ANTISENSE MODULATION OF FIBROBLAST GROWTH FACTOR RECEPTOR 4 EXPRESSION

Provided herein are methods, compounds, and compositions for reducing expression of fibroblast growth factor receptor 4 (FGFR4) mRNA and protein in an animal. Such methods, compounds, and compositions are useful to treat, prevent, delay, or ameliorate a metabolic disease, or a symptom thereof.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 13/631,437, filed Sep. 28, 2012, which is a continuation of U.S. patent application Ser. No. 13/525,197, filed Jun. 15, 2012, which claims priority under 35 U.S.C. §119(e) to U.S. Provisional Patent Application No. 61/497,921, filed Jun. 16, 2011, each of which is incorporated herein by reference in its entirety.

SEQUENCE LISTING

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0157USC2SEQ_ST25.txt created Dec. 4, 2014, which is 160 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.

FIELD

Provided herein are methods, compounds, and compositions for reducing expression of fibroblast growth factor receptor 4 (FGFR4) mRNA and protein in an animal. Such methods, compounds, and compositions are useful, for example, to treat, prevent, delay or ameliorate diseases associated with metabolic disorders, particularly disorders associated with obesity.

BACKGROUND

Obesity is considered a long-term metabolic disease. There are several serious medical sequelae related to obesity. There are over 1 billion overweight individuals worldwide with 100 million clinically obese. The increasing health care costs of treating obesity related diseases in the US alone are estimated at over $100 billion annually. Current methods for treating obesity include behavioral modification, diet, surgery (gastroplasty), administering pharmaceutical agents that block appetite stimulating signals or absorption of nutrients (fat), and administering agents that increase thermogenesis or fat metabolism. Some of these methods have disadvantages in that they rely on patient resolve, are invasive, or have unwanted side effects. An understanding of the mechanisms by which obesity is regulated may provide important therapeutic information.

Obesity is frequently associated with insulin resistance and together constitutes risk factors for later development of type 2 diabetes and cardiovascular diseases. Insulin resistance occurs well before development of type 2 diabetes, and insulin is overproduced to compensate for the insulin resistance and to maintain normal glucose levels. Type 2 diabetes ensues, as the pancreas can no longer produce enough insulin to maintain normal glucose levels. Early stages of type 2 diabetes are associated with elevated levels of insulin but as the disease progresses the pancreas may fail to produce insulin, resulting in increased blood glucose levels. Diabetes is a significant risk factor for both heart disease and stroke and is the leading cause of blindness and end-stage renal failure.

Diabetes is a disorder characterized by hyperglycemia due to deficient insulin action that may result from reduced insulin production or insulin resistance or both. Diabetes mellitus is a polygenic disorder affecting a significant portion of the people in the world. It is divided into two types. In type I diabetes, or insulin-dependent diabetes mellitus (IDDM), patients produce little or no insulin, the hormone that regulates glucose utilization. In type 2 diabetes, or noninsulin-dependent diabetes mellitus (NIDDM), patients often have plasma insulin levels that are the same compared to nondiabetic humans; however, these patients have developed a resistance to the insulin stimulating effect of glucose and lipid metabolism in the main insulin-sensitive tissues, i.e., muscle, liver and adipose tissues, and the plasma insulin levels are insufficient to overcome the pronounced insulin resistance. Additionally, glucotoxicity, which results from long-term hyperglycemia, induces tissue-dependent insulin resistance (Nawano et al., Am. J. Physiol. Endocrinol. Metab., 278, E535-543) exacerbating the disease. Type 2 diabetes accounts for over 90% of all diabetes cases. It is a metabolic disorder characterized by hyperglycemia leading to secondary complications such as neuropathy, nephropathy, retinopathy, hypertriglyceridemia, obesity, and other cardiovascular diseases generally referred to as metabolic syndrome.

Metabolic syndrome is a combination of medical disorders that increase one's risk for cardiovascular disease and diabetes. The symptoms, including high blood pressure, high triglycerides, decreased HDL and obesity, tend to appear together in some individuals. Metabolic syndrome is known under various other names, such as (metabolic) syndrome X, insulin resistance syndrome or Reaven's syndrome.

Diabetes and obesity (sometimes now collectively referred to as “diabesity”) are interrelated in that obesity is known to exacerbate the pathology of diabetes and greater than 60% of diabetics are obese. Most human obesity is associated with insulin resistance and leptin resistance. In fact, it has been suggested that obesity may have an even greater impact on insulin action than diabetes itself (Sindelka et al., Physiol Res., 51, 85-91). Additionally, several compounds on the market for the treatment of diabetes are known to induce weight gain, a very undesirable side effect to the treatment of this disease. Therefore, a compound that has the potential to treat both diabetes and obesity would provide a significant improvement over current treatments.

Fibroblast growth factor receptor 4 (also known as FGF receptor-4, TKF; tyrosine kinase related to fibroblast growth factor receptor; hydroxyaryl-protein kinase; tyrosylprotein kinase; Fgfr4; FGFR-4; FGFR4; CD334, FGFR4_HUMAN and JTK2) has high affinity for the acidic and/or basic fibroblast growth factors. (Armstrong et al., Genes Chromosomes Cancer, 4, 94-98).

Although FGFRs generally have been shown to have wide distribution throughout the body, to date, FGFR4 has only been found in a few tissues. Among a wide variety of cells and tissues tested, including human lymphocytes and macrophages, FGFR4 was found to be expressed in the lung and in some tumors of lung origin as well as in malignancies not derived from lung tissues. (Holtrich et al., Proc. Nat. Acad. Sci., 88, 10411-10415). FGFR4 has also been found to be expressed in the liver and in adipose tissues. (Patel et al., JCEM, 90(2), 1226-1232). FGFR4 has also been found to be expressed in certain carcinoma cell lines. (Bange et al., Cancer Res., 62, 840-847).

Additionally, FGFR4 has been shown to play a role in systemic lipid and glucose homeostasis. FGFR4-deficient mice on a normal diet exhibited features of metabolic syndrome that include increase mass of insulin resistance, in addition to hypercholesterolemia. FGFR4 deficiency was shown to alleviate high-fat diet-induced fatty liver in a certain obese mouse model, which is also a correlate of metabolic syndrome. Restoration of FGFR4, specifically in hepatocytes of FGFR4 deficient mice, decrease plasma lipid level and restored the high fat diet-induced fatty liver but failed to restore glucose tolerance and sensitivity to insulin. (Huang et al., Diabetes, 56, 2501-2510).

Antisense inhibition of FGFR4 provides a unique advantage over traditional small molecule inhibitors in that antisense inhibitors do not rely on competitive binding of the compound to the protein and inhibit activity directly by reducing the expression of FGFR4. A representative United States patent that teaches FGFR4 antisense inhibitors includes US. Pat. Publication No. US2010/0292140, of which is herein incorporated by reference in its entirety. Antisense technology is emerging as an effective means for reducing the expression of certain gene products and may therefore prove to be uniquely useful in a number of therapeutic, diagnostic, and research applications for the modulation of FGFR4.

There is a currently a lack of acceptable options for treating metabolic disorders. It is therefore an object herein to provide compounds and methods for the treatment of such diseases and disorder. This invention relates to the discovery of novel, highly potent inhibitors of FGFR4 gene expression.

All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated-by-reference for the portions of the document discussed herein, as well as in their entirety.

SUMMARY

Provided herein are methods, compounds, and compositions for modulating expression of FGFR4 and treating, preventing, delaying or ameliorating diseases associated with metabolic disorders, particularly disorders associated with obesity and/or a symptom thereof.

DETAILED DESCRIPTION

It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive described herein, as claimed. Herein, the use of the singular includes the plural unless specifically stated otherwise. As used herein, the use of “or” means “and/or” unless stated otherwise. Furthermore, the use of the term “including” as well as other forms, such as “includes” and “included”, is not limiting. Also, terms such as “element” or “component” encompass both elements and components comprising one unit and elements and components that comprise more than one subunit, unless specifically stated otherwise.

The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated-by-reference for the portions of the document discussed herein, as well as in their entirety.

DEFINITIONS

Unless specific definitions are provided, the nomenclature utilized in connection with, and the procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques can be used for chemical synthesis, and chemical analysis. Where permitted, all documents, or portions of documents, cited in this application, including, but not limited to, all patents, applications, published applications and other journal publications, GENBANK Accession Numbers and associated sequence information obtainable through databases such as National Center for Biotechnology Information (NCBI) and other data referred to throughout in the disclosure herein are incorporated by reference for the portions of the document discussed herein, as well as in their entirety.

Unless otherwise indicated, the following terms have the following meanings:

“2′-O-methoxyethyl” (also 2′-MOE and 2′-O(CH2)2—OCH3) refers to an O-methoxy-ethyl modification of the 2′ position of a furosyl ring. A 2′-O-methoxyethyl modified sugar is a modified sugar.

“2′-O-methoxyethyl nucleotide” means a nucleotide comprising a 2′-O-methoxyethyl modified sugar moiety.

“3′ target site” refers to the nucleotide of a target nucleic acid which is complementary to the 3′-most nucleotide of a particular antisense compound.

“5′ target site” refers to the nucleotide of a target nucleic acid which is complementary to the 5′-most nucleotide of a particular antisense compound.

“5-methylcytosine” means a cytosine modified with a methyl group attached to the 5′ position. A 5-methylcytosine is a modified nucleobase.

“About” means within ±10% of a value. For example, if it is stated, “a marker may be increased by about 50%”, it is implied that the marker may be increased between 45%-55%.

“Active pharmaceutical agent” means the substance or substances in a pharmaceutical composition that provide a therapeutic benefit when administered to an individual. For example, in certain embodiments an antisense oligonucleotide targeted to FGFR4 is an active pharmaceutical agent.

“Active target region” or “target region” means a region to which one or more active antisense compounds is targeted. “Active antisense compounds” means antisense compounds that reduce target nucleic acid levels or protein levels.

“Adipogenesis” means the development of fat cells from preadipocytes. “Lipogenesis” means the production or formation of fat, either fatty degeneration or fatty infiltration.

“Adipose tissue” or “body fat” or “fat depot” is loose connective tissue composed of adipocytes. Two types of adipose tissue exist: white adipose tissue (WAT) and brown adipose tissue (BAT).

“Adiposity” or “Obesity” refers to the state of being obese or an excessively high amount of body fat or adipose tissue in relation to lean body mass. The amount of body fat includes concern for both the distribution of fat throughout the body and the size and mass of the adipose tissue deposits. Body fat distribution can be estimated by skin-fold measures, waist-to-hip circumference ratios, or techniques such as ultrasound, computed tomography, or magnetic resonance imaging. According to the Center for Disease Control and Prevention, individuals with a body mass index (BMI) of 30 or more are considered obese. The term “Obesity” as used herein includes conditions where there is an increase in body fat beyond the physical requirement as a result of excess accumulation of adipose tissue in the body. The term “obesity” includes, but is not limited to, the following conditions: adult-onset obesity; alimentary obesity; endogenous or inflammatory obesity; endocrine obesity; familial obesity; hyperinsulinar obesity; hyperplastic-hypertrophic obesity; hypogonadal obesity; hypothyroid obesity; lifelong obesity; morbid obesity and exogenous obesity.

“Administered concomitantly” refers to the co-administration of two agents in any manner in which the pharmacological effects of both are manifest in the patient at the same time. Concomitant administration does not require that both agents be administered in a single pharmaceutical composition, in the same dosage form, or by the same route of administration. The effects of both agents need not manifest themselves at the same time. The effects need only be overlapping for a period of time and need not be coextensive.

“Administering” means providing an agent to an animal, and includes, but is not limited to, administering by a medical professional and self-administering.

“Agent” means an active substance that can provide a therapeutic benefit when administered to an animal. “First Agent” means a therapeutic compound provided herein. For example, a first agent can be an antisense oligonucleotide targeting FGFR4. “Second agent” means a second therapeutic compound described herein (e.g. a second antisense oligonucleotide targeting FGFR4) and/or a non-FGFR4 therapeutic compound.

“Amelioration” refers to a lessening of at least one indicator, sign, or symptom of an associated disease, disorder, or condition. The severity of indicators can be determined by subjective or objective measures, which are known to those skilled in the art.

“Animal” refers to a human or non-human animal, including, but not limited to, mice, rats, rabbits, dogs, cats, pigs, and non-human primates, including, but not limited to, monkeys and chimpanzees.

“Antisense activity” means any detectable or measurable activity attributable to the hybridization of an antisense compound to its target nucleic acid. In certain embodiments, antisense activity is a decrease in the amount or expression of a target nucleic acid or protein encoded by such target nucleic acid.

“Antisense compound” means an oligomeric compound that is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.

“Antisense inhibition” means reduction of target nucleic acid levels or target protein levels in the presence of an antisense compound complementary to a target nucleic acid compared to target nucleic acid levels or target protein levels in the absence of the antisense compound.

“Antisense oligonucleotide” means a single-stranded oligonucleotide having a nucleobase sequence that permits hybridization to a corresponding region or segment of a target nucleic acid.

“Bicyclic sugar” means a furosyl ring modified by the bridging of two non-geminal ring atoms. A bicyclic sugar is a modified sugar.

“Bicyclic nucleic acid” or “BNA” refers to a nucleoside or nucleotide wherein the furanose portion of the nucleoside or nucleotide includes a bridge connecting two carbon atoms on the furanose ring, thereby forming a bicyclic ring system.

“Biomarker” is meant to designate a gene or protein or protein fragment which is indicative of the effect of an FGFR4 inhibitor. That means the “biomarker” is used as a detection agent.

“Cap structure” or “terminal cap moiety” means chemical modifications, which have been incorporated at either terminus of an antisense compound.

“Chemically distinct region” refers to a region of an antisense compound that is in some way chemically different than another region of the same antisense compound. For example, a region having 2′-O-methoxyethyl nucleotides is chemically distinct from a region having nucleotides without 2′-O-methoxyethyl modifications.

“Chimeric antisense compound” means an antisense compound that has at least two chemically distinct regions.

“Co-administration” means administration of two or more agents to an individual. The two or more agents can be in a single pharmaceutical composition, or can be in separate pharmaceutical compositions. Each of the two or more agents can be administered through the same or different routes of administration. Co-administration encompasses parallel or sequential administration.

“Cholesterol” is a sterol molecule found in the cell membranes of all animal tissues. Cholesterol must be transported in an animal's blood plasma by lipoproteins including very low density lipoprotein (VLDL), intermediate density lipoprotein (IDL), low density lipoprotein (LDL), and high density lipoprotein (HDL). “Plasma cholesterol” refers to the sum of all lipoproteins (VDL, IDL, LDL, HDL) esterified and/or non-esterified cholesterol present in the plasma or serum.

“Complementarity” means the capacity for pairing between nucleobases of a first nucleic acid and a second nucleic acid.

“cEt” or “constrained ethyl” means a bicyclic sugar moiety comprising a bridge connecting the 4′-carbon and the 2′-carbon, wherein the bridge has the formula: 4′-CH(CH3)—O-2′.

“Constrained ethyl nucleoside” (also cEt nucleoside) means a nucleoside comprising a bicyclic sugar moiety comprising a 4′-CH(CH3)—O-2′ bridge.

“Contiguous nucleobases” means nucleobases immediately adjacent to each other.

“Deoxyribonucleotide” means a nucleotide having a hydrogen at the 2′ position of the sugar portion of the nucleotide. Deoxyribonucleotides may be modified with any of a variety of substituents.

“Diabetes mellitus” or “diabetes” is a syndrome characterized by disordered metabolism and abnormally high blood sugar (hyperglycemia) resulting from insufficient levels of insulin or reduced insulin sensitivity. The characteristic symptoms are excessive urine production (polyuria) due to high blood glucose levels, excessive thirst and increased fluid intake (polydipsia) attempting to compensate for increased urination, blurred vision due to high blood glucose effects on the eye's optics, unexplained weight loss, and lethargy.

“Diabetic dyslipidemia” or “type 2 diabetes with dyslipidemia” means a condition characterized by Type 2 diabetes, reduced HDL-C, elevated triglycerides, and elevated small, dense LDL particles.

“Diluent” means an ingredient in a composition that lacks pharmacological activity, but is pharmaceutically necessary or desirable. For example, the diluent in an injected composition can be a liquid, e.g. saline solution.

“Dyslipidemia” refers to a disorder of lipid and/or lipoprotein metabolism, including lipid and/or lipoprotein overproduction or deficiency. Dyslipidemias may be manifested by elevation of lipids such as cholesterol and triglycerides as well as lipoproteins such as low-density lipoprotein (LDL) cholesterol.

“Dosage unit” means a form in which a pharmaceutical agent is provided, e.g. pill, tablet, or other dosage unit known in the art. In certain embodiments, a dosage unit is a vial containing lyophilized antisense oligonucleotide. In certain embodiments, a dosage unit is a vial containing reconstituted antisense oligonucleotide.

“Dose” means a specified quantity of a pharmaceutical agent provided in a single administration, or in a specified time period. In certain embodiments, a dose can be administered in one, two, or more boluses, tablets, or injections. For example, in certain embodiments where subcutaneous administration is desired, the desired dose requires a volume not easily accommodated by a single injection, therefore, two or more injections can be used to achieve the desired dose. In certain embodiments, the pharmaceutical agent is administered by infusion over an extended period of time or continuously. Doses can be stated as the amount of pharmaceutical agent per hour, day, week, or month.

“Effective amount” or “therapeutically effective amount” means the amount of active pharmaceutical agent sufficient to effectuate a desired physiological outcome in an individual in need of the agent. The effective amount can vary among individuals depending on the health and physical condition of the individual to be treated, the taxonomic group of the individuals to be treated, the formulation of the composition, assessment of the individual's medical condition, and other relevant factors.

“Fibroblast growth factor 4” or “FGFR4” means any nucleic acid or protein of FGFR4.

“FGFR4 expression” means the level of mRNA transcribed from the gene encoding FGFR4 or the level of protein translated from the mRNA. FGFR4 expression can be determined by art known methods such as a Northern or Western blot.

“FGFR4 nucleic acid” means any nucleic acid encoding FGFR4. For example, in certain embodiments, a FGFR4 nucleic acid includes a DNA sequence encoding FGFR4, a RNA sequence transcribed from DNA encoding FGFR4 (including genomic DNA comprising introns and exons), and a mRNA sequence encoding FGFR4. “FGFR4 mRNA” means a mRNA encoding a FGFR4 protein.

“Fully complementary” or “100% complementary” means each nucleobase of a nucleobase sequence of a first nucleic acid has a complementary nucleobase in a second nucleobase sequence of a second nucleic acid. In certain embodiments, a first nucleic acid is an antisense compound and a target nucleic acid is a second nucleic acid.

“Gapmer” means a chimeric antisense compound in which an internal region having a plurality of nucleosides that support RNase H cleavage is positioned between external regions having one or more nucleosides, wherein the nucleosides comprising the internal region are chemically distinct from the nucleoside or nucleosides comprising the external regions. The internal region can be referred to as a “gap segment” and the external regions can be referred to as “wing segments.”

“Gap-widened” means a chimeric antisense compound having a gap segment of 12 or more contiguous 2′-deoxyribonucleosides positioned between and immediately adjacent to 5′ and 3′ wing segments having from one to six nucleosides.

“Glucose” is a monosaccharide used by cells as a source of energy and inflammatory intermediate. “Plasma glucose” refers to glucose present in the plasma.

“Hybridization” means the annealing of complementary nucleic acid molecules. In certain embodiments, complementary nucleic acid molecules include an antisense compound and a target nucleic acid.

“Hyperlipidemia” or “hyperlipemia” is a condition characterized by elevated serum lipids or circulating (plasma) lipids. This condition manifests an abnormally high concentration of fats. The lipid fractions in the circulating blood are cholesterol, low density lipoproteins, very low density lipoproteins and triglycerides.

“Hypertriglyceridemia” means a condition characterized by elevated triglyceride levels.

“Identifying” or “selecting an animal with metabolic” means identifying or selecting a subject having been diagnosed with a metabolic disease, or a metabolic disorder; or, identifying or selecting a subject having any symptom of a metabolic disease, including, but not limited to, metabolic syndrome, hyperglycemia, hypertriglyceridemia, hypertension increased insulin resistance, decreased insulin sensitivity, above normal body weight, and/or above normal body fat or any combination thereof. Such identification may be accomplished by any method, including but not limited to, standard clinical tests or assessments, such as measuring serum or circulating (plasma) blood-glucose, measuring serum or circulating (plasma) triglycerides, measuring blood-pressure, measuring body fat, measuring body weight, and the like.

“Immediately adjacent” means there are no intervening elements between the immediately adjacent elements.

“Individual” or “subject” or “animal” means a human or non-human animal selected for treatment or therapy.

“Inhibiting the expression or activity” refers to a reduction or blockade of the expression or activity of a RNA or protein and does not necessarily indicate a total elimination of expression or activity.

“Insulin resistance” is defined as the condition in which normal amounts of insulin are inadequate to produce a normal insulin response from fat, muscle and liver cells. Insulin resistance in fat cells results in hydrolysis of stored triglycerides, which elevates free fatty acids in the blood plasma. Insulin resistance in muscle reduces glucose uptake whereas insulin resistance in liver reduces glucose storage, with both effects serving to elevate blood glucose. High plasma levels of insulin and glucose due to insulin resistance often leads to metabolic syndrome and type 2 diabetes.

“Insulin sensitivity” is a measure of how effectively an individual processes glucose. An individual having high insulin sensitivity effectively processes glucose whereas an individual with low insulin sensitivity does not effectively process glucose.

“Internucleoside linkage” refers to the chemical bond between nucleosides.

“Intravenous administration” means administration into a vein.

“Linked nucleosides” means adjacent nucleosides which are bonded together.

“Lipid-lowering therapy” or “lipid lowering agent” means a therapeutic regimen provided to a subject to reduce one or more lipids in a subject. In certain embodiments, a lipid-lowering therapy is provided to reduce one or more of ApoB, total cholesterol, LDL-C, VLDL-C, IDL-C, non-HDL-C, triglycerides, small dense LDL particles, and Lp(a) in a subject. Examples of lipid-lowering therapy include statins, fibrates, and MTP inhibitors.

“Major risk factors” refers to factors that contribute to a high risk for a particular disease or condition. In certain embodiments, major risk factors for coronary heart disease include, without limitation, cigarette smoking, hypertension, low HDL-C, family history of coronary heart disease, age, and other factors disclosed herein.

“Metabolic disease” or “metabolic disorder” refers to a condition characterized by an alteration or disturbance in metabolic function. “Metabolic” and “metabolism” are terms well known in the art and generally include the whole range of biochemical processes that occur within a living organism. Metabolic diseases or disorders include, but are not limited to, obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

“Metabolic syndrome” means a condition characterized by a clustering of lipid and non-lipid cardiovascular risk factors of metabolic origin. In certain embodiments, metabolic syndrome is identified by the presence of any 3 of the following factors: waist circumference of greater than 102 cm in men or greater than 88 cm in women; serum triglyceride of at least 150 mg/dL; HDL-C less than 40 mg/dL in men or less than 50 mg/dL in women; blood pressure of at least 130/85 mmHg; and fasting glucose of at least 110 mg/dL. These determinants can be readily measured in clinical practice (JAMA, 2001, 285: 2486-2497).

“Mismatch” or “non-complementary nucleobase” refers to the case when a nucleobase of a first nucleic acid is not capable of pairing with the corresponding nucleobase of a second or target nucleic acid.

“Mixed dyslipidemia” means a condition characterized by elevated cholesterol and elevated triglycerides.

“Modified internucleoside linkage” refers to a substitution or any change from a naturally occurring internucleoside bond (i.e. a phosphodiester internucleoside bond).

“Modified nucleobase” refers to any nucleobase other than adenine, cytosine, guanine, thymidine, or uracil. An “unmodified nucleobase” means the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U).

“Modified nucleoside” means a nucleoside having, independently, a modified sugar moiety or modified nucleobase.

“Modified nucleotide” means a nucleotide having, independently, a modified sugar moiety, modified internucleoside linkage, or modified nucleobase. A “modified nucleoside” means a nucleoside having, independently, a modified sugar moiety or modified nucleobase.

“Modified oligonucleotide” means an oligonucleotide comprising at least one modified nucleotide.

“Modified sugar” refers to a substitution or change from a natural sugar.

“Motif” means the pattern of chemically distinct regions in an antisense compound.

“Naturally occurring internucleoside linkage” means a 3′ to 5′ phosphodiester linkage.

“Natural sugar moiety” means a sugar found in DNA (2′-H) or RNA (2′-OH).

“Non-alcoholic fatty liver disease” or “NAFLD” means a condition characterized by fatty inflammation of the liver that is not due to excessive alcohol use (for example, alcohol consumption of over 20 g/day). In certain embodiments, NAFLD is related to insulin resistance and the metabolic syndrome. NAFLD encompasses a disease spectrum ranging from simple triglyceride accumulation in hepatocytes (hepatic steatosis) to hepatic steatosis with inflammation (steatohepatitis), fibrosis, and cirrhosis.

“Nonalcoholic steatohepatitis” (NASH) occurs from progression of NAFLD beyond deposition of triglycerides. A “second hit” capable of inducing necrosis, inflammation, and fibrosis is required for development of NASH. Candidates for the second-hit can be grouped into broad categories: factors causing an increase in oxidative stress and factors promoting expression of proinflammatory cytokines

“Nucleic acid” refers to molecules composed of monomeric nucleotides. A nucleic acid includes ribonucleic acids (RNA), deoxyribonucleic acids (DNA), single-stranded nucleic acids, double-stranded nucleic acids, small interfering ribonucleic acids (siRNA), and microRNAs (miRNA). A nucleic acid can also comprise a combination of these elements in a single molecule.

“Nucleobase” means a heterocyclic moiety capable of pairing with a base of another nucleic acid.

“Nucleobase sequence” means the order of contiguous nucleobases independent of any sugar, linkage, or nucleobase modification.

“Nucleoside” means a nucleobase linked to a sugar.

“Nucleoside mimetic” includes those structures used to replace the sugar or the sugar and the base and not necessarily the linkage at one or more positions of an oligomeric compound such as for example nucleoside mimetics having morpholino, cyclohexenyl, cyclohexyl, tetrahydropyranyl, bicyclo or tricyclo sugar mimetics e.g. non furanose sugar units.

“Nucleotide” means a nucleoside having a phosphate group covalently linked to the sugar portion of the nucleoside.

“Nucleotide mimetic” includes those structures used to replace the nucleoside and the linkage at one or more positions of an oligomeric compound such as for example peptide nucleic acids or morpholinos (morpholinos linked by —N(H)—C(═O)—O— or other non-phosphodiester linkage).

“Oligomeric compound” or “oligomer” refers to a polymeric structure comprising two or more sub-structures and capable of hybridizing to a region of a nucleic acid molecule. In certain embodiments, oligomeric compounds are oligonucleosides. In certain embodiments, oligomeric compounds are oligonucleotides. In certain embodiments, oligomeric compounds are antisense compounds. In certain embodiments, oligomeric compounds are antisense oligonucleotides. In certain embodiments, oligomeric compounds are chimeric oligonucleotides.

“Oligonucleotide” means a polymer of linked nucleosides each of which can be modified or unmodified, independent one from another.

“Parenteral administration” means administration through injection or infusion. Parenteral administration includes subcutaneous administration, intravenous administration, intramuscular administration, intraarterial administration, intraperitoneal administration, or intracranial administration, e.g. intrathecal or intracerebroventricular administration. Administration can be continuous, or chronic, or short or intermittent.

“Peptide” means a molecule formed by linking at least two amino acids by amide bonds. Peptide refers to polypeptides and proteins.

“Pharmaceutical agent” means a substance that provides a therapeutic benefit when administered to an individual. For example, in certain embodiments, an antisense oligonucleotide targeted to FGFR4 is pharmaceutical agent.

“Pharmaceutical composition” means a mixture of substances suitable for administering to an individual. For example, a pharmaceutical composition can comprise one or more active agents and a sterile aqueous solution.

“Pharmaceutically acceptable carrier” means a medium or diluent that does not interfere with the structure of the oligonucleotide. Certain, of such carries enable pharmaceutical compositions to be formulated as, for example, tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspension and lozenges for the oral ingestion by a subject. For example, a pharmaceutically acceptable carrier can be a sterile aqueous solution.

“Pharmaceutically acceptable derivative” encompasses pharmaceutically acceptable salts, conjugates, prodrugs or isomers of the compounds described herein.

“Pharmaceutically acceptable salts” means physiologically and pharmaceutically acceptable salts of antisense compounds, i.e., salts that retain the desired biological activity of the parent oligonucleotide and do not impart undesired toxicological effects thereto.

“Phosphorothioate linkage” means a linkage between nucleosides where the phosphodiester bond is modified by replacing one of the non-bridging oxygen atoms with a sulfur atom. A phosphorothioate linkage is a modified internucleoside linkage.

“Portion” means a defined number of contiguous (i.e. linked) nucleobases of a nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of a target nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of an antisense compound.

“Prevent” refers to delaying or forestalling the onset or development of a disease, disorder, or condition for a period of time from minutes to indefinitely. Prevent also means reducing risk of developing a disease, disorder, or condition.

“Prodrug” means a therapeutic agent that is prepared in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals or conditions.

“Side effects” means physiological responses attributable to a treatment other than the desired effects. In certain embodiments, side effects include injection site reactions, liver function test abnormalities, renal function abnormalities, liver toxicity, renal toxicity, central nervous system abnormalities, myopathies, and malaise. For example, increased aminotransferase levels in serum can indicate liver toxicity or liver function abnormality. For example, increased bilirubin can indicate liver toxicity or liver function abnormality.

“Single-stranded oligonucleotide” means an oligonucleotide which is not hybridized to a complementary strand.

“Specifically hybridizable” refers to an antisense compound having a sufficient degree of complementarity between an antisense oligonucleotide and a target nucleic acid to induce a desired effect, while exhibiting minimal or no effects on non-target nucleic acids under conditions in which specific binding is desired, i.e. under physiological conditions in the case of in vivo assays and therapeutic treatments.

“Statin” means an agent that inhibits the activity of HMG-CoA reductase.

“Subcutaneous administration” means administration just below the skin.

“Targeting” or “targeted” means the process of design and selection of an antisense compound that will specifically hybridize to a target nucleic acid and induce a desired effect.

“Target nucleic acid,” “target RNA,” and “target RNA transcript” all refer to a nucleic acid capable of being targeted by antisense compounds.

“Target segment” means the sequence of nucleotides of a target nucleic acid to which an antisense compound is targeted. “5′ target site” refers to the 5′-most nucleotide of a target segment. “3′ target site” refers to the 3′-most nucleotide of a target segment.

“Therapeutically effective amount” means an amount of an agent that provides a therapeutic benefit to an individual.

“Therapeutic lifestyle change” means dietary and lifestyle changes intended to lower fat/adipose tissue mass and/or cholesterol. Such change can reduce the risk of developing heart disease, and may includes recommendations for dietary intake of total daily calories, total fat, saturated fat, polyunsaturated fat, monounsaturated fat, carbohydrate, protein, cholesterol, insoluble fiber, as well as recommendations for physical activity.

“Triglyceride” or “TG” means a lipid or neutral fat consisting of glycerol combined with three fatty acid molecules.

“Type 2 diabetes,” (also known as “type 2 diabetes mellitus” or “diabetes mellitus, type 2”, and formerly called “diabetes mellitus type 2”, “non-insulin-dependent diabetes (NIDDM)”, “obesity related diabetes”, or “adult-onset diabetes”) is a metabolic disorder that is primarily characterized by insulin resistance, relative insulin deficiency, and hyperglycemia.

“Treat” refers to administering a pharmaceutical composition to an animal to effect an alteration or improvement of a disease, disorder, or condition.

“Unmodified nucleotide” means a nucleotide composed of naturally occurring nucleobases, sugar moieties, and internucleoside linkages. In certain embodiments, an unmodified nucleotide is an RNA nucleotide (i.e. β-D-ribonucleosides) or a DNA nucleotide (i.e. β-D-deoxyribonucleoside).

Certain Embodiments

Certain embodiments provide methods, compounds, and compositions for inhibiting FGFR4 expression.

Certain embodiments provide antisense compounds targeted to a FGFR4 nucleic acid. In certain embodiments, the FGFR4 nucleic acid is any of the sequences set forth in GENBANK Accession No. NM002011.3 (incorporated herein as SEQ ID NO: 1), GENBANK Accession No: NT023133.11 truncated from nucleosides 21323018 to 21335213 (incorporated herein as SEQ ID NO: 2); and GENBANK Accession No. AB209631.1 (incorporated herein as SEQ ID NO: 3); and GENBANK Accession No NM022963.2 (incorporated herein as SEQ ID NO: 4). In certain embodiments, FGFR4 has the rhesus monkey sequence as set forth in GENBANK Accession No. NW001121000.1 truncated from nucleosides 3094000 to 3109000 (SEQ ID NO: 5). In certain embodiments, FGFR4 has the murine sequence as set forth in GENBANK Accession No. BC033313.1 (SEQ ID NO: 6).

