibit some processes that degrade proteins. However, metal ions also catalyze physical and chemical processes that degrade proteins and must be used cautiously.
Magnesium ions (10 -120 mM) can be used to inhibit isomerization of aspartic acid to isoaspartic acid. Ca+2 ions (up to 100 mM) can increase the stability of human deoxyribonuclease (rhDNase, Pulmozyme®). Mg+2, Mn+2, and Zn+2, however, can destabilize rhDNase. Similarly, Ca+2 and Sr+2 can stabilize Factor VIII, it can be destabilized by Mg+2,
Mn and Zn , Cu and Fe ' and its aggregation can be increased by Al ions, ix. Preservatives Preservatives typically are necessary when developing multi-dose parenteral formulations that involve more than one extraction from the same container. Their primary function is to inhibit microbial growth and ensure product sterility throughout the shelf-life or term of use of a product. Commonly used preservatives include benzyl alcohol, phenol and m-cresol. Although preservatives have a long history of use with small-molecule parenterals, the development of protein formulations that include preservatives can be challenging, because preservatives can have a destabilizing effect on proteins, such as aggregation, and this may limit their use, particularly in multi-dose formulations.
Several aspects of the preservation need to be considered in the development of preserved dosage forms. The effective preservative concentration in the product must be optimized. This requires testing a given preservative in the dosage form to determine concentrations that confer anti-microbial effectiveness without compromising protein stability. Different preservatives typically will be tested for their impact on stability over a range of concentrations, before selecting the best one for a given formulation.
As might be expected, development of liquid formulations containing preservatives is more challenging than developing lyophilized formulations, since freeze-dried products can be lyophilized without the preservative and then reconstituted with a preservative containing diluent at the time of use. As a result the preservative is in contact with the protein for a significantly shorter time in lyophilized formulations, significantly minimizing the associated stability risks. Indeed, with liquid formulations, preservative effectiveness and stability in the presence of the preservative both have to be maintained over the entire shelf- life of the product, which typically is 18 to 24 months for a pharmaceutical. An important point to note is that preservative effectiveness has to be demonstrated in the final formulation containing the active drug and all excipient components.
To date, most protein drugs have been formulated for single-use only. However, when multi-dose formulations are possible, they have the added advantage of enabling patient convenience, and increased marketability. A good example is human growth hormone (hGH), where the development of preserved formulations has led to commercialization of more convenient, multi-use injection pen presentations. At least four pen devices containing preserved formulations of hGH are currently available on the market. Norditropin® (liquid, Novo Nordisk), (Somatropin, Human Growth Hormone); Nutropin AQ® (liquid, Genentech), (Somatropin, Human Growth Hormone); and Genotropin (lyophilized - dual chamber cartridge, Pharmacia & Upjohn), (Somatropin, Human Growth Hormone) contain phenol, while Somatrope® (Eli Lilly), (Somatropin, Human Growth Hormone) is formulated with m- cresol. b. Formulations and Administration/Methods of Delivery Formulations may be designed in accordance with the invention for delivery by any suitable route, including but not limited to oral, aural, ophthalmic, rectal, and vaginal, and parenteral routes, including intravenous and intraarterial injection, intramuscular injection, and subcutaneous injection, to name just some. i. Formulations for Parenteral Administration
In embodiments, formulations for parenteral administration may be in the form of aqueous or non-aqueous isotonic sterile injection solutions or suspensions. In embodiments the solutions and/or suspensions are prepared from sterile powders and/or granules using one or more carriers and/or diluents suitable for use in pharmaceutical formulations.
When parenteral administration is contemplated, therapeutic compositions for use in embodiments of the invention may be in the form of a pyrogen- free, parenterally acceptable aqueous solution comprising the desired protein in a pharmaceutically acceptable vehicle. A particularly suitable vehicle for parenteral injection is sterile pure water in which the protein is formulated as a sterile, isotonic, buffered solution, often containing additional ingredients, such as excipients, such as but not limited to those described elsewhere herein. For instance but without limitation, in embodiments formulations for parenteral administration contain one or more substances that do one or more of adjust viscosity (such as carboxymethyl cellulose, sorbitol, or dextran), increase protein solubility (particularly solubility of the therapeutic protein), stabilize the therapeutic protein and/or other ingredients, adjust the tonicity and/or the ionic strength of the formulation, adjust and/or buffer the pH of the formulation, act as preservatives and/or otherwise have a beneficial effect on stability and storage, to mention just a few.
In embodiments proteins may be formulated in a variety of other "injectable" forms, including for example, but without limitation, injectable microspheres, bio-erodible particles, polymeric compounds (polylactic acid, polyglycolic acid), beads, or liposomes, including but not limited to those that provide for controlled or sustained release. In embodiments formulations may be introduced by implantable drug delivery devices, among others. ii. Formulations for Pulmonary Administration
In embodiments formulations are suitable for pulmonary administration. For pulmonary administration, the pharmaceutical composition may be administered, for example, in the form of an aerosol or with an inhaler including dry powder aerosol. For instance but without limitation, a binding agent may be formulated as a dry powder for inhalation. Inhalation solutions may also be formulated with a propellant for aerosol delivery. In embodiments in this regard, solutions for administration are nebulized. Various aspects regarding pulmonary administration useful in embodiments in this regard are described in PCT Application No. PCT/US94/001875, which is herein incorporated by reference in its entirety particularly regarding pulmonary delivery of therapeutic proteins. iii. Formulations for Oral Administration
In embodiments formulations are suitable for oral administration. In embodiments the formulation for oral administration may be in the form of, for example but without limitation, a tablet, capsule, suspension, or liquid. The formulation may be in the form of a dosage unit containing a particular amount of the active ingredient, e.g., the therapeutic protein. Examples of such dosage units are tablets or capsules. Formulations for oral administration in accordance with embodiments of the invention in this regard can be made conventionally. iv. Controlled-Release Formulations Also among embodiments are sustained- and controlled-delivery formulations.
Techniques for making such sustained- and controlled-delivery formulations that may be used in accordance with various aspects and embodiments of the invention are well-known to those skilled in the art. Among these are delivery methods that use liposome carriers, bio-erodible microparticles, porous beads, and semi-permeable polymer matrices, such as those described in PCT/US93/00829; U.S. 3,773,919; EP 58,481; Sidman et al, Biopotymers, 22:547-556 (1983); Langer et al., J. Biomed. Mater. Res., 15:167-277, (1981); Langer et al., Chem. Tech., 12:98-105(1982); EP 133,988; Eppstein et al., Proc. Natl. Acad. Sci. (USA), 82:3688-3692 (1985); EP 36,676; EP 88,046; and EP 143,949, each of which is incorporated herein by reference in its entirety, particularly in parts pertinent to sustained- and controlled-delivery pharmaceutical protein formulations as described and claimed herein. c. Sterilization
