NZ756260B2 - Liquid protein formulations containing viscosity-lowering agents - Google Patents

Liquid protein formulations containing viscosity-lowering agents

Info

Publication number
NZ756260B2
NZ756260B2 NZ756260A NZ75626014A NZ756260B2 NZ 756260 B2 NZ756260 B2 NZ 756260B2 NZ 756260 A NZ756260 A NZ 756260A NZ 75626014 A NZ75626014 A NZ 75626014A NZ 756260 B2 NZ756260 B2 NZ 756260B2
Authority
NZ
New Zealand
Prior art keywords
viscosity
protein
formulation
liquid pharmaceutical
pharmaceutical formulation
Prior art date
Application number
NZ756260A
Other versions
NZ756260A (en
Inventor
Alan Crane
Alexander M Klibanov
Robert S Langer
Alyssa M Larson
Kevin Love
Alisha K Weight
Original Assignee
Eagle Biologics 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 Eagle Biologics Inc filed Critical Eagle Biologics Inc
Publication of NZ756260A publication Critical patent/NZ756260A/en
Publication of NZ756260B2 publication Critical patent/NZ756260B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61GTRANSPORT, PERSONAL CONVEYANCES, OR ACCOMMODATION SPECIALLY ADAPTED FOR PATIENTS OR DISABLED PERSONS; OPERATING TABLES OR CHAIRS; CHAIRS FOR DENTISTRY; FUNERAL DEVICES
    • A61G3/00Ambulance aspects of vehicles; Vehicles with special provisions for transporting patients or disabled persons, or their personal conveyances, e.g. for facilitating access of, or for loading, wheelchairs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • 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
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/20Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing sulfur, e.g. dimethyl sulfoxide [DMSO], docusate, sodium lauryl sulfate or aminosulfonic acids
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/24Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing atoms other than carbon, hydrogen, oxygen, halogen, nitrogen or sulfur, e.g. cyclomethicone or phospholipids
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/54Improvements relating to the production of bulk chemicals using solvents, e.g. supercritical solvents or ionic liquids

Abstract

Concentrated, low-viscosity, low-volume liquid pharmaceutical formulations of antibodies comprising thiamine have been developed. Such formulations can be rapidly and conveniently administered by subcutaneous or intramuscular injection, rather than by lengthy intra-venous infusion.