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 12 to 30 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-6.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 12 to 30 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein consist of 12 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of any of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein can consist of 12 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of any of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compound or composition provided herein is or comprises ISIS NOs: 463588, 463589, 463690, 463691, 463835, 463837, or 464225.

In certain embodiments, the compounds or compositions provided herein consist of 12 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein consist of 12 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compound or composition is or comprises ISIS NO: 463588.

In certain embodiments, the compound or composition is or comprises ISIS NO: 463690.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 15 to 30 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein consist of 15 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of any of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein consist of 15 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of any of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compound or composition provided herein is or comprises ISIS NOs: 463588, 463589, 463690, 463691, 463835, 463837, or 464225.

In certain embodiments, the compounds or compositions provided herein consist of 15 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein consist of 15 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compound or composition provided herein is or comprise ISIS NO: 463588.

In certain embodiments, the compound or composition provided herein is or comprise ISIS NO: 463690.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 18 to 21 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 18 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of any of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 18 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of any of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 18 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 18 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 35 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 35 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 35 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein can consist of 20 to 35 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein can consist of 20 to 35 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 30 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 30 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 25 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 25 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 25 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 25 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 25 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions described herein comprise a modified oligonucleotide consisting of 20 to 24 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 24 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 24 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 24 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 24 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 23 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 23 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 23 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 23 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 23 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 22 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 22 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 22 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 221 inked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 22 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 21 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 21 linked nucleosides and having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 to 21 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 nucleosides having a nucleobase sequence complementary to an equal length portion of any of SEQ ID NOs: 1-4.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NOs: 7-322.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 16.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides and have a nucleobase sequence comprising at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 contiguous nucleobases of SEQ ID NO: 45.

In certain embodiments, the compounds or compositions provided herein comprise a salt of the modified oligonucleotide.

In certain embodiments, the compounds or compositions provided herein further comprise a pharmaceutically acceptable carrier or diluent.

In certain embodiments, the nucleobase sequence of the modified oligonucleotide is at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% complementary to any one of SEQ ID NOs: 1-4 as measured over the entirety of the modified oligonucleotide.

In certain embodiments, the nucleobase sequence of the modified oligonucleotide has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identity to any one of SEQ ID NOs: 7-322 as measured over the entirety of the modified oligonucleotide.

In certain embodiments, the nucleobase sequence of the modified oligonucleotide has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identity to any one of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138 as measured over the entirety of the modified oligonucleotide.

In certain embodiments, the nucleobase sequence of the modified oligonucleotide has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NO: 16 as measured over the entirety of the modified oligonucleotide.

In certain embodiments, the nucleobase sequence of the modified oligonucleotide has at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identity to SEQ ID NO: 45 as measured over the entirety of the modified oligonucleotide.

In certain embodiments, antisense compounds or modified oligonucleotides targets a region of a FGFR4 nucleic acid. In certain embodiments, such compounds or oligonucleotides targeted to a region of a FGFR4 nucleic acid have a contiguous nucleobase portion that is complementary to an equal length nucleobase portion of the region. For example, the portion can be at least an 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 contiguous nucleobases portion complementary to an equal length portion of a region recited herein. In certain embodiments, such compounds or oligonucleotide target the following nucleotide regions of SEQ ID NO: 1: 160-179, 191-210, 191-211, 191-212, 191-213, 192-211, 192-212, 192-213, 193-212, 193-213, 194-213, 196-215, 196-216, 197-216, 200-219, 202-221, 202-222, 203-222, 290-310, 290-309, 290-311, 290-312, 290-312, 291-310, 291-311, 291-312, 292-311, 292-312, 293-312, 309-328, 332-351, 338-357, 338-358, 339-358, 347-366, 349-368, 357-376, 368-387, 368-388, 368-389, 368-390, 368-391, 369-380, 369-389, 369-390, 369-391, 370-389, 370-390, 370-391, 371-390, 371-391, 372-391, 388-407, 388-408, 389-408, 392-411, 404-423, 431-450, 431-451, 432-451, 443-462, 443-463, 444-463, 601-620, 624-643, 734-753, 757-806, 787-807, 788-807, 790-809, 790-810, 791-810, 970-989, 1024-1043, 1024-1044, 1024-1045, 1024-1046, 1024-1047, 1024-1048, 1024-1105, 1025-1044, 1025-1045, 1025-1046, 1025-1047, 1025-1048, 1026-1045, 1026-1046, 1026-1047, 1026-1048, 1027-1046, 1027-1047, 1027-1048, 1028-1047, 1028-1048, 1029-1048, 1031-1050, 1031-1051, 1032-1051, 1084-1103, 1084-1105, 1086-1105, 1097-1116, 1097-1117, 1097-1122, 1100-1119, 1100-1120, 1100-1121, 1100-1122, 1101-1120, 1101-1121, 1101-1122, 1102-1121, 1102-1122, 1103-1122, 1105-1124, 1105-1125, 1106-1125, 1110-1029, 1110-1130, 1111-1130, 1115-1134, 1185-1204, 1255-1274, 1290-1309, 1290-1310, 1291-1310, 1301-1320, 1417-1436, 1468-1487, 1468-1488, 1469-1488, 1559-1578, 1562-1581, 1564-1583, 1619-1638, 2325-2344, 2325-2345, 2326-2345, 2438-2457, 2812-2831, 2816-2835, 2816-2836, 2816-2837, 2816-2838, 2817-2836, 2817-2837, 2817-2838, 2818-2837, 2818-2838, 2819-2838, 2822-2481, 2822-2842, 2822-2843, 2822-2844, 2823-2842, 2823-2843, 2823-2844, 2824-2843, 2824-2844, 2825-2844, 2951-2970, 2951-2971, 2951-2972, 2951-2973, 2951-2974, 2951-2975, 2951-3000, 2952-2971, 2952-2972, 2952-2973, 2952-2974, 2952-2975, 2953-2972, 2953-2973, 2953-2974, 2953-2975, 2954-2973, 2954-2974, 2954-2975, 2955-2974, 2955-2975, 2956-2975.

In certain embodiments, antisense compounds or modified oligonucleotides targets a region of a FGFR4 nucleic acid. In certain embodiments, such compounds or oligonucleotides targeted to a region of a FGFR4 nucleic acid have a contiguous nucleobase portion that is complementary to an equal length nucleobase portion of the region. For example, the portion can be at least an 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 contiguous nucleobases portion complementary to an equal length portion of a region recited herein. In certain embodiments, such compounds or oligonucleotide target the following nucleotide regions of SEQ ID NO: 2: 3165-3184, 3196-3215, 3197-3216, 3196-3217, 3196-3218, 3197-3216, 3197-3217, 3197-3218, 3198-3217, 3198-3218, 3199-3218, 3201-3220, 3201-3221, 3202-3221, 3205-3224, 3207-3226, 3207-3227, 3208-3227, 3991-4011, 3991-4010, 3991-4012, 3991-4013, 3992-4011, 3992-4012, 3992-4013, 3993-4012, 3993-4013, 3994-4013, 4010-4029, 4033-4052, 4039-4058, 4039-4059, 4040-4059, 4048-4067, 4050-4069, 4058-4077, 4069-4088, 4069-4089, 4069-4091, 4069-4091, 4069-4092, 4070-4091, 4070-4090, 4070-4091, 4070-4092, 4071-4090, 4071-4091, 4071-4092, 4072-4091, 4072-4092, 4073-4092, 4089-4108, 4089-4109, 4090-4109, 4093-4112, 4105-4124, 4132-4151, 4132-4152, 4133-4152, 4144-4163, 4144-4164, 4145-4164, 4506-4522, 4528-4547, 4638-4657, 5268-5290, 5271-5291, 5272-5291, 5274-5293, 5274-5294, 5275-5294, 5966-5985, 6020-6039, 6020-6040, 6020-6041, 6020-6042, 6020-6043, 6020-6044, 6020-6235, 6021-6040, 6021-6041, 6021-6042, 6021-6043, 6021-6044, 6022-6041, 6022-6042, 6022-6043, 6022-6044, 6023-6042, 6023-6043, 6023-6044, 6024-6043, 6024-6044, 6025-6044, 6027-6046, 6027-6047, 6028-6047, 6214-6235, 6214-6233, 6214-6235, 6216-6235, 6227-6246, 6227-6247, 6227-6252, 6230-6249, 6230-6250, 6230-6251, 6230-6252, 6231-6250, 6231-6251, 6231-6252, 6232-6251, 6232-6252, 6233-6252, 6235-6254, 6235-6255, 6236-6255, 6241-6260, 6245-6264, 6315-6334, 6784-6803, 6974-6993, 7025-7044, 7025-7045, 7026-7045, 7059-7081, 7221-7240, 7223-7242, 7278-7297, 10866-10885, 10866-10866, 10867-10886, 11108-11127, 11482-11501, 11486-11505, 11486-11506, 11486-11507, 11486-11508, 11487-11506, 11487-11507, 11487-11508, 11488-11507, 11488-11508, 11489-11508, 11492-11511, 11492-11512, 11492-11513, 11492-1151, 11493-11512, 11493-11513, 11493-11514, 11494-11513, 11494-11514, 11495-11514, 11621-11640, 11621-11641, 11621-11642, 11621-11643, 11621-11644, 11621-11645, 11621-11670, 11622-11641, 11622-11642, 11622-11643, 11622-11644, 11622-11645, 11623-11642, 11623-11643, 11623-11644, 11623-11645, 11624-11643, 11624-11644, 11624-11645, 11625-11644, 11625-11645, 11626-11645.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides wherein the linked nucleosides comprise at least an 8 contiguous nucleobase portion that is complementary to an equal length nucleobase portion within the region selected from nucleotides 191-210 or 369-388 of SEQ ID NO: 1. In certain embodiments, the modified oligonucleotide has at least a 9, at least a 10, at least an 11, at least a 12, at least a 13, at least a 14, at least a 15, at least a 16, at least a 17, at least an 18, at least 19 or at least a 20 contiguous nucleobase portion of which is complementary to an equal length portion within the region selected from nucleotides 191-210 or 369-388 of SEQ ID NO: 1. In certain embodiments, the modified oligonucleotide is 90%, 95%, 99%, or 100% complementary to a nucleic acid encoding human FGFR4, eg. SEQ ID No: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides wherein the linked nucleosides comprise at least an 8 contiguous nucleobase portion that is complementary to an equal length nucleobase portion within the region selected from nucleotides 3196-3215 or 4070-4089 of SEQ ID NO: 2. In certain embodiments, the modified oligonucleotide has at least a 9, at least a 10, at least an 11, at least a 12, at least a 13, at least a 14, at least a 15, at least a 16, at least a 17, at least an 18, at least 19 or at least a 20 contiguous nucleobase portion of which is complementary to an equal length portion within the region selected from nucleotides 3196-3215 or 4070-4089 of SEQ ID NO: 2. In certain embodiments, the modified oligonucleotide is 90%, 95%, 99%, or 100% complementary to a nucleic acid encoding human FGFR4, eg. SEQ ID No: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 60% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 788-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 60% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 70% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 188-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 70% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 80% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 188-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 80% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 90% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 188-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 90% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 95% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 188-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 95% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 188-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 100% complementary within the region selected from nucleotides 191-210, 193-212, 369-388, 370-389, 188-807, 790-809 and 2954-2973 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 100% complementary within the region selected from nucleotides 3196-3215, 3198-3217, 4070-4089, 4071-4090, 5272-5291, 5274-5293, and 11624-11643 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 60% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 60% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 70% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 70% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 80% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 80% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 90% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 90% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 95% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 95% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 100% complementary within nucleotides 191-210 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 100% complementary within the region selected from nucleotides 3196-3215 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 60% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 60% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 70% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 70% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 80% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 80% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 90% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 90% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 95% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 95% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 100% complementary within nucleotides 369-388 of SEQ ID NO: 1.

Certain embodiments provide compounds comprising a modified oligonucleotide consisting of 20 linked nucleosides 99% complementary within nucleotides 4070-4089 of SEQ ID NO: 2.

In certain embodiments, such compounds or oligonucleotides targeted to a region of a FGFR4 nucleic acid have a contiguous nucleobase portion that is complementary to an equal length nucleobase portion of the region 191-210 or 369-388 of SEQ ID NO: 1.

In certain embodiments, such compounds or oligonucleotides targeted to a region of a FGFR4 nucleic acid have a contiguous nucleobase portion that is complementary to an equal length nucleobase portion of the region 3196-3215 or 4070-4089 of SEQ ID NO: 2.

In certain embodiments, the following nucleotide regions of SEQ ID NO: 1, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 160-179, 191-210, 191-211, 191-212, 191-213, 192-211, 192-212, 192-213, 193-212, 193-213, 194-213, 196-215, 196-216, 197-216, 200-219, 202-221, 202-222, 203-222, 290-210, 290-309, 290-311, 290-312, 290-312, 291-310, 291-311, 291-312, 292-311, 292-312, 293-312, 309-328, 332-351, 338-357, 338-358, 339-358, 347-366, 349-368, 357-376, 368-387, 368-388, 368-389, 368-390, 368-391, 369-380, 369-389, 369-390, 369-391, 370-389, 370-390, 370-391, 371-390, 371-391, 372-391, 388-407, 388-408, 389-408, 392-411, 404-423, 431-450, 431-451, 432-451, 443-462, 443-463, 444-463, 601-620, 624-643, 734-753, 767-806, 787-807, 788-807, 790-809, 790-810, 791-810, 970-989, 1024-1043, 1024-1044, 1024-1045, 1024-1046, 1024-1047, 1024-1048, 1024-1105, 1025-1044, 1025-1045, 1025-1046, 1025-1047, 1025-1048, 1026-1045, 1026-1046, 1026-1047, 1026-1048, 1027-1046, 1027-1047, 1027-1048, 1028-1047, 1028-1048, 1029-1048, 1031-1050, 1031-1051, 1032-1051, 1074-1051, 1084-1103, 1084-1105, 1086-1105, 1097-1116, 1097-1117, 1097-1122, 1100-1119, 1100-1119, 1100-1120, 1100-1121, 1100-1122, 1101-1120, 1101-1121, 1101-1122, 1102-1121, 1102-1122, 1103-1122, 1105-1124, 1105-1125, 1106-1125, 1110-1029, 1110-1130, 1111-1130, 1115-1134, 1185-1204, 1255-1274, 1290-1309, 1290-1310, 1291-1310, 1301-1320, 1417-1436, 1468-1487, 1468-1488, 1469-1488, 1559-1578, 1562-1581, 1564-1583, 1619-1638, 2325-2344, 2325-2345, 2326-2345, 2438-2457, 2812-2831, 2816-2835, 2816-2836, 2816-2837, 2816-2838, 2817-2836, 2817-2837, 2817-2838, 2818-2837, 2818-2838, 2819-2838, 2822-2481, 2822-2842, 2822-2843, 2822-2844, 2822-2844, 2823-2842, 2823-2843, 2823-2844, 2824-2843, 2824-2844, 2825-2844, 2951-2970, 2951-2971, 2951-2972, 2951-2973, 2951-2974, 2951-2975, 2951-2975, 2951-3000, 2952-2971, 2952-2972, 2952-2973, 2952-2974, 2952-2975, 2953-2972, 2953-2973, 2953-2974, 2953-2975, 2954-2973, 2954-2974, 2954-2975, 2955-2974, 2955-2975, and 2956-2975.

In certain embodiments, the following nucleotide regions of SEQ ID NO: 2, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 3165-3184, 3196-3215, 3196-3216, 3196-3217, 3196-3218, 3197-3216, 3197-3217, 3197-3218, 3198-3217, 3198-3218, 3199-3218, 3201-3220, 3201-3221, 3202-3221, 3205-3224, 3207-3226, 3207-3227, 3208-3227, 3991-4011, 3991-4010, 3991-4012, 3991-4013, 3991-4014, 3992-4011, 3992-4012, 3992-4013, 3993-4012, 3993-4013, 3994-4013, 4010-4029, 4033-4052, 4039-4058, 4039-4059, 4040-4059, 4048-4067, 4050-4069, 4058-4077, 4069-4088, 4069-4089, 4069-4090, 4069-4091, 4069-4092, 4070-380, 4070-4090, 4070-4091, 4070-4092, 4071-4090, 4071-4091, 4071-4092, 4072-4091, 4072-4092, 4073-4092, 4089-4108, 4089-4109, 4090-4109, 4093-4112, 4105-4124, 4132-4151, 4132-4152, 4133-4152, 4144-4163, 4144-4164, 4145-4164, 4506-4522, 4528-4547, 4638-4657, 5268-5290, 5271-5291, 5272-5291, 5274-5293, 5274-5294, 5275-5294, 5966-5985, 6020-6039, 6020-6040, 6020-6041, 6020-6042, 6020-6043, 6020-6044, 6020-6045, 6021-6040, 6021-6041, 6021-6042, 6021-6043, 6021-6044, 6022-6041, 6022-6042, 6022-6043, 6022-6044, 6023-6042, 6023-6043, 6023-6047, 6024-6043, 6024-6044, 6025-6044, 6027-6046, 6027-6047, 6028-6047, 6214-6235, 6214-6233, 6214-6235, 6216-6235, 6227-6246, 6227-6247, 6227-6252, 6230-6249, 6230-6249, 6230-6250, 6230-6251, 6230-6252, 6231-6250, 6231-6251, 6231-6252, 6232-6251, 6232-6252, 6233-6252, 6235-6254, 6235-6255, 6236-6255, 6230-6260, 6241-6260, 6245-6264, 6315-6334, 6784-6803, 6974-6993, 7025-7044, 7025-7045, 7026-7045, 7221-7240, 7223-7242, 7278-7297, 10866-10885, 10866-10886, 10867-10886, 11008-11127, 11482-11501, 11486-11505, 11486-11506, 11486-11507, 11486-11508, 11487-11506, 11487-11507, 11487-11508, 11488-11507, 11488-11508, 11489-11508, 11492-11511, 11492-11512, 11492-11513, 11492-11514, 11493-11512, 11493-11513, 11493-11514, 11494-11513, 11494-11514, 11495-11514, 11621-11640, 11621-11641, 11621-11642, 11621-11643, 11621-11644, 11621-11645, 11621-11670, 11622-11641, 11622-11642, 11622-11643, 11622-11644, 11622-11645, 11623-11642, 11623-11643, 11623-11644, 11623-1645, 11624-11643, 11624-11644, 11624-11645, 11625-11644, 11625-11645, and 11626-11645.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NO: 1, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 59, 61, 62, 64, 65, 66, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, and 116.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NO: 1, when targeted by antisense compounds or oligonucleotides, displays at least 70% inhibition: 7, 14, 15, 16, 17, 18, 19, 20, 22, 23, 24, 27, 28, 29, 30, 32, 33, 34, 35, 38, 39, 43, 44, 45, 46, 47, 48, 49, 50, 51, 54, 59, 61, 64, 69, 70, 72, 73, 75, 77, 78, 79, 80, 81, 82, 83, 85, 86, 87, 89, 90, 91, 92, 94, 97, 98, 103, 105, 106, 111, 112, 113, and 116.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NO: 1, when targeted by antisense compounds or oligonucleotides, displays at least 75% inhibition: 7, 14, 16, 17, 22, 24, 28, 29, 30, 32, 33, 34, 39, 43, 44, 45, 46, 47, 49, 50, 59, 61, 69, 70, 72, 73, 75, 77, 78, 79, 80, 83, 85, 89, 90, 91, 92, 105, 106, 111, and 112.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NO: 1, when targeted by antisense compounds or oligonucleotides, displays at least 80% inhibition: 7, 14, 16, 17, 28, 29, 33, 39, 45, 47, 49, 50, 72, 80, 90, 91, and 106.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NO: 1, when targeted by antisense compounds or oligonucleotides, displays at least 85% inhibition 7, 14, 16, 29, 45, 50, 80, 90, and 91.

In certain embodiments, the following nucleotide regions of SEQ ID NOs: 1 or 2, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 908-927, 992-1011, 1138-1157, 1138-1161, 1142-1161, 1345-1364, 1386-1405, 1386-1413, 1394-1413, 1461-1480, 1461-1482, 1461-1484, 1461-1486, 1461-1490, 1463-1482, 1463-1484, 1463-1486, 1463-1490, 1465-1484, 1465-1486, 1465-1490, 1467-1486, 1467-1490, 1471-1490, 1542-1561, 1941-1960, 1941-1962, 1941-1964, 1943-1962, 1943-1964, 1945-1964, 2053-2072, 2104-2123, 2104-2125, 2104-2127, 2104-2129, 2104-2131, 2104-2133, 2104-2135, 2104-2137, 2106-2125, 2106-2127, 2106-2129, 2106-2131, 2106-2133, 2106-2135, 2106-2137, 2108-2127, 2108-2129, 2108-2131, 2108-2133, 2108-2135, 2108-2137, 2110-2129, 2110-2131, 2110-2133, 2110-2135, 2110-2137, 2112-2131, 2112-2133, 2112-2135, 2112-2137, 2114-2133, 2114-2135, 2114-2137, 2116-2135, 2116-2137, 2118-2137, 2271-2290, 2838-2857, 3122-3141, 3122-3144, 3125-3144, 3165-3184, 3325-3344, 3325-3346, 3325-3348, 3325-3350, 3325-3352, 3325-3354, 3325-3356, 3325-3358, 3325-3360, 3325-3362, 3325-3362, 3327-3346, 3327-3346, 3327-3348, 3327-3350, 3327-3352, 3327-3354, 3327-3356, 3327-3358, 3327-3360, 3327-3362, 3329-3348, 3329-3348, 3329-3350, 3329-3352, 3329-3354, 3329-3356, 3329-3358, 3329-3360, 3329-3362, 3331-3350, 3331-3352, 3331-3354, 3331-3356, 3331-3358, 3331-3360, 3331-3362, 3333-3352, 3333-3354, 3333-3356, 3333-3358, 3333-3360, 3333-3362, 3335-3354, 3335-3356, 3335-3358, 3335-3360, 3335-3362, 3337-3356, 3337-3358, 3337-3360, 3337-3362, 3339-3358, 3339-3360, 3339-3362, 3341-3360, 3341-3362, 3343-3362, 3386-3405, 3386-3413, 3386-3417, 3386-3419, 3386-3423, 3386-3427, 3386-3434, 3386-3434, 3394-3413, 3394-3417, 3394-3423, 3394-3427, 3394-3434, 3398-3417, 3398-3419, 3398-3423, 3398-3427, 3398-3434, 3400-3419, 3400-3423, 3400-3427, 3400-3434, 3404-3423, 3404-3427, 3404-3434, 3408-3427, 3408-3434, 3415-3434, 3445-3464, 3445-3466, 3445-3468, 3445-3470, 3447-3466, 3447-3468, 3447-3470, 3449-3468, 3449-3470, 3451-3470, 3499-3518, 3571-3590, 3571-3592, 3571-3594, 3571-3596, 3571-3598, 3573-3592, 3573-3594, 3573-3596, 3573-3598, 3575-3594, 3575-3596, 3575-3598, 3577-3596, 3577-3598, 3579-3598, 3772-3791, 3772-3793, 3772-3795, 3772-3797, 3772-3801, 3772-3807, 3772-3817, 3774-3793, 3774-3795, 3774-3797, 3776-3795, 3776-3797, 3778-3797, 3782-3801, 3782-3817, 3788-3807, 3788-3817, 3798-3817, 3993-4012, 4799-4818, 7684-7703, 7690-7709, 7692-7711.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 29, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, and 235.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2, when targeted by antisense compounds or oligonucleotides, displays at least 70% inhibition: 29, 117, 119, 120, 122, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 145, 146, 147, 150, 151, 152, 153, 154, 155, 156, 157, 158, 160, 161, 162, 163, 164, 165, 166, 167, 169, 170, 171, 174, 180, 183, 184, 185, 186, 187, 188, 189, 190, 193, 195, 198, 199, 200, 201, 202, 203, 206, 207, 208, 209, 210, 211, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 225, 226, 227, 228, 229, 231, 233, 234, and 235.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2, when targeted by antisense compounds or oligonucleotides, displays at least 75% inhibition: 29, 117, 120, 128, 129, 131, 132, 133, 135, 136, 137, 138, 139, 140, 141, 146, 152, 153, 154, 155, 156, 160, 161, 162, 163, 164, 165, 166, 167, 169, 174, 180, 186, 187, 188, 198, 199, 201, 202, 207, 208, 209, 213, 214, 215, 216, 217, 219, 220, 221, 223, 225, 227, 228, 229, 231, 233, and 235.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2, when targeted by antisense compounds or oligonucleotides, displays at least 80% inhibition: 29, 117, 131, 132, 133, 135, 136, 137, 138, 140, 141, 152, 153, 154, 155, 156, 160, 162, 163, 164, 174, 186, 187, 188, 199, 201, 202, 207, 208, 213, 214, 215, 216, 217, 219, 220, 221, 223, 227, 229, 231, and 233.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2, when targeted by antisense compounds or oligonucleotides, displays at least 85% inhibition: 29, 117, 132, 135, 136, 140, 141, 154, 156, 163, 164, 187, 188, 199, 201, 215, 216, 217, 219, 220, 221, 223, 227, 229, 231, and 233.

In certain embodiments, the following nucleotide regions of SEQ ID NOs: 1 or 2 or 3, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 101-120, 101-122, 101-124, 101-125, 101-126, 101-127, 102-126, 103-122, 103-124, 103-125, 103-126, 103-127, 105-124, 105-125, 105-127, 106-125, 106-126, 106-127, 107-126, 107-127, 108-127, 1122-1141, 1165-1184, 1193-1218, 1198-1217, 1199-1218, 1323-1342, 1323-1344, 1323-1346, 1323-1347, 1323-1352, 1325-1344, 1325-1346, 1325-1347, 1327-1346, 1327-1347, 1328-1347, 1333-1352, 1333-1354, 1333-1354, 1333-1356, 1333-1358, 1333-1360, 1335-1354, 1335-1356, 1335-1358, 1335-1360, 1337-1356, 1337-1358, 1337-1360, 1339-1358, 1339-1360, 1341-1360, 1392-1411, 1392-1417, 1393-1412, 1394-1413, 1394-1415, 1394-1417, 1396-1415, 1396-1417, 1398-1417, 1413-1432, 1413-1433, 1413-1434, 1414-1433, 1414-1434, 1415-1434, 1445-1464, 1445-1466, 1445-1468, 1445-1470, 1445-1471, 1447-1466, 1447-1468, 1447-1470, 1447-1471, 1449-1468, 1449-1470, 1449-1471, 1451-1470, 1451-1471, 1452-1471, 1462-1481, 1462-1481, 1462-1482, 1462-1483, 1462-1484, 1462-1485, 1462-1487, 1463-1482, 1463-1482, 1463-1483, 1463-1484, 1463-1485, 1463-1487, 1464-1483, 1464-1484, 1464-1485, 1464-1487, 1465-1484, 1465-1485, 1465-1487, 1466-1485, 1466-1487, 1468-1487, 1501-1521, 1501-1522, 1503-1522, 1569-1588, 1569-1589, 1569-1590, 1569-1591, 1569-1592, 1569-1593, 1569-1594, 1569-1596, 1569-1598, 1570-1589, 1570-1590, 1570-1591, 1570-1592, 1570-1593, 1570-1594, 1570-1596, 1570-1598, 1571-1590, 1571-1591, 1571-1591, 1571-1592, 1571-1592, 1571-1593, 1571-1593, 1571-1594, 1571-1594, 1571-1596, 1571-1596, 1571-1598, 1571-1598, 1572-1591, 1572-1592, 1572-1593, 1572-1594, 1572-1596, 1572-1598, 1573-1592, 1573-1593, 1573-1594, 1573-1596, 1573-1598, 1574-1593, 1574-1594, 1574-1596, 1574-1598, 1575-1594, 1575-1596, 1575-1598, 1577-1596, 1577-1598, 1579-1598, 1778-1797, 1778-1799, 1778-1805, 1778-1809, 1778-1811, 1778-1821, 1780-1799, 1786-1805, 1790-1809, 1790-1811, 1790-1821, 1792-1811, 1792-1821, 1802-1821, 1944-1963, 1996-2015, 2053-2072, 2074-2093, 2418-2437, 4988-5007, 5120-5139, 5121-5140, 5121-5146, 5122-5141, 5122-5142, 5122-5143, 5122-5144, 5122-5146, 5123-5142, 5123-5143, 5123-5144, 5123-5146, 5124-5143, 5124-5144, 5124-5144, 5124-5146, 5125-5146, 5127-5146, 5150-5169, 5150-5170, 5150-5171, 5151-5170, 5151-5171, 5152-5171, 7801-7820, 7801-7822, 7803-7822.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2 or 3, when targeted by antisense compounds or oligonucleotides, displays at least 65% inhibition: 29, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, and 322.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2 or 3, when targeted by antisense compounds or oligonucleotides, displays at least 70% inhibition: 29, 239, 240, 241, 242, 243, 244, 245, 247, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 268, 269, 270, 271, 272, 274, 275, 276, 277, 278, 279, 280, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 294, 295, 296, 297, 298, 299, 300, 301, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 317, 319, 320, and 322.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2 or 3, when targeted by antisense compounds or oligonucleotides, displays at least 75% inhibition: 29, 239, 240, 241, 242, 243, 244, 245, 247, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 264, 266, 269, 271, 272, 274, 275, 276, 277, 278, 279, 283, 284, 286, 287, 288, 291, 298, 299, 300, 305, 306, 307, 308, 309, 310, 312, 313, 315, 317, 319, and 320.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2 or 3, when targeted by antisense compounds or oligonucleotides, displays at least 80% inhibition: 29, 241, 242, 243, 244, 245, 247, 250, 253, 254, 255, 256, 259, 260, 261, 262, 264, 266, 271, 272, 274, 276, 278, 283, 284, 286, 287, 299, 300, 305, 306, 307, 308, 310, 312, 313, 317, and 320.

In certain embodiments, the nucleobase sequences recited in the following SEQ ID NOs of SEQ ID NOs: 1 or 2 or 3, when targeted by antisense compounds or oligonucleotides, displays at least 85% inhibition: 29, 241, 242, 243, 244, 247, 254, 256, 259, 260, 264, 272, 278, 299, 300, 305, 306, 307, 308, 310, 317, and 320.

In certain embodiments, the following antisense compounds target a region of SEQ ID NO: 1, a nucleic acid encoding human FGFR4, and demonstrate at least 65% inhibition of a FGFR4 mRNA: ISIS NOs: 299005, 299010, 299018, 299022, 299024, 299025, 299028, 299029, 299030, 463588, 463589, 463590, 463592, 463593, 463594, 463596, 463598, 463599, 463601, 463625, 463627, 463628, 463629, 463630, 463636, 463645, 463648, 463654, 463655, 463656, 463657, 463670, 463672, 463673, 463677, 463678, 463679, 463689, 463690, 463691, 463692, 463693, 463708, 463709, 463712, 463717, 463718, 463724, 463733, 463734, 463735, 463751, 463763, 463770, 463774, 463791, 463805, 463832, 463834, 463835, 463836, 463837, 463838, 463860, 463861, 463871, 463874, 463875, 463876, 463877, 463878, 463880, 463882, 463883, 463884, 463893, 463894, 463906, 463907, 463908, 463909, 463910, 463912, 463913, 463918, 463919, 463922, 463937, 463938, 463947, 463967, 463994, 464002, 464004, 464013, 464014, 464015, 464030, 464033, 464037, 464038, 464041, 464043, 464046, 464048, and 464049.

In certain embodiments, the following antisense compounds target a region of SEQ ID NO: 1, a nucleic acid encoding human FGFR4 and demonstrate at least 70% inhibition of a FGFR4 mRNA: ISIS NOs: 299005, 299029, 299030, 463588, 463589, 463590, 463592, 463593, 463596, 463598, 463599, 463627, 463628, 463629, 463630, 463645, 463648, 463654, 463655, 463670, 463672, 463679, 463689, 463690, 463691, 463692, 463693, 463708, 463709, 463712, 463724, 463751, 463763, 463774, 463834, 463835, 463837, 463838, 463861, 463874, 463875, 463876, 463877, 463878, 463880, 463882, 463884, 463893, 463894, 463907, 463908, 463909, 463910, 463913, 463922, 463937, 464002, 464013, 464014, 464038, 464041, 464043, and 464049.

In certain embodiments, the following antisense compounds target a region of SEQ ID NO: 1, a nucleic acid encoding human FGFR4 and demonstrate at least 75% inhibition of a FGFR4 mRNA: ISIS NOs: 299005, 299029, 463588, 463589, 463596, 463599, 463628, 463629, 463630, 463645, 463648, 463654, 463672, 463679, 463689, 463690, 463691, 463692, 463708, 463709, 463751, 463763, 463834, 463835, 463837, 463838, 463861, 463874, 463875, 463876, 463877, 463882, 463884, 463907, 463908, 463909, 463910, 464013, 464014, 464038, and 464041.

In certain embodiments, the following antisense compounds target a region of SEQ ID NO: 1, a nucleic acid encoding human FGFR4 and demonstrate at least 80% inhibition of a FGFR4 mRNA: ISIS NOs: 299005, 299029, 463588, 463589, 463628, 463629, 463648, 463672, 463690, 463692, 463708, 463709, 463837, 463877, 463908, 463909, and 464014.