A pharmaceutical composition for in vivo administration typically must be sterile. Sterilization may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution. A composition for parenteral administration may be stored in lyophilized form or in solution. In addition, parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle. d. Storage
Once a pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) requiring reconstitution prior to administration. e. Additional Pharmaceutical Agents
Compositions in embodiments of the invention can comprise in addition to an active protein of the composition, one or more additional pharmaceutical agents. Such agents may be proteins, or they may be other types of pharmaceutical agents. Included among such agents are those for prevention or treatment of any disorder or disease. Such agents include, for instance, antibiotics and antimycotics. They also include agents for treating human disorders, including but not limited to, agents for treating inflammatory diseases, cancers, metabolic disorders, neurological and renal disorders, to name just a few. Agents that may be used in the invention in this regard also include agents useful to augment the action of a composition and or prevent, ameliorate, or treat any undesirable side effects thereof. 4. Routes of Administration
In various embodiments, formulations in accordance with the invention may be administered by any suitable route, including those well-known to those skilled in the art of administering therapeutics to a subject. Such routes in a variety of embodiments include but are not limited to administration of the compositions orally, ocularly, mucosally, topically, rectally, pulmonarily, such as by inhalation spray, and epicutaneously. Parenteral routes of administration are useful in embodiments of the invention, including administration by intravenous, intraarterial, intracardiac, intraspinal, intrathecal, intraosseous, intraarticular, intrasynovial, intracutaneous, intradermal, subcutaneous, peritoneal, and/or intramuscular injection. In some embodiments intravenous, intraarterial, intracutaneous, intradermal, subcutaneous and/or intramuscular injection are used. In some embodiments intravenous, intraarterial, intracutaneous, subcutaneous, and/or intramuscular injection are used.
5. Doses
The amount of a protein formulation administered and the dosage regimen for treating a disease condition with the formulation depends on a variety of factors, including the age, weight, sex, and medical condition of the subject, the type of disease, the severity of the disease, the route and frequency of administration, and the particular formulation employed. In particular the amount will depend on the protein therapeutic being administered and any other therapeutic agents being administered in conjunction therewith. Dosages can be determined for formulations in accordance with the invention using well-established routine pharmaceutical procedures for this purpose.
6. Dosing Regimens
Formulations in various embodiments of the invention can be administered in dosages and by techniques well-known to those skilled in the medical and veterinary arts taking into consideration such factors as the age, sex, weight, and condition of the particular patient, and the formulation that will be administered (e.g., solid vs. liquid). Doses for humans, other mammals, and other subjects can be determined without undue experimentation by the skilled artisan, from this disclosure, the documents cited herein, and the knowledge possessed by those skilled in the pertinent art.
In accordance with some embodiments, proper dosages and dosing plans will depend on numerous factors, and may vary in different circumstances. The parameters that will determine the optimal dosage plans to be administered typically will include some or all of the following: the disease being treated and its stage; the species of the subject, their health, gender, age, weight, and metabolic rate; other therapies being administered; and expected potential complications from the subject's history or genotype.
The optimal dosing plan in a given situation also will take into consideration the nature of the formulation, the way it is administered, the distribution route following administration, and the rate at which it will be cleared both from sites of action and from the subject's body. Finally, the determination of optimal dosing preferably will provide an effective dose that is neither below the threshold of maximal beneficial effect nor above the threshold where the deleterious effects associated with the dose of the active agent outweighs the advantages of the increased dose. It will be appreciated that a "dose" may be delivered all at once, fractionally, or continuously over a period of time. The entire dose may be delivered to a single location or spread fractionally over several locations. Furthermore, doses may remain the same over a treatment, or they may vary.
In various embodiments, formulations in accordance with certain embodiments of the invention are administered in an initial dose, and thereafter maintained by further administrations. A formulation of the invention in some embodiments is administered by one method initially, and thereafter administered by the same method or by one or more different methods. The dosages of on-going administrations may be adjusted to maintain at certain values the levels of the active agents in the subject. In some embodiments the compositions are administered initially, and/or to maintain their level in the subject, by intravenous injection. In a variety of embodiments, other forms of administration are used.
Formulations of a variety of embodiments of the invention may be administered in many frequencies over a wide range of times, including any suitable frequency and range of times that delivers a treatment-effective dose. Suitable regimens for initial administration and further doses for sequential administrations may all be the same or may be variable.
Doses may be continuously delivered, administered every few hours, one or more times a day, every day, every other day or several times a week, more frequently or less frequently.
In embodiments they are administered over periods of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 or more days. In embodiments they are administered over periods of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more weeks. In embodiments they are administered over periods of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more months. In embodiments they are administered over periods of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or more years. Appropriate regimens can be ascertained by the skilled artisan, from this disclosure, the documents cited herein, and the knowledge in the art. Generally lengths of treatment will be proportional to the length of the disease process, the effectiveness of the therapies being applied, and the condition and response of the subject being treated.
7. Diseases and Treatments
Pharmaceutical protein compositions in accordance with the invention, in various embodiments, are useful to treat subjects suffering from a wide variety of disorders and diseases. As noted elsewhere herein, the invention provides, among others, compositions of pharmaceutical antibodies, antibody-derived pharmaceutical proteins, and antibody-related pharmaceutical proteins, that can comprise Fc effector functions and binding domains specific for a wide variety of disease-related targets that are particularly useful for treating disease. In embodiments the invention provides Fc fusion proteins. In embodiments the invention provides Fc-peptide fusion proteins. In embodiments the invention provides peptibodies. These proteins and compositions thereof are described at length herein, as well as their use in treating various disorders and diseases.
