<<

US 20140221285A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2014/0221285 A1 BLEY et al. (43) Pub. Date: Aug. 7, 2014

(54) STABILIZED PHARMACEUTICAL (22) Filed: Feb. 4, 2014 FORMULATIONS OF ANALOGUES AND/OR INSULIN DERIVATIVES Related U.S. Application Data (60) Provisional application No. 61/761,434, ?led on Feb. (71) Applicants:Oliver BLEY, Frankfurt am Main (DE); 6, 2013. Petra LOOS, Frankfurt am Main (DE); Bernd BIDLINGMAIER, Frankfurt am (30) Foreign Application Priority Data Main (DE); Walter KAMM, Frankfurt am Main (DE); Harald BERCHTOLD, Feb. 4, 2013 (EP) ...... 133051268 Frankfurt am Main (DE) Publication Classi?cation (72) Inventors: Oliver BLEY, Frankfurt am Main (DE); Petra LOOS, Frankfurt am Main (DE); (51) Int. Cl. Bernd BIDLINGMAIER, Frankfurt am A61K 38/28 (2006.01) Main (DE); Walter KAMM, Frankfurt (52) U.S. Cl. am Main (DE); Harald BERCHTOLD, CPC ...... A61K38/28 (2013.01) Frankfurt am Main (DE) USPC ...... 514/6.4 (73) Assignee: SANOFI, Paris (FR) (57) ABSTRACT Stabilized pharmaceutical formulations of insulin analogues (21) Appl. No.: 14/172,151 and/or insulin derivatives are disclosed. Patent Application Publication Aug. 7, 2014 US 2014/0221285 A1

0104)

FigureLC/MSofTRISbufferedformulation(formulation1:no. US 2014/0221285 A1 Aug. 7, 2014

STABILIZED PHARMACEUTICAL (Pharmaceutical Research, Vol. 7, No. 6, pp. 593-599, 1990) FORMULATIONS OF INSULIN ANALOGUES disclose chemical pathways of degradation. Patel et AND/OR INSULIN DERIVATIVES al. (Pharmaceutical Research, Vol. 7, No. 7, pp. 703-711, 1990) disclose chemical pathways of peptide degradation. CROSS REFERENCE TO RELATED Tyler-Cross et al. (Journal of Biological Chemistry, Vol. 266, APPLICATION No. 33, Issue ofNovember 25, pp. 22549-22556, 1991) dis [0001] This application is related to and claims the bene?t close effects of sequence, buffers, and ionic of US. Provisional Patent Application Ser. No. 61/761,434 strength on the rate and mechanism of of aspar ?led Feb. 6, 2013, the entire contents of which are incorpo agine residues in small . GB 840,870 discloses rated by reference herein. improvements in or relating to insulin preparations. US. Pat. No. 6,852,694 discloses stabilized insulin formulations. Gal INTRODUCTION loway et al. (DiabetesiThe Journal of the American Diabe tes Association, Vol. 21, No. Suppl.2, pp. 637-648, 1972) [0002] The present invention relates to a pharmaceutical disclose new forms of insulin. Jackson et al. disclose several formulation of at least one insulin analogue and/or insulin aspects with regard to neutral (DiabetesiThe derivative, a process for preparing the pharmaceutical formu Journal of the American Diabetes Association, Vol. 21 , No. 4, lation of at least on insulin analogue and/ or insulin derivative, pp. 235-245, 1972). Lill (PharmaZie in unserer Zeit, No. 1, pp. and a related kit. It also relates to the pharmaceutical formu 56-61, 2001) discloses general aspects in connection with lation of at least one insulin analogue and/ or insulin derivative insulin formulations. The German product speci?cation of and to the related kit for use in the treatment of diabetes the medicinal product Berlinsulin® H Normal 3 mL Pen mellitus, hyperglycemia, and/or for use in lowering blood discloses a formulation containing human insulin, meta glucose levels. The present invention also relates to the use of cresol, glycerol, water and optionally hydrochloric acid and a medical device for administering the pharmaceutical for sodium hydroxide. The German product speci?cation of the mulation of at least one insulin analogue and/or insulin medicinal product Actrapid® discloses a formulation con derivative to an animal and/ or human. taining human insulin, Zinc chloride, glycerol, metacresol, sodium hydroxide, hydrochloric acid and water. BACKGROUND OF THE INVENTION [0007] The solubility of insulin, insulin analogues and/or [0003] Diabetes mellitus is a metabolic disorder in which insulin derivatives in aqueous media depends on the pH value. the ability to utilize glucose is more or less completely lost. For example, the lowest solubility is shown close to the iso [0004] For decades, insulin has been used in the treatment electric point which for human insulin is around pH 5.3 and of diabetes mellitus. Several insulin formulations have been 5.4. Very good solubility can be observed at pH values below developed, e.g. insulin Zinc (Zn (II)) suspension, formula 4 and above 7. However, insulin suffers from degradation at tions containing , etc. Further, the active pharma strong acidic conditions and strong alkaline conditions. ceutical ingredient insulin itself has been modi?ed by devel Therefore, most of the medicinal products containing insulin, oping fast acting insulin analogues (e.g. , insulin insulin analogues and/or insulin derivatives have a pH value lispro, ) and long acting insulin analogues in the range of 7.2 to 7.4 and mostly buffering agents are used and derivatives (e.g. , , insulin to achieve and maintain the pH within this range. glargin). Fast acting insulin preparations are usually solutions [0008] It has now surprisingly been found that an altema of insulin, while long acting insulin preparations can be sus tive aqueous pharmaceutical formulation comprising at least pensions containing insulin in crystalline and/ or amorphous one insulin analogue and/or insulin derivative comprising form precipitated by the addition of Zinc (Zn(II)) salts (e.g. sodium chloride, without any additional buffering agent, Zinc chloride) alone or by addition of protamine or by a shows an excellent chemical and physical stability, which combination of both. quali?es this aqueous pharmaceutical formulation as a [0005] The chemical and physical stability of insulin for medicinal product having a de?ned shelf . mulations is very important. Insulin formulations are often administered by using pen injection devices or insulin pumps SUMMARY OF THE INVENTION in which an insulin formulation is stored in cartridges until the [0009] One embodiment of the present invention relates to entire cartridge is empty. Insulin formulations may also be a pharmaceutical formulation comprising stored in vials, requiring a stable formulation with respect to chemical and physical stability across the shelf life of the (a). at least one analogue and/ or derivative of insulin; and formulation. [0006] The chemical and/or physical stability of insulin, (b). Zn(II); and insulin analogues and/ or insulin derivatives strongly depends [0010] (c). sodium chloride; and on the pharmaceutical formulation, e.g. the solvent, the pH (d). optionally protamine; value and the . Brange et al. (Acta Pharm. Nord. 4(3), pp. 149-158, 1992) disclose several aspects in connec wherein the pharmaceutical formulation has a pH value in the tion with the chemical stability of insulin. WO 2004/080480 range of from 6.0 to 9.0 and is free of any additional buffering discloses pharmaceutical preparations comprising acid-sta agent. bilized insulin. GB 835,638 discloses insulin crystal suspen [0011] In another embodiment, the pharmaceutical formu sions having a protracted effect. WO 98/56406 discloses lation according to the present invention is an aqueous phar stable insulin formulations. US. Pat. No. 6,489,292 discloses maceutical composition. stable aqueous insulin preparations without phenol and [0012] In another embodiment, the pharmaceutical formu cresol. US. Pat. No. 6,211,144 discloses stable concentrated lation according to the present invention does not contain any insulin preparations for pulmonary delivery. Bhatt et al. additional buffering agent. US 2014/0221285 A1 Aug. 7, 2014

[0013] In another embodiment, the pharmaceutical formu In one embodiment, the pharmaceutical formulation accord lation according to the present invention does not contain any ing to the present invention comprises an analogue of insulin additional buffering agent other than the at least one analogue Which is insulin glulisine. and/ or derivative of insulin and the optionally present prota [0023] In another embodiment, the pharmaceutical formu mme. lation according to the present invention comprises a deriva [0014] In another embodiment, the pharmaceutical formu tive of insulin Which is selected from the group consisting of lation according to the present invention is free of any addi insulin detemir and/ or insulin degludec. In one embodiment, tional buffering agent selected from the group consisting of the pharmaceutical formulation according to the present 2-amino-2-hydroxymethyl-propane-1,3-diol (TRIS), phos invention comprises a derivative of insulin Which is insulin phate, or citrate salts, acetic acid and salts thereof, detemir. In one embodiment, the pharmaceutical formulation glycylglycine and methionin. according to the present invention comprises a derivative of insulin Which is insulin degludec. [0015] In another embodiment, the pharmaceutical formu lation according to the present invention does not contain any [0024] In another embodiment, the pharmaceutical formu additional buffering agent selected from the group consisting lation according to the present invention comprises at least of 2-amino-2-hydroxymethyl-propane-1,3-diol (TRIS), one analogue and/or derivative of insulin Which is present in phosphate, citric acid or citrate salts, acetic acid and salts a concentration from 10 U/mL to 1000 U/mL, from 10 U/mL thereof, glycylglycine and methionin. to 600 U/mL, from 10 U/mL to 300 U/mL, from 50 U/mL to 300 U/mL or 100 U/mL. [0016] In another embodiment, the pharmaceutical formu [0025] In another embodiment, the pharmaceutical formu lation according to the present invention comprises at least lation according to the present invention comprises an ana one analogue and/or derivative of insulin and optionally logue and/or derivative of insulin Which is present in a con protamine, Wherein the only component contributing any centration from 60 to 6000 nmol/mL, from 60 nmol/mL to buffering activity is the at least one analogue and/or derivative 3600 nmol/mL, from 60 nmol/mL to 1800 nmol/mL, from of insulin and optionally protamine. 300 nmol/mL to 1800 nmol/mL or 600 nmol/mL. [0017] In another embodiment, the pharmaceutical formu [0026] In another embodiment, the pharmaceutical formu lation according to the present invention comprises at least lation according to the present invention comprises Zn(II) one analogue and/or derivative of insulin and optionally Which is present in a concentration from 0.0100 mg/mL to protamine, Wherein the total concentration of any buffering 0.0600 mg/mL, from 0.0150 mg/mL to 0.0500 mg/mL, from agent in the present invention is in the range from 3.496 0.0150 mg/mL to 0.0300 mg/mL, from 0.0150 mg/mL to mg/mL (rounded: 3.5 mg/mL) to 3.996 mg/mL (rounded: 4.0 0.0200 mg/mL, from 0.0190 mg/mL to 0.0200 mg/mL, or mg/mL), from 3.496 mg/mL (rounded: 3.5 mg/mL) to 3.816 0.0196 mg/mL. mg/mL (rounded: 3.8 mg/mL) or the total concentration of [0027] In another embodiment, the pharmaceutical formu any buffering agent in the present invention is 3.496 mg/mL lation according to the present invention comprises Zn(II) (rounded: 3.5 mg/mL). Which is present in a concentration from 0.0100 mg/100 U to [0018] In another embodiment, the pharmaceutical formu 0.0600 mg/100 U, from 0.0150 mg/100 U to 0.0500 mg/100 lation according to the present invention consists of (a). at U, from 0.0150 mg/100 U to 0.0300 mg/100 U, from 0.0150 least one analogue and/or derivative of insulin; and (b). mg/100 U to 0.0200 mg/100 U, from 0.0190 mg/100 U to Zn(II); and (c). sodium chloride; and (d). optionally prota 0.0200 mg/100 U, or 0.0196 mg/100 U. mine; and (e). metacresol; (f). phenol; and (g). polysorbate [0028] In another embodiment, the pharmaceutical formu 20; and (h). sodium hydroxide and/or hydrochloric acid for lation according to the present invention comprises sodium pH adjustment to a pH value in the range of from 6.0 to 9.0; chloride Which is present in a concentration from 0.01 mg/mL and (i). water. to 15 mg/mL, from 0.1 mg/mL to 15 mg/mL, from 0.1 mg/mL [0019] In another embodiment, the pharmaceutical formu to 10 mg/mL, from 1 mg/mL to 10 mg/mL, from 2.0 mg/mL lation according to the present invention is an aqueous phar to 10 mg/mL, from 3.0 mg/mL to 9.0 mg/mL, from 4.0 maceutical formulation. mg/mL to 9.0 mg/mL, from 5.0 mg/mL to 9.0 mg/mL, from 6.0 mg/mL to 9.0 mg/mL, from 6.8 mg/mL to 8.3 mg/mL, 6.9 [0020] In another embodiment, the pharmaceutical formu mg/mL, 7.0 mg/mL, 7.1 mg/mL, 7.2 mg/mL, 7.3 mg/mL, 7.4 lation according to the present invention comprises at least mg/mL, 7.5 mg/mL, 7.6 mg/mL, 7.7 mg/mL, 7.8 mg/mL, 7.9 one analogue and/ or derivative of insulin Which has or have an mg/mL, 8.0 mg/mL, 8.1 mg/mL, 8.2 mg/mL or 8.3 mg/mL, or isoelectric point (IEP) in the range from 4.0 to 6.0, from 4.5 to 6.8 mg/mL. 6.0, from 4.5 to 5.5, from 5.0 to 5.5, from 5.0 to 5.2 or 5.1. [0029] In another embodiment, the pharmaceutical formu [0021] In another embodiment, the pharmaceutical formu lation according to the present invention comprises protamine lation according to the present invention has a pH value in the or Which is present in a concentration from range from 6.5 to 8.5, in the range from 7.0 to 8.0, from 7.0 to 0.10, 0.15, 0.20, 0.25, 0.30, 0.32, 0.35, 0.40, 0.45 or 0.5 7.8, from 7.1 to 7.6, 7.2, 7.3, 7.4 or 7.5, or 7.4. mg/mL. [0022] In another embodiment, the pharmaceutical formu [0030] In another embodiment, the pharmaceutical formu lation according to the present invention comprises at least lation according to the present invention comprises a stabi one analogue of insulin Which is selected from the group liZing agent, Which is in one embodiment a surfactant, a consisting of insulin aspart, and/or insulin polyoxyethylene derivative of sorbitan monolaurate (e.g. glulisine. In one embodiment, the pharmaceutical formula polysorbate 20), a polyethoxylethylene derivate of oleic acid tion according to the present invention comprises an analogue (e.g. polysorbate 80), poloxamer (Which is a polyoxyethyl of insulin Which is insulin lispro. In one embodiment, the ene-polyoxypropylene copolymer), or polysorbate 20 or pharmaceutical formulation according to the present inven polysorbate 80 or mixtures thereof. In another embodiment, tion comprises an analogue of insulin Which is insulin aspart. the stabiliZing agent, in one embodiment the surfactant, the US 2014/0221285 A1 Aug. 7, 2014