In certain embodiments, the following antisense compounds target a region of SEQ ID NO: 1, a nucleic acid encoding human FGFR4 and demonstrate at least 85% inhibition of a FGFR4 mRNA: ISIS NOs: 299005, 299029, 463588, 463629, 463690, 463709, 463877, 463908, and 463909.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2, a nucleic acid encoding human FGFR4, and demonstrate at least 65% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 299004, 464138, 464139, 464167, 464168, 464170, 464173, 299055, 464181, 464203, 464207, 464208, 464209, 464210, 464213, 464214, 464215, 464216, 464222, 464223, 464224, 464225, 464226, 464227, 464228, 464238, 464239, 464254, 464258, 464266, 464268, 464269, 464270, 464278, 464280, 464284, 464285, 464286, 464287, 464288, 464290, 464291, 464292, 464298, 464299, 464300, 464308, 464309, 464310, 464311, 464333, 464342, 464425, 464428, 464429, 464430, 464433, 464449, 464453, 464568, 464569, 464575, 464576, 464579, 464581, 464582, 464584, 464585, 464586, 464587, 464588, 464589, 464590, 464591, 464593, 464617, 464622, 464623, 464657, 464658, 464677, 464682, 464683, 464684, 464685, 464686, 464687, 464688, 464689, 464692, 464696, 464698, 464699, 464701, 464703, 464705, 464706, 464707, 464708, 464709, 464710, 464711, 464716, 464717, 464718, 464719, 464720, 464726, 464727, 464728, 464729, 464730, 464732, 464734, 464735, 464736, 464740, 464800, and 464801.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2, a nucleic acid encoding human FGFR4 and demonstrate at least 70% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 299004, 464138, 464139, 464168, 464203, 464207, 464208, 464209, 464210, 464213, 464214, 464215, 464216, 464222, 464223, 464224, 464225, 464226, 464227, 464228, 464238, 464258, 464266, 464268, 464278, 464280, 464284, 464285, 464286, 464287, 464288, 464290, 464291, 464298, 464299, 464300, 464308, 464309, 464310, 464311, 464333, 464425, 464428, 464429, 464449, 464579, 464584, 464585, 464586, 464587, 464588, 464589, 464590, 464591, 464622, 464657, 464682, 464683, 464684, 464685, 464686, 464687, 464692, 464696, 464698, 464699, 464701, 464703, 464706, 464707, 464708, 464709, 464710, 464711, 464716, 464717, 464718, 464719, 464720, 464727, 464728, 464729, 464730, 464732, 464735, 464740, 464800, and 464801.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2, a nucleic acid encoding human FGFR4 and demonstrate at least 75% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 299004, 464139, 464208, 464209, 464213, 464214, 464215, 464222, 464223, 464224, 464225, 464226, 464227, 464228, 464266, 464284, 464285, 464286, 464287, 464288, 464298, 464299, 464300, 464308, 464309, 464310, 464311, 464333, 464425, 464449, 464579, 464587, 464588, 464589, 464682, 464683, 464685, 464686, 464696, 464698, 464699, 464706, 464707, 464708, 464709, 464710, 464716, 464717, 464718, 464720, 464727, 464729, 464730, 464732, 464735, 464740, and 464801.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2, a nucleic acid encoding human FGFR4 and demonstrate at least 80% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 299004, 464213, 464214, 464215, 464222, 464223, 464224, 464225, 464227, 464228, 464284, 464285, 464286, 464287, 464288, 464298, 464300, 464308, 464309, 464449, 464587, 464588, 464589, 464683, 464685, 464686, 464696, 464698, 464706, 464707, 464708, 464709, 464710, 464716, 464717, 464718, 464720, 464729, 464732, 464735, and 464740.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2, a nucleic acid encoding human FGFR4 and demonstrate at least 85% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 299004, 464214, 464222, 464223, 464227, 464228, 464286, 464288, 464308, 464309, 464588, 464589, 464683, 464685, 464708, 464709, 464710, 464716, 464717, 464718, 464720, 464729, 464732, 464735, and 464740.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs:1 or 2 or 3, a nucleic acid encoding human FGFR4, and demonstrate at least 65% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 479530, 479532, 479533, 479534, 479535, 479536, 479537, 479538, 479539, 479540, 479541, 479542, 479543, 479544, 479545, 479546, 479547, 479548, 479549, 479550, 479551, 479552, 479553, 479554, 479555, 479556, 479557, 479558, 479560, 479561, 479562, 479564, 479565, 479566, 479567, 479568, 479569, 479570, 479572, 479573, 479574, 479576, 479577, 479582, 479583, 479584, 479585, 479594, 479596, 479597, 479608, 479613, 479614, 479622, 479625, 479626, 479641, 479682, 479689, 479690, 479691, 479692, 479693, 479694, 479696, 479697, 479698, 479699, 479703, 479704, 479705, 479706, 479716, 479721, 479722, 479725, 479731, 479732, 479736, 479737, 479738, 479739, 479740, and 479741.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2 or 3, a nucleic acid encoding human FGFR4 and demonstrate at least 70% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 479530, 479532, 479533, 479534, 479535, 479536, 479537, 479539, 479541, 479542, 479543, 479544, 479545, 479546, 479547, 479548, 479549, 479550, 479551, 479552, 479553, 479554, 479555, 479556, 479557, 479558, 479561, 479562, 479564, 479565, 479566, 479568, 479569, 479570, 479572, 479573, 479574, 479576, 479582, 479583, 479584, 479585, 479594, 479596, 479597, 479608, 479613, 479614, 479626, 479641, 479682, 479689, 479690, 479691, 479692, 479693, 479697, 479698, 479699, 479703, 479704, 479705, 479706, 479716, 479721, 479722, 479725, 479731, 479736, 479738, 479739, and 479741.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2 or 3, a nucleic acid encoding human FGFR4 and demonstrate at least 75% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 479530, 479532, 479533, 479534, 479535, 479536, 479537, 479539, 479542, 479543, 479544, 479545, 479546, 479547, 479548, 479549, 479550, 479551, 479552, 479553, 479554, 479556, 479558, 479562, 479565, 479566, 479568, 479569, 479570, 479572, 479573, 479574, 479583, 479584, 479594, 479596, 479597, 479614, 479690, 479691, 479692, 479698, 479699, 479703, 479704, 479705, 479706, 479721, 479722, 479731, 479736, 479738, and 479739.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2 or 3, a nucleic acid encoding human FGFR4 and demonstrate at least 80% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 479533, 479534, 479535, 479536, 479537, 479539, 479542, 479545, 479546, 479547, 479548, 479551, 479552, 479553, 479554, 479556, 479558, 479565, 479566, 479568, 479570, 479573, 479583, 479584, 479594, 479596, 479691, 479692, 479698, 479699, 479703, 479704, 479706, 479721, 479722, 479736, and 479739.

In certain embodiments, the following antisense compounds target a region of SEQ ID NOs: 1 or 2 or 3, a nucleic acid encoding human FGFR4 and demonstrate at least 85% inhibition of a FGFR4 mRNA: ISIS NOs: 463629, 479533, 479534, 479535, 479536, 479539, 479546, 479548, 479551, 479552, 479556, 479566, 479573, 479691, 479692, 479698, 479699, 479703, 479704, 479706, 479736, and 479739.

In certain embodiments, the compounds provided herein have a greater therapeutic potential than ISIS NO: 299005. In certain embodiments, the compounds provided herein have better in vivo inhibition over ISIS NO: 299005. In certain embodiments, the compounds provided herein have a better tolerability profile than ISIS NO: 299005.

In certain embodiments, the compound provided herein consists of a single-stranded modified oligonucleotide.

In certain embodiments, the modified oligonucleotide consists of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34 or 35 linked nucleosides. In certain embodiments, the modified oligonucleotide consists of 20 linked nucleosides. In certain embodiments, the modified oligonucleotide consists of 19 linked nucleosides. In certain embodiments, the modified oligonucleotide consists of 18 linked nucleosides. In certain embodiments, the modified oligonucleotide consists of 17 linked nucleosides. In certain embodiments, the modified oligonucleotide consists of 16 linked nucleosides.

In certain embodiments, at least one internucleoside linkage of the modified oligonucleotide is a modified internucleoside linkage. In certain embodiments, each internucleoside linkage is a phosphorothioate internucleoside linkage.

In certain embodiments, at least one nucleoside of said modified oligonucleotide comprises a modified nucleobase. In certain embodiments, the modified nucleobase is a 5-methylcytosine.

In certain embodiments, the modified oligonucleotide comprises: a) a gap segment consisting of linked deoxynucleosides; b) a 5′ wing segment consisting of linked nucleosides; and c) a 3′ wing segment consisting of linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment and each nucleoside of each wing segment comprises a modified sugar.

In certain embodiments, the modified oligonucleotide consists of 20 linked nucleosides, the gap segment consisting of ten linked deoxynucleosides, the 5′ wing segment consisting of five linked nucleosides, the 3′ wing segment consisting of five linked nucleosides, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine is a 5-methylcytosine.

In certain embodiments, the modified oligonucleotide consists of 17 linked nucleosides, the gap segment consisting of ten linked deoxynucleosides, the 5′ wing segment consisting of three linked nucleosides, the 3′ wing segment consisting of four linked nucleosides, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of any of SEQ ID NOs: 1-4, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of any of SEQ ID NO: 1, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 19 contiguous nucleobases of SEQ ID NOs: 7-322 wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 19 contiguous nucleobases of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 19 contiguous nucleobases of SEQ ID NO: 16, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 19 contiguous nucleobases of SEQ ID NO: 45, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 20 contiguous nucleobases of SEQ ID NOs: 7-322, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 20 contiguous nucleobases of SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine. In certain embodiments, the compound or composition comprises the compound of any of ISIS NOs: 463588, 463589, 463690, 463691, 463835, 463837, or 464225.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 20 contiguous nucleobases of SEQ ID NO: 16, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463588.

In certain embodiments, the compounds or compositions provided herein comprise a modified oligonucleotide consisting of 20 linked nucleosides having a nucleobase sequence comprising at least 20 contiguous nucleobases of SEQ ID NO: 45, wherein the modified oligonucleotide comprises: a) a gap segment consisting of ten linked deoxynucleosides; b) a 5′ wing segment consisting of five linked nucleosides; and c) a 3′ wing segment consisting of five linked nucleosides. The gap segment is positioned between the 5′ wing segment and the 3′ wing segment, each nucleoside of each wing segment comprises a 2′-O-methoxyethyl modified sugar, each internucleoside linkage is a phosphorothioate linkage and each cytosine residue is a 5-methylcytosine. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463690.

Certain embodiments provide methods, compounds, and compositions for inhibiting FGFR4 expression.

Certain embodiments provide a method of reducing FGFR4 expression in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 15 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 18 to 21 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 to 35 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 to 25 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 to 24 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 to 23 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 to 22 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 to 21 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4.

Certain embodiments provide a method of preventing, ameliorating or treating a metabolic disease in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. Examples of metabolic diseases or disorders include, but are not limited to obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

Certain embodiments provide a compound as described herein for use in preventing, ameliorating or treating a metabolic disease in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. Examples of metabolic diseases or disorders include, but are not limited to obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

Certain embodiments provide use of a compound as described herein in the manufacture of a medicament for preventing, ameliorating or treating a metabolic disease in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. Examples of metabolic diseases or disorders include, but are not limited to obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

Certain embodiments provide a method of preventing, ameliorating or treating obesity in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound or composition comprises the compound of ISIS NOs: 463588, 463589, 463690, 463691, 463835, 463837, or 464225. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463588. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463690.

Certain embodiments provide a compound as described herein for use in preventing, ameliorating or treating obesity in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound or composition comprises the compound of ISIS NOs: 463588, 463589, 463690, 463691, 463835, 463837, or 464225. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463588. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463690.

Certain embodiments provide use of a compound as described herein in the manufacture of a medicament for preventing, ameliorating or treating obesity in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound or composition comprises the compound of ISIS NOs: 463588, 463589, 463690, 463691, 463835, 463837, or 464225. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463588. In certain embodiments, the compound or composition comprises the compound of ISIS NO: 463690.

Certain embodiments provide a method of reducing body weight in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the body weight is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a compound as described herein for use in reducing body weight in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the body weight is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide use of a compound as described herein in the manufacture of a medicament for reducing body weight in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction of body weight in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the body weight is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a method of reducing adipose tissue in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, adiposity is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a compound as described herein for use in reducing adipose tissue in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, adiposity is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide use of a compound as described herein in the manufacture of a medicament for reducing adipose tissue in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction in adiposity in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, adiposity is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a method of increasing fatty acid oxidation in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats diabetes. In certain embodiments, increasing fatty acid oxidation an animal prevents, ameliorates or treats obesity. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, increasing fatty acid oxidation prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the fatty acid oxidation is increased by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a compound as described herein for use in increasing fatty acid oxidation in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats diabetes. In certain embodiments, increasing fatty acid oxidation an animal prevents, ameliorates or treats obesity. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, increasing fatty acid oxidation prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the fatty acid oxidation is increased by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide use of a compound as described herein in the manufacture of a medicament for increasing fatty acid oxidation in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats diabetes. In certain embodiments, increasing fatty acid oxidation an animal prevents, ameliorates or treats obesity. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, increasing fatty acid oxidation in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, increasing fatty acid oxidation prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the fatty acid oxidation is increased by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a method of reducing glucose levels in an animal comprising administering to the animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the glucose level is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide a compound as described herein for use in reducing glucose levels in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the glucose level is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

Certain embodiments provide use of a compound as described herein in the manufacture of a medicament for reducing glucose levels in an animal. In certain embodiments, the compound comprises a modified oligonucleotide 12 to 30 linked nucleosides in length targeted to FGFR4. In certain embodiments, the compound comprises a modified oligonucleotide 20 linked nucleosides in length targeted to FGFR4. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats a metabolic disease. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats diabetes. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats obesity. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats metabolic syndrome. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats insulin resistance. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats hyperglycemia. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats NAFLD. In certain embodiments, reduction of glucose levels in an animal prevents, ameliorates or treats diabetic dyslipidemia. In certain embodiments, the glucose level is reduced by at least 5%, 10%, 20%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.

In certain embodiments, FGFR4 has the sequence as set forth in any of the GENBANK Accession Numbers: GENBANK Accession No. NM002011.3 (incorporated herein as SEQ ID NO: 1), GENBANK Accession No: NT023133.11 truncated from nucleosides 21323018 to 21335213 (incorporated herein as SEQ ID NO: 2); GENBANK Accession No. AB209631.1 (incorporated herein as SEQ ID NO: 3); and GENBANK Accession No NM022963.2 (incorporated herein as SEQ ID NO: 4).). In certain embodiments, FGFR4 has the human sequence as set forth in SEQ ID NOs: 1-4. In certain embodiments, FGFR4 has the rhesus monkey sequence as set forth in GENBANK Accession No. NW001121000.1 truncated from nucleosides 3094000 to 3109000 (SEQ ID NO: 5). In certain embodiments, FGFR4 has the murine sequence as set forth in GENBANK Accession No. BC033313.1 (SEQ ID NO: 6).

In certain embodiments, the compounds or compositions provided herein comprise a salt thereof, and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138 or a salt thereof and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 to 25 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138 or a salt thereof and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 16, 17, 45, 46, 70, 72, or 138 or a salt thereof and a pharmaceutically acceptable carrier or diluent.

In certain embodiments, the compounds or compositions provided herein comprise a salt thereof, and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 16 or a salt thereof and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 to 25 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 16 or a salt thereof and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 16 or a salt thereof and a pharmaceutically acceptable carrier or diluent.

In certain embodiments, the compounds or compositions provided herein comprise a salt thereof, and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 45 or a salt thereof and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 to 25 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 45 or a salt thereof and a pharmaceutically acceptable carrier or diluent. In certain embodiments, the composition comprises a modified oligonucleotide consisting of 20 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 45 or a salt thereof and a pharmaceutically acceptable carrier or diluent.

Certain embodiments provide a method for treating an animal with a FGFR4 related disease or condition comprising: a) identifying said animal with the FGFR4 related disease or condition, and b) administering to said animal a therapeutically effective amount of a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides and having a nucleobase sequence at least 90% complementary to any of SEQ ID NOs: 1-4 as measured over the entirety of said modified oligonucleotide. In certain embodiments, the therapeutically effective amount of the compound administered to the animal treats or reduces the FGFR4 related disease or condition, or a symptom thereof, in the animal. In certain embodiments, the FGFR4 related disease or condition is obesity. In certain embodiments, the FGFR4 related disease or condition is diabetes.

Certain embodiments provide a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides and having a nucleobase sequence at least 90% complementary to any of SEQ ID NOs: 1-4 as measured over the entirety of said modified oligonucleotide for treating a FGFR4 related disease or condition. In certain embodiments, the FGFR4 related disease or condition is obesity. In certain embodiments, the FGFR4 related disease or condition is diabetes.

Certain embodiments provide use of a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides and having a nucleobase sequence at least 90% complementary to any of SEQ ID NOs: 1-4 as measured over the entirety of said modified oligonucleotide in the preparation of a medicament for treating a FGFR4 related disease or condition. In certain embodiments, the FGFR4 related disease or condition is obesity. In certain embodiments, the FGFR4 related disease or condition is diabetes.

Certain embodiments provide a method for treating an animal with a FGFR4 related disease or condition comprising: a) identifying said animal with the FGFR4 related disease or condition, and b) administering to said animal a therapeutically effective amount of a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides and having a nucleobase sequence at least 100% complementary to any of SEQ ID NOs: 1-4 as measured over the entirety of said modified oligonucleotide. In certain embodiments, the therapeutically effective amount of the compound administered to the animal treats or reduces the FGFR4 related disease or condition, or a symptom thereof, in the animal. In certain embodiments, the FGFR4 related disease or condition is obesity. In certain embodiments, the FGFR4 related disease or condition is diabetes.

Certain embodiments provide a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides and having a nucleobase sequence at least 100% complementary to any of SEQ ID NOs: 1-4 as measured over the entirety of said modified oligonucleotide for treating a FGFR4 related disease or condition. In certain embodiments, the FGFR4 related disease or condition is obesity. In certain embodiments, the FGFR4 related disease or condition is diabetes.

Certain embodiments provide use of a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides and having a nucleobase sequence at least 100% complementary to any of SEQ ID NOs: 1-4 as measured over the entirety of said modified oligonucleotide in the preparation of a medicament for treating a FGFR4 related disease or condition. In certain embodiments, the FGFR4 related disease or condition is obesity. In certain embodiments, the FGFR4 related disease or condition is diabetes.

Certain embodiments provide methods of treating, preventing, or ameliorating a metabolic disease. In certain embodiments the metabolic disease is obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

Certain embodiments provide compounds described herein for treating, preventing, or ameliorating a metabolic disease. In certain embodiments the metabolic disease is obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

Certain embodiments provide use of compounds described herein in the preparation of a medicament for treating, preventing, or ameliorating a metabolic disease. In certain embodiments the metabolic disease is obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof.

Certain embodiments provide methods comprising administering to an animal a compound as described herein to an animal. In certain embodiments, the method comprises administering to an animal a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138.

Certain embodiments provide a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138 for treating a metabolic disease, diabetes, and/or obesity.

Certain embodiments provide use of a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NOs: 16, 17, 45, 46, 70, 72, or 138 in the preparation of a medicament for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide methods comprising administering to an animal a compound as described herein to an animal. In certain embodiments, the method comprises administering to an animal a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence selected from among the nucleobase sequences recited in SEQ ID NO: 16.

Certain embodiments provide a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 16 for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide use of a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 16 in the preparation of a medicament for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide methods comprising administering to an animal a compound as described herein to an animal. In certain embodiments, the method comprises administering to an animal a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence selected from among the nucleobase sequences recited in SEQ ID NO: 45.

Certain embodiments provide a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 45 for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide use of a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence recited in SEQ ID NO: 45 in the preparation of a medicament for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide methods comprising administering to an animal a compound as described herein to an animal. In certain embodiments, the method comprises administering to an animal a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence selected from among the nucleobase sequences recited in ISIS NO: 463588.

Certain embodiments provide a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence of ISIS NO: 463588 for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide use of a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence of ISIS NO: 463588 in the preparation of a medicament for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide methods comprising administering to an animal a compound as described herein to an animal. In certain embodiments, the method comprises administering to an animal a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence selected from among the nucleobase sequences recited in ISIS NO: 463690.

Certain embodiments provide a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence of ISIS NO: 463690 for treating metabolic disease, diabetes, and/or obesity.

Certain embodiments provide use of a modified oligonucleotide consisting of 20 to 35 linked nucleosides and having a nucleobase sequence comprising at least 20 contiguous nucleobases of a nucleobase sequence of ISIS NO: 463690 in the preparation of a medicament for treating metabolic disease, diabetes, and/or obesity.

In certain embodiments, the animal is a human.

In certain embodiments, the administering prevents, treats, ameliorates, or slows progression of a metabolic disease as described herein.

In certain embodiments, the administering prevents, treats, ameliorates, or slows progression of obesity as described herein.

In certain embodiments, the administering prevents, treats, ameliorates, or slows progression of diabetes as described herein.

In certain embodiments, the compound is co-administered with a second agent.

In certain embodiments, the compound and the second agent are administered concomitantly.

In certain embodiments, the administering is parenteral administration.

Certain embodiments further provide a method to reduce FGFR4 mRNA or protein expression in an animal comprising administering to the animal a compound or composition as described herein to reduce FGFR4 mRNA or protein expression in the animal. In certain embodiments, the animal is a human. In certain embodiments, reducing FGFR4 mRNA or protein expression prevents, treats, ameliorates, or slows progression of metabolic disease. In certain embodiments, the metabolic disease or condition is diabetes. In certain embodiments, the metabolic disease or condition is obesity.

Certain embodiments provide a method for treating a human with a metabolic disease comprising identifying the human with the disease and administering to the human a therapeutically effective amount of a compound or composition as described herein. In certain embodiments, the treatment reduces a symptom selected from the group consisting of metabolic syndrome, hyperglycemia, hypertriglyceridemia, hypertension, increased glucose levels, increased insulin resistance, decreased insulin sensitivity, above normal body weight, and/or above normal body fat or any combination thereof.

Certain embodiments provide a method for treating a human with obesity comprising identifying the human with the disease and administering to the human a therapeutically effective amount of a compound or composition as described herein. In certain embodiments, the treatment reduces a symptom selected from the group consisting of metabolic syndrome, hyperglycemia, hypertriglyceridemia, hypertension, increased glucose levels, increased insulin resistance, decreased insulin sensitivity, above normal body weight, and/or above normal body fat or any combination thereof

Certain embodiments provide a method for treating a human with diabetes comprising identifying the human with the disease and administering to the human a therapeutically effective amount of a compound or composition as described herein. In certain embodiments, the treatment reduces a symptom selected from the group consisting of metabolic syndrome, hyperglycemia, hypertriglyceridemia, hypertension, increased glucose levels, increased insulin resistance, decreased insulin sensitivity, above normal body weight, and/or above normal body fat or any combination thereof

Further provided is a method for reducing or preventing metabolic disease comprising administering to a human a therapeutically effective amount compound or composition as described herein, thereby reducing or preventing metabolic disease.

Further provided is a method for reducing or preventing obesity comprising administering to a human a therapeutically effective amount compound or composition as described herein, thereby reducing or preventing diabetes.

Further provided is a method for reducing or preventing diabetes comprising administering to a human a therapeutically effective amount compound or composition as described herein, thereby reducing or preventing diabetes.

Further provided is a method for ameliorating a symptom of metabolic disease, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic disease in the human.

Further provided is a method for ameliorating a symptom of obesity, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic disease in the human.

Further provided is a method for ameliorating a symptom of diabetes, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic disease in the human.

Further provided is a method for ameliorating a symptom of metabolic syndrome, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic syndrome in the human.

Further provided is a method for ameliorating a symptom of metabolic disease, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic disease in the human.

Further provided is a method for ameliorating a symptom of obesity, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of obesity in the human.

Further provided is a method for ameliorating a symptom of diabetes, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of diabetes in the human.

Further provided is a method for ameliorating a symptom of metabolic syndrome, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic syndrome in the human.

Further provided is a method for ameliorating a symptom of metabolic disease, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic disease in the human.

Further provided is a method for ameliorating a symptom of obesity, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of obesity in the human.

Further provided is a method for ameliorating a symptom of diabetes, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of diabetes in the human.

Further provided is a method for ameliorating a symptom of metabolic syndrome, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby ameliorating a symptom of metabolic syndrome in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with metabolic disease, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of metabolic disease in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with obesity, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of obesity in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with diabetes, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of diabetes in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with metabolic syndrome, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 to 35 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of metabolic syndrome in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with metabolic disease, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of metabolic disease in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with obesity, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of obesity in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with diabetes, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of diabetes in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with metabolic syndrome, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of metabolic syndrome in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with metabolic disease, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of metabolic disease in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with obesity, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of obesity in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with diabetes, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of diabetes in the human.

Further provided is a method for reducing the rate of progression of a symptom associated with metabolic syndrome, comprising administering to a human in need thereof a compound comprising a modified oligonucleotide consisting of 20 linked nucleosides, wherein said modified oligonucleotide specifically hybridizes to SEQ ID NO: 1, 2, 3, 4 or 5, thereby reducing the rate of progression a symptom of metabolic syndrome in the human.

Also provided are methods and compounds for the preparation of a medicament for the treatment, prevention, or amelioration of metabolic disease.

Also provided are methods and compounds for the preparation of a medicament for the treatment, prevention, or amelioration of obesity.

Also provided are methods and compounds for the preparation of a medicament for the treatment, prevention, or amelioration of diabetes.

Also provided are methods and compounds for the preparation of a medicament for the treatment, prevention, or amelioration of metabolic syndrome.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, ameliorating, or preventing metabolic disease.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, ameliorating, or preventing obesity.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, ameliorating, or preventing diabetes.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, ameliorating, or preventing metabolic syndrome.

Certain embodiments provide a compound as described herein for use in treating, preventing, or ameliorating metabolic disease as described herein by combination therapy with an additional agent or therapy as described herein. Agents or therapies can be co-administered or administered concomitantly.

Certain embodiments provide a compound as described herein for use in treating, preventing, or ameliorating diabetes as described herein by combination therapy with an additional agent or therapy as described herein. Agents or therapies can be co-administered or administered concomitantly.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating metabolic disease as described herein by combination therapy with an additional agent or therapy as described herein. Agents or therapies can be co-administered or administered concomitantly.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating obesity as described herein by combination therapy with an additional agent or therapy as described herein. Agents or therapies can be co-administered or administered concomitantly.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating diabetes as described herein by combination therapy with an additional agent or therapy as described herein. Agents or therapies can be co-administered or administered concomitantly.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating diabetes as described herein by combination therapy with an additional agent or therapy as described herein. Agents or therapies can be co-administered or administered concomitantly.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating metabolic disease as described herein in a patient who is subsequently administered an additional agent or therapy as described herein.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating obesity as described herein in a patient who is subsequently administered an additional agent or therapy as described herein.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating diabetes as described herein in a patient who is subsequently administered an additional agent or therapy as described herein.

Certain embodiments provide the use of a compound as described herein in the manufacture of a medicament for treating, preventing, or ameliorating metabolic syndrome as described herein in a patient who is subsequently administered an additional agent or therapy as described herein.

Certain embodiments provide a kit for treating, preventing, or ameliorating metabolic disease as described herein wherein the kit comprises:

(i) a compound as described herein; and alternatively
(ii) an additional agent or therapy as described herein.

Certain embodiments provide a kit for treating, preventing, or ameliorating obesity as described herein wherein the kit comprises:

(i) a compound as described herein; and alternatively
(ii) an additional agent or therapy as described herein.

Certain embodiments provide a kit for treating, preventing, or ameliorating diabetes as described herein wherein the kit comprises:

(i) a compound as described herein; and alternatively
(ii) an additional agent or therapy as described herein.

Certain embodiments provide a kit for treating, preventing, or ameliorating metabolic syndrome as described herein wherein the kit comprises:

(i) a compound as described herein; and alternatively
(ii) an additional agent or therapy as described herein.

A kit as described herein may further include instructions for using the kit to treat, prevent, or ameliorate metabolic disease as described herein by combination therapy as described herein. In certain embodiments, the metabolic disease is obesity. In certain embodiments, the metabolic disease is diabetes.

In certain embodiments, a biomarker of the anti-obesity effect of an FGFR4 inhibitor is an increase in FGF15 and/or FGF19 protein levels. In certain embodiments, a biomarker of FGFR4 antisense oligonucleotide-caused anti-obesity effect is an increase in FGF15 and/or FGF19 gene expression levels. In certain embodiments, a biomarker of FGFR4 antisense oligonucleotide-caused anti-obesity effect is an increase in FGF15 and/or FGF19 protein levels. In certain embodiments, a biomarker of FGFR4 antisense oligonucleotide-caused anti-obesity effect is an increase in FGF15 and/or FGF19 gene expression levels. In certain embodiments, the FGF15 and/or FGF19 nucleic acid is any of the sequences set forth in GENBANK Accession No. NM008003.2 (incorporated herein as SEQ ID NO: 345), GENBANK Accession No: XM001100825.1 (incorporated herein as SEQ ID NO: 346); and GENBANK Accession No. NM005117.1 (incorporated herein as SEQ ID NO: 347).

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal by measuring an increase in ileum FGF15 and/or ileum FGF19 gene expression and plasma FGF15 and/or plasma FGF19 protein levels.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor by measuring an increase in ileum FGF15 and/or ileum FGF19 gene expression and plasma FGF15 and/or plasma FGF19 protein levels.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF15 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF19 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF15 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF19 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF15 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF19 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF15 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF19 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide a method for treating a metabolic disease, including obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof, comprising administering a first dose of a compound or composition as described herein to a subject having a baseline level of FGF15 or FGF19 mRNA or protein in the blood or a tissue and administering one or more additional doses of the compound or composition to the subject until the level of FGF15 or FGF19 in the blood or a tissue is not increased from the baseline level by a certain extent for a certain amount of time.

In some aspects, one or more additional doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is not increased from the baseline level for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks.

In certain aspects, one or more additional doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is not increased from the baseline level by at least about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103% 104%, 105%, 106%, 107%, 108%, 109%, 110%, 111%, 112%, 113%, 114%, 115%, 116%, 117%, 118%, 119%, 120%, 121%, 122%, 123%, 124%, 125%, 126%, 127%, 128%, 129%, 130%, 131%, 132%, 133%, 134%, 135%, 136%, 137%, 138%, 139%, 140%, 141%, 142%, 143%, 144%, 145%, 146%, 147%, 148%, 149%, 150%, 151%, 152%, 153%, 154%, 155%, 156%, 157%, 158%, 159%, 160%, 161%, 162%, 163%, 164%, 165%, 166%, 167%, 168%, 169%, 170%, 171%, 172%, 173%, 174%, 175%, 176%, 177%, 178%, 179%, 180%, 181%, 182%, 183%, 184%, 185%, 186%, 187%, 188%, 189%, 190%, 191%, 192%, 193%, 194%, 195%, 196%, 197%, 198%, 199%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1,000%, or any value in between any of the aforementioned percentages.