Methods for using the compositions, including formulation methods, administration methods, doses, and dosing methods are all described illustratively herein. The formulation and administration of any particular composition of the invention can be tailored to the treatment of a particular disease, using well-known and routine techniques in the arts for doing so, taken in light of the guidance provided by the present description of the invention. Among diseases usefully treated using pharmaceutical protein formulations in accordance with various aspects and embodiments of the invention are inflammatory diseases, cancers, metabolic disorders, neurological and renal disorders, to name just a few.
8. Packaging and Kits
The invention also provides kits comprising protein formulations, particularly kits comprising in one more containers, a pharmaceutical protein formulation and instructions regarding the use thereof, particularly such kits wherein the formulation is a pharmaceutically acceptable formulation for human use.
Among preferred kits are those comprising one or more containers of a protein formulation of the invention and one or more separate documents, information pertaining to the contents of the kit, and/or the use of its contents, particularly those wherein the protein is a biopharmaceutical protein, especially those wherein the protein is a biopharmaceutical protein formulated for the treatment of a disease in humans. Preferred proteins in this regard are discussed herein above and include, for instance, antibodies, proteins comprising an active peptide, Fc fusion proteins, proteins comprising an Fc region fused to an active peptide, and peptibodies, as discussed in greater detail throughout the present disclosure.
In certain aspects of the invention in this regard, preferred kits include kits as above further comprising one or more single or multi-chambered syringes (e.g., liquid syringes and lyosyringes) for administering one or more protein formulations of the invention. In certain aspects of the invention in this regard, certain of the particularly preferred kits further comprise preloaded syringes. In further particularly preferred embodiments in this regard, the kits comprise a pharmaceutical composition for parenteral administration, sealed in a vial under partial vacuum in a form ready for loading into a syringe and administration to a subject. In especially preferred embodiments in this regard, the composition is disposed therein under partial vacuum. In all of these regards and others, in certain further particularly preferred embodiments the kits contain one or more vials in accordance with any of the foregoing, wherein each vial contains a single unit dose for administration to a subject. In all these respects and others the invention further relates to kits comprising solutions, formulated as described herein and comprising, in particular embodiments in this regard, Fc fusion proteins, especially peptibodies. In all these respects and others the invention further relates to kits comprising lyophilates, disposed as above, that upon reconstitution provide compositions in accordance therewith. In this regard, the invention further provides in certain of its preferred embodiments, kits that contain a lyophilate in accordance with the invention and a sterile diluent for reconstituting the lyophilate.
Examples
The present invention is additionally described by way of the following illustrative, non-limiting Examples. The following tables provide illustrative formulations for antibodies and peptibodies in accordance with the invention as described above.
EXAMPLE 1 : Formulation and Stability of Antibodies
Antibodies were formulated in accordance with their pi as described, as shown in Table 2. The antibodies displayed good solubility in all cases. Proteins remained intact over a period of 6 months as measured by size-exclusion chromatography. EXAMPLE 2: Formulation and Stability of Peptibodies
Peptibodies were formulated in accordance with their pi as described, as shown in Table 3. The peptibodies displayed good solubility in all cases. Proteins remained intact over a period of 6 months as measured by size-exclusion chromatography.
TABLE 2
Table 2 is a list of IgG monoclonal antibodies formulated in accordance with the invention, showing for each antibody formulation, its "Isotype", pi ("Measured pi"), buffer composition ("Formulation Buffer"), the pH of the formulation ("pH") and the concentration of the antibody in the formulation in mg/ml ("Protein Concentration"). The pi for mAbl is the calculated value.
Abbreviations used for the "Formulation Buffer" are as follows: KP = 10 mM potassium phosphate; S = 5% w/v sorbitol; A = IO mM sodium acetate; Su = 9.0-9.5 % w/v sucrose; R = 3.4 % w/v arginine hydrochloride, T = 0.004 % w/v polysorbate20, * = calculated pi, Storage conditions: -20 0C or lower.
Stability was measured using size-exclusion chromatography for a period of 6 months or lon er.
Figure imgf000076_0001
TABLE 3
Table 3 is a list of peptibodies formulated in accordance with the invention, showing pi's of proteins ("pi with Fc") and active peptides ("pi Peptide Alone"), formulation buffers and constituents ("Formulation Buffer"), peptibody concentrations in the formulations in mg/ml ("Concentration"), and pH of the formulations ("pH").
Abbreviations used for the "Frozen Formulation" are as follows: KP = 10 mM potassium phosphate; S = 5% w/v sorbitol; A = IO mM sodium acetate; Su = 9.5% w/v sucrose; R = 3.4 % w/v arginine hydrochloride; T = 0.004% w/v polysorbate 20. Storage conditions -30 0C or lower.
Stability was measured using size-exclusion chromatography for a period of 6 months or longer.
Figure imgf000077_0001
Selected References
The following references are incorporated by reference herein in their entireties particularly in parts pertinent to the subject matter for which they are cited in the foregoing disclosure, as summarized below. (1) REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY, 21st
Edition, Hendrickson, R., Editor, Lippincott Williams, & Wilkins, Philadelphia (2006), which is incorporated by reference herein in its entirety in parts pertinent to pharmaceutical formulations, especially formulations of pharmaceutical peptides, polypeptides, and proteins, particularly as to considerations important in the consideration of formulating, dosing, and administering peptide, polypeptide, and protein pharmaceuticals, in particular as to the specific aspects thereof discussed herein above.
(2) ANSEL' S PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 8th Edition, Allen et al, Editors, Lippincott Williams & Wilkins, Philadelphia (2005), which is incorporated herein by reference in its entirety particularly in parts pertinent to pharmaceutical protein formulations, and to important aspects of such formulations and of making them and using them, in particular as to the specific aspects thereof discussed herein above.
(3) PHARMACEUTICAL FORMULATION DEVELOPMENT OF PEPTIDES AND PROTEINS, Frokjaer et al, Editors, CRC Press, Boca Raton (2000), which is incorporated herein by reference in its entirety particularly in parts pertinent to pharmaceutical protein formulations and to important aspects of such formulations and of making them and using them, in particular as to the specific aspects thereof discussed herein above.
(4) FORMULATION, CHARACTERIZATION AND STABILITY OF PROTEIN DRUGS - CASE HISTORIES, Pharmaceutical Biotechnology Series, Volume 9, Pearlman et al, Editors, Plenum Publishing Company, New York (1996), which is herein incorporated by reference in its entirety particularly in parts pertinent to the formulation of peptide and protein therapeutics, particularly as to the specific aspects thereof discussed herein above.
(5) PROTEIN FORMULATION AND DELIVERY, Drugs and the Pharmaceutical Sciences, Volume 93, E. J. McNaIIy, Editor, Marcel Dekker, New York (2000), which is herein incorporated by reference in its entirety particularly in parts pertinent to the formulation of peptide and protein therapeutics, particularly as to the specific aspects thereof discussed herein above.

