US 20100080773A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2010/0080773 A1 Geho et al. (43) Pub. Date: Apr. 1, 2010

(54) ORALLY BIOAVAILABLE LIPID-BASED Publication Classification CONSTRUCTS (51) Int. Cl. A638/2 (2006.01) (75) Inventors: W. Blair Geho, Wooster, OH (US); A6II 47/6 (2006.01) John R. Lau, Howard, OH (US) A 6LX 39/395 (2006.01) A638/22 (2006.01) Correspondence Address: A638/28 (2006.01) Kathryn Doyle, Ph.D., J.D. A6II 3/405 (2006.01) Drinker Biddle & Reath LLP A638/23 (2006.01) One Logan Square, 18th and Cherry Streets (52) U.S. Cl...... 424/85.4; 514/788; 424/130.1; Philadelphia, PA 19103-6996 (US) 514/12: 514/3: 514/415 (57) ABSTRACT (73) Assignee: SDG, Inc. The present invention is embodied by a composition capable (21) Appl. No.: 12/413,293 of chaperoning a typically non-orally available therapeutic or diagnostic agent through the environment of the digestive (22) Filed: Mar. 27, 2009 tract Such that the therapetuic or diagnostic agent is bioavail able. The composition may or may not be targeted to specific O O cellular receptors, such as hepatocytes. Therapeutic agents Related U.S. Application Data include, but are not limited to, , calcitonin, serotonin, (63) Continuation-in-part of application No. PCT/US08/ and other proteins. Targeting is accomplished with biotin or 77990, filed on Sep. 26, 2008. metal based targeting agents.

Lipid Sphere Fragment Lipid Planar Carrier 50 nm

1 Hepatic Targeting Molecule

insulin o Phospholipid Patent Application Publication Apr. 1, 2010 Sheet 1 of 14 US 2010/0080773 A1

Figure 1

Lipid Sphere Fragment Lipid Planar Carrier

1 Hepatic Targeting Molecule S Insulin o Phospholipid Patent Application Publication Apr. 1, 2010 Sheet 2 of 14 US 2010/0080773 A1

Figure 2

GUT-NJECTED ORAL COMPOSITION NON-TARGETED -A PORTAL VEIN - - FEMORAL VEN

1100

88O

660

440 5% OF DOSE IN BLOODAT 30 MINUTES

220 PM

15 3O MINUTES

Oral Absorption of Composition - Intact Fasted Rats

ESOC]=IO„LNEOBJEd

N 2 Az-z- Patent Application Publication Apr. 1, 2010 Sheet 4 of 14 US 2010/0080773 A1

Figure 4

BLOODLEVELS OF COMPOSITION FROMDRINKING WATER 1 O

BLOOD LEVELS

1 O O 15 30 45 MNUTES AFTER DRINKING Figure 5

ORAL ABSORPTION OF COMPOSITIONMP FRO NORMALRATSDRINKING WATER Patent Application Publication Apr. 1, 2010 Sheet 6 of 14 US 2010/0080773 A1

Figure 6

ORAL COMPOSITION INSULIN INALLOXAN-STREPTOZOTOCIN MICE

200

2 BASELINE 100 7 DAYS HTMI (402B) N RNKINGWATER 0.1U * p < 0.01 COMPARE TO 7DAY CONTROLS

CONTROL (11) HTMNSULIN (7) INSUINDEFICIENTMC Patent Application Publication Apr. 1, 2010 Sheet 7 of 14 US 2010/0080773 A1

Figure 7

EFFECT OF DUODENAL COMPOSITION-5HT ON HEPATIC GLUCOSE BALANCE IN VAGOTOMZED DOG 80 HEPATIC UPAKE 60

40 HEPATIC OUPUT

N SINGLE DOSE DUODENAL COMPOSITION-5HT 30mg/kg PORTAL VEINGLUCOSE 0.5g/kg/hr

O 15 30 55 60 65 80 100 110 MINUTES FASTED ANESHETIZED WAGOOMIZED DOG Patent Application Publication Apr. 1, 2010 Sheet 8 of 14 US 2010/0080773 A1

Figure 8

HYPOCALCEMIC EFFECT OF ORAL CALCITONIN IN THE RAT C COMPOSITION ORAL A CONTROL CA CITONIN SQ CACITONIN

100

95

90

85

80

75

70

65

60 Patent Application Publication Apr. 1, 2010 Sheet 9 of 14 US 2010/0080773 A1

Figure 9

90.0 SDP Set 1 Wome Distribution

20 3.9 7.8 15.5 30.6 60.7 1204, 238.8 4734, 9500 Size (nm) log Patent Application Publication Apr. 1, 2010 Sheet 10 of 14 US 2010/0080773 A1

08 09 09 07 09 OZ (SpueSnou) OS + eueSe UOJ OW blueue J3U Patent Application Publication Apr. 1, 2010 Sheet 11 of 14 US 2010/0080773 A1

Figure 11

Solution Containing Composition After insulin Load vs. Insulin Solution

-Composition. After Insulin Load -Insulin Control

0.5 -1 Composition. After insulin Load

0.4

0.2 insulin

Preservative 0,1

O 10 20 30 40 50 60 70 80 90 100 Time (min) Patent Application Publication Apr. 1, 2010 Sheet 12 of 14 US 2010/0080773 A1

Figure 12

Oral Delivery of IgGAntibodies Rats (n=8) with Intraduodenal Administration of 5ug 9.00 s AUC 0-120 min | Test 632: 251(SEM)

7.20 T Control 453 81 E N g 5.40 - A - SDG MAb - O - Control 3.60 MAb s

1 80 KAnalysis: T Tmax (mint SEM); Test 39-45.9 Control 52.5 it 4.6 A ? Cmax (g Gagmiplasma) Test 6.9 it 1.4 Control 5.2 it 4.6

O.OO O 15 30 45 60 75 90 105 120 Minutes Patent Application Publication Apr. 1, 2010 Sheet 13 of 14 US 2010/0080773 A1

Figure 13

Effect of Targeted Oral Thyroxine on Serum Cholesterol in Mice Dose Response, n = 8, 1 Week Treatment — serum - - Serum Cholesterol TG

1400 250

1300 s 200 C) CO H -H 5 1200 S. g 150 E o 3 1100 9. : Statistics: g g pK 0.01 for both Cholesterol 100 O OOO and TG for High vs Low doses E 50 C) 900 8

800

Oral Dose of Thyroxine, ugimouselday Patent Application Publication Apr. 1, 2010 Sheet 14 of 14 US 2010/0080773 A1

Figure 14

Oral Alfa Treatment of Hepatitis C Genotype 3 60,000 U Interferon Alfafday 2.00

18O

SO

1.40

120

1.00

O.80

0.60

O40

O.OO 2 3 4. Weeks Treatment US 2010/0080773 A1 Apr. 1, 2010

ORALLYBOAVAILABLE LPD-BASED 3-phosphocholine, 1,2-dimyristoyl-sn-glycero-3-phospho CONSTRUCTS choline, cholesterol, cholesterol oleate, dihexadecyl phosphate, 1,2-distearoyl-sn-glycero-3-phosphate, 1,2-di PRIORITY palmitoyl-sn-glycero-3-phosphate, 1,2-dimyristoyl-sn-glyc ero-3-phosphate, 1,2-distearoyl-sn-glycero-3-phosphoetha 0001. This application is a continuation in part of PCT nolamine, 1,2-dipalmitoyl-sn-glycero-3- application PCT/US08/77990, filed on Sep. 26, 2008, which phosphoethanolamine, 1,2-dimyristoyl-sn-glycero-3- in turn claims priority to U.S. patent application Ser. No. phosphoethanolamine, 1,2-dipalmitoyl-sn-glycero-3- 11/904.937, filed on Sep. 28, 2007, each of which is herein phospho-2-mercaptoethanol. 1,2-dipalmitoyl-sn-glycero-3- incorporated by reference in its entirety. phosphoethanolamine-N-(Succinyl), 1,2-dipalmitoyl-sn glycero-3-phospho-rac-(1-glycerol) (sodium salt), and BACKGROUND OF THE INVENTION triethylammonium 2,3-diacetoxypropyl 2-(5-((3aS,6aR)-2- 0002 One of the most preferred ways to deliver a pharma oxohexahydro-1H-thieno3,4-dimidazol-4-yl)pentana ceutical to a Subject is in an oral formulation. However, oral mido)ethyl phosphate. formulations of many pharmaceutical compounds are often 0008. In certain embodiments, the therapeutic agent is unavailable due to the pharmaceutical's incompatibility with selected from the group consisting of insulin, interferon, the harsh environment of the digestive tract. This is particu , parathyroid , calcitonin, serotonin, larly true for pharmaceutical compounds such as , rituximab, , uricase, tissue plasminogen activa proteins, certain Small molecules, and nucleic acids. Repre tor, thymoglobin, a , heparin or a heparin analog, sentative examples include calcitonin, serotonin, parathyroid antithrombin, III, filgrastin, pramilitide acetate, exanatide, hormone, GLP-1, erythropoietin, interferon of various types, epifibatide, antivenins, IgG, IgM, HGH, thyroxine, GLP-1. human , monoclonal antibodies, and many blood clotting Factors VII, VIII, IX, Kallikrein, Kininogen, others, the utilities of which have been extensively reviewed Hageman Factor (XII), plasma thromboplastin antecedent in the literature. Factor (XI), tissue factor, Stuart Factor (X), accelerin (V), 0003. Thus, what is needed in the field of oral drug deliv prothrombin (II), and fibrin stabilizing Factor (XIII); a mono ery is a composition that enables oral delivery of a wide range clonal antibody, and glycolipids that act as therapeutic agents. of pharmaceutical products and other therapeutic agents. The present invention meets and addresses this need. BRIEF DESCRIPTION OF THE DRAWINGS BRIEF SUMMARY OF THE INVENTION 0009. The foregoing summary, as well as the following detailed description of preferred embodiments of the inven 0004. The present invention includes compositions that tion, will be better understood when read in conjunction with facilitate and/or enable absorption of therapeutics which are the appended drawings. For the purpose of illustrating the not typically orally bioavailable. In one embodiment, a com invention, there are shown in the drawings embodiments position of the invention functions by associating with a which are presently preferred. It should be understood, how therapeutic agent and chaperoning or escorting the therapeu ever, that the invention is not limited to the precise arrange tic agent through the lumen of the gut into the portal blood ments and instrumentalities shown. flow and finally on to the systemic circulation. In certain embodiments, the composition of the invention possesses 0010 FIG. 1 is a schematic representation of a composi many unique and advantageous properties. One of these prop tion of the invention. erties is the ability to insert into intercellular gaps and pass 10011 FIG. 2 is a graph depicting the counts of ''C radio through the mammalian gut into the portal circulation. In labeled phospholipid found in the femoral and portal veins 15 certain embodiments, a composition of the invention may be and 30 minutes post injecting radio-labeled composition into targeted to specific cellular or extra-cellular receptors via one the duodenum of a fasted and anesthetized 230 gram rat. or more targeting agents. As an alternative to incorporation of I0012 FIG.3 is a bargraph depicting the distribution of ''C a targeting agent, or optionally in addition to a targeting radio-labeled phospholipid amongst the blood, liver, and agent, a composition of the invention may further include one spleen in the rats of FIG. 2, post-sacrifice. or more RES masking agents. 0013 FIG. 4 is a graph depicting the absorption of radio 0005. In a typical embodiment, an orally bioavailable labeled composition from drinking water at 15, 30, and 45 composition of the invention comprises various lipid-based minutes post-dosing. constituents, at least one therapeutic or diagnostic agent, an 0014 FIG. 5 is a bar graph depicting the distribution of the optional targeting agent, and/or an optional RES masking labeled composition amongst the blood, liver, and spleen in agent. the rats of FIG. 4, post-sacrifice. 0006. The various lipid-based constituents include, but are 0015 FIG. 6 is a graph depicting the efficacy of orally not limited to, dynamically sized liposomes, dynamically administered insulin in the form of a composition of the sized liposome fragments, and dynamically sized lipid par invention. ticles. A lipid particle comprises at least one, but preferably 0016 FIG. 7 is a bar graph depicting the efficacy of a more than one, molecule of a single lipid. A liposome or composition of the invention (at low dosages), in converting a liposome fragment comprise at least two structurally unique type 2 diabetic dog from hepatic glucose output to uptake lipid molecules. These lipid-based constituents may be during a portal glucose load. formed when lipids are combined according to the procedures (0017 FIG. 8 is a plot of blood calcium levels after the set forth herein. administration of calcitonin associated with a non-targeted 0007. In certain embodiments, the lipids are selected from composition of the invention. the group consisting of MPB-PE, MCC-PE, 1,2-distearoyl (0018 FIG. 9 is a graph of the size distribution of the sn-glycero-3-phosphocholine, 1,2-dipalmitoyl-sn-glycero constituent members of a composition of the invention. US 2010/0080773 A1 Apr. 1, 2010

0019 FIG.10 is a graph of the efficacy of a composition of used herein and the laboratory procedures in organic chem the invention comprising a biotin targeting agent and insulin istry and protein chemistry are those well known and com at reducing the effects of type 2 diabetes in humans. monly employed in the art. 0020 FIG. 11 is a chromatogram of a composition of the 0028. The articles “a” and “an are used herein to refer to invention showing the efficacy of insulin loading. one or to more than one (i.e. to at least one) of the grammatical 0021 FIG. 12 is a graph depicting the efficacy of oral object of the article. By way of example, “an element’ means delivery of IgG antibodies covalently linked to a composition one element or more than one element. of the invention versus oral absorption of non-associated (free) IgG antibodies. 0029. As used herein, amino acids are represented by the 0022 FIG. 13 is a graph depicting the effect of oral admin full name thereof, by the three-letter code as well as the istration of thyroxine associated with a composition of the one-letter code corresponding thereto, as indicated in the invention on serum cholesterol and triglycerides (“TG’) in following table: mice. 0023 FIG. 14 is a graph depicting the effect of oral admin istration of interferon associated with a composition of the 3 Letter 1-Letter invention on reducing viral load in humans Suffering from FullName Code Code hepatitis-C. Alanine Ala A. Arginine Arg R DETAILED DESCRIPTION OF THE INVENTION Asparagine ASn N Aspartic Asp D 0024. The present invention includes compositions that Aci facilitate and/or enable absorption of therapeutics which are Cysteine Cys C Cystine Cys-Cys C-C not typically orally bioavailable. The compounds of the Glutamic Glu E present invention may further act to enhance the oral bioavail Acid ability of compounds that are already orally bioavailable. In Glutamine Gln Q one embodiment, a composition of the invention functions by Glycine Gly G Histidine His H associating with a therapeutic agent and chaperoning the Isoleucine Ile I therapeutic agent through the lumen of the gut into the portal Leucine Leu blood flow and finally on to the systemic circulation. The Lysine Lys K composition of the invention possess many unique and advan Met M Phenylalanine Phe tageous properties. One of these properties is the ability to Proline Pro C insert into intercellular gaps and pass through the mammalian Serine Ser S gut into the portal circulation. In certain embodiments, a Threonine Thr T composition of the invention may be targeted to specific Tryptophan Trp W cellular or extra-cellular receptors via one or more targeting Tyrosine Tyr Y agents. As an alternative to incorporation of a targeting agent, Valine Wall V or optionally in addition to a targeting agent, a composition of the invention may further include one or more reticuloendot 0030. The term “lower', when used in reference to a helial system (“RES) masking agents. chemical structure, describes a group containing from 1 to 6 0025. Although the present invention bears some resem carbon atoms. blance to the composition disclosed in PCT/US06/19119, 0031. The term “alkyl, by itself or as part of another U.S. patent application Ser. No. 1 1/904,937, and PCT/US08/ Substituent means, unless otherwise stated, a straight, 77990, the compositions of the present invention may be branched or cyclic hydrocarbon having the number of carbon differentiated from all three applications. The present inven atoms designated (i.e. C-C means one to six carbons). tion may be differentiated from PCT/U06/19119 by the size Examples include: methyl, ethyl, propyl, isopropyl, butyl, of the composition as well as the use of covalent linkages to isobutyl, tert-butyl, pentyl, neopentyl, hexyl, cyclohexyl and tethera given therapeutic agent. The present invention may be cyclopropylmethyl. Most preferred is (C-C)alkyl, particu differentiated from PCT/US08/77990 and Ser. No. 1 1/904, larly ethyl, methyl and isopropyl. 937 by the chemical structure of the linker used to linka given 0032. The term “alkylene', by itself or as part of another therapeutic agent to the composition. The present invention Substituent means, unless otherwise stated, a straight, may be further differentiated from U.S. patent application branched or cyclic chain hydrocarbon having two Substitution Ser. No. 1 1/904,937 and PCT/US08/77990 by the therapeutic sites, e.g., methylene(-CH2—), ethylene(-CH2CH2—), iso agent associated with the composition. propylene(-C(CH)—CH ), etc. 0026. In a typical embodiment, an orally bioavailable composition of the invention comprises various lipid-based 0033. The term “aryl', employed alone or in combination constituents, at least one therapeutic or diagnostic agent, an with other terms, means, unless otherwise stated, a carbocy optional targeting agent, and/or an optional RES masking clic structure, with or without Saturation, containing one or agent. more rings (typically one, two or three rings) wherein said rings may be attached together in a pendant manner. Such as a biphenyl, or may be fused, such as naphthalene. Examples Definitions include phenyl, anthracyl, and naphthyl. The structure may be 0027. Unless defined otherwise, all technical and scien optionally substituted with one or more substituents, inde tific terms used herein generally have the same meaning as pendently selected from halogen; (C-C)alkyl, (C-C)alk commonly understood by one of ordinary skill in the art to enyl: (C-C)alkoxy; OH: NO; C=N; C(=O)C(C-C) which the invention belongs. Generally, the nomenclature alkyl: (C-C)alkylene-OR: phosphonato; NR; NHC US 2010/0080773 A1 Apr. 1, 2010

(=O)(C-C)alkyl; sulfamyl; carbamyl; OC(=O)(C-C) 0043. The term “insulin' refers to natural or recombinant alkyl, O(C-C)alkylene-N((C-C)alkyl); and (C-C) forms of insulin, synthetic insulin, and derivatives of the perfluoroalkyl. aforementioned . Examples of insulin include, but are 0034. The term “arylloweralkyl means a functional not limited to , , regular insulin, group wherein an aryl group is attached to a lower alkylene , insulin Zinc, human insulin Zinc extended, group, e.g., —CH2CH2-phenyl. isophane insulin, human buffered regular insulin, , recombinant human regular insulin, ultralente insu 0035. The term “alkoxy” employed alone or in combina lin, humulin, NPH insulin, Levemir, Novolog, and recombi tion with other terms means, unless otherwise stated, an alkyl nant human insulin isophane. Also included are animal insu group or an alkyl group containing a Substituent such as a lins, such as bovine or porcine insulin. hydroxyl group, having the designated number of carbon 0044) The terms 'glargine' and 'glargine insulin both atoms connected to the rest of the molecule via an oxygen refer to a recombinant human insulin analog which differs atom, such as, for example, —OCH(OH)— —OCH-OH, from human insulin in that the asparagine at methoxy(-OCH), ethoxy(-OCH2CH), 1-propoxy(- position A21 is replaced by glycine and two arginines are OCHCHCH), 2-propoxy(isopropoxy), butoxy(- added to the C-terminus of the B-chain. Chemically, it is OCH2CH2CHCH), pentoxy(-OCH2CH2CH2CHCH), 21A-Gly-30Ba-L-Arg-30Bb-L-Arg-human insulin and has and the higher homologs and isomers. the empirical formula C27HoN72O7sS and a molecular 0036. The term “acyl means a functional group of the weight of 6063. general formula —C(=O)—R, wherein —R is hydrogen, 0045. The term “recombinant human insulin isophane' alkyl, amino or alkoxy. Examples include acetyl(-C(=O) refers to a human insulin that has been treated with protamine. CH), propionyl(-C(=O)CHCH), benzoyl(-C(=O) 0046. The term “bioavailability” refers to a measurement CH), phenylacetyl(C(=O)CHCH), carboethoxy(- of the rate and extent that a pharmaceutical agent, Such as, but COCHCH), and dimethylcarbamoyl (C(=O)N(CH)). not limited to, insulin, reaches the systemic circulation and is available at its site of action. 0037. The terms “halo” or “halogen” by themselves or as 0047. As used herein, to “treat’ means reducing the fre part of another Substituent mean, unless otherwise stated, a quency with which symptoms of a disease, disorder, or fluorine, chlorine, bromine, or iodine atom. adverse condition, and the like, are experienced by a patient. 0038. The term “heterocycle” or “heterocyclyl or “het 0048. As used herein, the term “pharmaceutically accept erocyclic” by itself or as part of another substituent means, able carrier’ means a chemical composition with which the unless otherwise stated, a saturated or unsaturated, stable, active ingredient may be combined and which, following the mono or multicyclic ring system comprising carbon atoms combination, can be used to administer the active ingredient and at least one heteroatom selected from the group compris to a subject. ing N, O, and S, and wherein the nitrogen and Sulfur heteroa 0049. The term “lipid' or “lipids” means an organic com toms may be optionally oxidized, and the nitrogen atom may pound characterized by its preference for non-polar solvents. be optionally quaternized. Examples include pyridine, pyr A lipid may or may not possess an alkyl tail. Lipids according role, imidazole, benzimidazole, phthalein, pyridenyl, pyra to the present invention include, but are not limited to, the nyl, furanyl, thiazole, thiophene, oxazole, pyrazole, 3-pyrro class of compounds known in the art as phospholipids, cho line, pyrrollidene, pyrimidine, purine, quinoline, lesterols, and dialkyl phosphates. isoquinoline, carbazole, etc. Where substitution will result in 0050. As used herein, “cholesterol’ means the compound a stable compounds, the structure may be optionally Substi and all derivatives and analogs of the compound: tuted with one or more substituents, independently selected from halogen; (C-C)alkyl, (C-C)alkenyl: (C-C)alkoxy: OH, NO; C=N; C(=O)C(C-C)alkyl: (C-C)alkylene OR: phosphonato; NR: NHC(=O)(C-C)alkyl; sul HC famyl; carbamyl; OC(=O)(C-C)alkyl; O(C-C)alkylene CH CH N((C-C)alkyl), and (C-C)perfluoroalkyl. H 0039. The term "amphipathic lipid means a lipid mol CH CH3, ecule having a polar end and a non-polar end. 0040. A “complexing agent' is a compound capable of H forming a waterinsoluble coordination complex with a metal, e.g. a salt of chromium, Zirconium, etc., that is Substantially HO insoluble in water and soluble in organic solvents. 0041 Aqueous media” means media comprising water or wherein said derivatives and analogs include, but are not media comprising water containing at least one buffer or salt. limited to, thiocholesterol: 0042. The terms “associated,” or “associated with as well as variations thereof, when used in reference to a com position of the invention, means that the referenced material, H3C typically a therapeutic agent, is incorporated (or intercalated) CH CH3 into, or on the Surface of, or within a lipid-based constituent H comprising the composition of the present invention. Asso ciation may, however, refer to the situation wherein the ref CH CH3. erenced material, typically a therapeutic agent, is covalently attached to a lipid included in one of the various lipid-based H constituents comprising the composition of the invention. HS The applicability of the appropriate definition will be appre ciable from the context in which the terms is used. US 2010/0080773 A1 Apr. 1, 2010

0051. As used herein, “1,2-dipalmitoyl-sn-glycero-3- 0057 The lipid-based constituents comprising a compo phospho-2-mercaptoethanol' means the compound having sition of the invention include, but are not limited to, dynami the formula: cally sized liposomes, dynamically sized liposome frag ments, and dynamically sized lipid particles. A lipid particle comprises at least one, but preferably more than one, mol ecule of a single lipid. A liposome or liposome fragment comprise at least two structurally unique lipid molecules. 0.058 Traditionally, liposome, liposome fragments, and lipid particles comprised of amphipathic materials have been O limited to a lower size distribution of about 40 nanometers.

14 This limit was believed to be a function of the collective sizes of the constituent lipids (phospholipids, cholesterols, dialky lphosphates, etc.) that constituted the membrane structure. O 0- - - 0059. The lipid-based constituents of a composition of the CS invention, however, demonstrate heretofore unobserved ),- " dynamic sizing and size elasticity. Specifically, these struc tures exist in a dynamic equilibrium in aqueous media Such that, on average, these structures fluctuate in size from about 6 nanometers to about 80 nanometers in diameter, but may reach sizes as large as 200 nanometers. At any given time, as well as salts thereof. anywhere from about 5% to about 50% of the various lipid 0052. As used herein, “particle' comprises an agglomera based constituents exhibit an average diameter of about 20 tion of multiple units of one or more lipids. nanometers or less. Due to the nearly constant fluctuations in 0053 As used herein, “thyroxine' refers to the compound: sizes, the lipid-based constituents cannot be physically sepa rated by traditional fractionating means to form discrete populations. 0060. The composition of the invention may associate with one or more therapeutic agents or diagnostic agents. I When these associations are non-covalent, and without wish O I ing to be bound by any particular theory, it is believed that a NH2 given therapeutic agent is associated with a composition of the invention through various intramolecular forces. It is fur HO I OH ther believed that when a lipid-based constituent comprising the composition of the invention has a diameter of 20 nanom O I eters or less, it is sufficiently small to pass through intracel lular gaps and enable the transport of the associated therapeu tic agent from the lumen of the gut into the portal blood flow. Another mechanism of action may, however, account for the wherein the amino group may be in either the "D' or “L” observed activity. configuration. 0054 As used herein, “co-administration” or “co-admin Lipids istering as well as variations thereof, means administering a 0061 The lipids comprising the composition of the second therapeutic agent before, during, or after the admin present invention are selected from the group consisting of istration of a first therapeutic agent. The first and second 1.2-distearoyl-sn-glycero-3-phosphocholine, 1,2-dipalmi therapeutic agents may be the same or different. toyl-sn-glycero-3-phosphocholine, 1,2-dimyristoyl-sn-glyc 0055 As used herein, “interferon refers to all forms of ero-3-phosphocholine, cholesterol, cholesterol oleate, thio interferon, including, but not limited to, interferon-C. inter cholesterol, dihexadecyl phosphate, 1,2-distearoyl-sn feron-beta, interferon-gamma, as well as Sub-units thereof. glycero-3-phosphate, 1,2-dipalmitoyl-sn-glycero-3- phosphate, 1,2-dimyristoyl-sn-glycero-3-phosphate, 1.2- Description distearoyl-sn-glycero-3-phosphoethanolamine, 1.2- 0056. A composition of the present invention is comprised dipalmitoyl-sn-glycero-3-phosphoethanolamine, 1.2- of various lipid-based constituents, at least one therapeutic or dimyristoyl-sn-glycero-3-phosphoethanolamine, 1.2- diagnostic agent, optionally at least one targeting molecule, dipalmitoyl-sn-glycero-3-phospho-2-mercaptoethanol, 1.2- and optionally, at least one RES masking agent. A composi dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- tion of the present invention may further include gelatin as an (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac-(1- active component. When present, the gelatin actively revers glycerol) (sodium salt), triethylammonium 2,3- ibly interacts with one or more of the various lipid-based diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H constituents to stabilize the composition of the invention. The thieno3,4-dimidazol-4-yl)pentanamido)ethyl phosphate, at least one therapeutic agent and/or diagnostic agent is asso MPB-PE, MCC-PE, and derivatives thereof, including but ciated with a lipid-based constituent comprising the compo not limited to salts. Representative structures are presented in sition of the invention. Table 1. US 2010/0080773 A1 Apr. 1, 2010 5

TABLE 1.