Administration of one or more additional doses of the compound or composition described herein can continue until such increases in the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue relative to the baseline level does not occur for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain aspects, one or more additional doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 protein in the blood or a tissue is not increased from the baseline level by at least about 1 pg/mL, 5 pg/mL, 10 pg/mL, 15 pg/mL, 20 pg/mL, 25 pg/mL, 30 pg/mL, 35 pg/mL, 40 pg/mL, 45 pg/mL, 50 pg/mL, 55 pg/mL, 60 pg/mL, 65 pg/mL, 70 pg/mL, 75 pg/mL, 80 pg/mL, 85 pg/mL, 90 pg/mL, 95 pg/mL, 100 pg/mL, 105 pg/mL, 110 pg/mL, 115 pg/mL, 120 pg/mL, 125 pg/mL, 130 pg/mL, 135 pg/mL, 140 pg/mL, 145 pg/mL, 150 pg/mL, 155 pg/mL, 160 pg/mL, 165 pg/mL, 170 pg/mL, 175 pg/mL, 180 pg/mL, 185 pg/mL, 190 pg/mL, 195 pg/mL, 200 pg/mL, 205 pg/mL, 210 pg/mL, 215 pg/mL, 220 pg/mL, 225 pg/mL, 230 pg/mL, 235 pg/mL, 240 pg/mL, 245 pg/mL, 250 pg/mL, 255 pg/mL, 260 pg/mL, 265 pg/mL, 270 pg/mL, 275 pg/mL, 280 pg/mL, 290 pg/mL, 295 pg/mL, 300 pg/mL, 350 pg/mL, 400 pg/mL, 450 pg/mL, 500 pg/mL, 550 pg/mL, 600 pg/mL, 650 pg/mL, 700 pg/mL, 750 pg/mL, 800 pg/mL, 850 pg/mL, 900 pg/mL, 950 pg/mL, 1,000 pg/mL, 2,000 pg/mL, or any value in between any of the aforementioned concentrations. Administration of one or more additional doses of the compound or composition described herein can continue until such increases in the level of FGF15 or FGF19 protein in the blood or a tissue relative to the baseline level does not occur for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

It will be understood that one or more doses of the compound or composition described herein can be administered during the aforementioned time periods. For example, a subject may have been administered one or more doses of the compound or composition described herein during the at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In certain embodiments, additional doses of the compound or composition described herein are administered to the subject until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is not increased from the baseline level by at least about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103% 104%, 105%, 106%, 107%, 108%, 109%, 110%, 111%, 112%, 113%, 114%, 115%, 116%, 117%, 118%, 119%, 120%, 121%, 122%, 123%, 124%, 125%, 126%, 127%, 128%, 129%, 130%, 131%, 132%, 133%, 134%, 135%, 136%, 137%, 138%, 139%, 140%, 141%, 142%, 143%, 144%, 145%, 146%, 147%, 148%, 149%, 150%, 151%, 152%, 153%, 154%, 155%, 156%, 157%, 158%, 159%, 160%, 161%, 162%, 163%, 164%, 165%, 166%, 167%, 168%, 169%, 170%, 171%, 172%, 173%, 174%, 175%, 176%, 177%, 178%, 179%, 180%, 181%, 182%, 183%, 184%, 185%, 186%, 187%, 188%, 189%, 190%, 191%, 192%, 193%, 194%, 195%, 196%, 197%, 198%, 199%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1,000%, or any value in between any of the aforementioned percentages in the aforementioned time periods. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

Various embodiments are directed to a method of treating a metabolic disease, including obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof, comprising (a) obtaining the baseline level of FGF15 or FGF19 mRNA or protein in the blood or a tissue of a subject, (b) administering to the subject a dose of a compound or composition described herein, (c) obtaining the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue after the administration of the compound or composition described herein; and (d) repeating steps (b) and (c) until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue does not increase by a certain extent for a certain amount of time relative to baseline.

In several aspects, steps (b) and (c) are repeated until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue does not increase relative to baseline for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain aspects, one or more additional doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 protein in the blood or a tissue is not increased from the baseline level by at least about 1 pg/mL, 5 pg/mL, 10 pg/mL, 15 pg/mL, 20 pg/mL, 25 pg/mL, 30 pg/mL, 35 pg/mL, 40 pg/mL, 45 pg/mL, 50 pg/mL, 55 pg/mL, 60 pg/mL, 65 pg/mL, 70 pg/mL, 75 pg/mL, 80 pg/mL, 85 pg/mL, 90 pg/mL, 95 pg/mL, 100 pg/mL, 105 pg/mL, 110 pg/mL, 115 pg/mL, 120 pg/mL, 125 pg/mL, 130 pg/mL, 135 pg/mL, 140 pg/mL, 145 pg/mL, 150 pg/mL, 155 pg/mL, 160 pg/mL, 165 pg/mL, 170 pg/mL, 175 pg/mL, 180 pg/mL, 185 pg/mL, 190 pg/mL, 195 pg/mL, 200 pg/mL, 205 pg/mL, 210 pg/mL, 215 pg/mL, 220 pg/mL, 225 pg/mL, 230 pg/mL, 235 pg/mL, 240 pg/mL, 245 pg/mL, 250 pg/mL, 255 pg/mL, 260 pg/mL, 265 pg/mL, 270 pg/mL, 275 pg/mL, 280 pg/mL, 290 pg/mL, 295 pg/mL, 300 pg/mL, 350 pg/mL, 400 pg/mL, 450 pg/mL, 500 pg/mL, 550 pg/mL, 600 pg/mL, 650 pg/mL, 700 pg/mL, 750 pg/mL, 800 pg/mL, 850 pg/mL, 900 pg/mL, 950 pg/mL, 1,000 pg/mL, 2,000 pg/mL, or any value in between any of the aforementioned concentrations. Administration of one or more additional doses of the compound or composition described herein can continue until such increases in the level of FGF15 or FGF19 protein in the blood or a tissue relative to the baseline level does not occur for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain embodiments, a method of treating a metabolic disease and/or obesity comprises (a) obtaining the baseline level of FGF15 or FGF19 mRNA or protein in the blood or a tissue of a subject, (b) administering to the subject a dose of a compound or composition described herein, (c) obtaining the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue after the administration of the compound or composition described herein; and (d) repeating steps (b) and (c) until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue does not increase by about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103% 104%, 105%, 106%, 107%, 108%, 109%, 110%, 111%, 112%, 113%, 114%, 115%, 116%, 117%, 118%, 119%, 120%, 121%, 122%, 123%, 124%, 125%, 126%, 127%, 128%, 129%, 130%, 131%, 132%, 133%, 134%, 135%, 136%, 137%, 138%, 139%, 140%, 141%, 142%, 143%, 144%, 145%, 146%, 147%, 148%, 149%, 150%, 151%, 152%, 153%, 154%, 155%, 156%, 157%, 158%, 159%, 160%, 161%, 162%, 163%, 164%, 165%, 166%, 167%, 168%, 169%, 170%, 171%, 172%, 173%, 174%, 175%, 176%, 177%, 178%, 179%, 180%, 181%, 182%, 183%, 184%, 185%, 186%, 187%, 188%, 189%, 190%, 191%, 192%, 193%, 194%, 195%, 196%, 197%, 198%, 199%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1,000%, or any value in between any of the aforementioned percentages relative to baseline for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

Certain embodiments provide a method for treating a metabolic disease, including obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof, comprising administering a first dose of a compound or composition as described herein to a subject having a baseline level of FGF15 or FGF19 mRNA or protein in the blood or a tissue and administering one or more additional higher doses of the compound or composition to the subject until the level of FGF15 or FGF19 in the blood or a tissue is increased from the baseline level by a certain extent for a certain amount of time. In several aspects, such method further comprises administering additional doses of the compound or composition to the subject to maintain FGF15 or FGF19 mRNA or protein in the blood or a tissue at a certain level above the baseline level. It will be understood that the one or more additional higher doses can be relative to the first dose or the most recently administered additional higher dose.

In some aspects, one or more additional higher doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is increased from the baseline level for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks.

In several aspects, one or more additional higher doses of the compound or composition described herein is an amount at least about 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0, 2.1, 2.2, 2.3, 2.4, 2.5, 2.6, 2.7, 2.8, 2.9, 3.0, 3.1, 3.2, 3.3., 3.4, 3.5, 3.6, 3.7, 3.8, 3.9, 4.0, 4.1, 4.2, 4.3, 4.4, 4.5, 4.6, 4.7, 4.8, 4.9, 5.0, 5.1, 5.2, 5.3, 5.4, 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, 7.0, 7.1, 7.2, 7.3, 7.4, 7.5, 7.6, 7.7, 7.8, 7.9, 8.0, 8.1, 8.2, 8.3, 8.4, 8.5, 8.6, 8.7, 8.8, 8.9, 9.0, 9.1, 9.2, 9.3, 9.4, 9.5, 9.6, 9.7, 9.8, 9.9, or 10.0 fold greater than the first dose or most recently administered additional higher dose. In certain aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain aspects, one or more additional higher doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is increased from the baseline level by at least about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103% 104%, 105%, 106%, 107%, 108%, 109%, 110%, 111%, 112%, 113%, 114%, 115%, 116%, 117%, 118%, 119%, 120%, 121%, 122%, 123%, 124%, 125%, 126%, 127%, 128%, 129%, 130%, 131%, 132%, 133%, 134%, 135%, 136%, 137%, 138%, 139%, 140%, 141%, 142%, 143%, 144%, 145%, 146%, 147%, 148%, 149%, 150%, 151%, 152%, 153%, 154%, 155%, 156%, 157%, 158%, 159%, 160%, 161%, 162%, 163%, 164%, 165%, 166%, 167%, 168%, 169%, 170%, 171%, 172%, 173%, 174%, 175%, 176%, 177%, 178%, 179%, 180%, 181%, 182%, 183%, 184%, 185%, 186%, 187%, 188%, 189%, 190%, 191%, 192%, 193%, 194%, 195%, 196%, 197%, 198%, 199%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1,000%, or any value in between any of the aforementioned percentages.

Administration of one or more additional higher doses of the compound or composition described herein can continue until such increases in the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue relative to the baseline level occurs for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In certain aspects, additional doses of the compound or composition can be administered to the subject to maintain FGF15 or FGF19 mRNA or protein in the blood or a tissue at a certain level above the baseline level. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain aspects, one or more additional higher doses of the compound or composition described herein is administered to the subject until the level of FGF15 or FGF19 protein in the blood or a tissue is increased from the baseline level by at least about 1 pg/mL, 5 pg/mL, 10 pg/mL, 15 pg/mL, 20 pg/mL, 25 pg/mL, 30 pg/mL, 35 pg/mL, 40 pg/mL, 45 pg/mL, 50 pg/mL, 55 pg/mL, 60 pg/mL, 65 pg/mL, 70 pg/mL, 75 pg/mL, 80 pg/mL, 85 pg/mL, 90 pg/mL, 95 pg/mL, 100 pg/mL, 105 pg/mL, 110 pg/mL, 115 pg/mL, 120 pg/mL, 125 pg/mL, 130 pg/mL, 135 pg/mL, 140 pg/mL, 145 pg/mL, 150 pg/mL, 155 pg/mL, 160 pg/mL, 165 pg/mL, 170 pg/mL, 175 pg/mL, 180 pg/mL, 185 pg/mL, 190 pg/mL, 195 pg/mL, 200 pg/mL, 205 pg/mL, 210 pg/mL, 215 pg/mL, 220 pg/mL, 225 pg/mL, 230 pg/mL, 235 pg/mL, 240 pg/mL, 245 pg/mL, 250 pg/mL, 255 pg/mL, 260 pg/mL, 265 pg/mL, 270 pg/mL, 275 pg/mL, 280 pg/mL, 290 pg/mL, 295 pg/mL, 300 pg/mL, 350 pg/mL, 400 pg/mL, 450 pg/mL, 500 pg/mL, 550 pg/mL, 600 pg/mL, 650 pg/mL, 700 pg/mL, 750 pg/mL, 800 pg/mL, 850 pg/mL, 900 pg/mL, 950 pg/mL, 1,000 pg/mL, 2,000 pg/mL, or any value in between any of the aforementioned concentrations. Administration of one or more additional higher doses of the compound or composition described herein can continue until such increases in the level of FGF15 or FGF19 protein in the blood or a tissue relative to the baseline level occurs for at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In certain aspects, additional doses of the compound or composition can be administered to the subject to maintain FGF15 or FGF19 protein in the blood or a tissue at any of the aforementioned concentrations above the baseline level. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

It will be understood that one or more higher doses of the compound or composition described herein can be administered during the aforementioned time periods. For example, a subject may have been administered one or more doses of the compound or composition described herein during the at least about one week, two weeks, three weeks, four weeks, five weeks, six weeks, seven weeks, eight weeks, nine weeks, ten weeks, eleven weeks, twelve weeks, thirteen weeks, fourteen weeks, fifteen weeks, sixteen weeks, seventeen weeks, eighteen weeks, nineteen weeks, twenty weeks, twenty-one weeks, twenty-two weeks, twenty-three weeks, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty, forty-one, forty-two, forty-three, forty-four, forty-five, forty-six, forty-seven, forty-eight, forty-nine, or fifty weeks. In certain embodiments, additional higher doses of the compound or composition described herein are administered to the subject until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is increased from the baseline level by at least about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103% 104%, 105%, 106%, 107%, 108%, 109%, 110%, 111%, 112%, 113%, 114%, 115%, 116%, 117%, 118%, 119%, 120%, 121%, 122%, 123%, 124%, 125%, 126%, 127%, 128%, 129%, 130%, 131%, 132%, 133%, 134%, 135%, 136%, 137%, 138%, 139%, 140%, 141%, 142%, 143%, 144%, 145%, 146%, 147%, 148%, 149%, 150%, 151%, 152%, 153%, 154%, 155%, 156%, 157%, 158%, 159%, 160%, 161%, 162%, 163%, 164%, 165%, 166%, 167%, 168%, 169%, 170%, 171%, 172%, 173%, 174%, 175%, 176%, 177%, 178%, 179%, 180%, 181%, 182%, 183%, 184%, 185%, 186%, 187%, 188%, 189%, 190%, 191%, 192%, 193%, 194%, 195%, 196%, 197%, 198%, 199%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1,000%, or any value in between any of the aforementioned percentages in the aforementioned time periods. In certain aspects, additional doses of the compound or composition can be administered to the subject to maintain FGF15 or FGF19 mRNA or protein in the blood or a tissue at any of the aforementioned increased levels above the baseline level. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain embodiments, a method of treating a metabolic disease, including obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof, comprises (a) obtaining the baseline level of FGF15 or FGF19 mRNA or protein in the blood or a tissue of a subject, (b) administering to the subject a dose of a compound or composition described herein, (c) obtaining the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue after the administration of the compound or composition described herein; and (d) repeating steps (b) and (c) until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is increased by at least about 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103% 104%, 105%, 106%, 107%, 108%, 109%, 110%, 111%, 112%, 113%, 114%, 115%, 116%, 117%, 118%, 119%, 120%, 121%, 122%, 123%, 124%, 125%, 126%, 127%, 128%, 129%, 130%, 131%, 132%, 133%, 134%, 135%, 136%, 137%, 138%, 139%, 140%, 141%, 142%, 143%, 144%, 145%, 146%, 147%, 148%, 149%, 150%, 151%, 152%, 153%, 154%, 155%, 156%, 157%, 158%, 159%, 160%, 161%, 162%, 163%, 164%, 165%, 166%, 167%, 168%, 169%, 170%, 171%, 172%, 173%, 174%, 175%, 176%, 177%, 178%, 179%, 180%, 181%, 182%, 183%, 184%, 185%, 186%, 187%, 188%, 189%, 190%, 191%, 192%, 193%, 194%, 195%, 196%, 197%, 198%, 199%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, 1,000%, or any value in between any of the aforementioned percentages relative to the baseline level. In certain aspects, the dose administered in step (d) can be a higher dose than previously administered until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is increased by any of the aforementioned percentages. In certain aspects, such method further comprises administering additional doses of the compound or composition to the subject to maintain FGF15 or FGF19 mRNA or protein in the blood or a tissue at any of the aforementioned increased levels above the baseline level. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

In certain embodiments, a method of treating a metabolic disease, including obesity, diabetes, hyperglycemia, prediabetes, non-alcoholic fatty liver disease (NAFLD), metabolic syndrome, insulin resistance, diabetic dyslipidemia, or hypertriglyceridemia or a combination thereof, comprises (a) obtaining the baseline level of FGF15 or FGF19 protein in the blood or a tissue of a subject, (b) administering to the subject a dose of a compound or composition described herein, (c) obtaining the level of FGF15 or FGF19 protein in the blood or a tissue after the administration of the compound or composition described herein; and (d) repeating steps (b) and (c) until the level of FGF15 or FGF19 protein in the blood or a tissue is increased by at least about 1 pg/mL, 5 pg/mL, 10 pg/mL, 15 pg/mL, 20 pg/mL, 25 pg/mL, 30 pg/mL, 35 pg/mL, 40 pg/mL, 45 pg/mL, 50 pg/mL, 55 pg/mL, 60 pg/mL, 65 pg/mL, 70 pg/mL, 75 pg/mL, 80 pg/mL, 85 pg/mL, 90 pg/mL, 95 pg/mL, 100 pg/mL, 105 pg/mL, 110 pg/mL, 115 pg/mL, 120 pg/mL, 125 pg/mL, 130 pg/mL, 135 pg/mL, 140 pg/mL, 145 pg/mL, 150 pg/mL, 155 pg/mL, 160 pg/mL, 165 pg/mL, 170 pg/mL, 175 pg/mL, 180 pg/mL, 185 pg/mL, 190 pg/mL, 195 pg/mL, 200 pg/mL, 205 pg/mL, 210 pg/mL, 215 pg/mL, 220 pg/mL, 225 pg/mL, 230 pg/mL, 235 pg/mL, 240 pg/mL, 245 pg/mL, 250 pg/mL, 255 pg/mL, 260 pg/mL, 265 pg/mL, 270 pg/mL, 275 pg/mL, 280 pg/mL, 290 pg/mL, 295 pg/mL, 300 pg/mL, 350 pg/mL, 400 pg/mL, 450 pg/mL, 500 pg/mL, 550 pg/mL, 600 pg/mL, 650 pg/mL, 700 pg/mL, 750 pg/mL, 800 pg/mL, 850 pg/mL, 900 pg/mL, 950 pg/mL, 1,000 pg/mL, 2,000 pg/mL, or any value in between any of the aforementioned concentrations. In certain aspects, the dose administered in step (d) can be a higher dose than previously administered until the level of FGF15 or FGF19 mRNA or protein in the blood or a tissue is increased by any of the aforementioned percentages. In certain aspects, such method further comprises administering additional doses of the compound or composition to the subject to maintain FGF15 or FGF19 protein in the blood or a tissue at any of the aforementioned increased concentrations above the baseline level. In several aspects, each dose of compound or composition described herein can be about 50-2000 mg, about 50-400 mg, about 50-200 mg, about 50-100 mg, about 100-200 mg, or any amount in between any of the aforementioned ranges.

The level of FGF15 or FGF19 mRNA or protein in a blood or a tissue, such as liver tissue, may be obtained by several known assays. For instance, FGF15 or FGF19 mRNA levels can be obtained by quantitative RT-PCR. FGF15 or FGF19 protein levels can be obtained, for example, by using any of a number of well recognized immunological binding assays such as, but not limited to, an enzyme linked immunosorbent assay (ELISA), which is also known as a “sandwich assay”, an enzyme immunoassay, a radioimmunoassay (RIA), a fluoroimmunoassay (FIA), a chemiluminescent immunoassay (CLIA) a counting immunoassay (CIA), a filter media enzyme immunoassay (MEIA), or a fluorescence-linked immunosorbent assay (FLISA). Several commercial antibodies against FGF15 or FGF19 mRNA or protein are suitable for obtaining the level of FGF15 or FGF19 mRNA or protein in a blood or a tissue. Such commercially available antibodies can be obtained from Abcam or Santa Cruz Biotechnology, for example. FGF15 or FGF19 protein levels can be also be obtained by high performance liquid chromatography (HPLC), mass spectrometry, or surface plasmon resonance.

Antisense Compounds

Oligomeric compounds include, but are not limited to, oligonucleotides, oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics, antisense compounds, antisense oligonucleotides, and siRNAs. An oligomeric compound may be “antisense” to a target nucleic acid, meaning that is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.

In certain embodiments, an antisense compound has a nucleobase sequence that, when written in the 5′ to 3′ direction, comprises the reverse complement of the target segment of a target nucleic acid to which it is targeted. In certain such embodiments, an antisense oligonucleotide has a nucleobase sequence that, when written in the 5′ to 3′ direction, comprises the reverse complement of the target segment of a target nucleic acid to which it is targeted.

In certain embodiments, an antisense compound targeted to a FGFR4 nucleic acid is 12 to 30 nucleotides in length. In other words, antisense compounds are from 12 to 30 linked nucleobases. In other embodiments, the antisense compound comprises a modified oligonucleotide consisting of 8 to 80, 10 to 50, 15 to 30, 18 to 21, 20 to 80, 20 to 35, 20 to 30, 20 to 29, 20 to 28, 20 to 27, 20 to 26, 20 to 25, 20 to 24, 20 to 23, 20 to 22, 20 to 21 or 20 linked nucleobases. In certain such embodiments, the antisense compound comprises a modified oligonucleotide consisting of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 linked nucleobases in length, or a range defined by any two of the above values.

In certain embodiments, the antisense compound comprises a shortened or truncated modified oligonucleotide. The shortened or truncated modified oligonucleotide can have a single nucleoside deleted from the 5′ end (5′ truncation), or alternatively from the 3′ end (3′ truncation). A shortened or truncated oligonucleotide may have two nucleosides deleted from the 5′ end, or alternatively may have two subunits deleted from the 3′ end. Alternatively, the deleted nucleosides may be dispersed throughout the modified oligonucleotide, for example, in an antisense compound having one nucleoside deleted from the 5′ end and one nucleoside deleted from the 3′ end.

When a single additional nucleoside is present in a lengthened oligonucleotide, the additional nucleoside may be located at the 5′ or 3′ end of the oligonucleotide. When two or more additional nucleosides are present, the added nucleosides may be adjacent to each other, for example, in an oligonucleotide having two nucleosides added to the 5′ end (5′ addition), or alternatively to the 3′ end (3′ addition), of the oligonucleotide. Alternatively, the added nucleoside may be dispersed throughout the antisense compound, for example, in an oligonucleotide having one nucleoside added to the 5′ end and one subunit added to the 3′ end.

It is possible to increase or decrease the length of an antisense compound, such as an antisense oligonucleotide, and/or introduce mismatch bases without eliminating activity. For example, in Woolf et al. (Proc. Natl. Acad. Sci. USA 89:7305-7309, 1992), a series of antisense oligonucleotides 13-25 nucleobases in length were tested for their ability to induce cleavage of a target RNA in an oocyte injection model. Antisense oligonucleotides 25 nucleobases in length with 8 or 11 mismatch bases near the ends of the antisense oligonucleotides were able to direct specific cleavage of the target mRNA, albeit to a lesser extent than the antisense oligonucleotides that contained no mismatches. Similarly, target specific cleavage was achieved using 13 nucleobase antisense oligonucleotides, including those with 1 or 3 mismatches.

Gautschi et al (J. Natl. Cancer Inst. 93:463-471, March 2001) demonstrated the ability of an oligonucleotide having 100% complementarity to the bcl-2 mRNA and having 3 mismatches to the bcl-xL mRNA to reduce the expression of both bcl-2 and bcl-xL in vitro and in vivo. Furthermore, this oligonucleotide demonstrated potent anti-tumor activity in vivo.

Maher and Dolnick (Nuc. Acid. Res. 16:3341-3358, 1988) tested a series of tandem 14 nucleobase antisense oligonucleotides, and a 28 and 42 nucleobase antisense oligonucleotides comprised of the sequence of two or three of the tandem antisense oligonucleotides, respectively, for their ability to arrest translation of human DHFR in a rabbit reticulocyte assay. Each of the three 14 nucleobase antisense oligonucleotides alone was able to inhibit translation, albeit at a more modest level than the 28 or 42 nucleobase antisense oligonucleotides.

Antisense Compound Motifs

In certain embodiments, antisense compounds targeted to a FGFR4 nucleic acid have chemically modified subunits arranged in patterns, or motifs, to confer to the antisense compounds properties such as enhanced inhibitory activity, increased binding affinity for a target nucleic acid, or resistance to degradation by in vivo nucleases.

Chimeric antisense compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, increased binding affinity for the target nucleic acid, and/or increased inhibitory activity. A second region of a chimeric antisense compound may optionally serve as a substrate for the cellular endonuclease RNase H, which cleaves the RNA strand of an RNA:DNA duplex.

Antisense compounds having a gapmer motif are considered chimeric antisense compounds. In a gapmer an internal region having a plurality of nucleotides that supports RNaseH cleavage is positioned between external regions having a plurality of nucleotides that are chemically distinct from the nucleosides of the internal region. In the case of an antisense oligonucleotide having a gapmer motif, the gap segment generally serves as the substrate for endonuclease cleavage, while the wing segments comprise modified nucleosides. In certain embodiments, the regions of a gapmer are differentiated by the types of sugar moieties comprising each distinct region. The types of sugar moieties that are used to differentiate the regions of a gapmer may in some embodiments include β-D-ribonucleosides, β-D-deoxyribonucleosides, 2′-modified nucleosides (such 2′-modified nucleosides may include 2′-MOE and 2′-O—CH3, among others), and bicyclic sugar modified nucleosides (such bicyclic sugar modified nucleosides may include those having a constrained ethyl). In certain embodiments, wings may include several modified sugar moieties, including, for example 2′-MOE and constrained ethyl. In certain embodiments, wings may include several modified and unmodified sugar moieties. In certain embodiments, wings may include various combinations of 2′-MOE nucleosides, constrained ethyl nucleosides, and 2′-deoxynucleosides.

Each distinct region may comprise uniform sugar moieties, variant, or alternating sugar moieties. The wing-gap-wing motif is frequently described as “X-Y-Z”, where “X” represents the length of the 5′-wing, “Y” represents the length of the gap, and “Z” represents the length of the 3′-wing. “X” and “Z” may comprise uniform, variant, or alternating sugar moieties. In certain embodiments, “X” and “Y” may include one or more 2′-deoxynucleosides.“Y” may comprise 2′-deoxynucleosides. As used herein, a gapmer described as “X-Y-Z” has a configuration such that the gap is positioned immediately adjacent to each of the 5′-wing and the 3′ wing. Thus, no intervening nucleotides exist between the 5′-wing and gap, or the gap and the 3′-wing. Any of the antisense compounds described herein can have a gapmer motif. In certain embodiments, “X” and “Z” are the same, in other embodiments they are different. In certain embodiments, “Y” is between 8 and 15 nucleosides. X, Y, or Z can be any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30 or more nucleosides.

In certain embodiments, antisense compounds targeted to a FGFR4 nucleic acid possess a 5-10-5 gapmer motif.

In certain embodiments, antisense compounds targeted to a FGFR4 nucleic acid possess a 3-10-4 gapmer motif.

Target Nucleic Acids, Target Regions and Nucleotide Sequences

In certain embodiments, the FGFR4 nucleic acid is any of the sequences set forth in GENBANK Accession No. NM002011.3 (incorporated herein as SEQ ID NO: 1), GENBANK Accession No: NT023133.11 truncated from nucleosides 21323018 to 21335213 (incorporated herein as SEQ ID NO: 2); GENBANK Accession No. AB209631.1 (incorporated herein as SEQ ID NO: 3) and GENBANK Accession No NM022963.2 (incorporated herein as SEQ ID NO: 4). In certain embodiments, FGFR4 has the rhesus monkey sequence as set forth in GENBANK Accession No. NW001121000.1 truncated from nucleosides 3094000 to 3109000 (SEQ ID NO: 5 In certain embodiments, FGFR4 has the murine sequence as set forth in GENBANK Accession No. BC033313.1 (SEQ ID NO: 6).

It is understood that the sequence set forth in each SEQ ID NO in the Examples contained herein is independent of any modification to a sugar moiety, an internucleoside linkage, or a nucleobase. As such, antisense compounds defined by a SEQ ID NO may comprise, independently, one or more modifications to a sugar moiety, an internucleoside linkage, or a nucleobase. Antisense compounds described by Isis Number (Isis No) indicate a combination of nucleobase sequence and motif.

In certain embodiments, a target region is a structurally defined region of the target nucleic acid. For example, a target region may encompass a 3′ UTR, a 5′ UTR, an exon, an intron, an exon/intron junction, a coding region, a translation initiation region, translation termination region, or other defined nucleic acid region. The structurally defined regions for FGFR4 can be obtained by accession number from sequence databases such as NCBI and such information is incorporated herein by reference. In certain embodiments, a target region may encompass the sequence from a 5′ target site of one target segment within the target region to a 3′ target site of another target segment within the same target region.

Targeting includes determination of at least one target segment to which an antisense compound hybridizes, such that a desired effect occurs. In certain embodiments, the desired effect is a reduction in mRNA target nucleic acid levels. In certain embodiments, the desired effect is reduction of levels of protein encoded by the target nucleic acid or a phenotypic change associated with the target nucleic acid.

A target region may contain one or more target segments. Multiple target segments within a target region may be overlapping. Alternatively, they may be non-overlapping. In certain embodiments, target segments within a target region are separated by no more than about 300 nucleotides. In certain embodiments, target segments within a target region are separated by a number of nucleotides that is, is about, is no more than, is no more than about, 250, 200, 150, 100, 90, 80, 70, 60, 50, 40, 30, 20, or 10 nucleotides on the target nucleic acid, or is a range defined by any two of the preceeding values. In certain embodiments, target segments within a target region are separated by no more than, or no more than about, 5 nucleotides on the target nucleic acid. In certain embodiments, target segments are contiguous. Contemplated are target regions defined by a range having a starting nucleic acid that is any of the 5′ target sites or 3′ target sites listed herein.

Suitable target segments may be found within a 5′ UTR, a coding region, a 3′ UTR, an intron, an exon, or an exon/intron junction. Target segments containing a start codon or a stop codon are also suitable target segments. A suitable target segment may specifically exclude a certain structurally defined region such as the start codon or stop codon.

The determination of suitable target segments may include a comparison of the sequence of a target nucleic acid to other sequences throughout the genome. For example, the BLAST algorithm may be used to identify regions of similarity amongst different nucleic acids. This comparison can prevent the selection of antisense compound sequences that may hybridize in a non-specific manner to sequences other than a selected target nucleic acid (i.e., non-target or off-target sequences).

There may be variation in activity (e.g., as defined by percent reduction of target nucleic acid levels) of the antisense compounds within an active target region. In certain embodiments, reductions in FGFR4 mRNA levels are indicative of inhibition of FGFR4 expression. Reductions in levels of a FGFR4 protein are also indicative of inhibition of target mRNA expression. Further, phenotypic changes are indicative of inhibition of FGFR4 expression. In certain embodiments, reduced glucose levels, reduced lipid levels, and reduced body weight can be indicative of inhibition of FGFR4 expression. In certain embodiments, amelioration of symptoms associated with metabolic disease can be indicative of inhibition of FGFR4 expression. In certain embodiments, amelioration of symptoms associated with diabetes can be indicative of inhibition of FGFR4 expression. In certain embodiments, reduction of insulin resistance is indicative of inhibition of FGFR4 expression. In certain embodiments, reduction of diabetes biomarkers can be indicative of inhibition of FGFR4 expression. In certain embodiments, reduction of FGFR4 expression is accompanied by an increase in FGF15 and/or FGF19 gene expression and/or an increase in FGF15 and/or FGF19 protein levels. In certain embodiments, reduction of FGFR4 expression is accompanied by an increase in ileum FGF15 and/or ileum FGF19 gene expression and plasma FGF15 and/or plasma FGF19 protein levels. Therefore, certain embodiments provide methods of measuring reduction of FGFR expression by measuring an increase in ileum FGF15 and/or ileum FGF19 gene expression and plasma FGF15 and/or plasma FGF19 protein levels. In certain embodiments, a biomarker of the anti-obesity effect of an FGFR4 inhibitor is an increase in FGF15 and/or FGF19 gene expression levels. In certain embodiments, a biomarker of the anti-obesity effect of an FGFR4 inhibitor is an increase in FGF15 and/or FGF19 protein levels. In certain embodiments, a biomarker of FGFR4 antisense oligonucleotide-caused anti-obesity effect is an increase in FGF15 and/or FGF19 gene expression levels. In certain embodiments, a biomarker of FGFR4 antisense oligonucleotide-caused anti-obesity effect is an increase in FGF15 and/or FGF19 protein levels. In certain embodiments, a biomarker of FGFR4 antisense oligonucleotide-caused anti-obesity effect is an increase in FGF15 and/or FGF19 gene expression levels.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal by measuring an increase in ileum FGF15 and/or ileum FGF19 gene expression and plasma FGF15 and/or plasma FGF19 protein levels.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF15 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF19 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF15 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods of detecting the anti-obesity effect of a FGFR4 inhibitor in an animal comprising: (a) measuring FGF19 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF15 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF19 gene expression in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 gene expression after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 gene expression. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor by measuring an increase in ileum FGF15 and/or ileum FGF19 gene expression and plasma FGF15 and/or plasma FGF19 protein levels.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF15 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF15 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF15 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Certain embodiments provide methods for predicting responsiveness of an animal to an FGFR4 inhibitor comprising: (a) measuring FGF19 protein levels in an individual prior to administration of a FGFR4 inhibitor (b) administering an FGFR4 inhibitor (c) measuring FGF19 protein levels after administration of a FGFR4 inhibitor (d) detecting an increase of FGF19 protein levels. In certain embodiments, the FGFR4 inhibitor is a modified antisense oligonucleotide targeted to FGFR4.

Hybridization

In some embodiments, hybridization occurs between an antisense compound disclosed herein and a FGFR4 nucleic acid. The most common mechanism of hybridization involves hydrogen bonding (e.g., Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding) between complementary nucleobases of the nucleic acid molecules.

Hybridization can occur under varying conditions. Stringent conditions are sequence-dependent and are determined by the nature and composition of the nucleic acid molecules to be hybridized.

Methods of determining whether a sequence is specifically hybridizable to a target nucleic acid are well known in the art. In certain embodiments, the antisense compounds provided herein are specifically hybridizable with a FGFR4 nucleic acid.

Complementarity

An antisense compound and a target nucleic acid are complementary to each other when a sufficient number of nucleobases of the antisense compound can hydrogen bond with the corresponding nucleobases of the target nucleic acid, such that a desired effect will occur (e.g., antisense inhibition of a target nucleic acid, such as a FGFR4 nucleic acid).