Description

LIQUID PROTEIN FORMULATIONS CONTAINING VISCOSITY-LOWERING AGENTS CROSS-REFERENCE TO RELATED APPLICATIONS This application claims priority to and the benefit of U.S. Provisional Application No. ,521, filed July 29, 2014, entitled “Low- Viscosity Protein Formulations Containing Hydrophobic Salts; ” U.S. Provisional Application No. 62/026,497, filed July 18, 2014, entitled “LowwViscosity n Formulations Containing GRAS Viscosity-Reducing Agents; ” U.S.
Provisional Application No. 62/008,050, filed June 5, 2014, entitled “Low- Viscosity Protein Formulations Containing Ionic s; ” U.S. Provisional Application No. 61/988,005, filed May 2, 2014, entitled “Low-Viscosity ProteinllFormulations Containing plzosphatesf’ U.S. Provisional Application No. 61/946,436, filed February 28, 2014, entitled “Concentrated, Low-Viscosity Infliximab Formulations; ” US Provisional ation No. 61/943,197, filed February 21, 2014, entitled “Concentrated, Low~Viscosity, olecular—Weight—Protein Formulations; ” U.S. Provisional ation No. 61/940,227, filed February 14, 2014, entitled ”Concentrated, Low— Viscosity High~Molecular~ Weight Protein Formulations; ” and U.S. Provisional Application No. 61,876,621, filed September 11, 2013, entitled “Concentrated scosity, High— Molecular- Weight Protein Formulations, ” the disclosures ofwhich are expressly incorporated hereby by nce.
FIELD OF THE INVENTION The invention is generally in the field of inj ectable low-viscosity pharmaceutical formulations of highly concentrated proteins and methods of making and using thereof.
BACKGROUND OF THE INVENTION Monoclonal antibodies (mAbs) are important protein-based therapeutics for treating s human diseases such as cancer, infectious diseases, inflammation, and mune diseases. More than 20 mAb products have been approved by the U.S. Food and Drug Administration (FDA), and imately 20% of all biopharrnaceuticals currently being evaluated in al trials are mAbs (Daugherty et al. , Adv. Drug Deliv. Rev. 58:686-706, 2006; and Buss et al., Curr. Opinion in Pharmacol. 12:615-622, 2012). mAb-based therapies are y stered repeatedly over an extended period oftime and require l mg/kg dosing. Antibody solutions or sions can be administered via parenteral routes, such as by intravenous (IV) infusions, and subcutaneous (SC) or intramuscular (1M) injections. The SC or IM routes reduce the treatment cost, increase patient compliance, and improve ience for patients and healthcare providers during administration compared to the IV route. To be effective and phannaceutically able, parenteral formulations should preferably be sterile, stable, inj ectable (e.g., via a syringe), and non-irritating at the site of injection, in compliance with FDA guidelines. Because ofthe small volumes required for subcutaneous (usually under about 2 mL) and intramuscular (usually under about 5 mL) injections, these routes of administration for high-dose protein therapies require concentrated protein solutions. These high concentrations often result in very viscous formulations that are difficult to administer by injection, cause pain at the site of injection, are often imprecise, andfor may have decreased chemical and/or physical stability.
These characteristics result in manufacturing, storage, and usage requirements that can be challenging to achieve, in particular for formulations having high eonCentrations ofhigh—molecular-weight proteins, such as mAbs. All protein therapeutics to some extent are subject to physical and chemical instability, such as aggregation, denaturation, inking, deamidation, isomerization, oxidation, and clipping (Wang et all, J Pharm.
Sci. 96:1-26, 2007). Thus, optimal formulation development is paramount in the development of commercially viable n pharmaceuticals.
High protein trations pose challenges relating to the physical and chemical ity of the protein, as well as difficulty with manufacture, storage, and delivery of the protein formulation. One m is the tendency ofproteins to aggregate and form particulates during processing and/or storage, which makes manipulations during further sing and/or delivery difficult. Concentration-dependent degradation and/or aggregation are major challenges in developing protein formulations at higher concentrations. In addition to the potential for non-native protein ation and particulate formation, reversible ssociation in aqueous solutions 2014/055254 may occur, which butes to, among other things, increased viscosity that complicates delivery by injection. (See, for example, Steven J. Shire er al, .1 Pharm. Sci. 93:1390—1402, 2004.) Increased viscosity is one of the key challenges encountered in concentrated protein compositions affecting both production processes and the ability to readily r such compositions by conventional means. (See, for e, J. Jezek et (.11., Advanced Drug Delivery Reviews 63:1107—1117, 2011.) Highly viscous liquid formulations are difficult to manufacture, draw into a syringe, and inject subcutaneously or intramuscularly. The use of force in manipulating the viscous ations can lead to excessive frothing, which may further denature and inactivate the therapeutically active protein.
High viscosity solutions also require larger diameter needles for injection and produce more pain at the injection site.
Currently available commercial mAb products administered by SC or IM injection are. usually formulated in aqueous buffers, such as a phosphate or L-histidine buffer, with excipients or tants, such as mannitol, sucrose, lactose, trehalose, POLOXAMER® (nonionic triblock copolyrners composed of a central hydrophobic chain ofpolyoxypropylene (poly(propylene ) flanked by two hydrophilic chains of polyoxyethylene (poly(ethylene )) or POLYSORBATE® 80 (PEG(80)sorbitan monolaurate), to prevent aggregation and improve stability. Reported antibody concentrations formulated as described above are typically up to about 100 mg/mL (Wang et al, J. Pharm. Sci. 96: l-26, 2007).
US. Patent No. 7,758,860 describes reducing the viscosity in formulations of low-molecular-weight proteins using a buffer and a Viscosity-reducing inorganic salt, such as m de or magnesium de. These same salts, however, showed little effect on the viscosity of a high-molecular-weight antibody (IMA—63 8) formulation. As described in US. Patent No. 7,666,413, the viscosity of aqueous formulations of high- molecular-weight proteins has been reduced by the addition of such salts as arginine hydrochloride, sodium thiocyanate, ammonium thiocyanate, ammonium sulfate, ammonium de, calcium chloride, zinc chloride, or sodium acetate in a tration of greater than about 100 mM or, as described in US. Patent No. 7,740,842, by addition of organic or inorganic acids. However, these salts do not reduce the ity to a desired level and in some cases make the formulation so acidic that it is likely to cause pain at the site of injection.
US. Patent No. 7,666,413 describes reduced-viscosity formulations containing specific salts and a tituted anti-lgE mAb, but with a maximum antibody concentration of only up to about 140 mg/mL. US.
Patent No. 7,740,842 describes E25 anti-IgE mAb ations containing acetate/acetic acid buffer with antibody concentrations up to 257 mg/mL.
The addition of salts such as NaCl, CaClg, or MgCiz was demonstrated to decrease the dynamic Viscosity under high-shear conditions; however, at low-shear the salts produced an undesirable and dramatic increase in the c viscosity. Additionally, inorganic salts such as NaCl may lower solution ity and/0r decrease aggregation (EP 1981824).
Non-aqueous antibody or protein formulations have also been described. /071693 bes a non-aqueous suspension of up to 100 mg/mL mAb in a formulation having a Viscosity enhancer (polyvinyipyrrolidone, PVP) and a solvent (benzyl benzoate or PEG 400).
W02004/089335 describes 100 mg/mL non-aqueous lysozyme suspension formulations ning PVP, glycofurol, benzyl benzoate, benzyl alcohol, or PEG 400. /0226689A1 describes 100 mg/mL human growth hormone (hGH) single phase, three vehicle component (polymer, surfactant, and a solvent), non-aqueous, Viscous formulations. U.S. Patent No. 6,730,328 describes non-aqueous, hydrophobic, lar vehicles of low reactivity, such as perfluorodecalin, for protein formulations. These formulations are non-optimal and have high viscosities that impair processing, manufacturing and injection; lead to the presence of multiple vehicie components in the formulations; and present ial regulatory nges associated with using polymers not yet approved by the FDA.
Alternative non—aqueous protein or antibody formulations have been described using organic solvents, such as benzyl benzoate (Miller er £11., Langmuir 26:1067-1074, 2010), benzyl acetate, ethanol, or methyl ethyl ketone (Srinivasan er LIL, Pharm. Res. 30: 1749-1757, 2013). In both instances, viscosities of less than 50 centipoise (cP) were achieved upon 2014/055254 formulation at protein concentrations of at least about 200 mg/mL. U.S.
Patent No. 6,252,055 describes mAb formulations with concentrations ranging from 100 mg/mL up to 257 mg/mL. Formulations with concentrations greater than about 189 mg/mL demonstrated dramatically increased Viscosities, low recovery rates, and difficulty in processing. US.
Patent ation ation No. 20 1 2/0230982 describes dy formulations with concentrations of 100 mg/rnL to 200 mg/mL. None of these formulations are low enough viscosity for ease of injection.
Du and Klibanov (Biotechnology and Bioengineering 108:632—636, 2011) described reduced ity of concentrated aqueous solutions of bovine serum albumin with a maximum concentration up to 400 mg/mL and bovine gamma globulin with a maximum concentration up to 300 mg/mL.
Guo er al. (Pharmaceutical ch 29231026109, 2012) described low- ity aqueous solutions of four model rnAbs achieved using hydrophobic salts. The mAb formulation employed by Guo had an initialviscosity, prior to adding salts, no greater than 73 CF. The Viscosities of many pharmaceutically ant mAbs, on the other hand, can exceed 1,000 cP at therapeutically relevant concentrations.
It is not a trivial matter to l aggregation and viscosity in high— concentration mAb solutions (EP 253 8973). This is evidenced by the few mAb products currently on the market as high—concentration formulations (> 100 mg/rnL) (EP 253 8973).
The references cited above demonstrate that while many groups have attempted to prepare low-viscosity formulations of mAbs and other eutically important proteins, a truly usefiil formulation for many proteins has not yet been achieved. y, many ofthe above reports employ agents for which safety and toxicity profiles have not been fully established. These formulations would therefore face a higher regulatory burden prior to approval than formulations containing compounds known to be safe. Indeed, even if a compound were to be shown to substantially reduce viscosity, the compound may ultimately be unsuitable for use in a formulation intended for injection into a human.
Many pharmaceutically important high-molecular-weight proteins, such as rnAbs, are currently administered via IV infusions in order to deliver therapeutically effective amounts of n due to problems with high viscosity and other properties of concentrated solutions of large proteins.
For example, to provide a therapeutically effective amount ofmany high- molecular-«weight proteins, such as mAbs, in volumes less than about 2 mL, n concentrations greater than 150 mg/mL are often required.
It is, therefore, an object of the present ion to provide concentrated, low-viscosity liquid formulations of pharmaceutically important proteins, especially high-molecular—weight proteins, such as mAbs.
It is a further object of the present invention to e concentrated low—Viscosity liquid formulations of proteins, especially high-molecular" weight proteins, such as mAbs, capable of ring eutically effective amounts of these proteins in volumes useful for SC and 1M injections.
It is a further object of the present invention to provide the concentrated liquid formulations of proteins, especially olecular- weight proteins, such as mAbs, with low viscosities that can improve inj ectability and/or patient compliance, convenience, and comfort.
It is also an object of the present invention to provide methods for making and storing concentrated, low-viscosity formulations of proteins, especially high-moiecular-vveight proteins, such as mAbs.
It is an additional obj ect of the t invention to provide methods of administering iow-viscosity, concentrated liquid formulations of proteins, especially high-«molecular—weight proteins, such as mAbs. It is an additional object of the present invention to provide methods for processing reduced- viscosity, high-concentration biologics With tration and ion techniques known to those skilled in the art.
SUMMARY OF THE INVENTION Concentrated, low-viscosity, low-volume liquid pharmaceutical formulations of proteins have been developed. Such formulations can be rapidly and conveniently administered by subcutaneous (SC) or intramuscular (1M) ion, rather than by lengthy intravenous infiision.
These formulations include lecular-weight and/or olecular- Weight proteins, such as mAbs, and viscosity-lowering agents that are typically bulky polar organic compounds, such as many of the GRAS (US Food and Drug Administration’s list of nds generally regarded as safe), inactive inj ectable ingredients and FDA-approved therapeutics.
The concentration ofproteins is between about 10 mg/mL and about ,000 mg/mL, more preferably from about 100 mg/mL to about 2,000 mg/mL. In some embodiments, the concentration ofproteins is between about 100 mg/mL to about 500 mg/mL, more preferably from about 300 mg/mL to about 500 mg/mL. Formulations containing proteins and Viscosity-lowering agents are stable when stored at a temperature of 4° C, for a period of at least one month, preferably at least two months, and most preferably at least three months. The ity of the formulation is less than about 75 CF, preferably below 50 CF, and most preferably below 20 cP at about 25° C. In some embodiments, the viscosity is less than about 15 cP or even less than or about 10 C]? at about 25° C. In certain embodiments, the viscosity of the ation is about 10 cP. Formulations containing proteins and ity-lowering agents typically are measured at shear rates from about 0.6 s'1 to about 450 3'1, and preferably from about 2 s'1 to about 400 s", when ed using a cone and plate viscometer. Formulations containing ns and Viscosity—lowering agents typically are measured at shear rates from about 3 s'1 to about 55,000 5'}, and preferably from about 20 s"1 to about 2,0005'1,when measured using a microfluidic viscometer.
The viscosity of the protein formulation is reduced by the ce of one or more viscosity—lowering agents. Unless specifically stated otherwise, the term “viscosity-lowering agen ” includes both single compounds and es of two or more compounds. It is preferred that the viscosity- lowering agent is present in the formulation at a concentration less than about 1.0 M, preferably less than about 0.50 M, more preferably less than about 0.30 M, and most preferably less than about 0.15 M. In some embodiments, the viscosity-lowering agent is present in the formulation in concentrations as low as 0.01 M. The formulations can have a viscosity that ' is at least about 30% less, ably at least about 50% less, most preferably at least about 75% less, than the viscosity of the corresponding formulation under the same ions except for replacement of the Viscosity-lowering agent with an appropriate buffer or salt of about the same concentration. In some embodiments, a low-viscosity formulation is provided where the viscosity of the corresponding formulation without the viscosity—lowering agent is greater than about 200 cP, greater than about 500 GP, or even above about 1,000 cP. In a preferred embodiment, the shear rate of the formulation is at least about 0.5 5‘1, when measured using a cone and plate viscometer or at least about 1.0 s'l, when ed using a microfluidic viscometer.
For embodiments in which the protein is a “high-molecular-weight protein”, the high molecular weight protein may have a molecular weight between about 100 kDa and about 1,000 kDa, preferably between about 120 kDa and about 500 kDa, and most preferably n about 120 kDa and about 250 kDa. The high-molecular-weight protein can be an antibody, such as a mAb, or a PEGylated, or otherwise a derivatized form thereof. Preferred mAbs include natalizumab (TYSABRI®), cetuximab (ERBITUX®), bevacizumab (AVASTIN®), trastuzumab (HERCEPTIN®), infliximab (REMICADE®), rituxiinab AN®), paniturnumab (VECTIBIX®), ofatumumab (ARZERRA®), and biosimilars thereof. The olecular- weight protein, optionally PEGylated, can be an enzyme. Other proteins and es ofproteins may also be formulated to reduce their viscosity.
In some ments, the protein and viscosity—lowering agent are ed in a lyophilized dosage unit, sized for reconstitution with a sterile aqueous pharmaceutically acceptable vehicle, to yield the concentrated low- viscosity liquid formulations. The presence of the viscosity-lowering agent(s) facilitates and/or accelerates the titution of the iyophilized dosage unit compared to a lyophiiized dosage unit not containing a Viscosity- lowering agent.
Methods are ed herein for preparing concentrated, low- viscosity liquid ations of high-molecular—weight proteins such as mAbs, as well as methods for g the low~viscosity, high—concentration protein formulations, and for stration thereof to patients. In another embodiment, the viscosity-lowering agent is added to facilitate processing (e.g., pumping, concentration, and/or filtration) by reducing the viscosity of the protein solutions.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 depicts the ity in cP as a n of the protein concentration (in mg/mL) for solutions of biosimilar cetuximab (ERBITUX®) in 0.25 M phosphate buffer (PB; diamonds) and a solution containing 0.25 M camphorsulfonic acid L—lysine (CSAL; squares) at 25°C and final pH of 7.0. The data points incorporate standard deviations which, r, are often smaller than the symbols.
Figure 2 depicts the viscosity in CF as a on of the protein concentration (in mg/mL) for solutions of biosimilar bevacizumab (AVASTIN®) in 0.25 M phosphate buffer (PB; diamonds) and 0.25 M CSAL (squares) at 25°C and final pH of 7.0. The data points incorporate standard deviations which, however, are often smaller than the symbols.
Figure 3 is a graph of the Viscosity (cP) of aqueous solutions of 200 i 9 mg/mL biosimilar zumab (AVASTIN®) as a flinction ofpH along the x—axis containing either ate-citrate buffer or camphorsulfonic acid arginine (CSAA) at a concentration of 0.25 M.
Figure 4 is a bar graph comparing the fold reduction in Viscosity as a function ofpH for aqueous solutions containing biosimilar zumab (AVASTIN®; at approximately 200 mg/mL or 226 mg/mL) and 0.25 M camphorsulfonic acid arginine (CSAA). The fold ion is computed as the ratio of the viscosity (CF) in phosphate-citrate buffer to the viscosity (cP) in the 0.25 M CSAA solution.
Figure 5 is a graph of the viscosity (cP) of aqueous solutions of biosimiiar cetuximab (ERBITUX®; at 202 :t 5 mg/mL, 229 d: 5 mg/mL, or 253 :1: 4 mg/mL) containing 0.25 M CSAA as a function ofpH along the X- axis at 25°C.
Figure 6 is a size—exclusion chromatography trace depicting absorbance intensity (at 280 nm) as a function of n time (in minutes) for a 220 mg/InL aqueous solution of REMICADE® stored at 4°C for up to 100 days, ed to freshly reconstituted connnercial drug product.
Figure 7 depicts the Viscosity (cP) as a function of protein concentration (mg/mL) of aqueous solutions of biosimilar zumab (AVASTIN®) in 0.25 M phosphate buffer, 0.10 M or 0.25 M APMI*2HC1 ((1—(3 -arninopropyl)—2—methy1—1H~imidazole bis-HCI).
Figure 8 s the Viscosity (GP) as a function of protein tration ) of aqueous solutions of biosimilar bevacizumab (AVASTIN®) in 0.25 M phosphate buffer, 0.10 M thiamine pyrophosphate (TPP), or 0.10 M TPP1-(3-aminopropyl)—2—methyl—lH-imidazole (APMI).
Figure 9 depicts the Viscosity (0P) of aqueous solutions of golimumab NI ARIA®) as a on ofprotein concentration (mg/mL) with 0.15 M phosphate buffer or 0.15 M thiamine HCl.
DETAILED PTION OF THE INVENTION I. DEFINITIONS The term "protein," as generally used , refers to a polymer of amino acids linked to each other by peptide bonds to form a polypeptide for which the chain length is sufficient to produce at least a detectable tertiary structure. Proteins having a moleculm weight (expressed in kDa wherein “Da” stands for “Daltons” and 1 kDa = 1,000 Da) greater than about 100 kDa may be ated “high-molecular-weight proteins,” whereas ns having a molecular weight less than about 100 kDa may be designated “low- molecular-weight proteins.” The term “low-molecular-weight protein” excludes small peptides lacking the requisite of at least tertiary structure necessary to be considered a protein. Protein molecular weight may be determined using standard methods known to one skilled in the art, including, but not limited'to, mass spectrometry (e.g., ESI, MALDI) or calculation from known amino acid sequences and glycosylation. Proteins can be naturally occurring or non—naturally occurring, synthetic, or semi— synthetic.
“Essentially pure protein(s)” and "substantially pure protein(s)” are used interchangeably herein and refer to a composition comprising at least about 90% by weight pure protein, preferably at least about 95% pure protein by weight. “Essentially homogeneous” and antially homogeneous” are used interchangeably herein and refer to a composition wherein at least about 90% by weight of the protein present is a combination of the monomer and ible di- and oligo-meric associates (not irreversible aggregates), preferably at least about 95%.
The term “monoclonal antibody” or “mAb,” as generally used herein, refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual dies comprising the population are identical, except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single epitope. These are lly synthesized by culturing hybridoma cells, as described by Kohler et al. (Nature 256: 495, 1975), or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567), or ed from phage antibody libraries using the ques described in Clackson et al. (Nature 352: 624-628, 1991) and Marks et al. (J. Mol. Biol. 222: 7, 1991), for example. As used herein, “mAbs” ically include derivatized antibodies, antibody-drug conjugates, and “chimeric” antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is (are) identical with or homologous to corresponding sequences in antibodies derived from r species or belonging to another antibody class or subclass, as well as nts of such antibodies, so long as they exhibit the desired biological activity (U.S. Patent No. 4,816,567; on et al., Proc. Natl. Acad. Sci. USA 1-6855, 1984).
An “antibody fragment” comprises a portion of an intact antibody, including the antigen binding and/or the variable region of the intact antibody. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (see U.S. Patent No. 5,641,870; Zapata et al., Protein Eng. 8:1057-1062, 1995); single-chain antibody molecules; multivalent single domain antibodies; and multispecifie antibodies formed from antibody fragments.
“Humanized” forms ofnon-human (e.g., murine) dies are chimeric immunoglobulins, immunoglobulin chains, or fragments thereof (such as Fv, Fab; Fah', F(ab’)2, or other antigen-binding subsequences of antibodies) of mostly human ces, which contain minimal sequences derived from non-human immunoglobulin. (See, e.g.; Jones et al. , Nature 2-525, 1986; Reichmann at £11., Nature 332:323-329, 1988; and Presta, Curr. 0p. Struct. Biol. 2:593-596, 1992.) “Rheology” refers to the study of the deformation and flow of matter.
“Viscosity" refers to the resistance of a substance (typically a liquid) to flow. Viscosity is d to the concept of shear force; it can be understood as the effect of different layers of the fluid exerting shearng force on each other, or on other surfaces, as they move against each other.
There are several measures of Viscosity. The units of viscosity are Ns/rnz, known as Pascal-seconds (Pa-s). Viscosity can be "kinematic” or "absolute".
Kinematic viscosity is a measure of the rate at which momentum is transferred h a fluid. It is measured in Stokes (St). The kinematic viscosity is a measure of the ive flow of a fluid under the ce of gravity. When two fluids of equal volume and differing Viscosity are placed in identical capillary viscometers and allowed to flow by gravity; the more viscous fluid takes longer than the less viscous fluid to flow h the capillary. If, for example, one fluid takes 200 seconds (s) to te its flow and another fluid takes 400 s, the second fluid is called twice as viscous as the first on a kinematic viscosity scale. The dimension ofkinematic viscosity is lengch/time. Commonly, kinematic viscosity is expressed in tokes (cSt). The S1 unit of kinematic viscosity is mmzls, which is equal to 1 cSt. The "absolute viscosity,“ sometimes called ”dynamic viscosity" or ”simple Viscosity," is the product of tic Viscosity and fluid density.
Absolute ity is expressed in units of centipoise (CF). The SI unit of absolute viscosity is the milliPascal-second (mPa—s), where 1 OP = 1 mPa—s. ity may be measured by using, for example, a viscometer at a given shear rate or multiple shear rates. An “extrapolated zero-shear” viscosity can be determined by creating a best fit line of the four highest- shear points on a plot of absolute viscosity versus shear rate, and linearly extrapolating viscosity back to zero-shear. Alternatively, for a Newtonian fluid, viscosity can be ined by averaging viscosity values at multiple shear rates. Viscosity can aiso be measured using a microfluidic viscometer at single or multiple shear rates (also called flow , wherein absolute viscosity is derived from a change in pressure as a liquid flows through a channel. Viscosity equals shear stress over shear rate. Viscosities measured with microfluidic viscometers can, in some embodiments, be ly compared to extrapolated zero-shear viscosities, for example those extrapolated from viscosities measured at multiple shear rates using a cone and plate viscometer.
“Shear rate" refers to the rate of change of velocity at- which one layer of fluid passes over an adjacent layer. The ty gradient is the rate of change of velocity with ce from the plates. This simple case shows the uniform velocity gradient with shear rate (v1 - V2)/h in units of (cm/sec)/(cm) = llsec. Hence, shear rate units are reciprocal s or, in general, reciprocal time. For a microfluidic viscometer, change in pressure and flow rate are related to shear rate. "Shear rate" is to the speed with which a material is ed. ations containing ns and viscosity- lowering agents are typically measured at shear rates ranging from about 0.5 s‘1 to about 200 s'1 when measured using a cone and plate viscometer and a spindle appropriately chosen by one skilled in the art to accurately measure viscosities in the viscosity range of the sample of interest (i.e., a sample of 20 cP is most accurately measured on a CPE40 spindle affixed to a DV2T viscometer (Brookfield)); greater than about 20 s'1 to about 3,000 5'1 when measured using a microfluidic viscometer.
For classical nian” fluids, as generally used herein, viscosity is essentially independent of shear rate. For “non-Newtonian ” however, viscosity either decreases or increases with increasing shear rate, e.g., the fluids are "shear thinning" or "shear thickening", respectively. In the case of concentrated (i.e., high—concentration) protein solutions, this may manifest as plastic shear-thinning behavior, i.e., a decrease in viscosity with shear rate.
The term "chemical stability," as generally used herein, refers to the ability of the protein ents in a formulation to resist degradation via chemical pathways, such as ion, ation, or hydrolysis. A protein formulation is typically considered chemically stable if less than about 5% of the components are degraded after 24 months at 4° C.
The term "physical stability," as generally used herein, refers to the y of a n formulation to resist physical deterioration, such as aggregation. A ation that is physically stable forms only an acceptable percentage of irreversible aggregates (e.g., dimers, trimers, or other aggregates) of the bioactive protein agent. The ce of ates may be assessed in a number ofways, including by measuring the e particle size of the proteins in the formulation by means of dynamic light scattering.
A formulation is considered physicaily stable if less than about 5% irreversible aggregates are formed after 24 months at 4°C. Acceptable levels of aggregated contaminants ideally would be less than about 2%. Levels as low as about 0.2% are achievable, although approximately 1% is more typical.
The term "stable formulation," as generally used herein, means that a formulation is both chemically stable and physically stable. A stable formulation may be one in which more than about 95% of the bioactive protein molecules retain bioactivity in a formulation after 24 months of storage at 4° C, or equivalent solution conditions at an elevated temperature, such as one month storage at 40° C. Various analytical techniques for measuring protein stability are available in the art and are reviewed, for example, in Peptide and Protein Drug Delivery, 247—301, Vincent Lee, Ed, Marcel , Inc, New York, NY. (1991) and Jones, A., Adv. Drug Delivery Revs. 10:29-90, 1993. Stability can be measured at a selected temperature for a certain time period. For rapid screening, for example, the formulation may be kept at 40°C, for 2 weeks to one month, at which time 2014/055254 al biological activity is measured and compared to the initial condition to assess stability. When the formulation is to be stored at 2°C -8°C, generally the formulation should be stable at 30°C or 40°C for at least one month and/0r stable at 2°C -8°C for at least 2 years. When the formulation is to be stored at room temperature, about 25°C, generally the formulation should be stable for at least 2 years at about 25°C and/or stable at 40°C for at least about 6 months. The extent of aggregation following lyophilization and storage can be used as an indicator of protein stability. In some embodiments, the ity is assessed by measuring the le size of the proteins in the formulation In some embodiments, stability may be assessed by measuring the activity of a formulation using standard biological activity or binding assays well within the abilities of one ordinarily skilled in the art.
The term n cle size," as generally used herein, means the average diameter of the predominant population of bioactive molecule particulates, or particle size distributions thereof, in a formulation as determined by using well known particle sizing instruments, for e, dynamic light scattering, SEC (size ion chromatography), or other methods known to one ordinarily d in the art.
The term “concentrated” or "high—concentration", as generally used herein, describes liquid formulations having a final concentration of protein greater than about 10 mg/mL, preferably greater than about 50 mg/mL, more preferably greater than about 100 mg/rnL, still more preferably greater than about 200 mg/mL, or most preferably greater than about 250 mg/mL.
A “reconstituted formulation,” as generally used , refers to a formulation which has been prepared by dissolving a dry , lyophilized, spray-dried or solvent-precipitated protein in a diluent, such that the protein is dissolved or dispersed in aqueous solution for administration.
A “lyoprotectant” is a substance which, when combined with a protein, cantly reduces chemical and/or physical instability of the protein upon lyophilization and/or subsequent storage. Exemplary lyoprotectants include sugars and their corresponding sugar alcohols, such as sucrose, lactose, trehalose, dextran, erythritol, arabitol, xylitol, sorbitol, and mannitol; amino acids, such as arginine or histidine; lyotropic salts, such as magnesium sulfate; s, such as propylene glycol, glycerol, poly(ethylene glycol), or poly(propylene glycol); and combinations thereof.
Additional exemplary lyoprotectants include gelatin, ns, modified starch, and carboxymethyl cellulose. Preferred sugar ls are those compounds obtained by reduction of mono- and di-saccharides, such as e, trehalose, maltose, ose, and maltulose. onal examples of sugar alcohols are glucitol, maltitol, lactitol and isomaltulose. The lyoprotectant is generally added to the pre-lyophilized formulation in a “lyoprotecting amount.” This means that, following lyophilization of the protein in the presence of the lyoprotecting amount of the lyoprotectant, the protein essentially retains its physical and chemical stability and integrity.
A “diluent” or “carrier,” as generally used herein, is a pharmaceutically acceptable (i.e., safe and non-toxic for administration to a human or another mammal) and useful ingredient for the preparation of a liquid formulation, such as an aqueous formulation reconstituted after lyophilization. Exemplary diluents include sterile water, bacteriostatic water for injection , a pH ed solution (e.g., phosphate-buffered saline), steriie saline on, Ringer's solution or dextrose solution, and combinations f.
A “preservative” is a compound which can be added to the formulations herein to reduce contamination by and/or action of bacteria, fungi, or another infectious agent. The addition of a preservative may, for example, facilitate the production of a multi-use (multiple-dose) formulation. es ofpotential preservatives include cyldimethylbenzylammonium chloride, thonium chloride, benzalkonium chioride (a mixture of enzyldimethylammonium chlorides in which the aikyl groups are long-chained), and benzethonium chloride. Other types of preservatives include aromatic alcohols such as phenol, butyl and benzyl alcohol, alkyl parabens such as methyl or propyl paraben, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol.
A “bulking agent,” as generally used , is a compound which adds mass to a lized mixture and contributes to the physical structure ofthe lyophilized cake (e.g. facilitates the production of an essentially uniform lyophilized cake which maintains an open pore structure).
Exemplary bulking agents include mannitol, glycine, lactose, modified starch, poly(ethylene glycol), and sorbitol.
A “therapeutically effective amoun ” is the least tration required to effect a measurable improvement or prevention of any symptom or a particular condition or disorder, to effect a measurable enhancement of life expectancy, or to generally improve patient y of life. The therapeutically effective amount is dependent upon the specific biologically active molecule and the specific condition or disorder to be treated.
Therapeutically effective amounts ofmany proteins, such as the mAbs described herein, are well known in the art. The eutically ive amounts ofproteins not yet established or for treating specific disorders with known proteins, such as mAbs, to be clinically d to treat additional disorders may be determined by standard techniques which are well within the craft of a skilled artisan, such as a physician.
The term tability" or “syringeability,” as generally used herein, refers to the injection performance of a pharmaceutical formulation through an. syringe equipped with an 18-32 gauge needle, optionally thin walled.
Inj ectability depends upon factors such as pressure or force required for injection, evenness of flow, aspiration qualities, and fieedom from clogging.
Injectabiiity of the liquid pharmaceutical formulations may be ed by comparing the injection force of a reduced-Viscosity fomiulation to a standard formulation t added viscosity-lowering . The reduction in the injection force of the formulation containing a Viscosity-lowering agent reflects improved injectability of that formulation. The reduced Viscosity formulations have improved inj ectability when the injection force is reduced by at least 10%, preferably by at least 30%, more preferably by at least 50%, and most preferably by at least 75% when compared to a standard formulation having the same concentration ofprotein under ise the same conditions, except for replacement of the viscosity-lowering agent with an riate buffer of about the same concentration. Alternatively, inj ectability of the liquid pharmaceutical formulations may be assessed by comparing the time ed to inject the same volume, such as 0.5 mL, or more preferably about 1 mL, of different liquid protein formulations when the e is depressed with the same force.
The term “inj ecti0n force," as generally used herein, refers to the force ed to push a given liquid formulation through a given syringe equipped with a given needle gauge at a given injection speed. The injection force is typically reported in s. For example, the injection force may be measured as the force required to push a liquid formulation through a 1 mL plastic syringe having a 0.25 inch inside diameter, equipped with a 0.50 inch 27 gauge needle at a 250 nun/min ion speed. g equipment can be used to measure the ion force. When measured under the same conditions, a formulation with lower Viscosity will generally e an overall lower injection force.
The “viscosity gradient,” as used herein, refers to the rate of change ofthe viscosity of a protein solution as protein concentration increases. The viscosity gradient can be approximated from a plot of the Viscosity as a function of the protein concentration for a series of formulations that are otherwise the same but have different protein concentrations .u. The viscosity increases approximately exponentially with increasing protein concentration.
The viscosity gradient ata specific protein concentration can be approximated from the slope ‘of a line tangent to the plot of viscosity as a function of protein concentration. The viscosity gradient can be approximated from a linear approximation to the plot of ity as, a function of any protein concentration or over a narrow window of protein concentrations. In some ments a formulation is said to have a decreased viscosity gradient if, when the viscosity as a function of protein concentration is approximated as an ntial function, the exponent of the exponential function is smaller than the exponent obtained for the otherwise same formulation without the viscosity~lowering agent. In a similar manner, a formulation can be said to have a lower/higher ity gradient when compared to a second formulation if the exponent for the formulation'is lower/higher than the exponent for the second formulation.
The viscosity gradient can be numerically approximated from a plot of the ity as a function of protein concentration by other methods known to the skilled formulation researchers.
The term ed-visco sity ation,” as generally used herein, refers to a liquid formulation having a high concentration of a high- molecular—weight protein, such as a mAb, or a low-«molecular-weight n that is modified by the presence of one or more additives to lower the viscosity, as compared to a corresponding formulation that does not contain the viscosity-lowering additive(s).
The term “osmolarity,” as generally used , refers to the total number of dissolved components per liter. rity is similar to ty but includes the total number of moles of dissolved Species in solution. An osmolarity of l Osm/L means there is 1 mole of dissolved components per L of solution. Some solutes, such as ionic solutes that dissociate in solution, will contribute more than 1 mole of dissolved components per mole of solute in the solution. For example, NaCl dissociates into Na+ and CT in solution and thus provides 2 moles of ved components per 1 mole of ved NaCl in solution. Physiological osmolarity is typically in the range of about 280 mOsm/L to about 310 mOsrn/L.
The term “tonicity,” as generally used herein, refers to the osmotic re gradient resulting from the separation oftwo solutions by a semi- permeable membrane. In particular, tonicity is used to describe the osmotic pressure created across a cell membrane when a cell is exposed to an external on. Solutes that can cross the cellular membrane do not contribute to the final osmotic pressure nt. Only those dissolved species that do not cross the cell membrane will contribute to osmotic pressure differences and thus tonicity.
The term “hypertonic,” as generally used herein, refers to a solution with a higher concentration of solutes than is present on the inside of the cell.
When a cell is immersed into a hypertonic solution, the tendency is for water to flow out ofthe cell in order to balance the concentration of the solutes.
The term “hypotonic,” as generally used herein, refers to a solution with a lower tration of solutes than is present on the inside of the cell.
When a cell is immersed into a hypotonic solution, water flows into the cell in order to balance the concentration of the solutes.
The term “isotonic,” as generally used herein, refers to a solution wherein the osmotic re gradient across the cell membrane is essentially balanced. An ic formulation is one which has essentially the same osmotic pressure as human blood. Isotonic formulations will generally have an osmotic re from about 250 mOsm/kg to 350 mOsm/kg.
The term “liquid formulation,” as used herein, is a protein that is either ed in an acceptable pharmaceutical diluent or one that is reconstituted in an acceptable pharmaceutical diluent prior to stration to the patient.
The terms “branded” and “reference,” when used to refer to a protein or biologic, are used interchangeably herein to mean the single biological product licensed under section 351(a) of the US. Public Health Service Act (42 use. § 262).
The term “biosimilar,” as used herein, is generally used interchangeably with “a generic equivalent” or “follow-on.” For example, a “biosimilar'mAb” refers to a subsequent version of an innovator’s mAb typically made by a different company. “Biosimilar” When used in reference to a branded protein or branded biologic can refer to a biological product ted against the branded protein or branded biologic and licensed under section 351(k) of the US. Public Health e Act (42 U.S.C. § 262). A biosimilar mAb can be one that satisfies one or more guidelines adopted May , 2012 by the Committee for Medicinal Products for Human Use (CHMP) ofthe an Medicines Agency and published by the European Union as “Guideline on similar biological medicinal products containing monoclonal antibodies — non—clinical and clinical issues" (Document Reference EMA/CHMP/BMWP/403 543/2010).
Biosimilars can be produced by ial cells (prokaryotic, eukaryotic), cell lines of human or animal origin (e.g., mammalian, avian, insect), or tissues derived from animals or plants. The expression construct for a proposed biosimilar t will generally encode the same primary amino acid sequence as its reference product. Minor modifications, such as N- or C— terminal truncations that will not have an effect on safety, purity, or potency, may be present.