Common Name Chemical Name Structure

1,2-distearoyl 2,3- O Sn-glycero-3- bis(Stearoyloxy)propyl phosphocholine 2-(trimethylammonio) o 1-1-1-1-1-1--1a1a1a1 ethyl phosphate O O OEP-O N1--1a1a-n-n-n-n-n SH O O N-r-, CH3

1,2-dipalmitoyl 2,3- O Sn-glycero-3- bis(palmitoyloxy)propyl phosphocholine 2-(trimethylammonio) ethyl phosphate

1,2-dimyristoyl 2,3-bis Sn-glycero-3- (tetradecanoyloxy) phosphocholine propyl 2 (trimethylammonio) ethyl phosphate

O "ch n1n 1. 3

CH

Cholesterol 10,13-dimethyl-17 HC (6-methylheptan-2-yl)- 2,3,4,7,8,9,10,11,12,13 CH 14,15,16,17 3 CH tetradecahydro-1H H cyclopentaa. CH3 phenanthren-3-ol CH3 H

HO

MPB-PE (Na' 1,2-Dipalmitoyl-sn O salt) glycero-3- O G phosphoethanol amine-N-4-(p- 7 Na °-- O 14 maleimido)phenyl N O O butyrate O O O N 14 US 2010/0080773 A1 Apr. 1, 2010

TABLE 1-continued

Common Name Chemical Name Structure MCC-PE (Na' 1,2-Dipalmitoyl-sn salt) glycero-3- phosphoethanol amine-N-4-(p- Na o-p-g O maleimido- O O -" methyl)cyclohexane carboxamide N

O

0062 By way of non-limiting examples, the lipid-based formed from approximately 68 mole % 1.2 dipalmitoyl-sn constituents comprising the composition of the invention may glycero-3-phosphocholine, approximately 19 mole % be formed from about 40 to about 65 mol % 1.2 distearoyl dihexadecyl phosphate, approximately 10 mole % choles sn-glycero-3-phosphocholine; from about 10 to about 50 mol terol, and approximately 3 mole % MPB-PE, MCC-PE, or % dihexadecyl phosphate; from about 15 to about 35 mol % 1.2-distearoyl-sn-glycero-3-phosphoethanolamine. In cer cholesterol, and optionally up to about 15 mol %, preferably tain variations of this embodiment, at least about 25% of the less than about 5 mol %, and most preferably about 1 mol % cholesterol may be thiocholesterol. In a further variation, at of a targeting agent. The amount of targeting agent necessary least about 50% of the cholesterol may be thiocholesterol. In to target a given composition will be dictated by the size and another variation, at least about 75% of the cholesterol may be structure of the therapeutic agent. It is within the skill level of thiocholesterol. In a further variation, all of the cholesterol the orididnary skill artisan, based on the disclosure herein, to may be thiocholesterol. select and prepare the composition of the invention contain 0068. When any of the cholesterol in any variation of this ing the appropriate amount of targeting agent. embodiment is thiocholesterol, and either MPB-PE or MCC 0063. In a preferred embodiment, the lipid-based constitu PE is present, MPB-PE or MCC-PE will have been reacted ents comprising the composition of the invention are formed with an appropriate nucleophile prior to being exposed to from approximately 62 mole percent 1.2 distearoyl-sn-glyc thiocholesterol. ero-3-phosphocholine, approximately 22 mole percent 0069. In each of the above described embodiments, up to dihexadecyl phosphate, and approximately 16 mole percent about 10% of the 1.2 dipalmitoyl-sn-glycero-3-phosphocho cholesterol. In certain variations of this embodiment, at least line may be replaced with 1,2-distearoyl-sn-glycero-3-phos about 25% of the cholesterol may be thiocholesterol. In a phoethanolamine or 1,2-dipalmitoyl-sn-glycero-3-phospho further variation, at least about 50% of the cholesterol may be 2-mercaptoethanol. thiocholesterol. In yet another variation, at least about 75% of 0070. Each of the above described embodiments further the cholesterol may be thiocholesterol. In a further variation, includes at least one associated therapeutic agent or diagnos all of the cholesterol may be thiocholesterol. tic agent. In certain embodiments, the therapeutic agent may 0064. In another embodiment, the lipid-based constituents be non-covalently associated with the composition. In alter comprising the composition of the invention are formed from native embodiments, the associated therapeutic agent may be approximately 61 mole percent 1.2 distearoyl-sn-glycero-3- covalently linked to a lipid incorporated into a lipid-based phosphocholine, approximately 22 mole percent dihexadecyl constituent comprising the composition of the invention. The phosphate, approximately 16 mole percent cholesterol, and process of covalently linking a therapeutic agent to a lipid is about 1 mole percent of at least one targeting agent. Up to an described elsewhere herein. additional 1 mole percent of targeting agent may be added to 0071. When therapeutic agents are attached via covalent this embodiment. linkages, it is preferred that therapeutic agents are linked to no 0065. In a variation of this embodiment, at least about 25% more than about 10 mole % of the lipids comprising the of the cholesterol may be thiocholesterol. In a further varia composition of the invention. Even more preferably, thera tion, at least about 50% of the cholesterol may be thiocholes peutic agents are linked to no more than about 5 mole% of the terol. In another variation, at least about 75% of the choles lipids comprising the composition of the invention. Most terol may be thiocholesterol. In a further variation, all of the preferably, therapeutic agents are linked to no more than cholesterol may be thiocholesterol. about 2 mole % of the lipids comprising the composition of 0066. The lipid-based constituents comprising the compo the invention. Although the above described quantities are sition of the invention may also be formed from 40 to 75 mole preferred, a person of ordinary skill in the art will be able to %. 1.2 dipalmitoyl-sn-glycero-3-phosphocholine; from 5 to attenuate or titrate the amount of therapeutic agent present in 50 mole % dihexadecyl phosphate; from 5 to 15 mole % or on a given composition in order to affect the amount of cholesterol; from 1 to 6 mole % MPB-PE, MCC-PE, or 1,2- therapeutic agent delivered to a patient in need thereof. distearoyl-sn-glycero-3-phosphoethanolamine; and, option 0072 Any of the above described embodiments may fur ally, up to about 2 mole %, but preferably not more than 1 ther optionally include one or more RES masking agents. mole percent of a targeting agent. Typically, the one or more RES masking agents are covalently 0067. In a specific embodiment, the lipid-based constitu attached, either directly or indirectly, to one or more of the ents comprising the composition of the invention may be lipids comprising the composition of the invention as is US 2010/0080773 A1 Apr. 1, 2010

described elsewhere herein. They may, however, be non-co from about 6 to about 200 nanometers, with the optimal size Valently associated with a composition of the invention. range being about 6 to about 80 nanometers, and the average 0073. When covalently attached, RES masking agents are size in this range being about 50 to about 60 nanometers. A linked to no more than about 10 mole % of the lipids com significant percentage of the lipid-based constituents, are prising the composition of the invention. Even more prefer approximately 20 nanometers. Average sizing is measured by ably, RES masking agents are linked to no more than about 5 a Coulter N-4 Plus Sub-Micron Particle Size Analyzer. After mole 96 of the lipids comprising the composition of the inven microfluidization, the resulting constituents may be sterile tion. Most preferably, RES masking agents are linked to no filtered through a 0.8 micron to 0.2 micron gang SuportM more than about 2 mole % of the lipids comprising the com membrane at 50 to 70° C., preferably at about 60° C. FIG.9 position of the invention. represents repeated size analyses on the same sample as it remained stationary in the Coulter N-4 Plus Sub Micron 0074. When one or more RES masking agents are associ Particle Size Analyzer. This data demonstrates the dynamic ated with a composition of the invention non-covalently, any sizing and fluid nature of the lipid-based constituents formed of the above described embodiments may include up to about from the lipids comprising the invention. 10 mole % or greater of one or more RES masking agents. I0081. During the process of sub-micron particle forma 0075 Although it is preferred that a composition of the tion, hydrogen bonding, ionic bonding, van der Waal's inter invention contain about 18 mole % up to about 22 mole % actions, dipolar interactions, ion-dipole interactions, hydro dihexadecyl phosphate, a composition of the invention may phobic associations, and thermodynamic considerations contain up to 30 mole %, even up to 40 mole %, and even as dictate the manner in which the lipids assemble. While not much as 50 mole % dihexadecyl phosphate, inclusive of any wishing to be bound by any one particular theory, it is incremental amounts of dihexadecyl phosphate therein. This believed that the interaction of all of these forces, to varying increase in the amount of dihexadecyl phosphate requires a extents, under the conditions noted above, lead to a dynami concomitant reduction in the quantity of one or more other cally sized composition of the present invention. lipids in the composition by a total amount equivalent to the quantity of dihexadecylphosphate added in excess of 18 or 22 Incorporation of a Targeting Agent mole 96. I0082 In certain embodiments, a composition of the present invention may optionally comprise a targeting agent. Preparation Targeting agents alter the composition's bio-distribution and 0076 Generally, the composition of the present invention further enhances the efficacy of an associated therapeutic is formed when appropriate lipids and other ingredients (such agent. A composition of the present invention may incorpo as a targeting molecule) are homogenized in an aqueous rate one or more targeting agents that act to target the com media via microfluidization or other process involving cavi position, and associated therapeutic, to a specific cellular or tation. extracellular receptor. For example, a targeting agent may be 0077. In an embodiment of the invention, the lipids and used to target insulin associated with a composition of the other ingredients may be homogenized in 18 mM phosphate invention to hepatocytes in order to control post-prandial buffer at a pH of about 6.0 to a pH of about 8.0. Lipid glycogen storage. concentration in the phosphate buffer may range from about I0083. In one embodiment, a targeting agent facilitates 10 to about 200 mg/ml and any and all whole and partial delivery of a therapeutic agent to the liver and encompasses a integers therebetween. In one embodiment, the lipid concen class of molecules referred to as “hepatocyte target molecule' tration is about 30 to about 150 mg/ml. In more preferred (HTM). HTMexamples include, but are not limited to, biotin embodiment, the lipid concentration is about 15 to about 50 DHPE, biotin-X-DHPE, and metal derived targeting agents mg/ml. In a most preferred embodiment, the lipid concentra such as poly Cr-bis(N-2,6-diisopropylphenylcarbamoylm tion is about 28-30 mg/ml. ethyl iminodiacetic acid). Metal-derived targeting agents 0078 Homogenization of the aqueous media, lipids and and biotin derived targeting agents are discussed below and other ingredients may be accomplished via treatment in a arefully described in U.S. Pat. Nos. 7,169.410 and 4,603,044: device suitable for homogenization. Examples of suitable PCT application PCT/US06/19119; and U.S. patent applica devices include, but are not limited to, a Polytron(R) System tion Ser. Nos. 11/384,728, and 11/384,659. Additional PT 6100, an M-1 10-EH microfluidizer, an ultrasonic sonica examples of biotin-derived targeting agents are disclosed in tor, a high pressure membrane filtration apparatus, and a Table 2. homogenizer extruder. I0084. When the targeting agent comprises biotin, imino biotin, carboxybiotin, biocytin, or iminobiocytin, the biotin, 0079. In instances where a microfluidizer is used, the iminobiotin, carboxybiotin, biocytin, or iminobiocytin mol microfluidizer is preferably operated at a temperature that is ecules may be bound via an amide bond to the nitrogen of a greater than the highest transition temperature of the various phospholipid molecule Such as 1,2-dipalmitoyl-sn-glycero lipids and most preferably at a temperature greater than about 3-phosphoethanolamine. The compounds may likewise be 75° C. The elevated temperature allows any acyl and alkyl bound to a molecule Such as cholesterol through an ester chains present in the lipids to move fluidly as well as conform linkage. In the case of biocytin and iminobiocytin, the com to and associate with neighboring hydrocarbon moieties. pounds may be bound to benzoyl thioacetyl triglycine via an These non-covalent associations directly result in the forma amide bond between the terminal nitrogen of iminiobiocytin tion of a constituent of a composition of the present invention. and the terminal carbonyl of benzoyl thioacetyl triglycine. 0080 For the microfluidization process, up to about five Alternative bond connectivities to those described above are independent passes are required at 9000 psig in order to possible and considered to be within the scope of the present prepare compositions having lipid-based constituents sized invention. US 2010/0080773 A1 Apr. 1, 2010

ZHT8IVIL

I US 2010/0080773 A1 Apr. 1, 2010

OOOH

US 2010/0080773 A1 Apr. 1, 2010 10

US 2010/0080773 A1 Apr. 1, 2010 11

OOOH

US 2010/0080773 A1 Apr. 1, 2010 12

~

O

HO–O O O O

LI 8] US 2010/0080773 A1 Apr. 1, 2010 13

US 2010/0080773 A1 Apr. 1, 2010 14

US 2010/0080773 A1 Apr. 1, 2010 15

US 2010/0080773 A1 Apr. 1, 2010 16

O

CON US 2010/0080773 A1 Apr. 1, 2010 17

OOOH

US 2010/0080773 A1 Apr. 1, 2010 18

O || O

US 2010/0080773 A1 Apr. 1, 2010 19

*HO–O–NH HO O–OBN HOVHO HO US 2010/0080773 A1 Apr. 1, 2010 20

·1IIH

HO O OO *HO–O–NH

HOVHO O HO

HO O?EO HO |HO HOVHO OHO HO SOOHO HOVHO HO OHO

US 2010/0080773 A1 Apr. 1, 2010 21

HOVHO

O HO HOVHO HOHO ()H HO OO O HO HOVHO O*HO HO HOOHO HO HO US 2010/0080773 A1 Apr. 1, 2010 22

I0085 Names of Compounds 48-50: the transition metals. The transition and inner transition met 0086) 48. ((2R,5S)-3-acetamido-5-hydroxy-6-(hy als from which the metal is selected include: Sc (scandium), droxymethyl)-2,3,4,6-tetramethyl-4-((((2S.5R)-3,4,5-trihy Y (yttrium), La (lanthanum), Ac (actinium), the actinide droxy-6-(hydroxymethyl)-2,3,4,5,6-pentamethyltetrahydro series; Ti (titanium), Zr (zirconium), Hf (hafnium), V (vana 2H-pyran-2-yl)methoxy)methyl)tetrahydro-2H-pyran-2-yl) dium), Nb (niobium), Ta (tantalum), Cr (chromium), Mo methyl 5-((3aS,6aR)-2-oxohexahydro-1H-thieno 3,4-d (molybdenum), W (tungsten), Mn (manganese), Tc (techne imidazol-4-yl)pentanoate((2R,5S)-3-acetamido-5-hydroxy tium), Re (rhenium), Fe (iron), Co (cobalt), Ni (nickel), Ru 6-(hydroxymethyl)-2,3,4,6-tetramethyl-4-((((2S,5R)-3,4,5- (ruthenium), Rh(rhodium), Pd (palladium), Os (osmium), Ir trihydroxy-6-(hydroxymethyl)-2,3,4,5,6- (iridium), and Pt (platinum). The neighbors of the transition pentamethyltetrahydro-2H-pyran-2-yl)methoxy)methyl) metals from which the metal may be selected are: Cu (cop tetrahydro-2H-pyran-2-yl)methyl 5-((3aS,6aR)-2- per), Ag (silver), Au (gold), Zn (Zinc), Cd (cadmium), Hg oxohexahydro-1H-thieno3,4-dimidazol-4-yl)pentanoate. (mercury), Al (aluminum), Ga (gallium). In (indium), Tl I0087 49. (2R,3R,5S)-5-((((2S,3S,5S)-3-acetamido-5-hy (thallium), Ge (germanium), Sn (tin), Pb (lead), Sb (anti droxy-6-(hydroxymethyl)-2,4,6-trimethyl-4-((((2S,5R)-3,4, mony) and Bi (bismuth), and Po (polonium). Preferably, the 5-trihydroxy-6-(hydroxymethyl)-2,3,4,5,6-pentamethyltet metal is chromium. rahydro-2H-pyran-2-yl)methoxy)methyl)tetrahydro-2H 0091. Non-limiting examples of useful salts include chro pyran-2-yl)methoxy)methyl)-3,4-dihydroxy-2,4,5,6,6- mium chloride (III) hexahydrate; chromium (III) fluoride tet pentamethyltetrahydro-2H-pyran-2-yl 5-((3aS,6aR)-2- rahydrate; chromium (III) bromide hexahydrate; zirconium oxohexahydro-1H-thieno3,4-dimidazXol-4-yl)pentanoate (IV) citrate ammonium complex; zirconium (IV) chloride: I0088 50. (2S,5S)-3-acetamido-4-((((2R,5S)-5-((((2R, zirconium (IV) fluoride hydrate; zirconium (IV) iodide: 5S)-4,5-dihydroxy-6-(hydroxymethyl)-2,3,4,5,6-pentam molybdenum (III) bromide; molybdenum (III) chloride; ethyl-3-((((2S,5S)-3,4,5-trihydroxy-2,3,4,5,6,6-hexameth molybdenum (IV) sulfide; iron (III) hydrate; iron (III) phos yltetrahydro-2H-pyran-2-yl)methoxy)methyl)tetrahydro phate tetrahydrate, iron (III) sulfate pentahydrate, and the 2H-pyran-2-yl)methoxy)methyl)-3,4-dihydroxy-2, 3,4,5,6, like. 6-hexamethyltetrahydro-2H-pyran-2-yl)methoxy)methyl)- 0092. In addition to a metal, the metal derived targeting 5-hydroxy-6-(hydroxymethyl)-2,3,4,5,6- agent comprises one or more complexing agents. A complex pentamethyltetrahydro-2H-pyran-2-yl 5-((3aS,6aR)-2- ing agent is a compound capable of forming a water insoluble oxohexahydro-1H-thieno3,4-dimidazol-4-yl)pentanoate coordination complex with the preferred metal. There are I0089 Structures of iminobiotin compounds are not shown several families of Suitable complexing agents. in Table 2. However, the iminobiotin structures are analogs of 0093. A complexing agent may be selected from the fam the biotin structure where the biotin group is replaced by an ily of iminodiacetic acids of formula (1) wherein R is low iminobiotin group. An example is shown below. eralkyl, aryl, arylloweralkyl, or a heterocyclic Substituent.

(1) O O

HO-C-CH-N-CH-C-OH O Loweralkylene C-N-R N-O O H

O O N-hydroxysuccinimide biotin Suitable compounds of formula (1) include: 0094 N-(2,6-diisopropylphenylcarbamoylmethyl)imino diacetic acid; 0.095 N-(2,6-diethylphenylcarbamoylmethyl)iminodi O acetic acid; 0096 N-(2,6-dimethylphenylcarbamoylmethyl)iminodi acetic acid; N-O 0097 N-(4-isopropylphenylcarbamoylmethyl)iminodi acetic acid; O O 0.098 N-(4-butylphenylcarbamoylmethyl)iminodiacetic acid; N-hydroxysuccinimide iminobiotin 0099 N-(2,3-dimethylphenylcarbamoylmethyl)iminodi acetic acid; 0090. In an embodiment of the invention, metal derived 0100 N-(2,4-dimethylphenylcarbamoylmethyl)iminodi targeting agents may be polymeric or monomeric. Polymeric acetic acid; metal derived targeting agents are fully described in U.S. Pat. 0101 N-(2,5-dimethylphenylcarbamoylmethyl)iminodi No. 7,169.410. Monomeric metal derived targeting agents are acetic acid; described in U.S. Pat. No. 4,603,044. Whether polymeric or 0102 N-(3,4-dimethylphenylcarbamoylmethyl)iminodi monomeric, the compounds generally comprise a metal (typi acetic acid; cally purchased as an inorganic salt) that may be selected 0103 N-(3,5-dimethylphenylcarbamoylmethyl)iminodi from the transition and inner transition metals or neighbors of acetic acid; US 2010/0080773 A1 Apr. 1, 2010

0104 N-(3-butylphenylcarbamoylmethyl)iminodiacetic ylmethyl)iminodiacetic acid (NMIDA); iminodicarboxym acid; ethyl-2-naphthylketone phthalein complexone; 3 (3: 7a: 12a: 0105 N-(2-butylphenylcarbamoylmethyl)iminodiacetic trihydroxy-24-norcholanyl-23-iminodiacetic acid; benzimi acid; dazole methyliminodiacetic acid; and N-(5.pregnene-3-p-ol 0106 N-(4-tertiary butylphenylcarbamoylmethyl)imino 2-oyl carbamoylmethyl)iminodiacetic acid. diacetic acid; 0119 The complexing agent may also be selected from the 0107 N-(3-butoxyphenylcarbamoylmethyl)iminodiace family of amino acids of formula (4), tic acid; 0108 N-(2-hexyloxyphenylcarbamoylmethyl)iminodi acetic acid; (4) 0109 N-(4-hexyloxyphenylcarbamoylmethyl)iminodi acetic acid; 0110 Aminopyrrol iminodiacetic acid; 0111 N-(3-bromo-2,4,6-trimethylphenylcarbamoylm ethyl)iminodiacetic acid; R 0112 Benzimidazole methyl iminodiacetic acid; 0113 N-(3-cyano-4,5-dimethyl-2-pyrrylcarbamoylm where R, is an amino acid side chain; wherein Rs may be ethyl)iminodiacetic acid; loweralkyl, aryl, and arylloweralkyl; and wherein R is pyri 0114 N-(3-cyano-4-methyl-5-benzyl-2-pyrrylcarbam doxylidene. oylmethyl)iminodiacetic acid; and I0120 Suitable amino acids of the formula (4) are aliphatic 0115 N-(3-cyano-4-methyl-2-pyrrylcarbamoylmethyl) amino acids, including, but not limited to: glycine, alanine, iminodiacetic acid and other derivatives of N-(3-cyano-4- Valine, leucine, isoleucine; hydroxyamino acids, including methyl-2-pyrrylcarbamoylmethyl)iminodiacetic acid of serine, and threonine; dicarboxylic amino acids and their amides, including aspartic acid, asparagine, glutamic acid, formula (2), glutamine; amino acids having basic functions, including lysine, hydroxylysine, histidine, arginine; aromatic amino (2) acids, including phenylalanine, tyrosine, tryptophan, thyroX CH CN ine; and Sulfur-containing amino acids, including cystine and methionine. / \ CHCOOH I0121 The complexing agent may also be selected from amino acid derivatives including, but not limited to (3-ala R N N-C-CH-N nine-y-amino) butyric acid, O-diazoacetylserine (aZaserine), R CHCOOH homoserine, ornithine, citrulline, penicillamine and members of the pyridoxylidene class of compounds. Pyridoxylidene compounds include, but are not limited to: pyridoxylidene 0116 wherein R and R are the following: glutamate; pyridoxylidene isoleucine; pyridoxylidene phe nylalanine; pyridoxylidene tryptophan; pyridoxylidene-5- methyl tryptophan; pyridoxylidene-5-hydroxytryptamine; and pyridoxylidene-5-butyltryptamine. R2 R 0.122 The complexing agent may likewise be selected H iso-CHg from the family of diamines of formula (6): H CHCH-SCH, H CHCH-p-OH CH CH CH iso-CHg (6) CH CHCH-SCH, RCOOR-10 CH C6Hs CH CH2C6H5 R- localsley CH CH2C6H4-p-OCH RCOOR10

0117. Alternatively, the complexing agent may be selected wherein Rio is hydrogen, loweralkyl, or aryl; R is lower from the family of imino diacid derivatives of formula (3), alkylene or arylloweralky; R and R are independently wherein R. Rs, and R are independently selected at each selected at each occurrence and may behydrogen, loweralkyl, occurrence and may be hydrogen, loweralkyl, aryl, aryllow alkyl, aryl, arylloweralkyl, acylheterocyclic, toluene, Sulfo eralkyl, alkoxyloweralkyl, and heterocyclic. nyl or tosylate. I0123 Examples of suitable diamines of formula (6) include, but are not limited to, ethylenediamine-N,N diacetic (3) acid; ethylenediamine-N,N-bis(-2-hydroxy-5-bromophenyl) acetate; N'-acetylethylenediamine-N,N-diacetic acid; N'-ben R4 -O-C-loweralkylene-N-loweralkylene-C -O-R6 Zoyl ethylenediamine-N,N diacetic acid; N'-(p-toluenesulfo nyl)ethylenediamine-N,N diacetic acid; N'-(p-t- Rs butylbenzoyl)ethylenediamine-N,N diacetic acid; N'- (benzenesulfonyl)ethylenediamine-N,N diacetic acid; N'-(p- 0118 Suitable compounds of formula (3) include: N'-(2- chlorobenzenesulfonyl)ethylenediamine-N,N diacetic acid; acetylnaphthyl)iminodiacetic acid (NAIDA); N'-(2-naphth N'-(p-ethylbenzenesulfonyl ethylenediamine-N,N diacetic US 2010/0080773 A1 Apr. 1, 2010 24 acid; N'-acyl and N'-sulfonyl ethylenediamine-N,N diacetic 0.137 A1PDX Hedral Therapeutics acid; N'-(p-n-propylbenzenesulfonyl)ethylenediamine-N,N 0.138 A6—Angstrom diacetic acid; N'-(naphthalene-2-sulfonyl)ethylenediamine 0139 aaAT-III Genzyme N.N diacetic acid; and N'-(2,5-dimethylbenzenesulfonyl)eth 0140. Abciximab Centocor ylenediamine-N,N diacetic acid. 0.141 ABI.001—Atlantic BioPharmaceuticals 0124 Other, non-limiting examples of complexing com 0.142 ABT-828. Abbott pounds or agents include penicillamine; p-mercaptoisobu 0.143 Accutin tyric acid; dihydrothioctic acid; 6-mercaptopurine, kethoxal 0.144 Actinohivin bis(thiosemicarbazone); Hepatobiliary Amine Complexes, 0145 activin Biotech Australia, Human Therapeu 1-hydrazinophthalazine(hydralazine); Sulfonylurea; Hepato tics, Curis biliary Amino Acid Schiff Base Complexes; pyridoxylidene 0146 AD 439 Tanox glutamate; pyridoxylidene isoleucine; pyridoxylidene phe 0147 AD 519 Tanox nylalanine; pyridoxylidene tryptophan; pyridoxylidene 0.148 Adalimumab Cambridge Antibody Tech. 5-methyl tryptophan; pyridoxylidene-5-hydroxytryptamine; 0.149 Adenocarcinoma vaccine—Biomira NIS pyridoxylidene-5-butyltryptamine; tetracycline; 7-carboxy 0.150 Adenosine deanimase Enzond p-hydroxyquinoline; phenolphthalein; eosin I bluish; eosin I 0151. Adenosine A2B receptor antagonists—Adenos yellowish; verograffin: 3-hydroxyl-4-formyl-pyridene ine Therapeutics glutamic acid; AZO Substituted iminodiacetic acid; hepatobil 0152 ADP-001 Axis Genetics iary dye complexes, such as rose bengal; congo red; bromo 0153 AF 13948 Affymax sulfophthalein; bromophenol blue; toluidine blue; and 0154 Afelimomab Knoll indocyanine green; hepatobiliary contrast agents, such as 0.155 AFP-SCAN Immunomedics iodipamide; and ioglycamic acid; bile salts, such as bilirubin; 0156 AG 2195 Corixa cholgycyliodohistamine; and thyroxine; hepatobiliary thio 0157 agallsidase alfa Transkaryotic Therapies complexes, such as penicillamine; p-mercaptoisobutyric 0158 agallsidase beta Genzyme acid; dihydrothiocytic acid; 6-mercaptopurine; and kethoxal 0159 AGENT Antisoma bis(thiosemicarbazone); hepatobiliary amine complexes, 0.160 AI 300 AutoImmune Such as 1-hydrazinophthalazine(hydralazine); and Sulfonyl 0.161 AI-101 Teva urea; hepatobiliary amino acid Schiff Base complexes, (0162 AI-102 Teva including pyridoxylidene-5-hydroxytryptamine; and pyri 0.163 AI-201 AutoImmune doxylidene-5-butyltryptamine; hepatobiliary protein com 0.164 AI-301 Autoimmune plexes. Such as protamine; ferritin; and asialo-orosomucoid: 0.165 AIDS vaccine ANRS, CIBG, Hesed Biomed, and asialo complexes, such as lactosaminated albumin; Hollis-Eden, Rome, United Biomedical, American immunoglobulins, G, IgG, and hemoglobin. Home Products, Maxygen 0166 airway receptor ligand IC Innovations Non-Covalent Association of Therapeutic and Diagnostic (0167 AJVW 2–Ajinomoto Agents (0168 AK 30 NGF Alkermes 0.125. As noted previously, in certain embodiments, one or 0.169 Albuferon Human Genome Sciences more therapeutic agents may be associated with the compo 0170 albumin Biogen, DSM Anti-Infectives, Gen sition of the present invention. Examples of therapeutic Zyme Transgenics, PPL Therapeutics, agents include, but are not limited to, insulin, interferon, (0171 TranXenoGen. Welfide Corp. rituximab, trastuzumab, uricase, tissue plasminogen activa 0172 aldesleukin Chiron tor, thymoglobin, various , heparin, heparin analogs, 0173 alefacept Biogen antithrombin III, filgrastin, pramilitide acetate, exanatide, 0.174 Alemtuzumab epifibatide, antivenins, IgG, IgM, blood clotting Factors VII, 0.175 Allergy therapy—ALK-Abello/Maxygen, ALK VIII, IX, Kallikrein, Kininogen, Hageman Factor (XII), Abello/RP Scherer plasma thromboplastin antecedent Factor (XI), tissue factor, 0176 allergy vaccines—Allergy Therapeutics Stuart Factor (X), accelerin (V), prothrombin (II), and fibrin 0.177 Alnidofibatide Aventis Pasteur stabilizing Factor (XIII); HGH, GLP-1, erythropoietin, par 0.178 Alnorine SRC VB VECTO athyroid hormone, serotonin, D- or L-thyroxine, calcitonin, 0.179 ALP 242 Gruenentha monoclonal antibodies, as well as other therapeutic agents 0180 Alpha antitrypsin Arriva/Hyland Immuno/ that may include, but are not limited to: ProMetic/ Sciences 0.126 12AP1/E5 Viventia Biotech 0181 Alpha-1 antitrypsin Cutter, Bayer. PPL Thera 0127 1964 Aventis peutics, Profile, ZymoGenetics, Arriva I0128 20K growth hormone AMUR 0182 Alpha-1 protease inhibitor Genzyme Transgen 0129. 28P6/E6 Viventia Biotech ics, Welfide Corp. 0.130 3-Hydroxyphthaloyl-beta-lactoglobulin 0183 Alpha-galactose fusion protein Immunomed I0131 4-IBB ligand gene therapy 1CS (0132) 64-Cu MAb conjugate TETA-1A3 Mallinck 0.184 Alpha-galactosidase A Research Corporation rodt Institute of Radiology Technologies, Genzyme I0133) 64-Cu MAb conjugate TETA-cT84.66 0185 Alpha-glucosidase Genzyme, Novazyme 0.134 64-Cu Trastuzumab TETA conjugate Genen 0186 Alpha-lactalbumin tech 0187 Alpha-L-iduronidase Transkaryotic Therapies, I0135 A 200 Amgen BioMarin 013.6 A10255 Eli Lilly 0188 alteplase Genentech US 2010/0080773 A1 Apr. 1, 2010