polyoxyethylene derivative of sorbitan monolaurate (e.g. modulators, omega-3 fatty acids and derivatives polysorbate 20), the polyethoxylethylene derivate of oleic thereof, active substances for the treatment of obesity, such as acid (e.g. polysorbate 80), poloxamer (Which is a polyoxy sibutramine, tesofensine, orlistat, CB-l receptor antagonists, -polyoxypropylene copolymer), and in another MCH-l antagonists, MC4 receptor agonists and partial ago polysorbate 20 or polysorbate 80 or mixtures thereof are/is nists, NPY5 or NPY2 antagonists, NPY4 agonists, beta-3 present in a concentration from 0.01 to 0.05 mg/mL, 0.010 agonists, or leptin mimetics, agonists of the 5HT2c mg/mL, 0.015 mg/mL, 0.020 mg/mL, 0.025 mg/mL, 0.03 receptor, or the combinations of bupropione/ mg/mL, or 0.02 mg/mL. (CONTRAVE), bupropione/zonisamide (EMPATIC), bupro [0031] In another embodiment, the pharmaceutical formu pione/phentermine or /, QNEXA lation according to the present invention comprises more than (Phentermine+topiramate), lipase inhibitors, one analogue and/or derivative of insulin, Wherein one ana inhibitors, H3 antagonists, AgRP inhibitors, triple monoam logue and/or derivative of insulin is a fast acting insulin and ine uptake inhibitors (norepinephrine and acetylcholine), one analogue and/or derivative of insulin is a long acting MetAP2 inhibitors, nasal formulation of the calcium channel insulin. In another embodiment, the pharmaceutical formu blocker diltiazem, antisense oligonucleotides against produc lation according to the present invention comprises a fast tion of ?broblast 4, prohibitin targeting acting insulin selected from the group comprising insulin peptide- 1, drugs for in?uencing high blood pressure, chronic aspart, insulin lispro and/or insulin glulisine and a long acting or , such as angiotensin II recep insulin selected from the group comprising insulin glargin, tor antagonists, ACE inhibitors, ECE inhibitors, diuretics, insulin detemir and/or insulin degludec. beta-blockers, calcium antagonists, centrally acting hyper tensives, antagonists of the alpha-2-adrenergic receptor, [0032] In another embodiment, the pharmaceutical formu inhibitors of neutral endopeptidase, thrombocyte aggregation lation according to the present invention comprises one or inhibitors. more further active pharmaceutical ingredients. In one embodiment the further active pharmaceutical ingredient is [0033] In another embodiment, the pharmaceutical formu lation according to the present invention comprises more than an antidiabetic agent. In another embodiment, the pharma one analogue and/or derivative of insulin, Wherein one ana ceutical formulation according to the present invention com logue and/or derivative of insulin is a fast acting insulin and prises one or more antidiabetic agents as further active phar maceutical ingredients selected from the group comprising: one analogue and/or derivative of insulin is a long acting GLP-1 receptor agonists, dual GLP-l receptor/ insulin. In another embodiment the fast acting insulin is selected from the group comprising insulin aspart, agonists, human FGF-21, FGF-21 analogues, FGF lispro and/or insulin glulisine and the long acting insulin is 21 derivatives, , human insulin, analogues of insulin, selected from the group comprising insulin detemir and/or and derivatives of insulin. In another embodiment, the phar maceutical formulation according to the present invention insulin degludec. comprises one or more further active pharmaceutical ingre [0034] In another embodiment, the pharmaceutical formu dients selected from the group comprising: insulin and insulin lation according to the present invention consists of (a). 3 .496 derivatives, GLP-l, GLP-l analogues and GLP-1 receptor mg/mL insulin aspart (rounded: 3.5 mg/mL); and (b). 1.72 agonists, polymer bound GLP-1 and GLP-1 analogues, dual mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). GLPl/GIP agonists, dual GLPl/ agonists, 0.04087 mg/mL Zn(II); and (e). 6.8 mg/mL sodium chloride; PYY3-36 or analogues thereof, or and (f). 0.02 mg/mL polysorbate 20; and (g). sodium hydrox analogues thereof, glucagon receptor agonists or antagonists, ide and/or hydrochloric acid to adjust the pH to 7.4 and (h). GIP receptor agonists or antagonists, ghrelin antagonists or water. inverse agonists, Xenin and analogues thereof, DDP-IV [0035] In another embodiment, the pharmaceutical formu inhibitors, SGLT2 inhibitors, dual SGLT2/SGLT1 inhibitors, lation according to the present invention consists of (a). 3.496 , dual PPAR agonists, sulfony mg/mL insulin aspart (rounded: 3.5 mg/mL); and (b). 1.72 lureas, , alpha-glucosidase inhibitors, mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). and amylin analogues, GPR119 agonists, GPR40 agonists, 0.04087 mg/mL Zn(II); and (e). from 6.8 mg/mL to 8.3 GPR120 agonists, GPR142 agonists, systemic or loW-absorb mg/mL sodium chloride; (f). 0.02 mg/mL polysorbate 20; (g) able TGR5 agonists, Cycloset, inhibitors of 11-beta-HSD, from 0.1 mg/mL to 0.5 mg/mL protamine sulfate; and (h). activators of glucokinase, inhibitors of DGAT, inhibitors of sodium hydroxide and/or hydrochloric acid to adjust the pH tyrosinephosphatase 1, inhibitors of glucose-6-phos to a pH in the range from 7.1 to 7.6 and (i). water. phatase, inhibitors of fructose-1 ,6-bisphosphatase, inhibitors [0036] In another embodiment, the pharmaceutical formu of glycogen phosphorylase, inhibitors of phosphoenol pyru lation according to the present invention consists of (a). 3.496 vate carboxykinase, inhibitors of glycogen synthase kinase, mg/mL insulin aspart (rounded: 3.5 mg/mL); and (b). 1.72 inhibitors of pyruvate dehydrogenase kinase, alpha2-antago mg/mL metacresol; and (c). 1.50 mg/mL phenol; and (d). nists, CCR-2 antagonists, modulators of glucose transporter 0.04087 mg/mL Zn(II); and (e). 6.8 or 6.9 or 7.0 or 7.1 or 7.2 4, 3 agonists, HMG-CoA-reductase or 7.3 or 7.4 or 7.5 or 7.6 or 7.7 or 7.8 or 7.9 or 8.0 or 8.1 or inhibitors, ?brates, nicotinic acid and the derivatives thereof, 8.2 or 8.3 mg/mL sodium chloride; (f). 0.02 mg/mL polysor nicotinic acid receptor 1 agonists, PPAR-alpha, gamma or bate 20; (g) 0.1 or 0.15 or 0.2 or 0.25 or 0.3 or 0.32 or 0.35 or alpha/gamma) agonists or modulators, PPAR-delta agonists, 0.4 or 0.45 or 0.5 mg/mL protamine sulfate; and (h). sodium ACAT inhibitors, absorption inhibitors, bile acid hydroxide and/ or hydrochloric acid to adjust the pH to 7.4 and binding substances, IBAT inhibitors, MTP inhibitors, modu (i). water. lators of PCSK9, LDL receptor up-regulators by liver selec [0037] The present invention also provides a pharmaceuti tive thyroid hormone receptor B agonists, HDL-raising cal formulation for use in the treatment of diabetes mellitus, compounds, lipid metabolism modulators, PLA2 inhibitors, hyperglycemia and/ or for use in lowering blood glucose lev ApoA-I enhancers, cholesterol synthesis inhibitors, lipid els. US 2014/0221285 A1 Aug. 7, 2014