A biosimilar mAb is r to the reference mAb physiochemically or biologically both in terms of safety and efficacy. The biosimilar mAb can be ted against a reference mAb using one or more in vitro studies including assays ing g to target antigen(s); binding to isoforms of the Fc gamma receptors (Fm/R1, Fc'yRII, and FcyRIII), FcRn, and complement (Clq); Fab-associated functions (e.g. neutralization of a e ligand, receptor activation or blockade); or Fc-associated functions (6.g. antibody-dependent cell-mediated cytotoxicity, complement-dependent cytotoxicity, complement activation). In vitro comparisons may be combined with in vivo data demonstrating similarity of cokinetics, pharmacodynamics, and]or safety. Clinical evaluations of a biosimilar mAb against a nce mAb can include comparisons ofpharmacokinetic properties (e.g. AUCO_inf, AUCM, Cum, tmax, ; codynamic endpoints; or similarity of clinical efficacy (e.g. using randomized, parallel group ative clinical trials). The quality comparison between a biosimilar mAb anda reference mAb can be evaluated using established procedures, ing those described in the “Guideline on similar biological nal products containing biotechnology—derived ns as active substance: Quality issues” (EMEA/CHMP/BWP/49348/2005), and the “Guideline on development, production, characterization and specifications for monoclonal antibodies and related substances” (EMEA/CHMP/BWP/l 2007).
Differences between a biosimilar mAb and a reference mAb can include post-translational modification, 6.g. by attaching to the mAb other biochemical groups such as a phosphate, various lipids and carbohydrates; by proteolytic cleavage following translation; by changing the chemical nature of an amino acid (cg, formyiation); or by many other mechanisms. Other post—translational ations can be a consequence ofmanufacturing process operations — for example, glycation may occur with exposure ofthe product to reducing sugars. In other cases, storage conditions may be permissive for certain ation pathways such as oxidation, deamidation, or aggregation. As all of these product-related variants may be included in a biosimilar mAb.
The term “viscosity-lowering agent,” as used herein, refers to a nd which acts to reduce the viscosity of a solution relative to the Viscosity of the solution absent the Viscosity~lowering agent. The viscosity— lowering agent may be a single compound, or may be a mixture of one or more nds. When the viscosity—lowering agent is a mixture of two or more compounds, the listed concentration refers to each individual agent, WO 38818 unless otherwise specified. By way of example, a formulation containing about 0.25 M camphorsulfonic acid arginine as the viscosity-lowering agent is a solution having rsulfonic acid at a concentration of 0.25 M, and arginine at a concentration of 0.25 M.
Certain Viscosity-lowering agents contain acidic or basic onal groups. Whether or not these funCtional groups are fillly or partiaily ionized depends on the pH ofthe formulation they are in. Unless otherwise specified, reference to a formulation ning a ity-lowering agent having an ionizable functional group includes both theparent compound and any possible ionized states.
As used herein, the term “hydrogen bond donor” refers to a hydrogen atom connected to a vely electronegative atom, which creates a partial positive charge on the hydrogen atom.
As used , the term “hydrogen bond acceptor” refers to a relatively electronegative atom or functional group capable of cting with a hydrogen atom bearing a partial positive charge.
As used herein, the term “fieely rotating bond” refers to any singly bonded pair of non-hydrogen atoms.
As used herein, the term “molecular polar surface area” refers to the total exposed polar area on the surface of the molecule of interest.
As used herein, the term “molar volume” refers to the total volume that one mole of the molecule of st occupies in its native state (i.e. solid, liquid).
As used herein, the term “polarizability” refers to the induced dipole moment when the le of interest is placed in an electric field of unit strength.
As used herein, the term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable non-toxic acids and bases, ing inorganic acids and bases, and organic acids and bases. Suitable non-toxic acids include inorganic and organic acids such as , benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric acid, p-toluenesulfonic and the like.
Suitable positively charged counterions include sodium, potassium, lithium, calcium and magnesium.
As used herein, the term "ionic liquid” refers to a crystalline or amorphous salt, zwitterion, or mixture thereof that is a liquid at or near temperatures where most conventional salts are solids: at less than 200°C, preferably less than 100°C or more preferably less than 80°C. Some ionic liquids have melting temperatures around room temperature, e.g. between ' 10°C and 40°C, or between 15°C and 35°C. The term erion" is used herein to describe an overall lly charged le which carries formal positive and negative s on different chemical groups in the molecule.
Examples of ionic liquids are described in Riduan er al., Chem. Soc. Rev., 42:9055-9070, 2013; Rantwijk et at, Chem. Rev., 107:2757-2785, 2007; Earle er al, Pure Appl. Chem, 72(7): 1391-1398, 2000; and Sheldon et all, Green Chem, 4:147—151, 2002.
As used herein, the term “organophosphate” refers to a nd containing one or more phosphoryl groups at least one of which is covalently ted to an organic group through a phosphoester bond.
As used herein, a “water soluble organic dye” is an organic molecule having a molar solubility of at least 0.001 M at 25°C and pH 7, and that s certain wavelengths of light, preferably in the visible-to-infrared portion of the electromagnetic spectrum, While possibly transmitting or reflecting other wavelengths of light.
As used herein, the term ogen” refers to Group 16 elements, including oxygen, sulfur and selenium, in any oxidation state. For instance, unless specified otherwise, the term “chalcogen” also includes 802.
As used herein, term “alkyl group” refers to straight-chain, branched- chain and cyclic hydrocarbon groups. Unless specified otherwise, the term alkyl group embraces hydrocarbon groups ning one or more double or triple bonds. An alkyl group containing at least one ring system is a “cycloalkyl” group. An alkyl group containing at least one double bond is an “alkenyl group,” and an alkyl group ning at least one triple bond is an “alkynyl group.” The term as used herein, “Ary ” refers to aromatic carbon ring systems, including fused ring systems. In an “ary ” group, each of the atoms that form the ring are carbon atoms.
The term as used herein “Heteroaryl” refers to aromatic ring systems, ing fused ring systems, wherein at least one of the atoms that forms the ring is a heteroatom.
The term as used herein “Heterocycle” refers to ring systems that, including fused ring systems, are not aromatic, n at least one of the atoms that forms the ring is a heteroatorn.
The term as used herein, “heteroatom” is any non-carbon or non- hydrogen atom. Preferred atoms include , sulfur, and en.
Exemplary aryl and heterocyclyl rings include: benzimidazolyl, benzofilranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, , benzoxazolinyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH. carbazolyl, inyl, nyl, chromenyl, cinnolinyl, decahydroquinolinyl, 1,5,2- dithiazinyl, dihydrofuro [2,3 b]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lHnindazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H—indolyl, isatinoyl, isobenzofuranyl, isochrornanyl, isoindazolyl, olinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3—oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazoly1, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, nyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, zinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H- quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, ydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H—1,2,5- thiadiazinyl, 1,2,3—thiadiazolyl, 1,2,4-thiadiazoly1, 1,2,5-thiadiazolyl, 1,3,4- thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, and xanthenyl. 2014/055254 II. FORMULATIONS Biocompatible, low-viscosity protein solutions, such as those of mAbs, can be used to deliver therapeutically effective amounts of proteins in volumes useful for subcutaneous (SC) and intramuscular (1M) ions, typically less than or about 2 mL for SC and less than or about 5 mL for 1M, more preferably less than or about 1 mL for SC and less than or about 3 mL for IM. The ns can generally have any molecular weight, although in some ments high~molecular~weight proteins are preferred. In other embodiments the proteins are low-molecular-weight proteins.
Formulations may have protein concentrations between about 10 mg/mL and about 5,000 mg/mL. The formulations, including mAb ations, may have a protein concentration greater than 100 mg/mL, preferably greater than 150 mg/mL, more preferably r than about 175 mg/ml, even more preferably greater than about 200 mg/mL, even more preferably greater than about 225 mg/mL, even more preferably greater than about 250 mg/mL, and most preferably greater than or about 300 mg/mL. In the absence of a viscosity-lowering agent, the viscosity of a n formulation increases exponentially as the concentration is increased. Such protein formulations, in the absence of a viscosity-lowering agent, may have viseosities greater than 100 GP, greater than 150 cP, greater than 200 cP, greater than 300 cP, greater than 500 cP, or even greater than 1,000 cP, when ed at 25° C. Such formulations are often unsuitable for SC or IM injection. The use of one or more viscosity-lowering agents permits the preparation of formulations having a viscosity less than or about 100 cP, preferably less than or about 75 CF, more ably less than or about 50 CF, even more preferably less than or about 30 GP, even more preferably less than or about 20 cP, or most preferably less than or about 10 CF, when measured at 25° C.
Although the viscosity—lowering agents may be used to lower the viscosity of concentrated n formulations, they may be used in less» concentrated formulations as well. In some embodiments, formulations may have protein concentrations between about 10 mg/mL and about 100 mg/mL.
The formulations may have a protein concentration greater than about 20 mg/mL, greater than about 40 mg/mL, or greater than about 80 mg/mL.
For n ns, formulations not having a viscosity-lowering agent may have ities greater than about 20 cP, r than about 50 CF, or greater than about 80 CF. The use of one or more viscosity-lowering agents permits the preparation of formulations having a viscosity less than or about 80 CF, preferably less than or about 50 CF, even more preferably less than about 20 CR or most preferably less than or about 10 CF, when measured at 25° C.
In some embodiments, the aqueous protein formulations have a viscosity that is at least about 30% less than the analogous formulation Without the viscosity_lowering agent(s), when measured under the same conditions. In other embodiments, the formulations have a viscosity that is 40% less, 50% less, 60% less, 70% less, 80% less, 90% less, or even more than 90% less than the ous formulation t the viscosity-lowering agent(s). In a preferred embodiment, the formulation contains a therapeutically effective amount of the one or more high-molecular-weight proteins, such as mAbs, in a volume of less than about 2 mL, preferably less than about 1 mL, or more preferably less than about 0.75 mL.
The reduced-viscosity formulations have improved inj ectability and require less injection force compared to the analogous formulation without the viscosity—lowering agent (e.g., in phosphate buffer) under otherwise the same conditions. In some embodiments, the force of injection is decreased by more than about 20%, more than about 30%, more than about 40%, more than about 50%, or more than about 2 fold, as ed to standard formulations without the ity-lowering agent(s) under otherwise the same injection conditions. In some embodiments, the formulations s “Newtonian flow characteristics,” defined as having viscosity which is substantially independent of shear rate. The protein formulations can be readily injected through needles of about 18-32 gauge. red needle gauges for the delivery of the low-viscosity formulations include 27, 29, and 31 gauge, optionally thin walled.
The formulations may n one or more additional excipients, such as buffers, surfactants, sugars and sugar alcohols, other polyols, preservatives, antioxidants, and chelating agents. The formulations have a pH and osmolarity suitable for administration without causing significant adverse side effects. In some embodiments, the concentrated, low-viscosity formulations have a pH between 5 and 8, between 5 .5 and 7.6, n 6.0 and 7.6, between 6.8 and 7.6, or between 5.5 and 6.5.
The low-viscosity n formulations can allow for greater flexibility in formulation development. The low-viscosity formulations can exhibit changes in viscosity that are less dependent upon the protein concentration as compared to the otherwise same formulation t the viscosity-lowering agent. The low-viscosity protein formulations can allow for increased trations and decreased dosage frequencies ofthe protein.
In some embodiments the low-viscosity protein formulations contain 2 or more, 3 or more, or 4 or more different proteins. For example, combinations of 2 or more mAbs can be provided in a single low-viscosity protein formulation.
Because protein (such as mAb) formulations may be administered to patients at higher protein concentrations than otherwise. similar protein formulations not containing a viscosity~lowering agent, the dosing frequency of the protein can be reduced. For instance, ns previously requiring once daily administration may be administered once every two days, every three days, or even less frequently when the proteins are formulated with ity-lowering agents. Proteins which currently require multiple administrations on the same day (either at the same time or at ent times ofthe day) may be administered in fewer injections per day. In some instances, the fiequency may be reduced to a single injection once a day. By increasing the dosage administered per injection multiple-fold the dosing ncy can be decreased, for example from once every 2 weeks to once every 6 weeks.
In some embodiments, the liquid formulations have a logical osmolarity, for example, between about 280 mOsm/L to about 310 mOsm/L.
In some embodiments, the liquid formulations have an osmolarity greater than about 250 , greater than about 300 mOsm/L, greater than about 350 mOsm/L, greater than about 400 mOsm/L, or greater than about 500 mOsm/L. In some embodiments, the formulations have an osmolarity of about 200 mOsm/L to about 2,000 mOsm/L or about 300 mOsm/L to about 1,000 . In some embodiments, the liquid formulations are essentially isotonic to human blood. The liquid formulations can in some cases be hypertonic.
The additives, including the viscosity-lowering agents, can be included in any amount to achieve the d viscosity levels of the liquid formulation, as long as the amounts are not toxic or otherwise l, and do not substantially interfere with the chemical and/or physical stability of the formulation. The viscosity-lowering agent(s) in some embodiments can be independently present in a concentration less than about 1.0 M, preferably less than about 0.50 M, less than or equal to about 0.30 M or less ‘than or equal to 0.15 M. Especially preferred concentrations include about 0.15 M and about 0.30 M. For some embodiments having two or more viscosity~ lowering agents, the agents are preferably, but not necessarily, present at the same concentration.
The viscosity-lowering agents permit faster reconstitution of a lyophilized dosage unit. The dosage unit is a lyophilized cake of protein, ity—lowering agent and other excipients, to which water, saline or another pharmaceutically acceptable fluid is added. In the absence of viscosity—lowering agents, periods of 10 minutes or more are often ed in order to completely ve the lyophilized cake at high protein concentration. When the lyophilized cake contains one or more viscosity- lowering agents, the period required to completely ve the cake is often reduced by a factor oftwo, five or even ten. In certain ments, less than one minute is required to completely dissolve a lyophilized cake containing greater than or about 150, 200 or even 300 mg/mL of protein.
The low-viscosity protein formulations allow for greater flexibility in ation development. The low—viscosity formulations exhibit a viscosity that changes less with increasing protein trations as compared to the otherwise same formulation t the viscosity-lowering agent(s). The low—viscosity protein ations exhibit a decreased viscosity gradient as compared to the otherwise same formulation without the viscosity—lowering agent The viscosity gradient of the protein formulation may be 2-fold less,3-fold less, or even more than 3-fold less than the viscosity gradient of the otherwise same protein formulation without the viscosity-lowering agent(s). The viscosity gradient of the protein formulation may be less than 2.0 cP mL/mg, less than 1.5 cP mL/mg, less than 1.0 cP mL/mg, less than 0.8 0P mL/rng, less than 0.6 cP mL/mg, or less than 0.2 cP mL/mg for a n ation having a protein concentration between 10 mg/mL and 2,000 mg/mL. By reducing the viscosity gradient of the formulation, the protein concentration can be increased to a greater degree before an exponential l increase in viscosity is observed.
A. Proteins Any protein can be formulated, including recombinant, ed, or synthetic proteins, glycoproteins, or lipoproteins. These may be antibodies (including antibody fragments and recombinant antibodies), enzymes, growth factors or hormones, immunomodifiers, antiinfectives, antiproliferatives, es, or other therapeutic, prophylactic, or diagnostic ns. In certain embodiments, the protein has a lar weight greater than about 150 kDa, greater than 160 kDa, greater than 170 kDa, greater than 180 kDa, greater than 190 kDa or even greater than 200 kDa.
In certain embodiments, the protein can be a PEGylated protein. The term “PEGylated n,” as used herein, refers to a protein having one or more poly(ethylene glycol) or other stealth polymer groups ntly attached thereto, optionally through a chemical linker that may be ent from the one or more polymer groups. PEGylated proteins are characterized by their typically reduced renal filtration, decreased uptake by the reticuloendothelial system, and diminished enzymatic degradation leading to, for example, prolonged half-lives and enhanced ilability. Stealth polymers include poly(ethylene glycol); poly(propylene glycol); poly(amino acid) polymers such as poly(glutamic acid), poly(hydroxyethyl-L- gine), and poly(hydroxethyl-L-glutamine); poly(glycerol); poly(2— oxazoline) polymers such as poly(2—methyloxazoline) and poly(2-ethyl ine); poly(acrylamide); poly(vinylpyrrolidone); -(2~ hydroxypropyl)methacrylarnide); and copolymers and mixtures thereof. In preferred embodiments the h r in a PEGylated protein is poly(ethylene glycol) or a copolymer thereof. PEGylated proteins can be randomly PEGylated, 1'. e. having one or more stealth polymers covalently attached at non-specific site(s) on the protein, or can be PEGylated in a site- specific manner by ntly ing the stealth polymer to specific site(s) on the protein. Site-specific PEGylation can be accomplished, for example, using ted stealth polymers having one or more reactive onal groups. Examples are described, for instance, in Hoffman et all, Progress in Polymer Science, 32:922-932, 2007.
In the preferred embodiment, the protein is high-molecular-weight and an antibody, most preferably a mAb, and has a high viscosity in aqueous ed solution When concentrated sufficiently to inject a therapeutically effective amount in a volume not ing 1.0 to 2.0 mL for SC and 3.0 to .0 mL for 1M administration. High—molecular—weight proteins can include those described in Scolnik, mAbS 1:179-184, 2009; Beck, mAbs 110, . 2011; Baumann, Curr. Drug Math. 7:15-21, 2006; or Federici, Biologicals 41 :13 1-447, 2013. The proteins for use in the formulations described herein are preferably essentially pure and essentially homogeneous (i.e., substantially free from contaminating proteins and/or irreversible aggregates thereof).
Preferred mAbs herein include natalizumab RI®), cetuximab (ERBITUX®), bevacizurnab (AVASTIN®), trastuzumab P'1‘1N®), infliximab (REMICADE®), rituximab (RITUXAN®), panitumumab (VECTIBIX®), ofatumumab (ARZERRA®), and biosimilars thereof.
Exemplary high-molecular-weight proteins can include tocilizumab (ACTEMRA®), alemtuzurnab (marketed under several trade names), brodalumab (developed by Amgen, Inc (“Amgen”)), denosumab (PROLIA® and XGEVA®), and biosimilars thereof.
Exemplary molecular targets for antibodies described herein include CD ns, such as CD3, CD4, CD8, CD19, CD20 and CD34; members of the HER receptor family such as the EGF or, HERZ, HER3 or HER4 receptor; cell adhesion molecules, such as LFA—l, M01, p150,95, VLA-4, lCAM-l, VCAM, and (iv/[33 integrin, including either or or 0 subunits thereof (e.g., anti-CD1 1a, D18, or anti—CD1 lb antibodies); growth factors, such as VEGF; lgE; blood group antigens; flk2/flt3 receptor; y (OB) receptor; protein C; PCSKQ; etc.
Antibody eutics Currently on the Market Many n therapeutics currently on the market, ally antibodies as defined herein, are administered via IV ons due to high dosing requirements. Formulations can e one ofthe antibody therapeutics currently on the market or a biosimilar f. Some protein therapeutics currently on the market are not high—molecular—weight, but are still administered via IV infusion because high doses are needed for therapeutic y. In some embodiments, liquid formulations are provided of these low—molecular-weight proteins as defined herein with concentrations to deliver therapeutically effective amounts for SC or IM injections. dy therapeutics currently on the market include belimumab (BENLYSTA®), golimumab NIAR1A®), abciXimab (REOPRO®), the combination of tositumomab and iodine-131 tositurnomab, marketed as BEHAR‘E, alemtuzumab (CAMPATH® ,palivizumab (SYNAGIS®), basiliximab (SIMULECT®), ado-trastuzumab emtansine (KADCYLA®), pertuzumab (PERJETA®), capromab pendetide (PROSTASCJNT KIT® , caclizumab (ZENAPAX®), ibritumomab tiuxetan (ZEVALW®), eeulizumab (SOLIRIS®), ipiiimumab (YERVOY®), muromonab—CD3 (ORTHOCLONE , raxibacumab, nimotuzumab (THERACIM®), brentuximab vedotin (ADCETRIS®), adalimumab (HUMIRA®), mab (SIMPONI®), palivizumab (SYNAGIS®), ornalizumab (XOLAIR®), and ustekinumab (STELARA®).
Natalizumab, a humanized mAb against the cell adhesion molecule a4-integrin, is used in the treatment of multiple sclerosis and Crohn's disease.
Previously marketed under the trade name ANTEGREN®, natalizumab is currently eo—marketed as TYSABRI® by Biogen Idec (“Biogen”) and Elan Corp. (“Elan”) TYSABRI® is produced in murine myeloma cells. Each 15 mL dose contains 300 mg natalizumab; 123 mg sodium chloride, USP; 17.0 mg sodium phosphate, monobasic, monohydrate, USP; 7.24 mg sodium phosphate, dibasic, heptahydrate, USP; 3.0 mg polysorbate 80, USP/NF, in water for IV injection, USP at pH 6.1. zumab is lly stered by monthly intravenous (IV) infusions and has been proven effective in treating the symptoms of both multiple sis and Crohn's disease, as well as for preventing relapse, vision loss, cognitive decline, and significantly improving patient’s quality of life.
As used , the term “nataiizumab” es the InAb against the cell adhesion molecule a4-integrin known under the International Nonproprietary Name “NATALIZUMAB” or an antigen binding portion thereof. Natalizumab includes antibodies described in US. Patent No. ,840,299, us. Patent No. 6,033,665, U.S. Patent No. 6,602,503, us.
Patent No. 5,168,062, US Patent No. 5,385,839, and US. Patent No. ,730,978. Natalizumab includes the active agent in products marketed under the trade name TYSABR1® by Biogen Idec and Elan ation or a biosimilar product thereof.
Cetuximab is an epidermal growth factor receptor (EGFR) inhibitor used for the treatment of metastatic colorectal cancer and head and neck cancer. mab is a chimeric (mouse/human) mAb typically given by IV infusion. Cetuximab is marketed for IV use only under the trade name ERBITUX® by Bristol—Myers Squibb Company (North America; ol- Myers Squibb”), Eli Lilly and Company (North America; “Eli Lilly”), and Merck KGaA. ERBITUX® is produced in mammalian (murine myeloma) cell culture. Each single-use, 50me Vial ofERBITUX® contains 100 mg of cetuximab at a concentration of 2 mg/mL and is ated in a preservative— free solution containing 8.48 mg/mL sodium chloride, 1.88 mg/mL sodium phosphate dibasic ydrate, 0.42 mg/mL sodium phosphate monobasic monohydrate, and water for IV Injection, USP.
Cetuximab is indicated for the treatment of patients with epidermai growth factor receptor —expressing, KRAS wild-type atic colorectal cancer (mCRC), in combination With chemotherapy, and as a single agent in patients who have failed oxaliplatin— and ecan—based therapy or who are intolerant to irinotecan. Cetuximab is indicated for the treatment of patients with squamous cell carcinoma of the head and neck in combination with platinum-based herapy for the first-line treatment of recurrent and/or metastatic disease and in ation with radiation therapy for locally advanced disease. Approximately 75% of patients with metastatic colorectal cancer have an EGFR—expressing tumor and are, therefore, considered eligible for treatment with cetuximab or panitumumab, according to FDA guidelines.
As used herein, the term “cetuximab” includes the mAb known under the International Nonproprietary Name “CETUXIMAB” or an antigen binding portion thereof. Cetuximab includes antibodies described in US.
Patent No. 6,217,866. Cetuximab includes the active agent in products marketed under the trade name ERBITUX® and biosimilar ts f.
Biosimilars of ERBITUX® can include those currently being ped by Amgen, AlphaMab Co., Ltd. aMab”), and Actavis plc (“Actavis”).
Bevacizumab, a humanized mAb that ts vascular endothelial growth factor A (VEGF-A), acts as an ngiogenic agent. It is marketed under the trade name AVASTIN® by Genentech, Inc. (“Genentech”) and F.
Hoffinann-La Roche, LTD (“Roche”). It is licensed to treat various cancers, including colorectal, lung, breast de the U.S.A.), glioblastoma (USA. only), kidney and ovarian. AVASTIN® was approved by the FDA in 2004 for use in metastatic colorectal cancer when used with standard chemotherapy treatment (as first-line treatment) and With S-fluorouracil- based therapy for second-«line atic colorectal cancer. In 2006, the FDA approved N® for use in first-line advanced non-squamous non-small cell lung cancer in combination with carboplatin/paclitaxei chemotherapy.
AVASTIN® is given as an IV infusion every three weeks at the dose of either mg/kg or 7.5 mg/kg. The higher dose is usually given with carboplatin— based chemotherapy, whereas the lower dose is given with cisplatin-based chemotherapy. In 2009, the FDA approved AVASTIN® for use in metastatic renal cell carcinoma (a form of kidney ). The FDA also granted rated approval of AVASTIN® for the treatment ofrecurrent glioblastoma multiforme in 2009. Treatment for initial growth is still in phase III clinical trial.
The National Comprehensive Cancer Network (“NCCN”) recommends bevacizurnab as standard first-line treatment in ation with any platinum-based chemotherapy, followed by maintenance bevacizumab until disease progression. The NCCN updated its Clinical Practice ines for Oncology (NCCN Guidelines) for Breast Cancer in 2010 to affirm the recommendation regarding the use of bevacizuinab (AVASTIN®, Genentech/Roche) in the treatment of metastatic breast cancer.
As used herein, the term izumab” es the mAb that inhibits vascular endothelial growth factor A (VEGF-A) known under the International prietary Name/Common Name “BEVACIZUMAB” or an antigen binding portion thereof. Bevacizumab is described in U.S. Patent No. 297. zumab includes the active agent in products marketed under the trade name AVASTIN® and nilar products thereof.
Biosimilars of AVASTIN® can include those currently being developed by Amgen, Actavis, AlphaMab, and Pfizer, Inc (“Pfizer”). Biosimilars of AVASTIN® can include the biosimilar known as BCD-021 produced by Biocad and currently in clinical trials in the U.S.
Trastuzumab is a mAb that eres with the HER2/neu receptor.
Trastuzumab is marketed under the trade name HERCEPTIN® by Genentech, Inc. HERCEPTIN® is produced by a mammalian cell (Chinese r Ovary (CHO)) line. HERCEPTIN® is a sterile, white to pale- yellow, preservative-free lyophilized powder for IV administration. Each HERCEPTIN® vial contains 440 mg trastuzumab, 9.9 mg L—histidine HCl, 6.4 mg L—histidine, 400 mg a,a—trehalose dihydrate, and 1.8 mg polysorbate , USP. Reconstitution with 20 mL water yields a multi-dose solution containing 21 mg/mL zumab. HERCEPTIN® is currently administered via IV infusion as often as weekly and at a dosage ranging from about 2 mg/kg to about 8 mg/kg.
Trastuzumab is mainly used to treat certain breast cancers. The HER2 gene is amplified in 20-30% of stage breast cancers, which makes it overexpress epidermal growth factor '(EGF) receptors in the cell membrane.
Trastuzumab is generally administered as a maintenance therapy for patients with HERZ-positive breast cancer, typically for one year post~chemotherapy.
Trastuzumab is currently administered via IV infusion as often as weekly and at a dosage g from about 2 mg/kg to about 8 mg/kg.
As used herein, the term “trastuzumab” includes the mAb that interferes with the HERZ/neu receptor known under the International Nonproprietary Name/Common Name “TRASTUZUMAB” or an antigen binding portion thereof. Trastuzumab is described in U.S. Patent No. ,821,3 37. zumab includes the active agent in products ed under the trade name HERCEPTIN® and biosimilars thereof. The term “trastuzumab” es the active agent in biosimilar HERCEPTIN‘1D products marketed under the trade names HERTRAZ® by Mylan, Inc.
(“Mylan”) and CANMAB® by Biocon, Ltd. (“Biocon”). Trastuzurnab can include the active agent in biosimilar HERCEPTIN® products being developed by Amgen and by PlantForm Corporation, Canada.
Infliximab is a mAb against tumor necrosis factor alpha L) used to treat autoimmune diseases. It is marketed under the trade name REMICADE® by Janssen Global Services, LLC (“Janssen”) in the U.S., Mitsubishi Tanabe Pharma in Japan, Xian Janssen in China, and Merck & Co (“Merck”); elsewhere. Infliximab is a chimeric mouse/human monoclonal antibody with a high molecular weight of approximately 144 kDa. In some embodiments, the formulations contain a biosimilar ofREMICADE®, such as REMSIMATM or INFLECTRATM. Both REMSIMATM, developed by Celltrion, Inc. (“Celltrion”), and INFLECTRATM, developed by Hospira Inc, UK, have been recommended for regulatory approval in Europe. Celltrion has submitted a filing for REMSIMATM to the FDA. Infliximab is tly administered Via IV on at doses g from about 3 mg/kg to about mg/kg. lnfliximab contains approximately 30% murine variable region amino acid sequence, Which confers antigen-binding specificity to human TNFOL The remaining 70% correspond to a human IgG1 heavy chain constant region and a human kappa light chain constant region. rnab has high for human TNFu, which is a cytokine with multiple biologic actions . affinity including ion of inflammatory ses and modulation of the immune system.
Infliximab is a recombinant antibody generally produced and secreted from mouse a cells (SP2/O cells). The antibody is currently manufactured by continuous perfusion cell culture. The infliximab monoclonal antibody is expressed using ic antibody genes consisting ofthe variable region sequences cloned from the murine anti-TNFa oma A2, and human antibody constant region sequences supplied by the plasmid sion vectors. Generation ofthe murine anti-TNF or hybridoma is performed by immunization of BALB/c mice with purified , recombinant human TNFOL. The heavy and light chain vector ucts are linearized and transfected into the Sp2/0 cells by electroporation. Standard purification steps can include chromatographic purification, viral inactivation, nanofiltration, and ultrafiltration/diafiltration.
As used herein, the term “infliximab” es the chimeric mouse/human monoclonal dy known under the International Nonproprietary Name “INFLIXIMAB” or an antigen binding portion thereof. Infliximab neutralizes the biological ty of TNFa by binding with high y to the soluble and transmembrane forms ofTNFu and inhibits binding of TNFG with its receptors. Inflixirnab is described in US.
Patent No. 5,698,195. The term “Infliximab” includes the active agent in products marketed or proposed to be marketed under the trade names REMICADE® by multiple entities; REMSIMATM by Celltrion and INFLECTRATM by Hospira, Inc (“Hospira”). Infliximab is ed as a sterile lyophilized cake for reconstitution and dilution. Each vial of infliximab contains 100 mg infliximab and excipients such as monobasic sodium phosphate monohydrate, c sodium phosphate dihydrate, I _ sucrose, and polysorbate 80.
Denosumab A® and XGEVA®) is a human mAb - and the first RANKL inhibitor - approved for use in nopausal women with risk of osteoporosis and patients with bone metastases from solid .
Denosumab is in Phase II trials for the treatment of rheumatoid arthritis.
Panitumumab is a fully human mAb approved by the FDA for ent of EGFR-expressing metastatic cancer with disease progression.
Panitumumab is marketed under the trade name VECTIBIX® by Amgen.
VECTIBIX® is packaged as a 20 mg/ml panitumumab concentrate in 5 ml, ml, and 15 ml vials for IV infusion. When prepared according to the packaging instructions, the final panitumumab concentration does not exceed mg/ml. VECTIBIX® is stered at a dosage of 6 mg/kg every 14 days as an intravenous infilsion. As used herein, the term “panitumumab” includes the anti—human epidermal growth factor receptor known by the International Nonproprietary Name “PANITUMUMAB.” The term “panitumuma ” includes the active agent in ts marketed under the trade name VECTIBIX® by Amgen and biosimilars thereof. The term umumab” includes onal antibodies described in US. Patent No. 6,235,883. The term “panitumuma ” includes the active agent in biosimilar IX® products, including biosimilar IX® being developed by BioXpress, SA (“BioXpress”). mab (BENLYSTA®) is a human mAb with a molecular weight of about 151.8 kDa that inhibits B—cell activating factor (BAFF). Belimumab is approved in the United States, Canada, and Europe for treatment of systemic lupus erythematosus. Belimumab is currently administered to lupus patients by IV infusion at a 10 mg/kg dosage. A high-molecular-weight, low- ity protein formulation can e Belimurnab, preferably in a concentration of about 400 mg/mL to about 1,000 mg/mL. The preferred ranges are calculated based upon body weight of 40-100 kg (approximately 80-220 lbs) in a 1 mL volume.
Abciximab O®) is manufactured by Janssen Biologics BV and distributed by Eli Lilly & Company (“Eli Lilly”). Abciximab is a Fab fragment ofthe chimeric human-murine monoclonal antibody 7E3.
Abciximab binds to the rotein (GP) IIb/IIIa receptor of human platelets and inhibits platelet aggregation by ting the binding of fibrinogen, von Willebrand factor, and other adhesive molecules. It also binds to vitronectin (avB3) receptor found on platelets and vessel wall endothelial and smooth muscle cells. Abciximab is a platelet aggregation inhibitor mainly used during and after coronary artery procedures.
Abciximab is administered via IV infusion, first in a bolus of 0.25 mg/kg and followed by uous IV infusion of 0.125 meg/kg/minute for 12 hours.
Tositumomab (BEXXAR®) is a drug for the treatment of follicular lymphoma. It is an IgG2a anti-CD20 mAb derived from immortalized mouse cells. Tositumomab is administered in sequential infusions: cold mAb followed by iodine (1311) tositumomab, the same antibody covalently bound to the radionuclide iodine-131. Clinical trials have established the efficacy of the tositumomab/iodine tositumomab regimen in patients with relapsed refractory follicular lymphoma. BEXXAR® is currently administered at a dose of 450 mg via IV infusion.
Alemtuzumab (marketed as CAMPATH®, MABCAMPATH®, or H-IH® and currently under further development as LEMTRADA®) is a mAb used in the ent of chronic lymphocytic leukemia (CLL), cutaneous T—cell lymphoma (CTCL), and T—cell ma.
It is also used under clinical trial protocols for treatment of some autoimmune es, such as multiple sclerosis. zumab has a weight of approximately 145.5 kDa. It is administered in daily IV infusions of 30 mg for patients with B—cell chronic lymphocytic leukemia.
Palivizumab (SYNAGIS®) is a humanized mAb ed t an e in the A antigenic site of the F protein of respiratory syncytial virus. in two Phase III clinical trials in the pediatric popuiation, palivizumab reduced the risk of hospitalization due to respiratory syncytial virus infection by 55% and 45%. Palivizumab is closed once a month via IM injection of 15 mg/kg.
Ofatumumab is a human anti-CD20 mAb which appears to inhibit early~stage B lymphocyte activation. Ofaturnumab is marketed under the trade name ARZERRA® by GlaxoSmithKline, plc (“GlaxoSmithKline”).
ARZERRA® is distributed in single-use vials containing 100 mg/5 mL and 1,000 mg/SO mL ofatumumab for IV infusion. Ofatumumab is FDA- approved for treating chronic lymphocytic leukemia and has also shown potential in treating Follicular non-Hodgkin’s lymphoma, Diffuse large B cell lymphoma, rheumatoid tis, and relapsing remitting multiple sclerosis. Ofatumumab has a molecular weight of about 149 kDa. It is currently administered by IV infusion at an initial dose of 300 mg, followed by weekly infusions of 2,000 mg. AS used herein, the term “ofatumurnab” includes the anti-CD20 mAb known by the International Nonproprietary Name “OFATUMUMAB.” The term “ofatumumab” includes the active agent in ts marketed under the trade name ARZERRA® and biosimilars thereof. The term “ofatumumab” includes the active agent in biosimilar ARZERRA® products being developed by BioExpress. High- molecularuweight, low-viscosity liquid protein formulations can e ofatumumab, preferably in a concentration of about 300 mg/mL to about 2,000 mg/mL.
Trastuzumab ine (in the U.S., ado-trastuzumab emtansine, marketed as A®) is an antibody-drug ate consisting of the mAb trastuzumab linked to the cytotoxic agent mertansine (DM1®).
Trastuzumab, described above, stops growth of cancer cells by binding to the HERZ/neu receptor, Whereas rnertansine enters cells and ys them by binding to tubulin. In the United , trastuzumab emtansine was approved specifically for treatment of recurring HERZ-positive metastatic breast cancer. Multiple Phase III trials of trastuzumab emtansine are planned or ongoing in 2014. zumab emtansine is currently administered by IV infusion of 3.6 rug/kg. High—molecular—weight, low-viscosity liquid formulations can include trastuzumab emtansine, ably in a concentration of about 144 mg/mL to about 360 mg/mL.
Pertuzumab (PERJETA®) is a mAb that inhibits HER2 dimerization.
Pertuzumab received FDA approval for the treatment of HEM-positive metastatic breast cancer in 2012. The currently recommended dosage of umab is 420 mg to 840 mg by IV infusion. High-molecular-weight, low-viscosity liquid formulations can include pertuzumab, preferably in a concentration of about 420 mg/rnL to about 840 mg/mL.
Daclizumab is a humanized anti-CD25 mAb and is used to t rejection in organ transplantation, especially in kidney transplants. The drug is also under investigation for the ent of multiple sclerosis.
Daclizumab has a molecular weight of about 143 kDa. Daclizumab was marketed in the US. by Hoffmann—La Roche, Ltd. (“Roche”) as ZENAPAX® and administered by IV infusion of 1 rug/kg. Daclizumab High- Yield Process (DAC HYP; BIIBOl9; Biogen Idec (“‘Biogen”) and Abeie, Inc. e”)) is in phase III clinical trials as a 150 mg, once-monthly aneous injection to treat relapsing, remitting multiple-sclerosis. High- molecular-weight, low-viscosity liquid formulations. can include daclizumab, preferably in a concentration of about 40 mg/mL to about 300 mg/mL.
Eculizumab (SOLIRIS®) is a humanized mAb ed for the treatment of rare blood diseases, such as paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Eculizumab, with a molecular weight of about 148 kDa, is being developed by n Pharmaceuticals, Inc (“Alexion”). It is administered by IV infusion in the amount of about 600 mg to about 1,200 mg. High-molecular-weight, low- viscosity liquid formulations can e eculizumab, preferably in a concentration of about 500 mg/mL to about 1,200 mg/mL.
Tocilizumab (ACTEMRA®) is a humanized mAb against the interleukin-6 receptor. It is an immunosuppressive drug, mainly for the treatment of rheumatoid arthritis (RA) and systemic juvenile idiopathic arthritis, a severe form ofRA in children. Tocilizumab is commonly stered by IVinfusion in doses of about 6 mg/kg to about 8 mg/kg.
Highnmolecular-weight, low-viscosity liquid formulations can include tocilizumab, preferably in a concentration of about 240 mg/mL to about 800 mg/mL.
Rituximab (RITUXAN®) is a chimeric anti-CD20 mAb used to treat a y of diseases characterized by excessive numbers ofB cells, overactive B cells, or dysfunctional B cells. Rituxirnab is used to treat cancers of the White blood system, such as leukemias and lymphomas, including Hodgkin's ma and its lymphocyte-predominant subtype. It has been shown to be an effective toid arthritis treatment. Rituximab is Widely used off- label to treat difficult cases of multiple sclerosis, systemic lupus erythematosus, and autoimmune anemias.
Rituximab is jointly marketed in the US. under the trade name RITUXAN® by Biogen and Genentech and outside the US. under the trade name RA® by Roche. RITUXAN® is distributed in single-use vials containing 100 rug/10 mL and 500 mg/50 mL. RITUXAN® is typically administered by IV infusion of about 375 mg/mz. The term “rituximab,” as used herein, includes the anti—CD20 InAb known under the International Nonproprietary Name/Common Name IMAB.” Rituximab includes mAbs described in US. Patent No. 5,73 6,137. Rituximab includes the active agent in products marketed under the trade name N® and MABTHERA® and biosimilars thereof.
High-molecular-weight, low—viscosity liquid ations can include rituximab, ably in a concentration of about 475 mg/mL to about 875 rn mL (approximated using a body e area range of 1.3 to 2.3 square meters, derived from the Mosteller formula for persons ranging from 5 ft, 40 kg to 6 ft, 100 kg). Concentrations are calculated for a 1 mL formulation.
Ipilimumab is a human mAb developed by Bristol—Myers Squibb y (“Bristol-Myers Squibb”). Marketed as YERVOY®, it is used for the treatment of ma and is also undergoing clinical trials for the treatment of non-small cell lung carcinoma (NSCLC), small cell lung cancer (SCLC), and metastatic hormone—refractory prostate cancer. Ipilimurnab is currently administered by IV infusion of 3 mg/kg. High—molecular—weight, scosity liquid formulations can include ipilimumab, preferably in a concentration of about 120 mg/mL to about 300 mg/mL.
Raxibacumab (ABthrax®) is a human mAb intended for the prophylaxis and treatment of inhaled x. It is currently stered by IV infusion. The suggested dosage in adults and children over 50 kg is 40 mg/kg. High-molecularvweight, low-viscosity liquid formulations can include cumab, ably in a concentration of about 1,000 mg/mL to about 4,000 Ing/mL.
Nimotuzumab ‘(THERACIM®, BIOMAB EGFR®, THERALOC®, CIMAher®) is a humanized mAb with a molecular weight of about 151 kDa used to treat squamous cell carcinomas of the head and neck, recurrent or refractory high~grade malignant glioma, anaplastic astrocytomas, glioblastomas, and e intrinsic pontine glioma. zumab is typically administered by IV infusion of about 200 mg weekly. High~ lar-weight, low-viscosity liquid formulations can include nimotuzumab, preferably in a concentration of about 200 mg/mL.