(0189 alvircept sudotox NIH 0242 anticancer matrix Telios Integra 0.190 ALX1-11-sNPS Pharmaceuticals 0243 Anticancer monoclonal antibodies—ARIUS, 0191 Alzheimer's disease gene therapy ImmuneX (0192 AM-133 AMRAD 0244 anticancer peptides—Maxygen, Micrologix 0193 Amb a 1 immunostim conj. Dynavax 0245 Anticancer prodrug Tech. Alexion Antibody (0194 AMD 3100 AnorMED NIS Technologies (0195 AMD 3465. AnorMED NIS 0246 Anticancer Troy-Bodies—Affite Affitech (0196. AMD 3465. AnorMED NIS 0247 anticancer vaccine NIH 0.197 AMD Fab Genentech 0248 anticancers—Epimmune 0198 Amediplase Menarini, Novartis 0249 Anti-CCR5/CXCR4 sheep MAb KS Biomedix (0199 AMD Fab Genentech Holdings 0200 Amediplase Menarini, Novartis (0250 Anti-CD11a MAb KBA 0201 AM-F9 0251 Anti-CD11a MAb M17 0202 Amoebiasis vaccine 0252) Anti-CD11a MAb TA-3 0203 Octagene 0253 Anti-CD11a MAb WT.1 0204 anakinra—Amgen 0254 Anti-CD11b MAb Pharmacia 0205 analgesic Nobex 0255 Anti-CD11b MAb LM2 0206 Amgen 0256 Anti-CD154MAb Biogen 0207 AnergiX.RA Corixa, Organon 0257 Anti-CD16-anti-CD30 MAb Biotest 0208 Angiocidin. In Kine 0258 Anti-CD18 MAb Pharmacia Anti-CD19 MAb 0209 angiogenesis inhibitors—ILEX B43 0210 AngioMab—Antisoma 0259 Anti-CD19 MAb-liposomal sodium butyrate 0211 Angiopoietins Regeneron/Procter & Gamble conjugate 0212 angiostatin EntreMed 0260 Anti-CD147 0213 Angiostatin/endostatin gene therapy—Genetix 0261 Anti-CD19 MAb-Saporin conjugate Pharmaceuticals 0262 Anti-CD 19-dsFv-PE38-immunotoxin 0214) angiotensin-II, topical Maret 0263 Anti-CD2 MAb 12-15 0215. Anthrax EluSys Therapeutics/US Army Medi 0264 Anti-CD2 MAb B-E2 Diaclone cal Research Institute 0265 Anti-CD2 MAb OX34 0216 Anthrax vaccine 0266 Anti-CD2 MAb OX54 0217 Anti platelet-derived growth factor D human 0267 Anti-CD2 MAb OX55 monoclonal antibodies—CuraGen 0268 Anti-CD2 MAb RM2-1 0218 Anti-17-1A MAb 3622W94 GlaxoSmithKline 0269. Anti-CD2 MAb RM2-2 0219) Anti-2C4MAb—Genentech 0270 Anti-CD2 MAb RM2-4 0220 anti-4-1BB monoclonal antibodies—Bristol 0271 Anti-CD20 MAb BCA B20 Myers Squibb (0272 Anti-CD20-anti-Fc alpha RI bispecific MAb 0221) Anti-Adhesion Platform Tech. Cytovax Medarex, Tenovus 0222 Anti-adipocyte MAb Cambridge Antibody (0273 Anti-CD22 MAb-Saporin-6 comple Tech/ObeSys 0274 Anti-CD3 immunotoxi 0223) antiallergics—Maxygen (0275 Anti-CD3 MAb 145-2C11-Pharming 0224 antiallergy vaccine—Acambis 0276 Anti-CD3 MAb CD4IgG conjugate Genentech 0225 Anti-alpha-4-integrin MAb (0277 Anti-CD3 MAb humanised Protein Design, 0226 Anti-alphav3 integrin MAb—Applied Molecu RW Johnson lar Evolution 0278 Anti-CD3 MAb WT32 0227 Anti-angiogenesis monoclonal antibodies—KS 0279 Anti-CD3 MAb-ricin-chain-A conjugate Biomedix/Schering AG 0280 Anti-CD3 MAb-xanthirie-oxidase conjugate 0228 Anti-B4MAb-DC1 conjugate ImmunoGen 0281 Anti-CD30 MAb BerH2 Medac 0229 Anti-B7 antibody PRIMATIZED IDEC 0282 Anti-CD30 MAb-Saporin conjugate 0230 Anti-B7-1 MAb 16-10A1 0283 Anti-CD30-scFv-ETA'-immunotoxin 0231 Anti-B7-1 MAb 1 G10 0284 Anti-CD38 MAb AT13/5 0232 Anti-B7-2 MAb GL-1 0285 Anti-CD38 MAb-Saporin conjugate 0233 Anti-B7-2-gelonin immunotoxin 0286 Anti-CD3-anti-CD19 bispecific MAb 0234 Antibacterials/antifungals—Diversa/IntraBiot 0287 Anti-CD3-anti-EGFR MAb ics 0288 Anti-CD3-anti--2-receptor MAb 0235 Anti-beta-amyloid monoclonal antibodies— 0289 Anti-CD3-anti-MOv18 MAb Centocor Cambridge Antibody Tech., Wyeth-Ayerst 0290 Anti-CD3-anti-SCLC bispecific MAb 0236 Anti-BLyS antibodies—Cambridge Antibody 0291 Anti-CD4 idiotype vaccine Tech./Human Genome Sciences 0292 Anti-CD4 MAb Centocor, IDEC Pharmaceuti 0237) Antibody-drug conjugates—Seattle Genetics/ cals, Xenova Group Eos 0293 Anti-CD4. MAb 16H5 0238 Anti-C5 MAb BB5-1- Alexion 0294 Anti-CD4 MAb 4162W94 GlaxoSmithKline 0239) Anti-C5 MAb N19-8 Alexion 0295) Anti-CD4 MAb B-F5 Diaclone 0240 Anti-C8 MAb 0296 Anti-CD4. MAb GK1-5 0241 anticancer —BioPulse 0297 Anti-CD4. MAb KT6 US 2010/0080773 A1 Apr. 1, 2010 26

0298 Anti-CD4. MAb OX38 0353 Anti-GM2 MAb Kyowa 0299 Anti-CD4. MAb PAP conjugate Bristol-Myers 0354 Anti-GM-CSF receptor monoclonal antibod Squibb ies AMRAD 0300 Anti-CD4. MAb RIB 5-2 0355 Anti-gp130 MAb Tosoh 0301 Anti-CD4. MAb W3/25 0356 Anti-HCA monoclonal antibodies—AltaRex/ 0302 Anti-CD4. MAb YTA3.1.2 0303 Anti-CD4. MAb YTS 177-9 0357 Anti-hCG antibodies Abgenix/AVI BioPharma (0304 Anti-CD40 ligand MAb 5c8 Biogen 0358 Anti-heparanase human monoclonal antibod 0305 Anti-CD40 MAb ies—Oxford Glycosciences/Medarex 0306 Anti-CD40. MAb 5D 12 Tanox 0359 Anti-hepatitis C virus human monoclonal anti 0307 Anti-CD44 MAb A3D8 bodies—XTL Biopharmaceuticals 0308 Anti-CD44 MAb GKWA3 0360 Anti-HER-2 antibody gene therapy 0309 Anti-CD44 MAb IM7 0361 Anti-herpes antibody—Epicyte 0310 Anti-CD44 MAb KM81 0362 Anti-HIV antibody Epicyte 0311 Anti-CD44 variant monoclonal antibodies— 0363 anti-HIV catalytic antibody Hesed Biomed Corixa/Hebrew University 0364 anti-HIV fusion protein Idun 0312 Anti-CD45 MAb BC8-I-131 0365 anti-HIV proteins—Cangene 0313 Anti-CD45RB MAb 0366 Anti-HM1-24MAb Chugai 0314 Anti-CD48 MAb Hully-m3 0367 Anti-hR3 MAb 0315 Anti-CD48 MAb WM-63 0368 Anti-Human-Carcinoma-Antigen MAb—Epi 0316. Anti-CD5 MAb Becton Dickinson cyte 0317 Anti-CD5 MAb OX19 0369 Anti-ICAM-1 MAb Boehringer Ingelheim 0318 Anti-CD6 MAb 0370 Anti-ICAM-1 1A-29 Pharmacia 0319 Anti-CD7 MAb-PAP conjugate 0371 Anti-ICAM-1 MAb HA58 0320 Anti-CD7 MAb-ricin-chain-A conjugate 0372 Anti-ICAM-1 MAb YN1/1.7.4 0321) Anti-CD8 MAb Amerimmune, Cytodyn, Becton 0373 Anti-ICAM-3 MAb ICM3 ICOS Dickinson 0374 Anti-idiotype breast 1 1D10 0322 Anti-CD8 MAb 2-43 0375 Anti-idiotype breast cancer vaccine ACA14C5 0323 Anti-CD8 MAb OX8 0376 Anti-idiotype cancer vaccine ImClone Sys 0324 Anti-CD80 MAb P16C10 IDEC tems/Merck KGaA ImClone, Viventia Biotech 0325 Anti-CD80 MAb P7C10 ID Vaccine 0377 Anti-idiotype cancer vaccine 1A7 Titan 0326 Anti-CD8-idarubicin conjugate 0378 Anti-idiotype cancer vaccine3H1 Titan 0327. Anti-CEA MAb CE-25 0379 Anti-idiotype cancer vaccine TriAb Titan 0328. Anti-CEA MAb MN14 Immunomedics 0380 Anti-idiotype Chlamydia trachomatis vaccine 0329. Anti-CEA MAb MN14-PE40 conjugate Im 0381 Anti-idiotype colorectal cancer vaccine Novar munomedics tis 0330 Anti-CEA MAb T84.66-interleukin-2 conjugate 0382) Anti-idiotype colorectal cancer vaccine-Ony 0331 Anti-CEA sheep MAb KS Biomedix Holdings Wax 0332 Anti-cell surface monoclonal antibodies—Cam 0383 Anti-idiotype melanoma vaccine—IDEC Phar bridge Antibody Tech./Pharmacia maceuticals 0333 Anti-c-erbB2-anti-CD3 bifunctional MAb Ot 0384 Anti-idiotype ovarian cancer vaccine ACA 125 Suka 0385 Anti-idiotype ovarian cancer vaccine AR54—Al 0334 Anti-CMV MAb-Scotgen taRex 0335) Anti-complement 0386 Anti-idiotype ovarian cancer vaccine CA-125— 0336 Anti-CTLA-4 MAb AltaRex, Biomira 0337 Anti-EGFR catalytic antibody—Hesed Biomed 0387 Anti-IgE catalytic antibody Hesed Biomed 0338 anti-EGFR immunotoxin IVAX 0388 Anti-IgE MAb E26 Genentech 0339 Anti-EGFR MAb-Abgenix 0389 Anti-IGF-1 MAb 0340 Anti-EGFR MAb 528 0390 anti-inflammatory—GeneMax 0341 Anti-EGFR MAb KSB 107 KS Biomedix 0391 anti-inflammatory BTG 0342 Anti-EGFR MAb-DM1 conjugate Immuno 0392 anti-integrin peptides—Burnha Gen 0393 Anti-interferon-alpha-receptor MAb 64G12– 0343 Anti-EGFR MAb-LA1 Pharma Pacific Management 0344 Anti-EGFR sheep MAb KS Biomedix 0394 Anti-interferon-gamma MAb—Protein Design (0345 Anti-FAP MAb F19-1-131 Labs 0346) Anti-Fas IgM MAb CH11 0395 Anti-interferon-gamma polyclonal antibody— 0347 Anti-Fas MAb Jo2 Advanced Biotherapy 0348 Anti-Fas MAb RK-8 0396 Anti-interleukin-10 MAb 0349 Anti-Flt-1 monoclonal antibodies—ImClone 0397 Anti-interleukin-12 MAb 0350 Anti-fungal peptides—State University of New 0398 Anti-interleukin-1-beta polyclonal antibody— York R&D Systems 0351 antifungal tripeptides—BTG 0399 Anti-interleukin-2 receptor MAb 2A3 0352 Anti-ganglioside GD2 antibody-interleukin-2 (0400 Anti-interleukin-2 receptor MAb 33B3-1-Im fusion protein-Lexigen munotech US 2010/0080773 A1 Apr. 1, 2010 27

04.01 Anti-interleukin-2 receptor MAb ART-18 0451 Anti-Thy1 MAb 0402 Anti-interleukin-2 receptor MAb LO-Tact-1 0452 Anti-Thy 1.1 MAb 0403 Anti-interleukin-2 receptor MAb Mikbeta1 0453 Anti-tissue factor/factor VIIA sheep MAb KS 04.04 Anti-interleukin-2 receptor MAb NDS61 Biomedix 0405 Anti-interleukin-4 MAb 11B11 0454 Anti-TNF monoclonal antibodies—Centocor, 0406 Anti-interleukin-5 MAb Wallace Laboratories Chiron, Peptech, Pharacia, Serono 0407 Anti-interleukin-6 MAb–Centocor, Diaclone, 0455 Anti-TNF sheep MAb KS Biomedix Holdings Pharmadigm 0456 Anti-TNFalpha MAb Genzyme 04.08 Anti-interleukin-8 MAb Abgenix 0457 Anti-TNFalpha MAb B-C7 Diaclone 04.09 Anti-interleukin-8 MAb Xenotech 0458 Anti-tooth decay MAb Planet BioTech. 0410 Anti-JL1 MAb 0459 Anti-TRAIL receptor-1 MAb Takeda 0411 Anti-Klebsiella sheep MAb KS Biomedix 0460 Antitumour RNases NIH Holdings 0461 Anti-VCAM MAb 2A2. Alexion 0412 Anti-Laminin receptor MAb-liposomal doxoru 0462 Anti-VCAM MAb 3F4 Alexion bicin conjugate 0463 Anti-VCAM-1 MAb 0413 Anti-LCG MAb—Cytoclonal 0464 Anti-VECMAb ImClone 0414 Anti-lipopolysaccharide MAb VitaResc 0465 Anti-VEGF MAb Genentech 0415 Anti-L-selectin monoclonal antibodies—Protein 0466 Anti-VEGF MAb 2C3 Design Labs, Abgenix, Stanford University 0467 Anti-VEGF sheep MAb KS Biomedix Hold 0416 Anti-MBL monoclonal antibodies—Alexion/ ings Brigham and Women's Hospital 0468 Anti-VLA-4 MAb HP 1/2 Biogen 0417 Anti-MHC monoclonal antibodies 0469 Anti-VLA-4 MAb PS/2 0418 Anti-MIF antibody humanised IDEC, Cytok 0470 Anti-VLA-4 MAb R1-2 ine PharmaSciences 0471) Anti-VLA-4 MAb TA-2 0419 Anti-MRSA/VRSA sheep MAb KS Biomedix 0472 Anti-VAP-1 human MAb Holdings 0473 Anti-VRE sheep MAb KS Biomedix Holdings 0420 Anti-mu MAb- Novartis 0474) ANUP TranXenoGen 0421 Anti-MUC-1 MAb 0475 ANUP-1 Pharis 0422 Anti-MUC 18 0476 AOP-RANTES Senetek 0423 Anti-Nogo-A MAb IN1 0477 Apan-CH Praecis Pharmaceuticals 0424 Anti-nuclear autoantibodies—Procyon 0478 APC-8024 Demegen 0425 Anti-ovarian cancer monoclonal antibodies— 0479 ApoA-1—Milano, Pharmacia Dompe 0480 Apogen—Alexion 0426 Anti-p185 monoclonal antibodies 0481 apolipoprotein A1—AVanir 0427 Anti-p43 MAb 0482 Apollipoprotein E. Bio-Tech. General 0428 Antiparasitic vaccines 0483) Applaggin Biogen 0429 Anti-PDGF/bFGF sheep MAb KS Biomedix 0484 aprotinin ProdiGene 0430 Anti-properdin monoclonal antibodies—Ab 0485 APT-07OC AdProTech genix/Gliatech 0486 AR 177—Aronex Pharmaceuticals 0431 Anti-PSMA (prostrate specific membrane anti 0487 AR 209 Aronex Pharmaceuticals, Antigenics gen) 0488 ARGENT gene delivery systems—ARIAD 0432 Anti-PSMA MAb J591-BZL Biologics 0489 Arresten 0433 Anti-Rev MAb gene therapy 0490 ART-123 Asahi Kasei 0434 Anti-RSV antibodies—Epicyte, Intracell 0491 arylsulfatase B BioMarin 0435 Anti-RSV monoclonal antibodies—Medarex/ 0492 Arylsulfatase B. Recombinant human BioM MedImmune, Applied Molecular arin 0436 Evolution/MedImmune 0493 AS 1051—Ajinomoto 0437 Anti-RSV MAb, inhalation—Alkermes/MedIm 0494 ASI-BCL Intracell U 0495 Asparaginase-Merck 0438 Anti-RT gene therapy 0496 ATL-101—Alizyme 0439 Antisense K-ras RNA gene therapy 0497 Atrial Pharis 0440 Anti-SF-25 MAb 0498 Aurintricarboxylic acid-high molecular weight 0441 Anti-sperm antibody—Epicyte 0499 Autoimmune disorders—GPC Biotech/Morpho 0442 Anti-TacCFV)-PE38 conjugate Sys 0443 Anti-TAPA/CD81 MAbAMP1 0500 Autoimmune disorders and transplant rejec 0444 Anti-tat gene therapy tion Bristol-Myers Squibb/Genzyme Tra 0445 Anti-TCR-alphabeta MAb H57-597 0501 Autoimmune disorders/cancer Abgenix/Chi 0446 Anti-TCR-alphabeta MAb R73 ron, CuraGen 0447 Anti-tenascin MAb BC-4-I-131 0502. Autotaxin 0448 Anti-TGF-beta human monoclonal antibodies— 0503 Avicidin NeoRX Cambridge Antibody Tech., Genzyme 0504 axogenesis factor-1—Boston Life Sciences 0449) Anti-TGF-beta MAb 2G7 Genentech 0505 Axokine Regeneron 0450 Antithrombin III—Genzyme Transgenics, Aven 0506 B cell lymphoma vaccine Biomira tis, Bayer, Behringwerke, CSL, Myriad 0507 B7-1 gene therapy US 2010/0080773 A1 Apr. 1, 2010 28

0508 BABS proteins—Chiron 0568 calcitonin Inhale Therapeutics Systems, Aven 0509 BAM-002 Novelos Therapeutics tis, Genetronics, TranXenoGen, Unigene, Rhone Pou 0510) Basiliximab (anti CD25 MAb) Novartis lenc Rohrer 0511 Bay-16-9996 Bayer 0569 calcitonin oral Nobex, Emisphere, Pharma 0512 Bay-39-9437 Bayer ceutical Discovery 0513 Bay-50-4798 Bayer 0570 Calcitonin gene-related peptide Asahi Kasei 0514 BB-10153. British Biotech Unigene 0515 BBT-001- Bolder BioTech. 0571 calcitonin, human Suntory 0516 BBT-002. Bolder BioTech. 0572 calcitonin, nasal Novartis, Unigene 0517 BBT-003- Bolder BioTech. 0573 calcitonin, Panoderm Elan 0518 BBT-004 Bolder BioTech. 0574 calcitonin, Peptitrol Shire 0519 BBT-005 Bolder BioTech. 0575 calcitonin, salmon. Therapicon 0520 BBT-006 Bolder BioTech. 0576 calin Biopharm 0521 BBT-007 Bolder BioTech. 0577 Calphobindin I 0522 BCH-2763- Shire 0578 calphobindin I Kowa 0523 BCSF Millenium Biologix 0579 calreticulin NYU 0524 BDNF Regeneron Amgen 0580 Campath-1G 0525) Johnson & Johnson, Chiron 0581 Campath-1M 0526 Bectumomab-Immunomedics 0582 cancer therapy Cangene 0527 Beriplast Aventis 0583 cancer vaccine—Aixlie, Aventis Pasteur, Center 0528 Beta-adrenergic receptor gene therapy—Univer of Molecular Immunology, YM BioSciences, Cytos, sity of Arkansas Genzyme, Transgenics, Globemmune, Igeneon, 0529) bFGF Scios ImClone, Virogenetics, InterCell, Iomai, Jenner Bio 0530 BI 51013 Behringwerke AG therapies, Memorial Sloan-Kettering Cancer Center, 0531 BIBH 1—Boehringer Ingelheim Sydney Kimmel Cancer Center, NovaVax, Protein Sci 0532 BIM-23190 Beaufour-Ipsen ences, ArgoneX, SIGA 0533 birch pollen —Pharmacia 0584 Cancer vaccine ALVAC-CEA B7.1—Aventis 0534 bispecific fusion proteins—NIH Pasteur/Therion Biologics 0535 Bispecific MAb 2B1 Chiron 0585 Cancer vaccine CEA-TRICOM Aventis Pas 0536 Bitistatin teur/Therion Biologics 0537 BIWA 4 Boehringer Ingelheim 0586 Cancer vaccine gene therapy Cantab Pharma 0538 blood substitute Northfield, Baxter Intl. ceuticals 0539 BLP-25 Biomira 0587 Cancer vaccine HER-2/neu Corixa (0540 BLS-0597 Boston Life Sciences 0588 Cancer vaccine THERATOPE Biomira (0541 BLyS Human Genome Sciences 0589 cancer vaccine, PolyMASC Valentis (0542 BLyS radiolabelled Human Genome Sciences 0590 Candida vaccine Corixa, Inhibitex (0543 BM 06021-Boehringer Mannheim 0591 Canstatin ILEX (0544 BM-202 BioMarin 0592 CAP-18 Panorama 0545 BM-301 BioMarin 0593 Cardiovascular gene therapy—Collateral Thera (0546 BM-301 BioMarin peutics 0547 BM-302 BioMarin 0594 carperitide Suntory (0548 BMP 2–Genetics Institute/Medtronic-Sofamor 0595 Casocidin-1 Pharis Danek, Genetics Institute/Collagenesis, Genetics Insti 0596 CAT152 Cambridge Antibody Tech. tute/Yamanouch 0597 CAT 192 Cambridge Antibody Tech. 0549 BMP2 gene therapy 0598. CAT 213 Cambridge Antibody Tech. 0550 BMP 52 Aventis Pasteur, Biopharm 0599 Catalase Enzon 0551 BMP-2 Genetics Institute 0600 Cat-PAD Circassia 0552) BMS 182248 Bristol-Myers Squibb 0601 CB 0006 Celltech 0553 BMS 202448 Bristol-Myers Squibb 0602 CCK(27-32)—Akzo Nobel 0554 bone growth factors—IsoTis 0603 CCR2-64I NIH 0555 BPC-15 Pfizer 0604 CD, Procept Paligent 0556) brain natriuretic peptide 0605 CD154 gene therapy 0557 Breast cancer Oxford GlycoSciences/Medarex 0606 CD39. Immunex 0558 Breast cancer vaccine—Therion Biologics, 0607 CD39-L2 Hyseq Oregon 0608 CD39-L4 Hyseq 0559 BSSL PPL Therapeutics 0609 CD4 fusion toxin Senetek 0560 BST-2001- BioStratum 0610 CD4 IgG Genentech 0561 BST-3002- BioStratum 0611 CD4 receptor antagonists—Pharmacopeia/Pro 0562 BTI 322 genics 0563 butyrylcholinesterase Shire 0612 CD4 soluble Progenics 0564 C 6822 COR Therapeutics 0613 CD4. Soluble—Genzyme Transgenics 0565 C1 esterase inhibitor Pharming 0.614 CD40 ligand Immunex 0566 C3d adjuvant AdProTech 0615 CD4-ricin chain A Genentech 0567 CAB-2.1—Millennium 0616 CD59 gene therapy—Alexion US 2010/0080773 A1 Apr. 1, 2010 29

0617 CD8 TIL cell therapy—Aventis Pasteur 0673) CorSevin M 0618 CD8, soluble Avidex 0674) C-peptide analogues—Schwarz, 0619 CD95 ligand Roche 0675 CPI-1500 Consensus 0620 CDP 571 Celltech 0676 CRF Neurobiological Tech. 0621 CDP 850 Celltech 0677) cRGDfV pentapeptide 0622 CDP-860 (PEG-PDGF MAb) Celltech 0678 CRL 1095 CytRx 0623 CDP870 Celltech 0679) CRL 1336 CytRx 0624 CDS-1 Ernest Orlando 0680 CRL 1605 CytRx 0625 Cedelizumab Ortho-McNeil 0681 CS-560 Sankyo 0626 Cetermin—InSmed 0682 CSF ZymoGenetics 0627 CETP vaccine—Avant 0683 CSF-G Hangzhou, Dong-A, Hani (0628 Cetrorelix 0684 CSF-GM Cangene, Hunan, LG Chem 0629 0685 CSF-M Zarix 0630 CGH 400 Novartis 0686) CT 1579 Merck Frosst 0631 CGP 42934 Novartis 0687 CT 1786 Merck Frosst 0632 CGP 51901 Tanox 0688 CT-112 BTG 0633 CGRP Unigene 0689 CTB-134L Xenova 0634) CGS 27.913 Novartis 0690 CTC-111—Kaketsuken 0635 CGS 32359 Novartis 0691 CTGF FibroGen. 0636 Chagas disease vaccine—Corixa 0692 CTLA4-Ig Bristol-Myers Squibb 0637 chemokines—Immune Response 0693 CTLA4-Ig gene therapy 0638 CHH 380 Novartis 0694 CTP-37 AVI BioPharma 0639 chitinase Genzyme, ICOS 0695 C-type natriuretic peptide—Suntory 0640 Chlamydia pneumoniae vaccine—Antex Biolog 0696 CVS 995 Corvas Intl. ics 0697 CY 397 Nikko Kyodo 0641 Chlamydia trachomatis vaccine—Antex Biolog 0698 CY 1747 Epimmune ics 0699 CY 1748 Epimmune 0642 Chlamydia vaccine GlaxoSmithKline 0700 Cyanovirin-N 0643 Cholera vaccine CVD 103-HgR Swiss Serum 0701 Cystic fibrosis therapy CBR/IVAX and Vaccine Institute Berne 0702 CYT 351 0644 Cholera vaccine CVD 1 12 Swiss Serum and 0703 Traps—Regeneron Vaccine Institute Berne 0704 cytokines—Enzon, Cytoclonal 0645 Cholera vaccine inactivated oral SBL Vaccin 0705 Cytomegalovirus glycoprotein vaccine—Chiron, 0646 Chrysalin Chrysalis BioTech. Aquila Biopharmaceuticals, Aventis Pasteur, Virogenet (0647 CI-782 Hitachi Kase 1CS 0648 Ciliary neurotrophic factor Fidia, Roche 0706) Cytomegalovirus vaccine live—Aventis Pasteur 0649) CIM project—Active Biotech 0707 Cytosine deaminase gene therapy—GlaxoSmith 0650 CL 329753–Wyeth-Ayerst Kline 0651 CL22, Cobra—ML Laboratories 0708 DA-3003 Dong A 0652 Clenoliximab IDEC 0709) DAB389interleukin-6 Senetek 0653 Clostridium difficile antibodies—Epicyte 0710 DAB389interleukin-7 0654 clotting factors—Octagene 07.11 Daclizumab (anti-IL2R MAb) Protein Design 0655 CMB 401 Celltech Labs 0656 CNTF Sigma-Tau 0712 DAMP Incyte Genomics 0657 Cocaine abuse vaccine—Cantab, ImmuLogic, 0713 Daniplestim—Pharmacia Scripps 0714) -Amgen 0658 coccidiomycosis vaccine—Arizo 0715 DBI-3019 Diabetogen 0659 collagen Type I—Pharming 0716 DCC Genzyme 0660 Collagen formation inhibitors—FibroGen 0717 DDF Hyseq 0661 Collagen/hydroxyapatite/bone growth factor— 0718 decorin—Integra, Telios Aventis Pasteur, Biopharm, Orquest 0719 defensins—Large Scale Biology 0.662 collagenase-BioSpecifics 0720 DEGR-VIIa 0663 Colorectal cancer vaccine Wistar Institute 0721 Deimmunised antibody 3B6/22 AGEN 0664 Component B. Recombinant Serono 0722 Deimmunised anti-cancer antibodies—Biova 0665 Connective tissue growth factor inhibitors—Fib tion/Viragen roGen/Taisho 0723 Dendroamide A 0666 Contortrostatin 0724 Dengue vaccine Bavarian Nordic, Merck 0667 contraceptive vaccine—Zonagen 0725 denileukin diftitox—Ligand 0668 Contraceptive vaccine Hcg 0726 DES-1101- Desmos 0669 Contraceptive vaccine male reversible—IMMU 0727 desirudin Novartis CON 0728 desmopressin Unigene 0670) Contraceptive vaccine Zona pellucida—Zonagen 0729 Desmoteplase Merck, Schering AG 0671 Copper-64 labelled MAb TETA-1A3 NCI (0730 Destabilase 0672 Coralyne (0731 Diabetes gene therapy DeveloGen, Pfizer US 2010/0080773 A1 Apr. 1, 2010 30