[0038] The present invention also provides a process for tors, PPAR-delta agonists, ACAT inhibitors, cholesterol preparing the pharmaceutical formulation according to the absorption inhibitors, bile acid-binding substances, IBAT present invention, Wherein the components are mixed inhibitors, MTP inhibitors, modulators of PCSK9, LDL together in the form of a solution or suspension, the desired receptor up-regulators by liver selective thyroid hormone pH is adjusted and the mixture is made up to the ?nal volume receptor B agonists, HDL-raising compounds, lipid metabo With water. lism modulators, PLA2 inhibitors, ApoA-I enhancers, cho [0039] The present invention also relates to a kit or combi lesterol synthesis inhibitors, lipid metabolism modulators, nation comprising separate packages of the pharmaceutical omega-3 fatty acids and derivatives thereof, active substances formulation according to the present invention and a medical for the treatment of obesity, such as sibutramine, tesofensine, device. In one embodiment the medical device is selected orlistat, CB-lreceptor antagonists, MCH-l antagonists, MC4 from the group comprising: syringe, insulin injection system, receptor agonists and partial agonists, NPYS or NPY2 insulin infusion system, insulin pump, insulin pen injection antagonists, NPY4 agonists, beta-3-agonists, leptin or leptin device. mimetics, agonists of the 5HT2c receptor, or the combina [0040] The present invention also relates to a kit or combi tions of bupropione/naltrexone (CONTRAVE), bupropione/ nation comprising separate packages of the pharmaceutical zonisamide (EMPATIC), bupropione/phentermine or pram formulation according to the present invention, of at least one lintide/metreleptin, QNEXA (Phentermine+topiramate), further active pharmaceutical ingredient and optionally of a lipase inhibitors, angiogenesis inhibitors, H3 antagonists, medical device. In one embodiment the medical device is AgRP inhibitors, triple monoamine uptake inhibitors (nore selected from the group comprising: syringe, insulin injection pinephrine and acetylcholine), MetAP2 inhibitors, nasal for system, insulin infusion system, insulin pump, insulin pen mulation of the calcium channel blocker diltiazem, antisense injection device. oligonucleotides against production of ?broblast growth fac [0041] The present invention also relates to a kit or combi tor receptor 4, prohibitin targeting peptide- 1, drugs for in?u nation comprising separate packages of the pharmaceutical encing high blood pressure, chronic heart failure or athero formulation according to the present invention, of at least one sclerosis, such as angiotensin II receptor antagonists, ACE further active pharmaceutical ingredient and optionally of a inhibitors, ECE inhibitors, diuretics, beta-blockers, calcium medical device, Wherein the further active pharmaceutical antagonists, centrally acting hypertensives, antagonists of the alpha-2-adrenergic receptor, inhibitors of neutral endopepti ingredient is an antidiabetic agent. dase, thrombocyte aggregation inhibitors. [0042] The present invention also relates to a kit or combi nation comprising separate packages of the pharmaceutical [0043] The present invention also relates to a kit or combi formulation according to the present invention, of at least one nation comprising separate packages of the pharmaceutical further active pharmaceutical ingredient and optionally of a formulation according to the present invention, of at least one medical device, Wherein the further active pharmaceutical further active pharmaceutical ingredient and optionally of a ingredient is an antidiabetic agent selected from the group medical device, Wherein the kit comprises more than one comprising: GLP-1 receptor agonists, dual GLP-1 receptor/ analogue and/or derivative of insulin, Wherein one analogue glucagon receptor agonists, human FGF-Zl, FGF-Zl ana and/or derivative of insulin is a fast acting insulin and one logues, FGF-Zl derivatives, insulins, human insulin, ana analogue and/or derivative of insulin is a long acting insulin. logues of insulin, and derivatives of insulin. In another In one embodiment the fast acting insulin is selected from the embodiment, the pharmaceutical formulation according to group comprising insulin aspart, insulin lispro and/or insulin the present invention comprises one or more further active glulisine and Wherein the long acting insulin is selected from pharmaceutical ingredients selected from the group compris the group comprising insulin glargin, insulin detemir and/or ing: insulin and insulin derivatives, GLP- l, GLP-l analogues insulin degludec. and GLP-1 receptor agonists, polymer bound GLP-1 and [0044] The present invention also relates to a kit or combi GLP-1 analogues, dual GLPl/GIP agonists, dual GLPl/Glu nation comprising separate packages of the pharmaceutical cagon receptor agonists, PYY3-36 or analogues thereof, pan formulation according to the present invention, of at least one creatic polypeptide or analogues thereof, glucagon receptor further active pharmaceutical ingredient and optionally of a agonists or antagonists, GIP receptor agonists or antagonists, medical device for use in the treatment of diabetes mellitus, ghrelin antagonists or inverse agonists, Xenin and analogues hyperglycemia and/ or for use in lowering blood glucose lev thereof, DDP-IV inhibitors, SGLT2 inhibitors, dual SGLTZ/ els. SGLTl inhibitors, biguanides thiazolidinediones, dual PPAR agonists, , meglitinides, alpha-glucosidase [0045] In another embodiment, the present invention also inhibitors, amylin and amylin analogues, GPRll9 agonists, relates to a kit or combination comprising separate packages GPR40 agonists, GPR120 agonists, GPR142 agonists, sys of the pharmaceutical formulation according to the present temic or loW-absorbable TGRS agonists, Cycloset, inhibitors invention, of at least one further active pharmaceutical ingre of ll-beta-HSD, activators of glucokinase, inhibitors of dient and optionally of a medical device, Wherein the phar DGAT, inhibitors of protein tyro sinepho sphatase 1, inhibitors maceutical formulation according to the present invention of glucose-6-phosphatase, inhibitors of fructose-1,6-bispho and the further active pharmaceutical ingredient, in one sphatase, inhibitors of glycogen phosphorylase, inhibitors of embodiment an antidiabetic agent, are administered continu phosphoenol pyruvate carboxykinase, inhibitors of glycogen ously, separately, sequentially and/or stepWise. synthase kinase, inhibitors of pyruvate dehydrogenase [0046] The present invention also relates to the use of a kinase, alpha2-antagonists, CCR-2 antagonists, modulators medical device for administering the pharmaceutical formu of glucose transporter-4, somatostatin receptor 3 agonists, lation to an animal and/or human. In one embodiment, the HMG-CoA-reductase inhibitors, ?brates, nicotinic acid and medical device is selected from the group comprising: the derivatives thereof, nicotinic acid receptor 1 agonists, syringe, insulin injection system, insulin infusion system, PPAR-alpha, gamma or alpha/gamma) agonists or modula insulin pump, insulin pen injection device US 2014/0221285 A1 Aug. 7, 2014

DETAILED DESCRIPTION [0054] (d). Dual GLP-l/glucagon receptor agonists [0055] Examples of dual GLP-l/glucagon receptor ago [0047] As used herein, the singular forms a , an”, and “the” include plural reference unless the context clearly dic nists include but are not limited to OAP-189 (PF-05212389, tates otherwise. Thus, for example, reference to a ?ll material TKS-1225), TT-401/402, ZP2929, LAPS-HMOXM25, containing “a carrier” includes one or more carriers, reference MOD-6030). to “an additive” includes reference to one or more of such [0056] Other suitable active pharmaceutical ingredients additives. Which may be included in the pharmaceutical formulations of [0048] As used herein, the term “active pharmaceutical the invention include but are not limited to the folloWing: ingredient” (API) includes any pharmaceutically active [0057] Further gastrointestinal peptides such as peptideYY chemical or biological compound and any pharmaceutically 3-36 (PYY3-36) or analogues thereof and pancreatic acceptable salt thereof and any mixture thereof, that provides polypeptide (PP) or analogues thereof. some pharmacologic effect and is used for treating or pre [0058] Glucagon receptor agonists or antagonists, GIP venting a condition. Exemplary pharmaceutically acceptable receptor agonists or antagonists, ghrelin antagonists or salts include hydrochloric, sulfuric, nitric, phosphoric, hydro inverse agonists and xenin and analogues thereof. Dipeptidyl bromic, maleric, malic, ascorbic, citric, tartaric, pamoic, lau peptidase-IV (DPP-4) inhibitors, for example: /Ne ric, stearic, palmitic, oleic, myristic, lauryl sulfuric, naphtha sina®, /BI-135 6/ Ondero®/ Traj enta®/ Tradj enta®/ linesulfonic, linoleic, linolenic acid, and the like. As used Trayenta®, /Onglyza®, /Januvia®/Xe herein, the terms “active pharmaceutical ingredient”, “drug”, levia®/Tesavel®, sitagliptin+/Janumet®/ “active agent”, “active ingredient”, “active substance” and Velmetia®, aildagliptin, , aemigliptin, tenegliptin, “drug” are meant to be synonyms, i.e., have identical mean melogliptin, , DA-1229, MK-3102, KM-223, mg. KRP-104 and Ari-2243. [0049] In a one embodiment the active pharmaceutical [0059] Sodium-dependent glucose transporter 2 (SGLT2) ingredient is an antidiabetic agent. Examples of antidiabetic inhibitors, for example: canagli?ozin, dapagli?oZin, remogli agents are found in the Rote Liste 2012, chapter 12. Examples ?oZin, sergli?oZin, empagli?oZin, ipragli?oZin, tofogli?ozin of antidiabetic agents include but not limited to (a) insulin, (RO-4998452), luseogli?oZin, LX-4211, ertugli?ozin (PF insulin analogues and insulin derivatives, (b) glucagon-like 04971729), EGT-0001442 and DSP-3235. peptide 1 (GLP-1) and its analogues and receptor agonists, (c) [0060] Dual SGLT2/SGLT1 inhibitors. dual GLP-l/GIP agonists, and (d) dual GLP-l/glucagon [0061] Biguanides (e.g. metformin, , phen receptor agonists, as described in detail next. formin), thiazolidinediones (e.g. , , [0050] (a). Insulin, insulin analogues, and insulin deriva , ), dual PPAR agonists (e.g. alegli tives Examples of insulin, insulin analogues, and insulin tazar, muraglitazar, ), sulfonylureas (e.g. tolbuta derivatives include but are not limited to mide, , /Amaryl®, ), meg (Lantus®), insulin glulisine (Apidra®), insulin detemir litinides (e.g. , , ), alpha (Levemir®), insulin lispro (Humalog®/Liprolog®), insulin glucosidase inhibitors (e.g. , , ), degludec (Tresiba®), insulin aspart (NovoLog®/No amylin and amylin analogues (e.g. pramlintide/Symlin®). voRapid®), basal insulin and analogues (e.g. LY2605541, G-protein coupled receptor 119 (GPR119) agonists (e.g. LY2963016), PEGylated insulin lispro, Humulin®, Lin GSK-1292263, PSN-821, MBX-2982, jeta®, SuliXen®, NN1045, insulin plus Symlin®, fast-acting [0062] APD-597,ARRY-981). and short-acting insulins (e.g. Linjeta®, PH20 insulin, [0063] GPR40 agonists (e.g. TAK-875, TUG-424, P-1736, NN1218, HinsBet®), oral, inhalable, transdermal and sublin JTT-851, GW9508). GPR120 agonists and GPR142 agonists. gual insulins (e.g. Exubera®, Nasulin®, Afrezza®, insulin [0064] Systemic or loW-absorbable TGRS (GPBARIIG tregopil, TPM-02/Insulin, Capsulin®, Oral-lyn®, Cobal protein-coupled bile acid receptor 1) agonists (e.g. INT-777, amin® oral insulin, ORMD-0801, NN1953, VIAtab®). XL-475, SB756050). Additionally included are also those insulin derivatives Which [0065] /Cycloset®, inhibitors of 11-beta are bonded to albumin or another protein by a bifunctional hydroxysteroid dehydrogenase (11-beta-HSD) (e.g. linker. LY2523199, BMS770767, RG-4929, BMS816336, AZD [0051] (b). Glucagon-like-peptide 1 (GLP-l), GLP-l ana 8329, HSD-016, BI-135585), activators of glucokinase (e.g. logues and GLP-1 receptor agonists Examples of GLP-1, PF-04991532, TTP-399, GK1-399, ARRY-403 (AMG-151), GLP-l analogues and GLP-1 receptor agonists include but TAK-329, ZYGKI), inhibitors of diacylglycerol O-acyl are not limited to (AVE0010/ZP10/Lyxumia®), transferase (DGAT) (e.g. pradigastat (LCQ-908), LCQ-908), /exendin-4 (Byetta®/Bydureon®/ITCA 650, lira inhibitors of protein tyrosinephosphatase 1 (e.g. trodusquem glutide/Victoza®), , , , ine), inhibitors of glucose-6-phosphatase, inhibitors of fruc , rExendin-4, CJC-1134-PC, PB-1023, TTP-054, tose-1,6-bisphosphatase, inhibitors of glycogen phosphory HM-112600, CM-3, GLP-l Eligen, ORMD-0901, NN9924, lase, inhibitors of phosphoenol pyruvate carboxykinase, Nodexen, Viador-GLP-l, CVX-096, ZYOG-l, ZYD-l, inhibitors of glycogen synthase kinase, inhibitors of pyruvate MAR-701, ZP-2929, ZP-3022, CAM-2036, DA-15864,ARI dehydrogenase kinase, alpha2 adrenergic receptor antago 2651, ARI-2255, exenatide-XTEN and glucagon-XTEN, nists, C4C chemokine receptor type 2 (CCR-2) antagonists, AMX-8089+VRS-859 and polymer bound GLP-1 and modulators of glucose transporter-4 and somatostatin recep GLP-1 analogues. tor 3 agonists (e.g. MK-4256). [0052] (c). Dual GLP-l/glucose-dependent insulinotropic [0066] One or more lipid lowering agents are also suitable peptides (GIP) agonists as active pharmaceutical ingredients, such as for example: [0053] Examples of dual GLP-l/GIP agonists include but 3 -hydroxy-3 -methylglutaryl-coenzym-A-reductase (HMG are not limited to MAR701, MAR-709, BHM081/BHM089/ CoA-reductase) inhibitors (e.g. simvastatin, atorvastatin, BHM098). rosuvastatin), ?brates (e.g. beza?brate, feno?brate), nicotinic US 2014/0221285 A1 Aug. 7, 2014