Claims

What is claimed is:
1. A pharmaceutical formulation, comprising an Fc fusion protein comprised of an active peptide and an Fc region of an antibody, wherein: the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less; the concentration of the Fc fusion protein in the formulation is at least 20 mg/ml; and the formulation is stable for at least 18 months
2. A pharmaceutical formulation comprising an Fc fusion protein comprised of an active peptide and an Fc region of an antibody, wherein: the pH of the formulation is (i) 4.6 to 5.6 if the pi of the Fc fusion protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the Fc fusion protein is 6.0 or less; the concentration of the Fc fusion protein in the formulation is at least 20 mg/ml; and the formulation is stable for at least 18 months.
3. A formulation according to claim 1 or 2, wherein the Fc fusion protein is a peptibody.
4. A formulation according to claim 3, wherein the concentration of the peptibody in the formulation is at least 20 mg/ml.
5. A formulation according to claim 4, wherein the concentration of the peptibody is 20 to 500 mg/ml.
6. A formulation according to claim 3, wherein further the formulation comprises a buffering agent.
7. A formulation according to claim 6, wherein the buffering agent is acetate, phosphate, or histidine.
8. A formulation according to claim 6, wherein the formulation comprises a tonicifying agent.
9. A formulation according to claim 8, wherein the tonicifying agent is a polyol, arginine, or NaCl.
10. A formulation according to claim 9, wherein the polyol is sorbitol, mannitol, sucrose, trehalose, or glycerol.
11. A formulation according to claim 6, wherein the formulation comprises a surfactant.
12. A formulation according to claim 11, wherein the surfactant is polysorbate 20 or polysorbate 80.
13. A pharmaceutical formulation, comprising a peptibody, a surfactant, and a tonicifying agent, wherein: the pH of the formulation is (i) 4.6 to 5.6 if the pi of the peptibody is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the peptibody is 4.0 or less; the concentration of the peptibody is at least 20 mg/ml; and the formulation is stable for at least 18 months.
14. A formulation according to claim 13, wherein the Fc region is derived from an IgG antibody or is a derivative or a modification of an Fc region derived from an IgGl, IgG2, or IgG4 antibody.
15. A formulation according to any of claims 1 through 14, wherein the protein is selected from the group consisting of proteins that bind specifically to one or more of: CD3, CD4, CD8, CD19, CD20,CD22, CD30, CD34; HER2, HER3, HER4, the EGF receptor; LFA-I, MoI, pl50,95, VLA-4, ICAM-I, VCAM, alpha v/beta 3 integrin, OPGL, Ang2, Angl, stem cell factors, vascular endothelial growth factor ("VEGF"); growth hormone, thyroid stimulating hormone, follicle stimulating hormone, luteinizing hormone, growth hormone releasing factor, parathyroid hormone, mullerian-inhibiting substance, human macrophage inflammatory protein (MIP-I -alpha), erythropoietin (EPO), NGF, NGF-beta, platelet-derived growth factor (PDGF), aFGF, bFGF, epidermal growth factor (EGF), TGF-alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-beta4, TGF- beta5, IGF-I, IGF-II, des(l-3)-IGF-I (brain IGF-I), insulin, insulin A-chain, insulin B- chain, proinsulin, insulin-like growth factor binding proteins, factor VIII, tissue factor, von Willebrands factor, protein C, alpha- 1 -antitrypsin, plasminogen activators, such as urokinase and tissue plasminogen activator ("t-PA"), bombazine, thrombin, thrombopoietin, M-CSF, GM-CSF, G-CSF, albumin, IgE, flk2/flt3 receptor, obesity (OB) receptor, bone-derived neurotrophic factor (BDNF), NT-3, NT-4, NT-5, NT-6; relaxin A- chain, relaxin B-chain, prorelaxin; interferon-alpha, -beta, and -gamma; IL-I to IL-15; IL-I to IL- 15 receptors, AIDS envelope viral antigen; calcitonin, glucagon, atrial natriuretic factor, lung surfactant, tumor necrosis factor-alpha and -beta, enkephalinase, RANTES, mouse gonadotropin-associated peptide, DNAse, inhibin, and activin; protein A or D, bone morphogenetic protein (BMP), superoxide dismutase, decay accelerating factor (DAF), thrombopoietin receptor, TALL-I /B AFF/ AGP-3, myostatin, amyloid beta, thymic stromal lyphopoietin, RANKL (RANK ligand), c-kit, OX40L, glucagon receptor, IGF-I, B7RP-1, TNF, TNF receptor type I (p55), TRAIL-R2, activin A, CSF-I receptor, c-Kit and OX40L.
16. A method for making a pharmaceutical formulation comprising an Fc fusion protein that comprises an active peptide and an Fc region of an antibody, comprising formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less.
17. A method for making a pharmaceutical formulation comprising an Fc fusion protein that comprises an active peptide and an Fc region of an antibody, comprising formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less.