0732 Diabetes therapy Crucell 0792 EP-51216 Asta Medica 0733 Diabetes type 1 vaccine—Diamyd Therapeutics 0793 EP-51389 Asta Medica 0734 DiaCIM YMBioSciences 0794 EPH family ligands—Regeneron 0735 dialytic oligopeptides—Research Corp 0795 Hitachi Kasei, 0736 Diamyd Diamyd Therapeutics Johnson & Johnson 0737 DiaPep227—Pepgen 0796 Epidermal growth factor fusion toxin Senetek 0738 DiavaX—Corixa 0797 Epidermal growth factor-genistein 0739 Digoxin MAb—Glaxo 0798 EPI-HNE-4. Dyax 0740 Diphtheria tetanus pertussis-hepatitis B vac 0799 EPI-KAL2 Dyax cine—GlaxoSmithKline 0800 Epoetin-alfa Amgen, Dragon Pharmaceuticals, 0741 DIR therapy—Solis Therapeutics Nanjing Huaxin 0742 DNase—Genentech 0801 Epratuzumab Immunomedics 0743 Dornase alfa Genentech 0802 Epstein-Barr virus vaccine Aviron/SmithKline 0744 Dornase alfa, inhalation—Genentech Beecham, Bioresearch 0745 Doxorubicin-anti-CEAMAb conjugate Immu (0803 Eptacog alfa Novo Nordisk nomedics (0804 Eptifibatide COR Therapeutics 0746 DP-107 Trimeris 0805 erb-38 0747 drotrecogin alfa Eli Lilly 0806 Erlizumab Genentech 0748 DTctGMCSF 0807 erythropoietin Alkermes, ProLease, Dong-A, 0749 DTP-polio vaccine Aventis Pasteur Elanex, Genetics Institute, LG Chem, Protein Sciences, 0750 DU 257-KM231 antibody conjugate Kyowa Serono, Snow Brand, SRCVB VECTOR, Transkaryotic 0751 dural graft matrix—Integra Therapies 0752 Duteplase—Baxter Intl. (0808 Erythropoietin Beta Hoffman La Roche 0753 DWP-401-Daewoong 0809 Erythropoietin/ Chugai 0754) DWP-404 Daewoong 0810 Escherichia coli vaccine North American Vac 0755 DWP-408 Daewoong cine, SBL Vaccin, Swiss Serum and Vaccine 0756) Dx 88 (Epi-KAL2) Dyax 0811. Institute Berne 0757 Dx 890 (elastin inhibitors)—Dyax 0812 —Immunex 0758 Ecoli O157 vaccine NIH 0813 examorelin—Mediolanum 0759) E21-R-BresaGen 0814 Exendin 4 0760 Eastern equine encephalitis virus vaccine 0815 exonuclease VII 0761 Echicetin 0816 F 105 Centocor 0762 Echinhibin 1 0817) F-992 Fornix 0763 Echistatin—Merck 0818. Factor IX Alpha Therapeutics, Welfide Corp., 0764 Echitamine CSL, enetics Institute/AHP, Pharmacia, PPL Therapeu 0765 Ecromeximab Kyowa Hakko tics 0766 EC-SOD PPL Therapeutics 0819 Factor IX gene therapy Cell Genesy's 0767 Eculizumab (5G1.1)—Alexion 0820 Factor VII Novo Nordisk, Bayer, Baxter Intl. O768 EDF Ajinomoto 0821) Factor VIIa PPL Therapeutics, ZymoGenetics 0769 EDN derivative NIH 0822. Factor VIII Bayer Genentech, Beaufour-Ipsen, (0770 EDNA NIH CLB, Inex, Octagen, Pharmacia, Pharming 0771) Edobacomab XOMA 0823. Factor VIII PEGylated Bayer 0772 Edrecolomab-Centocor 0824 Factor VIII fragments—Pharmacia (0773) EF 5077 0825. Factor VIII gene therapy Targeted Genetics 0774 Efalizumab Genentech 0826 Factor VIII sucrose formulation Bayer, Genen 0775 EGF fusion toxin Seragen, Ligand tech 0776 EGF-P64k vaccine—Center of Molecular Immu 0827) Factor VIII-2 Bayer nology 0828 Factor VIII-3 Bayer 0777 EL 246—LigoCyte 0829. Factor Xa inhibitors Merck, Novo Nordisk, 0778 elastase inhibitor—Synergen Mochida 0779) elcatonin. Therapicon 0830) Factor XIII ZymoGenetics 0780 EMD 72000 Merck KGaA 0831 Factors VIII and IX gene therapy—Genetics 0781 Emdogain BIORA Institute/Targeted Genetics 0782 emfilermin AMRAD 0832 Famoxin Genset 0783 Emoctakin Novartis 0833) Fas (delta) TM protein-LXR BioTech. 0784 enamel matrix protein BIORA 0834) Fas TR Human Genome Sciences 0785 Endo III NYU 0835) Felvizumab Scotgen 0786) endostatin EntreMed, Pharis 0836 FFR-VIIa Novo Nordisk 0787 Enhancins—Micrologix 0837 FG-001-F-Gene 0788 Enlimomab-Isis Pharm. 0838 FG-002 F-Gene 0789 Enoxaparin Sodium Pharmuka 0839 FG-004 F-Gene 0790 linked antibody nutrient depletion 0840 FG-005 F-Gene therapy—KS Biomedix Holdings 0841 FGF+fibrin Repair 0791) Eosinophil-derived neutralizing agent 0842 Fibrimage—Bio-Tech. General US 2010/0080773 A1 Apr. 1, 2010 31

0843 fibrin-binding peptides—ISIS Innovation 0896 Growth Factor Chiron, Atrigel, Atrix, Innoge 0844 fibrinogen PPL Therapeutics, Pharming netics, ZymoGenetics, Novo 0845 Chiron, NYU, Ramot, 0897 growth factor peptides—Biotherapeutics ZymoGenetics 0898 growth hormone—LG Chem 0846 fibrolase conjugate—Schering AG 0899 growth hormone, Recombinant human Serono 0847 —Amgen 0900 GT4086 Gliatech 0848 filgrastim—PDA modified Xencor 0901 GW 353430 GlaxoSmithKline 0849 FLT-3 ligand Immunex 0902 GW-278884 GlaxoSmithKline 0850 FN18 CRM 0903 H 11 Viventia Biotech 0851 follistatin Biotech Australia, Human Therapeu 0904 H5N1 influenza A virus vaccine Protein Sci tics CCCS 0852 follitropin alfa Alkermes, ProLease, Powder (0905 haemoglobin Biopure Ject, Serono, Akzo Nobel (0906 haemoglobin 3011, Recombinant Baxter 0853 Follitropin Beta Bayer, Organon Healthcare 0854 FP 59 0907 haemoglobin crosfumaril—Baxter Intl. 0855 FSH Ferring 0908 haemoglobin stabilized Ajinomoto 0856 FSH+LH-Ferring 0909 haemoglobin, recombinant—Apex 0857 F-spondin CeNeS (0910 HAF Immune Response 0858 fusion protein delivery system UAB Research 0911 Hantavirus vaccine Foundation 0912 HB 19 0859 fusion toxins—Boston Life Sciences (0913) HBNF Regeneron 0860 G 5598 Genentech 0914 HCC-1 Pharis 0861 GA-II Transkaryotic Therapies 0915 hCG Milkhaus 0862 Gamma-interferon analogues—SRC VB VEC 0916 hCG vaccine—Zonagen TOR (0917 HE-317 Hollis-Eden Pharmaceuticals 0863 Ganirelix Roche 0918. Heat shock protein cancer and influenza vac 0864 gastric lipase—Meristem cines—StressGen 0865 Gavilimomab 0919 Helicobacter pylori vaccine Acambis, Astra 0866 G-CSF Amgen, SRC VB VECTOR Zeneca/CSL, Chiron, Provalis 0867 GDF-1 CeNeS 0920 Helistat-G GalaGen 0868 GDF-5 Biopharm 0921 Hemolink Hemosol 0869 GDNF (glial derived neurotrophic factor)—Am 0922 hepapoietin Snow Brand gen 0923 heparanase InSight 0870 gelsolin-Biogen 0924 heparinase I—Ibex 0871 Gemtuzumab ozogamicin Celltech 0925 heparinase III Ibex 0872 Gene-activated epoetin-alfa Aventis Pharma— 0926 Hepatitis A vaccine American Biogenetic Sci Transkaryotic Therapies CCCS 0927 Hepatitis A vaccine inactivated 0873 Glanzmann thrombasthenia gene therapy 0928 Hepatitis A vaccine Nothav Chiron 0874) Yeda 0929 Hepatitis A-hepatitis B vaccine GlaxoSmith 0875 glial growth factor 2 CeNeS Kline 0876 GLP-1—Amylin, Suntory. TheraTech, Watson 0930 hepatitis B therapy Tripep 0877 GLP-1 peptide analogues—Zealand Pharaceuti 0931 Hepatitis. B vaccine—Amgen, Chiron SpA. cals Meiji Milk, NIS, Prodeva, Powder Ject, Rhein Biotech 0878 glucagon—Eli Lilly, ZymoGenetics 0932 Hepatitis B vaccine recombinant Evans Vac 0879 Glucagon-like peptide-1 7-36 amide Suntory cines, Epitec Combiotech, Genentech, MedImmune, 0880 Glucogen-like peptide-Amylin Merck Sharp & Dohme, Rhein Biotech, Shantha Bio 0881 Glucocerebrosidase-Genzyme technics, Vector, Yeda 0882 glutamate decarboxylase-Genzyme Transgen 0933 Hepatitis B vaccine recombinant TGP 943– ics Takeda 0883 Glycoprotein S3—Kureha 0934 Hepatitis C vaccine Bavarian Nordic, Chiron, 0884 GM-CSF Immunex Innogenetics Acambis 0885 GM-CSF tumour vaccine—Powder Ject 0935 Hepatitis D vaccine Chiron Vaccines 0886 GnRH immunotherapeutic Protherics 0936 Hepatitis E. vaccine recombinant Genelabs/ 0887 Goserelin (LhRH antagonist)—AstraZeneca GlaxoSmithKline, NovaVax 0888 gp75 antigen-ImClone 0937 Panorama, Sosei 0889 gp96—Antigenics 0938 hepatocyte growth factor kringle fragments—En 0890 GPIO 100 Galenica treMed 0891 GR 4991 W93 GlaxoSmithKline 0939 Her-2/Neu peptides Corixa 0892 Granulocyte colony-stimulating factor— 0940 Herpes simplex glycoprotein DNA vaccine— Dong-A Merck, Wyeth-Lederle Vaccines-Malvern, Genentech, 0893 Granulocyte colony-stimulating factor conjugate GlaxoSmithKline, Chiron, Takeda 0894 grass allergy therapy—Dynavax 0941 Herpes simplex vaccine Cantab Pharmaceuti 0895 GRF1-44 ICN cals, CEL-SCI, Henderson Morley US 2010/0080773 A1 Apr. 1, 2010 32

0942 Herpes simplex vaccine live ImClone Systems/ Medarex, Eos/Xenerex, Exelixis/Protein Design Labs, Wyeth-Lederle, Aventis Pasteur ImmunoGen/Raven, Medarex/B.Twelve, MorphoSys/ (0943 HGF derivatives Dompe ImmunoGen, XTL Biopharmaceuticals/Dyax 0944 hIAPP vaccine Crucell 0989 Human monoclonal antibodies—Medarex/ (0945) Hib-hepatitis B vaccine Aventis Pasteur Northwest Biotherapeutics, Medarex/Seattle Genetics 0946 HIC 1 0990 human metrin-1 Exelixis 0947 HIP Altachem 0991 human papillomavirus antibodies—Epicyte 0948 Hirudins—Biopharma, Cangene, Dongkook, 0992 Human papillomavirus vaccine Biotech Aus Japan Energy Corporation, Pharmacia Corporation, SIR tralia, IDEC, StressGen International, Sanofi-Synthelabo, Sotragene, Rhein 0993 Human papillomavirus vaccine MEDI 501– Biotech MedImmune/GlaxoSmithKline 0949 HIV edible vaccine ProdiGene 0994) Human papillomavirus vaccine MEDI 503/ 0950 HIV gp120 vaccine—Chiron, Ajiomoto, Glaxo MEDI 504 MedImmune/GlaxoSmithKline SmithKline, ID Vaccine, Progenics, VaxGen 0995 Human papillomavirus vaccine TA-CIN 0951 HIV gp120 vaccine gene therapy Cantab Pharmaceuticals 0952 HIV gp160 DNA vaccine PowderJect, Aventis 0996 Human papillomavirus vaccine TA-HPV Pasteur. Oncogen, Hyland Immuno, Protein Sciences Cantab Pharmaceuticals 0953 HIV gp41 vaccine Panacos 0997 Human papillomavirus vaccine TH-GW 0954 HIV HGP-30W vaccine CEL-SCI Cantab/GlaxoSmithKline 0955 HIV immune globulin Abbott, Chiron 0998 human polyclonal antibodies—Biosite/Eos Bio 0956 HIV peptides American Home Products Tech/Medarex 0957 HIV vaccine Applied bioTech. Axis Genetics, 0999 human type II anti factor VIII monoclonal anti Biogen, Bristol-Myers Squibb, Genentech, Korea Green bodies—ThromboGenics Cross, NIS, Oncogen, Protein Sciences Corporation, 1000 humanised anti glycoprotein Ib murine mono Terumo, Tonen Corporation, Wyeth-Ayerst, Wyeth-Le clonal antibodies—ThromboGenics derle Vaccines-Malvern, Advanced BioScience Labora 1001 HumaRAD Intracell tories, Bavarian Nordic, Bavarian Nordic/Statens Serum 1002 HuMax EGFR Genmab Institute, GeneCure, Immune Response, Progenics, 1003 HuMax-CD4. Medarex Therion Biologics, United Biomedical, Chiron 1004 HUMax-IL 15 Genmab 0958 HIV vaccine vCP1433 Aventis Pasteur 1005 HYB 190 Hybridon 0959 HIV vaccine vCP 1452 Aventis Pasteur 1006 HYB 676. Hybridon 0960 HIV vaccine vCP205-Aventis Pasteur 1007 I-125 MAb A33 Celltech 0961 HL-9 American BioScience 1008 Ibritumomab tiuxetan IDEC 0962) HM-9239 Cytran 1009 IBT-9401 Ibex 0963 HML-103 Hemosol 1010 IBT-9402 Ibex 0964 HML-104 Hemosol 1011 IC 14-ICOS 0965 HML-105 Hemosol 1012 Idarubicin anti-Ly-2.1 0966 HML-109 Hemosol 1013 IDEC 114 IDEC 0967 HML-1 10 Hemosol 1014 IDEC 131-IDEC 0968 HML-121 Hemosol 1015 IDEC 152 IDEC 0969 hNLP Pharis 1016 IDM 1 IDM 0970 Hookworm vaccine 1017 IDPS Hollis-Eden Pharmaceuticals 0971 host-vector vaccines—Henogen 1018 iduronate-2-sulfatase Transkaryotic Therapies 0972 HPM 1- Chugai 1019 IGF/IBP-2-13 Pharis 0973 HPV vaccine MediCiene 1020. IGN-101-Igeneon 0974 HSA Meristem 1021 IK HIR02 Iketon 0975 HSF StressGen 1022. IL-11 Genetics Institute/AHP 0976. HSP carriers Weizmann, Yeda, Peptor 1023 IL-13-PE38 NeoPharm 0977 HSPPC-70 Antigenics 1024 IL-17 receptor Immunex 0978 HSPPC-96, pathogen-derived Antigenics 1025 IL-18BP Yeda 0979. HSV 863 Novartis 1026 IL-1Hy1-Hyseq 0980 HTLV-I DNA vaccine 1027 IL-1 B-Celltech 0981 HTLV-I vaccine 1028. IL-1B adjuvant Celltech 0982 HTLV-II vaccine Access 1029 IL-2 Chiron 0983 HU901 Tanox 1030 IL-2+IL-12 Hoffman LaRoche 0984 Hu23F2G ICOS 1031 IL-6/sIL-6R fusion Hadasit 0985 HuHMFG1 1032. IL-6R derivative Tosoh 0986 HumaLYM Intracell 1033 IL-7-Dap 389 fusion toxin Ligand 0987 Human krebs statika Yamanouchi 1034) IM-862 Cytran 0988 human monoclonal antibodies—Abgenix/Bio 1035 IMC-1C11 ImClone gen, Abgenix/Corixa, Abgenix/Immunex, Abgenix/ 1036) imiglucerase Genzyme Lexicon, Abgenix/Pfizer, Athersys/Medarex, Biogen/ 1037 Immune globulin intravenous (human)—Hoff MorphoSys, CAT/Searle, Centocor/Medarex, Corixa/ man LaRoche Kirin Brewery, Corixa/Medarex, Eos BioTech./ 1038 immune privilege factor Proneuron US 2010/0080773 A1 Apr. 1, 2010 33

1039 Immunocal Immunotec 1089 interferon (beta1A), inhale—Biogen 1040. Immunogene therapy—Briana Bio-Tech 1090 interferon (B1b) Chiron 1041. Immunoliposomal 5-fluorodeoxyuridine-di 1091 interferon (tau) Pepgen palmitate 1092] Interferon alfacon-1—Amgen 1042 immunosuppressant vaccine—Aixlie 1093 Interferon alpha-2a vaccine 1043 immunotoxin Antisoma, NIH 1094 Interferon Beta 1b Schering/Chiron, Inter 1044. ImmuRAIT-Re-188—Immunomedics Mune 1045 imreg-1—Imreg 1095 Boehringer Ingelheim, 1046 infertility—Johnson & Johnson, E-TRANS Sheffield, Rentschler, Hayashibara 1047 Infliximab Centocor 1096 interferon receptor, Type I Serono 1048 Influenza virus vaccine—Aventis Pasteur, Pro 1097 interferon(Gamma1B) Genentech tein Sciences 1098 Interferon-alpha-2b--ribavirin Biogen, ICN 1049 inhibin Biotech Australia, Human Therapeu 1099 Interferon-alpha-2b gene therapy—Schering tics Plough 1050 Inhibitory G protein gene therapy 1100 Interferon-con1 gene therapy 1051 INKP-2001 In Kine 1101 interleukin-1 antagonists—Dompe 1052 Inolimomab Diaclone 1102 Interleukin-1 receptor antagonist Abbott 1053 insulin—Autoimmune. Altea, Biobras, Bio Bioresearch, Pharmacia Sante, Bio-Tech. General, Chong Kun Dang, Emi 1103 Interleukin-1 receptor type I—Immunex sphere, Flamel, Provalis, Rhein Biotech, TranXenoGen 1104 interleukin-1 receptor Type II—Immunex 1054 insulin (bovine) Novartis 1105 Interleukin-1 trap—Regeneron 1055 insulin analogue Eli Lilly 1106 Interleukin-1-alpha Immunex/Roche 1056 Insulin Aspart Novo Nordisk 1107 interleukin-2-SRC VB VECTOR, Ajinomoto, 1057 Novo Nordisk Biomira, Chiron IL-2/diphtheria toxin Ligand 1058 insulin glargine Aventis 1108 Interleukin-3 Cangene 1059 insulin inhaled Inhale Therapeutics Systems, 1109 Interleukin-4 Immunology Ventures, Sanofi Alkermes Winthrop, Schering-Plough, Immunex/Sanofi Win 1060 insulin oral Inovax throp, Bayer, Ono 1061 insulin, AeroDose—AeroGen 1110 interleukin-4+TNF-Alpha NIH 1062 insulin, AERX Aradigm 1111 interleukin-4 agonist Bayer 1063 insulin, BEODAS Elan 1112 interleukin-4 fusion toxin-Ligand 1064 insulin, Biphasix Helix 1113 Interleukin-4 receptor Immunex, Immun 1065 insulin, buccal Generex 1114 Interleukin-6—Ajinomoto, Cangene, Yeda, 1066 insulin, I2R Flemington Genetics Institute, Novartis 1067 insulin, intranasal Bentley 1115 interleukin-6 fusion protein 1068 insulin, oral Nobex, Unigene 1116 interleukin-6 fusion toxin Ligand, Serono 1069 insulin, Orasome Endorex 1117 interleukin-7 IC Innovations 1070 insulin, ProMaxx Epic 1118 interleukin-7 receptor Immunex 1071 insulin, Quadrant Elan 1119 interleukin-8 antagonists—Kyowa Hakko/Mil 1072 insulin, recombinant—Aventis lennium/Pfizer 1073 insulin, Spiros Elan 1120 interleukin-9 antagonists—Genaera 1074 insulin, Transfersome IDEA 1121 Interleukin-10 DNAX, Schering-Plough 1075 insulin, Zymo, recombinant Novo Nordisk 1122 Interleukin-10 gene therapy 1076 insulinotropin Scios 1123 interleukin-12—Genetics Institute, Hoffman La 1077 Insulysin gene therapy Roche 1078 integrin antagonists—Merck 1124 interleukin-13—Sanofi 1079 interferon (Alpha2). SRC VB VECTOR, 1125 interleukin-13 antagonists—AMRAD Viragen, Dong-A, Hoffman La-Roche, Genentech 1126 Interleukin-13-PE38QQR 1080 interferon BioMedicines, Human Genome Sci 1127 interleukin-15—Immunex CCCS 1128 interleukin-16—Research Corp 1081 interferon (Alfa-n3)—Interferon Sciences Intl. 1129 interleukin-18 GlaxoSmithKline 1082 interferon (Alpha), Biphasix—Helix 1130 Interleukin-18 binding protein Serono 1083 interferon (Alpha) Amgen, BioNative, Novar 1131 Ior-P3—Center of Molecular Immunology tis, Genzyme Transgenics, Hayashibara, Inhale Thera 1132) IP-10 NIH peutics Systems, Medusa, Flamel, Dong-A, GeneTrol, 1133) IPF Metabolex Nastech, Shantha, Wassermann, LG Chem, Sumitomo, 1134 IR-501—Immune Response Aventis, Behring EGIS, Pepgen, Servier, Rhein Biotech 1135 ISIS 9.125 Isis Pharmaceuticals 1084 interferon (Alpha2A) 1136 ISURF No. 1554 Millennium 1085 interferon (Alpha2B)—Enzon, Schering-Plough, 1137 ISURF No. 1866–Iowa State Univer. Biogen, IDEA 1138 ITF-1697 Ital farmaco 1086 interferon (Alpha-N1)—GlaxoSmithKline 1139 IXC 162 Ixion 1087 interferon (beta) Rentschler, GeneTrol, Mer 1140 J 695 Cambridge Antibody Tech., Genetics istem, Rhein Biotech, Toray, Yeda, Daiichi, Mochida Inst., Knoll 1088 interferon (Beta 1A) Serono, Biogen 1141 Jagged--FGF Repair US 2010/0080773 A1 Apr. 1, 2010 34

1142 JKC-362—Phoenix Pharmaceuticals 1205 LY-355101 Eli Lilly 1143 JTP-2942—Japan Tobacce 1206 Lyme disease DNA vaccine Vical/Aventis Pas 1144 Juman monoclonal antibodies—Medarex/Raven teur 1145 K02—AXys Pharmaceuticals 1207 Lyme disease vaccine—Aquila Biopharmaceuti 1146 Keliximab IDEC cals, Aventis, Pasteur, Symbicom, GlaxoSmithKline, 1147 Keyhole limpet haemocyanin Hyland Immuno, MedImmune 1148 KGF Amgen 1208 Lymphocytic choriomeningitis virus vaccine 1149) KM871–Kyowa 1209 lymphoma vaccine Biomira, Genitope 1150 KPI 135 Scios 1210 LYP18 1151 KPI-022 Scios 1211 lys plasminogen, recombinant 1152) Kringle 5 1212 Lysosomal storage disease gene therapy—Avi 1153 KSB 304 gen 1154) KSB-201- KS Biomedix 1213 lysostaphin Nutrition 21 1155 L 696418 Merck 1214 M23—Gruenenthal 1156) L 703801 Merck 1215 M1 monoclonal antibodies—Acorda Therapeu 1157 L1-Acorda tics 1158] L-761191 Merck 1216 MA 16N7C2 Corvas Intl. 1159 lactoferrin—Meristem, Pharming, Agennix 1217 malaria vaccine—GlaxoSmithKline, AdPro 1160 lactoferrin cardio—Pharming Tech, Antigenics, Apovia, Aventis Pasteur, Axis Genet 1161. LAG-3—Serono ics, Behringwerke, CDCP, Chiron Vaccines, Genzyme 1162 LAIT GEMMA Transgenics, Hawaii, MedImmune, NIH, NYU, OXXon, 1163) LAK cell cytotoxin-Arizona Rochef Saramane, Biotech Australia, RX Tech 1164 lamellarins—PharmaMar/University of Malaga 1218 Malaria vaccine CDC/NIIMALVAC-1 1165 laminin A peptides NIH 1219 malaria vaccine, multicomponent 1166 lanoteplase-Genetics Institute 1220 mammaglobin Corixa 1167) laronidase BioMarin 1221 mammastatin-Biotherapeutics 1168) Lassa fever vaccine 1222 mannan-binding lectin-Natimmu 1169) LCAT NIH 1223 mannan-MUC1—Psiron 1170 LDP 01- Millennium 1224 MAP 30 1171 LDPO2- Millennium 1225 Marinovir Phytera 1172 Lecithinized Superoxide dismutase-Seikagaku 1226. MARstem Maret 1173 LeIF adjuvant Corixa 1227 MB-015 Mochida 1174) leishmaniasis vaccine Corixa 1228 MBP ImmuLogic 1175 lenercept—Hoffman La-Roche 1229 MCI-028 Mitsubishi-Tokyo 1176 —Aventis, Chugai 1230 MCIF Human Genome Sciences 1177 lepirudin Aventis 1231. MDC Advanced BioScience Akzo Nobel, 1178) —Amgen, IC Innovations ICOS 1179 Leptin gene therapy—Chiron Corporation 1232 MDX 11 Medarex 1180) leptin, 2nd-generation—Amgen 1233 MDX 210 Medarex 1181 leridistim—Pharmacia 1234 MDX 22 Medarex 1182 leuprolide, ProMaxx Epic 1235 MDX 22 1183. leuprorelin, oral—Unigene 1236 MDX 240 Medarex 1184 LeuTech Papatin 1237 MDX 33 1185) LEX 032 SuperGen 1238 MDX 44 Medarex 1186 LiDEPT Novartis 1239 MDX 447 Medarex 1187 Lintuzumab (anti-CD33 MAb) Protein Design 1240 MDX H210 Medarex Labs 1241 MDX RA Houston BioTech., Medarex 1188) lipase-Altus Biologics 1242 ME-104 Pharmexa 1189. lipid A vaccine EntreMed 1243 Measles vaccine 1190) lipid-linked anchor Tech. ICRT, ID Biomedical 1244 Cephalon/Chiron, Chiron 1191. liposome-CD4 Tech. Sheffield 1245 MEDI 488 MedImmune 1192) Listeria monocytogenes vaccine 1246 MEDI5OO 1193) LMB 1 1247 MEDI 507 BioTransplant 1194 LMB 7 1248 melanin concentrating hormone—Neurocrine 1195) LMB 9 Battelle Memorial Institute, NIH Biosciences 1196 LM-CD45 Cantab Pharmaceuticals 1249 melanocortins OMRF 1197 lovastatin Merck 1250 Melanoma monoclonal antibodies—Viragen 1198) LSA-3 1251 melanoma vaccine—GlaxoSmithKline, Akzo 1199) LT-B receptor—Biogen Nobel, Avant, Aventis Pasteur, Bavarian Nordic, Biovec 1200 lung cancer Vaccine—Corixa tor, CancerVax, Genzyme Molecular Oncology, Hum 1201) luSupultide Scios bolt, ImClone Systems, Memorial, NYU, OXXon 1202 L-Vax AVAX 1252 Melanoma vaccine Magevac Therion 1203 LY355455 Eli Lilly 1253 memory enhancers—Scios 1204 LY 366405 Eli Lilly 1254 meningococcal B vaccine Chiron US 2010/O080773 A1 Apr. 1, 2010 35