acid and derivatives thereof (e.g. niacin, including slow olmesartan, tasosartan, aZilsartan), angiotensin converting release formulations of niacin), nicotinic acid receptor 1 ago (ACE) inhibitors, endothelin converting enzyme nists (e.g. GSK-256073), peroxisome proliferator-activated (ECE) inhibitors, diuretics, beta-blockers, calcium antago receptors (PPAR-)(alpha, gamma or alpha/gamma) agonists nists, centrally acting hypertensives, antagonists of the alpha or modulators (e.g. ), PPAR-delta agonists, acetyl 2-adrenergic receptor, inhibitors of neutral endopeptidase, CoA-acetyltransferase (ACAT) inhibitors (e.g. avasimibe), thrombocyte aggregation inhibitors and others or combina cholesterol absorption inhibitors (e.g. ezetimibe), bile acid tions thereof are suitable. binding substances (e.g. cholestyramine, colesevelam), ileal [0071] As used herein, the terms “analogue of insulin” and bile acid transport inhibitors (IBAT) (e.g. GSK-2330672), “insulin analogue” refer to a polypeptide which has a molecu microsomal triglyceride transfer protein (MTP) inhibitors lar structure which formally can be derived from the structure (e.g. lomitapide (AEGR-733), SLx-4090, granotapide), of a naturally occurring insulin, for example that of human modulators of proprotein convertase subtilisin/kexin type 9 insulin, by deleting and/or exchanging at least one amino acid (PCSK9) (e.g. REGN727/SAR236553, AMG-145, LGT residue occurring in the naturally occurring insulin and/or 209, PF-04950615, MPSK3169A, LY3015014, ALD-306, adding at least one amino acid residue. The added and/or ALN-PCS, BMS-962476, SPC5001, ISIS-394814, 1B20, exchanged amino acid residue can either be codable amino LGT-210, 1D05, BMS-PCSK9Rx-2, SX-PCK9, RG7652), acid residues or other naturally occurring residues or purely LDL receptor up-regulators, for example liver selective thy synthetic amino acid residues. Examples of analogues of roid hormone receptor beta agonists (e.g. eprotirome (KB insulin include, but are not limited to, the following: 2115), MB07811, sobetirome (QRX-431), VIA-3196, (i). ‘Insulin aspart’ is created through recombinant DNA tech ZYTl), HDL-raising compounds such as: CETP inhibitors nology so that the amino acid B28 in human insulin (i.e. the (e.g. torcetrapib, anacetrapib (MK0859), dalcetrapib, evace amino acid no. 28 in the B chain of human insulin), which is trapib, JTT-302, DRL-17822, TA-8995, R-1658, , is replaced by ; LY-2484595) or ABC1 regulators, lipid metabolism modula (ii). ‘Insulin lispro’ is created through recombinant DNA tors (e.g. BMS-823778, TAP-301, DRL-21994, DRL technology so that the penultimate and proline residues 21995), phospholipase A2 (PLA2) inhibitors (e.g. darap on the C-terminal end of the B-chain of human insulin are ladib/Tyrisa®, varespladib, rilapladib), ApoA-I enhancers reversed (human insulin: ProBZSLysmg; insulin lispro: (e.g. RVX-208, CER-001, MDCO-216, CSL-112, VRX LySBZSPrOBZ9); HDL, VRX-1243, VIRxSYS), cholesterol synthesis inhibi (iii). “Insulin glulisine” differs from human insulin in that the tors (e.g. ETC-1002) and lipid metabolism modulators (e.g. amino acid at position B3 is replaced by lysine and BMS-823778, TAP-301, DRL-21994, DRL-21995) and the lysine in position B29 is replaced by ; omega-3 fatty acids and derivatives thereof (e.g. icosapent (iv). “Insulin glargine” differs from human insulin in that the ethyl (AMRlOl), Epanova®, AKR-063, NKPL-66). asparagine at position A21 is replaced by and the B [0067] Other suitable active pharmaceutical ingredients chain is extended at the carboxy terminal by two . which may be included in the pharmaceutical formulations [0072] As used herein, the term “aqueous” refers to a solu include one or more active substances for the treatment of tion in which the solvent is water and/or to a suspension in obesity, including but not limited to: which the external phase is water and/or to an emulsion in [0068] Sibutramine, tesofensine, orlistat, cannabinoid which the dispersed or continuous phase is water. receptor 1 (CB1) antagonists (e.g. TM-38837), -con [0073] As used herein, the term “buffering agent” refers to centrating hormone (MCH-l) antagonists (e.g. BMS a weak acid or base used to maintain the acidity (pH) of a 830216, ALB-127158(a)), MC4 receptor agonists and partial solution, a suspension and/ or an emulsion near a chosen value agonists (e.g. AZD-2820, RM-493), neuropeptide Y5 after the addition of another acid or base. The function of a (NPY5) or NPY2 antagonists (e.g. , S-234462), buffering agent is to prevent a rapid change in the pH value NPY4 agonists (e.g. PP-1420), beta-3-adrenergic receptor when acids or bases are added to the solution. In an aqueous agonists, leptin or leptin mimetics, agonists of the 5-hydrox solution, suspension and/or emulsion, a buffering agent is ytryptamine 2c (5HT2c) receptor (e.g. lorcaserin), or the present in a mixture of a weak acid and its conjugate base or combinations of bupropione/naltrexone (Contrave®), bupro a in a mixture of a weak base and its conjugated acid. pione/zonisamide (Empatic®), bupropione/phentermine or Examples of buffering agents include, but are not limited to, pramlintide/metreleptin, phentermine/topiramate the following: sodium bicarbonate; acetic acid or acetate salts (stmia®), lipase inhibitors (e.g. cetilistat/Cametor®), (e.g. sodium acetate, Zinc acetate); boric acid or boric salts; angiogenesis inhibitors (e.g. ALS-L1023), histamine H3 N-cyclohexyl-2-aminoethanesulfonic acid (CHES) or salts antagonists (e.g. HPP-404), AgRP (agouti related protein) thereof; 3 -[ [1 ,3 -dihydroxy-2-(hydroxymethyl)propan-2 -yl] inhibitors (e.g. TTP-435), triple monoamine uptake inhibi amino]propane-1-sulfonic acid (TAPS) or salts thereof; 2-(N tors (dopamine, norepinephrine and serotonin reuptake) (e. g. morpholino)ethanesulfonic acid (MES) and salts thereof; tesofensine), aminopeptidase 2 (MetAP2) inhibi piperaZine-N,N'-bis(2-ethanesulfonic acid (PIPES) and salts tors (e.g. beloranib), nasal formulations of the calcium chan thereof; N-(2-acetamido)-2-aminoethane-sulfonic acid nel blocker diltiazem (e.g. CP-404) and antisense oligonucle (ACES) and salts thereof; cholamine chloride; BES; 2-[[1,3 otides against production of ?broblast growth factor receptor dihydroxy-2-(hydroxymethyl)-propan-2-yl]amino]ethane 4 (FGFR4) (e.g. ISIS-FGFR4Rx) or prohibitin targeting pep sulfonic acid (TES) and salts thereof; 2-[4-(2-hydroxyethyl) tide-1 (e.g. Adipotide®). piperaZin-1-yl]ethanesulfonic acid (HEPES) and salts [0069] Further suitable active pharmaceutical ingredients thereof; acetamidoglycine; N-(2-hydroxy-1,1-bis(hydroxyl which may be included in the pharmaceutical formulations methyl)ethyl)glycine (tricine); glycinamide; 2-(bis(2-hy include but are not limited to: droxyethyl)amino)acetic acid (bicine) and salts thereof; pro [0070] Angiotensin II receptor antagonists (e.g. telmisar pionate salts; 3-[[1,3-dihydroxy-2-(hydroxymethyl)propan tan, candesartan, valsartan, losartan, eprosartan, irbesartan, 2-yl]-amino]-2-hydroxy-propane-1-sulfonic acid (TAPSO) US 2014/0221285 A1 Aug. 7, 2014

and salts thereof; 3-morpholinopropane-1-sulfonic acid [0079] As used herein, the term “formulation” refers to a (MOPS) and salts thereof; saline-sodium citrate (SSC) buffer; product comprising speci?ed ingredients in predetermined 2 -amino -2 -hydroxymethyl -propane-1 ,3 -diol (synonyms: amounts or proportions, as well as any product that results, TRIS, trisamine, THAM, tromethamine, trometamol, directly or indirectly, from combining speci?ed ingredients in tromethane); citric acid or citrate salts (e.g. sodium citrate); speci?ed amounts. In relation to pharmaceutical formula trisodium phosphate, disodium hydrogen phosphate, sodium tions, this term encompasses a product comprising one or dihydrogen phosphate, tripotassium phosphate, dipotassium more active ingredients, and an optional carrier comprising phosphate, monopotassium phosphate and/ or any other buff inert ingredients, as well as any product that results, directly ering agent containing phosphate. Amino acids (having free or indirectly, from combination, complexation or aggregation basic or acidic functional groups, e.g. methionin, ) or of any two or more of the ingredients, or from dissociation of peptides (having free basic or acidic functional groups) may one or more of the ingredients, or from other types of reac also be used as buffering agent. As used herein, the term tions or interactions of one or more of the ingredients. In “buffering agent” also comprises amino acids, peptides and general, pharmaceutical formulations are prepared by uni . As insulin analogues and/ or insulin derivatives and/ formly bringing the active pharmaceutical ingredient (i.e. the or protamine are peptides or derivatives of peptides (i.e. both analogue and/or derivative of insulin) into association with a contain amino acids having free basic or acidic functional liquid carrier or a ?nely divided solid carrier orboth, and then, groups), they may also have a certain buffering capacity, i.e. if necessary, shaping the product into the desired formulation. are also to be considered as buffering agent. The pharmaceutical formulation includes enough of the [0074] As used herein, the term “fast acting insulin” or active pharmaceutical ingredient to produce the desired effect “short acting insulin” refers to insulin analogues and/ or insu upon the progress or condition of diseases.As used herein, the lin derivatives, wherein the insulin-mediated effect begins term “formulation” may refer to a solution as well as to a within 5 to 15 minutes and continues to be active for 3 to 4 suspension or to an emulsion. As used herein, the terms “for hours. Examples of fast acting insulins include, but are not mulation” and “composition” are meant to be synonyms, i.e., limited to, the following: (i). insulin aspart; (ii). insulin lispro have identical meaning. The pharmaceutical compositions and (iii). insulin glulisine. are made following conventional techniques of pharmaceuti [0075] As used herein, the terms “free of additional buffer cal technology involving mixing, ?lling and dissolving the ing agent” or “buffer-free” means that there is no further ingredients, as appropriate, to give the desired oral, buffering agent next to the analogue and/ or derivative of parenteral, rectal, transdermal, or topical products. insulin and optionally protamine. As mentioned above, ana [0080] As used herein, the term “GLP-1 receptor agonist” logues and/ or derivatives of insulin as well as protamine refers to compounds which have an agonistic activity at the contain amino acids having acidic orbasic side chains and are glucagon-like peptide-1 receptor. Examples of GLP-1 recep therefore also to be considered as buffering agent. Indepen tor agonists include, but are not limited to, the following: dently from the absence of any buffering agents, the aqueous exenatide/exendin-4, , lixisenatide, dulaglutide, pharmaceutical formulation according to the present inven albiglutide, semaglutide, taspoglutide, rExendin-4, CJC tion may optionally comprise protamine. 1134-PC, PB-1023, TTP-054, HM-112600, CM-3, GLP-l [0076] As used throughout the description and the claims of Eligen, ORMD-0901, NN9924, Nodexen, Viador-GLP-l, this speci?cation, the word “comprise” and variations of the CVX-096,ZYOG-1, ZYD-l, MAR-701, ZP-2929, ZP-3022, word, such as “comprising” and “comprises” is not intended CAM-2036, DA-15864, ARI-2651, ARI-2255, exenatide to exclude other additives, components, integers or steps. XTEN and glucagon-XTEN, AMX-8089+VRS-859 and [0077] As used herein, the terms “derivative of insulin” and polymer bound GLP-1 and GLP-1 analogues. “insulin derivative” refer to a polypeptide which has a [0081] As used herein, the term “dual GLP-l receptor/glu molecular structure which formally can be derived from the cagon receptor agonist” refers to compounds which have structure of a naturally occurring insulin, for example that of agonistic activity at both the GLP-1 receptor and the glucacon human insulin, in which one or more organic substituents receptor. Examples of dual GLP-l receptor/glucagon recep (e. g. a fatty acid) is bound to one or more of the amino acids. tor agonist include, but are not limited to, the following: Optionally, one or more amino acids occurring in the natu , MAR701, MAR-709, and BHM081/ rally occurring insulin may have been deleted and/ or replaced BHM089/BHM098. by other amino acids, including non-codeable amino acids, or [0082] As used herein, the term “human insulin” refers to amino acids, including non-codable, have been added to the the human hormone whose structure and properties are well naturally occurring insulin. Examples of derivatives of insu known. Human insulin has two polypeptide chains (chains A lin include, but are not limited to, the following: and B) that are connected by disulphide bridges between (i). ‘Insulin detemir’ which differs from human insulin in that residues, namely the A-chain and the B-chain. The the C-terminal in position B30 is removed and a A-chain is a 21 amino acid peptide and the B-chain is a 30 fatty acid residue (myristic acid) is attached to the epsilon amino acid peptide, the two chains being connected by three amino function of the lysine in position B29. disulphide bridges: one between the cysteins in position 6 and (ii). ‘Insulin degludec’ which differs from human insulin in 1 1 of the A-chain; the second between the cysteine in position that the last amino acid is deleted from the B-chain and by the 7 of the A-chain and the cysteine in position 7 of the B-chain; addition of a glutamyl link from LysB29 to a hexadecandioic and the third between the cysteine in position 20 of the acid. A-chain and the cysteine in position 19 of the B-chain. [0078] As used herein, the term “FGF-21” means “?bro [0083] As used herein, the term “including” is used to mean blast growth factor 21”. FGF-21 compounds may be human “including but not limited to”. “Including” and “including but FGF-21, an analogue of FGF-21 (referred to “FGF-21 ana not limited to” are used interchangeably. logue”) or a derivative of FGF-21 (referred to “FGF-21 [0084] As used herein, the term “isoelectric point” (pl, IEP) derivative”). refers to the pH value at which a particular molecule carries US 2014/0221285 A1 Aug. 7, 2014