An antisense compound may hybridize over one or more segments of a FGFR4 nucleic acid such that intervening or adjacent segments are not involved in the hybridization event (e.g., a loop structure, mismatch or hairpin structure).

In certain embodiments, the antisense compounds provided herein, or a specified portion thereof, are, or are at least, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% complementary to a FGFR4 nucleic acid, a target region, target segment, or specified portion thereof. Percent complementarity of an antisense compound with a target nucleic acid can be determined using routine methods.

For example, an antisense compound in which 18 of 20 nucleobases of the antisense compound are complementary to a target region, and would therefore specifically hybridize, would represent 90 percent complementarity. In this example, the remaining non-complementary nucleobases may be clustered or interspersed with complementary nucleobases and need not be contiguous to each other or to complementary nucleobases. As such, an antisense compound which is 18 nucleobases in length having 4 (four) non-complementary nucleobases which are flanked by two regions of complete complementarity with the target nucleic acid would have 77.8% overall complementarity with the target nucleic acid and would thus fall within the scope of the present invention. Percent complementarity of an antisense compound with a region of a target nucleic acid can be determined routinely using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. Mol. Biol., 1990, 215, 403 410; Zhang and Madden, Genome Res., 1997, 7, 649 656). Percent homology, sequence identity or complementarity, can be determined by, for example, the Gap program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.), using default settings, which uses the algorithm of Smith and Waterman (Adv. Appl. Math., 1981, 2, 482 489).

In certain embodiments, the antisense compounds provided herein, or specified portions thereof, are fully complementary (i.e. 100% complementary) to a target nucleic acid, or specified portion thereof. For example, antisense compound may be fully complementary to a FGFR4 nucleic acid, or a target region, or a target segment or target sequence thereof. As used herein, “fully complementary” means each nucleobase of an antisense compound is capable of precise base pairing with the corresponding nucleobases of a target nucleic acid. For example, a 20 nucleobase antisense compound is fully complementary to a target sequence that is 400 nucleobases long, so long as there is a corresponding 20 nucleobase portion of the target nucleic acid that is fully complementary to the antisense compound. Fully complementary can also be used in reference to a specified portion of the first and/or the second nucleic acid. For example, a 20 nucleobase portion of a 30 nucleobase antisense compound can be “fully complementary” to a target sequence that is 400 nucleobases long. The 20 nucleobase portion of the 30 nucleobase oligonucleotide is fully complementary to the target sequence if the target sequence has a corresponding 20 nucleobase portion wherein each nucleobase is complementary to the 20 nucleobase portion of the antisense compound. At the same time, the entire 30 nucleobase antisense compound may or may not be fully complementary to the target sequence, depending on whether the remaining 10 nucleobases of the antisense compound are also complementary to the target sequence.

The location of a non-complementary nucleobase may be at the 5′ end or 3′ end of the antisense compound. Alternatively, the non-complementary nucleobase or nucleobases may be at an internal position of the antisense compound. When two or more non-complementary nucleobases are present, they may be contiguous (i.e. linked) or non-contiguous. In one embodiment, a non-complementary nucleobase is located in the wing segment of a gapmer antisense oligonucleotide.

In certain embodiments, antisense compounds that are, or are up to 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleobases in length comprise no more than 4, no more than 3, no more than 2, or no more than 1 non-complementary nucleobase(s) relative to a target nucleic acid, such as a FGFR4 nucleic acid, or specified portion thereof.

In certain embodiments, antisense compounds that are, or are up to 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleobases in length comprise no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 non-complementary nucleobase(s) relative to a target nucleic acid, such as a FGFR4 nucleic acid, or specified portion thereof.

The antisense compounds provided herein also include those which are complementary to a portion of a target nucleic acid. As used herein, “portion” refers to a defined number of contiguous (i.e. linked) nucleobases within a region or segment of a target nucleic acid. A “portion” can also refer to a defined number of contiguous nucleobases of an antisense compound. In certain embodiments, the antisense compounds, are complementary to at least an 8 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 12 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 13 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 14 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 15 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 16 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 17 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 18 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 19 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 20 nucleobase portion of a target segment. Also contemplated are antisense compounds that are complementary to at least a 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleobase portion of a target segment, or a range defined by any two of these values.

Identity

The antisense compounds provided herein may also have a defined percent identity to a particular nucleotide sequence, SEQ ID NO, or compound represented by a specific Isis number, or portion thereof. As used herein, an antisense compound is identical to the sequence disclosed herein if it has the same nucleobase pairing ability. For example, a RNA which contains uracil in place of thymidine in a disclosed DNA sequence would be considered identical to the DNA sequence since both uracil and thymidine pair with adenine. Shortened and lengthened versions of the antisense compounds described herein as well as compounds having non-identical bases relative to the antisense compounds provided herein also are contemplated. The non-identical bases may be adjacent to each other or dispersed throughout the antisense compound. Percent identity of an antisense compound is calculated according to the number of bases that have identical base pairing relative to the sequence to which it is being compared.

In certain embodiments, the antisense compounds, or portions thereof, are at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to one or more of the antisense compounds or SEQ ID NOs, or a portion thereof, disclosed herein.

Modifications

A nucleoside is a base-sugar combination. The nucleobase (also known as base) portion of the nucleoside is normally a heterocyclic base moiety. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to the 2′, 3′ or 5′ hydroxyl moiety of the sugar. Oligonucleotides are formed through the covalent linkage of adjacent nucleosides to one another, to form a linear polymeric oligonucleotide. Within the oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside linkages of the oligonucleotide.

Modifications to antisense compounds encompass substitutions or changes to internucleoside linkages, sugar moieties, or nucleobases. Modified antisense compounds are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target, increased stability in the presence of nucleases, or increased inhibitory activity.

Chemically modified nucleosides may also be employed to increase the binding affinity of a shortened or truncated antisense oligonucleotide for its target nucleic acid. Consequently, comparable results can often be obtained with shorter antisense compounds that have such chemically modified nucleosides.

Modified Internucleoside Linkages

The naturally occurring internucleoside linkage of RNA and DNA is a 3′ to 5′ phosphodiester linkage. Antisense compounds having one or more modified, i.e. non-naturally occurring, internucleoside linkages are often selected over antisense compounds having naturally occurring internucleoside linkages because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for target nucleic acids, and increased stability in the presence of nucleases.

Oligonucleotides having modified internucleoside linkages include internucleoside linkages that retain a phosphorus atom as well as internucleoside linkages that do not have a phosphorus atom. Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters, phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing linkages are well known.

In certain embodiments, antisense compounds targeted to a FGFR4 nucleic acid comprise one or more modified internucleoside linkages. In certain embodiments, the modified internucleoside linkages are phosphorothioate linkages. In certain embodiments, each internucleoside linkage of an antisense compound is a phosphorothioate internucleoside linkage.

Modified Sugar Moieties

Antisense compounds provided herein can optionally contain one or more nucleosides wherein the sugar group has been modified. Such sugar modified nucleosides may impart enhanced nuclease stability, increased binding affinity, or some other beneficial biological property to the antisense compounds. In certain embodiments, nucleosides comprise a chemically modified ribofuranose ring moiety. Examples of chemically modified ribofuranose rings include, without limitation, addition of substitutent groups (including 5′ and 2′ substituent groups); bridging of non-geminal ring atoms to form bicyclic nucleic acids (BNA); replacement of the ribosyl ring oxygen atom with S, N(R), or C(R1)(R)2 (R=H, C1-C12 alkyl or a protecting group); and combinations thereof. Examples of chemically modified sugars include, 2′-F-5′-methyl substituted nucleoside (see, PCT International Application WO 2008/101157, published on Aug. 21, 2008 for other disclosed 5′,2′-bis substituted nucleosides), replacement of the ribosyl ring oxygen atom with S with further substitution at the 2′-position (see, published U.S. Patent Application US2005/0130923, published on Jun. 16, 2005), or, alternatively, 5′-substitution of a BNA (see, PCT International Application WO 2007/134181, published on Nov. 22, 2007, wherein LNA is substituted with, for example, a 5′-methyl or a 5′-vinyl group).

Examples of nucleosides having modified sugar moieties include, without limitation, nucleosides comprising 5′-vinyl, 5′-methyl (R or S), 4′-S, 2′-F, 2′-OCH3, and 2′-O(CH2)2OCH3 substituent groups. The substituent at the 2′ position can also be selected from allyl, amino, azido, thio, O-allyl, O—C1-C10 alkyl, OCF3, O(CH2)2SCH3, O(CH2)2-O—N(Rm)(Rn), and O—CH2—C(═O)—N(Rm)(Rn), where each Rm and Rn is, independently, H or substituted or unsubstituted C1-C10 alkyl.

As used herein, “bicyclic nucleosides” refer to modified nucleosides comprising a bicyclic sugar moiety. Examples of bicyclic nucleosides include, without limitation, nucleosides comprising a bridge between the 4′ and the 2′ ribosyl ring atoms. In certain embodiments, antisense compounds provided herein include one or more bicyclic nucleosides wherein the bridge comprises a 4′ to 2′ bicyclic nucleoside. Examples of such 4′ to 2′ bicyclic nucleosides, include, but are not limited to, one of the formulae: 4′-(CH2)—O-2′ (LNA); 4′-(CH2)—S-2; 4′-(CH2)2—O-2′ (ENA); 4′-CH(CH3)—O-2′ and 4′-CH(CH2OCH3)—O-2′, and analogs thereof (see, U.S. Pat. No. 7,399,845, issued on Jul. 15, 2008); 4′-C(CH3)(CH3)—O-2′, and analogs thereof (see, published PCT International Application WO2009/006478, published Jan. 8, 2009); 4′-CH2—N(OCH3)-2′, and analogs thereof (see, published PCT International Application WO2008/150729, published Dec. 11, 2008); 4′-CH2—O—N(CH3)-2′ (see, published U.S. Patent Application US2004/0171570, published Sep. 2, 2004); 4′-CH2—N(R)—O-2′, wherein R is H, C1-C12 alkyl, or a protecting group (see, U.S. Pat. No. 7,427,672, issued on Sep. 23, 2008); 4′-CH2—C(H)(CH3)-2′ (see, Chattopadhyaya, et al., J. Org. Chem., 2009, 74, 118-134); and 4′-CH2—C(═CH2)-2′, and analogs thereof (see, published PCT International Application WO 2008/154401, published on Dec. 8, 2008). Also see, for example: Singh et al., Chem. Commun., 1998, 4, 455-456; Koshkin et al., Tetrahedron, 1998, 54, 3607-3630; Wahlestedt et al., Proc. Natl. Acad. Sci. U.S.A., 2000, 97, 5633-5638; Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222; Singh et al., J. Org. Chem., 1998, 63, 10035-10039; Srivastava et al., J. Am. Chem. Soc., 129(26) 8362-8379 (Jul. 4, 2007); Elayadi et al., Curr. Opinion Invens. Drugs, 2001, 2, 558-561; Braasch et al., Chem. Biol., 2001, 8, 1-7; Orum et al., Curr. Opinion Mol. Ther., 2001, 3, 239-243; U.S. Pat. Nos. 6,670,461, 7,053,207, 6,268,490, 6,770,748, 6,794,499, 7,034,133, 6,525,191, 7,399,845; published PCT International applications WO 2004/106356, WO 94/14226, WO 2005/021570, and WO 2007/134181; U.S. Patent Publication Nos. US2004/0171570, US2007/0287831, and US2008/0039618; and U.S. patent Ser. Nos. 12/129,154, 60/989,574, 61/026,995, 61/026,998, 61/056,564, 61/086,231, 61/097,787, and 61/099,844; and PCT International Application Nos. PCT/US2008/064591, PCT/US2008/066154, and PCT/US2008/068922. Each of the foregoing bicyclic nucleosides can be prepared having one or more stereochemical sugar configurations including for example α-L-ribofuranose and β-D-ribofuranose (see PCT international application PCT/DK98/00393, published on Mar. 25, 1999 as WO 99/14226).

In certain embodiments, bicyclic sugar moieties of BNA nucleosides include, but are not limited to, compounds having at least one bridge between the 4′ and the 2′ position of the pentofuranosyl sugar moiety wherein such bridges independently comprises 1 or from 2 to 4 linked groups independently selected from —[C(Ra)(Rb)]n—, —C(Ra)═C(Rb)—, —C(Ra)═N—, —C(═NRa)—, —C(═O)—, —C(═S)—, —O—, —Si(Ra)2—, —S(═O)x—, and —N(Ra)—;

wherein:

x is 0, 1, or 2;

n is 1, 2, 3, or 4;

each Ra and Rb is, independently, H, a protecting group, hydroxyl, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, heterocycle radical, substituted heterocycle radical, heteroaryl, substituted heteroaryl, C5-C7 alicyclic radical, substituted C5-C7 alicyclic radical, halogen, OJ1, NJ1J2, SJ1, N3, COOJ1, acyl (C(═O)—H), substituted acyl, CN, sulfonyl (S(═O)2-J1), or sulfoxyl (S(═O)-J1); and

each J1 and J2 is, independently, H, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, acyl (C(═O)—H), substituted acyl, a heterocycle radical, a substituted heterocycle radical, C1-C12 aminoalkyl, substituted C1-C12 aminoalkyl, or a protecting group.

In certain embodiments, the bridge of a bicyclic sugar moiety is, —[C(Ra)(Rb)]n—, —[C(Ra)(Rb)]n—O—, —C(RaRb)—N(R)—O— or, —C(RaRb)—O—N(R)—. In certain embodiments, the bridge is 4′-CH2-2′, 4′-(CH2)2-2′, 4′-(CH2)3-2′, 4′-CH2—O-2′, 4′-(CH2)2—O-2′, 4′-CH2—O—N(R)-2′, and 4′-CH2—N(R)—O-2′-, wherein each R is, independently, H, a protecting group, or C1-C12 alkyl.

In certain embodiments, bicyclic nucleosides are further defined by isomeric configuration. For example, a nucleoside comprising a 4′-2′ methylene-oxy bridge, may be in the α-L configuration or in the β-D configuration. Previously, α-L-methyleneoxy (4′-CH2—O-2′) BNA's have been incorporated into antisense oligonucleotides that showed antisense activity (Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372).

In certain embodiments, bicyclic nucleosides include, but are not limited to, (A) α-L-Methyleneoxy (4′-CH2—O-2′) BNA, (B) β-D-Methyleneoxy (4′-CH2—O-2′) BNA, (C) Ethyleneoxy (4′-(CH2)2—O-2′) BNA, (D) Aminooxy (4′-CH2—O—N(R)-2′) BNA, (E) Oxyamino (4′-CH2—N(R)—O-2′) BNA, (F) Methyl(methyleneoxy) (4′-CH(CH3)—O-2′) BNA, (G) methylene-thio (4′-CH2—S-2′) BNA, (H) methylene-amino (4′-CH2-N(R)-2′) BNA, (I) methyl carbocyclic (4′-CH2—CH(CH3)-2′) BNA, and (J) propylene carbocyclic (4′-(CH2)3-2′) BNA as depicted below.

wherein Bx is the base moiety and R is, independently, H, a protecting group or C1-C12 alkyl.

In certain embodiments, bicyclic nucleoside having Formula I:

wherein:

Bx is a heterocyclic base moiety;

-Qa-Qb-Qc- is —CH2—N(Rc)—CH2—, —C(═O)—N(Rc)—CH2—, —CH2—O—N(Rc)—, —CH2—N(Rc)—O—, or —N(Rc)—O—CH2;

Rc is C1-C12 alkyl or an amino protecting group; and

Ta and Tb are each, independently, H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety, or a covalent attachment to a support medium.

In certain embodiments, bicyclic nucleoside having Formula II:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently, H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety, or a covalent attachment to a support medium;

Za is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, substituted C1-C6 alkyl, substituted C2-C6 alkenyl, substituted C2-C6 alkynyl, acyl, substituted acyl, substituted amide, thiol, or substituted thio.

In one embodiment, each of the substituted groups is, independently, mono or poly substituted with substituent groups independently selected from halogen, oxo, hydroxyl, OJc, NJcJd, SJc, N3, OC(═X)Jc, and NJeC(═X)NJcJd, wherein each Jc, Jd, and Je is, independently, H, C1-C6 alkyl, or substituted C1-C6 alkyl and X is O or NJe.

In certain embodiments, bicyclic nucleoside having Formula III:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently, H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety, or a covalent attachment to a support medium;

Zb is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, substituted C1-C6 alkyl, substituted C2-C6 alkenyl, substituted C2-C6 alkynyl, or substituted acyl (C(═O)—).

In certain embodiments, bicyclic nucleoside having Formula IV:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety, or a covalent attachment to a support medium;

Rd is C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl;

each qa, qb, qc and qd is, independently, H, halogen, C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl, C1-C6 alkoxyl, substituted C1-C6 alkoxyl, acyl, substituted acyl, C1-C6 aminoalkyl, or substituted C1-C6 aminoalkyl;

In certain embodiments, bicyclic nucleoside having Formula V:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently, H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety, or a covalent attachment to a support medium;

qa, qb, qe and qf are each, independently, hydrogen, halogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C1-C12 alkoxy, substituted C1-C12 alkoxy, OJj, SJj, SOJj, SO2Jj, NJjJk, N3, CN, C(═O)OJj, C(═O)NJjJk, C(═O)Jj, O—C(═O)NJjJk, N(H)C(═NH)NJjJk, N(H)C(═O)NJjJk or N(H)C(═S)NJjJk;

or qe and qf together are ═C(qg)(qh);

qg and qh are each, independently, H, halogen, C1-C12 alkyl, or substituted C1-C12 alkyl.

The synthesis and preparation of the methyleneoxy (4′-CH2—O-2′) BNA monomers adenine, cytosine, guanine, 5-methyl-cytosine, thymine, and uracil, along with their oligomerization, and nucleic acid recognition properties have been described (see, e.g., Koshkin et al., Tetrahedron, 1998, 54, 3607-3630). BNAs and preparation thereof are also described in WO 98/39352 and WO 99/14226.

Analogs of methyleneoxy (4′-CH2—O-2′) BNA, methyleneoxy (4′-CH2—O-2′) BNA, and 2′-thio-BNAs, have also been prepared (see, e.g., Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222). Preparation of locked nucleoside analogs comprising oligodeoxyribonucleotide duplexes as substrates for nucleic acid polymerases has also been described (see, e.g., Wengel et al., WO 99/14226). Furthermore, synthesis of 2′-amino-BNA, a novel comformationally restricted high-affinity oligonucleotide analog, has been described in the art (see, e.g., Singh et al., J. Org. Chem., 1998, 63, 10035-10039). In addition, 2′-amino- and 2′-methylamino-BNA's have been prepared and the thermal stability of their duplexes with complementary RNA and DNA strands has been previously reported.

In certain embodiments, bicyclic nucleoside having Formula VI:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently, H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety, or a covalent attachment to a support medium;

each qi, qj, qk and ql is, independently, H, halogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C1-C12 alkoxyl, substituted C1-C12 alkoxyl, OJj, SJj, SOJj, SO2Jj, NJjJk, N3, CN, C(═O)OJj, C(═O)NJjJk, C(═O)Jj, O—C(═O)NJjJk, N(H)C(═NH)NJjJk, N(H)C(═O)NJjJk, or N(H)C(═S)NJjJk; and

qi and qj or ql and qk together are ═C(qg)(qh), wherein qg and qh are each, independently, H, halogen, C1-C12 alkyl, or substituted C1-C12 alkyl.

One carbocyclic bicyclic nucleoside having a 4′-(CH2)3-2′ bridge and the alkenyl analog, bridge 4′-CH═CH—CH2-2′, have been described (see, e.g., Freier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., J. Org. Chem., 2006, 71, 7731-7740). The synthesis and preparation of carbocyclic bicyclic nucleosides along with their oligomerization and biochemical studies have also been described (see, e.g., Srivastava et al., J. Am. Chem. Soc. 2007, 129(26), 8362-8379).

As used herein, “4′-2′ bicyclic nucleoside” or “4′ to 2′ bicyclic nucleoside” refers to a bicyclic nucleoside comprising a furanose ring comprising a bridge connecting the 2′ carbon atom and the 4′ carbon atom.

As used herein, “monocylic nucleosides” refer to nucleosides comprising modified sugar moieties that are not bicyclic sugar moieties. In certain embodiments, the sugar moiety, or sugar moiety analogue, of a nucleoside may be modified or substituted at any position.

As used herein, “2′-modified sugar” means a furanosyl sugar modified at the 2′ position. In certain embodiments, such modifications include substituents selected from: a halide, including, but not limited to substituted and unsubstituted alkoxy, substituted and unsubstituted thioalkyl, substituted and unsubstituted amino alkyl, substituted and unsubstituted alkyl, substituted and unsubstituted allyl, and substituted and unsubstituted alkynyl. In certain embodiments, 2′ modifications are selected from substituents including, but not limited to: O[(CH2)mO]mCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, OCH2C(═O)N(H)CH3, and O(CH2)nON[(CH2)nCH3]2, where n and m are from 1 to about 10. Other 2′-substituent groups can also be selected from: C1-C12 alkyl; substituted alkyl; alkenyl; alkynyl; alkaryl; aralkyl; O-alkaryl or O-aralkyl; SH; SCH3; OCN; Cl; Br; CN; CF3; OCF3; SOCH3; SO2CH3; ONO2; NO2; N3; NH2; heterocycloalkyl; heterocycloalkaryl; aminoalkylamino; polyalkylamino; substituted silyl; an RNA cleaving group; a reporter group; an intercalator; a group for improving pharmacokinetic properties; and a group for improving the pharmacodynamic properties of an antisense compound, and other substituents having similar properties. In certain embodiments, modifed nucleosides comprise a 2′-MOE side chain (see, e.g., Baker et al., J. Biol. Chem., 1997, 272, 11944-12000). Such 2′-MOE substitution have been described as having improved binding affinity compared to unmodified nucleosides and to other modified nucleosides, such as 2′-O-methyl, O-propyl, and O-aminopropyl. Oligonucleotides having the 2′-MOE substituent also have been shown to be antisense inhibitors of gene expression with promising features for in vivo use (see, e.g., Martin, P., Helv. Chim. Acta, 1995, 78, 486-504; Altmann et al., Chimia, 1996, 50, 168-176; Altmann et al., Biochem. Soc. Trans., 1996, 24, 630-637; and Altmann et al., Nucleosides Nucleotides, 1997, 16, 917-926).

As used herein, a “modified tetrahydropyran nucleoside” or “modified THP nucleoside” means a nucleoside having a six-membered tetrahydropyran “sugar” substituted in for the pentofuranosyl residue in normal nucleosides (a sugar surrogate). Modified THP nucleosides include, but are not limited to, what is referred to in the art as hexitol nucleic acid (HNA), anitol nucleic acid (ANA), manitol nucleic acid (MNA) (see Leumann, C J. Bioorg. & Med. Chem. (2002) 10:841-854), fluoro HNA (F-HNA), or those compounds having Formula X:

Formula X:

wherein independently for each of said at least one tetrahydropyran nucleoside analog of Formula X:

Bx is a heterocyclic base moiety;

T3 and T4 are each, independently, an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound or one of T3 and T4 is an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound and the other of T3 and T4 is H, a hydroxyl protecting group, a linked conjugate group, or a 5′ or 3′-terminal group;

q1, q2, q3, q4, q5, q6 and q7 are each, independently, H, C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl, or substituted C2-C6 alkynyl; and

one of R1 and R2 is hydrogen and the other is selected from halogen, substituted or unsubstituted alkoxy, NJ1J2, SJ1, N3, OC(═X)J1, OC(═X)NJ1J2, NJ3C(═X)NJ1J2, and CN, wherein X is O, S, or NJ1, and each J1, J2, and J3 is, independently, H or C1-C6 alkyl.

In certain embodiments, the modified THP nucleosides of Formula X are provided wherein qm, qn, qp, qr, qs, qt, and qu are each H. In certain embodiments, at least one of qm, qn, qp, qr, qs, qt, and qu is other than H. In certain embodiments, at least one of qm, qn, qp, qr, qs, qt and qu is methyl. In certain embodiments, THP nucleosides of Formula X are provided wherein one of R1 and R2 is F. In certain embodiments, R1 is fluoro and R2 is H, R1 is methoxy and R2 is H, and R1 is methoxyethoxy and R2 is H.

As used herein, “2′-modified” or “2′-substituted” refers to a nucleoside comprising a sugar comprising a substituent at the 2′ position other than H or OH. 2′-modified nucleosides, include, but are not limited to, bicyclic nucleosides wherein the bridge connecting two carbon atoms of the sugar ring connects the 2′ carbon and another carbon of the sugar ring and nucleosides with non-bridging 2′ substituents, such as allyl, amino, azido, thio, O-allyl, O—C1-C10 alkyl, —OCF3, O—(CH2)2—O—CH3, 2′-O(CH2)2SCH3, O—(CH2)2—O—N(Rm)(Rn), or O—CH2—C(═O)—N(Rm)(Rn), where each Rm and Rn is, independently, H or substituted or unsubstituted C1-C10 alkyl. 2′-modifed nucleosides may further comprise other modifications, for example, at other positions of the sugar and/or at the nucleobase.

As used herein, “2′-F” refers to a sugar comprising a fluoro group at the 2′ position.

As used herein, “2′-OMe” or “2′-OCH3” or “2′-O-methyl” each refers to a sugar comprising an —OCH3 group at the 2′ position of the sugar ring.

As used herein, “oligonucleotide” refers to a compound comprising a plurality of linked nucleosides. In certain embodiments, one or more of the plurality of nucleosides is modified. In certain embodiments, an oligonucleotide comprises one or more ribonucleosides (RNA) and/or deoxyribonucleosides (DNA).

Many other bicyclo and tricyclo sugar surrogate ring systems are also known in the art that can be used to modify nucleosides for incorporation into antisense compounds (see, e.g., review article: Leumann, J. C, Bioorganic & Medicinal Chemistry, 2002, 10, 841-854).

Such ring systems can undergo various additional substitutions to enhance activity.

Methods for the preparations of modified sugars are well known to those skilled in the art.

In nucleotides having modified sugar moieties, the nucleobase moieties (natural, modified, or a combination thereof) are maintained for hybridization with an appropriate nucleic acid target.

In certain embodiments, antisense compounds comprise one or more nucleotides having modified sugar moieties. In certain embodiments, the modified sugar moiety is 2′-MOE. In certain embodiments, the 2′-MOE modified nucleotides are arranged in a gapmer motif. In certain embodiments, the modified sugar moiety is a cEt. In certain embodiments, the cEt modified nucleotides are arranged throughout the wings of a gapmer motif

Modified Nucleobases

Nucleobase (or base) modifications or substitutions are structurally distinguishable from, yet functionally interchangeable with, naturally occurring or synthetic unmodified nucleobases. Both natural and modified nucleobases are capable of participating in hydrogen bonding. Such nucleobase modifications may impart nuclease stability, binding affinity or some other beneficial biological property to antisense compounds. Modified nucleobases include synthetic and natural nucleobases such as, for example, 5-methylcytosine (5-me-C). Certain nucleobase substitutions, including 5-methylcytosine substitutions, are particularly useful for increasing the binding affinity of an antisense compound for a target nucleic acid. For example, 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278).

Additional unmodified nucleobases include 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (—C≡C—CH3) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine.

Heterocyclic base moieties may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Nucleobases that are particularly useful for increasing the binding affinity of antisense compounds include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2 aminopropyladenine, 5-propynyluracil and 5-propynylcytosine.

In certain embodiments, antisense compounds targeted to a FGFR4 nucleic acid comprise one or more modified nucleobases. In certain embodiments, gap-widened antisense oligonucleotides targeted to a FGFR4 nucleic acid comprise one or more modified nucleobases. In certain embodiments, the modified nucleobase is 5-methylcytosine. In certain embodiments, each cytosine is a 5-methylcytosine.

Compositions and Methods for Formulating Pharmaceutical Compositions

Antisense oligonucleotides may be admixed with pharmaceutically acceptable active or inert substance for the preparation of pharmaceutical compositions or formulations. Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.

Antisense compound targeted to a FGFR4 nucleic acid can be utilized in pharmaceutical compositions by combining the antisense compound with a suitable pharmaceutically acceptable diluent or carrier. A pharmaceutically acceptable diluent includes phosphate-buffered saline (PBS). PBS is a diluent suitable for use in compositions to be delivered parenterally. Accordingly, in one embodiment, employed in the methods described herein is a pharmaceutical composition comprising an antisense compound targeted to a FGFR4 nucleic acid and a pharmaceutically acceptable diluent. In certain embodiments, the pharmaceutically acceptable diluent is PBS. In certain embodiments, the antisense compound is an antisense oligonucleotide.

Pharmaceutical compositions comprising antisense compounds encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other oligonucleotide which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to pharmaceutically acceptable salts of antisense compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents. Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.

Pharmaceutically acceptable salts of the compounds described herein may be prepared by methods well-known in the art. For a review of pharmaceutically acceptable salts, see Stahl and Wermuth, Handbook of Pharmaceutical Salts: Properties, Selection and Use (Wiley-VCH, Weinheim, Germany, 2002). Sodium salts of antisense oligonucleotides are useful and are well accepted for therapeutic administration to humans. Accordingly, in one embodiment the compounds described herein are in the form of a sodium salt.

A prodrug can include the incorporation of additional nucleosides at one or both ends of an antisense compound which are cleaved by endogenous nucleases within the body, to form the active antisense compound.

Conjugated Antisense Compounds

Antisense compounds may be covalently linked to one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the resulting antisense oligonucleotides. Typical conjugate groups include cholesterol moieties and lipid moieties. Additional conjugate groups include carbohydrates, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.

Antisense compounds can also be modified to have one or more stabilizing groups that are generally attached to one or both termini of antisense compounds to enhance properties such as, for example, nuclease stability. Included in stabilizing groups are cap structures. These terminal modifications protect the antisense compound having terminal nucleic acid from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5′-terminus (5′-cap), or at the 3′-terminus (3′-cap), or can be present on both termini. Cap structures are well known in the art and include, for example, inverted deoxy abasic caps. Further 3′ and 5′-stabilizing groups that can be used to cap one or both ends of an antisense compound to impart nuclease stability include those disclosed in WO 03/004602 published on Jan. 16, 2003.

Cell Culture and Antisense Compounds Treatment

The effects of antisense compounds on the level, activity or expression of FGFR4 nucleic acids can be tested in vitro in a variety of cell types. Cell types used for such analyses are available from commercial vendors (e.g. American Type Culture Collection, Manassas, Va.; Zen-Bio, Inc., Research Triangle Park, NC; Clonetics Corporation, Walkersville, Md.) and cells are cultured according to the vendor's instructions using commercially available reagents (e.g. Invitrogen Life Technologies, Carlsbad, Calif.). Illustrative cell types include, but are not limited to, HepG2 cells and primary hepatocytes.

In Vitro Testing of Antisense Oligonucleotides

Described herein are methods for treatment of cells with antisense oligonucleotides, which can be modified appropriately for treatment with other antisense compounds.

In general, cells are treated with antisense oligonucleotides when the cells reach approximately 60-80% confluence in culture.

One reagent commonly used to introduce antisense oligonucleotides into cultured cells includes the cationic lipid transfection reagent LIPOFECTIN® (Invitrogen, Carlsbad, Calif.). Antisense oligonucleotides are mixed with LIPOFECTIN® in OPTI-MEM® 1 (Invitrogen, Carlsbad, Calif.) to achieve the desired final concentration of antisense oligonucleotide and a LIPOFECTIN® concentration that typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.

Another reagent used to introduce antisense oligonucleotides into cultured cells includes LIPOFECTAMINE 2000® (Invitrogen, Carlsbad, Calif.). Antisense oligonucleotide is mixed with LIPOFECTAMINE 2000® in OPTI-MEM® 1 reduced serum medium (Invitrogen, Carlsbad, Calif.) to achieve the desired concentration of antisense oligonucleotide and a LIPOFECTAMINE® concentration that typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.

Another reagent used to introduce antisense oligonucleotides into cultured cells includes Cytofectin® (Invitrogen, Carlsbad, Calif.). Antisense oligonucleotide is mixed with Cytofectin® in OPTI-MEM® 1 reduced serum medium (Invitrogen, Carlsbad, Calif.) to achieve the desired concentration of antisense oligonucleotide and a Cytofectin® concentration that typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.

Another technique used to introduce antisense oligonucleotides into cultured cells includes electroporation.

Cells are treated with antisense oligonucleotides by routine methods. Cells are typically harvested 16-24 hours after antisense oligonucleotide treatment, at which time RNA or protein levels of target nucleic acids are measured by methods known in the art and described herein. In general, when treatments are performed in multiple replicates, the data are presented as the average of the replicate treatments.

The concentration of antisense oligonucleotide used varies from cell line to cell line. Methods to determine the optimal antisense oligonucleotide concentration for a particular cell line are well known in the art. Antisense oligonucleotides are typically used at concentrations ranging from 1 nM to 300 nM when transfected with LIPOFECTAMINE2000®, Lipofectin or Cytofectin. Antisense oligonucleotides are used at higher concentrations ranging from 625 to 20,000 nM when transfected using electroporation.