1255 meningococcal vaccine—CAMR 1309 MAb 50-6 1256 Meningococcal vaccine group B conjugate— 1310 MAb 50-61A Institut Pasteur North American Vaccine 1311 MAb 5A8 Biogen 1257 Meningococcal vaccine group B recombinant— 1312 MAb 791T/36-methotrexate conjugate BioCh em. Vaccines, Microscience 1313) MAb 7c11.e.8 1258 Meningococcal vaccine group Y conjugate— 1314 MAb 7E11 C5-selenocystamine conjugate North American Vaccine 1315 MAb 93KA9 Novartis 1259 Meningococcal vaccine groups A B and C conju 1316 MAb A5B7-cisplatin conjugate Biodynamics gate-North American Vaccine Research, Pharmacia 1260 Mepolizumab GlaxoSmithKline 1317 MAb A5B7-I-131 1261 Metastatin EntreMed, Takeda 1318 MAb A7 1262 Met-CkB7—Human Genome Sciences 1319 MAb A717 Exocell 1263 met-enkephalin TNI 1320 MAb A7-Zinostatin conjugate 1264 METH-1—Human Genome Sciences 1321 MAb ABX-RB2—Abgenix 1265 methioninase-AntiCancer 1322 MAb ACA 11 1266 Methionine lyase gene therapy—AntiCancer 1323 MAb AFP-I-131—Immunomedics 1267 Met-RANTES Genexa Biomedical, Serono 1324 MAb AP1 1268 1325) MAb AZ1 1269 Microtubule inhibitor MAb Immunogen/Ab 1326 MAb B3-LysPE40 conjugate genix 1327 MAb B4 United Biomedical 1270 MGDF Kirin 1328 MAb B43 Genistein-conjugate 1271 MGV Progenics 1329 MAb B43.13-Tc-99m Biomira 1272 micrin-Endocrine 1330 MAb B43-PAP conjugate 1273 microplasmin ThromboGenics 1331 MAb B4G7-gelonin conjugate 1274 MIF Genetics Institute 1332 MAb BCM 43-daunorubicin conjugate—BCM 1275 migration inhibitory factor NIH Oncologia 1276 Mim CD4.1—Xycte Therapies 1333 MAb BIS-1 1277 mirostipen-Human Genome Sciences 1334 MAb BMS 181170 Bristol-Myers Squibb 1278 Mitumomab (BEC-2)—ImClone Systems, 1335 MAb BR55-2 Merck KGaA 1336 MAb BW494 1279 MK 852 Merck 1337 MAb C242-DM1 conjugate ImmunoGen 1280 MLN 1202 (Anti-CCR2 monoclonal antibody)— 1338 MAb C242-PE conjugate Millenium Pharmaceuticals 1339 MAb c30-6 1281 Mobenakin. NIS 1340 MAb CA208-cytorhodin-S conjugate Hoechst 1282 molgramostim—Genetics Institute, Novartis Japan 1283 monoclonal antibodies—Abgenix/Celltech, 1341 MAb CC49. Enzon lmmusol/Medarex, Viragen/Roslin Institute, Cambridge 1342 MAb ch14.1 Antibody Tech./Elan 1343 MAb CH14.18-GM-CSF fusion protein Lexi 1284 MAb 108 gen 1285 MAb 10D5-eMAb 14.18-interleukin-2 immuno 1344 MAb chCE7 cytokine—Lexigen 1345 MAb CI-137 AMRAD 1286 MAb 14G2a 1346 MAb cisplatin conjugate 1287| MAb 15A1 O MAb 170 Biomira 1347 MAb CLB-CD19 1288 MAb 177LL CC49 MAb 17F9 1348 MAb CLB-CD19 V. 1289 MAb 1D7 1349 MAb CLL-1 Peregrine 1290 MAb 1 F7 Immune Network 1350 MAb CLL-1-GM-CSF conjugate 1291 MAb 1H10-doxorubicin conjugate 1351 MAb CLL-1-IL-2 conjugate Peregrine 1292 MAb 26-2F 1352 MAb CLN IgG doxorubicin conjugates 1293 MAb 2A11 1353) MAb conjugates—Tanox 1294 MAb 2E1 RW Johnson 1354 MAb D612 1295 MAb 2F5 1355 MAb Dal B02 1296 MAb 31.1—International Biolmmune Systems 1356 MAb DC101 - ImClone 1297 MAb 32 Cambridge Antibody Tech., Peptech 1357 MAb EA 1 1298 MAb 323A3 Centocor 1358 MAb EC708 Biovation 1299 MAb 3C5 1359 MAb EP-5C7 Protein Design Labs 1300 MAb 3F12 1360 MAb ERIC-1 ICRT 1301 MAb 3F8 1361) MAb F105 gene therapy 1302 MAb 42/6 1362 MAb FC 2.15 1303 MAb 425 Merck KGaA 1363 MAb G250 Centocor 1304 MAb 447-52D Merck Sharp & Dohme 1364 MAb GA6 1305 MAb 45-2D9 haematoporphyrin conjugate 1365 MAb GAT33 1306 MAb 4B4 1366 MAb Gliomab-H Viventia Biotech 1307 MAb 4E3-CPA conjugate—BCM Oncologia 1367 MAb HB2-saporin conjugate 1308 MAb 4E3-daunorubicin conjugate 1368 MAb HD 37 US 2010/0080773 A1 Apr. 1, 2010 36

1369 MAb HD37-ricin chain-A conjugate 1431 MAb TEC 11 1370 MAb HNK20 Acambis 1432 MAb TES-23 Chugai 1371) MAb huN901-DM1 conjugate ImmunoGen 1433 MAb TM31- Avant 1372 MAb I-131 CC49 Corixa 1434 MAb TNT-1 Cambridge Antibody Tech., Per 1373 MAb ICO25 egrine 1374 MAb ICR12-CPG2 conjugate 1435 MAb TNT-3 1375 MAb ICR-62 1436 MAb TNT-3 IL2 fusion protein 1376 MAb IRac-ricin A conjugate 1437 MAb TP3-At-211 1377 MAb K1 1438 MAb TP3-PAP conjugate 1378 MAb KS1-4-methotrexate conjugate 1439 MAb UJ13A ICRT 1379 MAb L6 Bristol-Myers Squibb, Oncogen 1440 MAb UN3 1380 MAb LiCO 16-88 1441 MAb ZME-018-gelonin conjugate 1381 MAb LL2-I-131—Immunomedics 1442 MAb-BC2 GlaxoSmithKline 1382) MAb LL2-Y-90 1443 MAb-DM1 conjugate ImmunoGen 1383 MAb LS2D617. Hybritech 1444 MAb-ricin-chain-A conjugate XOMA 1384) MAb LYM-1-gelonin conjugate 1445 MAb-temoporfin conjugates 1385 MAb LYM-1-I-131 1446 Monopharm C Viventia Biotech 1386) MAb LYM-1-Y-90 1447 monteplase-Eisai 1387 MAb LYM-2 Peregrine 1448 montirelin hydrate Gruenenthal 1388 MAb M195 1449 moroctocog alfa-Genetics Institute 1389) MAb M195-bismuth 213 conjugate Protein 1450 Moroctocog-alfa-Pharmacia Design Labs 1451 MP4 1390 MAb M195-gelonin conjugate 1452 MP-121 Biopharm 1391 MAb M195-I-131 1453 MP-52. Biopharm 1392 MAb M195-Y-90 1454) MRA Chugai 1393 MAb MA 33H1 Sanofi 1455 MS 281 68 Mitsui Chemicals, Nihon Schering 1394 MAb MAD11 1456 MSH fusion toxin Ligand 1395 MAb MGb2 1457 MSI-99 Genaera 1396 MAb MINTS 1458 MT 201 Micromet 1397 MAb MK2-23 1459 Muc-1 vaccine Corixa 1398 MAb MOC31 ETA(252-613) conjugate 1460 Mucosal tolerance—Aberdeen 1399 MAb MOC-31-In-111 1461 mullerian inhibiting subst 1400 MAb MOC-31-PE conjugate 1462 muplestim—Genetics Institute, Novartis, DSM 1401 MAb MR6 Anti-Infectives 1402 MAb MRK-16 Aventis Pasteur 1463 murine MAb KS Biomedix 1403 MAb MS11 G6 1464 Mutant somatropin—JCR Pharmaceutical 1404 MAb MX-DTPA BrE-3 1465 MV 833 Toagosei 1405 MAb MY9 1466 Mycoplasma pulmonis vaccine 1406 MAb Nd2 Tosoh 1467 Mycoprex XOMA 1407 MAb NG-1-Hygeia 1468 myeloperoxidase-Henogen 1408 MAb NM01 - Nissin Food 1469 myostatin-Genetics Institute 1409 MAb OC 125 1470 Nacolomab tafenatox Pharmacia 1410 MAb OC 125-CMA conjugate 1471) Nagrecor—Scios 1411 MAb OKI-1- Ortho-McNeil 1472 nagrestipen British Biotech 1412 MAb OX52 Bioproducts for Science 1473 NAP-5. Corvas Intl. 1413 MAb PMA5 1474) NAPc2 Corvas Intl. 1414 MAb PR1 1475 nartograstim—Kyowa 1415 MAb prost 30 1476 Natalizumab—Protein Design Labs 1416 MAb R-24 1477 Nateplase NIH, Nihon Schering 1417 MAb R-24C. Human GD3- Celltech 1478 nateplase—Schering AG 1418 MAb RFB4-ricin chain A conjugate 1479 NBI-3001- Neurocrine BioSci. 1419 MAb RFT5-ricin chain A conjugate 1480 NBI-5788 Neurocrine BioSci. 1420 MAb SC 1 1481 NBI-6024 Neurocrine BioSci. 1421 MAb SM-3 ICRT 1482 Nef inhibitors BRI 1422 MAb SMART 1D10 Protein Design Labs 1483) Neisseria gonorrhoea vaccine—Antex Biologics 1423 MAb SMART ABL 364 Novartis 1484 Neomycin B-arginine conjugate 1424 MAb SN6f 1485 Nerelimomab-Chiron 1425 MAb SN6f-deglycosylated ricin A chain conju 1486 —Amgen Chiron, Genen gate tech 1426 MAb SN6 1487 Nerve growth factor gene therapy 1427 MAb SN7-ricin chain A conjugate 1488 nesiritide citrate—Scios 1428 MAb T101-Y-90 conjugate Hybritech 1489 -2 CeNeS 1429 MAb T-88 Chiron 1490 neurocan NYU 1430 MAb TB94 Cancer ImmunoBiology 1491 neuronal delivery system CAMR US 2010/0080773 A1 Apr. 1, 2010 37

1492 Neurophil inhibitory Factor Corvas 1555 osteoporosis peptides—Integra, Telios 1493) Neuroprotective vaccine University of Auck 1556) —Amgen, SnowBrand land 1557) otitis media vaccines—Antex Biologics 1494 neurotrophic chimaeras—Regeneron 1558 ovarian cancer University of Alabama 1495 neurotrophic factor NSGene, CereMedix 1559 OX40-IgG fusion protein Cantab, Xenova 1496 NeuroVax Immune Response 1560 P 246 Diatide 1497 Genentech 1561 P 30 Alfacell 1498 neutral endopeptidase-Genentech 1562 p1025 Active Biotech 1499 NGF enhancers—NeuroSearch 1563 P-113 Demegen 1500 NHL vaccine—Large Scale Biology 1564 P-16 peptide Transition Therapeutics 1501 NIP45 Boston Life Sciences 1565 p43—Ramot 1502) NKI-B2O 1566 P-50 peptide Transition Therapeutics 1503 NM01 - Nissin Food 1567 p53+RAS vaccine NIH, NCI 1504 NMI-139 NitroMed 1568 PACAP(1-27) analogue 1505 NMMP Genetics Institute 1569 paediatric vaccines—Chiron 1506 NN-2211 Novo Nordisk 1570 Pafase ICOS 1507 Noggin-Regeneron 1571 PAGE-4 plasmid DNA IDEC 1508 Nonacog alfa 1572 PAI-2—Biotech Australia, Human Therapeutics 1509 Norelin Biostar 1573 (keratinocyte growth factor)—Amgen 1510 Norwalk virus vaccine 1574) Palivizumab MedImmune 1511 NRLU 10 NeORX 1575) PAM 4 Merck 1512 NRLU 1 OPE NeORX 1576 pamiteplase—Yamanouchi 1513 NT-3—Regeneron 1577) pancreatin, Minitabs—Eurand 1514 NT-4/5 Genentech 1578 Pangen—Fournier 1515 NU 3056 1579) Pantarin—Selective Genetics 1516 NU 3076 1580 Parainfluenza virus vaccine—Pharmacia, Pierre 1517 NX 1838 Gilead Sciences Fabre 1518 NY ESO-1/CAG-3 antigen NIH 1581 paraoxanase-Esperion 1519 NYVAC-7 Aventis Pasteur 1582 —Abiogen, Korea Green 1520 NZ-1002 Novazyme Cross 1521 obesity therapy Nobex 1583 Parathyroid hormone (1-34) Chugai/Suntory 1522 OC 10426 Ontogen 1584 Parkinson's disease gene therapy Cell Gene 1523 OC 144093 Ontogen SyS/Ceregene 1524 OCIF Sankyo 1585 Parvovirus vaccine MedImmune 1525 Oct-43—Otsuka 1586 PCP-Scan Immunomedics 1526 Odulimomab–Immunotech 1587 PDGF Chiron 1527 OK PSA liposomal 1588 PDGF cocktail Theratechnologies 1528 OKT3-gamma-1-ala-ala 1589 peanut allergy therapy—Dynavax 1529 OM991 1590 PEG anti-ICAMMAb Boehringer Ingelheim 1530 OM992 1591 PEG asparaginase Enzon 1531 Omalizumab Genentech 1592 PEG glucocerebrosidase 1532 oncoimmunin-L- NIH 1593 PEGhirudin Knoll 1533 Oncolysin B ImmunoGen 1594 PEG interferon-alpha-2a-Roche 1534 Oncolysin CD6—ImmunoGen 1595 PEG interferon-alpha-2b-tribavirin Biogen, 1535 Oncolysin M. ImmunoGen Enzon, ICN Pharmaceuticals, Schering-Plough 1536 Oncolysin S—ImmunoGen 1596. PEG MAb A5B7 1537 Oncophage—Antigenics 1597 Pegacaristim Amgen Kirin Brewery—Zy 1538 . Bristol-Myers Squibb moGenetics 1539 Onco Vax-CL Jenner Biotherapies 1598 Pegaldesleukin Research Corp 1540 Onco Vax-P Jenner Biotherapies 1599 pegaspargase Enzon 1541 onercept Yeda 1600 —Amgen 1542 onychomycosis vaccine—Boehringer Ingelheim 1601 PEG-interferon Alpha Viragen 1543 opebecan XOMA 1602 PEG-interferon Alpha 2A Hoffman LaRoche 1544 opioids—Arizona 1603 PEG-interferon Alpha 2B Schering-Plough 1545 Genetics Institute 1604 PEG-r-hirudin Abbott 1546 Oregovomab AltaRex 1605 PEG-rHuMGDF Amgen 1547 Org-33408 b–Akzo Nobel 1606 PEG-uricase Mountain View 1548 Orolip DP EpiCept 1607 Pegvisomant Genentech 1549 OryzacyStatin 1608 PEGylated proteins, PolyMASC Valentis 1550 OSA peptides—GenSci Regeneration 1609 PEGylated recombinant native human leptin— 1551 osteoblast-cadherin GF Pharis Roche 1552 Osteocalcin-thymidine kinase gene therapy 1610 Pemtumomab 1553 osteogenic protein-Curis 1611 Penetratin Cyclacel 1554 OraPharma 1612 Pepscan-Antisoma US 2010/0080773 A1 Apr. 1, 2010 38

1613 peptide G-Peptech, ICRT 1675 Pigment epithelium derived factor 1614 peptide vaccine NIH, NCI 1676 plasminogen activator inhibitor-1, recombi 1615 Pexelizumab nant—DuPont Pharmaceuticals 1616 pexiganan acetate-Genaera 1677 Plasminogen activators—Abbott Laboratories, 1617 Pharmaprojects No 3179 NYU American Home Products, Boehringer Mannheim, Chi 1618 Pharmaprojects No 3390 Ernest Orlando ron Corporation, DuPont Pharmaceuticals, Eli Lilly, 1619 Pharmaprojects No . 3417 Sumitomo Shionogi, Genentech, Genetics Institute, GlaxoSmith 1620 Pharmaprojects No . 3777 Acambis Kline, HemispherX Biopharma, Merck & Co, Novartis, 1621 Pharmaprojects No 4209 XOMA Pharmacia Corporation, Wakamoto, Yeda 1622 Pharmaprojects No . 4349 Baxter Intl. 1678 plasminogen-related peptides—Bio-Tech. Gen 1623 Pharmaprojects No 4651 eral/MGH 1624 Pharmaprojects No . 4915—Avanir 1679 platelet factor 4 RepliCen 1625 Pharmaprojects No . 5156—Rhizogenics 1680 Platelet-derived growth factor Amgen Zy 1626 Pharmaprojects No 5200 Pfizer moGenetics 1627 Pharmaprojects No . 5215—Origene 1681 Plusonermin Hayashibara 1628 Pharmaprojects No . 5216—Origene 1682 PMD-2850 Protherics 1629 Pharmaprojects No . 5218 Origene 1683 Pneumococcal vaccine—Antex Biologics, Aven 1630 Pharmaprojects No. 5267 ML Laboratories tis Pasteur 1631 Pharmaprojects No . 5373 MorphoSys 1684. Pneumococcal vaccine intranasal BioChem 1632 Pharmaprojects No . 5493- Metabolex Vaccines/Biovector 1633 Pharmaprojects No 5707 Genentech 1685 PR1A3 1634 Pharmaprojects No . 5728—Autogen 1686 PR-39 1635) Pharmaprojects No 5733. BioMarin 1687 prailmorelin—Kaken 1636 Pharmaprojects No 5757. NIH 1688 Pretarget-Lymphoma NeoRX 1637 Pharmaprojects No . 5765. Gryphon 1689 Priliximab Centocor 1638 Pharmaprojects No 5830 AntiCancer 1690 PRO 140 Progenics 1639 Pharmaprojects No . 5839. Dyax 1691 PRO 2000 Procept 1640 Pharmaprojects No .5849—Johnson & Johnson 1692 PRO 367 Progenics 1641 Pharmaprojects No. 5860 Mitsubishi-Tokyo 1693 PRO 542 Progenics 1642 Pharmaprojects No. 5869 Oxford Glyco 1694 pro-Apo A-I Esperion Sciences 1695 Genzyme 1643 Pharmaprojects N .5883–Asahi Brewery 1696 Prosaptide TX14(A) Bio-Tech. General 1644 Pharmaprojects N 5947 StressGen 1697 prostate cancer antbodies—Immunex, UroCor 1645 Pharmaprojects N . 5961 Theratechnologies 1698 prostate cancer antibody therapy—Genentech/ 1646 Pharmaprojects N 5962 NIH UroGenesys. Genotherapeutics 1647 Pharmaprojects N 5966 NIH 1648 Pharmaprojects N . 5994 Pharming 1699 prostate cancer immunotherapeutics—The 1649 Pharmaprojects N . 5995 Pharming PSMA Development Company 1650 Pharmaprojects N 6O23 IMMUCON 1700 prostate cancer vaccine Aventis Pasteur, Zon 1651 Pharmaprojects N . 6063—Cytoclonal agen, Corixa, Dendreon, Jenner Biotherapies. Therion 1652 Pharmaprojects N 6O73 SIDDCO Biologics 1653 Pharmaprojects N . 6115—Genzyme 1701 prostate-specific antigen EntreMed 1654 Pharmaprojects N 6227 NIH 1702 protein A RepliCen 1655 Pharmaprojects N 6230 NIH 1703 protein adhesives—Enzon 1656 Pharmaprojects N 6236 NIH 1704 protein C Baxter Intl. PPL Therapeutics, 1657 Pharmaprojects N 6243 NIH ZymoGenetics 1658 Pharmaprojects N 6244 NIH 1705 protein C activator Gilead Sciences 1659 Pharmaprojects N 6281- Senetek 1706 protein kinase Rantags—NIH 1660 Pharmaprojects N 6365 NIH 1707 protirelin Takeda 1661 Pharmaprojects N 6368 NIH 1708 protocadherin 2 Caprion 1662 Pharmaprojects N 6373 NIH 1709 Pro-urokinase Abbott, Bristol-Myers Squibb, 1663 Pharmaprojects N 6408 Pan Pacific Dainippon, Tosoh Welfide 1664 Pharmaprojects N ... 6410 Athersys 1710 P-selectin glycoprotein ligand-1—Genetics 1665) Pharmaprojects No. 6421—Oxford Glyco Institute Sciences 1711 pseudomonal infections—InterMune 1666 Pharmaprojects N . 6522—Maxygen 1712 Pseudomonas vaccine Cytovax 1667 Pharmaprojects N 6523- Pharis 1713 PSGL-Ig American Home Products 1668) Pharmaprojects N . 6538—Maxygen 1714 PSP-94 Procyon 1669 Pharmaprojects N 6554. APALEXO 1715 PTH 1-34 Nobex 1670 Pharmaprojects N 6560—Ardana 1716. Quilimmune-M Antigenics 1671 Pharmaprojects N . 6562 Bayer 1717 R 744 Roche 1672 Pharmaprojects N 6569 EOS 1718. R101933 1673 Phenoxazine 1719 R125224 Sankyo 1674 Phenylase Ibex 1720 RA therapy Cardion US 2010/0080773 A1 Apr. 1, 2010 39

1721 Rabies vaccine recombinant—Aventis Pasteur, 1778 rMANEI IVAX BioChem Vaccines, Kaketsuken Pharmaceuticals 1779 rmCRP Immtech 1722 RadioTheraCIM YM BioSciences 1780 RN-1001- Renovo 1723. Ramot project No. 1315 Ramot 1781 RN-3- Renovo 1724 Ramot project No. K-734A Ramot 1782 RNAse conjugate—Immunomedics 1725 Ramot project No. K-734B Ramot 1783 RO 631908 Roche 1726 (Anti-VEGF fragment)—Genen 1784) Rotavirus vaccine—Merck tech 1785 RP 431 DuPont Pharmaceuticals 1727 RANK Immunex 1786 RP-128 Resolution 1728 ranpirinase—Alfacell 1787 RPE65 gene therap 1729 ranpirinase-anti-CD22 MAb-Alfacell 1788 RPR 110173 Aventis Pasteur 1730 RANTES inhibitor Milan 1789 RPR 115135 Aventis Pasteur 1731 RAPID drug delivery systems ARIAD 1790 RPR 116258A Aventis Pasteur 1732 rasburicase—Sanofi 1791 rPSGL-Ig American Home Products 1733 rBPI-21, topical XOMA 1792 r-SPC surfactant Byk Gulden 1734 RC 529 Corixa 1793 RSV antibody Medimmune 1735 rCFTR Genzyme Transgenics 1794 Ruplizumab Biogen 1736 RD 621.98 1795 rV-HER-2/neu Therion Biologics 1737 rDnase Genentech 1796 SA 1042 Sankyo 1738 RDP-58 SangStat 1797 sacrosidase Orphan Medical 1739 RecepTox-Fce Keryx 1798 Sant 7 1740. RecepTox-GnRH Keryx, MTR Technologies 1799) —Immunex 1741 RecepTox-MBP Keryx, MTR Technologies 1800 Saruplase-Gruenenthal 1742 recFSH-Akzo Nobel, Organon 1801 Satumomab-Cytogen 1743 REGA 3G12 1802 SB 1–COR Therapeutics 1744 Regavirumab Teijin 1803 SB 207448 GlaxoSmithKline 1745 Connetics Corp 1804 SB 2.08651- GlaxoSmithKline 1746 Renal cancer vaccine Macropharm 1805 SB 240683–GlaxoSmithKline 1747 repifermin Human Genome Sciences 1806 SB 24.9415- GlaxoSmithKline 1748 Respiratory syncytial virus PFP-2 vaccine Wy 1807 SB 2494.17 GlaxoSmithKline eth-Lederle 1808 SB 6 COR Therapeutics 1749 Respiratory syncytial virus vaccine Glaxo 1809 SB RA31012 SmithKline, Pharmacia, Pierre Fabre 1810 SC 56929. Pharmacia 1750 Respiratory syncytial virus vaccine inactivated 1811 SCA binding proteins—Curis, Enzon 1751 Respiratory syncytial virus-parainfluenza virus 1812 sch v(14E1)-ETA Berlex Laboratories, Schering vaccine—Aventis Pasteur, Pharmacia AG, 1752 Reteplase Boehringer Mannheim, Hoffman 1813 LaRoche 1814 1753. Retropep Retroscreen, 1815 SCH 55700 Celltech 1754). RFB4 (dsFv) PE38 1816 Schistosomiasis vaccine—Glaxo Wellcome? 1755 RFI 641–American Home Products Medeva, Brazil 1756. RFTS UAB Research Foundation 1817 SCPF Advanced Tissue Sciences 1757 RG 12986 Aventis Pasteur 1818 scuPA-suPAR complex Hadasit 1758 RG 83852. Aventis Pasteur 1819 SD-9427. Pharmacia 1759 RG-1059 Repligen 1820 SDF-1 Ono 1760 rGCR NIH 1821 SDZ 215918 Novartis 1761 rCLP-1 Restoragen 1822 SDZ 28O125- Novartis 1762 rGRF Restoragen 1823 SDZ 891.04 Novartis 1763 rh Insulin Eli Lilly 1824 SDZ ABL 364 Novartis 1764. RHAMM targeting peptides—Cangene 1825 SDZ MMA 383 Novartis 1765 rHb1.1 Baxter Intl. 1826 Secretin-Ferring, Repligen 1766 rhCC10 Claragen 1827 serine protease inhibs—Pharis 1767 rhCG Serono 1828 sermorelin acetate—Serono 1768 Rheumatoid arthritis gene therapy 1829 SERP-1 Viron 1769 Rheumatoid arthritis vaccine Veterans Affairs 1830 sertenef Dainippon Medical Center 1831 serum albumin, Recombinant human Aventis 1770 rhLH Serono Behring 1771 Ribozyme gene therapy—Genset 1832 serum-derived factor Hadasit 1772 Rickettsial vaccine recombinant 1833 Sevirumab– Novartis 1773 RIGScan CR Neoprobe 1834) SGN 14 Seatle Genetics 1774 RIP-3 Rigel 1835 SGN 15 Seatle Genetics 1775 Rituximab Genentech 1836 SGN 17/19 Seattle Genetics 1776 RK-0202 RxKinetix 1837 SGN 30 Seatle Genetics 1777 RLT peptide Esperion 1838 SGN-10 Seatle Genetics US 2010/0080773 A1 Apr. 1, 2010 40