Brentuximab vedotin (ADCETRIS®) is an antibody-drug conjugate directed to the protein CD30, expressed in classical Hodgkin’s lymphoma and systemic anaplastic large cell lymphoma. It is administered by IV on of about 1.8 mg/kg. High-moleculanweight, low-viscosity liquid formulations can include brentuximab n, preferably in a concentration of about 80 mg/mL to about 200 mg/mL.
Itolizumab (ALZUMAB®) is a humanized IgGl mAb developed by Biocon. Itolizumab completed successful Phase III studies in patients with moderate to severe psoriasis. Itolizumab has received marketing approval in India; an application for FDA approval has not been submitted.
Obinutuzumab (GAZYVA®), originally developed by Roche and being further developed under a collaboration agreement with Biogen is a humanized anti-CD20 mAb approved for treatment of chronic lymphocytic leukemia. It is also being investigated in Phase III clinical trials for patients with s lymphomas. Dosages of about 1,000 mg are being administered via IV infilsion.
Certolizumab pegol (CIMZIA®) is a recombinant, humanized antibody Fab' fragment, with specificity for human tumor necrosis factor alpha (TNFa), conjugated to an approximately 40kDa polyethylene glycol (PEGZMAL40K). The molecular weight of certolizumab pegol is approximately 91 kDa.
Other antibody therapeutics that can be formulated with viscosity- lowering agents e CT—P6 from Celltrion, Inc. (Celltrion).
Antibody Therapeutics in Late-Stage Trials and Development The progression of antibody therapeutics to late-stage clinical development and regulatory review are proceeding at a rapid pace. In 2014, there are more than 300 mAbs in clinical trials and 30 commercially- sponsored antibody therapeutics undergoing evaluation in late-stage studies.
First marketing applications for two mAbs (vedolizumab and ramucinnnab) were recently submitted to the FDA. Arngen is currently sponsoring multiple ongoing Phase III trials on the use of brodalumab in patients with plaque psoriasis, with additional trials planned or recruiting patients. ch, Inc. has sponsored two Phase I clinical trials of MABpl (Xilonix) for patients with ed cancer or type—2 diabetes. Additional trials of MABpl are recruiting ts. le trials are sponsored by MedImmune, LLC (“Medlmrnune”) and underway or recruiting patients for the treatment of leukemia with moxetumomab pasudotox. Long-term safety and efficacy studies are underway for the use oftildrakizumab for the treatment of chronic plaque psoriasis. Multiple phase II trials have recently completed for the use of rilotumumab for the treatment of various cancers.
At least 28 mAbs are high-molecular-weight proteins currently in or having recently completed Phase III studies for the treatment of inflammatory or immunological disorders, cancers, high cholesterol, orosis, Alzheimer’s disease, and infectious diseases. The mAbs in or having recently completed Phase III trials e AMG 145, elotuzumab, zumab, farletuzumab (MORAb-OOB), erumab (RG1450), gevokizumab, inotuzumab ozogamicin, umab, ixekizumab, izuma’o, mepolizumab, omab natox, necitumuma‘o, mab, ocrelizumab, onartuzumab, racotumornab, ramucirurnab, umab, romosozumab, sariiumab, numab, mab, solanezumab, tabalumab, and vedoiizumab. A mAb mixture (actoxumab and bezlotoxumab) is also being evaluated in Phase III trials. See, e.g., Reichert, MES 5:1-4, 2013.
Vedolizurnab is a mAb being developed by Millennium Pharmaceuticals, Inc (“Miiiennium”; a subsidiary of Takeda Pharmaceuticals Company, Ltd. (“Takeda”)). Vedolizumab was found safe and highly effective for inducing and ining clinical remission in patients with moderate to severe tive colitis. Phase III clinical trials showed it to meet the objectives of inducing a clinical response and maintaining remission in Crohn's and ulcerative colitis patients. Studies evaluating long- term ciinicai es show close to 60% of patients achieving clinical remission. A common dose ofvedolizumab are 6 mg/kg by IV on.
Ramucirumab is a human mAb being developed for the treatment of umors. Phase III clinical trials are g for the treatment of breast cancer, metastatic gastric adenocarcinoma, non-small cell lung cancer, and other types of cancer. rumab, in some Phase III trials, is administered at about 8 mg/kg via IV infusion.
Rilotumumab is a human mAb that inhibits the action of hepatocyte growth factor/scatter factor. Developed by Amgen, it is in Phase III trials as a treatment for solid tumors. An open Phase III study of rilotumumab treatment in patients with advanced or metastatic esophageal cancer will administer mumab at about 15 mg/kg Via IV infusion.
Evolocumab (AMG 145), also developed by Amgen, is a mAb that binds to PCSK9. Evolocumab is indicated for hypercholesteroiemia and hyperlipidemia.
Alirocumab (REGN727) is a human mAb from Regeneron Pharmaceuticals, Inc. (“Regeneron”) and Sanofi—Aventis U.S. LLC (“Sanofi”), indicated for hypercholesteroiemia and acute coronary syndrome.
Naptumomab estafenatox, AER-217620 from Active Biotech AB (“Active Biotech”) is a mAb ted for renal cell carcinoma.
Racotumomab from CIMAB, SA (“CIMAB”); Laboratorio Elea S.A.C.I.F.y A. is a mAb indicated for non-smali cell lung cancer.
Other antibodies which may be formulated with viscosity-lowering agents include bocociznmab (PF-04950615) and tanezumab; ganitumab, blinatumomab, trebananib from Amgen; x immune globulin from Cangene Corporation; teplizurnab from MacroGenics, Inc.; MK-3222, MK- 6072 from Merck & Co (“Merck”); girentuximab from Wilex AG; RIGScan from Navidea nnaceuticals (“Navidea”); PF-05280014 from Pfizer; SA237 from Chugai Pharmaceutical Co. Ltd. ("Chugai"); guselkumab from Janssen/ Johnson and Johnson Services, Inc. (“J&J”); Antithrombin Gamma (KW-3357) from Kyowa; and CT-PlO from Ceiltrion.
Antibodies ianarlySrage al Trials Many mAbs have recently entered, or are entering, clinical trials.
They can include proteins currently stered Via IV infilsion, preferably those having a molecular weight greater than about 120 kDa, typically fiom about 140 kDa to about 180 kDa. They can also include such high- molecular—weight proteins such as Albumin-conjugated drugs or peptides that are also entering clinical trials or have been approved by the FDA.
Many mAbs from Amgen are currently in clinical trials. These can be high- molecular—weight proteins, for example, AMG 557, which is a human monoclonal antibody ped jointly by Arngen and AstraZeneca and currently in Phase I trials for treatment of lupus. Likewise, AMG 729 is a humanized mAb developed by Amgen and currently in Phase I trials for the treatment of lupus and rheumatoid tis. In addition, AMG 110 is a mAb for epithelial cell adhesion le; AMG 157, jointly developed by Amgen and AstraZeneca, is a human mAb currently in Phase I for the treatment of asthma; AMG 167 is a humanized mAb that has been evaluated in multiple Phase I trials for the treatment of osteopenia; AMG 334, having ted Phase I dosing studies and currently in in Phase II studies for the treatment of migraines and hot , is a human mAb that inhibits Calcitonin Gene-Related Peptide; AMG 780 is a human anti-angiopoietin mAb that inhibits the interaction between the endothelial cell-selective TieZ or and its ligands Angl and AngZ, and recently completed Phase I trials as a cancer treatment; AMG 811 is a human monoclonal antibody that ts interferon gamma being investigated as a treatment for systemic lupus erythematosus; AMG 820 is a human mAb that inhibits c~fms and decreases tumor associated macrophage (TAM) function and is being investigated as a cancer treatment; AMG 181, jointly developed by Amgen and AstraZeneca, is a human mAb that inhibits the action of alpha4fbeta7 and is in Phase II trials as a treatment for tive colitis and Crohn's disease.
Many mAbs are currently in clinical trials for the treatment of autoimmune disorders. These mAbs can be included in low-Viscosity, high- molecular-weight liquid formulations. RG7624 is a fully human mAb ed to specifically and selectively bind to the human interleukin-17 family of nes. A Phase I clinical trial evaluating RG7624 for autoimmune disease is ongoing. BIIB033 is an INGO-l mAb by Biogen currently in Phase II trials for treatingmultiple sclerosis.
High-molecular—weight proteins also can include AGS-009, a mAb targeting pha developed by Argos Therapeutics, Inc. that recently completed phase I trials for the treatment of lupus. Patients are administered up to 30 mg/kg ofAGS—009 via IV infilsion. BT-06l , developed by Abeie, is in PhaSe II trials for patients with rheumatoid arthritis. Certolizumab pegol (CIMZIA®) is a mAb in Phase II trials for ankylosing spondylitis and juvenile toid arthritis. Clazakizumab, an anti-1L6 mAb, is in Phase II trials by Bristol—Myers Squibb.
(INTO-I36 (sirukumab) and CNTO~1959 are mABs having ly completed Phase II and Phase III trials by Janssen. Daclizumab (previously ed as X® by Roche) is currently in or has recently completed multiple Phase III trials by Abeie for the treatment of multiple sclerosis.
Epratuzumab is a zed mAb in Phase III trials for the ent of lupus. Canakinurnab (ILARIS®) is a human mAb targeted at interleukin-1 beta. It was approved for the treatment of cryopyrin—associated periodic syndromes. Canakinurnab is in Phase I trials as a possible treatment for chronic obstructive puimonary disease, gout and coronary artery disease.
Mavriiimumab is a human mAb designed for the treatment of rheumatoid arthritis. Discovered as CAM—3001 by Cambridge Antibody Technology, mavrilimumab is being developed by Medlmmune.
MEDI—546 are 70 are mAbs currently in Phase I and Phase II trials by AstraZeneca for the ent of lupus. MEDI—546 is administered in the Phase II study by regular IV infusions of 300~l,000 mg. MEDI—SS 1, another mAb being developed by AstraZeneca for numerous indications, is also currently administered by IV infusion. NN8209, a mAb blocking the C5aR receptor being developed by Novo Nordisk A/S( “Novo Nordisk”), has completed a Phase II dosing study for treatment of rheumatoid arthritis.
NN8210 is another antiCSaR mAb being developed by Novo k and currently is in Phase I trials. IPH2201 (NN8765) is a humanized mAb targeting NKGZA being developed by Novo Nordisk to treat patients with inflammatory conditions and autoimmune diseases. NN8765 ly completed Phase I trials.
Olokizumab is a humanized mAb that potently targets the cytokine IL-6. IL—6 is involved in several autoimmune and atory pathways.
Olokizumab has completed Phase II trials for the treatment of rheumatoid arthritis. Otelixizumab, also known as TRX4, is a mAb, which is being developed for the treatment oftype 1 diabetes, rheumatoid arthritis, and other autoimmune diseases. Ozoralizumab is a humanized mAb that has completed Phase II trials.
Pfizer currently has Phase Itrials for the mAbs PD-3 60324 and PF— 04236921 for the treatment of lupus. A rituximab biosimilar, 805 86, has been developed by Pfizer and is in Phase e II trials for rheumatoid arthritis.
Rontalizumab is a humanized InAb being developed by Genentech. It recently completed Phase II trials for the treatment of lupus. 244 (anti-CXCRS) is a mAb by Sanofi in Phase I trials. Sifalimumab (anti—IFNn alpha mAb) is a mAb in Phase II trials for the treatment of lupus.
A high-moiecular—weight low-viscosity liquid formulation can include one of the mAbs in early stage clinical development for treating various blood disorders. For example, Belirnumab (BENLYSTA®) has recently ted Phase I trials for patients with itis. Other mAbs in early-stage trials for blood disorders include 075 from nger Ingelheim GmbH inger Ingelheim”, ferroportin mAb and hepcidin WO 38818 mAb from Eli Lily, and SelGl from Selexys Pharmaceuticals, Corp. xys”).
One or more mAbs in early-stage development for treating various cancers and related ions can be included in a scosity, high- molecular-weight liquid formulation. United Therapeutics, Corporation has two mAbs in Phase I , 8H9 mAb and ch14.18 mAb. The mAbs ABT- 806, enavatuzurnab, and volociXimab from Abeie are in early-stage development. Actinium Pharmaceuticals, Inc has conducted early-stage trials for the mAbs ActimabuA (M195 mAb), anti-CD45 mAb, and Iomab—B.
Seattle Genetics, Inc. (“Seattle cs”) has several InAbs in early—stage trials for cancer and related conditions, including anti-CD22 ADC (RG7593; pinatuzumab vedotin), anti-CD79b ADC (RG7596), anti-STEAPI ADC (RG7450), ASG—SME and ASG-ZZME from Agensys, Inc. (“Agensys”) the antibody-drug conjugate RG7458, and vorsetuzumab mafodotin. The early- stage cancer therapeutics from Genentech can be included in low-viscosity formulations, including ALT-83 6, the dy-drug conjugates RG7600 and DEDN6526A, anti-CD22 ADC (RG7593), anti-EGFL7 InAb (RG7414), anti-HER3/EGFR DAF mAb (RG7597), anti—PD~L1 mAb (RG7446), DFRF4539A, an MINT1526A. l-Myers Squibb is developing early- stage mAbs for cancer therapeutics, including those identified as anti- CXCR4, anti-PD-Ll, IL-21 (EMS—982470), lirilumab, and urelumab (anti- CD137). Other mAbs in early-stage trials as cancer therapeutics include (hu14.18-IL2) from Apeiron Biologics AG, AV-203 from AVEO ceuticals, Inc. (“AVEO”), AVX701 and AVX901 from AlphaVaX, BAX-69 from Baxter International, Inc. (“Baxter”), BAY 79-4620 and BAY -10112 from Bayer HealthCare AG, BHQ880 from Novartis AG, 212-Pb- TCMCtrastuzumab from AREVA Med, AbGn-7 from AbGenomics International Inc, and ABIO-OSOI (TALL-104) from Abiogen Phanna S.p.A.
Other antibody therapeutics that can be formulated with viscosity- lowering agents include b, GA101, daratumumab, imab, ALX- 0061, ALX—0962, ALX-0761, bimagumab (BYM338), CT-Oll (pidilizurnab), ,actoxumab/bezlotoxumab (MK-3515A), MIC-3475 (pembrolizumab), dalotuzumab (MK-0646), mab (IMC-18F1, LY3012212), AMG 139 (MED12070), SAR339658, dupilumab (REGN668), SAR156597, SAR256212, 356, SAR3419, SAR153192 (REGN421, enoticumab), SAR307746 (nesvacumab), SAR650984, SAR566658, SAR391786, SAR228810, SAR252067, SGN—CDIQA, SGN-CD33A, SGN— LIVIA, ASG 15MB, Anti-LINGO, BIIB037, ALXN1007, teprotumumab, concizumab, anrukinzumab (IMA—63 8), ponezumab (PF-04360365), PF— 03446962, PF-06252616, etrolizumab (RG7413), quilizumab, ranibizumab, lampalizumab, onclacumab, gentenerumab, umab (RG7412), IMC— RONS (narnatumab), tremelimumab, vantictumab, eemcizumab, ozanezumab, mumab, tralokinumab, XmAbS871, XmAb7195, cixutumumab (LY3 012217), LY2541546 (blosozumab), olaratmnab (LY3012207), MEDI4893, MEDIS73, MEDIO639, MEDI3617, MEDI473 6, MEDI6469, 80, MED15872, PF-05236812 03), PF- 05082566, BI 1034020, RG7116, RG7356, RG7155, RG7212, RG7599, RG7636, , RG7652 (MPSK3169A), RG7686, HuMaxTFADC, MOR103, BT061, MORZOS, OMP59R5 (anti-notch 2/3), VAY736, MOR202, BAY94-9343, LJM716, OMP52M51, GSK933776, GSK249320, GSK1070806, NN8828, CEP-37250/KHK2804 M8F, AGS-16C3F, LY3016859, 655, LY28753 58, and LY2812176.
Other early stage InAbs that can be formulated with Viscosity- lowering agents include benralizumab, 968, anifrolumab, MEDI7183, sifalimumab, MEDI—575, tralokinumab from AstraZeneca and Medlmmune; BAN2401 from Biogen Idec/Eisai Co. LTD ("Eisai")/ BioArctic cience AB; CDP7657 an anti~CD40L monovalent pegylated Fab antibody fragment, STX-IOO an anti-avB6 mAb, BIIBOS 9, Anti-TWEAK (BHB023), and BIIB022 from Biogen; umab from Janssen and Amgen; BI—204/RG741 8 from BioInvent International/Genentech; BT-062 (indatuximab ravtansine) from Biotest ceuticals Corporation; XInAb from Boehringer eim/Xencor; anti-11310 from Bristol—Myers Squibb; J 591 LII-177 from BZL Biologics LLC; CDX-Oll (glembatumumab vedotin), CDX~0401 from Celldex Therapeutics; foravirumab from Cmceli; tigatuzumab from Daiichi Sankyo Company Limited; 004, MORAb-009 (amatuximab) from Eisai; LY23 82770 from Eli Lilly; DIl7E6 from EMD Serono Inc; zanolimumab from Emergent BioSolutions, 1110.; FG—3019 from FibroGen, 1nc.; catumaxomab from Fresenius SE & Co. KGaA; pateclizumab, rontalizumab from Genentech; fresolimumab from Genzyme & Sanofi; GS-6624 (simtuzumab) from Gilead; 28; bapineuzumab (AAB-OOI), ab, 36 from n; K3003 from KaloBios Pharmaceuticals, Inc.; ASKP1240 from Kyowa; RN—307 from Labrys Biologics 1n0.; ecromeximab from Life Science Pharmaceuticals; LY249565S, LY292805 7, LY3015014, LY2951742 from Eli Lilly; MBL-HCVl from 'MassBiologics; AME-133V fiom MENTRIK Biotech, LLC; abituzumab from Merck KGaA; MM~121 from Merrimack Pharmaceuticals, 1110.; MCSl 10; QAX576, QBX258, QGE031 from Novartis AG; HCD122 from Novartis AG and XOMA Corporation (“XOMA”); NN8555 from Novo Nordisk; bavituximab, cotara from Peregrine Pharmaceuticals, 1110.; PSMA~ADC from Progenics Pharmaceuticals, 1110.; omab from Quest Pharmatech, 1110.; fasinumab (REGN475), REGN1033, 893, REGN846 from Regeneron; RG7160, CIM331, RG7745 from Roche; ibalizumab (TMB-355) from TaiMed Biologics 1110.; TCN-032 from Theraclone Sciences; TRClOS from TRACON Pharmaceuticals, Inc.; UB—421 from United Biomedical Inc; from ia Bio, 1110.; ART-110 from Abeie; Caplaciiumab, Ozoralizumab from Ablynx; PRO 140 from CytoDyn, Inc.; GS-CDAI, MDX-1388 from Medarex, 1110.; AMG 827, AMG 888 from Amgen; ublituximab from TG Therapeutics 1110.; TOLlOl from Tolera Therapeutics, Inc.; —DM1 (lorvotuzuma‘o mertansine) from ImmunoGen 1nc.; epratuzumab Y-90/veltuzumab combination (1MMU-102)from Immunomedics, 1110.; anti-fibrin mAb/ 3B6/22 T0-99m from Agenix, Limited; ALD403 from Alder Biopharmaceuticals, 1110.; RN6G/ PF- 23 from Pfizer; CG201 from CG Therapeutics, Inc. ; KBOOl-A from KaloBios Pharmaceuticals/Sanofi; KRN-23 fiom Kyowa.; Y—90 hPAM 4 from Immunomedics,lnc.; Tarextumab from Morphosys AG & OncoMed Pharmacetuicals, 1110.; LFG316 from Morphosys AG & Novartis AG; CNTO3157, CNT06785 from Morphosys AG & Jannsen; RG6013 from Roche & Chugai; MM—l 1 1 from Merrimack Pharmaceuticals, Inc.
(“Merrimack”); GSK2862277 from GlaxoSmithKline; AMG 282, AMG 172, AMG 595, AMG 745, AMG 761 from Amgen; BVX—20 from Biocon; CT- P19, CT-P24, CT-PZS, CT-P26, , CT—P4 from Celltrion; GSK284933, GSK2398852, GSK2618960, GSK1223249, GSK933776A from GlaxoSmithKline; anetumab ravtansine from sys AG & Bayer AG; BI-836845 from Morphosys AG & Boehringer Ingelheim; NOV-7, NOV— 8 from Morphosys AG & Novartis AG; MM—302, MM—3 10, MM—141, MM-131, MM~151 from Merrimack, RG7882 from Roche & e Genetics; RG7841 from Roche/ Genentech; PF-06410293, PF—0643 8179, PF-0643953 5, PF-04605412, PF-05280586 from Pfizer; RG7716, RG793 6, gentenerumab, RG7444 from Roche; 47, 65, MED11814, MEDI4920, MED18897, MEDI-4212, MEDI-5117, MEDI—7814 from Astrazeneca; ulocuplurnab, PCSK9 adnectin froin Bristol-Myers Squibb; FPAOOQ, FPA145 from FivePrime Therapeutics, Inc.; GS—5745 from Gilead; BIW—8962, 3, KHK6640 from Kyowa Hakko Kirin; MM-141 from Merck KGaA; REGN1154, REGN1193, REGN1400, REGN 1 500, REGN1908-1909, REGN2009, REGN2176-3, REGN728 from Regeneron; SAR307746 from Sanofi; SGN—CD70A from Seattle Genetics; ALX—0141, ALX—Ol 71 from Ablynx; milatuzumab-DOX, milatuzhmab, TF2, from Immunornedics, Inc.; MLN0264 from Millennium; ABT—981from Abeie; AbGn-l68H from AbGenomics International Inc.; ficlatuzumab from AVEO; BI—505 from Biolnvent ational; CDX-l 127, CDX-301 from Celldex Therapeutics; CLT-008 from Cellerant Therapeutics Inc.; VGX~100 from Circadian; U3-1565 from Daiichi Sankyo Company d; DKN—Ol from Dekkun Corp.; flanvotumab (TYRPI protein), IL—l I?» antibody, IMC- CS4 from Eli Lilly; VEGFR3 mAb, IMC-TRl (LY3022859) from Eli Lilly and ImClone, LLC; Anthim from Elusys Therapeutics Inc.; HuL2G7 from Galaxy Biotech LLC; IMGB853, IMGN529 from ImmunoGen Inc.; CNTO- , ONTO—5825 from n; KD-247 from Kaketsuken; K3004 from KaloBios Pharmaceuticals; MGA27I, MGAH22 from MacroGenics, Inc.; XmAbSS74 from MorphoSys AG/Xencor; ensituximab (NPC—l C) from ix Oncology, Inc.; LFA102 from is AG and XOMA; ATI355 from Novartis AG; SAN-300 from Santarus Inc.; SelGl from Selexys; /rGel from Targa Therapeutics, Corp. ; VX15 from Teva 2014/055254 Pharmaceuticals, Industries Ltd. (“Teva”) and Vaccinex Inc.; TCN—202 from Theraclone es; XmAb2513, XmAb5872 from Xencor; XOMA 3AB from XOMA and National Institute for Allergy and Infectious Diseases; neuroblastoma antibody vaccine from MabVax Therapeutics; Cytolin from CytoDyn, Inc; Thravixa from Emergent utions Inc; and FE 301 from Cytovance Biologics; rabies mAb from Janssen and Sanofi; flu mAb from Janssen and partly funded by National Institutes of Health; MB-003 and ZMapp from Mapp Biopharmaceutical, Inc; and ZMAb from s Inc.
Other Protein Therapeutics The n can be an , a fusion protein, a stealth or ted protein, vaccine or otherwise a biologically active protein (or protein e). The term “enzyme,” as used herein, refers to the protein or functional fragment thereof that catalyzes a mical transformation of a target molecule to a desired product. s as drugs have at least two ant features, namely i) often bind and act on their targets with high affinity and specificity, and ii) are catalytic and convert multiple target molecules to the desired products.
In certain embodiments, the protein can be PEGylated, as defined herein.
The term “fusion protein,” as used herein, refers to a protein that is created from two different genes encoding for two separate proteins. Fusion proteins are generally produced through recombinant DNA techniques known to those skilled in the art. Two proteins (or protein fragments) are fused together covalently and exhibit properties from both parent proteins.
There are a number of fusion proteins that are on the market.
ENBREL® (Etanercept), is a fusion protein marketed by Amgen that competitively inhibits TNF.
ELOCTATE®, Antihemophilic Factor (Recombinant), Fc Fusion Protein, is a recombinant DNA derived, mophilic factor indicated in adults and children with Hemophilia A (congenital Factor VIII deficiency) for control and tion of bleeding episodes, perioperative management, routine prophylaxis to prevent or reduce the frequency of bleeding es.
EYLEA® (aflibercept) is a recombinant fusion protein consisting of portions of human VEGF receptors 1 and 2 extracellular domains fused to the Fc portion of human IgG1 formulated as an iso-osmotic solution for intravitreal administration. EYLEA (aflibercept) is a inant fusion protein consisting of portions of human VEGF receptors 1 and 2 extracellular domains fused to the Fc portion of human IgG1 formulated as an iso-osmotic solution for intravitreal administration. Aflibercept is a dimeric glycoprotein with a protein molecular weight of 97 ltons (kDa) and contains glycosylation, constituting an additional 15% of the total molecular mass, resulting in a total molecular weight of 115 kDa. Aflibercept is produced in recombinant Chinese hamster ovary (CHO) cells, marketed by ron.
ALPROLIXTM, Coagulation Factor IX (Recombinant), Fe Fusion Protein, is a inant DNA derived, ation Factor IX concentrate is indicated in adults and children With hemophilia B for control and prevention ofbleeding episodes, perioperative management, routine prophylaxis to prevent or reduce the frequency of bleeding episodes.
Pegloticase (KRYSTEXXA®) is a drug for the treatment of severe, treatment-refractory, c gout, developed by Savient Pharmaceuticals, Inc. and is the first drug approved for this indication. Pegloticase is a pegylated recombinant porcine-like uricase with a molecular weight of about 497 kDa. Pegloticase is currently administered by IV ons of about 8 mg/kg. High—molecular-weight, low-viscosity liquid formulations can include pegloticase, preferably in a concentration of about 300 Ing/mL to about 800 mg/mL.
Alteplase (ACTIVASE®) is a tissue plasminogen tor produced by recombinant DNA technology. It is a purified glycoprotein comprising 527 amino acids and sized using the complementary DNA (cDNA) for natural human tissue-type plasminogen activator obtained from a human melanoma cell line. ase is administered via IV infusion of about 100 mg immediately following ms of a stroke. In some embodiments, low—viscosity formulations are provided containing ase, preferably in a concentration of about 100 mg/mL.
Glucarpidase AZE®) is a FDA—approved drug for the treatment of elevated levels of methotrexate (defined as at least 1 ol/L) during treatment of cancer patients who have impaired kidney function. Glucarpidase is administered via IV in a single dose of about 50 IU/kg. In some embodiments, low-viscosity formulations are provided containing glucarpidase.
Alglucosidase alfa (LUMIZYME®) is an enzyme replacement therapy orphan drug for treatment ofPompe disease gen storage disease type II), a rare lysosomal storage disorder. It has a molecular weight of about 106 kDa and is tly administered by IV infusions of about 20 mg/kg. In some embodiments, a low-viscosity pharmaceutical formulation of alglucosidase alfa is provided, preferably with a concentration of about 100 mg/mL to about 2,000 mg/mL.
Pegdamase bovine (ADAGEN®) is a modified enzyme used for enzyme replacement therapy for the treatment of severe ed immunodeficiency disease (SCID) associated with a deficiency of adenosine deaminase. Pegdamase bovine is a conjugate of numerous strands of monomethoxypolyethylene glycol (PEG), molecular weight 5,000 Da, covalently attached to adenosine deaminase enzyme that has been derived fi‘om bovine ine. u-Galactosidase is a lysosomal enzyme that catalyses the hydrolysis ofthe glycolipid, globotriaosylceramide (GL6), to galactose and ceramide dihexoside. Fabry disease is a rare inheritable lysosomal storage disease characterized by subnormal enzymatic activity of u—Galactosidase and resultant accumulation of GL—3. Agalsidase alfa GAL®) is a human a-galactosidase A enzyme produced by a human cell line. dase beta (FABRAZYME®) is a inant human a—gaiactosidase expressed in a CHO'cell line. Replagal is stered at a dose of 0.2 mg/kg every other week by intravenous infusion for the treatment of Fabry disease and, off label, for the treatment of Gaucher disease. FABRAZYME® is administered at a dose of 1.0 mg/kg body weight every other week by IV infusion. Other lysosomal enzymes can also be used. For example, the protein can be a lysosomal enzyme as bed in US 2012/0148556.
RasburicaSe (ELITEK®) is a recombinant urate-oxidase indicated for initial management of plasma uric acid levels in pediatric and adult patients with leukemia, lymphoma, and solid tumor malignancies who are receiving anti-cancer therapy ed to result in tumor lysis and subsequent elevation of plasma uric acid. ® is administered by daily IV infusion at a dosage of 0.2 mg/kg.
Imiglucerase (CEREZYME®) is a recombinant analogue ofhuman B- glucocerebrosidase. Initial dosages range from 2.5 U/kg body weight 3 times a week to 60 U/kg once every 2 weeks. ME® is administered by IV infusion.
Abraxane, paclitaxel—conjugated albumin, is approved for metastatic breast cancer, all cell lung cancer, and late stage pancreatic cancer.
Taiiglucerase alfa (ELEYSO®) is a hydrolytic lysosomal glucocerebroside-specific enzyme indicated for long-term enzyme replacement therapy for Type 1 Gaucher disease. The reconnnended dose is 60 U/kg of body weight administered once every 2 weeks Via intravenous infusion.
Laronidase (ALDURAZYME®) is a polymorphic t of the human enzyme o-L—iduronidase that is produced via CHO cell line. The recommended dosage regimen ofALDURAZYME® is 0.58 mg/kg administered once weekly as an intravenous infusion.
Elosufase alfa (VIMIZIM®) is a human N-acetylgalactcsamine-QS- ase produced by CI-IO cell line by BioMarin Pharmaceuticals Inc (“BioMarin”). It was ed by the FDA on February 14, 2014 for the treatment of Mucopolysaccharidosis Type NA. It is administered weekly via intravenous infusion at a dosage of 2 mg/kg.
Other biologics which may be formulated with viscosity-lowering agents include asparaginase erwinia chrysanthemi (ERWINAZE®), incobotulinumtoxin A (XEOMIN®), EPOGEN® (epoetin Alta), PROCRIT® (epoetin Alfa), ARANESP® (darbepoetin alfa), ORENCLA® cept), BATASERON® (interferon beta-1b), NAGLAZYME® (galsulfase); ELAPRASE® ulfase); MYOZYME® (LUMIZYME®, algucosidase alfa); VPRIV® lucerase), abobotulinumtoxin A (DYSPORT®); BAX- 326, Octocog alfa from Baxter; Syncria from mithKline; liprotamase from Eli Lilly; Xiaflex (collagenase clostridium histolyticum) from Auxilium and BioSpecifics Technologies Corp. ; anakinra from Swedish Orphan Biovitrum AB; eptin from l-Myers Squibb; Avonex, Plegridy (BIIBOl 7) from Biogen; NN1841, NN7008 from Novo Nordisk; KRN321 poetin alfa), AMG531 (romiplostim), KRNlZS (pegfilgrastim), KW- 0761 ulizumab) from Kyowa; IB 1001 from Inspiration Biopharmaceuticals; Iprivask from Canyon Pharmaceuticals Group.
Protein Therapeutics in Development Versartis, Inc.’s VRS—3 17 is a recombinant human growth hormone (hGH) fusion protein utilizing the XTEN ife extension technology. It aims to reduce the frequency ofhGH injections necessary for patients with hGH deficiency. VRS-317 has completed a Phase II study, comparing its efficacy to daily injections of non-derivatized hGH, with positive results.
Phase III s are planned. lysin is a proteolytic enzyme secreted by the Gram-negative marine rganism, Vibrio proreolytz'cus. This endoprotease has specific affinity for the hydrophobic s of proteins and is capable of cleaving proteins adjacent to hydrophobic amino acids. Vibriolysin is currently being igated by Biomarin for the cleaning and/or treatment of burns. lysin formulations are described in patent W0 02/092014.
PEG-PAL (PEGylated recombinant phenylalanine ammonia lyase or “‘PAL”) is an investigational enzyme substitution therapy for the treatment of phenylketonuria (PKU), an ted metabolic disease caused by a ncy ofthe enzyme phenylalanine hydroxylase (PAH). PEG—PAL is being ped as a potential treatment for patients whose blood phenylalanine (Phe) levels are not adequately controlled by KUVAN®. PEG- PAL is now in Phase 2 clinical development to treat patients who do not adequately respond to KUVAN®.
Other n therapeutics which may be formulated with viscosity- lowering agents include Alprolix/ rFlXFc, Eloctate/ rFVIIIFc, BMN—l90; BMN-250; Lamazyme; Galazyme; ZA-Ol 1 ; Sebelipase alfa; SEC-103; and HGT—1 l 10. Additionally, fusionuproteins containing the XTEN half-life extension technology including, but not limited to: VRS-317 GH~XTEN; Factor VIIa, Factor VIII, Factor IX; PF05280602, VRS-859; Exenatide- XTEN; AMX—256; GLP2~2GlXTEN; and AMX-l 79 Folate-XTEN—DMl can be formulated with visco sity-lowering agents.
Other late—stage protein therapeutics which can be ated with viscosity-lowering agents include CM-AT from CureMark LLC; NN7999, NN7088, Liraglutide (NN8022), NN9211, Semaglutide (NN9535) from Novo Nordisk; AMG 386, Fiigrastim from Amgen; 4, Factor VIII from CSL Behring; LA-EP2006 (pegfilgrastim biosimilar) from is AG; Multikine (leukocyte interleukin) from CEL-SCI Corporation; LY260554], Teriparatide (recombinant PTH 1—34) from Eli Lilly; NU—lOO from Nuron Biotech, Inc.; Calaspargase Pegol from Sigma-Tau Pharmaceuticals, Inc.; ADI-PEG-20 from Polaris Pharmaceuticals, Inc.; BMN—l 10, BMN—702 from BioMarin; NGR—TNF from Molmed S.p.A.; recombinant human C1 se inhibitor from Pharming Group/Santarus Inc. ; Somatropin biosimilar from LG Life Sciences LTD; a from NPS Pharmaceuticals, Inc.; ART123 from Asahi Kasei Corporation; BAX-11 1 from Baxter; OBI-1 from Inspiration Biopharrnaceuticals; Wilate from arma AG; Talactoferrin alfa from Agennix AG; eplase from Lundbeck; Cinryze from Shire; RG7421 and Roche and Exelixis, Inc.; aurin (PKC412) from Novartis AG; Damoctocog alfa pegol, BAY 86- 6150, BAY 94-9027 from Bayer AG; Peginterferon lambda-1a, Nulojix (Belatacept) from Bristol-Myers Squibb; Pergoveris, Corifollitrcpin alfa (MK-8962) from Merck KGaA; recombinant coagulation Factor IX Fc fusion protein c; BIIB029) and recombinant coagulation Factor VIII Fc fusion protein (rFVIIIFc; BIIB031) from Biogen; and Myalept from AstraZenec'a.
Other early stage protein biologics which can be ated with viscosity-lowering agents include Alferon LDO from Hemispherx BioPharma, Inc.; SL-40l from Stemline Therapeutics, Inc.; PRX-lOZ from Protalix Biotherapeutics, Inc.; KTP-OOI from Kaketsuken/Teijin Pharma Limited; Vericiguat from Bayer AG; BMN—l 11 from in; ACC—OOl (PFw05236806) from Janssen;LY2510924, LY2944876 from Eli Lilly; NN9924 from Novo Nordisk; INGAP peptide from Exsulin; ART-122 from Abbvie; AZD9412 from AstraZeneca; STIN (BGOOOIO) from Biogen; Luspatercept (ACE-536), Sotatercept (ACE-011) from Celgene ation; PRAME immunotherapeutic from GlaXOSmithKline; Plovamer acetate (PI-2301) from Merck KGaA; PREMIPLEX (607) from Shire; BMN~701 from BioMarin; Ontak from Eisai; rHuPHZO/insulin from Halozyme, Inc.; PB-1023 from PhaseBio Pharmaceuticals, Inc.; ALV-003 from Alvine Pharmaceuticals Inc. and ; NN8717 from Novo Nordisk; PRT-201 from Proteon Therapeutics Inc.; PEGPH20 from Halozyme, Inc.; A1nevive® ept from Astellas Pharma Inc.; F-627 from Regeneron; AGN—214868 (semebotase) from Allergan, Inc.; BAX-817 from Baxter; PRT4445 from Portola Pharmaceuticals, Inc.; VENlOO from Ventria Bioscience; Onconase/ ranpirnase from Tamir Biotechnology Inc.; interferon alpha«2b infusion from Medtronic,1nc; pase alfa from Synageva BioPharma; IRX-2 from IRX eutics, Inc; GSK2586881 from GlaxoSmithKline; 3 from Seikagaku Corporation; ALXN1101, asfotase alfa from Alexion; SHP611, SHP609 ase, idursulfase) from - Shire; PF-04856884, 80602 from Pfizer; ACE—031, Daiantercept from Acceleron Pharma; ALT-801 from Altor BioScience Corp; BA—210 from BioAxone Biosciences, Inc.; WTI immunotherapeutic from GlaxoSmithKiine; GZ402666 from Sanofi; MSBOOIO445, Atacicept from Merck KGaA; Leukine (sargramostim) from Bayer AG; KUR—le from Baxter; fibroblast growth factor-1 from CardioVascular BioTherapeutics Inc.; SPI—2012 from Hanrni Pharmaceuticals Co., LTD ruIn Pharmaceuticals; FGF-18 (sprifermin) fiom Merck KGaA; MK—i 293 from Merck; interferon-alpha—Zb from HanAll Biopharma; CYT107 fiom Cytheris SA; RTOOI from Revance Therapeutics; Inc.; MEDI6012 from AztraZeneca; E2609 from Biogen; BMN—190, BMN-270 from BioMarin; ACE-661 from Acceleron Pharrna; AMG 876 from Amgen; GSK3 052230 from GlaxoSmithKline; RG7813 from Roche; SAR342434, Lantus from Sanofi; A201 from Aliozyne Inc. ; ARX424 from Ambrx, Inc.; FP-1040, FP-1039 from FivePrime eutics, Inc.; ATX-MS-1467 from Merck KGaA; XTEN fusion proteins from Amunix Operating Inc.; entolimod (CBLBSOZ) from Cleveland BioLabs, Inc.; HGT2310 from Shire; HM10760A from Hannu' Pharmaceuticals Co., LTD; ALXNl 102/ ALXN1103 from n; CSL-689, CSL-627 from CSL Behring; glial growth factor 2 from Acorda Therapeutics, Inc. ; NXOOI from Nephrx Corporation; NN8640, NN1436, NN1953, NN9926, NN9927, NN9928 from Novo Nordisk; NHS-IL 12 from EMD Serono; 3K3A-APC from 22 Biotech LLC; PB-1046 from PhaseBio Pharmaceuticals, Inc. ; RU-IOI from R—Tech Ueno, Ltd.; insulin WO 38818 lispro/BC106 from Adocia; l from Iconic Therapeutics,_lnc.; PRT-IOS from Protalix BioTherapeutics, Inc; PF-04856883, CVX-096 from Pfizer; ACP-501 from AlphaCore Pharma LLC; BAX-855 fiom Baxter; CDX-1135 from Celldex Therapeutics; PRM-151 from Promedior, Inc.; T801 from Thrombolytic Science International; TT-173 from Thrombotargets Corp; QBI—139 from Quintessence Biosciences, Inc.; Vatelizumab, GBRSOO, GBR600, GBR830, and GBR900 from rk Phannaceuticals; and 91 from Cytimmune Sciences, Inc.
Other Biologic Agents Other biologic drugs that can be formulated with Viscosity-lowering agents include PF-05285401, 31023, RN317 (PF-05335810), PF- 06263507, PF-05230907, Dekavil, PF-06342674, PF06252616, RG7598, RG7842, RG7624d, OMP54F28, GSK1995057, 9470, IMC—3G3, IMC-18F1, IMO-3 5C, IMC~20D7S, 80605, PF—0_6647263, PF- 06650808, PF»05335810 (RN317) PD-0360324, PF—00547659 from Pfizer; MK—8237 from Merck; B1033 from Biogen; GZ402665, SAR43 8584/ REGN2222 fiom Sanofi; IMC-lSFl; and Icrucumab, IMC—3G3 from ImClone LLC; Ryzodeg, Tresiba, Xultophy from Novo Nordisk; Touj60 (U300), LiXiLan, Lyxumia (lixisenatide) from Sanofi; MAGE-A3 immmotherapeutic from GlaxoSmithKline; Tecemotide from Merck KGaA; Sereleaxin (RLX030) from Novartis AG; Erythropoietin; rastim; LY2963016, Dulaglutide (LY2182965) from Eli Lilly; and Insulin Glargine from Boehringer Ingelheim.
B. Viscosity-lowering Agents The ity of liquid protein formulations, including low- lar-weight and/or high-molecular-weight proteins, is reduced by the addition of one or more ity-lowering agents. The pharmaceutical formulations may be converted from non-Newtonian to ian fluids by the addition“ of an effective amount of one or more Viscosity-lowering agents.
When employed in a ation intended for administration to a human or other mammal, the Viscosity-lowering agents, like the formulation itself, must be pharmaceutically able. The viscosity~lowering agents are typically organic compounds containing at least one non-carbon, non- hydrogen atom. Preferably, the Viscosityulowering agents contain hydrogen, carbon, oxygen and at least one other type of atom. In certain embodiments, the Viscosity-lowering agents are characterized by at least one of the following: 1) organic nds having at least four carbon and four hydrogen atoms, and at least one sulfur, oxygen, nitrogen, or phosphorus atom; 2) a molecular weight between about 85 and 1,000 Da; 3) the presence of at least one charged, or other hydrophilic, moiety; 4) the presence of at least one, preferably two, and more preferably three, freely rotating bonds; ) the presence of at least one substituted ring; - 6) a molecular polar Surface area of at least 24 43, ably at least 50 A2, and more preferably at least 80 A2; 7) a molar volume of at least 75 cm3, preferably at least 85 cm3, more preferably at least 100gcm3, and most preferably at least 120 cms; a polarizability of at least 10 cm3, preferably at least 15 cm3, more ably at least 20 01113, and most preferably at least 25 cm3; and 9) the presence of at least one, preferably two, and more preferably three ' hydrogen bond donors and/or acceptors.
In certain embodiments, the viscosity-lowering agent is characterized by at least two, three, four, five, Six, seven, eight or all nine of the above listed attributes. In certain embodiments, the viscosity-lowering agent is r characterized in that it does not contain an aldehyde or carbon—carbon triple bond functional group.
In other embodiments, the viscosity-lowering agent is a combination oftwo or more compounds, each of which is characterized by at least two, three, four, five, six, seven, eight or all nine of the above listed attributes.
In some embodiments, the viscosity-lowering agents are listed as GRAS by the US. Food and Drug Administration (“the FDA”), as of September 11, 2014. "GRAS" is an acronym for the phrase generally Recognized As fiafe. Under sections 201(s) and 409 of the Federal Food, Drug, and Cosmetic Act (the Act), any substance that is intentionally added to food is‘a food additive and is subject to premarket review and approval by FDA unless the substance is generally recbgnized, among ed experts, as having been adequately shown to be safe under the conditions of its intended use, or unless the use of the substance is otherwise excluded from - the definition of a food additive. Another source of compounds is the Inactive ient Guide of the FDA (HG), and equivalents listed by the International Pharmaceutical Excipients Council (IPEC) and the European Medicines Agency (EMA), as of September 11, 2014. The substances used in formulations must be safe for injection. Preferably, the GRAS—listed viscosity~lowering agent is characterized by at least two, three, four, five, six, seven, eight or all nine of the above listed utes.
In other embodiments, the viscosity-lowering agent is an FDA- or EMA-approved drug product as of September 11, 2014. Like compounds drawn from the GRAS and HG lists, the toxicity and safety profiles of FDA- and EMA-approved drug ts are well established. In addition to lowering the viscosity of the protein solution, the use of an FDA- or EMA- approved drug product provides the opportunity for combination therapies.
Preferably a FDA— or EMA-approved drug product Viscosity-lowering agent is characterized by at least two, three, four, five, six, seven, eight or all nine of the above listed utes.
In some embodiments, the viscosityJowering agent includes at least one compound ofFormula (I): (R3); A , (R3); (3 3).: (Ra): Formula (1), or a pharmaceutically acceptable salt f; wherein ii represents either a single or double bond, A is a ed from O, s, 802, NR3, C(R3)2 or: (at): _ (R32; ”is. M wherein R3 is independently selected from hydrogen, R2, -OH, NH;, - F, -Cl, -Br, -1, -N02, —CN, —C(=O)R4a, -C(=NR4a)R4, -C(=O)OH, OR4, )R4, -OC(=O)OR4, _s03H, -SOgN(R43 2, soar, -SOgNR4aC(=O)R4, -PO3H2, ~R4aC(=NR4a)N(R4a 2, -NHC(=NR4a)NH-CN, -NR4“C(=O)R4, - NR4aSO2R4, ~NR43C(=NR43)NR4aC(=NR4a)N(R4a 2, (=O)N(R4a)2, - C(=0)NH2, -C(=O)N(R4a)2, -OR4, -s12.4a, and -N(R4a 2; wherein R2 is independently selected from Cuzalkyl, C3-1gcycloalkyl, C5_;2aryl, C1-1zheteroary1 and C2_12heterocyclyl; wherein each szalkyl may be substituted one or more times with C3420ycloalkyl, C5.12aryl, C1-12heter0aryl, C2-12heterocyclyi, -OH, NH;, (:0), (=NR4a), -F, _c31, ~Br, —1,-No2, -CN, -C(=O)R4a, -C(=NR4E)R4, — C(=O)0H, -C(=O)OR4, -OC(=O)R4, -OC(:0)0R4, -SO3H, -so2N(R“a 2, — so2R4, -SOgNR4aC(=O)R4, -P02H2, -R43C(=NR4a)N(R4a 2, — NHC(=NR4a)NH-CN, —NR4aC(:0)R4, «114330216, - , NR4aC(=NR4a)NR4aC(=NR4a)N(R4a)2, -NR4aC(=O)N(R4" 2, —C(:0)NH2, C(=O)N(R4a)2, -OR4, s11“, or -N(R4a)2; wherein each C3.12cycloalky1 may be substituted one or more times with C1_12a1ky1, Ca-lzflI‘YL C1-12heteroary1, C2_;2heterocyclyl, -OH, NH;, —F, - (:1, Br, -1, -No2, -CN, -C(=0)R4“, -C(=NR4a)R4, -C(=O)OH, -C(=O)OR4, - OC(=O)R4, »OC(:O)OR4, -so2H, -so2N(R“a 2, —so2R4, 2so2NR4aC(=0)R4, —P02H2, -R4“C(=NR43)N(R4a 2, —NHC(=NR4a)NH-CN, -NR4aC(=O)R4, — NR4aso2R“, -NR4“C(=NR4a)NR4aC(=NR4a)N(R4a 2, (:O)N(R4a)2, - C(=0)NH2, -C(=O)N(R4a)2, -0R4, s11“, or —N(R4a 2; wherein each ry1 may be substituted one or more times with C1- Igalkyl, C3.1gcycloa1kyl, C1_1;2heteroaryl, erocyclyl, -OH, NHg, -F, -Cl, -Br, -1, ~N02, -CN, -C(=O)R4a, -C(=NR43)R4, -C(=O)OH, —C(=0)0R4, - OC(=O)R4, -OC(=O)OR4, -802H, —s02N(R4a 2, , -so2NR4aC(=0)R4, -P03H2, -R4aC(=NR4a)N(R4a 2, NR4a)NH-CN, —NR4aC(=O)R4, - NR4aSOgR4, -NR4aC(:NR4")NR4aC(=NR4a)N(R4a)2, -NR4aC(=O)N(R4a 2, — C(=0)NH2, -C(=O)N(R4a)2, -0R4, -SR4a, or —N(R4a)2; wherein each C1.1zheteroaryl may be substituted one or more times with C1.12a1kyl, cloalkyl, €5-12aryl, C2_1gheterocyc1yl, -OH, NHz, -F, - Cl, —Br, -1, -N02, ~CN, -C(=O)R4a, -C(=NR4a)R4, -C(=O)OH, -C(=0)0R4, — OC(=0)R4, )OR4, —so2H, R4a 2, -so2R“, -SOzNR4aC(=O)R4, -P03H2, -R4aC(=NR4“)N(R4a)2, ~NHC(=NR4a)NH-CN, -NR4aC(=O)R4, - 2R4, (=NR4a)NR4aC(=NR4a)N(R4a 2, -NR43C(=O)N(R4a)2, - C(=O)NH2, —C(:0)N(R4“)2, -0R4, -SR4a, or -N(R4a)2; wherein each C2.12heterocyclyl may be substituted one or more times with C}-;2aikyl, ycloalkyl, C5-1garyl, C1.12heteroaryl, -OH, NHz, ~F, —Cl, -Br, -I, -N02, -CN, -C(=O)R4a, -C(=NR4a)R4, —C(=O)OH, -C(=O)OR4, - OC(=O)R4, —OC(=0)0R4, -s03H, -SO;N(R4a)2, , -SO;NR4aC(=O)R4, —P03H2, -R4aC(=NR4a)N(R4a 2, NR4a)NH-CN, -NR4aC(=O)R4, — NR4as02R4, -NR‘iaC(=NR4a)NR4aC(=NR4a)N(R4a 2, —NR4aC(=O)N(R4a 2, - C(=O)NH2, -C(=0)N(R4a 2, ~0R4, -SR4a, or -N(R4a 2; wherein R4 is independently selected from C1-12a1kyl, C3_1gcycloalkyl, C6_1zaryl, C1_12heteroaryl and C2-lgheterocyclyl, each Of which may be substituted one or more times by -OH, -NH2, -F, -Cl, -Br, -1, -N02, -CN, — C(=0)OH, -SO3H, -PO3H2, or -C('—“O)NH2; wherein K“ may be R4 or hydrogen; wherein any two or more of R2, R3 , R4 and R43 groups may er form a ring; wherein when two R3 groups are bonded to the same carbon atom, the two R3 groups may together form an (=0), (=NR4a or (:C(R4a)2); wherein z is in each case independently selected from 1 or 2, provided that when the (15(3)z tuent is connected to an sp2 hybridized carbon, 2 is 1, and when the (R3)z substituent is connected to an sp3 hybridized carbon, 2 is 2.