no net electrical charge. The isoelectric point can be deter contains amino acids having free basic side chains, it has a mined by using isoelectric focusing, which is a technique for certain buffering capacity and is therefore considered to be a separating different molecules by differences in their isoelec buffering agent. Protamine may be used as protamine sulfate tric point and which is well known in the art. It can also be or protamine hydrochloride. calculated (see e.g. Levene and Sims, “Calculation of iso [0094] Concentrations, amounts, solubilities, particle size, electric point” J. Biol. Chem., 1923, pp. 801-813). wavelength, pH values, weight mass, molecular weight, per [0085] As used herein, the term “kit” refers to a product cent and other numerical date may be expressed or presented (e.g. medicament, kit-of-parts) comprising one package or herein in a range format. It is to be understood that such a one or moreseparate packages of: range format is used merely for convenience and brevity and (i). A pharmaceutical formulation containing an active phar thus should be interpreted ?exibly to include not only the maceutical ingredient and at least one further active pharma numerical values explicitly recited as the limits of the range, ceutical ingredient and optionally a medical device. The at but also to include all the individual numerical values or least one further active pharmaceutical ingredient may be sub-ranges encompassed within that range as if each numeri present in said pharmaceutical formulation, i.e. the kit may cal value and sub-range is explicitly recited. comprise one or more packages, wherein each package com [0095] As used herein, the term “long acting insulin” refers prises one pharmaceutical formulation which comprises two to insulin analogues and/ or insulin derivatives, wherein the or more active pharmaceutical ingredients. The further active insulin-mediated effect begins within 0.5 to 2 hours and con pharmaceutical ingredient may also be present in a further tinues to be active for about or more than 24 hours. Examples pharmaceutical formulation, i.e. the kit may comprise sepa of fast acting insulins include, but are not limited to, the rate packages of two or more pharmaceutical formulations, following: (i). insulin glargin; (ii). insuline detemir and (iii). wherein each pharmaceutical formulation contain one active insulin degludec. pharmaceutical ingredient. Or [0096] As used herein, the term “stability” refers to the (ii). A pharmaceutical formulation containing an active phar chemical and/or physical stability of active pharmaceutical maceutical ingredient and medical device. ingredients, in particular of insulin analogues and/or deriva [0086] A kit may comprise one package only or may com tives. The purpose of stability testing is to provide evidence prise one or more separate packages For example, the kit may on how the quality of an active pharmaceutical ingredient or be a product (e. g. medicament) containing two or more vials dosage form varies with time under the in?uence of a variety each containing a de?ned pharmaceutical formulation, of environmental factors such as temperature, humidity, and wherein each pharmaceutical formulation contains at least light, and to establish a shelf life for the active pharmaceutical one active pharmaceutical ingredient. For example, the kit ingredient or dosage form and recommended storage condi may comprise (i.) a vial containing a de?ned pharmaceutical tions. Stability studies can include testing of those attributes formulation and (ii). further a tablet, capsule, powder or any of the active pharmaceutical ingredient that are susceptible to other oral dosage form which contains at least one further change during storage and are likely to in?uence quality, active pharmaceutical ingredient. The kit may further com safety, and/ or e?icacy. The testing can cover, as appropriate, prise a package lea?et with instructions for how to administer the physical, chemical, biological, and microbiological the pharmaceutical formulation and the at least one further attributes, preservative content (e.g., antioxidant, antimicro active pharmaceutical ingredient. bial preservative), and functionality tests (e. g. for a dose [0087] As used herein, the term “medical device” means delivery system). Analytical procedures can be fully vali any instrument, apparatus, implant, in vitro reagent or similar dated and stability indicating. In general, signi?cant changes or related article that is used to diagnose, prevent, or treat a for an active pharmaceutical ingredient and/or dosage form disease of other condition, and does not achieve its purpose with regard to stability are de?ned as: through pharmacological action within or on the body. [0097] a 5% change in assay from its initial value; or [0088] As used herein, a medical device may be a syringe, failure to meet the acceptance criteria for potency when an insulin injection system, an insulin infusion system, an using biological or immunological procedures; insulin pump or an insulin pen injection device. As used [0098] any degradation products exceeding its accep herein, a medical device may be mechanically or electrome tance criterion; chanically driven. [0099] failure to meet the acceptance criteria for appear [0089] As used herein, unless speci?cally indicated other ance, physical attributes, and functionality test (e. g., wise, the conjunction “or” is used in the inclusive sense of color, phase, separation, resuspendibility, caking, hard “and/or” and not the exclusive sense of “either/ or”. ness, dose delivery per actuation); however, some [0090] As used herein, the term “pH” and “pH value” refer changes in physical attributes (e. g. softening of supposi to the decimal logarithm of the reciprocal of the hydrogen ion tories, melting of creams) may be expected under accel activity in a solution. erated conditions; and, as appropriate for the dosage [0091] As used herein, the term “pharmaceutical” refers to form: the intended use in the medical diagnosis, cure, treatment [0100] failure to meet the acceptance criterion for pH; or and/ or prevention of diseases. [0101] failure to meet the acceptance criteria for disso [0092] As used herein, the term “pharmaceutically accept lution for 12 dosage units. able” refers to physiologically well tolerated by a mammal or [0102] The signi?cant changes may also be evaluated a human. against established acceptance criteria prior to starting the [0093] As used herein, the term “protamine” refers to a evaluation of the stability. mixture of strongly basic peptides. It was originally isolated [0103] Acceptance criteriacan be derived from the mono from the sperm of salmon and other species of ?sh but is now graphs (e.g. monographs for the European Pharrnacopeia, of produced primarily recombinant through biotechnology. It the United States Pharmacopeia, of the British Pharrnacopeia, contains more than two-thirds of L-arginine. As protamine or others), and from the analytical batches of the active phar US 2014/0221285 A1 Aug. 7, 2014

maceutical ingredient and medicinal product used in the pre [0107] Further embodiments of the present invention clinical and clinical studies. Acceptable limits should be pro include the following: posed and justi?ed, taking into account the levels observed in [0108] In one aspect, the invention provides a pharmaceu material used in preclinical and clinical studies. Product char tical formulation comprising (a). at least one analogue and/or acteristics may be visual appearance, purity, color and clarity derivative of insulin; and (b). Zn(II); and (c). sodium chloride; for solutions/ suspensions, visible particulates in solutions, and (d). optionally protamine; wherein the pharmaceutical and pH. As a non-limiting example, suitable acceptance cri formulation has a pH value in the range of from 6.0 to 9.0 and teria for insulin aspart formulations are shown below: is free of any additional buffering agent. [0109] In one aspect, the pharmaceutical formulation of the invention is an aqueous pharmaceutical formulation. Test item Acceptance criteria for clinical trials [01 1 0] In one aspect, the pharmaceutical formulation of the Appearance of solution (visual) invention has a pH value in the range from 7.0 to 7.8. Clarity and degree of opalescence Monitoring [01 1 1] In one aspect, the pharmaceutical formulation of the Degree ofcoloration Monitoring invention comprises an analogue of insulin selected from the Assay insulin aspart units (HPLC) 90.0 insulin aspart units/mL to group consisting of insulin aspart, insulin lispro and insulin 110.0 insulin aspart units/mL glulisine. Related impurities (HPLC) [0112] In one aspect, the pharmaceutical formulation of the B28isoAsp insulin aspart equal or below to 2.5% invention comprises a derivative of insulin which is insulin Total of A2 lAsp insulin aspart, equal or below to 5.0% detemir and/or insulin degludec. B3Asp insulin aspart and B3isoAsp insulin aspart [0113] In one aspect, the pharmaceutical formulation of the Any other unspeci?ed, unidenti?ed equal or below to 2.0% invention comprises an analogue and/or derivative of insulin impurity which is present in a concentration from 10 U/mL to 1000 Total of other impurities equal or below to 3.5% U/mL. High molecular weight proteins equal or below to 1.5% (HPSEC) [0114] In one aspect, the pharmaceutical formulation of the pH 7.0 to 7.8 invention comprises Zn(II) in a concentration from 0.0100 to Particulate matter (visible particles) Practically free of visible particles 0.0600 mg/100 U of the analogue and/or derivative of insulin. Particulate matter (subvisible Number of particles per container: particles) equal or larger to 10 pm: [0115] In one aspect, the pharmaceutical formulation of the equal or below to 6000 invention comprises sodium chloride in a concentration from equal or larger to 25 pm: 0.01 to 15 mg/mL. equal or below to 600 Assay m—cresol 1.55 to 1.89 [mg/mL] [01 1 6] In one aspect, the pharmaceutical formulation of the Assay phenol 1.35 to 1.65 [mg/mL] invention comprises sodium chloride in a concentration from Zinc (Zn(II)) (AAS) below 40 pg per 100 units 6.8 to 8.3 mg/mL. insulin aspart [0117] In one aspect, the pharmaceutical formulation of the invention comprises protamine sulfate which in a concentra [0104] The acceptance criteria shown above are based on tion from 0.1 to 0.5 mg/mL. monographed acceptance limits (e. g. British Pharmacopoeia, [0118] In one aspect, the pharmaceutical formulation of the Volume III, 2012 or Pharrnacopoeial Forum, Volume 36(6), invention is free of any additional buffering agent selected November-December 2010) and/ or are derived from exten from the group consisting of 2-amino-2-hydroxymethyl-pro sive experience in the development of insulin formulations. pane-1,3-diol (TRIS), phosphate, citric acid, citrate, acetic acid, acetate, glycylglycine and methionine. [0105] As used herein, the term “treatment” refers to any treatment of a mammalian, for example human condition or [0119] In one aspect, the pharmaceutical formulation of the disease, and includes: (1) inhibiting the disease or condition, invention comprises one or more further active pharmaceuti i.e., arresting its development, (2) relieving the disease or cal ingredients. condition, i.e., causing the condition to regress, or (3) stop [0120] In one aspect, the pharmaceutical formulation of the ping the symptoms of the disease. invention comprises a further active pharmaceutical ingredi ent which is an antidiabetic agent. [0106] As used herein, the unit of measurement “U” and/or “international units” refers to the blood glucose lowering [0121] In one aspect, the pharmaceutical formulation of the activity of insulin and is de?ned (according to the World invention comprises a further active pharmaceutical ingredi Health Organization, WHO) as follows: 1 U corresponds to ent which is an antidiabetic agent selected from the group the amount of highly puri?ed insulin (as de?ned by the WHO) consisting of (a) a GLP-1 receptor agonist; (b) a dual GLP-l which is suf?cient to lower the blood glucose level of a rabbit receptor/glucagon receptor agonist; (c) human FGF-2l; (d) (having a body weight of 2-2.5 kg) to 50 mg/100 mL within 1 an FGF-21 analogue; (e) an FGF-21 derivative; (f) insulin; (g) hour and to 40 mg/100 mL within 2 hours. For human insulin, human insulin; (h) an analogue of insulin; and (i) a derivative 1 U corresponds to approximately 35 kg (Lill, PharmaZie in of insulin. unserer Zeit, No. 1, pp. 56-61, 2001). For insulin aspart, 100 [0122] In one aspect, the pharmaceutical formulation of the U correspond to 3.5 mg (product information NovoRapid®). invention comprises more than one analogue and/or deriva For insulin lispro, 100 U correspond to 3.47 mg (product tive of insulin, wherein one analogue and/or derivative of information Humalog®). For insulin glulisine, 100 U corre insulin is a fast acting insulin and one analogue and/or deriva spond to 3.49 mg (product information Apidra® cartridges). tive of insulin is a long acting insulin. For insulin determir, 100 U correspond to 14.2 mg (product [0123] In one aspect, the pharmaceutical formulation of the information Levemir®). For insulin glargin, 100 U corre invention comprises a fast acting insulin is selected from the spond to 3.64 mg (product information Lantus®). group consisting of insulin aspart, insulin lispro, and insulin US 2014/0221285 A1 Aug. 7, 2014