1839 SGN-11 Seatle Genetics 1890 superoxide dismutase-2—OXIS 1840 SH 306 DuPont Pharmaceuticals 1891 suppressin UAB Research Foundation 1841 Shanvac-B–Shantha 1892 SY-161-P5 ThromboGenics 1842 Shigella flexneri vaccine—Avant, Acambis, 1893. SY-162 ThromboGenics NovaVax 1894 Systemic lupus erythematosus vaccine Med 1843 Shigella Sonnei vaccine Clone/VivoRX 1844 sICAM-1—Boehringer Ingelheim 1895 T cell receptor peptides—Xoma 1845 Silteplase Genzyme 1896 T cell receptor peptide vaccine 1846 SIV vaccine—Endocon, Institut Pasteur 1897 T4N5 liposomes AGI Dermatics 1847 SK 896 Sanwa Kagaku Kenkyusho 1898 TACI, soluble ZymoGenetics 1848 SK-827—Sanwa Kagaku Kenkyusho 1899 targeted apoptosis—Antisoma 1849 Skeletex—CellFactors 1900 tasonermin Boehringer Ingelheim 1850 SKF 106160 GlaxoSmithKline 1901 TASP 1851 S-nitroso-AR545C 1902 TASP-V 1852 SNTP Active Biotech 1903 Tat peptide analogues—NIH 1853 somatomedin-1 GroPep, Mitsubishi-Tokyo, NIH 1904 TBPI Yeda 1854 Somatomedin-1 carrier protein Insmed 1905 TBP II 1855 Somatostatin-Ferring 1906 TBV25H NIH 1856 Somatotropin/Human Growth Hormone—Bio 1907 Tc 99m ior ceal Center of Molecular Immunol Tech. General, Eli Lilly Ogy 1857 somatropin Bio-Tech. General, Alkermes, Pro 1908 Tc 99m P 748 Diatide Lease, Aventis Behring, BioVector, Cangene, Dong-A, 1909 Tc 99m votumumab–Intracell Eli Lilly, Emisphere, Enact, Genentech, Genzyme 1910 Tc-99m rh-Annexin V Theseus Imaging Transgenics, Grandis/InfiMed, CSL, InfiMed, Mac 1911 teceleukin-Biogen roMed, Novartis, Novo Nordisk, Pharmacia Serono, 1912 tenecteplase Genentech TranXenoGen 1913 Armour Pharmaceuticals, Asahi 1858 Somatropin derivative—Schering AG Kasei, Eli Lilly 1859 somatropin, AIR Eli Lilly 1914 terlipressin Ferring 1860 Somatropin, inhaled Eli Lilly/Alkermes 1915 testisin AMRAD 1861 Somatropin, Kabi Pharmacia 1916 Tetrafibricin Roche 1862 somatropin, Orasome Novo Nordisk 1917 TFPI EntreMed 1863 Sonermin-Dainippon Pharmaceutical 1918 tgD-IL-2 Takeda 1864 SP(V5.2)C Supertek 1919. TGF-Alpha ZymoGenetics 1865 SPf66 1920 TGF-R-Kolon 1866 sphingomyelinase-Genzyme 1921 TGF-32. Insmed 1867 SR 29001- Sanofi 1922. TGF-B3 OSI 1868 SR 41476 Sanofi 1923 Thalassaemia gene therapy Crucell 1869 SR-29001- Sanofi 1924. TheraCIM-h-R3–Center of Molecular Immu 1870 SS1(dsFV)-PE38 NeoPharm nology, YM BioSciences 1871 32 microglobulin—AvideX 1925. Theradigm-HBV Epimmune 1872 B2-microglobulin fusion proteins—NIH 1926. Theradigm-HPV Epimmune 1873 B-amyloid peptides CeNeS 1927. Theradigm-malaria Epimmune 1874 B-defensin Pharis 1928. Theradigm-melanoma Epimmune 1875 Staphylococcus aureus infections—InhibiteX/ 1929. TheraFab Antisoma ZLB 1930. ThCRF 1-29. Theratechnologies 1876 Staphylococcus aureus vaccine conjugate—Nabi 1931. ThCRF 1-44. Theratechnologies 1877 Staphylococcus therapy Tripep 1932. Thrombin receptor activating peptide Abbott 1878 Staphylokinase—Biovation, Prothera, Thrombo 1933 thrombomodulin—Iowa, Novocastra genetics 1934 Dragon Pharmaceuticals, 1879 Streptococcal A vaccine M6 Pharmaceuticals, Genentech North American Vaccine 1935 thrombopoietin, Pliva Receptron 1880 Streptococcal B vaccine—Microscience 1936. Thrombospondin 1881 Streptococcal B vaccine recombinant Biochem 1937 thrombostatin Thromgen Vaccines 1938 thymalfasin SciClone 1882 Streptococcus pyogenes vaccine 1939 thymocartin Gedeon Richter 1883 STRL-33 NIH 1940 thymosin Alpha1- NIH 1884 Subalin SRC VB VECTOR 1941 thyroid stimulating hormone—Genzyme 1885 SUIS United Biomedical 1942 tICAM-1- Bayer 1886 SUIS-LHRH United Biomedical 1943. Tick anticoagulant peptide—Merck 1887 SUN-E3001 Suntory 1944 TIF Xoma 1888 super high affinity monoclonal antibodies YM 1945 Tifacogin Chiron, NIS, Pharmacia BioSciences 1946 Tissue factor Genentech 1889 Superoxide dismutase Chiron, Enzon, Ube 1947 Tissue factor pathway inhibitor Industries, Bio-Tech, Yeda 1948. TJN-135 Tsumura US 2010/0080773 A1 Apr. 1, 2010 41

1949 M27 Avant 2009 V 20 GLYCODesign 1950 M29 Avant 2010 V2 vasopressin receptor gene therapy 1951 MC-151—Tanabe Seiyaku 2011 vaccines—Active Biotech 1952 NF tumour necrosis factor Asahi Kasei 2012 Varicella Zoster glycoprotein vaccine Research 1953 NF Alpha CytImmune Corporation Technologies 1954 NF antibody—Johnson & Johnson 2013 Varicella Zoster virus vaccine live Cantab Phar 1955 NF binding protein—Amgen maceuticals 1956 NF degradation product Oncotech 2014 Vascular endothelial growth factor Genentech, 1957 NF receptor Immunex University of California 1958 TNF receptor 1, soluble-Amgen 2015 Vascular endothelial growth factors—R&D Sys 1959 TNF Tumour necrosis factor-alpha Asahi tems Kasei, Genetech, Mochida 2016 vascular targeting agents—Peregrine 1960 TNF-Alpha inhibitor Tripep 2017 Vasopermeation enhancement agents—Peregrine 1961 TNFR:Fc gene therapy Targeted Genetics 2018 vasostatin NIH 1962 TNF-SAM2 2019 VCL. Bio-Tech. General 1963 ToleriMab—Innogenetics 2020 VEGF Genentech, Scios 1964 Toxoplasma gondii vaccine GlaxoSmithKline 2021 VEGF inhibitor Chugai 1965 TP 9201 Telios 2022 VEGF-2 Human Genome Sciences 1966 TP10 Avant 2023 VEGF-Trap Regeneron 1967 TP20 Avant 2024 Viscumin, recombinant—Madaus 1968 tA Centocor 2025 Vitaxin 1969 Scios 2026 Vitrase—ISTA Pharmaceuticals 1970 TRAIL/Apo2L-Immunex 2027 West Nile virus vaccine—Bavarian Nordic 1971) TRAIL-R1 MAb Cambridge Antibody Tech 2028 WP 652 nologies 2029 WT1 vaccine—Corixa 1972 transferrin-binding proteins—CAMR 2030 WX-293 Wilex BioTech. 1973 Transforming growth factor-beta-1—Genentech 2031 WX-360 Wilex BioTech. 1974 transport protein Genesis 2032 WX-UK1- Wilex BioTech. 1975 Trastuzumab Genetech 2033 XMP-5OO XOMA 1976 TRH Ferring 2034 XomaZyme-791- XOMA 1977 Triabin Schering AG 2035 XTL 001—XTL Biopharmaceuticals 1978 Triconal 2036 XTL 002 XTL Biopharmaceuticals 1979 Triflavin 2037 yeast delivery system—Globemmune 1980 troponin I Boston Life Sciences 2038 Yersinia pestis vaccine 1981) TRP-2 NIH 2039 YIGSR-Stealth—Johnson & Johnson 1982 trypsin inhibitor Mochida 2040 Yissum Project No. D-0460 Yissum 1983 TSP-1 gene therapy 2041 YM 207 Yamanouchi 1984 TT-232 2042 YM337 Protein Design Labs 1985 TTS-CD2 Active Biotech 2043) Yttrium-90 labelled biotin 1986 Tuberculosis vaccine—Aventis Pasteur, Genesis 2044 Yttrium-90-labeled anti-CEA MAb T84.66 1987 Tumor Targeted Superantigens Active Bio 2045 ZD 0490 AStraZeneca tech Pharmacia 2046 Ziconotide-Elan 1988 tumour vaccines—PhotoCure 2047 ZK 157138 Berlex Laboratories 1989 tumour-activated prodrug antibody conjugates— 2048 Zolimomab aritox Millennium/ImmunoGen 2049 Zorcell—Immune Response 1990 tumstatin ILEX 2050 ZRXL peptides Novartis 1991 Tuvirumab Novartis 2051. In certain embodiments, a therapeutic agent such as 1992 TV-4710 Teva insulin is associated with a composition of the invention. 1993 TWEAK receptor Immunex Association of insulin with the lipid-based constituents com 1994 TXU-PAP prising a composition of the invention is achieved via com 1995 TY-10721 TOA Eiyo bination of a low molarity solution of insulin with an aqueous 1996 Type I diabetes vaccine Research Corp Suspension of the lipid-based constituents. In this embodi 1997 Typhoid vaccine CVD 908 ment, the number of lipid molecules involved in the assembly 1998 U 143677 Pharmacia of the lipid-based constituents comprising the composition 1999 U 81749 Pharmacia far surpasses the number of molecules of insulin. This high 2000 UA 1248 Arizona lipid to insulin ratio minimizes the molecular interactions 2001 UGIF Sheffield between insulin and the lipids, insuring that the self-assembly 2002 UIC 2 and self-organization process of the lipid-based constituents 2003 UK 101 are not disrupted. This high ratio also facilitates the formation 2004 UK-279276 Corvas Intl of a stable insulin/composition construct. 2005 urodilatin Pharis 2052 Without wishing to be bound by a particular theory, 2006 urofollitrophin Serono it is believed that the quantity of therapeutic agent(s) associ 2007 Urokinase Abbott ated with the composition of the present invention appears to 2008 uteroferrin—Pepgen be a function of loading time, lipid concentration, and buffer US 2010/0080773 A1 Apr. 1, 2010 42 molarity. As the lipid concentration in aqueous media is cent 1,2-distearoyl-sn-glycero-3-phosphocholine, approxi increased, additional therapeutic agents associate with a com mately 22 mole percent dihexadecyl phosphate, and approxi position of the present invention. The time required for load mately 16 mole percent cholesterol. ing the therapeutic agent may be anywhere from several hours 2059 Thyroxine, although orally bioaviable, is not selec to about one week. tive when taken orally. In an embodiment of the invention, 2053. The low concentration of therapeutic agent relative though, thyroxine may associate with the composition of the to the concentration of the composition is unique among lipid invention giving a constituent/thyroxine construct that may particle delivery systems. Typically, liposome or liposome be specifically targeted to the liver, restricting thyroxine's like delivery systems have employed a much larger quantity action to that of lowering blood lipids and cholesterol. Pref of therapeutic agent. The efficacy of this embodiment shows erably, the lipids selected to form the composition for asso that it is possible to utilize less therapeutic agent while still ciating thyroxine include approximately 62 mole percent, obtaining a pharmacologically desirable result in the patient. 1.2-distearoyl-sn-glycero-3-phosphocholine, approximately This embodiment of the invention therefore provides an 22 mole percent dihexadecyl phosphate, approximately 16 advantageous therapeutic option. mole percent cholesterol, and approximately 1 mole percent 2054. In other embodiments the addition of a higher con Biotin DHPE. centration of therapeutic agent may be both desirable and 2060 Blood clotting Factors VII, VIII, IX, and X act in advantageous. The composition of the present invention is either the contact activation (intrinsic), tissue factor (extrin capable of associating with, and tolerating, higher molarity sic), or common pathways for blood clotting. These proteins Solutions of any given therapeutic agent. are not presently orally bioavailable for treatment of diseases 2055. A diagrammatic example of insulin associated with Such as hemophilia. In an embodiment of the present inven a composition of the invention is depicted in FIG. 1. tion, blood clotting factors VII, VIII, IX, and X may associate with a composition of the invention. Preferably the lipids 2056 Serotonin, like insulin, may also be delivered to the selected to form the composition for associating one of fac liver utilizing a composition including an HTM. Serotonin tors VII, VIII, IX, or X include approximately 62 mole per acts jointly with insulin at the level of the liver to activate cent, 1,2-distearoyl-sn-glycero-3-phosphocholine, approxi hepatic glucose storage during a portal (oral) glucose load. In mately 22 mole percent dihexadecyl phosphate, order to achieve the desired effect, serotonin must be deliv ered to the liver. Non-targeted serotonin, introduced via injec approximately 16 mole percent cholesterol, and approxi tion or oral delivery in pharmacologically acceptable doses mately 1 mole percent Biotin DHPE. cannot effectively induce the desired activity. Therefore, an 2061 Although the invention has been described in terms embodiment of the invention includes a composition com of specific therapeutic agents and lipids noted above, any of prising an HTM with associated serotonin. This embodiment the therapeutic agents described herein may associate with a provides a highly desirable delivery mechanism for this composition of the invention, comprising any of the combi important gluco-regulatory hormone. In an embodiment of nation of lipids disclosed herein. the invention designed for the delivery of serotonin, the lipids comprising the composition are approximately 61 mole per Covalent Association of Therapeutic and Diagnostic Agents cent, 1,2-distearoyl-sn-glycero-3-phosphocholine, approxi 2062. In embodiments of the invention, a therapeutic or mately 22 mole percent dihexadecyl phosphate, approxi diagnostic agent is covalently attached to a lipid. Examples of mately 16 mole percent cholesterol and about 1 mole percent lipids to which the therapeutic agents may be attached of a targeting agent. include, for example, cholesterol, thiocholesterol, MPB-PE, 2057 Calcitonin is a hormone that regulates bone metabo MCC-PE, and 1,2-distearoyl-sn-glycero-3-phosphoethano lism. Due to the high prevalence of diseases Such as lamine, and 1,2-dipalmitoyl-sn-glycero-3-phospho-2-mer osteoporosis, an oral formulation of this hormone is highly captoethanol. Examples of therapeutic agents that may be desirable. Presently calcitonin is only deliverable via injec covalently bound to a lipid include, but are not limited to, tion. In an embodiment of the invention designed for the poly-peptides and/or proteins, such as, but not limited to, delivery of calcitonin, the lipids selected to form the compo GLP-1, insulin, calcitonin, interferon, uricase, tissue plasmi sition include approximately 62 mole percent, 1,2-distearoyl nogenactivator, thymoglobin, various vaccines, heparin, hep sn-glycero-3-phosphocholine, approximately 22 mole per arin analogs, antithrombin III, filgrastin, pramilitide acetate, cent dihexadecyl phosphate, and approximately 16 mole , epifibatide, and antivenins, blood clotting factors percent cholesterol. including, but not limited to, Factors VII, VIII, IX, Kallikrein, 2058 GLP-1 is a peptide that acts at both the liver and Kininogen, Hageman Factor (XII), plasma thromboplastin pancreas. In the liver, GLP-1 acts to stimulate glycogen accu antecedent Factor (XI), tissue factor, Stuart Factor (X), acce mulation during a meal. However, prior art administration lerin (V), prothrombin (II), and fibrin stabilizing Factor methods where GLP-1 is administered orally evidence poor (XIII); various Small molecules, such as, for example, D or L bioavailability and reduced efficacy upon oral dosing. In an thyroxine or serotonin, nucleic acids, DNA or RNA embodiment of the present invention, GLP-1 associates with sequences, immunoglobulins, such as, but not limited to, IgG a constituent of a composition of the invention form a consti and IgM, and a variety of monoclonal antibodies. Such as but tutent/GLP-1 construct. The constituent/GLP-1 construct not limited to, rituximab, trastuzumab, and glycolipids that may further include a targeting agent. Preferably, the lipid act as therapeutic agents, and in addition, other larger pro components selected to form the constituents of the compo teins, such as, for example, human growth hormone sition including GLP-1 include approximately 62 mole per (“HGH), erythropoietin, and parathyroid hormone. Various US 2010/0080773 A1 Apr. 1, 2010

other therapeutic agents have been described elsewhere 2067. Additional linker precursors that may be used herein. Each of these therapeutic agents may likewise include compounds according to formula I: covalently associate with a composition of the invention. 2063. Examples of diagnostic agents that may be covalently bound to a lipid include diagnostic contrast agents such as, but not limited to, gold, TEMPO (2-diacyl-sn-glyc erol-3-phospho-TEMPO-choline), Fe" oxide, Fe" oxide, and gadolinium. Other diagnostic agents include radioactive materials such as radioactive isotopes of common atoms including, but not limited to, 'C, Ge, F, and 'I. These contrast and radioactive agents may be covalently attached to a lipid or to the optionally present targeting agent. Alterna wherein 'A' corresponds to tively, and where chemically appropriate, the diagnostic agent may be bound to a ligand Such as DADO (2'-deoxyad enosine), which is itself covalently attached to a lipid or the Gl Gl optional targeting agent. Alternatively, diagnostic agents, O O such as those described above, may be covalently linked to an O antibody or small molecule. These antibodies or small mol N N NH ecules may then associate with a composition of the invention s’, --- for subsequent oral delivery. O O 2064. In one embodiment, a therapeutic or diagnostic agent may be directly attached to a lipid. In this embodiment, or NH-NH : “J” corresponds to (CH), or a free carboxylate or aldehyde on a therapeutic agent is con densed with a lipid bearing an amine using known proce dures. Alternatively, the carboxylate may forman ester with a lipid bearing a free alcohol using known esterification proce dures. In an alternative embodiment, a free thiol on a thera peutic agent may form a disulfide linkage with a lipid also presenting a free thiol. 2065. More typically, however, a therapeutic agent is attached to a given lipid via a linker. As an example, a thera peutic agent may be attached to a lipid as follows: (therapeu and G' is either Hor SO-Na. Subscript “a” is independently, tic agent)-N-C(O)(CH), S-lipid. In this embodiment, the at each occurrence, 1, 2, 3, 4, 5, 6, 7, or 8. Common examples linker is —C(O)(CH), S -. This linker is derived from reac of linker precurors according to formula I include, but are not tion of a Succinimidyl based linker precursor, Succinimidyl limited to, N-succinimidyl-3-(2-pyridyldithio)proprionate O—C(O)(CH),SR. Preferably, n is an integer between 1 and (“SPDP). Succinimidyl 6-(3-2-pyridyldithio-propiona 10. Even more preferably, n is 1, 2, or 3. R is typically a mido)hexanoate (“LC-SPDP), Sulfosuccinimidyl 6-(3'-2- protecting group Such as —C(O)CH. Other appropriate thiol pyridyldithio-propionamido)hexanoate (“Sulfo-LC protecting groups may be found in Green's Protective Groups SPDP), 4-Succinimidyloxycarbonyl-methyl-a-2- in Organic Synthesis, Wuts, et al, 4" edition, 2007. pyridyldithiotoluene (“SMPT), 4-Sulfosuccinimidyl-6- 2066 Generally speaking, the linker precursor reacts with methyl-a-(2-pyridyldithio)toluamidohexanoate) ("Sulfo a nucleophilic amine, alcohol, or thiol present on the thera LC-SMPT), and 3-(2-pyridyldithio)propionyl hydrazide peutic agent, displacing N-hydroxysuccinimide, to form an (“PDPH), each of which are known and described in the amide, ester, or thioester. Preferably, the nucleophile is a literature. 2068. When a compound of formula I is used (and 'A' is primary amine. After the linker is bound to the therapeutic not NH-NH) a free nitrogen on atherapeutic agent reacts with agent, the protecting group, R, is removed from the linker to the compound of formula I to forman amide bond by displac reveal a thiol. Preferably, the protecting group is removed ing N-hydroxysuccinimide or a related derivative. Subse under conditions that do not perturb the now attached thera quently, the disulfide bridge present in the linker precursor is peutic agent. This thiol may then undergo a Michael reaction reduced under mild conditions using tris(2-carboxyethyl) with a lipid such as MPB-PE or MCC-PE. Preferably, lipids phosphine (TCEP) or other known reducing agents. The MPB-PE and/or MCC-PE are already incorporated into a resulting free thiol can then react with a lipid such as MPB-PE composition of the invention, however, the Michael reaction or MCC-PE, either before or after the lipid is incorporated may take place pior to incorporating these lipids into a com into a composition of the invention. Preferably, the resulting position of the invention. The order of reactions will depend free thiol is reacted with the lipid after the lipid has been upon the therapeutic agent's ability to tolerate microfluidiza incorporated into a composition of the invention. tion, aqueous environments, and elevated temperatures. In the 2069. Alternatively, a compound of formula I may react case of complex proteins which may denature at high tem with a nucleophile Such as 1,2-distearoyl-sn-glycero-3-phos peratures, it is preferable to perform the Michael reaction phethanolamine, or related derivative, to displace Succinim after MPB-PE and/or MCC-PE have been incorporated into a ide. The disulfide in the resulting product may then be composition of the invention. reduced using TCEP or other mild reductant to provide a free US 2010/0080773 A1 Apr. 1, 2010 44 thiol. The resulting thiol compound may then be oxidatively 2074 Common examples of linker precursors according coupled to a free thiol in a therapeutic agent. Preferably, the to formula II include, but are not limited to, Succinimidyl resulting free thiol is reacted with the therapeutic agent after 4-N-maleimidomethylcyclohexane-1-carboxylate the lipid has been incorporated into a composition of the (“SMCC), Sulfosuccinimidyl 4-N-maleimidomethylcy invention, however it need not be, depending upon the stabil clohexane-1-carboxylate ("Sulfo-SMCC), m-Maleimido ity of the therapeutic agent. benzoyl-N-hydroxysuccinimide ester (“MBS”), m-Maleimi 2070. When A is NH-NH , the nucleophilic nitrogen of dobenzoyl-N-hydroxysulfosuccinimide ester (“Sulfo the hydrazide reacts with a ketone, aldehyde, activated ester, MBS”), N-Succinimidyl-4-iodoacetylaminobenzoate a carboxylic acid, or leaving group on a therapeutic agent to (“SIAB), N-Sulfosuccinimidyl 4-iodoacetylaminoben form a therapeutic agent/linker conjugate. When reacting Zoate ("Sulfo-SIAB), succinimidyl-4-(((iodoacetyl)amino) with an aldehyde or ketone, the reaction is typically a reduc methyl)cyclohexane-1-carboxylate (“SIAC), succinimidyl tive amination, but may be a simple condensation without 4-p-maleimidophenylbutyrate (“SMPB), sulfosuccinim concomitant reduction, resulting in the formation of an enam idyl 4-p-maleimidophenylbutyrate ("Sulfo-SMPB), and ine. When the hydrazide reacts with a carboxylic acid to form Succinimidyl-6-((((4-(iodoacetyl)amino)methyl)cyclohex a hydrazone, the reaction is mediated by a crosslinking ane-1-carbonyl)amino)-hexanoate (“SIACX'), each of reagent, Such as EDC, EDCI, or other crosslinking reagent which are known and described in the literature. 2075. When a compound of formula II is used, a free now known or hereafter developed. nitrogen on a therapeutic agent reacts with the compound of 2071 As above, the disulfide bridge is then reduced under formula II to form an amide bond by displacing N-hydrox mild conditions. The resulting free thiol can then react with a ySuccinimide or Sulfo-N-hydroxySuccinimide. The resulting lipid such as MPB-PE or MCC-PE, either before or after the therapeutic agent/linker conjugate is then preferably reacted lipid is incorporated into a composition of the invention. with a composition of the invention containing a lipid bearing Preferably, the resulting free thiol is reacted with the lipid a free thiol (such as, for example, thiocholesterol or 1.2- after the lipid has been incorporated into the composition. dipalmitoyl-sn-glycero-3-phospho-2-mercaptoethanol). The 2072. In another embodiment, the linker precursor may be free thiol undergoes a Michael reaction into the double bond a compound according to formula II of a maleimide group, or displaces I in a displacement reac tion. Although it is preferred that the therapeutic agent/linker conjugate is reacted with a lipid presenting a free thiol that has II already been incorporated into a composition of the inven Gl tion, the therapeutic agent/linker conjugate may be reacted O O with a lipid presenting a free thiol prior to the lipid being incorporated into a composition of the invention. N 1. 2076. In another embodiment, the linker precursor may be No1 a compound according to formula III O

III

O No1 N-1 G4 O wherein G' is either Hor SONa, G' is maleimidyl, HNC (O)CHI, or NHC(O)(CH)NHC(O)CHI and “a” is, inde pendently at each occurrence, 1,2,3,4, 5, 6, 7, or 8. A double dashed bond connected to an oxygen indicates that a given HNC(O)CHI, CH-NHC(O)(CH)NHC(O)CHI, carbon is optionally a carbonyl. Thus, in formula III, the —CHHNC(O)CHI; "Q" is optional and, when present, is double dashed bond connected to the noted carbon indicates —C(O)(CH)NH ; “K” is optional, and when present, is that the bond connectivity at that carbon is —C(O)— or —(CH) ; and “a” as used in formula II, “Q', or “K” is —CH2—, Common examples of linker precursors according independently, at each occurrence 1,2,3,4,5,6,7, or 8. When to formula III include, but are not limited to, N-g-maleimi 'A' is not present, the oxygen of the N-hydroxysuccinimidyl dobutyryloxysuccinimide ester (“GMBS”), N-g-maleimi group is bound directly to the carbon of the carbonyl adjacent dobutyryloxysulfosuccinimide ester ("Sulfo-GMBS”), suc to A. cinimidyl-6-((iodoacetyl)amino)hexanoate (“SIAX'), and 2073 Informula II, the bond notation “trindicates that Succinimidyl-6-(6-(((iodoacetyl)amino)hexanoyl)amino) the bond may be a single or a double bond. Preferably, when hexanoate (“SIAXX'), each of which are known and one bond according to the above described notation repre described in the literature. sents a double bond, all bonds according to that notation 2077. When a compound of formula III is used, a free represent double bonds. Similarly, if any bond according to nitrogen on a therapeutic agent reacts with the compound of the above described notation represents a single bond, it is formula III to form an amide bond by displacing N-hydrox preferred that all bonds according to that notation represent a ySuccinimide or Sulfo-N-hydroxySuccinimide. The resulting single bond. therapeutic agent/linker conjugate is then preferably reacted US 2010/0080773 A1 Apr. 1, 2010 with a composition of the invention containing a lipid bearing sents a single bond, it is preferred that all bonds according to a free thiol (such as, for example, thiocholesterol or 1.2- that notation represent a single bond. dipalmitoyl-sn-glycero-3-phospho-2-mercaptoethanol). The 2080 When a compound of formula V is used, the nucleo free thiol undergoes a Michael reaction into the double bond philic nitrogen of the hydrazide reacts with a ketone, alde of a maleimide group, or displaces I in a displacement reac hyde, activated ester, a carboxylic acid, or leaving group on a tion. Although it is preferred that the therapeutic agent/linker therapeutic agent to form a therapeutic agent/linker conju conjugate is reacted with a lipid presenting a free thiol that has gate. When reacting with an aldehyde or ketone, the reaction already been incorporated into a composition of the inven is typically a reductive amination. The reaction may, however, tion, the therapeutic agent/linker conjugate may be reacted be a simple condensation without concomitant reduction, with a lipid presenting a free thiol prior to the lipid being resulting in the formation of an enamine. When the hydrazide incorporation into a composition of the invention. is reacted with a carboxylic acid, the reaction is mediated by 2078. In another embodiment, the linker precursor may be a crosslinking reagent, Such as EDC (1-ethyl-3.3-dimethy compounds according to formula IV laminopropylcarbodiimide), EDCI (1-(3-dimethylaminopro pyl)-3-ethylcarbodiimide), or other crosslinking reagent now IV known or hereafter developed. 2081. The resulting therapeutic agent/linker conjugate is then preferably reacted with a composition of the invention containing a lipid bearing a free thiol (such as, for example, thiocholesterol or 1,2-dipalmitoyl-sn-glycero-3-phospho-2- mercaptoethanol). The free thiol undergoes a Michael reac tion into the double bond of the maleimide portion of the When a compound of formula IV is used, a free nitrogen on a conjugate. Although it is preferred that the therapeutic agent/ therapeutic agent reacts with the compound of formula IV to linker conjugate is reacted with a lipid presenting a free thiol form an amide bond by displacing the p-nitrophenyl group. that has already been incorporated into a composition of the The resulting therapeutic agent/linker conjugate is then pref invention, the therapeutic agent/linker conjugate may be erably reacted with a composition of the invention containing reacted with a lipid presenting a free thiol prior to the lipid a lipid bearing a free thiol (such as, for example, thiocholes being incorporated into a composition of the invention. terol or 1,2-dipalmitoyl-sn-glycero-3-phospho-2-mercapto 2082 In a further embodiment, the linker precursor may ethanol). The free thiol displaces I in a displacement reac be a compound according to formula VI: tion. Although it is preferred that the therapeutic agent/linker conjugate is reacted with a lipid presenting a free thiol that has already been incorporated into a composition of the inven tion, the therapeutic agent/linker conjugate may be reacted VI with a lipid presenting a free thiol prior to the lipid being incorporated into a composition of the invention. 2079. In a further embodiment, the linker precursor is a compound of formula V.