RNA Isolation

RNA analysis can be performed on total cellular RNA or poly(A)+mRNA. Methods of RNA isolation are well known in the art. RNA is prepared using methods well known in the art, for example, using the TRIZOL® Reagent (Invitrogen, Carlsbad, Calif.) according to the manufacturer's recommended protocols.

Analysis of Inhibition of Target Levels or Expression

Inhibition of levels or expression of a FGFR4 nucleic acid can be assayed in a variety of ways known in the art. For example, target nucleic acid levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or quantitative real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. Northern blot analysis is also routine in the art. Quantitative real-time PCR can be conveniently accomplished using the commercially available ABI PRISM® 7600, 7700, or 7900 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif. and used according to manufacturer's instructions.

Quantitative Real-Time PCR Analysis of Target RNA Levels

Quantitation of target RNA levels may be accomplished by quantitative real-time PCR using the ABI PRISM® 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions. Methods of quantitative real-time PCR are well known in the art.

Prior to real-time PCR, the isolated RNA is subjected to a reverse transcriptase (RT) reaction, which produces complementary DNA (cDNA) that is then used as the substrate for the real-time PCR amplification. The RT and real-time PCR reactions are performed sequentially in the same sample well. RT and real-time PCR reagents are obtained from Invitrogen (Carlsbad, Calif.). RT, real-time-PCR reactions are carried out by methods well known to those skilled in the art.

Gene (or RNA) target quantities obtained by real time PCR are normalized using either the expression level of a gene whose expression is constant, such as cyclophilin A, or by quantifying total RNA using RIBOGREEN® (Invitrogen, Inc. Carlsbad, Calif.). Cyclophilin A expression is quantified by real time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RIBOGREEN® RNA quantification reagent (Invitrogen, Inc. Eugene, Oreg.). Methods of RNA quantification by RIBOGREEN® are taught in Jones, L. J., et al, (Analytical Biochemistry, 1998, 265, 368-374). A CYTOFLUOR® 4000 instrument (PE Applied Biosystems) is used to measure RIBOGREEN® fluorescence.

Probes and primers are designed to hybridize to a FGFR4 nucleic acid. Methods for designing real-time PCR probes and primers are well known in the art, and may include the use of software such as PRIMER EXPRESS® Software (Applied Biosystems, Foster City, Calif.).

Analysis of Protein Levels

Antisense inhibition of FGFR4 nucleic acids can be assessed by measuring FGFR4 protein levels. Protein levels of FGFR4 can be evaluated or quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked immunosorbent assay (ELISA), quantitative protein assays, protein activity assays (for example, caspase activity assays), immunohistochemistry, immunocytochemistry or fluorescence-activated cell sorting (FACS). Antibodies directed to a target can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, Mich.), or can be prepared via conventional monoclonal or polyclonal antibody generation methods well known in the art. Antibodies useful for the detection of human and rat FGFR4 are commercially available.

In Vivo Testing of Antisense Compounds

Antisense compounds, for example, antisense oligonucleotides, are tested in animals to assess their ability to inhibit expression of FGFR4 and produce phenotypic changes. Testing may be performed in normal animals, or in experimental disease models. For administration to animals, antisense oligonucleotides are formulated in a pharmaceutically acceptable diluent, such as phosphate-buffered saline. Administration includes parenteral routes of administration. Following a period of treatment with antisense oligonucleotides, RNA is isolated from tissue and changes in FGFR4 nucleic acid expression are measured. Changes in FGFR4 protein levels are also measured.

Certain Indications

In certain embodiments, provided herein are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has a metabolic disease.

As shown in the examples below, compounds targeted to FGFR4, as described herein, have been shown to reduce the severity of physiological symptoms of a metabolic disease, including obesity or adiposity, metabolic syndrome, diabetes mellitus, insulin resistance, diabetic dyslipidemia, and hypertriglyceridemia. In certain of the experiments, the compounds reduced body weight, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In certain of the experiments, the compounds reduced body fat, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In certain of the experiments, the compounds reduced adipose tissue, e.g., the animals continued to experience symptoms, but the symptoms were less severe compared to untreated animals. In other of the experiments, however, the compounds appear to reduce the symptoms of obesity; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the compounds appear to reduce the symptoms of diabetes; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the compounds appear to inhibit weight gain; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the compounds appear to reduce glucose levels; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. In other of the experiments, however, the compounds appear to increase fatty acid oxidation; e.g., animals treated for a longer period of time experienced less severe symptoms than those administered the compounds for a shorter period of time. The ability of the compounds exemplified below to restore function therefore demonstrates that symptoms of the disease may be reversed by treatment with a compound as described herein.

Obesity is characterized by numerous physical and physiological symptoms. Any symptom known to one of skill in the art to be associated with obesity can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the symptom is a physical symptom selected from the group consisting of increased adipose tissue mass or weight, increased weight gain, increased fat pad weight, imbalance with caloric intake and energy expenditure, increase in body fat, increase in body mass, having a body mass index (BMI) of 30 or higher, increase in body frame, increased sweating, sleep apnea, difficulty in sleeping, inability to cope with sudden physical activity, lethargy, back and joint problems, increase in breathlessness, increase in breast region adiposity, increase in abdomen size or fat, extreme hunger, or extreme fatigue.

In certain embodiments, the symptom is a physiological symptom selected from the group consisting of high blood pressure, hypertension, high cholesterol levels, type 2 diabetes, stroke, cardiac insufficiency, heart disease, coronary artery obstruction, breast cancer in women, gastro-oesophageal reflux disease, hip and knee arthrosis, and reduced life expectancy.

In certain embodiments, the physical symptom is excess body weight. In certain embodiments, the symptom is excess fat mass. In certain embodiments, the symptom is a body mass index of 30 or higher. In certain embodiments, the symptom is breathlessness. In certain embodiments, the symptom is increased sweating. In certain embodiments, the symptom is sleep apnea. In certain embodiments, the symptom is difficulty in sleeping. In certain embodiments, the symptom is inability to cope with sudden physical activity. In certain embodiments, the symptom is lethargy. In certain embodiments, the symptom is back and joint problems.

In certain embodiments, the physiological symptom is high blood pressure. In certain embodiments, the symptom is hypertension. In certain embodiments, the symptom is high cholesterol levels. In certain embodiments, the symptom is type 2 diabetes. In certain embodiments, the symptom is stroke. In certain embodiments, the symptom is cardiac insufficiency. In certain embodiments, the symptom is heart disease. In certain embodiments, the symptom is coronary artery obstruction. In certain embodiments, the symptom is breast cancer in women. In certain embodiments, the symptom is gastro-oesophageal reflux disease. In certain embodiments, the symptom is hip and knee arthrosis. In certain embodiments, the symptom is reduced life expectancy.

Diabetes mellitus is characterized by numerous physical and physiological symptoms. Any symptom known to one of skill in the art to be associated with Type 2 diabetes can be ameliorated or otherwise modulated as set forth above in the methods described above. In certain embodiments, the symptom is a physical symptom selected from the group consisting of increased glucose levels, increased weight gain, frequent urination, unusual thirst, extreme hunger, extreme fatigue, blurred vision, frequent infections, tingling or numbness at the extremities, dry and itchy skin, weight loss, slow-healing sores, and swollen gums.

In certain embodiments, the symptom is a physiological symptom selected from the group consisting of increased insulin resistance, increased glucose levels, increased fat mass, decreased metabolic rate, decreased glucose clearance, decreased glucose tolerance, decreased insulin sensitivity, decreased hepatic insulin sensitivity, increased adipose tissue size and weight, increased body fat, and increased body weight.

In certain embodiments, the physical symptom is increased weight gain. In certain embodiments, the symptom is frequent urination. In certain embodiments, the symptom is unusual thirst. In certain embodiments, the symptom is extreme hunger. In certain embodiments, the symptom is extreme fatigue. In certain embodiments, the symptom is blurred vision. In certain embodiments, the symptom is frequent infections. In certain embodiments, the symptom is tingling or numbness at the extremities. In certain embodiments, the symptom is dry and itchy skin. In certain embodiments, the symptom is weight loss. In certain embodiments, the symptom is slow-healing sores. In certain embodiments, the symptom is swollen gums. In certain embodiments, the symptom is increased insulin resistance. In certain embodiments, the symptom is increased fat mass. In certain embodiments, the symptom is decreased metabolic rate. In certain embodiments, the symptom is decreased glucose clearance. In certain embodiments, the symptom is decreased glucose tolerance. In certain embodiments, the symptom is decreased insulin sensitivity. In certain embodiments, the symptom is decreased hepatic insulin sensitivity. In certain embodiments, the symptom is increased adipose tissue size and weight. In certain embodiments, the symptom is increased body fat. In certain embodiments, the symptom is increased body weight.

In certain embodiments, provided are methods of treating an individual comprising administering one or more pharmaceutical compositions as described herein. In certain embodiments, the individual has metabolic related disease.

In certain embodiments, administration of an antisense compound targeted to a FGFR4 nucleic acid results in reduction of FGFR4 expression by at least about 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values.

In certain embodiments, pharmaceutical compositions comprising an antisense compound targeted to FGFR4 are used for the preparation of a medicament for treating a patient suffering or susceptible to a metabolic disease.

In certain embodiments, the methods described herein include administering a compound comprising a modified oligonucleotide having a contiguous nucleobases portion as described herein of a sequence recited in SEQ ID NO: 16.

In certain embodiments, the methods described herein include administering a compound comprising a modified oligonucleotide having a contiguous nucleobases portion as described herein of a sequence recited in SEQ ID NO: 45.

Administration

In certain embodiments, the compounds and compositions as described herein may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical, pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal, oral or parenteral. The compounds and compositions as described herein can be administered directly to a tissue or organ.

In certain embodiments, the compounds and compositions as described herein are administered parenterally. “Parenteral administration” means administration through injection or infusion. Parenteral administration includes subcutaneous administration, intravenous administration, intramuscular administration, intraarterial administration, intraperitoneal administration, or intracranial administration, e.g. intracerebral administration, intrathecal administration, intraventricular administration, ventricular administration, intracerebroventricular administration, cerebral intraventricular administration or cerebral ventricular administration. Administration can be continuous, or chronic, or short or intermittent.

In certain embodiments, parenteral administration is by injection. The injection can be delivered with a syringe or a pump. In certain embodiments, the injection is a bolus injection. In certain embodiments, the injection is administered directly to a tissue or organ.

In certain embodiments, the compounds and compositions as described herein are administered parenterally.

In certain embodiments, parenteral administration is subcutaneous.

In further embodiments, the formulation for administration is the compounds described herein and saline.

In certain embodiments, an antisense oligonucleotide is delivered by injection or infusion once every month, every two months, every 90 days, every 3 months, every 6 months, twice a year or once a year.

Certain Combination Therapies

In certain embodiments, one or more pharmaceutical compositions described herein are co-administered with one or more other pharmaceutical agents. In certain embodiments, such one or more other pharmaceutical agents are designed to treat the same disease, disorder, or condition as the one or more pharmaceutical compositions described herein. In certain embodiments, such one or more other pharmaceutical agents are designed to treat a different disease, disorder, or condition as the one or more pharmaceutical compositions described herein. In certain embodiments, such one or more other pharmaceutical agents are designed to treat an undesired side effect of one or more pharmaceutical compositions as described herein. In certain embodiments, one or more pharmaceutical compositions are co-administered with another pharmaceutical agent to treat an undesired effect of that other pharmaceutical agent. In certain embodiments, one or more pharmaceutical compositions are co-administered with another pharmaceutical agent to produce a combinational effect. In certain embodiments, one or more pharmaceutical compositions are co-administered with another pharmaceutical agent to produce a synergistic effect.

In certain embodiments, a first agent and one or more second agents are administered at the same time. In certain embodiments, the first agent and one or more second agents are administered at different times. In certain embodiments, the first agent and one or more second agents are prepared together in a single pharmaceutical formulation. In certain embodiments, the first agent and one or more second agents are prepared separately.

In certain embodiments, the second compound is administered prior to administration of a pharmaceutical composition described herein. In certain embodiments, the second compound is administered following administration of a pharmaceutical composition described herein. In certain embodiments, the second compound is administered at the same time as a pharmaceutical composition described herein. In certain embodiments, the dose of a co-administered second compound is the same as the dose that would be administered if the second compound was administered alone. In certain embodiments, the dose of a co-administered second compound is lower than the dose that would be administered if the second compound was administered alone. In certain embodiments, the dose of a co-administered second compound is greater than the dose that would be administered if the second compound was administered alone.

In certain embodiments, the co-administration of a second compound enhances the effect of a first compound, such that co-administration of the compounds results in an effect that is greater than the effect of administering the first compound alone. In certain embodiments, the co-administration results in effects that are additive of the effects of the compounds when administered alone. In certain embodiments, the co-administration results in effects that are supra-additive of the effects of the compounds when administered alone. In certain embodiments, the first compound is an antisense compound. In certain embodiments, the second compound is an antisense compound.

In certain embodiments the FGFR4 antisense oligonucleotide is delivered concomitant with delivery of the second agent. Alternatively, delivery can be in the same formulation or can be administered separately. In certain embodiments, FGFR4 antisense oligonucleotide is administered prior to the treatment with the second agents. In a certain embodiment, the FGFR4 antisense oligonucleotide is administered after treatment with an obesity inducing drug or agent is ceased.

In certain embodiments, second agents include, but are not limited to, a glucose-lowering agent. The glucose lowering agent can include, but is not limited to, a therapeutic lifestyle change, PPAR agonist, a dipeptidyl peptidase (IV) inhibitor, a GLP-1 analog, insulin or an insulin analog, an insulin secretagogue, a SGLT2 inhibitor, a human amylin analog, a biguanide, an alpha-glucosidase inhibitor, or a combination thereof. The glucose-lowering agent can include, but is not limited to metformin, sulfonylurea, rosiglitazone, meglitinide, thiazolidinedione, alpha-glucosidase inhibitor or a combination thereof. The sulfonylurea can be acetohexamide, chlorpropamide, tolbutamide, tolazamide, glimepiride, a glipizide, a glyburide, or a gliclazide. The meglitinide can be nateglinide or repaglinide. The thiazolidinedione can be pioglitazone or rosiglitazone. The alpha-glucosidase can be acarbose or miglitol.

In some embodiments, the glucose-lowering therapeutic is a GLP-1 analog. In some embodiments, the GLP-1 analog is exendin-4 or liraglutide.

In other embodiments, the glucose-lowering therapeutic is a sulfonylurea. In some embodiments, the sulfonylurea is acetohexamide, chlorpropamide, tolbutamide, tolazamide, glimepiride, a glipizide, a glyburide, or a gliclazide.

In some embodiments, the glucose-lowering drug is a biguanide. In some embodiments, the biguanide is metformin, and in some embodiments, blood glucose levels are decreased without increased lactic acidosis as compared to the lactic acidosis observed after treatment with metformin alone.

In some embodiments, the glucose-lowering drug is a meglitinide. In some embodiments, the meglitinide is nateglinide or repaglinide.

In some embodiments, the glucose-lowering drug is a thiazolidinedione. In some embodiments, the thiazolidinedione is pioglitazone, rosiglitazone, or troglitazone. In some embodiments, blood glucose levels are decreased without greater weight gain than observed with rosiglitazone treatment alone.

In some embodiments, the glucose-lowering drug is an alpha-glucosidase inhibitor. In some embodiments, the alpha-glucosidase inhibitor is acarbose or miglitol.

In a certain embodiment, a co-administered glucose-lowering agent is ISIS 113715.

In a certain embodiment, glucose-lowering therapy is therapeutic lifestyle change.

In certain embodiments, second agents include, but are not limited to, lipid-lowering agents. The lipid-lowering agent can include, but is not limited to atorvastatin, simvastatin, rosuvastatin, and ezetimibe. In certain such embodiments, the lipid-lowering agent is administered prior to administration of a pharmaceutical composition described herein. In certain such embodiments, the lipid-lowering agent is administered following administration of a pharmaceutical composition described herein. In certain such embodiments the lipid-lowering agent is administered at the same time as a pharmaceutical composition described herein. In certain such embodiments the dose of a co-administered lipid-lowering agent is the same as the dose that would be administered if the lipid-lowering agent was administered alone. In certain such embodiments the dose of a co-administered lipid-lowering agent is lower than the dose that would be administered if the lipid-lowering agent was administered alone. In certain such embodiments the dose of a co-administered lipid-lowering agent is greater than the dose that would be administered if the lipid-lowering agent was administered alone.

In certain embodiments, a co-administered lipid-lowering agent is a HMG-CoA reductase inhibitor. In certain such embodiments the HMG-CoA reductase inhibitor is a statin. In certain such embodiments the statin is selected from atorvastatin, simvastatin, pravastatin, fluvastatin, and rosuvastatin.

In certain embodiments, a co-administered lipid-lowering agent is a cholesterol absorption inhibitor. In certain such embodiments, cholesterol absorption inhibitor is ezetimibe.

In certain embodiments, a co-administered lipid-lowering agent is a co-formulated HMG-CoA reductase inhibitor and cholesterol absorption inhibitor. In certain such embodiments the co-formulated lipid-lowering agent is ezetimibe/simvastatin.

In certain embodiments, a co-administered lipid-lowering agent is a microsomal triglyceride transfer protein inhibitor (MTP inhibitor).

In certain embodiments, a co-administered lipid-lowering agent is an oligonucleotide targeted to ApoB.

In certain embodiments, second agents include, but are not limited to an anti-obesity drug or agent. Such anti-obesity agents include but are not limited to Orlistat, Sibutramine, or Rimonabant, and may be administered as described above as adipose or body weight lowering agents. In certain embodiments, the antisense compound may be co-administered with appetite suppressants. Such appetite suppressants include but are not limited to diethylpropion tenuate, mazindol, orlistat, phendimetrazine, phentermine, and sibutramine and may be administered as described herein. In certain embodiment, the anti-obesity agents are CNS based such as, but not limited to, sibutramine or GLP-1 based such as, but not limited to, liraglutide.

In certain embodiments, second agents include, but are not limited to an antipsychotic drug or agent. Such antipsychotic agents therapeutics may be administered as described above to reduce metabolic abnormalities associated with treatment with antipsychotic agents. In a particular embodiment administering of the FGFR4 antisense compound results in increased metabolic rate or decreasing adiposity or decreasing body weight or all three without affecting the CNS effects of the psychotherapeutic agent

Due to the ability of FGFR4 antisense oligonucleotides to increase metabolic rate and insulin sensitivity and reduce adiposity and weight gain, these compounds can be administered to reduce metabolic abnormalities associated with treatment with antipsychotic agents. In certain embodiments the FGFR4 antisense oligonucleotide is delivered in a method of reducing metabolic abnormalities associated with the therapeutic use of psychotherapeutic agents. Such weight inducing antipsychotic agents include, but are not limited to clozapine, olanzapine, aripiprazole, risperidone and ziprasidone.

Further provided is a method of administering an antisense compound targeted to a FGFR4 nucleic acid via injection and further including administering a topical steroid at the injection site.

Further examples of pharmaceutical agents that may be co-administered with a pharmaceutical composition of the present invention include, but are not limited to, corticosteroids, including but not limited to prednisone; immunoglobulins, including, but not limited to intravenous immunoglobulin (IVIg); analgesics (e.g., acetaminophen); anti-inflammatory agents, including, but not limited to non-steroidal anti-inflammatory drugs (e.g., ibuprofen, COX-1 inhibitors, and COX-2, inhibitors); salicylates; antibiotics; antivirals; antifungal agents; antidiabetic agents (e.g., biguanides, glucosidase inhibitors, insulins, sulfonylureas, and thiazolidenediones); adrenergic modifiers; diuretics; hormones (e.g., anabolic steroids, androgen, estrogen, calcitonin, progestin, somatostan, and thyroid hormones); immunomodulators; muscle relaxants; antihistamines; osteoporosis agents (e.g., biphosphonates, calcitonin, and estrogens); prostaglandins, antineoplastic agents; psychotherapeutic agents; sedatives; poison oak or poison sumac products; antibodies; and vaccines.

In certain embodiments, the pharmaceutical compositions of the present invention may be administered in conjunction with a lipid-lowering therapy. In certain such embodiments, a lipid-lowering therapy is therapeutic lifestyle change. In certain such embodiments, a lipid-lowering therapy is LDL apheresis.

Formulations

The compounds provided herein may also be admixed, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds, as for example, liposomes, receptor-targeted molecules, or other formulations, for assisting in uptake, distribution and/or absorption. Representative United States patents that teach the preparation of such uptake, distribution and/or absorption-assisting formulations include, but are not limited to, U.S. Pat. Nos. 5,108,921; 5,354,844; 5,416,016; 5,459,127; 5,521,291; 5,543,158; 5,547,932; 5,583,020; 5,591,721; 4,426,330; 4,534,899; 5,013,556; 5,108,921; 5,213,804; 5,227,170; 5,264,221; 5,356,633; 5,395,619; 5,416,016; 5,417,978; 5,462,854; 5,469,854; 5,512,295; 5,527,528; 5,534,259; 5,543,152; 5,556,948; 5,580,575; and 5,595,756, each of which is herein incorporated by reference.

The antisense compounds provided herein encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof.

The term “pharmaceutically acceptable salts” refers to physiologically and pharmaceutically acceptable salts of the compounds provided herein: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. The term “pharmaceutically acceptable salt” includes a salt prepared from pharmaceutically acceptable non-toxic acids or bases, including inorganic or organic acids and bases. For oligonucleotides, preferred examples of pharmaceutically acceptable salts and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety. Sodium salts have been shown to be suitable forms of oligonucleotide drugs.

The term “pharmaceutically acceptable derivative” encompasses, but is not limited to, pharmaceutically acceptable salts, solvates, hydrates, esters, prodrugs, polymorphs, isomers, isotopically labeled variants of the compounds described herein.

The present invention also includes pharmaceutical compositions and formulations which include the antisense compounds provided herein. The pharmaceutical compositions described herein may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intracerebral administration, intrathecal administration, intraventricular administration, ventricular administration, intracerebroventricular administration, cerebral intraventricular administration or cerebral ventricular administration.

Parenteral administration, is preferred to target FGFR4 expression in the liver and plasma. Oligonucleotides with at least one 2′-O-methoxyethyl modification are believed to be particularly useful for oral administration. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful.

The pharmaceutical formulations described herein, which may conveniently be presented in unit dosage form, may be prepared according to conventional techniques well known in the pharmaceutical industry. Such techniques include the step of bringing into association the active ingredients with the pharmaceutical carrier(s) or excipient(s). In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.

The compositions described herein may be formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules, gel capsules, liquid syrups, soft gels, suppositories, and enemas. The compositions described herein may also be formulated as suspensions in aqueous, non-aqueous or mixed media. Aqueous suspensions may further contain substances which increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.

Pharmaceutical compositions described herein include, but are not limited to, solutions, emulsions, foams and liposome-containing formulations. The pharmaceutical compositions and formulations described herein may comprise one or more penetration enhancers, carriers, excipients or other active or inactive ingredients.

Emulsions are typically heterogenous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 μm in diameter. Emulsions may contain additional components in addition to the dispersed phases, and the active drug which may be present as a solution in the aqueous phase, oily phase or itself as a separate phase. Microemulsions are included as an embodiment described herein. Emulsions and their uses are well known in the art and are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

Formulations include liposomal formulations. As used in the present invention, the term “liposome” means a vesicle composed of amphiphilic lipids arranged in a spherical bilayer or bilayers. Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior that contains the composition to be delivered. Cationic liposomes are positively charged liposomes which are believed to interact with negatively charged DNA molecules to form a stable complex. Liposomes that are pH-sensitive or negatively-charged are believed to entrap DNA rather than complex with it. Both cationic and noncationic liposomes have been used to deliver DNA to cells.

Liposomes also include “sterically stabilized” liposomes, a term which, as used herein, refers to liposomes comprising one or more specialized lipids that, when incorporated into liposomes, result in enhanced circulation lifetimes relative to liposomes lacking such specialized lipids. Liposomes and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

In another embodiment, formulations include saline formulations. In certain embodiments, a formulation consists of the compounds described herein and saline. In certain embodiments, a formulation consists essentially of the compounds described herein and saline. In certain embodiments, the saline is pharmaceutically acceptable grade saline. In certain embodiments, the saline is buffered saline. In certain embodiments, the saline is phosphate buffered saline (PBS).

In certain embodiments, a formulation excludes liposomes. In certain embodiments, the formulation excludes sterically stabilized liposomes. In certain embodiments, a formulation excludes phospholipids. In certain embodiments, the formulation consists essentially of the compounds described herein and saline and excludes liposomes.

The pharmaceutical formulations and compositions may also include surfactants. Surfactants and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

In one embodiment, the present invention employs various penetration enhancers to affect the efficient delivery of nucleic acids, particularly oligonucleotides. Penetration enhancers and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

One of skill in the art will recognize that formulations are routinely designed according to their intended use, i.e. route of administration.

Formulations for topical administration include those in which the oligonucleotides provided herein are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Preferred lipids and liposomes include neutral (e.g. dioleoylphosphatidyl DOPE ethanolamine, dimyristoylphosphatidyl choline DMPC, distearolyphosphatidyl choline) negative (e.g. dimyristoylphosphatidyl glycerol DMPG) and cationic (e.g. dioleoyltetramethylaminopropyl DOTAP and dioleoylphosphatidyl ethanolamine DOTMA).

Compositions and formulations for parenteral administration, including intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular injection or infusion, or intracranial may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives such as, but not limited to, penetration enhancers, carrier compounds and other pharmaceutically acceptable carriers or excipients.

Certain embodiments provided herein provide pharmaceutical compositions containing one or more oligomeric compounds and one or more other chemotherapeutic agents which function by a non-antisense mechanism. Examples of such chemotherapeutic agents include but are not limited to cancer chemotherapeutic drugs such as daunorubicin, daunomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, esorubicin, bleomycin, mafosfamide, ifosfamide, cytosine arabinoside, bis-chloroethylnitrosurea, busulfan, mitomycin C, actinomycin D, mithramycin, prednisone, hydroxyprogesterone, testosterone, tamoxifen, dacarbazine, procarbazine, hexamethylmelamine, pentamethylmelamine, mitoxantrone, amsacrine, chlorambucil, methylcyclohexylnitrosurea, nitrogen mustards, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-azacytidine, hydroxyurea, deoxycoformycin, 4-hydroxyperoxycyclophosphoramide, 5-fluorouracil (5-FU), 5-fluorodeoxyuridine (5-FUdR), methotrexate (MTX), colchicine, taxol, vincristine, vinblastine, etoposide (VP-16), trimetrexate, irinotecan, topotecan, gemcitabine, teniposide, cisplatin and diethylstilbestrol (DES). When used with the compounds provided herein, such chemotherapeutic agents may be used individually (e.g., 5-FU and oligonucleotide), sequentially (e.g., 5-FU and oligonucleotide for a period of time followed by MTX and oligonucleotide), or in combination with one or more other such chemotherapeutic agents (e.g., 5-FU, MTX and oligonucleotide, or 5-FU, radiotherapy and oligonucleotide). Anti-inflammatory drugs, including but not limited to nonsteroidal anti-inflammatory drugs and corticosteroids, and antiviral drugs, including but not limited to ribivirin, vidarabine, acyclovir and ganciclovir, may also be combined in compositions provided herein. Combinations of antisense compounds and other non-antisense drugs are also within the scope of this invention. Two or more combined compounds may be used together or sequentially.

In another related embodiment, compositions provided herein may contain one or more antisense compounds, particularly oligonucleotides, targeted to a first nucleic acid and one or more additional antisense compounds targeted to a second nucleic acid target. Alternatively, compositions provided herein may contain two or more antisense compounds targeted to different regions of the same nucleic acid target. Numerous examples of antisense compounds are known in the art. Two or more combined compounds may be used together or sequentially.

Dosing

Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Optimum dosages may vary depending on the relative potency of individual oligonucleotides, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 μg to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or at desired intervals. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligonucleotide is administered in maintenance doses, ranging from 0.01 μg to 100 g per kg of body weight, once or more daily.

While the present invention has been described with specificity in accordance with certain of its preferred embodiments, the following examples serve only to illustrate the invention and are not intended to limit the same. Each of the references, GENBANK accession numbers, and the like recited in the present application is incorporated herein by reference in its entirety.

Certain Compounds

About one thousand four hundred and fifty four newly designed and previously disclosed antisense compounds of various lengths, motifs and backbone composition were tested for their effect on human FGFR4 mRNA in vitro in several cell types. The new compounds were compared with nine previously designed compounds, including ISIS 299005, ISIS 299010, ISIS 299018, ISIS 299022, ISIS 299024, ISIS 299025, ISIS 299028, ISIS 299029, and ISIS 299030 which have previously been determined to be some of the most potent antisense compounds in vitro (see e.g., U.S. Patent Publication No. US2010/0292140). Of the one thousand four hundred and fifty four newly designed and the nine previously designed antisense compounds, fifty three compounds were selected for further study based on in vitro potency. The selected compounds were tested for dose dependent inhibition in HepG2 (Examples 8 and 9).

Certain oligonucleotides were then tested for tolerability in a CD1 mouse model, as well as a Sprague-Dawley rat model. The oligonucleotides tested for tolerability include oligonucleotides, ISIS 299005 (SEQ ID NO: 7), ISIS 463588 (SEQ ID NO: 16), ISIS 463589 (SEQ ID NO: 17), ISIS 463628 (SEQ ID NO: 28), ISIS 463690 (SEQ ID NO: 45), ISIS 463691 (SEQ ID NO: 46), ISIS 463835 (SEQ ID NO: 70), ISIS 463837 (SEQ ID NO: 72), ISIS 464222 (SEQ ID NO: 135), ISIS 464225 (SEQ ID NO: 138), ISIS 464228 (SEQ ID NO: 141), ISIS 464286 (SEQ ID NO: 154), ISIS 464308 (SEQ ID NO: 163), ISIS 464449 (SEQ ID NO: 174), ISIS 464587 (SEQ ID NO: 186), ISIS 464588 (SEQ ID NO: 187), ISIS 464589 (SEQ ID NO: 188), ISIS 464718 (SEQ ID NO: 221), ISIS 479533 (SEQ ID NO: 241), ISIS 479551 (SEQ ID NO: 259), ISIS 479691 (SEQ ID NO: 299), ISIS 479692 (SEQ ID NO: 300), ISIS 479698 (SEQ ID NO: 305), ISIS 479699 (SEQ ID NO: 306), ISIS 479703 (SEQ ID NO: 307), ISIS 479704 (SEQ ID NO: 308), ISIS 479706 (SEQ ID NO: 310), and ISIS 479736 (SEQ ID NO: 317). By virtue of their complementary sequence, the compounds are complementary to the regions 192-211, 191-210, 193-212, 291-310, 369-388, 370-389, 788-807, 790-809, 2951-2970, 2954-2973, and 2981-3000 of SEQ ID NO: 1; 11621-11640, 11624-11643, 11651-11670, 1463-1482, 3325-3344, 7802-7821, 2110-2129, 2112-2131, 2114-2133, 3575-3594, 2111-2130, 3570-3589, 11623-11639, 11624-11640, 11652-11668, 11653-11669, 2113-2129, 2114-2130, 2116-2132, and 3571-3587 of SEQ ID NO: 2; and 103-122, 1569-1588, 5122-5138, 5123-5139, 5151-5167, 5152-5168, 105-121, 106-122, 108-124, and 1570-1586 of SEQ ID NO: 3.

In the in vivo models, the liver function markers, such as alanine transaminase, aspartate transaminase and bilirubin, and kidney function markers, such as BUN and creatinine were measured. (Example 11).

Eight oligonucleotides having a nucleobase sequence of a sequence recited in SEQ ID NO: 7 (ISIS 299005), 16 (ISIS 463588), 17 (ISIS 463589), 45 (ISIS 463690), 46 (ISIS 463691), 70 (ISIS 463835), 72 (ISIS 463837) and 138 (ISIS 464225) were tested. By virtue of their complementary sequence, the compounds are complementary to the regions 192-211, 191-210, 193-212, 369-388, 370-389, 788-807, 790-809, and 2954-2973 of SEQ ID NO: 1. In certain embodiments, the compounds targeting the listed regions, as further described herein, comprise a modified oligonucleotide having some nucleobase portion of the sequence recited in the SEQ ID NOs, as further described herein, In certain embodiments, the compounds targeting the listed regions or having a nucleobase portion of a sequence recited in the listed SEQ ID NOs can be of various length, as further described herein, and can have one of various motifs, as further described herein. In certain embodiments, a compound targeting a region or having a nucleobase portion of a sequence recited in the listed SEQ ID NOs has the specific length and motif, as indicated by the ISIS NOs: 299005, 463588, 463589, 463690, 463691, 463835, 463837, and 464225.