glulisine, and wherein the long acting insulin is one or more one analogue and/or derivative of insulin is a fast acting insulin selected from the group consisting of insulin detemir insulin and one analogue and/or derivative of insulin is a long and insulin degludec. acting insulin. [0124] In one aspect, the pharmaceutical formulation of the invention consists of: [0163] In one aspect, the kit of the invention comprises a [0125] (a). 3.5 mg/mL insulin aspart; fast acting insulin selected from the group consisting of insu [0126] (b). 1.72 mg/mL metacresol; lin aspart, insulin lispro and nsulin glulisine, and a long acting [0127] (c). 1.50 mg/mL phenol; insulin selected from the group consisting of insulin glargin, [0128] (d). 0.04087 mg/mL Zn(II); insulin detemir and insulin degludec. [0129] (e). 6.8 mg/mL sodium chloride; [0164] In one aspect, the invention provides a pharmaceu [0130] (f). 0.02 mg/mL polysorbate 20; tical formulation or kit for use in the treatment of diabetes [0131] (g). sodium hydroxide and/or hydrochloric acid mellitus. to adjust the pH to 7.4, and [0132] (h). water. [0165] In one aspect, the invention provides a pharmaceu [0133] In one aspect, the pharmaceutical formulation of the tical formulation or kit for use in the treatment of hypergly invention consists of: cemia. [0134] (a). 3.5 mg/mL insulin aspart; [0166] In one aspect, the invention provides a pharmaceu [0135] (b). 1.72 mg/mL metacresol; tical formulation or kit for use in lowering blood glucose [0136] (c). 1.50 mg/mL phenol; level. [0137] (d). 0.04087 mg/mL Zn(II); [0138] (e). from 6.8 mg/mL to 8.3 mg/mL sodium chlo [0167] In one aspect, the invention provides a method of ride; treating diabetes mellitus in a subject in need thereof com [0139] (f). 0.02 mg/mL polysorbate 20; prising administering a pharmaceutical formulation of the [0140] (g). from 0.1 mg/mL to 0.5 mg/mL protamine invention. sulfate; [0141] (h). sodium hydroxide and/or hydrochloric acid [0168] In one aspect, the invention provides a method of to adjust the pH to a pH in the range from 7.1 to 7.6; and treating hyperglycemia in a subject in need thereof compris ing administering a pharmaceutical formulation of the inven [0142] (i). water. tion. [0143] In one aspect, the invention provides a process for preparing the pharmaceutical formulation of the invention, [0169] In one aspect, the invention provides a method of wherein the components are mixed together in the form of a lowering blood glucose levels in a subject in need thereof solution or suspension, the pH is adjusted to reach the desired comprising administering a pharmaceutical formulation of pH, and water is added to reach the ?nal volume. the invention. [0144] In one aspect, the invention provides a kit compris ing one or more separate packages of: [0170] In one aspect, the invention provides a medical [0145] (a). a pharmaceutical formulation of the inven device for administering a pharmaceutical formulation of the tion; and invention to an animal and/ or human. [0146] (b). a medical device. [0171] The present invention is illustrated by the following [0147] In one aspect, the invention provides a kit compris Examples. However, it should be understood that the present ing one or more separate packages of: invention is not limited to the speci?c details of these [0148] (a). a pharmaceutical formulation of the inven examples. tion; and [0149] (b). at least one further active pharmaceutical ingredient; EXAMPLES [0150] (c). and optionally a medical device. [0151] In one aspect, the kit of the invention comprises a Example 1 further active pharmaceutical ingredient which is an antidia betic agent. Manufacturing Process [0152] In one aspect, the kit of the invention comprises a further active pharmaceutical ingredient which is an antidia betic agent selected from the group consisting of: (a) Polysorbate Solution [0153] (a). a GLP-1 receptor agonist; [0154] (b). a dual GLP-l receptor/glucagon receptor [0172] Polysorbate Solution was prepared by dissolving agonist; 1.00 g polysorbate 20 in water for injection (according to Ph. [0155] (c). human FGF-21; Eur.) and by ?lling up with water for injection to ?nal volume [0156] (d). an FGF-21 analogue; of 1000 mL. [0157] (e). an FGF-21 derivative; [0158] (f). insulin; (b) Zinc Chloride Solution [0159] (g). human insulin; [0160] (h). an analogue of insulin; and [0173] Zinc Chloride Solution (containing Zn(II)) was pre [0161] (i). a derivative of insulin. pared by dissolving 2.00 g Zinc chloride in water for injection [0162] In one aspect, the kit of the invention comprises and by ?lling up with water for injection to ?nal volume of more than one analogue and/ or derivative of insulin, wherein 1000 mL. US 2014/0221285 Al Aug. 7, 2014

(0) Solution A [0184] Final Solution was prepared as described in the fol lowing: [0174] The ?nal composition of SolutionA is given in Table [0185] 1. It was started with approximately 300 mL 1: water for injection (according to Ph. Eur.). [0186] 2. 3.496 g (rounded 3.5 g) insulin aspart were TABLE 1 added to the 300 mL water for injection while stirring Composition of SolutionA constantly (a suspension of insulin aspart in water for injection is formed). Composition Composition Composition [0187] 3. pH value was checked. per 200 mL per 400 mL per 1000 mL [0188] 4. pH value was changed to approximately 3.1 to 1. Sodium chloride 6.8 g 13.60 g 34.00 g 3.2 by adding hydrochloric acid 0.1 N or sodium hydrox 2. Phenol 1.5 g 3.0 g 7.5 g ide solution 0.02 N to dissolve the insulin aspart. 3. m—Cresol 1.72 g 3.44 g 8.6 g [0189] 5. Solution was stirred for approximately 15 min 4. Sodium hydroxide ad pH 8.65 ad pH 8.65 ad pH 8.65 using a magnetic stirrer. solution (rounded: (rounded: (rounded: pH 9.0) pH 9.0) pH 9.0) [0190] 6. 40.866 g Zinc Chloride Solution was added to 5. Hydrochloric acid ad pH 8.65 ad pH 8.65 ad pH 8.65 the solution while stirring constantly. (rounded: (rounded: (rounded: [0191] 7. 40 g Polysorbate Solution was added while pH 9.0) pH 9.0) pH 9.0) 6. Water for Injection ad 200 mL = ad 400 mL = ad 1000 mL = stirring constantly. 205.2 g 410.4 g 1026 g [0192] 8. Solution was ?lled up to 600 g with water for injection. [0193] 9. 410.4 g SolutionA was added slowly and care [0175] SolutionA was prepared as described in the follow fully while stirring constantly. ing: [0194] 10. pH was adjusted to 7.4 (range 7.2 to 7.6) using [0176] 1. It was started with approximately 500 mL hydrochloric acid 0.1 N or sodium hydroxide solution water for injection. 0.02 N. [0177] 2. 34.00 g sodium chloride, 7.5 g phenol and 8.6 [0195] 11. Solution was ?lled up to 2010 g (corresponds g m-cresol were dissolved while stirring constantly. to 100% of the Final Solution). [0178] 3. Solution was ?lled up to approximately 900 g [0196] Quality control: Final solution was a clear and with water for injection. uncolored solution, showed a pH value of 7.4 (plus/minus 0.2; [0179] 4. Solution was stirred for approximately 15 min at 20-25° C.). using a magnetic stirrer. [0197] The Final Solution was applied to sterile ?ltration [0180] 5. pH was checked (pH should be 8.65, rounded: using “Sartopore Minisart high ?ow” ?lter (?lter material: pH 9.0). prH value is not 8.65, the pH was adjusted to polyethersulfone; pore size: 0.2 km; supplier: Sartorius). said range using hydrochloric acid 1 N or sodium [0198] The Final Solution after sterile ?ltration was a clear hydroxide solution 1 N. and uncolored solution and showed an osmolality of 260 [0181] 6. Solution was ?lled up to 1026 g with water for mOsmol/kg (plus/minus 30). injection. [0199] The Final Solution after sterile ?ltration was ?lled into appropriate vials (volume: 5 and 10 mL; 13 mm; clear (d) Final Solution glas; glas type 1). [0200] The vials4containing the Final Solution after ster [0182] The ?nal composition of Final Solution is given in ile ?ltrationiwas stored between +2o C. and +8° C. and Table 2: protected from light. TABLE 2 Example 2