18. A method according to claim 16, further comprising predicting or measuring the pi of the active peptide and then formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the pi of the active peptide is 4.0 or more, and (ii) 7.0 to 8.0 if the pi of the active peptide is 4.0 or less, wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
19. A method according to claim 17, further comprising predicting or measuring the pi of the protein and then formulating the protein such that the pH of the formulation is (i) 4.6 to 5.6 if the protein is 6.0 or more, and (ii) 7.0 to 8.0 if the pi of the protein is 6.0 or less, wherein the resulting formulation is a pharmaceutically acceptable protein formulation.
20. A method according to claim 16 or 17, wherein the Fc fusion protein is a peptibody.
21. A method according to claim 20, wherein the pi of the active peptide substantially determines the pi of the protein.
22. A method according to claim 20, wherein the concentration of the protein in the formulation is at least 20 mg/ml.
23. A method according to claim 22 wherein the concentration of the protein in the formulation is 20 to 500 mg/ml.
24. A method according to claim 22, wherein further the formulation comprises a buffering agent.
25. A method according to claim 24, wherein the buffering agent is acetate, phosphate, or histidine.
26. A method according to claim 24, wherein the formulation comprises a tonicifying agent.
27. A method according to claim 26, wherein the tonicifying agent is a polyol, arginine, or NaCl.
28. A method according to claim 27, wherein the polyol is sorbitol, mannitol, sucrose, trehalose, or glycerol.
29. A method according to claim 26, wherein further the formulation comprises a surfactant.
30. A method according to claim 29, wherein the surfactant is polysorbate 20 or polysorbate 80.
31. A method according to claim 29, wherein further the formulation comprises a polyol.
32. A method according to claim 31, wherein the polyol is one or more of sorbitol, mannitol, sucrose, trehalose, or glycerol.
33. A method according to claim any of claims 16 through 32, wherein the protein is selected from the group consisting of proteins that bind specifically to one or more of:
CD3, CD4, CD8, CD19, CD20, CD22, CD30, CD34; HER2, HER3, HER4, the EGF receptor; LFA-I, MoI, pl50,95, VLA-4, ICAM-I, VCAM, alpha v/beta 3 integrin, OPGL, Ang2, Angl, stem cell factors, vascular endothelial growth factor ("VEGF"); growth hormone, thyroid stimulating hormone, follicle stimulating hormone, luteinizing hormone, growth hormone releasing factor, parathyroid hormone, mullerian-inhibiting substance, human macrophage inflammatory protein (MIP-I -alpha), erythropoietin (EPO), NGF, NGF-beta, platelet-derived growth factor (PDGF), aFGF, bFGF, epidermal growth factor (EGF), TGF-alpha, TGF-betal, TGF-beta2, TGF-beta3, TGF-beta4, TGF- beta5, IGF-I, IGF-II, des(l-3)-IGF-I (brain IGF-I), insulin, insulin A-chain, insulin B- chain, proinsulin, insulin-like growth factor binding proteins, factor VIII, tissue factor, von Willebrands factor, protein C, alpha- 1 -antitrypsin, plasminogen activators, such as urokinase and tissue plasminogen activator ("t-PA"), bombazine, thrombin, thrombopoietin, M-CSF, GM-CSF, G-CSF, albumin, IgE, flk2/flt3 receptor, obesity (OB) receptor, bone-derived neurotrophic factor (BDNF), NT-3, NT-4, NT-5, NT-6; relaxin A- chain, relaxin B-chain, prorelaxin; interferon-alpha, -beta, and -gamma; IL-I to IL-15; IL-I to IL- 15 receptors, AIDS envelope viral antigen; calcitonin, glucagon, atrial natriuretic factor, lung surfactant, tumor necrosis factor-alpha and -beta, enkephalinase, RANTES, mouse gonadotropin-associated peptide, DNAse, inhibin, and activin; protein A or D, bone morphogenetic protein (BMP), superoxide dismutase, decay accelerating factor (DAF), thrombopoietin receptor, TALL-I /B AFF/ AGP-3, myostatin, amyloid beta, thymic stromal lyphopoietin, RANKL (RANK ligand), c-kit, OX40L, glucagon receptor, IGF-I, B7RP-1, TNF, TNF receptor type I (p55), TRAIL-R2, activin A, CSF-I receptor, c-Kit and OX40L receptor.
34. A method for treating a subject, comprising administering to a subject in an amount and by a route effective for said treatment, a formulation according to any of claims 1 through 14.
35. A method for treating a subject comprising administering to a subject in an amount and by a route effective for said treatment, a formulation made by a method according to any of claims 16 through 33.
PCT/US2008/071209 2007-07-25 2008-07-25 Pharmaceutical compositions comprising fc fusion proteins WO2009015345A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US93507307P 2007-07-25 2007-07-25
US60/935,073 2007-07-25