When the substituent -NR43C(=NR4a)NR4aC(=NR43)N(R4a)2 is t, it is preferred that R421 is selected so as to give - NHC(=NH)NHC(=NH)NH2.
In n embodiments, the compound of Formula (1) ns at least one substituent selected from -C(=O)OH, -SO3H, -SOgNHC(=O)R4, and —PO3H2. In some embodiments, the compound of Formula (1) contains at least one —SO3H group.
In certain embodiments, one or more of the R3 substituents may be: O {Rag H x ,gJLNncasbzt-ew N CR3” “N2}‘ ‘5 a," fir‘ was H R o R33. 7:510/(Gama); R3“ / 4:: (033”2 x—-N’ N\ ‘57» Y FR“ R32: 01' 0 wherein R3a and R3b are independently ed from hydrogen, C1-1231ky1, C3.120ycloalkyl, ryl, C1_1zheteroaryl and C2-12heterocyclyl, C(=O)R4a, — H, -C(=O)OR4, -SO3H, —5102N(R4a 2, ~SOZR4, -SOgNHC(=O)R-4, C(=O)NH2, -C(=O)N(R4a)2, -0R4, -SR“, and -N(R43 2, and when any two R3b are bonded to the same carbon atom, the two R3ID groups may together form an (=0), (:NR“), or a)2); wherein each C1_12alkyl, C3.1gcycloalkyl, C5_uary1, C1_12heteroaryl and C2.1gheterocyclyl may be substituted one or more times with -OH, NHz, -F, - (:1, -Br, -1, -NO;, -CN, -C(=O)R4a, -C(=NR4a)R4, -C(=O)OH, —C(=0)0R4, - OC(=0)R4, -OC(=O)OR4, -503H, ~S02N(R4” 2, -soZR4, -SO;NR4‘1C(:O)R4, -P03H2, -R4aC(=NR4a)N(R4a 2, -NHC(=NR4a)NH-CN, ~NR4aC(=0)R4, - NR43802R4, -NR4ac(:NR“)NR4*‘C(:NR43)N(R43)2, (=O)N(R4a)2, — C(=O)NH2, -C(:O)N(R4a)2, -0R4, -SR4a, or -N(R43 2; wherein R4 and R4a are as defined above; wherein x is ed from 1, 2, 3, 4, 5, 7, 8, 9 or 10; and wherein any two or more of R3 , R3a, R4 and RA"1 groups may together form a ring.
In certain embodiments, the compound of Fom1ula(l) may be represented by either the compound of Formula (la) or (lb): Formula (la) Formula (1b) wherein R3 has the meanings given above.
In certain embodiments, the compound of Formula (la) may be represented by the compounds of Formulas (la-i—iv): R3 Ea R3 /(CR352)KW\ :12 R38 R3 Formula (la-i) Formula (1 a-ii), Formula (la-iii), Formula (la—iv) n R3 is independently selected from hydrogen, NHz, CH3, C1, OR“ and NHR“; wherein x is 1 or 2; wherein R33 and R3‘) are independently Selected from hydrogen and C142 alkyl; n said lkyl may be substituted one or more times by C3- lgcycloalkyl, C5-12aryl, Cuzheteroaryl, €2.12heterocyclyl, ~OH, NHz, ~F, -Cl, —Br, -1, -N02, -CN, -C(=O)R4a, -C(=NR4"‘)R“, OH, —C(=0)0R4, _ OC(=0)R“, -0C(=0)0R4, sogH, ~SOZN(R4a 2, 60212“, -802NR4aC(=0)R4u -P03H2, - 4aC(=NR4a)N(R4a)2, -NHC(=NR4B)NH-CN, -NR4aC(=O)R4, — NR4aS02K: — R4aC(=NR4&)NR4“C(:NR43)N(R43 2, -NR4‘*C(:O)N(R43)2, - C(=O)NH2, —C(=O)N(R4a 2, -0114, $1142 or -N(R43 2; R4 and 11’” are as defined above; and wherein any two or more R33, R3b, R4 R451 may together form a ring.
The compound of Formula (1) may be represented by the compound of Formula (1 a-V, Vi or Vii): R3 R3 R3! R3 R3 R3 R3 R3 R3 ' R3 R3 (1 a—v), R3 R3 (la-vi), R3 R3 (vii) wherein R3f is selected from -C(=O)OH, —so3H, -SOgNHC(=O)R4, and — P03H2, and R3 is as defined above. In certain preferred embodiments, R3 is independently selected from en, OH, NHZ, C1_5alkyl and COOH.
In other embodiments, the compound of Formula (1) may be represented by any of the compounds of ae (10), (1d), (1e) or (if): Formula (1d) 3 a R R3 R38 a (le) Formula (It), wherein R3 has the meanings given above.
In other embodiments, the compound of Formula (1) may be represented by a compound of Formula (lg): R3:! 3' R33 R3138 6 Formula (1 g), wherein R3c is independently selected from hydrogen and R2, wherein R2 has the meanings given above; wherein R3d is independently ed from hydrogen, OH, NH;, NH(C1-5alky1), N(C1_6alky1)2; NHC(=O)(Cl_5alkyl), COOH and CHZOH; or any two R“ and R3d groups connected to the same carbon may together form an oxo (=0), imino (=NR43), or an olefin (=C(R4a)2), wherein R4a has the meanings given above; WO 38818 wherein R3'3 is selected from hydrogen, —OH or 0R4; and wherein R4 has the meanings given above.
In n embodiments, the viscosity-lowering agent includes a compound of a (1 g-i): R301! 0 R313 £10 0H 0“ Formula (1 g-i), n R3e is selected from OH and -OC1.12alkyl, which is further substituted with at least one OH and at least one COOH: and wherein R3d is selected from COOH and CHZOH.
In some embodiments, the viscosity—lowering agent includes a compound of Formula (2): ea): (R3): Formula (2), or a phannaceutically acceptable salt thereof; wherein i represents a single or double bond; X is ndently selected from chalcogen, N(R3)z and C(R3)z; X1 is absent, or is chalcogen, N(R3)Z, C(R3)Z or: (R3): “a.“M1,; wherein R3 has the meanings given for the compound of Formula (1); provided that when the (R3)Z tuent is connected to an sp2 hybridized nitrogen, z is 0 or 1, when the (R3)z substituent is connected to an sp2 hybridized carbon or an sp3 hybridized nitrogen, z is 1, and when the (R3)Z substituent is ted to an sp3 hybridized carbon, 2 is 2; wherein at least one ofX or X1 is chalcogen or N(R3)z.
In certain embodiments, the compound may be an aromatic ring.
Exemplary aromatic rings include the compounds of Formulas (2a-e): R3 1}? R3 “a. X 93%)4 R3 ”2% R3 Formula (2a), R3 Formula (2b), 0 w' R3 - X2 R3 R3 Formula (2c), R3 Formula (2d), XQ/ X k 1 9-4? R3 X2 X2 Formula (26), n R3 and X have the meanings above, and X2 is ed from N(R3)z and C(R3)z.
In certain embodiments, the viscosity-lowering agent is a compound of Formula : 3" N (Formula 2a—i), wherein R4 is as defined above and is ably hydrogen or CH3; wherein R6 is C1.12heteroaryl, which may be substituted one or more times by C1-5alkyl; wherein said C1.6alkyl may be substituted one or more times by OH, -NH2, «F, -c1, -Br, —1, ~N02, —CN, —C(=0)R4, —C(=NR4a)R4, -C(=O)OH, — C(=O)OR4, -s03H, —s02NR4-, -S02R4, $0,117,, —NHC(=O)R4, — NHC(=O)N(R4)2, -C(:0)NH2, -C(=O)N(R4)2, -OR4b, -SR4b, -N(R4b)2, wherein R4 has the meanings given above; or . (iii-{O SR7 a/ij/AMR‘ wherein R4 is as defined above, and R7 is selected from SR4 and —C(=0)R4.
The double bond in the group above may be in either the E or Z geometry. 2014/055254 In preferred embodiments, R6 is a heterocycle having the structure: rug/AXi R53 R63 n X4 is a chalcogen and R6&1 is hydrogen or C1-5alkyl, wherein the C1. fialkyl may be substituted one or more times by ~0H, —NH2, -F, -Cl, -Br, -I, - N02, -CN, —C(=0)OH. In an even more preferred ment, R6 is a heterocycle having the structure: {Rug/\sC425 Rue R63 wherein R6a is selected from unsubstituted C1_6alkyl and C1.5alkyl substituted one or more times with IQH.
The viscosity—lowering agent may be an imidazole of a (Zb-i) R3 R“ Formula (2134), wherein R3 is as defined above. In certain embodiments, R3 is independently selected from hydrogen, N02, and R4. In certain preferred embodiments, the compound of Formula (2b-i) has the structure: N(Luna wherein R3 is independently ed from CH; alkyl, which may be unsubstituted or substituted one or more times with a group selected from 0H, NH;, SR4, F,C1, Br and I; and R3g is either hydrogen or N02.
In other embodiments, the viscosity—lowering agent has the structure of Formula (2a-ii) or Formula (EC-i): 2014/055254 R3 Formula (2a-ii), R3 R3 Formula (204), wherein R3 is independently selected from OH, Cl, Br, F, I, N(R4a)2, C(IO)OH, H2.
In further embodiments, at least one R3 substituent is NHR4, n R4 is a C1_5alky1, optionally substituted by one or more groups selected from Cl, Br, F, I, OH, C(=O)OH, NHz, NH(C1_5alkyl) and N(C1.5alkyl)2.
In other embodiments, the Viscosity-lowering agent is a pyridinium salt of Formula (2a—iii): R3 / P/Rfi R3 R3 wherein R3 and R4 are as defined above.
In other embodiments, the heteroeyclie ring is not a aryl ring.
Exemplary non—aromatic rings include the compounds of Formulas (2f—k): - R3 )4 R3 R3 R3 R3 R3 R3 R3 R3 Ra Ra R3 R3 R3 x3 R3 3R3 XXX R3 Formula (2f) R333 R Formula (2g), P.3 Ra Formula (2h) Formula (Zj), Formula (2k) wherein R5 and X have the meanings above, and X3 is gen or N(R3)z.
In certain embodiments, the compound of a (2f) is a beta- lactam of Formula (Zf-i), R3 R3 R3itR3 0 R3 Formula (2fni).
The beta lactam of Formula (2f—i) includes penicillin-type compounds, as well cephalosporin-type and cephamycin—type compounds of the Formula (2f-ii) and (2f—iii): will?“a a 9.ng R3 0::37 3 X R; - R3}? 0 R3 as Formula (2f-ii) - R3 Formula (2m). wherein X and R3 are as defined above. In preferred embodiments, X is sulfur.
In n embodiments, the compound of Formula (2i) is a compound ofFormula : or;Formula (2i-i) wherein X and R3 are as defined above. In certain embodiments, X is in both cases NR4, whereinR4 has the meanings given above, and R3 is in both cases hydrogen.
In other embodiments, the compound of Formula (2) is represented by a compound of Formula (2i-ii): F?3 x R3 R3 R" x R3 Formula (21-111), wherein X, X1 and R3 are as defined above.
The nd ula (Zj) may be represented by the compound ofFormula (2j ~i): WO 38818 IRE.
R3(Formu1a (Zj-i), wherein X3 and R3 are as defined above, and R8 is selected from the NHC(=0)R2 and OC(=O)R2. In preferred embodiments, X3 is N+(CH3)2, R3 are both hydrogen, or R3 together form an epoxide or double bond.
The compound ula (2k) may be represented by the compound of Formula (2k-i): a3 Formula , wherein X3 and R8 are as defined above.
In other embodiments, the viscosity-lowering agent includes a compound of the structure of Formula (3): stif"R€ R5 Formula (3), or a pharmaceutically acceptable salt thereof; wherein R5 is in each case independently selected from hydrogen, and R2, R5, is either R5 or absent; providing that at least one R5 substituent is not hydrogen, n R2 has the same meanings given for the compound or Formula (1).
In certain embodiments, the viscosity-lowering agent is a mixture of two or more compounds selected from nds of Formula (1), Formula (2) and Formula (3).
In preferred embodiments, the viscosity—lowering agent is camphorsulfonic acid (CSA), or a pharmaceutically acceptable salts thereof, such as an alkaline or alkaline earth metal salt. The camphorsulfonic acid or salt thereof is combined with one or more compounds of Formula (1), (2) or (3) to give es such as CSA-piperazine, GSA-TRIS, CSA—4-amino pyridine, CSA—l-(c-tclyl)biguanide, ocaine, CSA-Na— 2014/055254 aminocyclohexane carboxylic acid, CSA-Na—creatinine and GSA-Na- omidazole. Other red viscosity-lowering agents include thiamine, procaine, biotin, nine, metoclopramide, amine, cimetidine, chloroquine phosphate, mepivacaine, granisetron, sucralose, HEPES-tris, nicotinamide, ionic acid-TRIS, glucuronic acid-TRIS, sulfacetamide, CSA-4~aminopyridine, CSA—piperazine and cefazolin. Any two or more of the viscosity-lowering agents listed above may further be ed in the same formulation.
In other embodiments, the viscosity-lowering agent is an organosulfonic acid. Exemplary organosulfonic acids include, but are not limited to, camphorsulfonic acid, naphthalene—Z-sulfonic acid, benzenesulfonic acid, toluenesulfonic acid, cyclohexylsuflonic acid, xylenesulfonic acids (including p-Xylene-Z-sulfonic acid, m-xylene-Z- sulfonic acid, m-Xylenesulfonic acid and o-Xylene-3 —sulfonic acid), esulfonic acid, 1,2 ethane disulfonic acid, 4-(2-hydroxyethyl)—l- piperazine ethane sulfonic acid, 2-hydroxyethanesulfonic acid, 3— hydroxypropane-l-sulfonic acid, cymenesulfonic acid, 4-hydroxybutane sulfonic acid and pharmaceutically acceptable salts thereof. The sulfonic acid may be in the form of an ne or alkaline earth metal salt, such as lithium, sodium, potassium, magnesium, and calcium salt. The organosulfonic acid (or salt thereof) may be combined With one or more compounds ula (2) or Formula (3).
In certain embodiments, the yiscosity—lowering agent contains at least one carboxylic acid. The carboxylic acid may be in the form of an alkaline or alkaline earth metal salt, such, as lithium, , potassium, magnesium, and calcium salt. Exemplary carboxylic acid compounds include lactobionic acid, glucuronic acid, l-aminocyclohexane carboxylic acid, biotin, brocrinat, cyclopentane propionic acid, ynaphthoic acid, phenyipropionic acid, gentisic acid, salicylic acid, camphoric acid, mandelic acid, sulfosalicyclic acid, hydroxybenzoyl benzoic acid, phenyl acetic acid, acetyl salicylic acid, cinnamic acid, t-butyl acetic acid, ic acid, trimethylacetic acid, anthrallic acid and pharmaceutically acceptable salts thereof. The carboxylic acid (or salt thereof) may be combined with one or more compounds of Formula (2) or Formula (3).
The following compounds may also be used as Viscosity-lowering : colistin, articaine, tetracaine, etacaine, metoclopramide, procaine, lidccaine, cyclomethylcaine, piperocaine, chloroprocaine, etidocaine, aine, phenylephrine, bupivacaine, mepivacaine, cinchocaine, mixtures thereof and and pharmaceutically acceptable salts thereof.
Other agents which may be employed as viscosity-lowering agents include lmaminocyclohexane carboxylic acid, 1-(o-tolyl)biguanide, benzethonium chloride, benzoic acid, brocrinat, calcium carrageenan, calcium cyclamate, calcobutrol, caloxetic acid, camphorsulfonic acid, creatinine, dalfampridine, dehydroacetic acid, diazolidinyl urea, dichlorobenzyl alcohol, dimethyl isosorbide, epitetracycline, ethyl maltol, ethyl vanillin, ornidazole, gentisic acid ethanolamide, HEPES (4~(2- hydroxyethyl)-l-piperazine ethane sulfonic acid), gentisic acid, glucuronic acid, iodoxamic acid, menthol, galactose, medronic acid, m—cresol, glutathione, lactobionic acid, maltitol, octisalate, oxyquinoline, pentetic acid, piperazine, yl guaethol, propyl gallate, propylene carbonate, propylparaben, protamine e, QUATERNHIM-15, QUATERNIUM-SZ, satialgine H, sodium 1,2—ethanedisulfonate, sodium cocoyl sarcosinate, sodium lauroyl sarcosinate, sodium polymetaphosphate, sodium pyrophosphate, pyroglutamic acid, sodium aphosphate, sodium tripolyphosphate, sorbitan, tartaric acid, lactic acid, mine, sucralcse, 1— (4—pyridyl)pyridinium de, aminobenzoic acid, sulfacetamide sodium, naphthalenenZ—sulfonic acid, tert-butylhydroquinone, thimerosal, trolamine, tromantadine, in, versetamide, nioxime, amide, methylisothiazolinone, mannose D, maltose, lidofenin, lactose, lactitol, isomalt esulfonic acid, xylenesulfonic , imidurea, gluconolactone, acid, sulfobutylether B—cyclodextrin and pharmaceutically acceptable salts In certain embodiments, the Viscosity-lowering agent includes an organic base. Exemplary organic bases include N-methylglucamine, line, piperidine, and primary, secondary, tertiary, and quaternary amines, substituted amines, and cyclic amines. For example, they can be isopropylamine, trimethylamine, lamine, triethylamine, tripropylarnine, ethanolamine, 2-diethylaminoethanoi, trimethamine, dicyclohexylamine, lysine, ne, histidine, caffeine, procaine, lidocaine, hydrabamine, cholines, betaines, choline, e, nediamine, theobrornine, purines, piperazine, N—ethylpiperidine, N- rnethylpiperidinepolyamine. Particularly preferred organic bases are arginine, histidine, lysine, ethanolamine, thiamine, 2-amin0—2- hydroxymethyl-propane~ l l (TRIS), 4~aminopyridine, aminocyclohexane carboxylic acid, lybiguanide, ornidazole, urea, njctoinamide, benzethonium chloride, S-amino—l-pentanol, 2—(2- arninoethoxy)ethanol, trans—cyclohexaneul,4udiamine, cyclohexane— 1R, 2R-diamine, ethylenediamine, propane-1,3-diarnine, butane-1,4-diamine, pentane—1,5~diamine, -1,6—diamine, octane—1,8—diamine, 5-amino pentanol, 2-(2-aminoethoxy)ethanarnine, 2-(2-(2-aminoeth0xy)- )ethanamine, 3-(4-(3-aminopropoxy)-butoxy)propan—1-arnine, 3-(2-(2— (3 propoxy)-ethoxy)-ethoxy)propan—l-amine, N—(2-(2— thylamino)ethy1)ethane-1 ,2-diamine, N—(2-aminoethy1)ethane-1 ,2— e, N— l -(2-(2-(2-aminoethylamino)ethylamino)-ethyl)ethane- 1,2— e, N,N-dimethylhexane-1,6-diamine, N,N,N,N—tetramethylbutane-l ,4- diarnine, phenyltrimethylammonium salts, isopropylarnine, diethylamine, ethanolamine, trimethamine, choline, 1-(3-aminopropy1)—2-methyl-lH- imidazole, piperazine, 1—(2-aminoethyl)piperazine, 1-[3- (dimethylamino)propy1]piperazine, l-(2-aminoethyl)piperidine, 2-(2— aminoethyl-l-methylpyrrolidine, mixtures thereof, and ceutically acceptable salts thereof.
Exemplary beta-lactams include benzylpenicillin (penicillin G), phenoxymethylpenicillin (penicillin V), cloxacillin, dicloxacillin, flucloxacillin, methicillin, nafcillin, oxacillin, temocillin, amoxicillin, ampicillin, mecillinarn, carbenicillin, ticarcillin, llin, mezlocillin, piperacillin, cefoxitin, cefazolin, cephalexin, cephalosporin C, cephalothin, cefaclor, cefamandole, cefuroxime, cefotetan, cefixime, cefotaxime, cefpodoxime, ceftazidime, ceftriaxone, cefepime, cefpirome, ceftobiprole, biapenem, doripenem, ertapenem‘faropenem, imipenem, meropenem, panipenem, razupenem, tebipenem, thienamycin, aztreonam, tigemonam, nocardicin a, inine, clavulanic acid, clavulanic acid, tazobactam, sulbactam and pharmaceutically acceptable salts thereof.
Other viscosity-lowering agents include tropane N-heterocycles, such as atropine, amine, scopolamine, and salts thereof, as well as tiotropium and ipratropium salts, thiamine, iamine, prosultiamine, fursultiamine, benfotiamine, iamine, quaternium 15; 1-(3-aminopropyl)methyl-1H- imidazole dihydrochloride; creatinine; biotin, cimetidine, piperocaine, cyclomethylcaine, granisetron, moxifloxacin, quine, mepivacaine, levetiracetam, bupivacaine, caine, clindamycin and pharmaceutically able salts thereof. Thiamine is an especially preferred viscosity-lowering agent.
In certain formulations, the following compounds are not preferred: creatinine, cadaverine, lidocaine, ne and lysine, and are excluded from the scope of the foregoing formulas and definitions of useful viscosity-lowering agents.
C. Excipients A wide variety of pharmaceutical excipients useful for liquid protein formulations are known to those skilled in the art. They include one or more additives, such as liquid solvents or vents; sugars or sugar alcohols such as mannitol, trehalose, e, sorbitol, fructose, maltose, lactose, or dextrans; surfactants such as TWEEN® 20, 60, or 80 (polysorbate 20, 60, or 80); buffering agents; preservatives such as benzalkonium chloride, benzethonium chloride, tertiary ammonium salts, and chlorhexidinediacetate; carriers such as poly(ethylene glycol) (PEG); antioxidants such as ascorbic acid, sodium metabisulfite, and nine; chelating agents such as EDTA or citric acid; or biodegradable polymers such as water soluble polyesters; cryoprotectants; lyoprotectants; bulking agents; and stabilizing agents.
Other pharmaceutically acceptable carriers, excipients, or stabilizers, such as those described in Remington: “The Science and Practice of cy”, 20th edition, Alfonso R. Gennaro, Ed., cott Williams & Wilkins (2000) may also be included in a protein formulation described herein, provided that they do not adversely affect the desired characteristics of the formulation.
The viscosity-lowering agents described herein can be combined with one or more other types of Viscosity-lowering agents, for example, organophosphates described in co—filed PCT application entitled “LIQUID PROTEIN FORMULATIONS NING ORGANOPHOSPI-IATES” by Arsia Therapeutics; water soluble dyes described in d PCT application entitled “LIQUID N FORNIULATIONS CONTAINING WATER SOLUBLE ORGANIC DYES” by Arsia Therapeutics; ionic liquids described in co-filed PCT application entitled “LIQUID PROTEIN FORMULATIONS CONTAINING IONIC LIQUIDS” by Arsia eutics.
III. Methods of Making The protein, such as a mAb, to be ated may be produced by any known technique, such as by culturing cells transformed or transfected with a vector containing one or more nucleic acid sequences encoding the protein, as is well known in the art, or through synthetic techniques (such as recombinant techniques and peptide synthesis or a combination of these ques), or may be isolated from an endogenous source of the n.
Purification of the protein to be formulated may be conducted by any suitable technique known in the art, such as, for example, ethanol or um sulfate precipitation, reverse phase HPLC, chromatography on silica or -exchange resin (e.g., ellulose), dialysis, chromatofocusing, gel filtration using protein A OSE® columns (cg, SEPHADEX® (1-75) to remove contaminants, metal chelating columns to bind epitope-tagged forms, and ultrafiltration/diafiltration (non- lirniting examples include centrifilgal filtration and tangential flow filtration (TFFD. inclusion of Viscosity-lowering agents at viscosity—reducing concentrations such as 0.010 M to 1.0 M, preferably 0.050 M to 0.50 M, most preferably 0.10 M to 0.30 M, allows a solution ofthe pharmaceutically active mAb to be purified and/or concentrated at higher mAb concentrations using common methods known to those skilled in the art, including but not limited to tangential flow filtration, centrifugal tration, and dialysis.
In some embodiments, iyophilized formulations of the proteins are provided and/or are used in the preparation and manufacture of the low- ity, trated protein formulations. In some embodiments, the pre— lyophilized protein in a powder form is reconstituted by dissolution in an formulation is filled into a aqueous solution. In this embodiment, the liquid c dosage unit container such as a vial or pre—filled mixing e, lyophilized, optionally with lyoprotectants, vatives, antioxidants, and other typical pharmaceutically acceptable excipients, then stored under sterile storage ions until shortly before use, at which time it is reconstituted with a defined volume of diluent, to bring the liquid to the desired concentration and viscosity.
The formulations described herein may be stored by any suitable method known to one d in the art. Non-limiting examples of methods for preparing the protein formulations for storage include freezing, lyophilizing, and spray drying the liquid protein formulation. In some cases, the lyophilized formulation is frozen for storage at subzero temperatures, such as at about °C or in liquid nitrogen. In some cases, a lyophilized or aqueous formulation is stored at 2-8°C. miting examples of diluents useful for reconstituting a lyophilized formulation prior to injection include sterile water, bacteriostatic water for injection (BWFI), a pH buffered solution (e.g., phosphate—buffered saline), sterile saline solution, Ringer's solution, dextrose solution, or aqueous solutions of salts and/or buffers. In some cases, the formulation is spray—dried and then stored.
IV. Administration to an Individual in Need Thereof The protein formulations, including, but not d to, reconstituted formulations, are administered to a person in need thereofby intramuscular, intraperitoneal (i.e., into a body cavity), erobrospinal, or subcutaneous injection using an 18-32 gauge needle (optionally a thin-walled needle), in a volume of less than about 5 mL, less that about 3 mL, preferably less than about 2 mL, more preferably less than about 1 mL.
The appropriate dosage (“therapeutically ive amount”) of the protein, such as a mAb, will depend on the condition to be treated, the severity and course of the disease or ion, Whether the protein is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the protein, the type of protein used, and the discretion of the ing physician. The protein is suitably stered at one time in single or multiple injections, or over a series of treatments, as the sole treatment, or in conjunction with other drugs or therapies.
Dosage formulations are designed so that the injections cause no significant signs of irritation at the site of injection, for example, wherein the primary irritation index is less than 3 when evaluated using a Draize scoring system. In an alternative embodiment, the ions cause macroscopically similar levels of irritation when compared to injections of equivalent volumes of saline solution. In another embodiment, the bioavailability of the protein is higher when compared to the otherwise same formulation without the viscosity-lowering agent(s) administered in the same way. In r embodiment, the formulation is at least approximately as effective pharmaceutically as about the same dose ofthe protein administered by enous on.
In a preferred ment, the formulation is injected to yield increased levels of the eutic protein. For example, the AUC value may be at least 10%, preferably at least 20%, larger than the same value computed for the otherwise same formulation without the viscosity-lowering agent(s) administered in the same way.
The viscosity-lowering agent may also affect bioavailability. For example, the t bioavailability of the protein may be at least 1.1 times, preferably at least 1.2 times the percent bioavailability of the otherwise same formulation without the ity-lowering agent(s) administered in the same way.
The Viscosity-lowering agent may also affect the pharmacokinetics.
For example, the CM after SC or IM ion may be at least 10%, preferably at least 20%, less than the CM of an approximately equivalent phannaceutically effective intravenously administered dose.
In some embodiments, the proteins are administered at a higher dosage and a lower frequency than the otherwise same formulations without the viscosity-lowering agent(s).
WO 38818 The lower viscosity ations require less injection force. For example, the ion force may be at least 10%, preferably at least 20%, less than the ion force for the otherwise same formulation without the viscosity-lowering agent(s) administered in the same way. In one embodiment, the injection is administered with a 27 gauge needle and the injection force is less than 30 N. The formulations can be administered in most cases using a very small gauge needle, for example, between 27 and 31 gauge, typically 27, 29 or 31 gauge.
The ity-lowering agent may be used to prepare a dosage unit formulation suitable for reconstitution to make a liquid pharmaceutical formulation for aneous or intramuscular injection. The dosage unit may contain a dry powder of one or more ns; one or more viscosity— lowering agents; and other excipients. The ns are present in the dosage unit such that after titution in a pharmaceutically acceptable solvent, the resulting formulation has a protein concentration from about 100 mg to about 2,000 mg per 1 mL (mg/mL). Such reconstituted formulations may have an absolute viscosity of from about 1 cP to about 50 cP at 25°C.
The low viscosity formulation can be provided as a solution or in a dosage unit form where the protein is lyophilized in one vial, with or without the Viscosity-lowering agent and the other ents, and the solvent, with or without the viscosity-lowering agent and other excipients, is provided in a second vial. In this embodiment, the solvent is added to the protein shortly before or at the time of injection to ensure uniform mixing and dissolution.
The viscosity-lowering agent(s) are present in the formulations at concentrations that cause no significant signs of toxicity andlor no irreversible signs of toxicity when administered via subcutaneous, intramuscular, or other types of injection. As used , “significant signs of toxicity” include intoxication, lethargy, behavioral modifications such as those that occur with damage to the central nervous system, infertility, signs of serious cardiotoxicity such as cardiac arrhythmia, cardiomyopathy, myocardial infarctions, and cardiac or congestive heart failure, kidney failure, liver failure, difficulty breathing, and death.
In preferred embodiments the formulations cause no significant irritation when administered not more than twice daily, once daily, twice weekly, once weekly or once monthly. The protein ations can be administered causing no significant signs of tion at the site of injection, as measured by a primary irritation index of less than 3, less than 2, or less than 1 when evaluated using a Draize scoring system. As used herein, “significant signs of irritation” include erythema, redness, and!or swelling at the site of injection having a diameter of greater than 10 cm, greater than 5 cm, or greater than 2.5 cm, necrosis at the site of injection, exfoliative dermatitis at the site of injection, and severe pain that prevents daily ty and/or es l attention or hospitalization. In some embodiments, injections of the protein formulations cause copically similar levels of irritation when ed to injections of equivalent volumes of saline solution.
The protein formulations can exhibit increased bioavailability compared to the ise same protein formulation without the viscosity- lowering agent(s) when administered via subcutaneous or intramuscular injection. “Bioavailability” refers to the extent and rate at which the bioactive species such as a mAb, s circulation or the site of action. The overall bioavailability can be increased for SC or IM injections as compared to the otherwise same formulations without the viscosity-lowering agent(s).
“Percent ilability” refers to the fraction of the stered dose of the bioactive species which enters circulation, as determined with respect to an intravenously administered dose. One way ofmeasuring the bioavailability is by comparing the “area under the curve” (AUC) in a plot of the plasma concentration as a function of time. The AUC can be ated, for example, using the linear trapezoidal rule. “AUCOD”, as used herein, refers to the area under the plasma concentration curve from time zero to a time where the plasma concentration returns to baseline levels. “AUCM”, as used herein, refers to the area under the plasma tration curVe from time zero to a time, t, later, for example to the time of reaching baseline. The time will typically be measured in days, although hours can also be used as will be apparent by context. For example, the AUC can be increased by more than %, 20%, 30%, 40%, or 50% as compared to the otherwise same formulation without the viscosity-lowering agent(s) and administered in the same way.
As used herein, “tum” refers to the time after administration at Which the plasma concentration reaches a maximum.
As used herein, “ max” refers to the maximum plasma concentration after dose administration, and before administration of a subsequent dose.
As used herein, "Cm-n" or "Cmugh" refers to the minimum plasma tration after dose administration, and before administration of a subsequent dose.
The Cmax after SC or IM injection may be less, for example, at least %, more preferably at least 20%, less than the Cmax of an intravenously administered dose. This reduction in Cm may also result in decreased toxicity.
The pharmacokinetic and pharmacodynamic parameters may be imated across species using approaches that are known to the skilled artisan. The pharmacokinetics and pharmacodynamics of antibody therapeutics can differ markedly based upon the specific antibody. An approved murine mAb was shown to have a half-life in humans of~ 1 day, while a human mAb Will typically have a half-life of ~ 25 days (Waldmann et at, Int. Immunol, 2001, 13 : 1551—1559). The pharmacokinetics and eodynarnics of antibody therapeutics can differ markedly based upon the route of administration. The time to reach maximal plasma concentration after IM or SC injection of lgG typically ranges from 2 to 8 days, although shorter or longer times may be encountered (Wang et at, Clin. Pharm. Ther., 2008, 84(5):548-558). The pharmacokinetics and pharmacodynamics of antibody eutics can differ markedly based upon the formulation.
The low-viscosity protein formulations can allow for greater flexibility in dosing and decreased dosing ncies compared to those protein formulations without the Viscosity—lowering agent(s). For example, by increasing the dosage stered per injection le-fold, the dosing frequency can in some embodiments be decreased from once every 2 weeks to once every 6 weeks. The protein formulations, ing, but not limited to, reconstituted formulations, can be administered using a heated and/0r self-mixing syringe or j ector. The protein formulations can also be ated in a separate warming unit prior to filling the syringe. i. Heated es The heated syringe can be a standard syringe that is pre-heated using a syringe warmer. The syringe warmer will generally have one or more gs each capable of receiving a syringe ning the protein formulation and a means for heating and maintaining the syringe at a specific (typically above the ambient) temperature prior to use. This will be ed to herein as a pre-heated syringe. Suitable heated syringe warmers e those available from Vista Dental Products and Inter-Med. The warmers are e of accommodating various sized syringes and heating, typically to within 1°C, to any temperature up to about 130°C. In some embodiments the syringe is pre-heated in a heating bath such as a water bather maintained at the desired temperature.
The heated syringe can be a self-heating syringe, i.e. capable of g and maintaining the liquid formulation inside the syringe at a specific ature. The self-heating syringe can also be a standard medical syringe having attached thereto a heating device. Suitable heating devices capable of being ed to a syringe include e heaters or syringe heater tape available from Watlow Electric Manufacturing Co. of St. Louis, MO, and syringe heater blocks, stage heaters, and in—line perfusion heaters available from Warner Instruments of Hamden, CT, such as the SW—61 model syringe warmer. The heater may be controlled through a central controller, e.g. the TC-324B or TC-344B model heater controllers available from Warner Instruments.
The heated syringe maintains the liquid protein formulation at a specified temperature or to within 1°C, within 2°C, or within 5°C of a specified temperature. The heated syringe can maintain the protein ation at any temperature from room temperature up to about 80°C, up to about 60°C, up to about 50°C, or up to about 45°C as long as the protein formulation is sufficiently stable at that temperature. The heated syringe can maintain the protein formulation at a temperature n 20°C and 60°C, between 21°C and 45°C, between 22°C and 40°C, between 25° C and 40° C, or between 25°C and 37°C. By maintaining the protein formulations at an elevated temperature during injection, the viscosity of the liquid ation is decreased, the lity of the protein in the formulation is increased, or WO 38818 both. _ 1 ii. Self—Mixing es 1 The syringe can be ixing or can have a mixer attached. The mixer can be a static mixer or a dynamic mixer. Examples of static mixers include those disclosed in US. Patent Nos. 5,819,988, 6,065,645, 6,394,314, 6,564,972, and 622. Examples of some dynamic mixers can include those disclosed in U.S. Patent Nos. 6,443,6l2 and 6,457,609, as well as U.S.
Patent Application Publication No. US 2002/0190082.The syringe can include multiple barrels for mixing the components of the liquid protein formulation. US. Patent No. 5,819,998 describes syringes with two barrels and a mixing tip for mixing two-component viscous substances. iii. Autoiniectors and Pre-ijiled Syringes of n Formulations The liquid protein formulation can be administered using a pre-filled syringe autoinj ector or a needleless injection device. Autoinj ectors e a handheld, often pen—like, cartridge holder for holding replaceable pre—filled cartridges and a spring based or analogous mechanism for subcutaneous or intramuscular injections of liquid drug dosages from a pre-filled cartridge.
Autoinj ectors are typically ed for self-administration or administration by untrained personnel. Autoinj ectors are available to dispense either single dosages or multiple dosages from a pre-filled cartridge. Autoinj ectors enable different user settings including inter alia ion depth, injection speed, and the like. Other injection systems can include those described in U. S.
Patent No. 8,500,681.
The lyophilized protein formulation can be provided in pre-filled or unit-dose syringes. US. Patent Nos. 3,682,174; 4,171,698; and 5,569,193 describe sterile es containing ambers that can be pre-filled With a dry formulation and a liquid that can be mixed immediately prior to injection. U.S. Patent No. 5,779,668 describes a syringe system for lyophilization, reconstitution, and stration of a pharmaceutical composition. In some embodiments the protein formulation is provided in lyophilized form in a pre-filled or unit-dose syringe, tituted in the syringe prior to administration, and administered as a single subcutaneous 0r intramuscular injection. Autoinj ectors for delivery of unit-dose lized drugs are described in W0 10,832. Auto injectors such as the Safe WO 38818 Click LyoTM (marketed by Future Injection Technologies, Ltd, Oxford, UK.) can be used to administer a unit-dose protein formulation where the formulation is stored in lyophilized form and reconstituted just prior to administration. In some embodiments the protein formulation is provided in unit-dose cartridges for lyophilized drugs (sometimes referred to as Vetter cartridges). Examples of suitable cartridges can include those described in U.S. Patent Nos. 5,334,162 and 5,454,786.
V. Methods of cation and Concentration The viscosity-lowering agents can also be used to assist in protein purification and concentration. The viscosity-lowering agent(s) and excipients are added to the protein in an effective amount reduce the viscosity of the protein solution. For example, the Viscosity-lowering agent is added to a concentration of between about 0.01 M and about 1.0 M, preferably between about 0.01 M and about 0.50 M, and most preferably between about 0.01 M and about 0.25 M.
The viscosity-lowering agent solution containing protein is then purified or concentrated using a method selected from the group consisting of ultrafiltration/diafiltration, tial flow filtration, centrifugal concentration, and dialysis.
Examples The foregoing will be r understood by the following non- lim‘iting examples.
All Viscosities of well-mixed aqueous mAb solutions were measured using either a mVROC uidic viscometer (RheoSense) or a DV2T cone and plate viscometer (Brookfield; “C & P”) after a 5 minute equilibration at 25°C (unless otherwise ted). The mVROC viscometer was equipped with an “A” or “B” chip, each manufactured with a 50 micron channel. Typically, 0.10 mL ofprotein solution was back-loaded into a gastight ab instrument syringe (Hamilton; 100 uL), affixed to the chip, and measured at multiple flow rates, approximately 20%, 40%, and 60% of the maximum re for each chip. For example a sample of approximately 50 GP would be measured at around 10, 20, and 30 nL/min ximately 180, 350, and 530 5'1, respectively, on an “A” chip) until viscosity stabilized, typically after at least 30 seconds. An average absolute viscosity and standard deviation was then calculated from at least these three measurements. The C & P viscometer was equipped with a CPE40 or CPESZ spindle (cone angle of 08° and 3.0°, respectively) and 0.50 mL samples were measured at multiple shear rates between 2 and 400 5'1.
Specifically, samples were measured for 30 seconds each at 22.58, 24.38, 26.25, 28.13, 30, 31.88, 45, 67.5, 90, 112.5,135, 157.5, 180, 202.5, 247, 270, 292.5, 315, 337.5, 360, 382, 400 3'], starting at a shear rate that gave at least % torque, and continuing until instrument torque reached 100%. An extrapolated zero-shear ity was then determined from a plot of c viscosity versus shear rate for the samples measured on a DV2T cone and plate viscometer. The extrapolated zero—shear viscosities reported are the average and standard deviation of at least three measurements.
Example 1: Effect of a viscosity-lowering agent, camphorsulfonic acid lysine (CSAL), on the viscosity of solutions of ilar ERBITUX® Materials and Methods A commercially—obtained biosimilar ERBITUX® (100-400 mg) containing pharmaceutical ents (Polysorbate 80, phosphate buffer, and NaCl) was purified. First, Polysorbate 80 was removed using DETERGENT-OUT® TWEEN® Medi Columns (G—Biosciences). Next, the resulting solutions were extensively buffer-exchanged into 20 mM sodium phosphate buffer (PB; pH 7.0) or 20 mM CSAL (pH 7.0) and trated to a final volume of less than 10 mL on Jumbosep centrifiigal concentrators (Pall Corp). The ted n solution was freeze-dried. The dried protein cakes, containing protein and buffer salts or agent, were reconstituted to a final volume of 0.15 - 1.3 mL. These samples were reconstituted using additional PB (pH 7.0) or CSAL (pH 7.0) sufficient to bring the final tration ofPB or CSAL to 0.25 M. The final concentration ofmAb in solution was determined by light absorbance at 280 nm. Reported n concentrations represent the range of all protein samples included in each Table or Figure. Specifically, ed values are the median plus or minus e range. Extrapolated zero-shear using an experimentally determined extinction coefficient of 1.4 L/g'cm and viscosities reported were ed on a DV2T cone and plate viscometer. 2014/055254 Results The data in Figure 1 demonstrate the viscosityulowering effect of CSAL on aqueous solutions ofbiosimilar ERBITUX®. The Viscosity of a solution of biosimilar ERBITUX® in phosphate buffer (PB) increases exponentially with increasing mAb concentration. The ity of a solution of biosimilar ERBITUX® in the presence of CSAL is seen to increase exponentially with increasing mAb concentration, but to a lesser extent than the ation in PB i.e. the ity gradient is reduced. The data in Figure 1 show that the higher the concentration of mAb, the greater ' the viscosity-lowering . The magnitude of viscosity-lowering s afforded by the replacement ofPB with CSAL varied from 1.1-fold at 100 i mg/mL to 10.3-fold at 227 i 5 mg/mL mAb.
Example 2: Viscosity-lowering effect of a Viscosity-lowering agent, camphorsulfonic acid lysine (CSAL), as a function of concentration of biosimilar AVASTIN® Materials and Methods A biosimilar AVASTIN® ed commercially and containing pharmaceutical excipients (Polysorbate 20, phosphate buffer, citrate buffer, mannitol, and NaCl) was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 1.7 Mg cm at 280 nm). The protein was formulated to contain either 0.25 M phosphate buffer or 0.25 M CSAL.
Results Figure 2 depicts the ity of aqueous mAb solutions as a function ofmAb concentration in aqueous buffered solution and with CSAL. The viscosity of biosimilar N® in aqueous phosphate buffer and in the presence of CSAL increases exponentially with increasing concentration; however, as in the case of ilar X®, this increase is much less marked for the CSAL—containing formulation, i.e. the viscosity gradient is reduced. In general, the higher the mAb concentration, the greater the viscosity-lowering effect ed. The magnitude of viscosity-lowering effects afforded by the replacement of PB with CSAL varied from 2.1-fold at 80 mg/mL to 3.7-fold at 230 i 5 mg/mL mAb.
Example 3: Viscosity-lowering effect as a function of CSAL concentration for aqueous solutions of biosimilar ERBITUX® Materials and s Samples were purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed similarly to Example 1 above. The final concentration of CSAL upon reconstitution in an aqueous CSAL solution ranged from 0.25 M to 0.50 M.
Results Table 1 shows the Viscosity of solutions of biosimilar ERBITUX® formulated in 0.25 M phosphate buffer (no CSAL as a l) and with g concentrations of CSAL. The viscosity-lowering effect of CSAL is seen to rise from 8.4- to 12.1-fold with increasing viscosity-lowering agent concentration. The data in Table 1 ShOW that the higher the concentration of CSAL, the greater the Viscosity-lowering effect, at least Within the agent concentration range tested.
Table I. Viscosities of aqueous solutions of biosimilar ERBITUX® (155 i 5 mglmL, pH 7.9) in the presence of different concentrations of CSAL at 25°C.
Fold Viscosity ion red to no CSAL [CSAL], M Viscosity, cP present) 0 154 d: 0 l 0.25 18.3 i 0.0 8.4 0.38 14.9 $0.1 10.3 0.50 12.7 i 0.1 12.1 Example 4: Viscosities of solutions of biosimilar ERBITUX® as a on of temperature in the presence of various viscosity—lowering agents Materials and Methods Aqueous solutions ofbiosimilar ERBITUX® containing various viscosity-lowering agents were prepared as described in Example 1.