Compo ition ofFinal Solution Control of the Formulation Composition Composition Composition Excipient per mL per 1000 mL per 2000 mL (a) Analytical Procedures 1. Insulin aspart 3.496 mg 3.496 g 6.992 g [0201] Tests were carried out using compendial analytical (rounded: (rounded: (rounded: test methods, where applicable. The quality control concept 3.5 mg) 3.5 g) 7.0 g) 2. 211(11) 40.87 [1g 0.04087 g 0.08174 g has been established taking into account the cGMP require 3. Sodium chloride 6.80 mg 6.80 g 13.60 g ments as well as the current status of the ICH process. 4. Phenol 1.50 mg 1.50 g 3.00 g [0202] The non-compendial and chromatographic analyti 5. m—Cresol 1.72 mg 1.72 g 3.44 g cal procedures used to control the formulation are summa 6. Polysorbat 20 0.02 mg 0.02 g 0.04 g 7. Sodium hydroxide ad pH 7.4 ad pH 7.4 ad pH 7.4 rized in the following: solution 8. Hydrochloric acid ad pH 7.4 ad pH 7.4 ad pH 7.4 Description 9. Water for Injection ad 1 mL = ad 1000 mL = ad 2000 mL = 1.005 g 1005 g 2010 g [0203] Visually examined a number of containers for con formance to the acceptance criteria. [0183] In the following, preparation of the composition per Identi?cation (HPLC) 2000 mL is described. Other volumes (e.g. composition per 1000 mL) can be prepared in the same way (using the corre [0204] The identity of the active ingredient was ensured by sponding amount of insulin aspart and excipients). comparing the retention time of the drug formulation sample US 2014/0221285 A1 Aug. 7, 2014

with the retention time of the reference standard using a Antimicrobial Preservative Assay reverse phase HPLC method. The method was also used for the determination of assay of the active ingredient, for the [0216] The same chromatographic conditions as for “Assay determination of the related compounds and impurities, and (HPLC)” were used for the determination of assay of for quantifying the preservatives m-cresol and phenol. m-Cresol and of phenol m-cresol and phenol were calculated by external standardization. Assay (HPLC) [0205] The test was carried out by reverse phase liquid (b) Validation of Analytical Procedures chromatography (HPLC). The method was also used for the identi?cation, the determination of assay of the active ingre [0217] The HPLC analytical procedure for the formulation dient, for the determination of the related compounds and for the determination of identi?cation, assay, and related impurities, and for quantifying the preservatives m-cresol and compounds and impurities was validated to demonstrate phenol. Column: Lichrosorb RP18, particle size 5 pm, pore speci?city, linearity, limit of detection and limit of quanti? size 100 A (250 mm><4.0 mm), thermostated at +350 C. cation, accuracy, precision and range. Autosampler: Thermostated at o C. Mobile phase A: Sodium sulfate solved in water, 14 g/mL, adjusted with phosphoric (c) Justi?cation of the Acceptance Criteria Tests and accep acid and sodium hydroxide to a pH of 3.4. tance criteria, as previously presented, were selected based on [0206] Mobile phase B: Water/acetonitrile (50:50 v/v). ICH Q6B and on published monographs, analytical results Gradient is shown in Table 3. obtained, precision of procedures used, Pharmacopoeial and/ or regulatory guidelines, and were in agreement with the TABLE 3 standard limits at this stage of development.

HPLC gradient Example 3 Time [min] Mobile phaseA [%] Mobile phase B [%]

O to 42 57.7 42.3 Stability of the Formulation 42 to 47 linear to 20 80 47 to 52 2O 80 52 to 53 linear to 57.7 42.3 (a) Stability of the Formulation 53 to 60 equilibration 57.7 42.5 [0218] Stability studies for the formulation were initiated [0207] Flow rate: 1.0 mL/min. Injection volume: 10 uL. according to the stability protocol summary described in the Detection: 214 nm (for the active ingredient) and following table. The composition and manufacturing method [0208] 260 nm (for m-cresol and phenol). Typical run time: of the stability batches were representative of the material. 60 min. The stability pro?le was assessed for storage under long term, [0209] Assay of the active ingredient, m-cresol and phenol accelerated, and stress testing conditions according to ICH were calculated by external standardization. guidelines. Samples were packed and stored in glass vials [0210] Impurities were calculated using the peak area per with ?anged cap with inserted disc and ?ip-off lid. The sta cent method. bility data obtained using this packaging material were rep [021 1] Test solution: The formulation was used without any resentative for the preliminary shelf life and storage direction dilution or further treatment. for both packaging con?gurations (10 mL glass vials and 3 mL cartridges). Up to now, 12 months stability data are avail Related Compounds and Impurities (HPLC) able from a batch ?lled into 10 mL vials and 12 months of a batch ?lled into 3 mL cartridges ongoing stability studies of [0212] The same chromatographic conditions as for “Assay the formulation. (HPLC)” were used for the determination of related com pounds and impurities. Related compounds and Impurities TABLE 4 were calculated using the peak area percent method. Storage Conditions

High Molecular Weight Proteins (HMWPs) Storage Condition Sampling Intervals Container [0213] The high molecular weight proteins were deter Long Term mined using high pressure size exclusion chromatography (HPSEC). Column: Waters Insulin HMWP, particle size 5-10 +5O C.:3O C. 1,2,3,6, 9, and 10 vaials pm, pore size 12-12.5 nm (300 mm><7.8 mm), thermostated at 12 months room temperature. Autosampler: thermostated at s+8° C. +5° C. 13° C. 1,2,3, 6,9, 12, 3mL vials 18, 24 and 36 months Mobile phase: 650 mL of arginine solution (1 g/ L) was mixed Accelerated with 200 mL of acetonitrile and 150 mL of glacial acetic acid. Isocratic elution Flow rate: 1.0 mL/min. Injection volume: +25O C. r 1, 2, 3, and 6 months 10 mL vials and 100 uL. 20 C./60% : 5% RH 3 mL cartridges [0214] Detection: 276 nm. Typical run time: 35 min. Stress [0215] HMWPs were calculated using the peak area per +40O C. r 1 month 10 mL vials and cent method. Test solution: The formulation was used without 50 C./75% : 5% RH 3 mL cartridges any dilution or further treatment. US 2014/0221285 A1 Aug. 7, 2014

TABLE 4-continued wherein batch no. ‘ i318” refers to a formulation according to the present invention ?lled into 3 mL cartridges. Storage Conditions (b) Comparison of Stability of the Buffer-Free Formulation Storage Condition Sampling Intervals Container Against Buffer Formulations Photostability [0223] A buffered formulation containing a phosphate buffer showed physical instability by formation of anorganic Sun test according to ICH 1 day 3 mL cartridges guidelines* particles (formation of sodium Zinc phosphate hydrate, Na6 Indoor light** 14 days 3 mL cartridges (ZnPO4)6.8 H20, Sodalite-type crystal structure). Another buffer formulation containing citrate buffer showed physical *Overall illumination ofnot less than 1.2 million lux hours and an integrated near ultraviolet instability as well, by turning turbid after exposing to slight energy ofnot less than 200 watt hours/m2. A dark control sample is stored under the same conditions to eliminate any effects due to local temperature changes physical stress. A buffered formulation containing 2-amino **Variolux, Heraeus, standard ?uorescent tubes, GE-Lightening, Type F40/33, irradiance approximately 8 W/m2, 2000 Lux. A dark control sample is stored under the same conditions 2-hydroxymethyl-propane-1,3-diol (synonym: TRIS) to evaluate any effects due to local temperature changes showed an increase of the related impurities when stored under accelerated conditions. Additionally, the buffered for [0219] The following tests were performed during stability mulation containing 2-amino-2-hydroxymethyl-propane-1, testing: appearance, assay, related impurities, high molecular weight proteins, pH, particulate matter (visible and subvisible 3-diol (TRIS) showed three additional impurites (identi?ed particles), assay of antimicrobial preservatives (m-cresol and by LC/MS) when stored under stress conditions (+40o phenol), content of Zinc. The investigations on physical and C./75% RH), i.e. a TRIS-adduct +103 Da; a corresponding Desamido-adduct +104 Da; and a Formaldehyde-adduct +12 chemical properties after 12 months of storage at the long Da (see FIG. 1). The formulation according to the invention term storage condition of +5° C. con?rm the stability of the does not show these impurities. formulation when stored at the recommended storage condi tion. Only very slight changes of the related impurities could [0224] The relevant items tested and the analytical results after one month stability, at long term, accelerated and stress be observed. conditions regarding the physico-chemical stability in com [0220] When stored at accelerated conditions for 3 months parison to the formulation according to the invention are at +25° C./ 60% RH the related impurities and high molecular listed in Table 13. weight proteins increased, however stayed within the current [0225] After storage of the formulations at long-term stor acceptance limit. When stored at stress conditions (1 month at age conditions (1 month at +5° C.), the formulations were +400 C./75% RH) one of the related impurities increased applied to mechanical stress (shaking) and thermal stress above the acceptance criterion. The content of the active (+37o C.). The turbidity was measured (nephelometric inves ingredient, m-cresol and phenol remained basically tigation). unchanged under accelerated conditions. [0226] The stability of the buffer-free formulation (i.e., the [0221] Due to the present results of the stability studies of alternative aqueous pharmaceutical formulation comprising the formulation, the chemical and physical stability of the at least one insulin analogue and/or insulin derivative com formulation can be con?rmed. prising sodium chloride and without any additional buffering [0222] Tables 5-12 show the long term stability results, agent) shows an excellent chemical and physical stability wherein batch no. “i0105” is referring to a formulation which quali?es said aqueous pharmaceutical formulation as according to the present invention ?lled into 10 mL vials and medicinal product having a de?ned shelf life. TABLE 5

Long term stability +5O C. — batch i0105

Acceptance Time [months]

Test item criteria Initial 2 3 6 9 12

Appearance of solution (visual)

Clarity Monitoring

B28isoAsp insulin aspart 52.5% 0.20% 0.29% 0.36% 0.42% 0.53% 0.63% 0.79% A21 Asp insulin aspart Monitoring 1.29% 1.14% 1.09% 1.14% 1.19% 1.23% 0.44% US 2014/0221285 A1 Aug. 7, 2014 l 4

TABLE 5-continued

Long term stability +5O C. — batch i0105

Acceptance Time [months]

Test item criteria Initial 1 2 3 6 9 12

B3 Asp insulin aspart Monitoring

Total ofA21 Asp insulin 55.0% 1.37% 1.23% 1.19% 1.27% 1.32% 2.16% 1.61% aspart, B3Asp insulin aspart and B3isoAsp insulin aspart Any other impurity 52.0% 0.69% 0.73% 0.59% 0.65% 0.83% 0.58% 0.46% Total ofother impurities 53.5% 0.87% 0.92% 0.75% 0.84% 0.91% 0.81% 0.62% High molecular weight 51.5% 0.24% 0.24% 0.26% 0.28% 0.31% 0.33% 0.32% proteins (HPSEC) pH Between 7.33 7.33 7.35 7.34 7.31 7.38 7.49 7.0 to 7.8 Particulate matter Practically complies complies complies complies complies complies Complies (visible particles) free from visible particles Assay m—cresol 1.55 to 1.89 1.72 QOL 1.72 mg/mL 1.71 QOL 1.69 mg/mL 1.69 QOL 1.68 QOL 1.71mg/mL mg/mL (90.0% (100.0%) (100.0%) (99.2%) (98.3%) (98.3%) (97.7%) (99.2%) to 110.0% of label claim) Assay phenol 1.35 to 1.65 1.49 QOL 1.48 mg/mL 1.49 QOL 1.47 mg/mL 1.47 QOL 1.48 QOL 1.49 mg/mL mg/mL (90.0% (99.5%) (98.6%) (99.3%) (98.0%) (98.0%) (98.6%) (99.5%) to 110.0% of label claim) Zinc (AAS) <40 pg per 19.7 pg per Not tested Not tested Not tested Not tested Not tested 19.5 pg per 100 units 100 units 100 units insulin aspart insulin aspart insulin aspart (20.6 ug/mL) (20.1 ug/mL) Nephelometry Monitoring 0.77 Not tested 0.77 Not tested <1 (water Turbid after clear) 3 days

TABLE 6

Accelerated stability +25O C./60%RH — batch i0105

Acceptance criteria Time

Test item for clinical trials Initial results 1 month 2 months 3 months 6 months Appearance of solution (visual)

Clarity and degree of Monitoring <1 (water <1 (water <1 (water <1 (water <1 (water opalescence clear) clear) clear) clear) clear) Degree of coloration Monitoring B9 B9 B9 B9 B9 Assay insulin aspart 90.0 insulin 104.6 104.6 104.3 103.7 101.8 units (HPLC) aspart units/mL units/mL units/mL units/mL units/mL units/mL to 110.0 insulin aspart units/mL Related impurities (HPLC)

B28isoAsp insulin aspart 52.5% 0.20% 0.84% 1.49% 2.24% 4.11% Total ofA21Asp insulin aspart, 55.0% 1.37% 1.50% 1.74% 2.14% 2.90% B3Asp insulin aspart and B3isoAsp insulin aspart Any other unspeci?ed, 52.0% 0.69% 0.88% 0.93% 1.26% 2.16% unidenti?ed impurity Total ofother impurities 53.5% 0.87% 1.08% 1.11% 1.50% 2.40% High molecular weight 51.5% 0.24% 0.35% 0.48% 0.67% 1.07% proteins (HPSEC) pH 7.0 to 7.8 7.3 7.3 7.4 7.3 7.2 Particulate matter Practically free conforms conforms conforms conforms conforms (visible particles) ofvisible particles Particulate matter Number of particles (subvisible particles) per container 210 pm: 56000 33 not tested not tested not tested 3 225 pm: 5600 1 0 Assay m—cresol 1.55 to 1.89 1.72 QOL 1.72 mg/mL 1.71 QOL 1.70 mg/mL 1.69 mg/mL US 2014/0221285 A1 Aug. 7, 2014 15