Publications (1)

Publication Number Publication Date
WO2009015345A1 true WO2009015345A1 (en) 2009-01-29

Family

ID=40011280

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/071209 WO2009015345A1 (en) 2007-07-25 2008-07-25 Pharmaceutical compositions comprising fc fusion proteins

Country Status (1)

Country Link
WO (1) WO2009015345A1 (en)

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102399292A (en) * 2011-09-14 2012-04-04 盛剑鹏 Recombinant stem cell factor-immunoglobulin (rSCF-Ig) fusion protein and preparation thereof
EP2496691A2 (en) * 2009-11-02 2012-09-12 The University Of Washington Therapeutic nuclease compositions and methods
CN101993496B (en) * 2009-08-20 2013-06-05 重庆富进生物医药有限公司 Dual blood sugar and blood fat adjusting fusion protein and preparation method and application thereof
WO2013106589A1 (en) * 2012-01-10 2013-07-18 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
US8937157B2 (en) 2011-04-29 2015-01-20 University Of Washington Therapeutic nuclease compositions and methods
WO2015159254A1 (en) * 2014-04-16 2015-10-22 Biocon Ltd. Stable protein formulations comprising a molar excess of sorbitol
JP2016512546A (en) * 2013-03-13 2016-04-28 アムジェン インコーポレイテッド Proteins specific for BAFF and B7RP1 and uses thereof
US9340612B2 (en) 2001-03-19 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9365646B2 (en) 2012-12-05 2016-06-14 Novartis Ag Compositions and methods for antibodies targeting EPO
WO2016133372A3 (en) * 2015-02-17 2016-10-13 한미약품 주식회사 Long-acting insulin or insulin analogue complex
RU2599848C2 (en) * 2011-02-11 2016-10-20 Сведиш Орфан Биовитрум Аб (Пабл) Citrate free pharmaceutical compositions comprising anakinra
CN106084062A (en) * 2015-04-28 2016-11-09 烟台荣昌生物工程有限公司 The bispecific fusion protein of bridging
RU2613905C2 (en) * 2010-10-26 2017-03-21 Ханми Сайенс Ко., Лтд. Liquid composition of long-acting alpha interferon conjugate
US9938353B2 (en) 2011-06-24 2018-04-10 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
US10421824B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1
US10988745B2 (en) 2013-10-31 2021-04-27 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
US11013785B2 (en) 2016-11-10 2021-05-25 Keros Therapeutics, Inc. Activin receptor type IIA variants and methods of use thereof
US11071782B2 (en) 2016-04-20 2021-07-27 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11103552B2 (en) 2018-05-10 2021-08-31 Regeneron Pharmaceuticals, Inc. High concentration VEGF receptor fusion protein containing formulations
CN113521015A (en) * 2020-10-21 2021-10-22 山东省妇幼保健院 Fusion protein 4D5Fv-PE25 freeze-dried agent and preparation method and application thereof
US11208452B2 (en) 2015-06-02 2021-12-28 Novo Nordisk A/S Insulins with polar recombinant extensions
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11292825B2 (en) 2015-10-01 2022-04-05 Novo Nordisk A/S Protein conjugates
CN114533653A (en) * 2020-11-26 2022-05-27 上海凯茂生物医药有限公司 Human erythropoietin hybrid Fc fusion protein phase change type preparation and preparation method thereof
US11471537B2 (en) 2017-04-05 2022-10-18 Novo Nordisk A/S Oligomer extended insulin-Fc conjugates
US11484573B2 (en) 2017-11-09 2022-11-01 Keros Therapeutics, Inc. Activin receptor type IIa variants and methods of use thereof
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
US11725246B2 (en) 2015-08-12 2023-08-15 Novartis Ag Methods of treating ophthalmic disorders
US11884715B2 (en) 2018-01-12 2024-01-30 Keros Therapeutics, Inc. Activin receptor type IIB variants and methods of use thereof
US11912754B2 (en) 2017-10-12 2024-02-27 Immunowake Inc. VEGFR-antibody light chain fusion protein

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004058988A2 (en) * 2002-12-20 2004-07-15 Amgen, Inc. Binding agents which inhibit myostatin
WO2007014073A2 (en) * 2005-07-22 2007-02-01 Amgen Inc. Concentrated protein lyophilates, methods, and uses
WO2007092772A2 (en) * 2006-02-03 2007-08-16 Medimmune, Inc. Protein formulations

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004058988A2 (en) * 2002-12-20 2004-07-15 Amgen, Inc. Binding agents which inhibit myostatin
WO2007014073A2 (en) * 2005-07-22 2007-02-01 Amgen Inc. Concentrated protein lyophilates, methods, and uses
WO2007092772A2 (en) * 2006-02-03 2007-08-16 Medimmune, Inc. Protein formulations