Specifically, 20 mM solutions ofthe ity-lowering agents of interest were used for buffer ge, and the lyophilized cakes were reconstituted to 0.25 M of each viscosity-lowering agent. For the sample containing CSA— APMI, biosimilar ERBITUX® was extensively buffer exchanged into 2 mM PB (pH 7.0), and concentrated to a final volume of less than 10 mL on Jumbosep centrifugal concentrators (Pall Corp). The sample was first aliquoted. Then, an appropriate amount of CSAAPMI solution (pH 7.0) was added to each aliquot such that upon reconstitution with water, the final excipient concentration is 0.25 M. The protein solutions were then freeze- dried. The dried protein cakes, containing protein and ity-lowering agent (and a negligible amount of buffer salts) were reconstituted to a final volume of approximately 0.10 mL and ity—lowering agent concentration as previously described.
Results Table 2 shows viscosity data for biosimilar ERBITUX® in the ce of six ity-lowering agents — cainphorsulfonic acid iysine (CSAL), camphorsulfonic acid arginine (CSAA), esulfonic acid lysine (BSAL), benzenesulfonic acid arginine (BSAA), naphthalenesulfonic acid arginine (NSAA), and camphorsulfonic acid 1-(3-aminopropyl)methyl— 1H—imidazole (CSAAPMI). The data in Table 2 Show a reduction in viscosity of at least about 9—fold for all six ity-lowering agents compared to a solution of biosimilar X® in phosphate buffer under otherwise the same conditions. The most efficacious viscosity-lowering agent — CSAAPMI — lowered viscosity by >40-fold. onally, the data in Table 3 show that at multiple temperatures ranging from 20°C to 30°C, a 225 mg/mL solution of biosimilar ERBITUX® prepared with 0.25 M CSAA had the lowest Viscosity ofthe five viscosity- lowering agents. Thus, the observed trends in Viscosities at 25°C seem to be predictive of those at temperatures of at least 20°C and 30°C.
Table 2. Reduction in viscosity of aqueous solutions of ilar ERBITUX® (226 i 6 mg/mL, pH 7.0) formulated with various 0.25 M viscosity-lowering agents, as ed to that in 0.25 M sodium phosphate buffer (PB) at 25°C.
Agent Viscosity, cP Fold reduction PB 1130 d: 7 1 CSAL 109-3: 1 10.4 CSAA 58.0 d: 0.3 19.5 BSAL 126 :i: 1 9.0 BSAA 61.3 :1: 0.9 18.4 NSAA 69.4 i 0.6 16.3 CSAAPMI 25.7 i 1.5 44.0 Table 3. Viscosities of aqueous solutions of biosimilar ERBITUX® (225 i mg/mL, pH 7.0) formulated with various 0.25 M viscosity-lowering Viscosity, cP Temp. . Agent PB CSAL CSAA BSAL BSAA NSAA 1810i 796i . 85.2 :I: °C 10 166 :I: 2 0.9 193 i 0 0.6 103 :I: 0 1130i 58.0i 61.3 i 69.4 i °C 7 109 ii 0.3 126 $1 0.9 0.6 46.9 :t: 50.5 :I: 60.9 i °C 723 :t 0 78.4 i 1.5 0.6 89.8 A: 0.8 1.9 4.3 Example 5: The effect of temperature on viscosity of aqueous solutions of biosimilar AVASTIN® formulated with various viscosity-lowering agents Materials and Methods Solutions of biosimilar AVAST1N® containing different viscosity- lowering agents were prepared as described in Example 1 above. In particular, 20 mM solutions of the viscosity-lowering agents of interest were used for buffer ge, and the lyophilized cakes were reconstituted to 0.15 or 0.25 M viscositynlowering agent.
Results As seen in Table 4, 0.25 M CSAL lowered the ity of a 230 i 5 mg/mL on of biosimilar N® at all three temperatures between and 30°C. Furthermore, 0.15 M CSAL reduces viscosity to approximately the same absolute value as 0.25 M CSAL at 20 and 25°C and is equally effective at 30°C.
The data in Table 5 compare the effects of CSAL and BSAL at a tration of 0.15 M. CSAL is a superior ity—lowering agent compared to BSAL at all three temperatures.
Table 4. ities of aqueous solutions of biosimilar AVASTIN® (230 i mg/mL, pH 7.0) formulated with 0.25 and 0.15 M CSAL at different temperatures.
Viscosity, cP Temperature 0.25 M PB 0.25 M CSAL 0.15 M CSAL °C 563i2 152i0 157i0 °C 397i2 107i4 113i0 °C 311i4 95.5i5.4 91.7133 Table 5. Viscosities of aqueous solutions of biosimilar AVASTIN® (230 i mg/mL, pH 7.0) formulated with 0.15 M CSAL and BSAL at ent temperatures.
Viscosity, cP Temperature 0.25 M PB 0.15 M CSAL 0.15 M BSAL °C 56312 157i0 395i3 °C 397i2 113i0 227i5 °C 311d:4 91.7i3.3 175:1:7 Example 6: Removal of CSAL reverses viscosity-lowering effect in mAb solutions Materials and Methods Three samples each ofbiosimilar ERBITUX® and biosimilar AVASTIN® were prepared. First, Polysorbate was removed from the commercially obtained mAb solutions. The resulting on With remaining pharmaceutical excipients was either (i) concentrated on a fugal device with a lOO-kDa molecular weight cutoff (MWCO) (Pall Corp.) as a control sample (original excipients), (ii) buffer exchanged into 0.25 M CSAL as described in Example 1, or (iii) buffer exchanged into 0.25 M CSAL as described in e 1, reconstituted, and then r exchanged into 0.25 M PB. In this third instance, exchange into 0.25 M phosphate buffer proceeded first by overnight dialysis against 20 mM PB (50-kDa MWCO, Spectrum Labs). The lly dialyzed samples were then diluted to 60 mL in 0.25 M PB and subjected to centrifugal concentration (30-kDa MWCO Jumbosep (Pall Corp), ed by a lOO-kDa MWCO Macrosep device (Pall Corp.)). The viscosities of these three aqueous solutions were determined as described in Example 1 above.
Results The viscosities of aqueous solutions of both biosiinilar ERBITUX® and biosimilar AVASTIN® decreased in the presence of CSAL — 2.7- and 1.5—fold, respectively — but then increased when CSAL was removed (see Tables 6 and 7). Furthermore, upon removal of CSAL, mAb solution viscosities returned to approximately the same level as the original solutions, ting that CSAL does not damage the protein and showing that it is ary for the observed Viscosity ion.
Table 6. Viscosities of aqueous solutions of biosimilar X® (80 i mg/mL, pH 7.0) at 25°C.
Formulation Viscosity, cP Original excipients 8.30 :l: 0.04 0.25 M CSAL 3.08 a 0.18 0.25 M CSAL exchanged into 0.25 M PB 9.43 d: 0.04 Table 7. Viscosities of aqueous ons of biosimilar AVASTIN® (101 i mg/InL, pH 7.0) at 25°C.
Formulation Viscosity, cP Original excipients 6.08 d: 0.19 0.25 M CSAL 4.03 :I: 0.24 0.25 M CSAL ged into 0.25 M PB 6.61 i 0.08 Example 7: rsulfonic acid-containing viscosity-lowering agents provide large viscosity reductions in aqueous solutions ofAVASTIN® and biosimilar AVASTIN® Materials and Methods AVASTIN® and a biosimilar AVASTIN® obtained commercially and containing ceutical excipients (AVASTIN®: trehalose, sodium phosphate buffer, and Polysorbate 20; biosimilar AVASTIN®2 Polysorbate , phosphate buffer, citrate buffer, mannitol, and NaCl) were purified, buffer exchanged, trated, freeze-dried, and reconstituted as bed above. Samples in Table 8 were ed as described in Example 1 above (using the protein extinction coefficient of 1.7 L/g-cm at 280 nm) and measured on a C & P viscometer. Viscosity-reduced samples in Table 9 were prepared as described in Example 4 above, but mAb was extensively buffer exchanged into 2 mM PB. Subsequently, the appropriate amount of viscosity-lowering agent was added to result in a final viscosity-lowering agent concentration of 015—03 5 M upon reconstitution. Viscosities were ed using a RheoSense mVROC uidic viscometer equipped with an “A” or "B” chip. Results The data in Tables 8 and 9 demonstrate the viscosity-lowering effect of different viscosity-lowering agents 011 aqueous solutions of biosimilar AVASTIN®. Viscosity reductions up to 25-fold (compared to mAb solutions in PB) are ed for aqueous solutions of biosimilar AVASTIN® in the presence of viscosity-lowering agents containing CSA.
Table 8. Viscosities of aqueous solutions of biosimiar AVASTIN® (200 :i: mg/mL, pH 7.0) at 25°C with various viscosity-lowering agents.
Agent [Salt] (M of anion) Viscosity (cP) PB 0.25 96.8 i 0.9 NaCl 0.25 121 i 8 Arginine-HCI 0.25 83.2 i 2.8 Argininc-HCI 0.3 71.8 d: 2.2 Lysine-HCI 0.25 137 d: 2 BSA sodium salt 0.25 133 d: 3 CSA sodium salt 0.25 55.7 i 0.2 BSAA 0.25 75.3 d: 0.4 Bcnzoic acid arginine 0.15 52.2 i 0.5 Benzoic acid nc 0.25 51.4 d: 0.5 CSAA 0.25 48.5 i 1.9 CSA betaine* 0.25 66.0 i 0.7 diCSA cadaverine 0.25 85.5 i- 5.2 diCSA cadavcrine 0.35 65.6 i 1.6 CSA canavanine 0.15 60.5 i 0.6 CSA nine 0.25 75.6 i 3.0 CSA camitine* 0.25 72.4 :J: 1.7 CSA dimethylpiperazinc 0.25 47.4 i 1.3 CSA dimethylpiperazine 0.35 51.7 i 0.9 CSAL 0.25 54.9 :I: 0.9 Chlorotheophyllinc arginine 0.25 104.5 i 6.5 Ethandisulfonate nine* 0.15 77.1 d: 0.3 Ethandisulfonate diarginine* 0.25 105 i 4 MSA arginine 0.25 93.1 d: 0.9 Toluenesulfonic acid arginine 0.25 159 i 5 Toluenesulfonic acid lysine 0.25 118 i l * Contains equimolar NaCl; CSA = Camphorsulfonic acid, BSA = Benzenesu1fonic acid, MSA = Methanesulfonic acid, PB = Phosphate buffer Table 9. Viscosities of aqueous solutions of biosimilar AVASTIN® (pH 7.0) at 25°C with 0.15 M viscosity—lowering agents s ise noted). milar .
Agent AVASTIN] Viscosity (0P) (mg/mL) 0.25 M PB 220 213 i 10 0.25 M PB 200 96.8 :1: 0.9 CSA—piperazine 212 64.5 :1: 13.1 Lactobionic acid-tris 219 109 d: 5 CSA-4—aminopyridine 229 86.4 i 1.1 Glucuronic acid-tris 221 151 i 5 The viscosity of a 200 i 9 mg/mL aqueous solution of biosimilar AVASTIN® with CSAA was measured as a on of pH as depicted in Figure 3. As pH increases, the magnitude of the Viscosity-lowering effect resulting from the presence of CSAA in aqueous solutions of biosimilar AVAST1N® also increases, reaching a minimum Viscosity and maximum viscosity-lowering effect around pH 7. The Viscosity reduction by CSAA was compared as a function ofpH for two different concentrations of biosimilar AVASTIN®. Figure 4 demonstrates that 0.25 M CSAA s in a greater reduction in viscosity with increasing (i) tration of the biosimilar AVASTIN® and (ii) pH.
Table 10 es the viscosity reduction of biosimilar AVASTIN® to that of branded AVASTIN® with and without CSAL. The branded AVASTIN® solution has a muchhigher viscosity than a solution ofthe biosimilar mAb in the absence ofthe agent. However, the presence of 0.25 M CSAL results in a 1.8- and 33-fold reduction in viscosity of the ilar and branded AVASTIN® respectively; the viscosities of biosirnilar and d AVASTIN® are seen to be similar in the presence of 0.25 M CSAL.
Table 10. ities of aqueous solutions ning 205 :I: 5 mg/mL of biosimilar N® or branded AVASTIN® with or without 0.25 M .
CSAL measured at 25°C and pH 7.0.
Salt Biosimilar AVASTIN® Branded AVASTIN® (GP) (9P) Phosphate Buffer 96.8 d: 0.9 154 i 4 0.25 M CSAL 54.9 :I: 0.9 46.7 d: 0.9 CSAL = camphorsulfonic acid lysine As demonstrated in Table 11, CSA l-(3-aminopropyl)-2—methyl-1H- imidazole (CSAAPMI) with HCl provides superior viscosity reduction than CSAL, reducingr the viscosity more than 5—fold as compared to the PB control for a solution of 210 mg/mL biosimilar AVASTIN®.
Table 11. Viscosities of aqueous solutions of biosimilar AVASTIN® with s viscosity~lowering agents at 25°C and pH 7.0.
Agent [Agent], M [23:13:] ’ Viscosity, cP PB 0.25 220 213:1:10 CSAL 0.25 210 63.0 d: 1.8 I—ZHCI 0.25 210 40.9 d: 0.5 APMI = 1-(3—amjnopropyl)methyl-1H—imidazole For a solution containing ~ 230 mg/mL biosimilar AVASTIN®, Table 12 demonstrates Viscosity reduction of approximately 5—fold with sulfosalicylic ontaining viscosity-lowering agents as well as for CSAAPMI and CSA ne.
Table 12. Viscosities of aqueous solutions containing 228 :l: 5 mg/mL biosimilar AVASTIN® with viscosity-lowering agents at 25°C and pH 7.0.
Agent Viscosity Agent Concentrati (UP) on [M] PB 0.25 397 :l: 2 CSAA 0.25 116i2 CSAL 0.25 113 d: 0 Sulfosalicylic acid diarginine 0.15 81.6i1.7 Sulfosalicylic acid dilysine 0.25 73.45204 CSAAPMI—ZHC] 0.25 71.8i3.2 CSAthiamine- 2NaCl 0.15 83.721: 2.2 APMI = 1-(3—aminopropyl)methyl-1H—imidazole; CSA = camphorsulfonic acid Example 8. The effect of Viscosity—lowering agents on aqueous solutions of ERBITUX® and biosimilar ERBITUX® Materials and Methods Aqueous solutions of biosimilar and branded ERBITUX® ning various Viscosity-lowering agents were ed as described in Example 1.
Specifically, 20 mM solutions ofthe salts of interest were used for buffer exchange, and the lyophilized cakes were reconstituted to contain 0.25 M of each agent. ViSCosities were measured using either a RheoSense mVROC microfluidic viscometer equipped with an “A” or "B” chip or a DVZT cone and plate viscometer.
Results Table 13 shows data for biosimilar ERBITUX® (222 i 5 mg/mL) in the presence of five Viscosity-lowering agents: CSAA, CSAL, BSAA, BSAL, and NSAA. Table 14 es the Viscosity reduction of biosimilar ERBITUX® solutions using CSAA and CSAL to arginine or lysine alone.
Table 13. ities of aqueous solutions of biosimilar ERBITUX® (222 :l: 5 mglmL, pH 7.0) with 0.25 M Viscosity-lowering agents at 25°C.
Agent Viscosity (cP) Fold Reduction ate Buffer 1130 i 7 1.0 CSA Arginine 52.5 d: 1.0 21.5 CSA Lysine 109 :1: 1 10.4 BSA Arginine 53.4:t: 5.5 21.2 BSA Lysine 126 i 1 9.0 NSA Arginine 69.4 :t 0.6 16.3 Table 14. ities of aqueous solutions of biosimilar ERBITUX® (222 :1: 5 mg/mL, pH 7.0) with 0.25 M viscosity—lowering agents at 25°C.
Agent Viscosity (cP) Fold Reduction Phosphate Buffer 1130 i 7 1.0 CSAA 52.5 :1: 1.0 21.5 CSA Sodium 393 i 14 2.9 Arginine HCl 45.3 :1: 0.5 24.9 CSAL 109 i 1 10.4 Lysine HCI 128 i 2 8.8 The data in Table 13 Show a ion in ity of at least 90-fold for all five Viscosity-lowering agents compared to an aqueous solution of biosimilar ERBITUX® in phosphate buffer under otherwise the same conditions. The most efficacious Viscosity—lowering agents, CSAA and BSAA, lowered the solution Viscosity some 21-fold. The viscosities of aqueous solutions of biosimilar ERBITUX® containing 0.25 M CSAA were compared as a function ofpH at varying protein concentrations. Figure 5 demonstrates thata Viscosity minimum is observed around pH 7.0 for all protein concentrations. The effect ofpH on viscosity is most nced for higher protein trations (253 mg/mL in the example).
As seen in Table 15, the s solutions of biosimilar and branded ERBITUX® have similar viscosities in the presence ofthe arginine salt BSAA at 0.25 M .
Table 15. Viscosities of 224 :l: 4 mg/mL aqueous solutions of biosimilar ERBITUX® or branded X® with or Without 0.25 M BSAA at °C and pH 7.0.
Agent Biosimilar ERBITUX® Branded ERBITUX® Viscosity (0P) Viscosity (0P) Phosphate Buffer 1130 i: 7 556 i- 20 0.25 M BSAA 53.4 d: 5.5 44.1 i 0.5 The impact of the Viscosity-lowering agents on the formation of irreversible protein aggregates was examined for biosimilar ERBITUX®.
Aqueous liquid formulations were prepared of (i) biosimilar ERBITUX® and .(ii) ilar ERBITUX® containing 0.25 M CSAL. These ons were stored for 90 days at 4°C and pH 5.4 and 7.0, tively. The stored samples were examined using size exclusion tography (column: Tosoh TSngl W Aggregate; mobile phase: 0.1 M potassium phosphate/0.1 M sodium sulfate, pH 6.8 at 0.8 mL/min; injection: 20 pL of a mg/mL mAb solution). The data in Table 16 reveal no significant aggregate formation in either the cial drug product or high- concentration viscosity~lowered formulation.
Table 16. Percentage of protein aggregate formation after 90 days of storage at 4°C as measured by size exclusion chromatography for aqueous solutions containing of biosimilar ERBITUX® with or without 0.25 M CSAL.
Sample % Monomer % Dimer % Aggregate Biosimilar ERBITUX® 5 mg/mL 99.0 1.0 ' 0.0 Biosimilar ERBITUX® 210 mg/mL 98.4 0.9 0.7 with 0.25 M CSAL e 9. The effect of viscosity—lowering agents on aqueous solutions of DE® Materials and Methods Commercially-obtained REMICADE® containing pharmaceutical excipients (sucrose, Polysorbate 80, sodium phosphate buffer) was prepared as per instructions in the ibing information sheet. uently, the aqueous drug product was d, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 1.4 L/g*cm at 280 nm). Viscosities were measured using either a RheoSense mVROC microfluidic viscometer equipped with an “A” or “B” chip.
Results The data for aqueous REMICADE® solutions in Table 17 demonstrate that (i) visco sity—lowering agents containing a bulky cyclic group e greater than 15-fold viscosity reductions, and (ii) CSAA, CSAAPMI, and sulfosalicylic acid diarginine (SSA DiArg) e the greatest ity reduction of about 29-fold. Solution viscosities in the presence of ArgHCl alone are significantly higher than those with the bulky cyclic groups.
Table 17. Viscosities of aqueous solutions of REMICADE® containing 0.25 M viscosity-lowering agents at 25°C and pH 7.0.
[REMICADE®]_Viscosity (cP) (mg/mL) ArgHCl CSAA CSA BSAA CSAL SSA APMI DiArg 1557 486 i 53.7 :1: 56.3 4 92.3 i 95.3 i 222% 559$ 1.8 i 22 34 9.3 2.7 1.4 1.1 5132b 19.1 d: 31.721: 26.7:E 27.43: 166i4 110:1:1 27.1i0.3 0.2 0.3 1.2 0.2 PB = phosphate buffer; ArgHCl = argim'ne HCI; CSAA = camphorsulfonic acid arginine; CSA APMI = camphorsulfonic acid l-(3 -aminopropyl)methyl—1H- imidazole; BSAA = benzene sulfonic acid arginine; CSAL = orsulfonic acid lysine; SSA DiArg Z sulfosalicylic acid di—arginine.
The dependence of the viscosity reduction on the agent concentration was examined for aqueous solutions ofREMICADE® in the presence of CSAA. The results presented in Table 18 demonstrate that viscosity reduction increases with increasing agent concentration. The Viscosity reduction, for example, is more than twice as large (the viscosity is less than halt) with 0.35 M agent as compared to 0.20 M agent.
Table 18. Viscosity of an aqueous solution of REMICADE® (215 :l: 5 mglmL) in the presence of various trations of CSAA measured at °C and pH 7.0.
, (M) Viscosity (0P) 0 1557 2k 22 0.20 81.3 :I: 1.0 0.25 53.7 a: 9.3 0.35 38.2 :t: 0.9 Biophysical properties of solutions CADE® formulated with 0.25 M CSAA were assessed over 90 days. Samples ofREMTCADE® formulated with 0.25 M CSAA were prepared as described in Example 1 above. As seen in Table 19 and Figure 6, the monomer t of trated ons of REMICADE® in 0.25 M CSAA as determined by size exclusion chromatography (Tosoh TSngl UltraSW Aggregate column; 0.1 M potassium phosphate/0.1 M sodium sulfate buffer pH 6.8 at 0.8 mL/mjn; 20 uL injection of ~4.5 mg/mL solutions), is similar to the drug product at all time points and no detectable ation is observed after storage for 100 days at 4°C. The viscosity, as measured using a microfluidic viscometer, was demonstrated to remain stable after storage for 30 days at 4°C (Table 20). Additionally, antigen binding of this processed REMICADE® protein was ed with a REMICADE®-specific ELISA assay and no decrease in binding was seen between days 0 and 100 (Table ). Similarly, the monomer content (Table 21) and antigen binding (normalized to that of the drug t, Table 22) of concentrated solutions ofREMICADE® in 0.25 M CSAA are comparable to the drug t after 1 week of storage at room temperature. Lastly, Table 23 demonstrates that storage of a lyophilized cake containing CSAA at 4°C for 75 days has no negative effects on the solution Viscosity or extent of protein aggregation when the sample is tituted. The s in Tables 19-23 and Figure 6 demonstrate the biophysical stability ofREMICADE® formulated With CSAA before and after storage for at least 100 days at 4°C.
Table 19. N0 increased ation red to drug product) is observed in an aqueous solution of REMICADE® (227 mg/mL, pH 7) after formulation with 0.25 M CSAA and storage at 4°C.
Day % monomer Drug Product 99.9 i 0.03 0 99.7 i 0.07 99.7 i 0.04 100 . 99.9 i 0.1 Table 20. Reduced viscosity and antigen binding are retained over time in an aqueous solution of REMICADE® (227 mg/mL, pH 7) after formulation with 0.25 M CSAA and storage at 4°C.
Viscosity % binding (0P) (ELISA) 0 65.2 a 0.7 105 i 14 62.2 :I: 1.4 98 :i: 12 100 nd. 101 i 5 Table 21. No increased aggregation (compared to drug product) is observed in an aqueous solution of REMICADE® (219 mg/mL, pH 7) after formulation with 0.25 M CSAA and storage at room temperature. % monomer Drug Product (Elia/1 D 99.7 i 0.1 99.9 :J: 0.1 4 99.9 d: 0.1 97.9 i O 7 100 i 0 100 i 0 Table 22. Antigen binding persists in an aqueous solution of REMICADE® (219 mg/mL, pH 7) after formulation with 0.25 M CSAA and storage at room temperature. % binding (normalized to drug t) Drug Product }: 0 1002!: 12 88.6i5.2 7 100:1:28 “43:22.4 Table 23. RENEGADE® stored as a lyophilized powder retains low viscosity and monomer content upon reconstitution after e at 4°C for 75 days Storage time (days) ity, cP % Monomer (SEC) 0 65.2i0.7 99..7:|:0.1 75 59.3i1.0 98.9:|:0.1 WO 38818 Example 10. The effect of Viscosity-lowering agents on aqueous solutions of HERCEPTIN® Materials and Methods Commercially-obtained HERCEPTIN® containing pharmaceutical excipients (histidine buffer, trehalose, Polysorbate 20) was prepared as per instructions in the prescribing information sheet. Subsequently, the aqueous drug product was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 1.5 L/g*cm at 280 nm). Viscosities were measured using a RheoSense mVROC microfluidic viscometer ed with an “A” or “B” chip.
The data ted in Table 24 show that the Viscosity of an aqueous solution of HERCEPTIN® containing viscosity-lowering agents - compared to those containing PB - is lowest in the presence of CSAA. At higher n trations (i.e. >250 Ing/mL) Arginine HCl alone reduces Viscosity significantly and CSA further enhances the effect.
Table 24. Viscosities of aqueous solutions of HERCEPTIN® containing 0.25 M salts at 25°C and pH 7.0.
[HERCEPTIN®] Viscosity (0P) (mg/mm PB ArgHCl CSAA BSAA 270% 40014 7 96.7:t4.7 115m 254i3 172i5 116i24 78.04: 8.7 75.44: 5.0 216 :i: o n.d. 44.8 $1.1 55.7 :I: 2.3 n.d.
PB = phosphate buffer; ArgHCl = arginine HCl; n.d. = not determined Example 11. The effect of viscosity-lowering agents on aqueous solutions of TYSABRI® Materials and Methods Commercially-Obtained TYSABR1® containing pharmaceutical excipients (sodium phosphate , sodium chloride, Polysorbate 80) was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in e 1 above (using the extinction coefficient of 1.5 L/g*cm at 280 nm). Viscosities were measured using a RheoSense mVROC microfluidio viscometer equipped with an “A” or “B” chip.
Results The data presented in Table 25 show that the ity ion of an aqueous solution of TYSABRI® containing viscosity~lowering agents is approximately 25-fold (compared to solution containing PB) near 276 mg/mL protein.
Table 25. Viscosities of aqueous solutions of TYSABRI® containing 0.25 M viscosity-lowering agents at 25°C and pH 7.0.
[TYSABRI®] ity (cP) (mg/mL) PB ArgHCl CSAA BSAA 276 :t: s 255 d: 5 97.2 :t: 5.7 92.9 :1: 2.6 n.d. 237 a 4 182 :l: 5 52.3 a: 4.5 47.1 :I: 2.1 n.d. 230 :I: 2 n.d. 37.0 a 0.1 n.d. 34.9 :I: 1.3 PB = phosphate ; ArgHCI : arginine HCl; n.d. = not determined. e 12. The effect of viscosity-lowering agents on aqueous solutions of biosimilar RITUXAN® Materials and Methods cially-obtained biosimilar RITUXAN® containing phantnaceutical excipients (citrate buffer, sodium chloride, and TWEEN® 80) was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 1.7 L/g*cm at 280 nm). ities were measured using a RheoSense mVROC microfluidie viscometer equipped with an “A” or “B” chip.
Rails The data presented in Table 26 Show that the Viscosity reduction for an s solution of biosimilar R1TUXAN® containing ity—lowering agents is over 13-fold at approximately 213 mg/mL protein and over 5-fold at approximately 202 mg/mL, compared to the mAb formulated in PB.
Table 26. Viscosities of aqueous solutions of biosimilar RITUXAN® with viscosity-lowering agents at 25°C and pH 7.0.
Arg SSA SSA CSA CSA CSA CSAA HCl diArg diAPMI Na APMI DMP 0.45 0.25 0.25 M 0.25 M 0.25 M 0.25 M 0.25 M M M 86.8i 533$ 45m: 211i2 103% 76.13: 78.421: ' ' . . 1.3 0.3 * [RITUXAN®] is 220 mg/mL DMP = dimethylpiperazine Example 13. The effect of viscosity-lowering agents on aqueous solutions of IX® Materials and Methods Commercially-obtained VECTIBIX® containing pharmaceutical excipients was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 1.25 L/g*cm at 280 nrn). ities were measured using a nse mVROC microfluidic viscometer equipped with an “A” or “B” chip.
Rinks The data presented in Table 27 show that the Viscosity reduction of an aqueous solution of VECTIBIX® containing Viscosity-lowering agents is approximately 2-fold at 291 mg/mL and 3-fold at 252 mg/mL, compared to solutions with PB but no viscosity—lowering agents.
Table 27. Viscosities of aqueous solutions of VECTIBIX® with 0.25 M viscositynlowering agents at 25°C and pH 7.0.
BIX®] ity (0P) (mg/mm ' PB ArgHCl CSAA 291 :b 3 32s :I: 12 n.d. 162 3:1 264 n.d mi. 44.3 3: 2.3 252 a 3 80.3 :I: 3.3 36.2 a 1.0 27.4 3: 1.2 233i4 38.73: 1.8 24.73: 1.3 26.2i6.5 Example 14. The effect of viscosity-lowering agents on aqueous solutions of ARZERRA® als and Methods Commercially-obtained ARZERRA® containing pharmaceutical ents was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 1.5 L/g*cm at 280 nm). Viscosities were measured using a RheoSense mVROC microfluidic viscometer equipped with an “A” or “B” chip.
Results ' The data presented in Table 28 show that the viscosity reduction of an s solution ofARZERRA® containing viscosity-lowering agents is approximately 3-fold at 274 mg/mL and 2-fold at 245 mg/mL, compared to solutions with PB but no viscosity-lowering agents.
Table 28. Viscosities of aqueous ons of ARZERRA® with 0.25 M viscosity—lowering agents at 25°C and pH 7.0.
[ARZERRA® ] Viscosity (0P) (mg/mL) PB CSAA CSAAPMI 274 :i: 10 349 a 2 125 :l: 7 98.9 :I: 0.7 245 :l: 4 120 :i: 4 n.d. 53.6 :I: 0.6 Example 15. Comparison of Different Methods for Measuring Viscosity Materials and Methods Aqueous solutions containing 220 mg/mL DE® and 0.25 M CSAA were prepared as bed above Example 1. The viscosities at 25°C and pH 7.0 are reported in Table 29 as extrapolated zero-shear viscosities from cone and plate viscometer measurements and as absolute ities measured with a microfluidic viscometer. The cone and plate measurements used a DV2T cone and plate viscometer (Brookfield) equipped with a CPE40 or CPESZ spindle measured at multiple shear rates between 2 and 400 5'1.
An extrapolated zero-shear viscosity was determined from a plot of absolute viscosity Versus shear rate. The microfluidic viscometer measurements were performed using a RheoSense mVROC microfluidic viscometer equipped with an “A” or “B” chip at multiple flow rates (approximately 20%, 40%, and 60% of the maximum pressure for each chip).
Results The data in Table 29 demonstrates that the absolute Viscosities from the microfluidic eter can be directly compared to the extrapolated zero-shear ities determined from the cone and plate viscometer.
Table 29. Viscosities of s solutions of REMICADE® (220 mg/mL) with 0.25 M CSAA at 25 °C and pH 7.0 measured on two different viscometers.
Instrument Viscosity (0?) Cone and plate 62.3 :1: 0'1 viscometer (C&P) Microfluidic eter 53 7 i. 9 3. on a chip (mVROC) In order to compare a broader range of Viscosities and protein concentrations, aqueous solutions of a model antibody, bovine gamma globulin, were prepared with and Without 0.25 M CSAL. The viscosities were measured as described above at protein concentrations ranging from 110 mg/mL to 310 mg/mL. The data presented in Table 30 demonstrates that the absolute viscosities from the microfluidic viscometer can be directly compared to the olated zero—shear viscosities for both low and high viscosity n solutions.
Table 30. Viscosities of aqueous gamma globulin solutions with and without 0.25 M CSAL at 25°C and pH 7.0 measured on two ent eters.
Viscosity (cP) [gamma globulin] without CSAL With CSAL (mg/mm microfiuIdlc. . . C & P microfluldic. . .
C & P 110 3.81s: 0.19 2.66 :t 0.01 n.d. n.d. 170 12.0i0.6 11.0i0.1 10.3 :t 1.0 10.6i0.1 260 167 3:1 161 :1: 1 93.5 $1.2 85.3 i 0.3 310 399i1 377322 223i1 203i2 Example 16. Viscosity—lowering agents show no signs of toxicity when injected subcutaneously Materials and Methods Thirty 11-week old euDaney rats were separated into 6 groups of 5 rats each (3 saline control groups and 3 CSAA groups). The rats - ~ - were injected subcutaneously with 0.5 mL of either endotoxin—free phosphate—buffered saline or endotoxin—free 0.25 M CSAA according to the following schedule: One group from each condition was injected once on day 1 and then sacrificed 1 hour later; one group from each condition was injected once on day 1 and once on day 2 and then sacrificed 24 hours after the second injection; and one group from each ion was injected once on day 1, once on day 2, and once on day 3, and then sacrificed 24 hours after the third injection.
Clinical observations were recorded for any co-toxicological signs pre-dose, immediately postudose, at l and 4 hours (3: 15 minutes) post- dose, and daily thereafter. Irritation, if any, at injection sites was scored using the Draize evaluation scores pre-dose, immediately post—dose, at 1 hour (3:15 minutes) post close, and prior to sacrifice.
Results Overall, the observed consequences of the injections of saline and CSAA were macroscopically r throughout the course of the study.
Both induced from no irritation to slight irritation with edema scores of 0-2 at various time points. Microscopic examination of injection sites suggests a very minor, clinically insignificant, irritative effect with CSAA that was no longer evident by day 4.
Example 17. Concentrated aqueous solutions of REMICADE® formulated with viscosity-lowering agents t low syringe extrusion forces and high monomer content when expelled through various gauge needles.
Materials and Methods Commercially-obtained REMICADE® containing pharmaceutical excipients (sucrose, Polysorbate 80, sodium phosphate buffer) was prepared per instructions in the prescribing information sheet. Subsequently, the aqueous drug product was purified, buffer exchanged, concentrated, dried, reconstituted, and ed as described in Example 1 above (using the extinction cient of 1.4 L/g*cm at 280 nm). 20 mM solutions of either phosphate buffer, CSAAPMI or CSAA were used for buffer exchange, and the lized cakes were tituted to 0.25 M of each Viscosity- lowering agent. Following reconstitution, the Viscosity of each solution was measured using the microfluidic viscometer as described in previous examples. The solutions were then back-loaded into 1 mL BD n syringes with 27, 29, or 31 gauge fixed s. The force required to extrude the concentrated REMICADE® solutions was then ed using an Instron at a rate of displacement lent to a fluid flow rate of 3 mL/min. The expelled solution was collected from the syringe and analyzed by size-exclusion chromatography.
RLHI’ES All REMICADE® solutions containing viscosity-lowering agents were able to be expelled h the syringes at vely low extrusion forces (Table 31). The solution containing phosphate buffer could not be expelled due to high viscosity. Both solutions containing ity—lowering agents retained high monomer content post-extrusion regardless of needle WO 38818 gauge, as indicated in Table 31.
Table 31. Syringeability of concentrated aqueous solutions of REMICADE® extruded through various gauge needles.
[REMICADE ®I Needle Syringe Agent (mg/mL) % Monomer gauge Force (N) (viscosity in c?) 0.25 M 27 ate 220 (1,500) could not be Buffer -29 extruded 0.25 M 230(90.8:t8.4) cs FMI 29 99.0 :I: 0.0 0.25 M 99.5 d: 0.1 224 (60.9 2121.1) CSAA 29 99.4 i 0.2 24.9 Example 18: Viscosity-lowering agents reduce the viscosity of concentrated aqueous solutions of biosimilar AVASTIN® Materials and Methods A commercially—obtained biosimilar AVASTIN® containing ceutical excipients (Polysorbate 20, phosphate and citrate buffers, mannitol, and NaCl) \vas purified. First, rbate 20 was removed using DETERGENT-OUT® TWEEN Medi Columns (G-Biosciences). Next, the resulting solutions were ively buffer—exchanged into 20 mM sodium phosphate buffer (PB) for PB samples and 2 mM PB for viscosity-lowering agent samples, and concentrated to a final volume of less than 10 mL on 2014/055254 Jumbosep centrifugal concentrators (Pall Corp). The viscosity-lowering agent was then added to the 2 mM PB samples as described in Example 4 above. The Viscosity-lowering agent(s) were added in an amount sufficient to give concentration upon reconstitution as specified below. In cases of combinations of agents, the concentration of each component is 0.15 M. The protein solutions were then freeze-dried. The dried protein cakes were reconstituted in phosphate buffer (for PB samples) or water (for s containing viscosity-«lowering agents) to a final volume of approximately 0.10 mL. The final tration ofmAb in solution was determined by either a Coomassie protein quantification assay by comparing unknown concentrations of s to a standard curve of biosimilar AVASTIN® or by A280 using the extinction coefficient of 1.7 L/g*cm, when possible.
Viscosities reported were measured on a RheoSense mVROC microfluidic eter. Results are reported in Table 32.
Many GRAS, 116, and API compounds are capable ofreducing the viscosity of concentrated biosimilar AVASTIN® solutions relative to phosphate-buffered samples. Of those nds included in Table 32, local anesthetics such as procaine and lidocaine, as well as GRAS agents such as biotin are among the most efficacious Viscosity reducing exeipients.
Table 32: Effect of Viscosity-Lowering Agents on ons of Biosimilar AVASTIN®.
[Biosimilar AVASHNfi, mg/ml IIIIHHHHHIIIIII 0.25 M Phosphate Buffer CSA-l t01ybiguanide :1: CSA--Na-aminocyclonexane 01 carboxylic acid Ethane disulfonate-diTris— 2Na 219 >150 CSA—piperazineT 212 t 0 64.5 Sulfacetamide-Na 214 1 13 Trimetaphosphate-3Na 21 1 121 Creatinine (0.6 M) Creatinine (0.3 M) —--- Glucuronic acid-Tris Procaine HCl Lidocaine HCl N—(4-Pyridiy1)pyridinium Cl Creatim'ne Thiamine HCl Pyridoxine Wm —-II "III Chloroquine Phosphate (0.10— 586II 718II Scopolamine HBr III _imetidineHCI 203 iptan Succinate (0.25 vir hydrate (0.02 M)- Piperacillin sodium salt Colistin sulfate salt Ceftriaxone sodium salt Cefazolin Granisetron HCI 'i'Average of two biological replicates CSA = camphorsulfonic acid.
Example 19. Viscosity Reduction is an Agent—Concentration-Dependent Effect Materials and Methods Aqueous solutions of a commercially-obtained biosimilar AVASTIN® were ed as described in Example 4. The dried protein cakes were reconstituted in phosphate buffer or water to a final volume of about 0.10 mL and a final 1-(3 —a.r_n_inopropy1)_—2_-methy_l-1H—imidaz_ole dihydrochloride (APMI*2HCl) concentration of either 0.10 or 0.25 M. The final concentration ofmAb in solution was determined by a Coomassie protein quantification assay by comparing unknown trations of samples to a standard curve of biosimilar AVASTIN®. Viscosities reported were measured on a RheoSense mVROC microfluidic viscometer.
Results As depicted in Figure 7, Viscosity-lowering effect was increased as the concentration of HCI was increased.
Example 20. A single viscosity-lowering agent lowers the ity of many therapeutically relevant monoclonal antibodies Materials and Methods Aqueous ons of a commercially-obtained biosimilar AVASTIN® were prepared as described in Example 4. The dried protein cakes were reconstituted in phosphate buffer or water to a final volume of about 0.10 mL and a final thiamine HCl concentration of 0.10 or 0.25 M.
The final concentration ofmAb in solution was determined by a Coomassie protein fication assay by comparing unknown concentrations of samples to a standard curve of biosimilar AVASTIN®.
Commercially—obtained TYSABRI® containing pharmaceutical excipients (sodium ate buffer, NaCl, Polysorbate 80) was purified, buffer exchanged, concentrated, dried, reconstituted, and ed in the same manner. Commercially-obtained HERCEPTTN® containing ceutical excipients (sodium phosphate buffer, NaCl, Polysorbate 80) was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed in the same manner. Commercially obtained biosimilar ERBITUX® containing pharmaceutical excipients (Polysorbate 80, phosphate buffer, and NaCl) was purified, buffer exchanged, concentrated, dried, tituted and analyzed in the same manner. Commercially—obtained REMICADE® containing pharmaceutical ents (sucrose, Polysorbate 80, sodium phosphate buffer) was prepared as per instructions in the prescribing ation sheet. Subsequently, the aqueous drug product was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in the same manner. Viscosities reported were measured on a RheoSense mVROC microfluidic viscometer. _ Results The data in Table 33 demonstrate that thiamine HCl can lower the viscosity of concentrated aqueous solutions ofmany eutically relevant mAbs. Thiamine HCl can produce a greater than 4—fold Viscosity reduction for each mAb.
Table 33: Effect of Thiamine HCl on Solution Viscosity.
[Excipient] , Viscosity, cP Biosimilar ' Thiamine TYSABRI® Thiamine HERCEPTIN® Thiamine Biosimiiar 235 1370i 3 ERBITUX® Thlamme. , 0.15 HCl 245 29.5 :i: 0.9 PB 0.25 176 432 i 30 Thiamine 0.15 H01 178 40.7 :I: 0.3 Examples 21~24. Viscosity~lowering agents reduce the viscosity of aqueous ons of many therapeutically relevant monoclonal antibodies Materials and Methods Aqueous ons of commercially-obtained ilar RITUXAN®, TYSABRI®, HERCEPTIN®, biosimilar ERBITUX®, and REMICADE® were prepared as described in es 18 and 19. Tables 34-38 demonstrate that viscosity-lowering agents can be advantageously employed for many different monoclonal antibodies.
Results Table 34: Viscosities of Aqueous Solutions of Biosimilar RITUXAN® in the Presence of 0.15 M Viscosi —Lowerin_ A_ents [biosirnilar RITUXAN®], Viscosity, cP mg/ml 0.25 M Phosphate Buffer 215 CSA— 1-o-tolybiguanide 190 HEPES- Tris 191 CSA—Na—Creatinine (0.3 M) 190 CSA-Na—aminocyciohexane carboxylic acid 191 61.3.-d: 2.5 Ethane disulfonate- diTris- 2Na 191 80.3.-i 16.0 CSA—piperazine 191 57.5 :1: Sulfacetamide-Na 181 64.1 A: CSA-Tris 191 59.1 d: Creatim'ne (0.6 M) 197 28.4 i --EI CSA—4-amino pyridine “III I—III I—III Glucummc ris Lu U1 CSA-Na—Omidazole Proceu'ne HCl Metoclopramide HCl Scopolamine HBI‘ --lm ———fll Chloroquine Phosphate (0 10 M) Penicillin G sodium salt -11 Piperacillin sodium salt -: Moxifloxacin HCl Ceftriaxone sodium salt 2014/055254 mycin Phosphate —19 Colistin sulfate salt _22 TAverage of 1two biological replicates Table 35: Viscosities of Aqueous Solutions of TYSABRI® in the Presence of 0.15 M Viscosity-Lowering Agents (Unless Otherwise Indicated).
[TYSABRI®}, Vlscosfiy, 0P. .
Agent mg/mL 3 10 71 5 i 106 PB 278 255 " :l: 5 23 7 182 :1: 6 Creatinine (0.30 M) 219 40.8 i 1.8 Procaine HCI 228 45.1 i 1.5 Biotin Na 233 75 .8 i 0.4 Thiamine HCl (0.10 M) 244 43 .4 i 0.7 Table 36: Viscosities of Aqueous Solutions ofHERCEPTIN® in the Presence of 0.15 M ity—Lowering Agents (Unless Otherwise Indicated).
[HERCEPTINCE], Agent Viscosity, cP mg/mL 272 400 i 4 253 172 :l: 5 239 122 i 17 218 71.6 H. 3.9 Creatinine (0.3 M) 222 45.7 0.3 Procaine HCl 222 41.8 0.6 CSA piperazine 236 50.3 0.6 GSA-Na Olrnidazole 232 60.1 0.6 Biotin-Na 230 69.9 2.3 Thiamine HCl (0.10 M) 245 41.5 444444 0.5 Table 37: Viscosities of Aqueous Solutions of ERBITUX® in the Presence of 0.15 M Viscosity~Lowering Agents (Unless Otherwise Indicated).
[ERBITUX®], Agent Viscosity, cP mg/mL 235 1370 0* ' Creatinine (0.30 M) 240 131 :t 4 Procaine HCl 230 35.9 3: 0.3 ine HCl 223 33.8 d: 0.4 Niootinamide 232 292 :1: 10 RiboflavinPhosphate (0.1 0 M) 237 492 3: 9 djne HCl 183 19.7 :I: 0.2 Metocloprmhide HCl 172 23.0 :I: 0.2 Granisetron HCl 180 23.0 :1- 0.2 Scopolamjne HBr 173 23.4 :1: 0.6 caine HCl 182 27.8 3: 0.2 Clindamycin Phosphate 209 36.5 3; 0.0 179 37.4 :I: 0.9 Chloroquine Phosphate (0.10 M) 199 54.8?02 Phenylephrine HCl 183 54.1 :I: 2.9 Moxifloxacin HCl 186 66.7 :I: 1.0 Piperacillin sodium salt 182 75.3 at 1.6 Penicillin G sodium salt Levetiracetam Fosphenytoin disodium salt Ceftriaxone sodium salt Colistin sulfate salt 203 138 :l: 4 Cefoxitin sodium salt 194 166 :l: 8 Aztreonam (0.02 M) 179 256 :l: 4 Cidofovir hydrate (0.02 M) 189 284 :1: 5 Table 38: Viscosities of Aqueous Solutions of REMICADE® in the Presence of 0.15 M Viscosity-Lowering Agents s Otherwise Indicated).
[REMICADE®], Agent Viscosity, CF mg/mL PB 176 432 30 Creatinine 144 37.1 0.5 Procaine HCl 174 23.4 0.2 Thiamine HCI 178 40.7 0.3 Example 25. Viscosity-lowering effect of TPP and TPPAPNII, as a function of concentration of biosimilar AVASTIN® Aqueous ons of a commercially-obtained biosimilar N® were prepared as bed in Example 1 above. The protein was formulated to contain either 0.25 M phosphate buffer, 0.10 M ne pyrophosphate (TPP), or 0.10 M TPP- 1-(3-aminopropyl)methyi-1H- imidazole (TPPAPMI).
Figure 8 depicts the viscosity of aqueous biosirnilar AVASTIN® solutions as a function ofmAb concentration with either ate buffer, TPP, or TPPAPMI. The viscosity of nilar AVASTIN® in phosphate buffer increases exponentially within the tested protein concentration range.
In the presence of TPP-containing excipients, the se in viscosity is attenuated i.e. the viscosity gradient is reduced.
Example 26: Viscosity-reducing effect of a viscosity—lowering agent, thiamine HCl, as a function of tration of biosimilar SIMPONI ARIA® Materials and s SIMPONI ARIA® ed commercially and containing pharmaceutical excipients (Histidine, ol, Polysorbate 80) was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in e 1 above (using the extinction coefficient of 1.4 L/g-crn at 280 nm). The protein was formulated to contain either 0.15 M phosphate buffer or 0.15 M thiamine HCl.
Results Figure 9 depicts the viscosity of aqueous SIMPONI AR1A® solutions as a function ofmAb concentration with either phosphate buffer or thiamine HCl. The viscosity of I ARIA® in ate buffer increases exponentially within the tested protein concentration range. In the presence ofthiamine HCl, the increase in viscosity is attenuated i.e. the Viscosity gradient is reduced.
Example 27. Viscosity-lowering effect of Thiamine I-ICl, as a function of concentration ofENBREL® Materials and Methods ® obtained commercially and containing pharmaceutical excipients (Mannitol, Sucrose, Tromethamine) was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1 above (using the extinction coefficient of 0.96 L/g-crn at 280 um). The protein was formulated to contain either 0.15 M phosphate buffer or 0.15 M Thiamine HCl.
Realms Table 39 depicts the Viscosity of aqueous ENBREL® solutions with either phosphate buffer or thiamine HCl. The addition ofthiamine HCI reduces the ity ofENBREL® up to about 2-fold.
Table 39: Viscosities of s Solutions of ENBREL® in the Presence of 0.15 M PB or Thiamin HCl [ENBREL] 0.15 M Th1am1n 0.15 M PB ,mg/mL HCl 271m 1120 1:26 626:}:32 250a3 439 in 305i7 212M 316 in l41:|:3 Example 28. Isotonic solutions of viscosity-lowering excipients reduce the viscosity of concentrated solutions of REMICADE® als and Methods Commercially-obtained REMICADE® containing pharmaceutical excipients (sucrose, Polysorbate 80, sodium phosphate buffer) was prepared as per instructions in the ibing ation sheet. uently, the aqueous drug product was purified, buffer exchanged, concentrated, dried, reconstituted, and analyzed as described in Example 1, except that isotonic amounts of d hydrophobic compounds were added.
Results As demonstrated in Table 40, isotonic amounts of both CSAA and CSAAPMI are capable of substantially reducing the Viscosity of concentrated solutions ofREMICADE®, in some cases by up to about 10- fold.
Table 40. Viscosities of ons of REMICADE® in the presence of isotonic (0.3 molal) viscosity-lowering excipients Salt [RIiEgrfigEqa] Viscosity (GP) PB 171 432 :l: 30 CSAAPMI 167 41.4 :1: 0.7 PB 131 175 d: 15 CSAAPMI 124 16.4 :I: 1.2 CSAA 128 25.8 :I: 0.8 Unless sly defined otherwise above, all technical and scientific terms used herein have the same meanings as commonly understood by one of skill in the art. Those skilled in the art will recognize, or will be able to ascertain using no morehthan routine mentation, many equivalents to the specific embodiments ofthe invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims (26)