O wherein G is selected from the group consisting of C(O) O G7, C(O)NHNH, C(O)C(O)H, C(O)NH(CH)NH, C(O)NH(CH.)NHC(O)CHI, C(O)NH(CH),C(O) GE) X-z Z-N G7, NO, -(CH)NHC(O)G", NH(CH),C(O)C7, HN-NH —(CH)SSC(O)G7, C(O)NH(CH),SS(CH),C(O)C7. X O —(CH),C(O)C7, and –C(O)NH(CH)NHC(O)(CH) O G”; “a” is independently at each occurrence 1,2,3,4,5,6,7, or 8; and G' is selected from the group consisting —OH, In formula V. “Z” is independently optional at each occur - NO, -H, and –C(O)G7. G7 is rence, and when present is (CH2). Subscript 'a' is indepen dently, at each occurrence, 1, 2, 3, 4, 5, 6, 7, or 8. Although structure V is shown as the salt, compounds of formula V may be either a salt or a free base. Examples of linker precursors according to formula V include, but are not limited to, 4-(4- N-Maleimidophenyl)butyric acid hydrazide hydrochloride (“MBPH) and 4-(N-maleimidophenyl)cyclohexane-1-car bonyl-hydrazidehydrochloride (“MCH). Informula V, the bond notation"-r'indicates that the bond may be a single or a double bond. Preferably, when one bond according to the above described notation represents a double bond, all bonds wherein G' is either H or -SONa; provided that G' is according to that notation represent double bonds. Similarly, —C(O)C7 only when G is NO and that G is NO only if any bond according to the above described notation repre when G is C(O)G7. G’ is US 2010/0080773 A1 Apr. 1, 2010 46

reaction may, however, be a simple condensation without concomitant reduction, resulting in the formation of an enam ine. When the nucleophilic nitrogen reacts with a carboxylic acid, the reaction is mediated by a crosslinking reagent, Such as EDC, EDCI, or other crosslinking reagent now known or 21 ry hereafter developed. 2088. The resulting therapeutic agent/linker conjugate is 2083 Examples of linkers according formula VI include, then preferably irradiated with UV in the presence of a but are not limited to, N-Hydroxysuccinimidyl-4-azidosali Substantial excess of a lipid, as described above, to form a cylic acid (“NHS-ASA'), N-hydroxysulfosuccinimidyl-4- therapeutic agent/linker/lipid conjugate. This conjugate can azidosalicylic acid ("Sulfo-NHS-ASA'), sulfosuccinimidyl then be incorporated into a composition. In an alternative 4-azidosalicylamido-hexanoate ("Sulfo-NHS-LC-ASA), procedure, the therapeutic agent/linker conjugate may be irra N-hydroxysuccinimidyl-4-azidobenzoate (“HSAB), N-hy diated in the presence of composition of the invention. droxysulfosuccinimidyl-4-azidobenzoate ("Sulfo-HSAB), N-5-azido-2-nitrobenzoyloxysuccinimide (ANB-NOS), 2089. In yet another embodiment, the linker precursor of N-Succinimidyl-6-(4'-azido-2'-nitrophenylamino)hexanoate formula VI may be irradiated in the presence of a lipid or a (“SANPAH), N-sulfosuccinimidyl-6-(4-azido-2'-nitrophe composition of the invention prior to reaction with a thera nylamino)hexanoate ("Sulfo-SANPAH), N-succinimidyl peutic agent. This process results in the formation of a lipid/ (4-azidophenyl)-1,3'-dithioproprionate (“SADP), N-Sulfo linker conjugate or a composition/linker conjugate. The lipid/ succinimidyl(4-azidophenyl)-1,3'-dithioproprionate ("Sulfo linker conjugate is Subsequently incorporated into a SADP), sulfosuccinimidyl-2-(p-azidosalicylamido)-ethyl composition of the invention according to the procedures set 1,3'-dithiopropionate (“SASD), 1-(p-azidosalicylamido)-4- forth elsewhere herein. The composition/linker conjugate (iodoacetamido)butane (ASIB), N-4-(p- may then be reacted with a therapeutic agent with a nucleo azidosalicylamido)butyl-3'-(2-pyridyldithio)propionamide philic nitrogen according to the displacement chemistry (“APDP), p-azidobenzoyl hydrazide (ABH'), 4-p-azi dosalicylamidobutylamine (ASBA), p-azidophenyl gly described previously. oxal (APG'), and sulfosuccinimidyl-4-(p-azidophenyl)bu 2090. In an alternative embodiment, a linker precursor tyrate ("Sulfo-SAPB), each of which are known and according to formula VI may be irradiated and reacted with a described in the literature. therapeutic agent to form a therapeutic agent/linker conju 2084 Linker precursors according to formula VI may be gate. When the conjugate contains a group according to G'. used in various ways. In an example of a first method of the conjugate may then be reacted with a compound Such as attachment wherein G or G' is a group containing G', a free 1.2-distearoyl-sn-glycero-3-phosphoethanolamine, wherein nitrogen on antherapeutic agent reacts with the linker pre the free nitrogen of the ethanolamine reacts with the activated cursor giving atherapeutic agent/linker conjugate by displac hydroxy succinimidyl ester of G'. If the conjugate contains a ing N-hydroxySuccinimide or Sulfo-N-hydroxysuccinimide. “CHI''' functionality, the conjugate may be reacted with a The resulting conjugate is then irradiated with UV light in the presence of a substantial excess of a lipid. The UV light lipid such as thiocholesterol. If the conjugate contains a dis induces nitrene formation. This nitrene Subsequently reacts ulfide, this disulfide may be selectively reduced, whereupon with the lipid in a non-selective manner to formatherapeutic the resulting free thiol bound to the conjugate may react with agent/linker/lipid conjugate. This conjugate can then be a compound such as MPB-PE or MCC-PE. Preferably, the incorporated into a composition of the invention. lipids used to bind the therapeutic agent/linker conjugate have 2085. In an alternative process, a therapeutic agent/linker already been incorporated into a composition of the inven conjugate may be irradiated with UV light in the presence of tion. a composition of the invention. The UV light induces nitrene 2091. In any of the above described procedures, the order formation. This nitrene can then react with any lipid present in the composition. of reactions and the choice of coupling partner will be deter 2086. In another embodiment, the linker precursor accord mined by the stability of the therapeutic agent under a par ing to formula VI may be irradiated in the presence of a lipid ticular set of reaction conditions. It is within the skill of the or a composition of the invention prior to reaction with a ordinarily skilled artisan to determine the appropriate order of therapeutic agent. This process results in the formation of a reactions to arrive at the desired bond connectivity. lipid/linker conjugate or a composition/linker conjugate. The 2092. In a further embodiment, the linker precursor may lipid/linker conjugate is Subsequently incorporated into a be a compound according to formula VII or VIII: composition of the invention according to the procedures set forth elsewhere herein. The composition/linker conjugate may then be reacted with a therapeutic agent presenting a VII nucleophilic nitrogen according to the displacement chemis Gl try described previously herein. 2087. When G’ in formula VI is a group containing a O nucleophilic nitrogen, this nucleophilic nitrogen may react O --> NH with a ketone, aldehyde, activated ester, a carboxylic acid, or N S 1. leaving group on a therapeutic agent to form a therapeutic s--- O21 NC8 agent/linker conjugate. When reacting with an aldehyde or O ketone, the reaction is typically a reductive amination. The US 2010/0080773 A1 Apr. 1, 2010 47

2094. In a further embodiment, the linker precursor may -continued be a compound according to formula X: VIII O O Gl G8 N-O

O wherein G' is either Hor SONa and G is selected from the group consisting of 2-nitrophenyl-5-azido and wherein G' is selected from the group consisting of C(O)C (N)H and C(N)CF. When a linker precursor according to formula X is used, the linker precursor is first reacted with a therapeutic agent containing a free primary amine in order displace p-nitrophenol. This results in a therapeutic agent/ linker conjugate. Subsequently, the conjugate is irridated to form a carbene. When G' is C(O)C(N)H, the conjugate is irradiated in the presence of a compound containing a nucleo philic amine. Such as, for example, 1,2-distearoyl-sn-glycero Subscript 'a' is independently, at each occurrence, 1, 2, 3, 4, 3-phosphoethanolamine. When G' is C(N)CF, the conju 5, 6, 7, or 8. Examples of compounds according to formula gate is irradiated in the presence of lipid or a composition of VII and VIII include, but are not limited to, sulfosuccinim idyl-2-(m-azido-o-nitrobenzamido)-ethyl-1,3'-proprionate the invention. (“SAND'''), sulfosuccinimidyl 2-7-amino-4-methylcou 2095 Alternatively, the compound according to Formula marin-3-acetamido)ethyl-1,3'dithiopropionate (“SAED), X may first be reacted with a lipid such as 1,2-distearoyl-sn and Sulfo-Succinimidyl 7-azido-4-methylcoumarin-3-acetate glycero-3-phosphoethanolamine to displace p-nitrophenol ("Sulfo-SAMCA'). Linker precursors according to formula and then irridated to form a carbene. When G' is C(O)C(N) VII and VIII may be utilized in substantially the same ways as H, the conjugate is irradiated in the presence of a therapeutic described with respect to linker precursors of formula VI. agent containing a nucleophilic amine. When G' is C(N) 2093. In a further embodiment, the linker precursor may CF, the conjugate is irradiated in the presence of a therapeu be a compound according to formula IX: tic agent. 2096 As with other reactions described herein, the order of reactions and the choice of coupling partner will be deter IX mined by the stability of the therapeutic agent under a par ticular set of reaction conditions. It is within the skill of the ordinarily skilled artisan to determine the appropriate order of reactions to arrive at the desired bond connectivity. G10 RES Masking and Avoidance 2097. In addition to an optional targeting molecule, the composition of the invention may further include a reticu loendothelial sytem (RES) avoidance molecule. The RES wherein G' is selected from the group consisting of maleim avoidance molecule gives the composition a longer halflife in idyl and NC(O)CHI. Examples of linker precursors accord the systemic circulation by shielding the composition from ing to formula IX include, but are not limited to, benzophe none-4-iodoacetamide and benzophenone-4-maleimide. macrophage detection. When using a linker precursor of formula IX, the free thiol of 2098 RES avoidance molecules may be incorporated into thiocholesterol displaces I in a displacement reaction to a composition of the invention by binding to a lipid compris form a linker/lipid conjugate. Subsequently, the linker/lipid ing the composition of the invention. For example, U.S. Pat. conjugate is irradiated with UV light in the presence of a No. 6, 177,099 describes a process wherein B-methoxy therapeutic agent to formatherapeutic agent/linker/lipid con neuraminic acid was modified to contain a free thiol that was jugate. This compound may then be incorporated into a com subsequently reacted with MPB-PE via a Michael reaction, as position of the invention. shown in Scheme 1.

US 2010/0080773 A1 Apr. 1, 2010

-continued -continued AcO O OAc OH OH COH HO 1,2-distearoyl-sn-glycero-3- phosphoethanolamine H OH NaCNBH N O OH O He

HO O 5-glycolamido-8,9-di-O-acetyl-3,5-dideoxy-D-glycerol-D-galacto-2- nonulopyranose-1-onic acid (“8-9-di-O-acetyl-N-glycolylneuraminic acid) OH

HO \ OH O OH COH H N O OH

HO O 5-glycolamido-8-O-methyl-3,5-dideoxy-D-glycerol-D-galacto-2- nonulopyranose-1-onic acid (“N-glycolyl-8-O-methylneuraminic acid')

2101. In an alternative embodiment, an N-acyl neuraminic acid derivative may be reacted with a phosgene equivalent 5-glycolamido-8-O-sulfo-3,5-dideoxy-D-glycerol-D-galacto-2- such as N,N'-disuccinimidylcarbonate (DSC). In this nonulopyranose-1-onic acid (“N-glycolyl-8-O-sulfoneuraminic acid') embodiment, an alcohol on the neuraminic acid derivative C-2-8-poly-N-acetyl-neuraminic acid (colominic acid) and reacts with DSC to produce an intermediate containing an derivatives thereof activated carbonyl. This intermediate can then be reacted with a lipid presenting a free primary or secondary amine. A non limiting example of an amine bearing lipid is 1,2-distearoyl Although not shown, further examples of neuraminic acid sn-glycero-3-phosphoethanolamine. derivatives include those in which the nitrogen is not acylated. 2102. In a further embodiment, cholesterol may react with 2100. The above described neuraminic acid derivatives DSC to form intermediate that may be subsequently reacted may be linked to a lipid of the invention via various method with neuraminic acid derivative presenting a free amine. 2103. In still another embodiment, a neuraminic acid ologies. In one embodiment, an N-acyl neuraminic acid derivative presenting a free amine may be condensed with derivative containing a 1.2 diol functionality may be cleaved formaldehyde to generate an iminium, which may be to an aldehyde using NaIO under known conditions. The quenched by nucleophilic attack at the formaldehyde carbon resulting aldehyde may then undergo reductive amination with an appropriate nucleophile. Appropriate nucleophiles with the primary amine of a lipid such as 1,2-distearoyl-sn include primary and secondary amines, an example of which glycero-3-phosphoethanolamine according to known proce includes, but is not limited to, 1,2-distearoyl-sn-glycero-3- dures. An example of this chemistry is shown in Scheme 2. phosphoethanolamine. 2104 As discussed elsewhere herein, RES masking agents may also be associated with a composition of the Scheme 2 invention via non-covalent interactions. In the non-covalent O embodiment, up to about 10 mole percent of the composition may comprise one or more RES masking agents. OH HO O Stability us O OH NaIO4,() 1 mM 2105. Although a composition of the invention is formu N OH Oo C. lated in aqueous media, the composition does not exhibit long H term stability in water. Specifically, water aids hydrolysis of any acyl chains present in any of the lipids present in the HO composition. The aqueous environment also allows for the OH ready oxidation of any unsaturated acyl chains present in any of these lipids. In a preferred embodiment of the present US 2010/0080773 A1 Apr. 1, 2010

invention, the composition of the present invention may be hereinafter “composition’ for oral administration may be protected for long term storage via interaction with a pro prepared, packaged, or sold in the form of a discrete solid teoglycan Such as a modified collagen, known generically as dose unit including, but not limited to, a tablet, a hard or soft dry granulated gelatin. Dry granulated gelatin, when con capsule, a cachet, a troche, or a lozenge, each containing a tacted with an aqueous Suspension of a composition of the predetermined amount of the active ingredient. Other formu invention, reacts with the water, and stabilizes the composi lations suitable for oral administration include, but are not tion. limited to, a powdered or granular formulation, aqueous Sus 2106 The reaction of dried granulated gelatin with an aqueous Suspension of a composition of the present invention pensions, or emulsions. results in a semi-solid colloidal gel that shields the composi 2112 A tablet comprising the composition of the present tion from direct interaction with water. Any water not asso invention, for example, be made by compressing or molding ciated with gelatin is slowly evaporated via refrigerated Stor the composition optionally with one or more additional ingre age at about 2 to about 8°C. The water may, however, be dients. Compressed tablets may be prepared by compressing, removed via techniques including, but not limited to, freeze in a suitable device, the composition in a free-flowing form drying and spray drying. Such as a powder or granular preparation, optionally mixed 2107 This results in a pellet like “dry” composition/gela with one or more of a binder, a lubricant, an excipient, a tin complex. In the complex, the composition of the invention Surface active agent, and a dispersing agent. Molded tablets is partially dehydrated in a reversible manner and sequestered may be made by molding, in a Suitable device, the composi by the proteinaceous lattice of dry gelatin. This sequestration tion, a pharmaceutically acceptable carrier, and at least Suf is enabled by structured water, structured lipid and structured ficient liquid to moisten the mixture. gelatin all interacting through hydrogen bonding, ionic bond 2113 Pharmaceutically acceptable excipients used in the ing, van der Waal's interactions, and hydrophobic bonding manufacture of tablets include, but are not limited to, inert between the lipids, water, and protein structures, such as, for diluents, granulating and disintegrating agents, binding example, insulin. This evidences that gelatin is not acting as agents, and lubricating agents. Known dispersing agents an emulsifying or Suspending agent. As a result, the “dry” include, but are not limited to, potato starchand Sodium starch pellet is stable for long term storage because the activity of glycollate. Known Surface active agents include, but are not water has been mitigated. These pellets can be further pro limited to, Sodium lauryl Sulphate. Known diluents include, cessed to a granulated or free-flowing powder for final cap but are not limited to, calcium carbonate, Sodium carbonate, Sule filling or tabletting, while maintaining their stability. lactose, microcrystalline cellulose, calcium phosphate, cal 2108. Upon oral administration to a patient, the “dry” cium hydrogen phosphate, and sodium phosphate. Known pellet becomes hydrated and once again assumes a semi-solid granulating and disintegrating agents include, but are not colloidal gel state. Upon further exposure to the gastric envi limited to, corn starchandalginic acid. Known binding agents ronment, the gel becomes liquid as gelatin is solubilized. include, but are not limited to, gelatin, acacia, pre-gelatinized Once the gelatin is completely solubilized, the composition of maize starch, polyvinylpyrrolidone, and hydroxypropyl the invention rehydrates, resulting in the formation of a new methylcellulose. Known lubricating agents include, but are Suspension within the gastric environment. The reconstituted not limited to, magnesium Stearate, Stearic acid, silica, and composition may then be absorbed into the portal blood flow. talc. 2109. It is important to realize that the role of gelatin in this 2114 Tablets may be non-coated or they may be coated aspect of the invention is as an active stabilizer of the com using known methods to achieve delayed disintegration in the position and not an inert filler as is commonly found in oral gastrointestinal tract of a Subject, thereby providing Sustained formulations of many other pharmaceutical compositions. release and absorption of the composition. By way of That said, the additional use of gelatin as an inert filler in example, a material Such as glyceryl monostearate or glyceryl addition to the aforementioned use is also contemplated. distearate may be used to coat tablets. Further by way of 2110 Although gelatin is used in a preferred embodiment example, tablets may be coated using methods described in of the invention, other gelatin like compounds may be used as U.S. Pat. Nos. 4,256,108; 4,160,452; and 4,265,874 to form well. Examples of agents that will act as active stabilizers osmotically-controlled release tablets. Tablets may further include, but are not limited to, acacia (gum arabic), agar comprise a Sweetening agent, a flavoring agent, a coloring (agar-agar, vegetable gelatin: gelosa; Chinese or Japanese agent, a preservative, or some combination of these in order to gelatin), alginic acid, Sodium alginate (alginic acid; sodium provide pharmaceutically elegant and palatable preparation. salt; algin, Manucol, Norgine; Kelgin), carbomer (carboxy polymethylene), carrageenan, carboxymethylcellulose 2115 Hard capsules comprising the composition may be sodium (carbose D; carboxymethocel S. CMC; cellulose made using a physiologically degradable composition, Such gum), powdered cellulose (Degussa), hydroxyethyl cellulose as gelatin. Such hard capsules comprise the active ingredient, (cellulose: 2-hydroxyethyl ether; Cellosize; Natrosol), and may further comprise additional ingredients including, hydroxypropyl cellulose (cellulose: 2-hydroxypropyl ether; for example, an inert Solid diluent such as calcium carbonate, Klucel), hydroxypropyl methylcellulose (cellulose: 2-hy calcium phosphate, kaolin or cellulose acetate hydrogen droxypropyl methyl ether), methycellulose (cellulose; phthalate. methyl ether Methocel), povidone(2-pyrrolidinone: 1-ethe 2116 Soft gelatin capsules comprising the composition nyl-, homopolymer, polyvinylpyrrolidone), tragacanth (gum may be made using a physiologically degradable composi tragacanth; Hog Gum; Goat's Thorn), and Xanthan gum tion, Such as gelatin. (Keltrol). Like gelatin, and where appropriate, these com 2117 Liquid formulations of the composition which are pounds may also be used as inert fillers. Suitable for oral administration may be prepared, packaged, and sold either in liquid form or in the form of a dry product Formulations intended for reconstitution with water or another suitable 2111. A formulation of a composition of the invention and vehicle prior to use, subject to the stability limitations dis therapeutic agent (with or without the targeting agent)— closed earlier. US 2010/0080773 A1 Apr. 1, 2010 52

2118 Liquid Suspensions may be prepared using conven therapeutic agent with the composition of the invention, and tional methods to achieve Suspension of the constituents in an treat a disease or condition susceptible to treatment with the aqueous vehicle. Aqueous vehicles include, for example, therapeutic agent. water and isotonic saline. Oily vehicles may only be used to the extent that such solvents are not incompatible with the Kits constituents of the composition of the present invention. To the extent that an oily suspension is not incompatible with the 2126. The invention also includes a comprising a com constituents of the composition of the present invention, an position of the invention and an instructional material which oily Suspension may further comprise a thickening agent. describes administering the composition to a mammal. As used herein, an “instructional material” includes a publica 2119 Liquid Suspensions may further comprise one or tion, a recording, a diagram, or any other medium of expres more additional ingredients to the extent that said ingredients sion which can be used to communicate the usefulness of the do not disrupt the structures of the constituents of the com position of the invention. Examples of additional ingredients composition of the invention in the kit for effecting allevia include, but are not limited to, Suspending agents, dispersing tion of the various diseases or disorders recited herein. or wetting agents, emulsifying agents, demulcents, preserva 2127 Optionally, or alternatively, the instructional mate tives, buffers, salts, flavorings, coloring agents, and Sweeten rial may describe one or more methods of alleviating the ing agents. diseases or disorders in a cell or a tissue of a mammal. The instructional material of the kit may, for example, be affixed 2120 Known Suspending agents include, but are not lim to a container which contains the invention or be shipped ited to, Sorbitol syrup, sodium alginate, polyvinylpyrroli together with a container which contains the invention. Alter done, gum tragacanth, gum acacia, and cellulose derivatives natively, the instructional material may be shipped separately Such as Sodium carboxymethylcellulose, methylcellulose, from the container with the intention that the instructional hydroxypropylmethylcellulose. material and the compound be used cooperatively by the 2121 Known emulsifying agents include, but are not lim recipient. ited to acacia. Known preservatives include, but are not lim ited to, methyl, ethyl, or n-propyl-para-hydroxybenzoates, ascorbic acid, and Sorbic acid. Known Sweetening agents EXPERIMENTAL, EXAMPLES include, for example, glycerol, propylene glycol, Sorbitol, 2128. The invention is now described with reference to the Sucrose, and saccharin. following examples. These examples are provided for the 2122 Powdered and granular formulations of a pharma purpose of illustration only and the invention should in no ceutical preparation of the invention may be prepared using way be construed as being limited to these examples but known methods. Such formulations may be administered rather should be construed to encompass any and all varia directly to a subject, used, for example, to form tablets, to fill tions which become evident as a result of the teaching pro capsules, or to prepare an aqueous Suspension or Solution by vided herein. addition of an aqueous vehicle thereto. Each of these formu lations may further comprise one or more of dispersing or wetting agent, a suspending agent, and a preservative. Addi Experiment 1—Administration of Compositions Not tional excipients, such as fillers and Sweetening, flavoring, or Containing a Targeting Agent coloring agents, may also be included in these formulations. 2129. A composition prepared from a mixture of lipids including approximately 62 mole percent 1,2-distearoyl-sn Methods of Treating Diseases glycero-3-phosphocholine, approximately 22 mole percent 2123 Diseases, such as diabetes, may be treated by orally dihexadecyl phosphate, approximately 16 mole percent cho administering a composition of the invention wherein insulin lesterol, and no targeting agent was prepared according to the is the associated therapeutic agent. Similarly, diabetes may be microfluidization procedure generally described herein. A treated by orally administering a compound of the invention known portion of the lipid component comprised ''C labeled wherein insulin is the associated therapeutic and wherein phospholipid. Following filtration through a 0.2 micron filter, another form of insulin is co-administered. Routes of co the average constituent size was less than 100 nm as measured administration include, but are not limited to, oral adminis with a Coulter Sub-micron Particle Size Analyzer. tration, intramuscular injection, inhalation, intravenous 2130. A 10 mg/kg body weight sample of the composition injection, intra-arterial injection, as well as any other form of (containing 85,000 cpm of ''C radio-label) was then injected administration. into the duodenum of an anesthetized 230 gram fasted, but 2124. Although a physician will be able to select the otherwise normal, rat. Blood was taken from the portal and appropriate dose for a given patient, the range of doses that femoral veins at 15 and 30 minutes post-dosing for counting may be delivered in a given formulation of a compound of the (FIG.2). At 30 minutes post-dosing, the rat was sacrificed and invention is from about 1 to about 40 units, but may be 5, 10. representative samples of blood, liver, and spleen were 15, 20, 25, 30, or 35 units. A given formulation may, however, removed for analysis (FIG. 3). contain any whole or partial integer therebetween and may 2131 Labeled composition, as measured by ''C, was exceed 40 units. found in both portal and femoral blood of the rat. The portal 2125. Of course, diseases other than diabetes may be blood levels of ''C labeled composition was higher than the treated by orally administering a composition of the invention femoral blood levels (FIG. 2). At 30 minutes post-dosing, with a different associated therapeutic agent. A person of approximately 15% of the composition that was injected into ordinary skill in the art, armed with the disclosure herein, will the gut was found in the blood. Approximately 4% of the be able to select a given therapeutic agent, associate that counts were found in the liver and about 1% were found in the US 2010/0080773 A1 Apr. 1, 2010 spleen. Considering the relative sizes of the liver and spleen, sition is shown in FIG. 6. Mice receiving the composition had the splenic uptake was much higher than liver uptake on a a statistically significant reduction in blood glucose on day weight basis. seven (p<0.01) compared to mice receiving regular insulin, whose blood glucose was not altered at all. Experiment 2-Hepatocyte Targeting Example 4 2132) To demonstrate the absorption of the composition from the gut, a composition comprising insulin and approxi In Vivo Administration of Serotonin mately 61 mole percent 1.2 distearoyl-sn-glycero-3-phos phocholine, approximately 22 mole percent dihexadecyl 2137 The hepatic action of a composition comprising phosphate, approximately 16 mole percent cholesterol, and serotonin and approximately 61 mole percent 1.2 distearoyl approximately 1 mole percent polyCr-bis(N-2,6-diisopropy sn-glycero-3-phosphocholine, approximately 22 mole per lphenylcarbamoylmethyl iminodiacetic acid) (wherein a cent dihexadecyl phosphate, approximately 16 mole percent known portion of the phospholipid component comprised ''C cholesterol, and 1 mole percent of polyCr-bis(N-2,6-diiso labeled phospholipid) was prepared as recited in the general propylphenylcarbamoylmethyl iminodiacetic acid) was preparation disclosed herein. Prior to dosing the labeled com demonstrated in a type 2 diabetic dog (truncal vagotomy). position to rats, the rats were fasted from food for 24 hours The dog was fasted, and then anesthetized. Blood sampling and from water for 4 hours. The fasted rats were then permit catheters were placed in the hepatic and portal veins to enable ted to drink water from a graduated water bottle containing simultaneous blood sampling. Glucose was infused into the the composition. The drinking water bottle was removed from portal system at a rate of 0.5 g/kg/hour. Next, the above the cage after 15 minutes, the amount of water ingested from described composition was administered intraduodenally in a the drinking bottle was measured, and the amount of compo single dose of 30 ug/kg body weight. Results are depicted in sitioningested was calculated. The rats' blood was sampled at FIG. 7 and demonstrate that serotonin (also referred to as 15, 30, and 45 minutes and the radiolabel in each sample was 5-hydroxytryptamine or 5-HT), administered intraduode counted (FIG. 4). At 45 minutes the rats were sacrificed and nally as a composition of the invention is effective at low the livers were counted for radio-label (FIG. 5). doses in converting a type 2 diabetic dog from hepatic glucose 2133 As is shown in FIG. 4, approximately 8% of the output to uptake during a portal glucose load. ingested dose was found in the rats’ blood 15 minutes after the water had been removed from the cage. The quantity in the Example 5 rats' blood remained constant between 15 and 45 minutes. Liver uptake was approximately 8% at 45 minutes. Splenic In Vivo Administration of Calcitonin uptake at 45 minutes was approximately 1% of the ingested 2138 Normal, fasted, control rats were given a dose of dose (FIG. 5). The total absorption was approximately 17% salmon calcitonin via Subcutaneous injection Such that an (including blood, liver, and spleen). initial 10% reduction in blood calcium was observed. Blood calcium levels were then measured for six hours post injec Experiment 3-Hepatocyte Targeting with a tion. An experimental group of rats was given the same effec Composition in Alloxan-Streptozotocin Treated Mice tive dose of calcitonin by oral gavage, in the form of a com 2134 Mice used in the present experiment were made position comprising calcitonin and approximately 62 mole diabetic by administering streptozotocin and alloxan. The percent 1.2 distearoyl-sn-glycero-3-phosphocholine, diabetic animals were then divided into two groups. The approximately 22 mole percent dihexadecyl phosphate, and control group (11 mice) was orally dosed with regularinsulin. approximately 16 mole percent cholesterol. Blood calcium The experimental group (7 mice) was orally dosed with a levels were followed for six hours (FIG. 8). A blood calcium composition comprising insulin and approximately 61 mole reduction of up to 20% was observed in the non-control rats. percent 1.2 distearoyl-sn-glycero-3-phosphocholine, This difference was statistically significant (FIG. 8). approximately 22 mole percent dihexadecyl phosphate, approximately 16 mole percent cholesterol, and approxi Example 6 mately 1 mole percent poly Cr-bis(N-2,6-diisopropylphenyl carbamoylmethyliminodiacetic acid) (whereina known por Clinical Trial with Targeted Insulin in Type 2 Diabe tion of the phospholipid component comprised ''C labeled tes Mellitus Subjects phospholipid). Dosing was accomplished utilizing the water 2139 Capsules containing a composition of the invention bottle dosing method described in Experiment 2. were prepared. The composition comprised insulin as the 2135. After being made diabetic, rats in both groups were therapeutic agent, gelatin, and approximately 61 mole per treated identically over a 7 day period and fed with plain food cent 1.2 distearoyl-sn-glycero-3-phosphocholine, approxi and plain water. Following this 7 day period, rats in the mately 22 mole percent dihexadecyl phosphate, approxi control group were treated for an additional 7 day experimen mately 16 mole percent cholesterol, and about 1 mole percent tal period with food and regular insulin in the available drink of the sodium salt of Biotin-HDPE. Each capsule contained 2 ing water at 0.1 U/ml. Over the same 7 day experimental U of insulin. period, the experimental group was fed regular food with the 2140 Six well characterized Type 2 diabetes patients par composition of the invention available in the drinking water at ticipated in the controlled study. The patients were main 0.1 U/ml. At the end of each 7-day period, blood glucose was tained on their customary Type 2 oral anti-diabetes therapy. measured in a tail-vein sample of blood by a Beckman Blood Study participants were also given either placebo capsules or Glucose Analyzer. the above described capsules 30 minutes before a 60 gram 2136 The pharmacologic efficacy of orally administered carbohydrate meal at breakfast, lunch and dinner. Blood insulin in the group dosed with the above described compo samples were drawn at frequent intervals over a 13 hour US 2010/0080773 A1 Apr. 1, 2010 54 period and the Incremental Area Under the Curve for the blood glucose values was calculated for each Subject. TABLE 3 2141 At 0.1 U/kg body weight/meal, the same dose that is frequently used with Subcutaneous injection of insulin at a Liver given meal, a statistically significant reduction in AUC for Treatment Dose GLP-1 mg?rat Glycogen mgg liver each of the three meals was observed. FIG. 10 depicts the Control Oral GLP-1 O.O1 40 22 Intraperitoneal GLP-1 O.O1 59 44 results of the trial in graphical format. Oral Associated GLP-1 O.OOS 7356* Example 7 Oral Associated GLP-1 O.O1 90- 75* Insulin Concentration *p = 0.05 compared to Control Oral GLP-1 2142 Insulin U-500 contains 500 units of insulin/ml=0.5 Example 10 units/1 ul 2143 Add 3.36 ml of U-500 insulin to 70 ml of con Oral Thyroxine stituent suspension in 18 mM phosphate buffer (a pH 2153. Thyroxine is known to lower blood cholesterol and 7.01. triglyceride levels. However, at the doses required to treat 2144 (3,360 ul)*(0.5units of insulin/ul)=1,680 units of high cholesterol and triglyceride, thyroxine causes hyperthy insulin total in 73.36 ml roidism as an unwanted side effect. The goal of this study was 2145 (1,680 units of insulin)/(73.36 ml)=22.9 units of to demonstrate that orally administered targeted thyroxine insulin/ml—or 34.35 units of insulin/1.5 ml associated with a compound of the invention would act at the 2146 Load insulin for 21 hours: liver with the result of lowering blood lipids without inducing 2147 Post loading, chromatograph 1.5 ml of sample the unwanted hyperthyroidism. over a 1.5 cmx25 cm column with Sepharose CL-6B gel 2154 Normal laboratory mice, on high caloric diets, were equilibrated with 18 mM phosphate buffer (a pH 7.01 administered low oral doses (0.2 to 1.0 g) thyroxine in the 2148 0% of free insulin recovered from column; The form of a composition comprising thyroxine and constituents recovery of 0% of the total loaded insulin implies that generated from a mixture of lipid components comprising 100% of the total “loaded’ insulin is associated with a approximately 61 mole percent 1.2 distearoyl-sn-glycero-3- constituent of the composition. phosphocholine, approximately 22 mole percent dihexadecyl 2149 34.35 units of insulinx100%–34.35 units of insu phosphate, approximately 16 mole percent cholesterol, and lin bound or associated with the constituents of the approximately 1 mole percent of the sodium salt of Biotin invention. HDPE, a liver-targeting agent. FIG. 11 depicts the above described chromatography. A trace 2155 The mice, in groups of 4, were dosed daily by oral showing the elution time of free insulin is included for pur gavage for one week in a dose response study. Blood choles poses of comparison. As can be seen from the chromatogram, terol and triglycerides were measured after one week treat insulin is associated with the constituents of the invention and ment. Baseline values for cholesterol and triglycerides for all no free insulin is in solution. A preservative included with the groups were similar. The dose responses, shown in FIG. insulin does not associate with the constituents of the com 13, demonstrates the efficacy of orally administered, hepatic position of the invention and is visible in the chromatogram. targeted thyroxine associated with a composition of the invention. Blood levels of thyroid hormone did not increase Example 8 with the dosing of hepatic targeted oral thyroxine, demon Oral Delivery of GLP-1 strating the safety of the product. 2156. Other published studies (Erion, M., et al., PNAS 2150 Rats were fasted overnight. Subsequently, 800 mg Sep. 25, 2007 vol 104, #39, pp 15490-15495) with hepatic each of alloxan and streptozotocin were dissolved in 40 mL of targeted thyroxine analogs required doses at least 10 fold PBS (pH 7, 0.01 M). The fasted rats were then treated imme higher than those described herein to elicit similar reductions diately with a 0.5 mL IP dose to induce insulin deficiency. The in blood cholesterol and triglycerides. animals were then stabilized overnight with water and food. Following stabilization, the rats were fasted overnight to Example 11 deplete liver glycogen. 2151 Subsequently, the rats were administered 1.5 g glu Oral Interferon cose/kg body weight and GLP-1 in the form of a GLP-1 2157. A composition was prepared comprising inter associated with a composition comprising approximately 62 feron-C. as the therapeutic agent and approximately 61 mole mole percent 1.2 distearoyl-sn-glycero-3-phosphocholine, percent 1.2 distearoyl-sn-glycero-3-phosphocholine, approximately 22 mole percent dihexadecyl phosphate, and approximately 22 mole percent dihexadecyl phosphate, approximately 16 mole percent cholesterol (“associated approximately 16 mole percent cholesterol, and about 1 mole GLP-1). In separate experiments, the amount of associated percent of the sodium salt of Biotin-HDPE. GLP-1 was varied. Liverglycogen was measured chemically 2158 Six patients with Hepatitis C, genotype 3, were at 2 hours post dosing. treated with an aqueous Suspension of the above described 2152. As a control, unassociated GLP-1 was gavaged in composition and Ribivirin daily for 8 weeks. The inter place of associated GLP-1. In a separate control, GLP-1, in a feron-C. dose in the aqueous Suspension of the composition dose similar to that orally gavaged, was injected intraperito was 60,000 Units/day. neally. As is shown in Table 3, below, substantially enhanced 2159 Hepatitis C viral loads were measured at the begin oral efficacy was observed for the associated GLP-1 versus ning of the study, then at weeks 1, 2, 4, and 8. See FIG. 14. The non-associated GLP-1. data demonstrates the ability of the aqueous Suspension of a US 2010/0080773 A1 Apr. 1, 2010