These eight compounds, ISIS 299005 (SEQ ID NO: 7), ISIS 463588 (SEQ ID NO: 16), ISIS 463589 (SEQ ID NO: 17), ISIS 463690 (SEQ ID NO: 45), ISIS 463691 (SEQ ID NO: 46), ISIS 463835 (SEQ ID NO: 70), ISIS 463837 (SEQ ID NO: 72), and ISIS 464225 (SEQ ID NO: 138), were assayed for long-term effects on tolerability in a CD/1GS rat model for 13 weeks (Example 12). Body weights and organ weights, the liver function markers, such as alanine transaminase, aspartate transaminase and bilirubin, and kidney function markers, such as BUN and creatinine were measured. The eight compounds were also tested for their viscosity. (Example 14)

ISIS 463588, ISIS 463589, and ISIS 463690 which demonstrated very good tolerability in all three in vivo models, were tested for their half-life in CD1 mouse liver (Example 13).

These eight compounds, ISIS 299005 (SEQ ID NO: 7), ISIS 463588 (SEQ ID NO: 16), ISIS 463589 (SEQ ID NO: 17), ISIS 463690 (SEQ ID NO: 45), ISIS 463691 (SEQ ID NO: 46), ISIS 463835 (SEQ ID NO: 70), ISIS 463837 (SEQ ID NO: 72), and ISIS 464225 (SEQ ID NO: 138), were tested for efficacy, pharmacokinetic profile and tolerability in cynomolgus monkeys (Example 15). The inhibition studies in these monkeys indicated that treatment with some of these compounds caused reduction of FGFR4 mRNA in the liver tissues. Specifically, treatment with ISIS 463588 caused significantly greater reduction of FGFR4 mRNA in liver and kidney tissues, respectively compared to treatment with the previously disclosed compound, ISIS 299005. It was noted that ISIS 463588 caused the highest reduction of FGFR4 mRNA compared to the PBS control, irrespective of the primer probe set used. Hence, in terms of potency, treatment with ISIS 463588 was the most effective in the monkey study. Treatment with ISIS 463690 also caused a greater reduction of FGFR4 mRNA in liver and kidney tissues, respectively compared to treatment with the previously disclosed compound, ISIS 299005.

FGF19 has been known to reduce adiposity and improve insulin sensitivity in transgenic mice (Fu, L. et al., Endocrinology. 145: 2594-2603, 2004). FGF19 is also characterized as a high affinity ligand for FGFR4 (Xie, M.-H. et al., Cytokine. 11: 729-735, 1999). However, treating mice with FGF19 protein induces hepatocyte proliferation consistent with the increased hepatocyte proliferation and liver tumor formation observed in FGF19 transgenic mice (Wu, X. et al., JBC 285(8): 5165-5170, 2010). Leptin is a hormone which has been found to be present at very high levels in obese individuals compared to normal-weight individuals (Considine, R. V. et al., N. Engl. J. Med. 334: 292-295, 1996). Evaluation of FGF19 mRNA and plasma levels demonstrated the significant increase in FGF19 mRNA and protein levels in all the treatment groups. Specifically, monkeys treated with ISIS 463588 had the most significant increase in FGF19 levels. Evaluation of leptin plasma levels demonstrated a significant decrease in monkeys treated with ISIS 463588 or ISIS 463690. Tolerability studies in cynomolgus monkeys (Example 15) were conducted after treatment with the ISIS oligonucleotides. This included measurement plasma levels of liver metabolites, kidney metabolites, pro-inflammatory factors, such as C-reactive protein, complement C3 and cytokines. The results indicated that treatment with the ISIS oligonucleotides in Example 15 remained within acceptable levels for antisense oligonucleotides and were therefore tolerable to the monkeys. In particular, treatment with ISIS 463588 was very well-tolerated in this model.

Pharmacokinetic studies of the three most well-tolerated ISIS oligonucleotides, ISIS 463588, ISIS 463589, and ISIS 463690, was also performed in the monkeys and indicated that the pharmacokinetics of all three were optimal.

Hence, the in vivo studies, particularly in the cynomolgus monkeys, indicate that ISIS 463588, ISIS 463589, and ISIS 463690, were a more potent oligonucleotide compared to ISIS 299005 and was also considerably more tolerable.

Accordingly, provided herein are antisense compounds with any one or more of the improved characteristics. In a certain embodiments, provided herein are compounds comprising a modified oligonucleotide as further described herein targeted to or specifically hybridizable with the region of nucleotides of SEQ ID NO: 1.

In certain embodiments, the compounds as described herein are efficacious by virtue of having at least one of an in vitro IC50 of less than 1.5 μM, less than 1.4 μM, less than 1.3 μM, less than 1.2 μM, less than 1.1 μM, less than 1.0 μM, less than 0.9 μM, less than 0.8 μM, less than 0.7 μM, less than 0.6 μM when delivered to a HepG2 cell line using electroporation as described in Example 8. In certain embodiments, the compounds as described herein are efficacious in vivo, as demonstrated by decreasing the levels of FGFR4 mRNA by 60%, 65%, 70%, 75% or 80%. In further embodiments, the compounds are efficacious in vivo, as demonstrated by increasing the levels of FGF15 and FGF19 mRNA and protein by 100%, 200%, 300%, 400%, 500%, 600%, 700%, 800%, 900%, or 1000%. In other embodiments, the compounds are efficacious in vivo, as demonstrated by decreasing plasma levels of leptin by 30%, 35%, or 40%.

In certain embodiments, the compounds as described herein are highly tolerable, as demonstrated by having at least one of an increase an ALT or AST value of no more than 4 fold, 3 fold, or 2 fold over saline treated animals; or an increase in liver, spleen or kidney weight of no more than 30%, 20%, 15%, 12%, 10%, 5% or 2%.

EXAMPLES Non-Limiting Disclosure and Incorporation by Reference

While certain compounds, compositions and methods described herein have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds described herein and are not intended to limit the same. Each of the references recited in the present application is incorporated herein by reference in its entirety.

Example 1 Antisense Inhibition of Human Fibroblast Growth Factor Receptor (FGFR4) in HepG2 Cells

Antisense oligonucleotides were designed targeting a FGFR4 nucleic acid and were tested for their effects on FGFR4 mRNA in vitro. Cultured HepG2 cells at a density of 20,000 cells per well were transfected using electroporation with 4,500 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human primer probe set RTS3232 (forward sequence TCATCAACGGCAGCAGCTT, designated herein as SEQ ID NO: 327; reverse sequence AGCTATTGATGTCTGCAGTCTTTAGG, designated herein as SEQ ID NO: 328; probe sequence AGCCGACGGTTTCCCCTATGTGCA, designated herein as SEQ ID NO: 329) was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells. A total of 458 oligonucleotides were tested. Only those oligonucleotides demonstrating greater than 65% inhibition in vitro or which were used in subsequent assays are shown in Table 1.

Some of the antisense oligonucleotides were also tested with an additional primer probe set RTS1325 (forward sequence TTGCTGTGCCGTGTCCAA, designated herein as SEQ ID NO: 330; reverse sequence TCCAAGAAGCCGAGCAGAAC, designated herein as SEQ ID NO: 331; probe sequence AGCTGCCGTGCCTGTGTCCTGAT, designated herein as SEQ ID NO: 332). ‘n/a.’ indicates that particular antisense oligonucleotide was not tested with RTS1325.

The newly designed chimeric antisense oligonucleotides in Table 1 were designed as 5-10-5 MOE gapmers. The gapmers are 20 nucleosides in length, wherein the central gap segment comprises of ten 2′-deoxynucleosides and is flanked by wing segments on the 5′ and 3′ directions comprising five nucleosides each. Each nucleotide in the 5′ wing segment and each nucleotide in the 3′ wing segment has a 2′-MOE modification. The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine residues throughout each gapmer are 5-methylcytosines. “Start site” indicates the 5′-most nucleoside to which the gapmer is targeted in the human gene sequence. “Stop site” indicates the 3′-most nucleoside to which the gapmer is targeted in the human gene sequence. Each gapmer listed in Table 1 is targeted to the human FGFR4 mRNA, designated herein as SEQ ID NO: 1 (GENBANK Accession No. NM002011.3).

TABLE 1 Inhibition of human FGFR4 mRNA levels by chimeric antisense oligonucleotides targeted to SEQ ID NO: 1 % % SEQ Start Stop ISIS inhibition inhibition ID Site Site Sequence No (RTS3232) (RTS1325) NO 192 211 GGCACACTCAGCAGGACCCC 299005 85 n/a 7 304 323 AGGCTGCCCAAGGGCTACTG 299010 65 n/a 8 597 616 GTCCAGTAGGGTGCTTGCTG 299018 68 n/a 9 727 746 CCCATGAAAGGCCTGTCCAT 299022 68 n/a 10 757 776 GCGCAGCCGAATGCCTCCAA 299024 68 65 11 785 804 TCTCCATCACGAGACTCCAG 299025 65 59 12 969 988 TACACCTTGCACAGCAGCTC 299028 68 66 13 1027 1046 GCTGCTGCCGTTGATGACGA 299029 91 61 14 1032 1051 CCGAAGCTGCTGCCGTTGAT 299030 72 19 15 191 210 GCACACTCAGCAGGACCCCC 463588 87 n/a 16 193 212 AGGCACACTCAGCAGGACCC 463589 83 n/a 17 194 213 CAGGCACACTCAGCAGGACC 463590 72 n/a 18 196 215 CCCAGGCACACTCAGCAGGA 463592 73 n/a 19 197 216 GCCCAGGCACACTCAGCAGG 463593 71 n/a 20 198 217 GGCCCAGGCACACTCAGCAG 463594 69 n/a 21 200 219 GAGGCCCAGGCACACTCAGC 463596 78 n/a 22 202 221 TGGAGGCCCAGGCACACTCA 463598 72 n/a 23 203 222 CTGGAGGCCCAGGCACACTC 463599 78 n/a 24 205 224 GACTGGAGGCCCAGGCACAC 463601 69 n/a 25 287 306 CTGTCAGCTCCTGCTCTTGC 463625 65 n/a 26 290 309 CTACTGTCAGCTCCTGCTCT 463627 74 n/a 27 291 310 GCTACTGTCAGCTCCTGCTC 463628 82 n/a 28 292 311 GGCTACTGTCAGCTCCTGCT 463629 93 n/a 29 293 312 GGGCTACTGTCAGCTCCTGC 463630 75 n/a 30 299 318 GCCCAAGGGCTACTGTCAGC 463636 69 n/a 31 309 328 CGCACAGGCTGCCCAAGGGC 463645 75 n/a 32 332 351 GCTCAGCCCGCCCACAGCAC 463648 81 n/a 33 338 357 CACCACGCTCAGCCCGCCCA 463654 77 n/a 34 339 358 CCACCACGCTCAGCCCGCCC 463655 73 n/a 35 340 359 GCCACCACGCTCAGCCCGCC 463656 69 n/a 36 341 360 GGCCACCACGCTCAGCCCGC 463657 65 n/a 37 347 366 ACCAGTGGCCACCACGCTCA 463670 73 n/a 38 349 368 GTACCAGTGGCCACCACGCT 463672 81 n/a 39 350 369 TGTACCAGTGGCCACCACGC 463673 69 n/a 40 355 374 CTCCTTGTACCAGTGGCCAC 463677 67 n/a 41 356 375 CCTCCTTGTACCAGTGGCCA 463678 66 n/a 42 357 376 CCCTCCTTGTACCAGTGGCC 463679 76 n/a 43 368 387 CCAGGCGACTGCCCTCCTTG 463689 76 n/a 44 369 388 GCCAGGCGACTGCCCTCCTT 463690 85 n/a 45 370 389 TGCCAGGCGACTGCCCTCCT 463691 78 n/a 46 371 390 GTGCCAGGCGACTGCCCTCC 463692 81 n/a 47 372 391 GGTGCCAGGCGACTGCCCTC 463693 70 n/a 48 388 407 CCGTACACGGCCAGCAGGTG 463708 80 n/a 49 389 408 CCCGTACACGGCCAGCAGGT 463709 85 n/a 50 392 411 AGCCCCGTACACGGCCAGCA 463712 73 n/a 51 397 416 CCTCCAGCCCCGTACACGGC 463717 66 n/a 52 398 417 CCCTCCAGCCCCGTACACGG 463718 66 n/a 53 404 423 GGCGGCCCCTCCAGCCCCGT 463724 70 n/a 54 414 433 GCAATCTCTAGGCGGCCCCT 463733 65 n/a 55 415 434 GGCAATCTCTAGGCGGCCCC 463734 69 n/a 56 416 435 TGGCAATCTCTAGGCGGCCC 463735 67 n/a 57 431 450 CCTCAGGTAGGAAGCTGGCA 463750 56 n/a 58 432 451 TCCTCAGGTAGGAAGCTGGC 463751 76 n/a 59 443 462 AGCGGCCAGCATCCTCAGGT 463762 58 n/a 60 444 463 TAGCGGCCAGCATCCTCAGG 463763 77 n/a 61 599 618 GTGTCCAGTAGGGTGCTTGC 463770 66 n/a 62 601 620 GTGTGTCCAGTAGGGTGCTT 463771 32 n/a 63 624 643 AGTTTCTTCTCCATGCGCTG 463774 72 n/a 64 717 736 GCCTGTCCATCCTTAAGCCA 463791 68 n/a 65 732 751 TTCTCCCCATGAAAGGCCTG 463805 65 n/a 66 734 753 GGTTCTCCCCATGAAAGGCC 463807 60 n/a 67 784 803 CTCCATCACGAGACTCCAGT 463832 65 76 68 787 806 GCTCTCCATCACGAGACTCC 463834 78 59 69 788 807 CGCTCTCCATCACGAGACTC 463835 78 67 70 789 808 ACGCTCTCCATCACGAGACT 463836 69 66 71 790 809 CACGCTCTCCATCACGAGAC 463837 80 75 72 791 810 CCACGCTCTCCATCACGAGA 463838 76 67 73 968 987 ACACCTTGCACAGCAGCTCC 463860 66 67 74 970 989 GTACACCTTGCACAGCAGCT 463861 76 74 75 1021 1040 GCCGTTGATGACGATGTGCT 463871 65 46 76 1024 1043 GCTGCCGTTGATGACGATGT 463874 77 52 77 1025 1044 TGCTGCCGTTGATGACGATG 463875 78 42 78 1026 1045 CTGCTGCCGTTGATGACGAT 463876 78 10 79 1028 1047 AGCTGCTGCCGTTGATGACG 463877 90 54 80 1029 1048 AAGCTGCTGCCGTTGATGAC 463878 73 22 81 1031 1050 CGAAGCTGCTGCCGTTGATG 463880 74 3 82 1084 1103 GCTATTGATGTCTGCAGTCT 463882 76 67 83 1085 1104 AGCTATTGATGTCTGCAGTC 463883 68 56 84 1086 1105 GAGCTATTGATGTCTGCAGT 463884 75 61 85 1097 1116 CCTCCACCTCTGAGCTATTG 463893 74 73 86 1098 1117 ACCTCCACCTCTGAGCTATT 463894 71 71 87 1099 1118 GACCTCCACCTCTGAGCTAT 463906 66 55 88 1100 1119 GGACCTCCACCTCTGAGCTA 463907 77 90 89 1101 1120 AGGACCTCCACCTCTGAGCT 463908 89 47 90 1102 1121 CAGGACCTCCACCTCTGAGC 463909 89 74 91 1103 1122 ACAGGACCTCCACCTCTGAG 463910 79 55 92 1105 1124 GTACAGGACCTCCACCTCTG 463912 69 75 93 1106 1125 GGTACAGGACCTCCACCTCT 463913 71 73 94 1111 1130 CCGCAGGTACAGGACCTCCA 463918 67 72 95 1112 1131 TCCGCAGGTACAGGACCTCC 463919 65 37 96 1115 1134 CGTTCCGCAGGTACAGGACC 463922 70 72 97 1185 1204 GCAGACTGGTAGGAGAGGCC 463937 74 82 98 1186 1205 GGCAGACTGGTAGGAGAGGC 463938 66 7 99 1214 1233 GGTCCTCCTCTGGCAGCACC 463947 68 55 100 1301 1320 GCAGGAGCACAGCCAAGGCC 463967 67 77 101 1329 1348 GCCTGCCCTCGATACAGCCC 463994 68 n/a 102 1417 1436 GCCTGACTCCAGGGAGAACT 464002 73 n/a 103 1419 1438 GAGCCTGACTCCAGGGAGAA 464004 68 n/a 104 1468 1487 GGAGGAGAGACGCACGCCTC 464013 77 n/a 105 1469 1488 TGGAGGAGAGACGCACGCCT 464014 82 n/a 106 1470 1489 CTGGAGGAGAGACGCACGCC 464015 68 n/a 107 1502 1521 GACTCACGAGGCCGGCGAGC 464030 68 n/a 108 1505 1524 CTAGACTCACGAGGCCGGCG 464033 68 n/a 109 1558 1577 CCCAAGCACCAGCCTGTCCC 464037 67 n/a 110 1559 1578 TCCCAAGCACCAGCCTGTCC 464038 79 n/a 111 1562 1581 GCTTCCCAAGCACCAGCCTG 464041 75 n/a 112 1564 1583 GGGCTTCCCAAGCACCAGCC 464043 74 n/a 113 1616 1635 CCATGCCAAAGGCCTCTGCA 464046 65 n/a 114 1618 1637 GTCCATGCCAAAGGCCTCTG 464048 68 n/a 115 1619 1638 GGTCCATGCCAAAGGCCTCT 464049 73 n/a 116

Example 2 Dose-Dependent Antisense Inhibition of Human FGFR4 in HepG2 Cells

Gapmers from Example 1 exhibiting significant in vitro inhibition of human FGFR4 were tested at various doses in HepG2 cells. Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.6 nM, 1.3 nM, 2.5 nM, 5.0 nM, and 10.0 μM concentrations of antisense oligonucleotide, as specified in Table 2. After a treatment period of approximately 16 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human FGFR4 primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells.

The half maximal inhibitory concentration (IC50) of each oligonucleotide is also presented in Table 2 and was calculated by plotting the concentrations of oligonucleotides used versus the percent inhibition of FGFR4 mRNA expression achieved at each concentration, and noting the concentration of oligonucleotide at which 50% inhibition of FGFR4 mRNA expression was achieved compared to the control. As illustrated in Table 2, FGFR4 mRNA levels were significantly reduced in a dose-dependent manner in antisense oligonucleotide treated cells.

TABLE 2 Dose-dependent antisense inhibition of human FGFR4 in HepG2 cells using electroporation 0.6 1.3 2.5 5.0 10.0 IC50 ISIS No μM μM μM μM μM (μM) 299005 45 63 77 92 95 0.7 463588 43 66 86 90 97 0.6 463589 41 67 85 92 95 0.6 463628 55 68 87 94 92 0.3 463629 25 40 63 76 91 1.8 463648 36 51 71 85 96 1.1 463672 19 46 74 90 96 1.5 463690 30 66 86 94 97 0.9 463691 31 50 78 89 96 1.1 463692 33 56 75 90 94 1.1 463708 11 45 63 77 94 1.9 463709 35 50 73 86 96 1.1 463750 24 42 54 80 93 1.8 463762 57 76 90 95 98 <0.6 463771 53 44 66 83 88 1.4 463807 13 36 56 87 96 2.0 463834 32 44 68 90 97 1.3 463835 37 59 82 91 97 0.9 463837 28 61 77 92 97 1.1 463838 29 50 72 88 95 1.3 463861 44 52 78 90 97 0.8 463893 29 33 65 84 95 1.6 464013 27 34 50 75 86 2.1 464014 16 33 55 78 90 2.1 464038 37 55 74 90 96 1.0

Example 3 Antisense Inhibition of Human Fibroblast Growth Factor Receptor (FGFR4) in HepG2 Cells

Additional antisense oligonucleotides were designed targeting a FGFR4 nucleic acid and were tested for their effects on FGFR4 mRNA in vitro. Some of the antisense oligonucleotides described in Example 1 were also included in the assay for comparison. Cultured HepG2 cells at a density of 20,000 cells per well were transfected using electroporation with 4,500 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells. A total of 772 oligonucleotides were tested. Only those oligonucleotides demonstrating greater than 65% inhibition are shown in Tables 3 and 4.

The newly designed chimeric antisense oligonucleotides in Table 3 were designed as 5-10-5 MOE gapmers. The gapmers are 20 nucleosides in length, wherein the central gap segment comprises of ten 2′-deoxynucleosides and is flanked by wing segments on the 5′ and 3′ directions comprising five nucleosides each. Each nucleotide in the 5′ wing segment and each nucleotide in the 3′ wing segment has a 2′-MOE modification. The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine residues throughout each gapmer are 5-methylcytosines. “Start site” indicates the 5′-most nucleoside to which the gapmer is targeted in the human gene sequence. “Stop site” indicates the 3′-most nucleoside to which the gapmer is targeted in the human gene sequence.

Each gapmer listed in Table 3 is targeted to either the human FGFR4 mRNA, designated herein as SEQ ID NO: 1 (GENBANK Accession No. NM002011.3) or the human FGFR4 genomic sequence, designated herein as SEQ ID NO: 2 (GENBANK Accession No: NT023133.11 truncated from nucleosides 21323018 to 21335213), or both. Some of the antisense oligonucleotides were designed to target variant gene sequences and are listed in Table 4. Each gapmer in Table 4 is listed to either SEQ ID NO: 3 (GENBANK Accession No. AB209631.1)

TABLE 3 Inhibition of human FGFR4 mRNA levels by chimeric antisense oligonucleotides targeted to SEQ ID NO: 1 and SEQ ID NO: 2 Start Stop Start Stop Site on Site on Site on Site on SEQ SEQ SEQ SEQ SEQ ID ID ISIS % ID ID ID NO: 1 NO: 1 No Sequence inhibition NO: 2 NO: 2 NO 160 179 299004 CAGCAGCCGCATCTCCTTCT 88 3165 3184 117 2497 2516 299055 GCTGAAGACAGAATCGCTGG 65 11167 11186 118 292 311 463629 GGCTACTGTCAGCTCCTGCT 87 3993 4012 29 2325 2344 464138 CAGCACTCACGCATCAGCCC 72 10866 10885 119 2326 2345 464139 CCAGCACTCACGCATCAGCC 75 10867 10886 120 2437 2456 464167 GGGTCCGAAGGTCAGGCGGA 68 11107 11126 121 2438 2457 464168 AGGGTCCGAAGGTCAGGCGG 70 11108 11127 122 2440 2459 464170 ATAGGGTCCGAAGGTCAGGC 67 11110 11129 123 2443 2462 464173 GGAATAGGGTCCGAAGGTCA 69 11113 11132 124 2582 2601 464181 GTGCCTGCACAGCCTTGAGC 66 11252 11271 125 2812 2831 464203 TCTCCAGCCAGGCTCAGCCA 72 11482 11501 126 2816 2835 464207 CAGCTCTCCAGCCAGGCTCA 72 11486 11505 127 2817 2836 464208 GCAGCTCTCCAGCCAGGCTC 78 11487 11506 128 2818 2837 464209 AGCAGCTCTCCAGCCAGGCT 79 11488 11507 129 2819 2838 464210 TAGCAGCTCTCCAGCCAGGC 70 11489 11508 130 2822 2841 464213 GCATAGCAGCTCTCCAGCCA 82 11492 11511 131 2823 2842 464214 AGCATAGCAGCTCTCCAGCC 85 11493 11512 132 2824 2843 464215 TAGCATAGCAGCTCTCCAGC 84 11494 11513 133 2825 2844 464216 TTAGCATAGCAGCTCTCCAG 72 11495 11514 134 2951 2970 464222 CCAGCTTCTCTGGGCTCAGG 88 11621 11640 135 2952 2971 464223 TCCAGCTTCTCTGGGCTCAG 86 11622 11641 136 2953 2972 464224 TTCCAGCTTCTCTGGGCTCA 81 11623 11642 137 2954 2973 464225 CTTCCAGCTTCTCTGGGCTC 82 11624 11643 138 2955 2974 464226 GCTTCCAGCTTCTCTGGGCT 79 11625 11644 139 2956 2975 464227 GGCTTCCAGCTTCTCTGGGC 87 11626 11645 140 2981 3000 464228 ACGCCATTTGCTCCTGTTTT 89 11651 11670 141 n/a n/a 464238 TGCGAATCAATGGGTCCCGA 73 908 927 142 n/a n/a 464239 GGTGCGAATCAATGGGTCCC 67 910 929 143 n/a n/a 464254 CCGCCGGCGCGAAGACAGCC 66 984 1003 144 n/a n/a 464258 CATCTCTGCCGCCGGCGCGA 71 992 1011 145 n/a n/a 464266 CTGACCGCTGACCGACCACC 76 1138 1157 146 n/a n/a 464268 GCTGCTGACCGCTGACCGAC 73 1142 1161 147 n/a n/a 464269 CTGCCCTGATATCAGAGTCC 65 1180 1199 148 n/a n/a 464270 GGCTGCCCTGATATCAGAGT 65 1182 1201 149 n/a n/a 464278 CTCAGATACTGCTGTCTCTG 71 1345 1364 150 n/a n/a 464280 TGCCCATCCCTCTGTGCCCC 72 1386 1405 151 n/a n/a 464284 TGCTCTCTTGCCCATCCCTC 82 1394 1413 152 n/a n/a 464285 CTCTTTGGTCACACCGTCTG 82 1461 1480 153 n/a n/a 464286 ATCTCTTTGGTCACACCGTC 90 1463 1482 154 n/a n/a 464287 CTATCTCTTTGGTCACACCG 82 1465 1484 155 n/a n/a 464288 GCCTATCTCTTTGGTCACAC 88 1467 1486 156 n/a n/a 464290 CGCTGCCTATCTCTTTGGTC 70 1471 1490 157 n/a n/a 464291 AGCTTGCAAGCCCTTAATGG 70 1542 1561 158 n/a n/a 464292 CCAGCTTGCAAGCCCTTAAT 69 1544 1563 159 n/a n/a 464298 ACCTTCATCTTCCAGCAGAG 80 1941 1960 160 n/a n/a 464299 CAACCTTCATCTTCCAGCAG 76 1943 1962 161 n/a n/a 464300 TTCAACCTTCATCTTCCAGC 81 1945 1964 162 n/a n/a 464308 CAGCTTTGCTCAGCCCAGCA 90 3325 3344 163 n/a n/a 464309 TCCAGCTTTGCTCAGCCCAG 87 3327 3346 164 n/a n/a 464310 TTTCCAGCTTTGCTCAGCCC 78 3329 3348 165 n/a n/a 464311 CCTTTCCAGCTTTGCTCAGC 78 3331 3350 166 n/a n/a 464333 CCAGGTCCACAGTCCAGGGC 75 4799 4818 167 n/a n/a 464342 ACTTGCCAGAGAGTAGCAGA 66 4836 4855 168 n/a n/a 464425 GCCATAGCACCTCCTCCAGG 75 7684 7703 169 n/a n/a 464428 CCCAATGCCATAGCACCTCC 73 7690 7709 170 n/a n/a 464429 GTCCCAATGCCATAGCACCT 70 7692 7711 171 n/a n/a 464430 TAGTCCCAATGCCATAGCAC 65 7694 7713 172 n/a n/a 464433 TTCTATTAGTCCCAATGCCA 69 7700 7719 173 n/a n/a 464449 GTCACTTGCCAGGGTCAGGA 81 7802 7821 174 n/a n/a 464453 GCTCAGAAGTCACTTGCCAG 68 7810 7829 175 n/a n/a 464568 GTCCATCTGGCTTCCCCTGC 68 2031 2050 176 n/a n/a 464569 CAGTCCATCTGGCTTCCCCT 68 2033 2052 177 n/a n/a 464575 CCACTCCACTTCCAGTCCAT 65 2045 2064 178 n/a n/a 464576 TGCCACTCCACTTCCAGTCC 68 2047 2066 179 n/a n/a 464579 GGTCACTGCCACTCCACTTC 78 2053 2072 180 n/a n/a 464581 CCTTGGTCACTGCCACTCCA 68 2057 2076 181 n/a n/a 464582 GGAAGCCTATCACACCTCCT 67 2080 2099 182 n/a n/a 464584 GTGTCTCTGGATCTACCCTG 71 2104 2123 183 n/a n/a 464585 TGGTGTCTCTGGATCTACCC 74 2106 2125 184 n/a n/a 464586 ACTGGTGTCTCTGGATCTAC 72 2108 2127 185 n/a n/a 464587 GCACTGGTGTCTCTGGATCT 83 2110 2129 186 n/a n/a 464588 TGGCACTGGTGTCTCTGGAT 88 2112 2131 187 n/a n/a 464589 GGTGGCACTGGTGTCTCTGG 88 2114 2133 188 n/a n/a 464590 TGGGTGGCACTGGTGTCTCT 74 2116 2135 189 n/a n/a 464591 TATGGGTGGCACTGGTGTCT 74 2118 2137 190 n/a n/a 464593 GGCCTATGGGTGGCACTGGT 68 2122 2141 191 n/a n/a 464617 GTCAGGCTGTGATGTACACA 69 2261 2280 192 n/a n/a 464622 TGCTGTTACTGTCAGGCTGT 73 2271 2290 193 n/a n/a 464623 GCCAGTCACCTCTGGTTCGG 68 2292 2311 194 n/a n/a 464657 AGCAGTTTTGGGATTCTTTT 72 2838 2857 195 n/a n/a 464658 AAAGCAGTTTTGGGATTCTT 67 2840 2859 196 n/a n/a 464677 TCCAAGTCCCTGGCCAGGCT 65 2993 3012 197 n/a n/a 464682 ATCCTTTCCAGCTTTGCTCA 78 3333 3352 198 n/a n/a 464683 GGATCCTTTCCAGCTTTGCT 90 3335 3354 199 n/a n/a 464684 AAGGATCCTTTCCAGCTTTG 72 3337 3356 200 n/a n/a 464685 GCAAGGATCCTTTCCAGCTT 88 3339 3358 201 n/a n/a 464686 GGGCAAGGATCCTTTCCAGC 82 3341 3360 202 n/a n/a 464687 CTGGGCAAGGATCCTTTCCA 71 3343 3362 203 n/a n/a 464688 GCCTGGGCAAGGATCCTTTC 69 3345 3364 204 n/a n/a 464689 GTGGTTGAGCCCTGCCCTGC 67 3380 3399 205 n/a n/a 464692 GTCTCAGTGGTTGAGCCCTG 71 3386 3405 206 n/a n/a 464696 CTGACTGAGTCTCAGTGGTT 82 3394 3413 207 n/a n/a 464698 GGCACTGACTGAGTCTCAGT 84 3398 3417 208 n/a n/a 464699 CAGGCACTGACTGAGTCTCA 79 3400 3419 209 n/a n/a 464701 AAGCCAGGCACTGACTGAGT 72 3404 3423 210 n/a n/a 464703 CTGGAAGCCAGGCACTGACT 70 3408 3427 211 n/a n/a 464705 GCTTGCTGGAAGCCAGGCAC 67 3413 3432 212 n/a n/a 464706 ATGCTTGCTGGAAGCCAGGC 80 3415 3434 213 n/a n/a 464707 GTCCTCTCTCGCAGACACAG 84 3445 3464 214 n/a n/a 464708 CAGTCCTCTCTCGCAGACAC 86 3447 3466 215 n/a n/a 464709 GCCAGTCCTCTCTCGCAGAC 86 3449 3468 216 n/a n/a 464710 AGGCCAGTCCTCTCTCGCAG 90 3451 3470 217 n/a n/a 464711 GAGCTCACCACCAGCTCTGC 70 3499 3518 218 n/a n/a 464716 GCTGCCTGGACCTCCTAGGT 90 3571 3590 219 n/a n/a 464717 ATGCTGCCTGGACCTCCTAG 85 3573 3592 220 n/a n/a 464718 ACATGCTGCCTGGACCTCCT 89 3575 3594 221 n/a n/a 464719 ACACATGCTGCCTGGACCTC 73 3577 3596 222 n/a n/a 464720 CCACACATGCTGCCTGGACC 88 3579 3598 223 n/a n/a 464726 GCAAATGCCACACTCTTGGG 67 3770 3789 224 n/a n/a 464727 GGGCAAATGCCACACTCTTG 78 3772 3791 225 n/a n/a 464728 CAGGGCAAATGCCACACTCT 71 3774 3793 226 n/a n/a 464729 CCCAGGGCAAATGCCACACT 87 3776 3795 227 n/a n/a 464730 CACCCAGGGCAAATGCCACA 78 3778 3797 228 n/a n/a 464732 GCCACACCCAGGGCAAATGC 87 3782 3801 229 n/a n/a 464734 GGATGCCACACCCAGGGCAA 66 3786 3805 230 n/a n/a 464735 GCGGATGCCACACCCAGGGC 87 3788 3807 231 n/a n/a 464736 CTGCGGATGCCACACCCAGG 67 3790 3809 232 n/a n/a 464740 GCCACATGCTGCGGATGCCA 88 3798 3817 233 n/a n/a 464800 GGACTTCCCACCAACTGCCT 71 3122 3141 234 n/a n/a 464801 GCTGGACTTCCCACCAACTG 77 3125 3144 235

TABLE 4 Inhibition of human FGFR4 mRNA levels by chimeric antisense oligonucleotides targeted to SEQ ID NO: 3 Target Start ISIS  % SEQ ID Site Sequence No inhibition NO 1502 CAAGGAGCTCACCACCAGCT 464713 83 236 1504 GGCAAGGAGCTCACCACCAG 464714 76 237 1506 CAGGCAAGGAGCTCACCACC 464715 69 238

Example 4 Dose-Dependent Antisense Inhibition of Human FGFR4 in HepG2 Cells

Gapmers from Example 3 which caused significant inhibition of FGFR4 mRNA were further tested at various doses in HepG2 cells. Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.6 μM, 1.3 μM, 2.5 μM, 5.0 μM, and 10.0 μM concentrations of antisense oligonucleotide, as specified in Table 5. After a treatment period of approximately 16 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human FGFR4 primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells.