We claim :
1. A liquid pharmaceutical formulation for ion comprising: (i) at least about 100 mg/ml of an antibody; (ii) thiamine or a pharmaceutically acceptable salt f; and (iii) a pharmaceutically able solvent; wherein the liquid pharmaceutical formulation, when in a volume suitable for injection, has an te viscosity of from about 1 cP to about 100 cP at 25°C as measured using a cone and plate viscometer or a microfluidic viscometer; and the absolute ity of the liquid pharmaceutical formulation is less than the absolute viscosity of a control formulation comprising the antibody and the pharmaceutically acceptable solvent, but without the thiamine or a pharmaceutically acceptable salt thereof; wherein the absolute viscosity is an extrapolated zero-shear viscosity.
2. The liquid pharmaceutical formulation of claim 1, wherein the antibody is a monoclonal antibody.
3. The liquid pharmaceutical formulation of claim 1 or 2, wherein the antibody has a molecular weight of from about 120 kDa to about 250 kDa.
4. The liquid pharmaceutical formulation of any one of the previous claims, comprising from about 100 mg/ml to about 300 mg/ml of the antibody.
5. The liquid pharmaceutical formulation of any one of the previous claims, comprising from about 178 mg/ml to about 245 mg/ml of the antibody.
6. The liquid pharmaceutical ation of any one of the us claims, wherein the pharmaceutically acceptable solvent is s.
7. The liquid pharmaceutical formulation of any one of the previous claims, wherein the ne or a pharmaceutically acceptable salt thereof is present at a concentration of from about 0.01 M to about 1.0 M.
8. The liquid pharmaceutical formulation of any one of the previous claims, wherein the thiamine or a pharmaceutically acceptable salt thereof is present at a concentration of from about 0.15 M to about 0.25 M.
9. The liquid pharmaceutical formulation of any one of the previous claims, further sing one or more pharmaceutically acceptable excipients sing a sugar, sugar l, buffering agent, preservative, carrier, antioxidant, ing agent, natural polymer, synthetic polymer, cryoprotectant, lyoprotectant, tant, bulking agent, izing agent, or any combination thereof.
10. The liquid pharmaceutical formulation of claim 9, wherein the sugar alcohol is sorbitol or mannitol.
11. The liquid pharmaceutical formulation of claim 9, wherein the one or more pharmaceutically acceptable excipients comprise a polysorbate, mer 188, sodium lauryl sulfate, a polyol, a poly(ethylene glycol), ol, a propylene glycol, or a poly(vinyl alcohol).
12. The liquid ceutical formulation of any one of the previous claims in a unitdose vial, multidose vial, cartridge, or pre-filled syringe.
13. The liquid pharmaceutical formulation of any one of the previous claims, wherein the liquid pharmaceutical formulation is reconstituted from a lyophilized composition.
14. The liquid pharmaceutical formulation of any one of the previous claims, wherein the liquid pharmaceutical formulation is isotonic to human blood serum.
15. The liquid pharmaceutical formulation of any one of the previous , wherein the absolute viscosity is measured at a shear rate of at least about 0.5 s-1 when measured using a cone and plate viscometer, or a shear rate of at least about 1.0 s-1 when measured using a microfluidic eter.
16. Use of the liquid pharmaceutical formulation of any one of the previous claims in the manufacture of a medicament for administering to a subject a therapeutically effective amount of an antibody, wherein the medicament is formulated for subcutaneous or intramuscular injection.
17. The use of claim 16, wherein the medicament is formulated for injection with a syringe.
18. The use of claim 17, wherein the syringe is a heated syringe, a self-mixing syringe, an auto-injector, a pre-filled syringe, or combinations thereof.
19. The use of claim 18, wherein the syringe is a heated syringe and the medicament is formulated to have an administration temperature between 25°C and 40°C.
20. The use of any one of claims 16-19, wherein the medicament is formulated to produce a primary irritation index of less than 3 when evaluated using a Draize scoring system.
21. The use of any one of claims 16-20, n the medicament is formulated to be administered with an ion force that is at least 10% less than an injection force for a control composition comprising the antibody and the ceutically able solvent, but without the thiamine or a pharmaceutically acceptable salt thereof, when administered in the same way.
22. The use of any one of claims 16-20, wherein the medicament is formulated to be administered with an injection force that is at least 20% less than an injection force for a control composition comprising the antibody and the pharmaceutically acceptable solvent, but without the thiamine or a pharmaceutically acceptable salt thereof, when stered in the same way.
23. The use of any one of claims 16-22, wherein the medicament is ated for administration with a needle between 27 and 31 gauge in diameter and with an injection force less than 30 N with the 27 gauge needle.
24. A method of preparing the liquid pharmaceutical formulation of any one of claims 1- 15, comprising the step of combining the dy, the pharmaceutically acceptable solvent, and the thiamine or a ceutically acceptable salt thereof.
25. A lyophilized composition comprising: (i) an antibody; (ii) ne or a pharmaceutically acceptable salt thereof; and (iii) a pharmaceutically acceptable excipient.
26. The lyophilized composition of claim 25, wherein, once reconstituted, the antibody has a concentration of at least 100 mg/ml.
NZ756260A 2013-09-11 2014-09-11 Liquid protein formulations containing viscosity-lowering agents NZ756260B2 (en)