Cited By (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9340612B2 (en) 2001-03-19 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
CN101993496B (en) * 2009-08-20 2013-06-05 重庆富进生物医药有限公司 Dual blood sugar and blood fat adjusting fusion protein and preparation method and application thereof
KR102378465B1 (en) 2009-11-02 2022-03-28 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
US10000745B2 (en) 2009-11-02 2018-06-19 University Of Washington Therapeutic nuclease compositions and methods
CN106399276B (en) * 2009-11-02 2021-08-10 华盛顿大学 Therapeutic nuclease compositions and methods
EP2496691A4 (en) * 2009-11-02 2013-07-24 Univ Washington Therapeutic nuclease compositions and methods
US8841416B2 (en) 2009-11-02 2014-09-23 University Of Washington Therapeutic nuclease compositions and methods
US9790479B2 (en) 2009-11-02 2017-10-17 University Of Washington Therapeutic nuclease compositions and methods
KR102624939B1 (en) 2009-11-02 2024-01-16 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
US11306297B2 (en) 2009-11-02 2022-04-19 University Of Washington Therapeutic nuclease compositions and methods
KR20190002735A (en) * 2009-11-02 2019-01-08 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
KR20220038546A (en) * 2009-11-02 2022-03-28 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
EP3460056A1 (en) * 2009-11-02 2019-03-27 University Of Washington Therapeutic nuclease compositions and methods
KR20120112446A (en) * 2009-11-02 2012-10-11 유니버시티 오브 워싱톤 Therapeutic nuclease compositions and methods
EP2496691A2 (en) * 2009-11-02 2012-09-12 The University Of Washington Therapeutic nuclease compositions and methods
KR102118584B1 (en) 2009-11-02 2020-06-05 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
EP3202898A1 (en) * 2009-11-02 2017-08-09 University of Washington Therapeutic nuclease compositions and methods
KR20200063266A (en) * 2009-11-02 2020-06-04 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
CN106399276A (en) * 2009-11-02 2017-02-15 华盛顿大学 Therapeutic nuclease compositions and methods
KR101934923B1 (en) 2009-11-02 2019-04-10 유니버시티 오브 워싱톤 스루 이츠 센터 포 커머셜리제이션 Therapeutic Nuclease Compositions and Methods
RU2613905C2 (en) * 2010-10-26 2017-03-21 Ханми Сайенс Ко., Лтд. Liquid composition of long-acting alpha interferon conjugate
US9669105B2 (en) 2010-10-26 2017-06-06 Hanmi Science Co., Ltd. Liquid formulations of long-acting interferon alpha conjugates
US10179162B2 (en) 2011-02-11 2019-01-15 Swedish Orphan Biovitrum Ab (Publ) Citrate free pharmaceutical compositions comprising anakinra
RU2599848C2 (en) * 2011-02-11 2016-10-20 Сведиш Орфан Биовитрум Аб (Пабл) Citrate free pharmaceutical compositions comprising anakinra
US8937157B2 (en) 2011-04-29 2015-01-20 University Of Washington Therapeutic nuclease compositions and methods
US10202588B2 (en) 2011-04-29 2019-02-12 The University Of Washington Therapeutic nuclease compositions and methods
US11034944B2 (en) 2011-04-29 2021-06-15 University Of Washington Therapeutic nuclease compositions and methods
US9938353B2 (en) 2011-06-24 2018-04-10 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
CN102399292A (en) * 2011-09-14 2012-04-04 盛剑鹏 Recombinant stem cell factor-immunoglobulin (rSCF-Ig) fusion protein and preparation thereof
US10478508B2 (en) 2012-01-10 2019-11-19 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
WO2013106589A1 (en) * 2012-01-10 2013-07-18 The Regents Of The University Of Colorado, A Body Corporate Compositions, methods and uses for alpha-1 antitrypsin fusion molecules
US9724414B2 (en) 2012-09-07 2017-08-08 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10772960B2 (en) 2012-09-07 2020-09-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9782480B2 (en) 2012-09-07 2017-10-10 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9770507B2 (en) 2012-09-07 2017-09-26 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9789185B2 (en) 2012-09-07 2017-10-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9808525B2 (en) 2012-09-07 2017-11-07 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9861695B2 (en) 2012-09-07 2018-01-09 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9757454B2 (en) 2012-09-07 2017-09-12 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9737600B2 (en) 2012-09-07 2017-08-22 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9340611B2 (en) 2012-09-07 2016-05-17 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10155039B2 (en) 2012-09-07 2018-12-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10159733B2 (en) 2012-09-07 2018-12-25 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10159732B2 (en) 2012-09-07 2018-12-25 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9731009B2 (en) 2012-09-07 2017-08-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9731008B2 (en) 2012-09-07 2017-08-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10195275B2 (en) 2012-09-07 2019-02-05 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9724415B2 (en) 2012-09-07 2017-08-08 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10207000B2 (en) 2012-09-07 2019-02-19 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9707293B2 (en) 2012-09-07 2017-07-18 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9682145B2 (en) 2012-09-07 2017-06-20 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10286072B2 (en) 2012-09-07 2019-05-14 Coherus Biosciences, Inc. Methods of manufacturing stable aqueous formulations of adalimumab
US10286071B2 (en) 2012-09-07 2019-05-14 Coherus Biosciences, Inc. Syringe containing stable aqueous formulations of adalimumab