TABLE 6-continued

Accelerated stability +25O C./60%RH — batch i0105

Acceptance criteria Time

Test item for clinical trials Initial results 1 month 2 months 3 months 6 months

Assay phenol 1.35 to 1.65 1.49 QOL 1.48 mg/mL 1.49 QOL 1.47 mg/mL 1.47 mg/mL [mg/mL] Zinc (MS) <40 pg per 100 19.7 119 not tested not tested not tested testing units insulin 100 U ongoing aspart

TABLE 7

Stress stability +40O C./75%RH — batch i0105

Acceptance criteria Time

Test item for clinical trials Initial result 1 months Appearance of solution (visual) Clarity and degree of Monitoring

B28isoAsp insulin aspart 52.5% 0.20% 4.44% Total ofA21Asp insulin 55.0% 1.37% 2.81% aspart, B3Asp insulin aspart and B3isoAsp insulin aspart Any other unspeci?ed, 52.0% 0.69% 2.62% unidenti?ed impurity Total ofother impurities 53.5% 0.87% 2.97% High molecular weight 51.5% 0.24% 1.28% proteins (HPSEC) pH 7.0 to 7.8 7.3 7.3 Particulate matter Practically free conforms conforms (visible particles) ofvisible particles Particulate matter (subvisible Number of particles per container particles) 210 pm: 56000 33 8 225 pm: 5600 1 1 Assay m—cresol 1.55 to 1.89 [QOL] 1.72 QOL 1.70 mg/mL Assay phenol 1.35 to 1.65 [QOL] 1.49 QOL 1.47 mg/mL Zinc (Zn(II)) (AAS) <40 pg per 100 units insulin aspart 19.7 ugIOO U 20.9 ug100 U

TABLE 8

Long term stability +5O C. — batch i318 — up to 12 months Container/closure: 3 mL cartridges Storage condition: +5O C. 1 30 C. Storage orientation' Hori ontal

Acceptance Time

Test item criteria Initial results 1 month 3 months 6 months 9 months 12 months Appearance of solution (visual)

Clarity Monitoring

TABLE 8-continued

Long term stability +5O C. — batch i318 — up to 12 months Container/closure: 3 mL cartridges Storage condition: +5O C. 1 30 C. Storage orientation: Horizontal

Acceptance Time

Test item criteria Initial results 1 month 3 months 6 months 9 months 12 months Related compounds(HPLC)

B28isoAsp insulin aspart 52.5% 0.22% 0.40% 0.45% 0.59% 0.61% 0.74% Total ofA21Asp insulin 55.0% 0.68% 1.01% 1.04% 1.11% 0.62% 0.80% aspart, B3Asp insulin aspart and B3isoAsp insulin aspart Any other unspeci?ed, 52.0% 0.45% 0.62% 0.65% 0.67% 0.87% 0.34% unidenti?ed impurity Total ofother impurities 53.5% 0.53% 0.72% 0.74% 0.84% 0.99% 0.53% High molecular weight 51.5% 0.19% 0.24% 0.24% 0.23% 0.22% 0.26% proteins (HPSEC) pH 7.0 to 7.8 7.31 7.37 7.41 7.20 7.39 7.35 Sterility Complies complies complies Particulate matter Practically free complies complies complies complies complies complies (visible particles) from visible particles Particulate matter (subvisible particles)

Number of particles per 56000 60 not tested not tested not tested not tested 213 container 210 pm: Number ofparticles per 5600 0 0 container 225 pm: Antimicrobial preservative assay m—cresol 1.55 QOL to 1.69 QOL 1.70 mg/mL 1.69 QOL 1.70 mg/mL 1.67 QOL 1.69 mg/mL 1.89 QOL (98.3%) (98.8%) (98.3%) (101.2%) (97.2%) (98.4%) (90.0% to 110.0% of label claim) phenol 1.35 QOL to 1.48 QOL 1.49 mg/mL 1.49 QOL 1.48 mg/mL 1.46 QOL 1.49 mg/mL 1.65 QOL (98.7%) (99.3%) (99.3%) (98.7%) (97.2%) (99.4%) (90.0% to 110.0% of label claim) Zinc (AAS) <40 ug/ 100 19.4 ug/ not tested not tested not tested not tested not tested units insulin 100 units aspart insulin aspart (19.6 ugmL) Preservative ef?cacy Complies With Complies Complies Ph. Eur/USP With Ph. Eur With Ph. Eur criteria A criteria A and With USP and With USP

'1 The content of insulin aspart is calculated together With B28isoAsp insulin aspart, A21Asp insulin aspart, B3Asp insulin aspart, and B3isoAsp insulin aspart. 1 unit is equivalent to 0.0350 mg of insulin aspart

TABLE 9

Long term stability +5O C. — batch i318 — 18 to 36 months Container/closure: 3 mL cartridges Storage condition: +5O C. 1 30 C. Storage orientation: Horizontal

Time

Test item Acceptance criteria Initial results 18 months 24 months 36 months Appearance of solution (visual)

Clarity Monitoring <1 (water clear) not yet available not yet available not yet available Color Monitoring B9 not yet available not yet available not yet available Assay in insulin aspart 90.0 to 110.0 insulin 100.9 insulin not yet available not yet available not yet available units (HPLC) " aspart units/mL aspart units/mL Related compounds(HPLC)

B28isoAsp insulin aspart 52.5% 0.22% not yet available not yet available not yet available Total ofA21Asp insulin 55.0% 0.68% not yet available not yet available not yet available aspart, B3Asp insulin aspart and B3isoAsp insulin aspart US 2014/0221285 A1 Aug. 7, 2014 17

TABLE 9-continued

Long term stability +5O C. — batch i318 — 18 to 36 months Container/closure: 3 mL cartridges Storage condition: +5O C. 1 30 C. Storage orientation: Horizontal

Time

Test item Acceptance criteria Initial results 18 months 24 months 36 months

Any other unspeci?ed, 52.0% 0.45% not yet available not yet available not yet available unidenti?ed impurity Total of other impurities 53.5% 0.53% not yet available not yet available not yet available High molecular weight 51.5% 0.19% not yet available not yet available not yet available proteins (HPSEC) pH 7.0 to 7.8 7.31 not yet available not yet available not yet available Sterility Complies not yet available not yet available Particulate matter Practically free from complies not yet available not yet available not yet available (visible particles) visible particles Particulate matter (subvisible particles)

Number of particles per 56000 60 not yet available not yet available not yet available container 210 um: Number ofparticles per 5600 0 container 225 um: Antimicrobial preservative assay m—cresol 1.55 mg/mL to 1.89 mg/mL 1.69 QOL not yet available not yet available not yet available (90.0% to 110.0% (98.3%) oflabel claim) phenol 1.35 mg/mL to 1.65 mg/mL 1.48 QOL not yet available not yet available not yet available (90.0% to 110.0% (98.7%) oflabel claim) Zinc (AAS) <40 ug100 units 19.4 rig/100 units not yet available insulin aspart insulin aspart (19.6 ugmL) Preservative ef?cacy Complies With not yet available not yet available Ph. Eur/U SP

‘1 The content ofinsulin aspart is calculated together With B28isoAsp insulin aspart, A21Asp insulin aspart, B3Asp insulin aspart, and B3isoAsp insulin aspart. 1 unit is equivalent to 0.0350 mg ofinsulin aspart

TABLE 10

Accelerated stability +25O C./60% RH — batch i318 Container/closure: 3 mL cartridges Storage condition: +25O C. 1 2O C./60% : 5% RH Storage orientation' Hori outal

Time

Test item Acceptance criteria Initial results 1 month 3 months 6 months Appearance of solution (visual)

Clarity Monitoring <1 (water clear) <1 (water clear) <1 (water clear) <1 (water clear) Color Monitoring B9 B9 B9 B9 Assay in insulin aspart 90.0 to 110.0 insulin 100.9 insulin 100.5 insulin 99.2 insulin 99.8 insulin units (HPLC) " aspart units/mL aspart units/mL aspart units/mL aspart units/mL aspart units/mL Related compounds(HPLC)

B28isoAsp insulin aspart 52.5% 0.22% 1.05% 2.34% 4.06% b Total ofA21Asp insulin 55.0% 0.68% 1.35% 1.97% 2.91% aspart, B3Asp insulin aspart and B3isoAsp insulin aspart Any other unspeci?ed, 52.0% 0.45% 0.82% 1.15% 1.64% unidenti?ed impurity Total ofother impurities 53.5% 0.53% 0.95% 1.36% 2.12% High molecular weight 51.5% 0.19% 0.34% 0.51% 0.62% proteins (HPSEC) pH 7.0 to 7.8 7.31 7.35 7.39 7.22 Particulate matter Practically free from complies complies complies Complies (visible particles) visible particles Particulate matter (subvisible particles)

Number ofparticles per 56000 60 not tested not tested 164 container 210 um: US 2014/0221285 A1 Aug. 7, 2014 18

TABLE 10-continued

Accelerated stability +25O C./60% RH — batch i318 Container/closure: 3 mL cartridges Storage condition: +25O C. 1 2O C./60% : 5% RH Storage orientation: Horizontal

Time

Test item Acceptance criteria Initial results 1 month 3 months 6 months

Number ofparticles per 5600 0 0 container 225 pm: Antimicrobial preservative assay m—cresol 1.55 mg/mL to 1.89 mg/mL 1.69 QOL 1.69 mg/mL 1.68 QOL 1.67 mg/mL (90.0% to 110.0% (98.3%) (98.3%) (97.6%) (97.1%) oflabel claim) phenol 1.35 mg/mL to 1.65 mg/mL 1.48 QOL 1.49 mg/mL 1.49 QOL 1.47 mg/mL (90.0% to 110.0% (98.7%) (99.3%) (99.3%) (98.0%) oflabel claim) Zinc (MS) <40 pg 100 units 19.4 ug/ 100 units not tested not tested 20.5 ug/ 100 units insulin aspart insulin aspart insulin aspart (19.6 ugmL) (20.5 ug/mL)

‘1 The content of insulin aspart is calculated together With B28isoAsp insulin aspart, A21Asp insulin aspart, B3Asp insulin aspart1 and B3isoAsp insulin aspart. 1 unit is equivalent to 0.0350 mg ofinsulin aspart b OOS result

TABLE 11

Stress stability +40O C./75% RH — batch i318 Container/closure: 3 mL cartridges Storage condition: +40O C. 1 2O C./75% : 5% RH Storage orientation: Horizontal

Acceptance Time

Test item criteria Initial result 1 month Appearance of solution (visual) Clarity Monitoring <1 (water clear)

B28isoAsp insulin aspart 52.5% 0.22% 5.05% b Total ofA21Asp insulin 55.0% 0.68% 2.64% aspart, B3Asp insulin aspart and B3isoAsp insulin aspart Any other unspeci?ed, 52.0% 0.45% 2.38% 1’ unidenti?ed impurity Total ofother impurities 53.5% 0.53% 2.79% High molecular weight 51.5% 0.19% 1.16% proteins (HPSEC) pH 7.0 to 7.8 7.31 7.24 Particulate matter Practically free from complies complies (visible particles) visible particles Particulate matter (subvisible particles)

Number ofparticles per 56000 60 132 container 210 pm: Number ofparticles per 5600 0 0 container 225 pm: Antimicrobial preservative assay m—cresol 1.55 mg/mL to 1.89 mg/mL 1.69 QOL 1.68 QOL (90.0% to 110.0% (98.3%) (97.7%) oflabel claim) phenol 1.35 mg/mL to 1.65 mg/mL 1.48 QOL 1.48 QOL (90.0% to 110.0% (98.7%) (98.7%) oflabel claim)