Applications Claiming Priority (17)

Application Number Priority Date Filing Date Title
US201361876621P 2013-09-11 2013-09-11
US61/876,621 2013-09-11
US201461940227P 2014-02-14 2014-02-14
US61/940,227 2014-02-14
US201461943197P 2014-02-21 2014-02-21
US61/943,197 2014-02-21
US201461946436P 2014-02-28 2014-02-28
US61/946,436 2014-02-28
US201461988005P 2014-05-02 2014-05-02
US61/988,005 2014-05-02
US201462008050P 2014-06-05 2014-06-05
US62/008,050 2014-06-05
US201462026497P 2014-07-18 2014-07-18
US62/026,497 2014-07-18
US201462030521P 2014-07-29 2014-07-29
US62/030,521 2014-07-29
NZ717944A NZ717944B2 (en) 2013-09-11 2014-09-11 Liquid protein formulations containing viscosity-lowering agents

Publications (2)

Publication Number Publication Date
NZ756260A NZ756260A (en) 2021-06-25
NZ756260B2 true NZ756260B2 (en) 2021-09-28

Family

ID=

Similar Documents

Publication Publication Date Title
AU2020202407B2 (en) Liquid protein formulations containing viscosity-lowering agents
US20240181063A1 (en) Liquid Protein Formulations Containing Viscosity-Lowering Agents
NZ756260B2 (en) Liquid protein formulations containing viscosity-lowering agents
RU2782765C2 (en) Liquid protein compositions containing viscosity-reducing agents
NZ717944B2 (en) Liquid protein formulations containing viscosity-lowering agents
NZ717918B2 (en) Liquid protein formulations containing ionic liquids
BR112016005309B1 (en) LIQUID PHARMACEUTICAL FORMULATION COMPRISING CIMETIDINE OR ITS PHARMACEUTICALLY ACCEPTABLE SALT AND USE THEREOF
NZ756401B2 (en) Liquid protein formulations containing ionic liquids
BR122021023271B1 (en) LIQUID PHARMACEUTICAL FORMULATION COMPRISING THIAMINE OR ITS PHARMACEUTICALLY ACCEPTABLE SALT AND USE THEREOF
BR122021023267B1 (en) LIQUID PHARMACEUTICAL FORMULATION, ITS USE AND ITS PREPARATION METHOD