US9346880B2 (en) 2012-09-07 2016-05-24 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9382317B2 (en) 2012-09-07 2016-07-05 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10799585B2 (en) 2012-09-07 2020-10-13 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10786566B2 (en) 2012-09-07 2020-09-29 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10780163B2 (en) 2012-09-07 2020-09-22 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10772959B2 (en) 2012-09-07 2020-09-15 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10688183B2 (en) 2012-09-07 2020-06-23 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716852B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716854B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10716853B2 (en) 2012-09-07 2020-07-21 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US10722579B2 (en) 2012-09-07 2020-07-28 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9782479B2 (en) 2012-09-07 2017-10-10 Coherus Biosciences, Inc. Stable aqueous formulations of adalimumab
US9365646B2 (en) 2012-12-05 2016-06-14 Novartis Ag Compositions and methods for antibodies targeting EPO
US10106605B2 (en) 2012-12-05 2018-10-23 Novartis Ag Compositions and methods for antibodies targeting Epo
US10385127B2 (en) 2012-12-05 2019-08-20 Novartis Ag Compositions and methods for antibodies targeting EPO
US11492417B2 (en) 2013-03-13 2022-11-08 Amgen Inc. Proteins specific for BAFF and B7RP1 and uses thereof
US10421823B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1 and uses thereof
JP2016512546A (en) * 2013-03-13 2016-04-28 アムジェン インコーポレイテッド Proteins specific for BAFF and B7RP1 and uses thereof
US10421824B2 (en) 2013-03-13 2019-09-24 Amgen Inc. Proteins specific for BAFF and B7RP1
US10988745B2 (en) 2013-10-31 2021-04-27 Resolve Therapeutics, Llc Therapeutic nuclease-albumin fusions and methods
WO2015159254A1 (en) * 2014-04-16 2015-10-22 Biocon Ltd. Stable protein formulations comprising a molar excess of sorbitol
US11058768B2 (en) 2014-04-16 2021-07-13 Biocon Ltd. Stable protein formulations comprising a molar excess of sorbitol
CN106456784A (en) * 2014-04-16 2017-02-22 拜康有限公司 Stable protein formulations comprising a molar excess of sorbitol
WO2016133372A3 (en) * 2015-02-17 2016-10-13 한미약품 주식회사 Long-acting insulin or insulin analogue complex
US10894089B2 (en) 2015-02-17 2021-01-19 Hanmi Pharm. Co., Ltd. Long-acting insulin or insulin analogue conjugate
CN106084062B (en) * 2015-04-28 2021-04-02 荣昌生物制药(烟台)有限公司 Bridged bispecific fusion proteins
CN106084062A (en) * 2015-04-28 2016-11-09 烟台荣昌生物工程有限公司 The bispecific fusion protein of bridging
US11208452B2 (en) 2015-06-02 2021-12-28 Novo Nordisk A/S Insulins with polar recombinant extensions
US11725246B2 (en) 2015-08-12 2023-08-15 Novartis Ag Methods of treating ophthalmic disorders
US11292825B2 (en) 2015-10-01 2022-04-05 Novo Nordisk A/S Protein conjugates
US11229702B1 (en) 2015-10-28 2022-01-25 Coherus Biosciences, Inc. High concentration formulations of adalimumab
US11576971B2 (en) 2016-04-20 2023-02-14 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11071782B2 (en) 2016-04-20 2021-07-27 Coherus Biosciences, Inc. Method of filling a container with no headspace
US11090361B2 (en) 2016-11-10 2021-08-17 Keros Therapeutics, Inc. Activin receptor type IIa variants and methods of use thereof
US11013785B2 (en) 2016-11-10 2021-05-25 Keros Therapeutics, Inc. Activin receptor type IIA variants and methods of use thereof
US11717558B2 (en) 2016-11-10 2023-08-08 Keros Therapeutics, Inc. Nucleic acid molecules encoding activin receptor type IIA variants
US11471537B2 (en) 2017-04-05 2022-10-18 Novo Nordisk A/S Oligomer extended insulin-Fc conjugates
US11912754B2 (en) 2017-10-12 2024-02-27 Immunowake Inc. VEGFR-antibody light chain fusion protein
US11484573B2 (en) 2017-11-09 2022-11-01 Keros Therapeutics, Inc. Activin receptor type IIa variants and methods of use thereof
US11884715B2 (en) 2018-01-12 2024-01-30 Keros Therapeutics, Inc. Activin receptor type IIB variants and methods of use thereof
US11103552B2 (en) 2018-05-10 2021-08-31 Regeneron Pharmaceuticals, Inc. High concentration VEGF receptor fusion protein containing formulations
US11634485B2 (en) 2019-02-18 2023-04-25 Eli Lilly And Company Therapeutic antibody formulation
CN113521015B (en) * 2020-10-21 2022-12-06 山东省妇幼保健院 Fusion protein 4D5Fv-PE25 freeze-dried agent and preparation method and application thereof
CN113521015A (en) * 2020-10-21 2021-10-22 山东省妇幼保健院 Fusion protein 4D5Fv-PE25 freeze-dried agent and preparation method and application thereof
CN114533653A (en) * 2020-11-26 2022-05-27 上海凯茂生物医药有限公司 Human erythropoietin hybrid Fc fusion protein phase change type preparation and preparation method thereof
CN114533653B (en) * 2020-11-26 2023-12-22 上海凯茂生物医药有限公司 Human erythropoietin hybrid Fc fusion protein phase-change type preparation and preparation method thereof

Similar Documents

Publication Publication Date Title
WO2009015345A1 (en) Pharmaceutical compositions comprising fc fusion proteins
US20160319011A1 (en) Self-buffering protein formulations
US20200276313A1 (en) Pharmaceutical Formulations
JP7411615B2 (en) Viscosity reduction of pharmaceutical formulations
WO2013096835A1 (en) Stable protein formulations
Ghosh et al. A systematic review of commercial high concentration antibody drug products approved in the US: formulation composition, dosage form design and primary packaging considerations
AU2017202889B2 (en) Self-buffering protein formulations
AU2012272950B2 (en) Self-buffering protein formulations

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 08796635

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08796635

Country of ref document: EP

Kind code of ref document: A1