composision of the invention to lower viral load with a mini was stopped, and excess XIII removed, by the addition of a mal dose of interferon. Side effects were likewise minimized. 50x molar excess of N-ethylmaleimide. 2164 Although the above example was described with Example 12 respect to IgG, it is equally applicable to any therapeutic agent with a nucleophilic nitrogen. 2160. In an example of a covalent interaction, IgG (human immunoglobulin, mixture of antibodies) was covalently Example 13 linked to MPB-PE to forma IgG construct. IgG is an antibody Administration of Covalent Oral IgG that is not normally orally bioavailable. In this embodiment of the invention, the lipids selected to form the composition of 2165 Human IgG antibodies were covalently attached to a the invention included approximately 68 mole percent 1.2- constituent of the invention, as described in Example 12. dipalmitoyl-sn-glycero-3-phosphocholine, approximately 19 Subsequently, eight 250 gram laboratory rats were prepared mole percent dihexadecylphosphate, approximately 10 mole with intra-duodenal catheters for the administration of cova percent cholesterol, and approximately 3 mol percent MPB lent IgG. After an overnight fast, 5 lug of covalent IgG was PE. infused into the duodenal catheter. The catheter was subse 2161 In order to form the composition of the invention, quently washed with 0.5 ml buffer. Blood samples were taken 1.2-dipalmitoyl-sn-glycero-3-phosphocholine, dihexadecyl at 15, 30, 60 and 120 minutes to assay the plasma concentra phosphate, and cholesterol were microfluidized as set forth tion of human IgG antibodies by ELISA reaction. earlier herein to form constituents with an average size of 2166. In a control experiment, 5ug of free IgG was infused between 50 and 60 nanometers. This suspension of constitu into the duodenal catheter. The catheter was subsequently ents was then transferred to around bottom flask that had been washed with 0.5 ml buffer. Blood samples were taken at 15, coated with a thin film of MPB-PE. The suspension was 30, 60 and 120 minutes to assay the plasma concentration of heated to about 62°C., with the temperature not falling below human IgG antibodies by ELISA reaction. The results of both 60° C. or exceeding 65° C. The heated suspension was sub studies are shown in FIG. 12. sequently stirred for 15 minutes until all of the MPB-PE had 2167 As can be seen in FIG. 12, covalent IgG provided been incorporated into the lipid construct. enhanced plasma concentration of human IgG (AUC) as com 2162 Separately, IgG was reacted with a 10 fold excess of pared to free IgG. Likewise, covalent IgG enhanced Tmax the time to maximum concentration, and Cmax the maxi linker precursor XI to form XII, per Scheme III. mum plasma concentration observed upon dosing. The enhanced efficacy of covalent IgG, as compared to free IgG, Scheme III thus demonstrates the ability of a compound of the invention to enhance oral absorption of very large proteins into the O systemic circulation. O O 2168) The disclosures of each and every patent, patent O n N application, and publication cited herein are hereby incorpo rated herein by reference in their entirety. us S O 2169 While this invention has been disclosed with refer XI ence to specific embodiments, it is apparent that other O O IgG embodiments and variations of this invention may be devised M by others skilled in the art without departing from the true NH spirit and scope of the invention. The appended claims are S intended to be construed to include all such embodiments and XII equivalent variations. What is claimed is: 1. An orally bioavailable composition comprising: one or more lipids selected from the group consisting of MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho line, 1.2-dimyristoyl-sn-glycero-3-phosphocholine, Compound XII was then purified using a 2.5x25 cm Sepha cholesterol, thiocholesterol, cholesterol oleate, dex G-25 column equilibrated with 18 mM phosphate buffer decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos plus 1.0 mM EDTA buffer at pH 7.4. Next, the acetyl protect phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- ing group on compound XII was removed by stirring com dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn pound XII with 50 mM hydroxylamine hydrochloride in 18 glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn mM sodium phosphate buffer containing 1.0 mM EDTA (pH glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn 7.4) for 2 hours at ambient temperature. The resulting free glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn thiol. XIII, was purified on 2.5x25 cm Sephadex G-25 col glycero-3-phospho-2-mercaptoethanol, 1.2- umn, as set forth for compound XII. dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- 2163 Immediately following purification, 200 u-moles of (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac compound XIII was mixed with 10 ml of the composition (1-glycerol) (sodium salt), and triethylammonium 2,3- prepared earlier. The reaction mixture was stirred for 15 min diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H utes, during which time compound XIII underwent a Michael thieno3,4-dimidazol-4-yl)pentanamido)ethyl reaction with the maleimide functionality of the MBP-PE phosphate; incorporated in the lipid construct. The conjugation reaction at least one therapeutic or diagnostic agent; US 2010/0080773 A1 Apr. 1, 2010 56

an optional reticuloendothelial system (“RES) masking glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn agent; and glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn an optional targeting agent, glycero-3-phospho-2-mercaptoethanol, 1.2- wherein said at least one therapeutic or diagnostic agent is dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- covalently bound to one or more of said one or more lipids (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac through a linker derived from a linker precursor according to (1-glycerol) (sodium salt), and triethylammonium 2,3- formula I, diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H thieno 3,4-dimidazol-4-yl) pentanamido)ethyl phosphate; at least one therapeutic or diagnostic agent; N21 an optional reticuloendothelial system (“RES) masking agent; and O i N an optional targeting agent, wherein said at least one therapeutic or diagnostic agent is Aul J 1. covalently bound to one or more of said one or more lipids through a linker derived from a linker precursor according to wherein formula II, “A.” is II Gl Gl Gl O O O O O N 1. N N NH No1 s’, S.-- O O O or NH-NH : wherein “J” corresponds to (CH), or G' is either Hor SONa; G is maleimidyl,

G' is either Hor SONa; and 'a' is independently, at each occurrence, 1, 2, 3, 4, 5, 6, 7, or 8. HNC(O)CHI, CH-NHC(O)(CH)NHC(O)CHI, 2. The composition according to claim 1, wherein the CHHNC(O)CHI; linker precursor according to formula I is N-Succinimidyl-3- "Q is optional and, when present, is C(O)(CH)NH; (2-pyridyldithio)proprionate (“SPDP). Succinimidyl 6-(3- “K” is optional and, when present, is —(CH)—, and 2-pyridyldithio-propionamido)hexanoate (“LC-SPDP), wherein “a” is independently, at each occurrence 1, 2, 3, 4, Sulfosuccinimidyl 6-(3'-2-pyridyldithio-propionamido) 5, 6, 7, or 8. hexanoate ("Sulfo-LC-SPDP), 4-Succinimidyloxycarbo 4. The composition according to claim 3, wherein the nyl-methyl-a-2-pyridyldithiotoluene (“SMPT), 4-Sulfos linker precursor according to formula II is Succinimidyl uccinimidyl-6-methyl-a-(2-pyridyldithio)toluamido 4-N-maleimidomethylcyclohexane-1-carboxylate hexanoate) ("Sulfo-LC-SMPT), or 3-(2-pyridyldithio) (“SMCC), Sulfosuccinimidyl 4-N-maleimidomethylcy propionyl hydrazide (“PDPH'). clohexane-1-carboxylate ("Sulfo-SMCC), m-Maleimido 3. An orally bioavailable composition comprising: benzoyl-N-hydroxysuccinimide ester (“MBS”), m-Maleimi one or more lipids selected from the group consisting of dobenzoyl-N-hydroxysulfosuccinimide ester (“Sulfo MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos MBS”), N-Succinimidyl-4-iodoacetylaminobenzoate phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho (“SIAB), N-Sulfosuccinimidyl 4-iodoacetylaminoben line, 1,2-dimyristoyl-sn-glycero-3-phosphocholine, Zoate ("Sulfo-SIAB), succinimidyl-4-(((iodoacetyl)amino) cholesterol, thiocholesterol, cholesterol oleate, dihexa methyl)cyclohexane-1-carboxylate (“SIAC), succinimidyl decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos 4-p-maleimidophenylbutyrate (“SMPB), sulfosuccinim phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- idyl 4-p-maleimidophenylbutyrate ("Sulfo-SMPB), or dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn Succinimidyl-6-((((4-(iodoacetyl)amino)methyl)cyclohex glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn ane-1-carbonyl)amino)-hexanoate (“SIACX”). US 2010/0080773 A1 Apr. 1, 2010 57

5. An orally bioavailable composition comprising: glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn one or more lipids selected from the group consisting of glycero-3-phospho-2-mercaptoethanol, 1,2-dipalmi MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos toyl-sn-glycero-3-phosphoethanolamine-N-(Succinyl), phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho 1,2-dipalmitoyl-sn-glycero-3-phospho-rac-(1-glyc erol) (sodium salt), and triethylammonium 2,3-diac line, 1,2-dimyristoyl-sn-glycero-3-phosphocholine, etoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H cholesterol, thiocholesterol, cholesterol oleate, dihexa decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos thieno3,4-dimidazol-4-yl)pentanamido)ethyl phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- phosphate; dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn at least one therapeutic or diagnostic agent; glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn an optional reticuloendothelial system (“RES) masking glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn agent; and glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn an optional targeting agent, glycero-3-phospho-2-mercaptoethanol, 1.2- wherein said at least one therapeutic or diagnostic agent is dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- covalently bound to one or more of said one or more lipids (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac through a linker derived from a linker precursor according to (1-glycerol) (sodium salt), and triethylammonium 2,3- formula IV. diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H thieno3,4-dimidazol-4-yl)pentanamido)ethyl IV phosphate; at least one therapeutic or diagnostic agent; an optional reticuloendothelial system (“RES) masking agent; and an optional targeting agent, wherein said at least one therapeutic or diagnostic agent is covalently bound to one or more of said one or more lipids 8. An orally bioavailable composition comprising: through a linker derived from a linker precursor according to one or more lipids selected from the group consisting of MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos formula III, phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho line, 1.2-dimyristoyl-sn-glycero-3-phosphocholine, III cholesterol, thiocholesterol, cholesterol oleate, dihexa decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn glycero-3-phospho-2-mercaptoethanol, 1.2- dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- wherein (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac G' is either Hor SONa; (1-glycerol) (sodium salt), and triethylammonium 2,3- diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H G' is maleimidyl, HNC(O)CHI, or NHC(O)(CH) thieno3,4-dimidazol-4-yl)pentanamido)ethyl NHC(O)CHI; and phosphate; a' is independently at each occurrence 1, 2, 3, 4, 5, 6, 7, or 8. at least one therapeutic or diagnostic agent; 6. The composition according to claim 5, wherein the an optional reticuloendothelial system (“RES) masking linker precursor according to formula III is N-g-maleimi agent; and dobutyryloxysuccinimide ester (“GMBS”), N-g-maleimi an optional targeting agent, dobutyryloxysulfosuccinimide ester ("Sulfo-GMBS”), suc wherein said at least one therapeutic or diagnostic agent is cinimidyl-6-((iodoacetyl)amino)hexanoate (“SIAX'), or covalently bound to one or more of said one or more lipids Succinimidyl-6-(6-(((iodoacetyl)amino)hexanoyl)amino) through a linker derived from a linker precursor according to hexanoate (“SIAXX'). formula V. 7. An orally bioavailable composition comprising: one or more lipids selected from the group consisting of V MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos O phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho O line, 1,2-dimyristoyl-sn-glycero-3-phosphocholine, cholesterol, thiocholesterol, cholesterol oleate, dihexa GE) X-z Z-N decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos HN-NH phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- X dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn G O glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn US 2010/0080773 A1 Apr. 1, 2010 58 wherein G7 is Z is independently optional at each occurrence, and, when present, (CH), and O wherein “a” is independently at each occurrence 1, 2, 3, 4, Gl 5, 6, 7, or 8. 9. The composition according to claim 8, wherein the N-O linker precursor according to formula V is 4-(4-N-Maleimi dophenyl)butyric acid hydrazide hydrochloride (“MBPH), O 4-(N-maleimidophenyl)cyclohexane-1-carbonyl-hy drazide hydrochloride (“MCH). wherein G' is either H or - SONa; and 10. An orally bioavailable composition comprising: G is one or more lipids selected from the group consisting of MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho NN line, 1,2-dimyristoyl-sn-glycero-3-phosphocholine, cholesterol, thiocholesterol, cholesterol oleate, dihexa decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos 1s-y- phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn with the proviso that is G is —C(O)C7 only when G is glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn - NO, and that G is - NO, only when G is C(O)C7. glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn 11. The composition according to claim 10, wherein the linker precursor according to formula VI is N-Hydroxysuc glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn cinimidyl-4-azidosalicylic acid (“NHS-ASA'), N-hydrox glycero-3-phospho-2-mercaptoethanol, 1.2- ysulfosuccinimidyl-4-azidosalicylic acid ("Sulfo-NHS dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- ASA'), SulfoSuccinimidyl-4-azidosalicylamido-hexanoate (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac ("Sulfo-NHS-LC-ASA), N-hydroxysuccinimidyl-4-azido (1-glycerol) (sodium salt), and triethylammonium 2,3- benzoate (“HSAB), N-hydroxysulfosuccinimidyl-4-azido diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H benzoate ("Sulfo-HSAB), N-5-azido-2-nitrobenzoylox thieno3,4-dimidazol-4-yl)pentanamido)ethyl y succinimide (ANB-NOS), N-succinimidyl-6-(4-azido phosphate; 2'-nitrophenylamino)hexanoate (“SANPAH), N-sulfosuccinimidyl-6-(4-azido-2'-nitrophenylamino)hex at least one therapeutic or diagnostic agent; anoate ("Sulfo-SANPAH), N-succinimidyl(4-azidophe an optional reticuloendothelial system (“RES) masking nyl)-1,3'-dithioproprionate (“SADP), N-Sulfosuccinimidyl agent; and (4-azidophenyl)-1,3'-dithioproprionate ("Sulfo-SADP), an optional targeting agent, SulfoSuccinimidyl-2-(p-azidosalicylamido)-ethyl-1,3'- dithiopropionate (“SASD), 1-(p-azidosalicylamido)-4-(io wherein said at least one therapeutic or diagnostic agent is doacetamido)butane (ASIB), N-4-(p-azidosalicylamido) covalently bound to one or more of said one or more lipids butyl-3'-(2-pyridyldithio)propionamide (“APDP), through a linker derived from a linker precursor according to p-azidobenzoylhydrazide (ABH'), 4-p-azidosalicylamido formula VI, butylamine (ASBA), p-azidophenylglyoxal (APG'), or SulfoSuccinimidyl-4-(p-azidophenyl)butyrate (“Sulfo SAPB). VI 12. An orally bioavailable composition comprising: one or more lipids selected from the group consisting of MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho line, 1.2-dimyristoyl-sn-glycero-3-phosphocholine, cholesterol, thiocholesterol, cholesterol oleate, dihexa decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos wherein phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- G is selected from the group consisting of C(O)G", dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn C(O)NHNH, C(O)C(O)H, C(O)NH(CH) glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn NH, C(O)NH(CH.)NHC(O)CHI, C(O)NH glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn (CH),C(O)G7 - NO, -(CH)NHC(O)G7 - NH glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn (CH),C(O)G7, -(CH)SSC(O)C7, C(O)NH(CH) glycero-3-phospho-2-mercaptoethanol, 1.2- dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- SS(CH),C(O)C 7, -(CH),C(O)C7, and C(O)NH (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac (CH)NHC(O)(CH-)G: (1-glycerol) (sodium salt), and triethylammonium 2,3- 'a' is independently at each occurrence 1,2,3,4,5,6,7, or diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H 8: thieno3,4-dimidazol-4-yl)pentanamido)ethyl G is selected from the group consisting —OH, NO, phosphate; —H, and —C(O)G': at least one therapeutic or diagnostic agent; US 2010/0080773 A1 Apr. 1, 2010 59

an optional reticuloendothelial system (“RES) masking agent; and an optional targeting agent, VIII wherein said at least one therapeutic or diagnostic agent is O O covalently bound to one or more of said one or more lipids Gl G8 through a linker derived from a linker precursor according to N-O formula VIII,

O VII Gl wherein G' is either Hor SONa; O O -> G is selected from the group consisting of 2-nitrophenyl s--- S es 5-azido and O wherein G' is either Hor SONa: G is selected from the group consisting of 2-nitrophenyl 5-azido and O O N.

15. The composition according to claim 14, wherein the H linker precursor according to formula VIII is Sulfo-Succinim C idyl 7-azido-4-methylcoumarin-3-acetate (“Sulfo SAMCA). > NY 16. An orally bioavailable composition comprising: O O N3; and one or more lipids selected from the group consisting of MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos wherein “a” is independently, at each occurrence, 1, 2, 3, 4, 5, phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho 6, 7, or 8. line, 1.2-dimyristoyl-sn-glycero-3-phosphocholine, 13. The composition according to claim 12, wherein the cholesterol, thiocholesterol, cholesterol oleate, dihexa linker precursor according to formula VII is SulfoSuccinim decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos idyl-2-(m-azido-o-nitrobenzamido)-ethyl-1,3'-proprionate phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- (“SAND”) or sulfosuccinimidyl 2-7-amino-4-methylcou dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn marin-3-acetamido)ethyl-1,3'dithiopropionate (“SAED). glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn 14. An orally bioavailable composition comprising: glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn one or more lipids selected from the group consisting of glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos glycero-3-phospho-2-mercaptoethanol, 1.2- phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- line, 1,2-dimyristoyl-sn-glycero-3-phosphocholine, (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac cholesterol, thiocholesterol, cholesterol oleate, dihexa (1-glycerol) (sodium salt), and triethylammonium 2,3- decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- thieno3,4-dimidazol-4-yl)pentanamido)ethyl dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn phosphate; glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn at least one therapeutic or diagnostic agent; glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn an optional reticuloendothelial system (“RES) masking glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn agent; and glycero-3-phospho-2-mercaptoethanol, 1.2- an optional targeting agent, dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- wherein said at least one therapeutic or diagnostic agent is (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac covalently bound to one or more of said one or more lipids (1-glycerol) (sodium salt), and triethylammonium 2,3- through a linker derived from a linker precursor according to diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H formula IX, thieno3,4-dimidazol-4-yl)pentanamido)ethyl phosphate; at least one therapeutic or diagnostic agent; an optional reticuloendothelial system (“RES) masking agent; and an optional targeting agent, wherein said at least one therapeutic or diagnostic agent is covalently bound to one or more of said one or more lipids through a linker derived from a linker precursor according to formula VIII, US 2010/0080773 A1 Apr. 1, 2010 60 wherein 22. A method of preparing an orally bioavailable compo G' is selected from the group consisting of maleimidyl sition comprising: and NC(O)CHI. one or more lipids selected from the group consisting of 17. The composition according to claim 16, wherein the MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos linker precursor according to formula IX is benzophenone-4- phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho iodoacetamide or benzophenone-4-maleimide. line, 1.2-dimyristoyl-sn-glycero-3-phosphocholine, 18. An orally bioavailable composition comprising: cholesterol, thiocholesterol, cholesterol oleate, dihexa one or more lipids selected from the group consisting of decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn line, 1,2-dimyristoyl-sn-glycero-3-phosphocholine, glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn cholesterol, thiocholesterol, cholesterol oleate, dihexa glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- glycero-3-phospho-2-mercaptoethanol, 1.2- dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn (1-glycerol) (sodium salt), and triethylammonium 2,3- glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn diacetoxypropyl 2-(5-((3aS,6aR)-2-oxohexahydro-1H glycero-3-phospho-2-mercaptoethanol, 1.2- thieno3,4-dimidazol-4-yl)pentanamido)ethyl dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- phosphate; (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac at least one therapeutic or diagnostic agent; (1-glycerol) (sodium salt), and triethylammonium 2,3- an optional RES masking agent; and diacetoxypropyl 2-(5-((3 aS,6aR)-2-oxohexahydro-1H an optional targeting agent, thieno3,4-dimidazol-4-yl)pentanamido)ethyl wherein said at least one therapeutic or diagnostic agent is phosphate; covalently bound to one or more of said one or more lipids at least one therapeutic or diagnostic agent; either directly or through a linker derived from a linker pre an optional reticuloendothelial system (“RES) masking cursor according to formula I, said method comprising the agent; and steps of: an optional targeting agent, a. Selecting one or more of said lipids; wherein said at least one therapeutic or diagnostic agent is b. mixing said lipids to form a mixture of lipid-based con covalently bound to one or more of said one or more lipids stituents; through a linker derived from a linker precursor according to c. Selecting said therapeutic agent; formula X, d. reacting said therapeutic agent with a linker-precursor of formula I to form a therapeutic agent/linker conjugate; and e. adding said conjugate to said mixture to form said com position. 23. An orally bioavailable composition comprising: one or more lipids selected from the group consisting of MCC-PE, MPB-PE, 1,2-distearoyl-sn-glycero-3-phos phocholine, 1,2-dipalmitoyl-sn-glycero-3-phosphocho line, 1.2-dimyristoyl-sn-glycero-3-phosphocholine, wherein G' is selected from the group consisting of C(O)C cholesterol, thiocholesterol, cholesterol oleate, dihexa (N)H and C(N)CF. decyl phosphate, 1,2-distearoyl-sn-glycero-3-phos 19. The composition of claim 1, wherein said therapeutic phate, 1,2-dipalmitoyl-sn-glycero-3-phosphate, 1.2- agent is selected from the group consisting of insulin, inter dimyristoyl-sn-glycero-3-phosphate, 1,2-distearoyl-sn feron, erythropoietin, parathyroid hormone, calcitonin, sero glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn tonin, rituximab, trastuzumab, uricase, tissue plasminogen glycero-3-phosphoethanolamine, 1,2-dimyristoyl-sn activator, thymoglobin, a vaccine, heparin or a heparin ana glycero-3-phosphoethanolamine, 1,2-dipalmitoyl-sn log, antithrombin III, filgrastin, pramilitide acetate, glycero-3-phospho-2-mercaptoethanol, 1.2- exanatide, epifibatide, antivenins, IgG, IgM, HGH, thyrox dipalmitoyl-sn-glycero-3-phosphoethanolamine-N- ine, GLP-1, blood clotting Factors VII and VIII, IX, X, a (Succinyl), 1,2-dipalmitoyl-sn-glycero-3-phospho-rac monoclonal antibody, and glycolipids that act as therapeutic (1-glycerol) (sodium salt), and triethylammonium 2,3- agents. diacetoxypropyl 2-(5-((3 aS,6aR)-2-oxohexahydro-1H 20. The composition of claim 1 wherein the lipids are 1.2 thieno3,4-dimidazol-4-yl)pentanamido)ethyl distearoyl-sn-glycero-3-phosphocholine, dihexadecyl phos phosphate; and phate, and cholesterol. at least one therapeutic agent selected from the group con 21. The composition of claim 20 wherein said 1.2 dis sisting of blood clotting factors VII, VIII, IX, Kallikrein, tearoyl-sn-glycero-3-phosphocholine is present in approxi Kininogen, Hageman Factor (XII), plasma thrombo mately 62 mole percent, said dihexadecyl phosphate is plastin antecedent Factor (XI), tissue factor, Stuart Fac present in approximately 22 mole percent, and said choles tor (X), accelerin (V), prothrombin (II), and fibrin sta terol is present in approximately 16 mole percent, wherein, bilizing Factor (XIII). optionally, at least about 25% of the cholesterol is thiocho lesterol. c c c c c