The half maximal inhibitory concentration (IC50) of each oligonucleotide is also presented in Table 5. As illustrated in Table 5, FGFR4 mRNA levels were significantly reduced in a dose-dependent manner in antisense oligonucleotide treated cells.

TABLE 5 Dose-dependent antisense inhibition of human FGFR4 in HepG2 cells using electroporation 0.6 1.3 2.5 5.0 10.0 IC50 ISIS No μM μM μM μM μM (μM) 299004 20 44 73 87 96 1.5 463629 23 54 80 87 96 1.0 464138 0 32 57 84 91 2.3 464208 28 37 58 76 87 1.8 464209 22 30 64 79 80 2.0 464213 21 40 54 79 90 1.9 464214 14 31 55 84 93 2.1 464215 35 38 67 85 94 1.4 464222 29 53 73 89 93 1.2 464223 16 0 63 76 88 3.2 464225 36 43 74 85 88 1.2 464227 29 56 64 86 90 1.3 464228 52 76 82 91 92 0.3 464284 21 44 67 83 91 1.6 464285 27 36 57 81 93 1.9 464286 35 47 70 89 95 1.2 464287 26 50 68 85 90 1.4 464288 19 49 55 83 90 1.7 464300 25 34 47 75 93 2.1 464308 35 57 77 94 97 1.0 464309 4 25 65 89 95 2.1 464425 2 31 52 71 81 2.7 464449 32 59 78 88 95 1.0 464587 25 52 75 88 91 1.3 464588 26 74 84 93 93 1.0 464589 29 62 83 90 93 1.0 464683 10 35 50 71 90 2.4 464685 14 42 42 62 88 2.6 464686 12 44 66 81 95 1.8 464696 22 43 68 85 94 1.6 464698 12 10 20 44 71 5.9 464706 16 52 46 84 92 1.8 464707 26 40 69 84 93 1.5 464708 18 46 57 84 94 1.7 464709 6 14 32 58 84 3.7 464710 12 30 44 65 86 2.7 464713 9 28 47 78 92 2.4 464716 21 45 64 86 93 1.6 464717 13 37 57 86 94 2.0 464718 22 56 80 93 97 1.2 464720 15 33 49 77 92 2.2 464729 15 20 35 69 84 3.0 464732 19 55 73 85 93 1.4 464735 27 45 62 89 94 1.5 464740 10 44 65 82 89 1.9 464801 17 53 56 81 92 1.7

Example 5 Dose-Dependent Antisense Inhibition of Human FGFR4 in HepG2 Cells

Gapmers from the studies described above which caused significant inhibition of FGFR4 mRNA were further tested at various doses in HepG2 cells. Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.3 μM, 0.6 μM, 1.3 μM, 2.5 μM, 5.0 μM, and 10.0 μM concentrations of antisense oligonucleotide, as specified in Table 6. After a treatment period of approximately 16 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human FGFR4 primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells.

The half maximal inhibitory concentration (IC50) of each oligonucleotide is also presented in Table 6. As illustrated in Table 6, FGFR4 mRNA levels were significantly reduced in a dose-dependent manner in antisense oligonucleotide treated cells.

TABLE 6 Dose-dependent antisense inhibition of human FGFR4 in HepG2 cells using electroporation 0.3 0.6 1.3 2.5 5.0 10.0 IC50 ISIS No μM μM μM μM μM μM (μM) 299004 0 20 14 49 70 89 2.5 299005 7 25 52 76 92 96 1.3 463588 26 22 43 84 94 98 1.3 463589 13 24 52 74 92 95 1.3 463628 27 45 57 76 94 95 0.8 463629 24 36 67 85 93 96 0.9 463648 14 21 38 54 75 90 1.9 463672 8 28 41 57 86 95 1.6 463690 22 17 59 74 91 97 1.3 463691 10 24 45 60 86 87 1.6 463692 0 10 33 56 76 92 2.2 463709 12 22 36 66 85 95 1.6 463762 0 22 16 29 0 84 >10.0 463771 0 29 38 49 66 89 2.2 463834 14 24 43 52 79 94 1.7 463835 18 35 40 58 82 94 1.4 463837 8 22 53 73 89 97 1.4 463838 12 23 44 56 77 91 1.7 463861 25 41 41 61 76 90 1.3 463907 0 25 51 68 84 95 1.6 463909 19 39 54 82 93 97 1.0 464038 8 22 36 44 72 89 2.2

Example 6 Dose-Dependent Antisense Inhibition of Human FGFR4 in HepG2 Cells

Gapmers from the study described in Example 4 exhibiting significant in vitro inhibition of FGFR4 mRNA were selected and tested at various doses in HepG2 cells. Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.6 μM, 1.3 μM, 2.5 μM, 5.0 μM and 10.0 μM concentrations of antisense oligonucleotide, as specified in Table 7. After a treatment period of approximately 16 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human FGFR4 primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells.

The half maximal inhibitory concentration (IC50) of each oligonucleotide is also presented in Table 7. As illustrated in Table 7, FGFR4 mRNA levels were significantly reduced in a dose-dependent manner in antisense oligonucleotide treated cells.

TABLE 7 Dose-dependent antisense inhibition of human FGFR4 in HepG2 cells using electroporation 0.6 1.3 2.5 5.0 10.0 IC50 ISIS No μM μM μM μM μM (μM) 463629 44 73 86 96 98 <0.6 464222 40 65 81 88 96 0.7 464225 47 76 84 92 86 <0.6 464228 56 80 86 92 95 <0.6 464284 24 47 62 78 90 1.6 464286 23 60 73 86 93 1.2 464287 19 62 69 89 91 1.3 464308 38 54 78 90 96 1.0 464449 27 69 80 91 94 0.9 464587 24 68 74 88 91 1.1 464588 42 75 81 88 92 <0.6 464589 36 69 78 90 92 0.8 464716 52 60 75 90 95 0.6 464718 38 61 76 91 95 0.9 464732 30 39 65 85 94 1.5

Example 7 Antisense Inhibition of Human Fibroblast Growth Factor Receptor (FGFR4) in HepG2 Cells

Additional antisense oligonucleotides were designed targeting a FGFR4 nucleic acid and were tested for their effects on FGFR4 mRNA in vitro. ISIS 463629 and ISIS 463762 were also included in the assay for comparison. Cultured HepG2 cells at a density of 20,000 cells per well were transfected using electroporation with 4,500 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells. A total of 230 oligonucleotides were tested. Only those oligonucleotides demonstrating greater than 65% inhibition are shown in Table 8.

The newly designed chimeric antisense oligonucleotides in Table 8 were designed as 5-10-5 MOE gapmers or 3-10-4 MOE gapmers. The 5-10-5 gapmers are 20 nucleosides in length, wherein the central gap segment comprises of ten 2′-deoxynucleosides and is flanked by wing segments on the 5′ and 3′ directions comprising five nucleosides each. The 3-10-4 gapmers are 17 nucleosides in length, wherein the central gap segment comprises of ten 2′-deoxynucleosides and is flanked by a wing segment in the 5′ direction comprising three nucleosides and a wing segment in the 3′ direction comprising four nucleosides. Each nucleotide in the 5′ wing segment and each nucleotide in the 3′ wing segment has a 2′-MOE modification. The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine residues throughout each gapmer are 5-methylcytosines. “Start site” indicates the 5′-most nucleoside to which the gapmer is targeted in the human gene sequence. “Stop site” indicates the 3′-most nucleoside to which the gapmer is targeted in the human gene sequence.

Each gapmer listed in Table 8 is targeted to either the human FGFR4 mRNA, designated herein as SEQ ID NO: 1 (GENBANK Accession No. NM002011.3) or the human FGFR4 genomic sequence, designated herein as SEQ ID NO: 2 (GENBANK Accession No: NT023133.11 truncated from nucleosides 21323018 to 21335213), or SEQ ID NO: 3 (GENBANK Accession No. AB209631.1), or all three.

TABLE 8 Inhibition of human FGFR4 mRNA levels by chimeric antisense oligo- nucleotides targeted to SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3 Start Start Start Site on Site on Site on SEQ SEQ SEQ SEQ ID NO: ID NO: ID NO: ISIS % ID 1 2 3 Motif Sequence No inhibition NO 292 3993 1996 5-10-5 GGCTACTGTCAGCTCCTGCT 463629 93 29 118 3123 1122 5-10-5 TGGACTTCCCACCAACTGCC 479530 77 239 n/a 2109 101 5-10-5 CACTGGTGTCTCTGGATCTA 479532 78 240 n/a 2111 103 5-10-5 GGCACTGGTGTCTCTGGATC 479533 89 241 n/a 2113 105 5-10-5 GTGGCACTGGTGTCTCTGGA 479534 92 242 n/a 2115 107 5-10-5 GGGTGGCACTGGTGTCTCTG 479535 88 243 n/a 3334 1333 5-10-5 GATCCTTTCCAGCTTTGCTC 479536 91 244 n/a 3336 1335 5-10-5 AGGATCCTTTCCAGCTTTGC 479537 84 245 n/a 3338 1337 5-10-5 CAAGGATCCTTTCCAGCTTT 479538 65 246 n/a 3340 1339 5-10-5 GGCAAGGATCCTTTCCAGCT 479539 88 247 n/a 3342 1341 5-10-5 TGGGCAAGGATCCTTTCCAG 479540 68 248 n/a 3393 1392 5-10-5 TGACTGAGTCTCAGTGGTTG 479541 71 249 n/a 3395 1394 5-10-5 ACTGACTGAGTCTCAGTGGT 479542 80 250 n/a 3397 1396 5-10-5 GCACTGACTGAGTCTCAGTG 479543 76 251 n/a 3399 1398 5-10-5 AGGCACTGACTGAGTCTCAG 479544 77 252 n/a 3414 1413 5-10-5 TGCTTGCTGGAAGCCAGGCA 479545 83 253 n/a 3446 1445 5-10-5 AGTCCTCTCTCGCAGACACA 479546 88 254 n/a 3448 1447 5-10-5 CCAGTCCTCTCTCGCAGACA 479547 80 255 n/a 3450 1449 5-10-5 GGCCAGTCCTCTCTCGCAGA 479548 92 256 n/a 3502 1501 5-10-5 AAGGAGCTCACCACCAGCTC 479549 76 257 n/a n/a 1503 5-10-5 GCAAGGAGCTCACCACCAGC 479550 79 258 n/a 3570 1569 5-10-5 CTGCCTGGACCTCCTAGGTC 479551 95 259 n/a 3572 1571 5-10-5 TGCTGCCTGGACCTCCTAGG 479552 85 260 n/a 3574 1573 5-10-5 CATGCTGCCTGGACCTCCTA 479553 80 261 n/a 3576 1575 5-10-5 CACATGCTGCCTGGACCTCC 479554 80 262 n/a 3578 1577 5-10-5 CACACATGCTGCCTGGACCT 479555 71 263 n/a 3580 1579 5-10-5 ACCACACATGCTGCCTGGAC 479556 87 264 n/a 3775 1778 5-10-5 CCAGGGCAAATGCCACACTC 479557 71 265 n/a 3777 1780 5-10-5 ACCCAGGGCAAATGCCACAC 479558 83 266 n/a 3783 1786 5-10-5 TGCCACACCCAGGGCAAATG 479560 67 267 n/a 3787 1790 5-10-5 CGGATGCCACACCCAGGGCA 479561 70 268 n/a 3789 1792 5-10-5 TGCGGATGCCACACCCAGGG 479562 78 269 n/a 3799 1802 5-10-5 AGCCACATGCTGCGGATGCC 479564 71 270 n/a 1393 n/a 5-10-5 GCTCTCTTGCCCATCCCTCT 479565 81 271 n/a 1462 n/a 5-10-5 TCTCTTTGGTCACACCGTCT 479566 90 272 n/a 1464 n/a 5-10-5 TATCTCTTTGGTCACACCGT 479567 67 273 n/a 1466 n/a 5-10-5 CCTATCTCTTTGGTCACACC 479568 83 274 n/a 1468 n/a 5-10-5 TGCCTATCTCTTTGGTCACA 479569 76 275 n/a 1944 n/a 5-10-5 TCAACCTTCATCTTCCAGCA 479570 80 276 n/a 3324 1323 5-10-5 AGCTTTGCTCAGCCCAGCAG 479572 75 277 n/a 3326 1325 5-10-5 CCAGCTTTGCTCAGCCCAGC 479573 85 278 n/a 3328 1327 5-10-5 TTCCAGCTTTGCTCAGCCCA 479574 79 279 n/a 7801 n/a 5-10-5 TCACTTGCCAGGGTCAGGAG 479576 70 280 n/a 7803 n/a 5-10-5 AGTCACTTGCCAGGGTCAGG 479577 65 281 n/a 1462 n/a 3-10-4 CTTTGGTCACACCGTCT 479582 74 282 n/a 1463 n/a 3-10-4 TCTTTGGTCACACCGTC 479583 84 283 n/a 1464 n/a 3-10-4 CTCTTTGGTCACACCGT 479584 82 284 n/a 1465 n/a 3-10-4 TCTCTTTGGTCACACCG 479585 71 285 n/a 3326 1325 3-10-4 GCTTTGCTCAGCCCAGC 479594 80 286 n/a 3328 1327 3-10-4 CAGCTTTGCTCAGCCCA 479596 81 287 n/a 3329 1328 3-10-4 CCAGCTTTGCTCAGCCC 479597 78 288 161 3166 1165 3-10-4 GCAGCCGCATCTCCTTC 479608 70 289 194 3199 1198 3-10-4 GCACACTCAGCAGGACC 479613 72 290 195 3200 1199 3-10-4 GGCACACTCAGCAGGAC 479614 78 291 349 4050 2053 3-10-4 CCAGTGGCCACCACGCT 479622 67 292 369 4070 2073 3-10-4 AGGCGACTGCCCTCCTT 479625 68 293 370 4071 2074 3-10-4 CAGGCGACTGCCCTCCT 479626 71 294 602 4506 2418 3-10-4 GTGTCCAGTAGGGTGCT 479641 70 295 2819 11489 4988 3-10-4 CAGCTCTCCAGCCAGGC 479682 71 296 2951 11621 5120 3-10-4 GCTTCTCTGGGCTCAGG 479689 72 297 2952 11622 5121 3-10-4 AGCTTCTCTGGGCTCAG 479690 78 298 2953 11623 5122 3-10-4 CAGCTTCTCTGGGCTCA 479691 87 299 2954 11624 5123 3-10-4 CCAGCTTCTCTGGGCTC 479692 87 300 2955 11625 5124 3-10-4 TCCAGCTTCTCTGGGCT 479693 71 301 2956 11626 5125 3-10-4 TTCCAGCTTCTCTGGGC 479694 67 302 2958 11628 5127 3-10-4 GCTTCCAGCTTCTCTGG 479696 65 303 2981 11651 5150 3-10-4 CCATTTGCTCCTGTTTT 479697 73 304 2982 11652 5151 3-10-4 GCCATTTGCTCCTGTTT 479698 88 305 2983 11653 5152 3-10-4 CGCCATTTGCTCCTGTT 479699 92 306 n/a 2113 105 3-10-4 GCACTGGTGTCTCTGGA 479703 88 307 n/a 2114 106 3-10-4 GGCACTGGTGTCTCTGG 479704 95 308 n/a 2115 107 3-10-4 TGGCACTGGTGTCTCTG 479705 78 309 n/a 2116 108 3-10-4 GTGGCACTGGTGTCTCT 479706 90 310 n/a 3395 1394 3-10-4 GACTGAGTCTCAGTGGT 479716 71 311 n/a 3415 1414 3-10-4 CTTGCTGGAAGCCAGGC 479721 82 312 n/a 3416 1415 3-10-4 GCTTGCTGGAAGCCAGG 479722 82 313 n/a 3446 1445 3-10-4 CCTCTCTCGCAGACACA 479725 70 314 n/a 3452 1451 3-10-4 GCCAGTCCTCTCTCGCA 479731 78 315 n/a 3453 1452 3-10-4 GGCCAGTCCTCTCTCGC 479732 69 316 n/a 3571 1570 3-10-4 GCCTGGACCTCCTAGGT 479736 97 317 n/a 3572 1571 3-10-4 TGCCTGGACCTCCTAGG 479737 69 318 n/a 3573 1572 3-10-4 CTGCCTGGACCTCCTAG 479738 76 319 n/a 3574 1573 3-10-4 GCTGCCTGGACCTCCTA 479739 88 320 n/a 3575 1574 3-10-4 TGCTGCCTGGACCTCCT 479740 66 321 n/a 3576 1575 3-10-4 ATGCTGCCTGGACCTCC 479741 72 322

Example 8 Dose-Dependent Antisense Inhibition of Human FGFR4 in HepG2 Cells

Gapmers from Examples 5, 6 and 7 exhibiting significant in vitro inhibition of FGFR4 mRNA were further selected and tested at various doses in HepG2 cells. Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0.6 μM, 1.3 μM, 2.5 μM, 5.0 μM, and 10.0 μM concentrations of antisense oligonucleotide, as specified in Table 9. After a treatment period of approximately 16 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human FGFR4 primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells.

The half maximal inhibitory concentration (IC50) of each oligonucleotide is also presented in Table 9. As illustrated in Table 9, FGFR4 mRNA levels were significantly reduced in a dose-dependent manner in antisense oligonucleotide treated cells.

TABLE 9 Dose-dependent antisense inhibition of human FGFR4 in HepG2 cells using electroporation 0.6 1.3 2.5 5.0 10.0 IC50 ISIS No μM μM μM μM μM (μM) 299005 31 50 66 89 95 1.3 463588 25 48 70 91 97 1.4 463589 33 46 69 87 96 1.3 463628 49 67 77 90 97 <0.6 463629 36 58 70 88 92 1.6 463648 34 41 49 64 84 1.9 463672 16 34 68 84 94 1.8 463690 42 58 75 88 97 0.8 463691 27 38 73 83 96 1.5 463692 3 39 57 76 94 2.2 463709 22 43 64 82 95 1.6 463762 13 29 46 74 90 2.5 463771 40 31 51 78 91 1.7 463834 23 44 55 78 93 1.8 463835 30 39 65 83 95 1.5 463837 29 43 72 87 95 1.4 463838 23 40 59 77 93 1.8 463861 9 33 61 82 97 2.1 464038 19 25 42 61 88 2.8 464222 30 56 75 87 95 1.1 464225 40 60 79 85 90 0.8 464228 50 72 86 91 94 <0.6 464284 30 52 59 84 90 1.4 464286 50 65 83 92 95 <0.6 464287 24 50 67 89 92 1.4 464308 36 56 76 90 97 1.0 464449 44 73 85 93 95 <0.6 464587 33 54 79 92 98 1.0 464588 53 76 89 95 95 <0.6 464589 30 66 80 93 95 0.9 464716 33 41 69 86 95 1.4 464718 33 56 77 93 98 1.0 464732 27 43 61 86 95 1.6 479533 68 84 89 93 95 <0.6 479534 67 74 92 95 97 <0.6 479535 54 72 81 91 95 <0.6 479536 38 68 86 96 98 0.7 479539 39 52 77 92 98 1.0 479546 32 70 78 91 98 0.9 479548 49 71 81 93 96 <0.6 479551 72 82 91 95 97 <0.6 479556 36 63 83 90 97 0.9

Example 9 Dose-Dependent Antisense Inhibition of Human FGFR4 in HepG2 Cells

Gapmers from Examples 7 and 8 exhibiting significant in vitro inhibition of FGFR4 mRNA were further selected and tested at various doses in HepG2 cells. Cells were plated at a density of 20,000 cells per well and transfected using electroporation with 0156 μM, 0.31 μM, 0.63 μM, 1.25 μM, 2.50 μM and 5.00 μM concentrations of antisense oligonucleotide, as specified in Table 10. After a treatment period of approximately 16 hours, RNA was isolated from the cells and FGFR4 mRNA levels were measured by quantitative real-time PCR. Human FGFR4 primer probe set RTS3232 was used to measure mRNA levels. FGFR4 mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN®. Results are presented as percent inhibition of FGFR4, relative to untreated control cells.

The half maximal inhibitory concentration (IC50) of each oligonucleotide is also presented in Table 10. As illustrated in Table 10, FGFR4 mRNA levels were significantly reduced in a dose-dependent manner in antisense oligonucleotide treated cells.

TABLE 10 Dose-dependent antisense inhibition of human FGFR4 in HepG2 cells using electroporation 0.156 0.31 0.63 1.25 2.50 5.00 IC50 ISIS No μM μM μM μM μM μM (μM) 463629 19 30 48 66 84 91 0.7 479533 18 33 63 67 84 86 0.6 479534 18 25 34 63 82 86 0.9 479535 24 28 43 62 68 81 0.9 479536 25 28 29 62 78 90 0.8 479539 8 16 36 48 75 88 1.1 479546 0 27 33 63 77 87 1.1 479548 8 39 30 62 74 85 0.9 479551 27 44 59 80 86 89 0.4 479556 16 29 32 53 71 87 1.0 479566 19 27 29 63 81 88 0.9 479584 3 22 30 58 80 88 1.0 479596 4 20 32 54 71 88 1.1 479691 18 11 50 62 80 91 0.8 479692 12 26 49 61 79 90 0.8 479698 23 40 57 73 87 92 0.5 479699 17 37 60 76 90 93 0.5 479703 18 20 41 67 82 89 0.8 479704 31 43 66 80 90 92 0.4 479706 26 18 36 58 76 90 0.9 479736 36 48 71 86 93 94 0.3

Example 10 Tolerability of Antisense Oligonucleotides Targeting Human FGFR4 in CD1 Mice

CD1® mice (Charles River, MA) are a multipurpose mice model, frequently utilized for safety and efficacy testing. The mice were treated with ISIS antisense oligonucleotides selected from studies described above and evaluated for changes in the levels of various markers.

Treatment

Groups of five male CD1 mice were injected subcutaneously twice a week for 6 weeks with 50 mg/kg of ISIS 299005, ISIS 463588, ISIS 463589, ISIS 463628, ISIS 463690, ISIS 463691, ISIS 463835, ISIS 463837, ISIS 464222, ISIS 464225, ISIS 464228, ISIS 464286, ISIS 464308, ISIS 464449, ISIS 464587, ISIS 464588, ISIS 464589, ISIS 464718, ISIS 479533, ISIS 479551, ISIS 479691, ISIS 479692, ISIS 479698, ISIS 479699, ISIS 479703, ISIS 479704, ISIS 479706, or ISIS 479736. One group of male CD1 mice was injected subcutaneously twice a week for 6 weeks with PBS. Mice were euthanized 48 hours after the last dose, and plasma were harvested for further analysis. Treatment with ISIS 479691 caused death of the mice and that ISIS oligonucleotide was therefore removed from further study.

Plasma Chemistry Markers

To evaluate the effect of ISIS oligonucleotides on metabolic function, plasma concentrations of transaminases, bilirubin, albumin, creatinine, and BUN were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). The transaminase levels are expressed as IU/L; the bilirubin, creatinine, and BUN levels are expressed as mg/dL; and the albumin is expressed in g/dL. The results are presented in Table 11. ISIS oligonucleotides that caused adverse changes in the levels of any of the plasma chemistry markers were excluded in further studies.

TABLE 11 ALT, AST, Bilirubin, BUN, Creatinine and Albumin levels in CD1 mouse plasma at week 6 ALT AST (IU/ (IU/ Bilirubin BUN Creatinine Albumin L) L) (mg/dL) (mg/dL) (mg/dL) (g/dL) PBS 34 59 0.2 33 0.16 3.3 ISIS 299005 50 72 0.1 27 0.12 2.8 ISIS 463588 55 72 0.1 30 0.12 3.0 ISIS 463589 64 79 0.2 28 0.10 2.7 ISIS 463628 48 83 0.1 27 0.13 3.0 ISIS 463690 71 93 0.2 29 0.13 3.0 ISIS 463691 145 134 0.2 26 0.10 3.0 ISIS 463835 159 113 0.2 26 0.11 3.0 ISIS 463837 59 78 0.1 27 0.09 2.8 ISIS 464222 559 564 0.2 23 0.09 2.7 ISIS 464225 83 88 0.1 25 0.09 2.8 ISIS 464228 58 93 0.1 29 0.10 2.8 ISIS 464286 139 154 0.1 21 0.05 2.8 ISIS 464308 2533 1673 0.2 28 0.11 3.3 ISIS 464449 748 451 0.2 24 0.08 3.0 ISIS 464587 183 159 0.1 25 0.11 3.0 ISIS 464588 256 726 0.2 21 0.03 2.0 ISIS 464589 142 126 0.2 27 0.09 2.9 ISIS 464718 789 608 0.2 19 0.03 2.7 ISIS 479533 61 76 0.1 22 0.09 2.9 ISIS 479551 81 104 0.2 26 0.13 3.0 ISIS 479692 847 1026 0.3 26 0.10 3.1 ISIS 479698 92 133 0.2 29 0.12 2.7 ISIS 479699 57 95 0.1 20 0.09 2.6 ISIS 479703 158 108 0.1 23 0.11 3.0 ISIS 479704 38 56 0.2 23 0.10 3.2 ISIS 479706 700 642 0.5 26 0.12 3.1 ISIS 479736 204 134 0.1 25 0.11 2.9

Example 11 Tolerability of Antisense Oligonucleotides Targeting Human FGFR4 in Sprague-Dawley Rats

Sprague-Dawley rats are a multipurpose model used for safety and efficacy evaluations. The rats were treated with ISIS antisense oligonucleotides from the study described in Example 10 and evaluated for changes in the levels of various plasma chemistry markers.

Treatment

Seven week old male Sprague-Dawley rats were maintained on a 12-hour light/dark cycle and fed ad libitum with Purina normal rat chow, diet 5001. Groups of four Sprague-Dawley rats each were injected subcutaneously twice a week for 4 weeks with 50 mg/kg of ISIS 299005, ISIS 463588, ISIS 463589, ISIS 463628, ISIS 463690, ISIS 463691, ISIS 463835, ISIS 463837, ISIS 464222, ISIS 464225, ISIS 464228, ISIS 464286, ISIS 464308, ISIS 464449, ISIS 464587, ISIS 464718, ISIS 479533, ISIS 479551, ISIS 479691, ISIS 479692, ISIS 479698, ISIS 479699, ISIS 479703, ISIS 479704, ISIS 479706, or ISIS 479736. A group of rats were injected subcutaneously twice a week for 4 weeks with PBS. Forty eight hours after the last dose, rats were euthanized and plasmas were harvested for further analysis.

Liver Function

To evaluate the effect of ISIS oligonucleotides on hepatic function, plasma concentrations of transaminases were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). Plasma concentrations of ALT (alanine transaminase) and AST (aspartate transaminase) were measured and the results are presented in Table 12, expressed in IU/L. Plasma levels of bilirubin were also measured using the same clinical chemistry analyzer and the results are also presented in Table 12, expressed as mg/dL. ISIS oligonucleotides that caused adverse changes were excluded in further studies.

TABLE 12 Effect of antisense oligonucleotide treatment on ALT, AST, and Bilirubin in the liver of Sprague-Dawley rats ALT AST Bilirubin (IU/L) (IU/L) (g/dL) PBS 52 206 0.15 ISIS 299005 72 387 0.16 ISIS 463588 56 305 0.13 ISIS 463589 82 553 0.15 ISIS 463628 351 351 0.13 ISIS 463690 81 367 0.14 ISIS 463691 83 368 0.13 ISIS 463835 90 345 0.13 ISIS 463837 67 301 0.11 ISIS 464222 231 322 0.19 ISIS 464225 66 241 0.11 ISIS 464228 77 359 0.57 ISIS 464286 96 207 0.11 ISIS 464308 59 295 0.12 ISIS 464449 158 509 0.15 ISIS 464587 414 373 0.29 ISIS 464588 215 278 0.40 ISIS 464589 282 482 0.32 ISIS 464718 280 577 0.43 ISIS 479533 391 457 0.29 ISIS 479551 1360 1300 0.41 ISIS 479691 383 439 0.35 ISIS 479692 674 675 0.24 ISIS 479698 354 775 0.86 ISIS 479699 145 455 0.90 ISIS 479703 779 781 0.54 ISIS 479704 790 1243 0.41 ISIS 479706 570 680 0.36 ISIS 479736 499 644 0.24

Kidney Function

To evaluate the effect of ISIS oligonucleotides on kidney function, plasma concentrations of blood urea nitrogen (BUN) and creatinine were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). Results are presented in Table 13, expressed in mg/dL.

TABLE 13 Effect of antisense oligonucleotide treatment on renal function markers (mg/dL) of Sprague-Dawley rats BUN Creatinine Saline 18 0.29 ISIS 299005 20 0.33 ISIS 463588 23 0.35 ISIS 463589 19 0.32 ISIS 463628 19 0.33 ISIS 463690 19 0.35 ISIS 463691 18 0.34 ISIS 463835 19 0.34 ISIS 463837 18 0.32 ISIS 464222 21 0.37 ISIS 464225 20 0.29 ISIS 464228 22 0.32 ISIS 464286 22 0.36 ISIS 464308 18 0.32 ISIS 464449 16 0.32 ISIS 464587 23 0.38 ISIS 464588 23 0.27 ISIS 464589 24 0.35 ISIS 464718 22 0.32 ISIS 479533 28 0.31 ISIS 479551 21 0.36 ISIS 479691 29 0.36 ISIS 479692 25 0.40 ISIS 479698 30 0.34 ISIS 479699 30 0.35 ISIS 479703 28 0.31 ISIS 479704 31 0.42 ISIS 479706 26 0.38 ISIS 479736 22 0.37

Example 12 Tolerability of Antisense Oligonucleotides Targeting Human FGFR4 in CD/IGS Rats

CD/IGS rats are a multipurpose model used for safety and efficacy evaluations. The rats were treated with ISIS antisense oligonucleotides selected from the study described in Examples 10 and 11 and evaluated for changes in the levels of various markers.

Treatment

Ten-twelve week old male CD/IGS rats were maintained on a 12-hour light/dark cycle and fed ad libitum with Purina normal rat chow, diet 5001. Groups of four CD/IGS rats each were injected subcutaneously twice a week for 12 weeks with 30 mg/kg of ISIS 299005, ISIS 463588, ISIS 463589, ISIS 463690, ISIS 463691, ISIS 463835, ISIS 463837, or ISIS 464225. A group of 6 rats was injected subcutaneously twice a week for 12 weeks with PBS and served as a control group. Urine and blood samples were collected at various time points. Forty eight hours after the last dose, body weights were taken, rats were euthanized and organs and plasma were harvested for further analysis.

Liver Function

To evaluate the effect of ISIS oligonucleotides on hepatic function, plasma concentrations of various liver function markers were measured on week 8 and week 12 using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). Plasma concentrations of ALT (alanine transaminase) and AST (aspartate transaminase) were measured and the results are presented in Tables 14 and 15, expressed in IU/L. Plasma levels of bilirubin and BUN were also measured using the same clinical chemistry analyzer and the results are also presented in Tables 14 and 15, expressed as mg/dL. ISIS oligonucleotides that caused adverse changes in the levels of any of the markers of liver function were excluded in further studies.

TABLE 14 ALT, AST, Bilirubin and BUN of CD/IGS rats on week 8 ALT AST Bilirubin BUN (IU/L) (IU/L) (mg/dL) (mg/dL) PBS 31 71 0.16 13.6 ISIS 299005 60 121 0.15 17.4 ISIS 463588 57 103 0.19 18.6 ISIS 463589 46 136 0.14 16.8 ISIS 463690 79 91 0.24 18.1 ISIS 463691 80 93 0.18 18.8 ISIS 463835 103 118 0.18 16.6 ISIS 463837 52 101 0.14 20.7 ISIS 464225 48 253 0.14 18.9

TABLE 15 ALT, AST, TBIL, and BUN levels in the liver of CD/IGS rats on week 12 ALT AST TBIL BUN (IU/L) (IU/L) (mg/dL) (mg/dL) PBS 38 60 0.10 18.2 ISIS 299005 79 150 0.10 20.0 ISIS 463588 66 146 0.13 23.3 ISIS 463589 47 106 0.10 18.3 ISIS 463690 66 65 0.10 20.3 ISIS 463691 72 68 0.13 20.3 ISIS 463835 63 76 0.10 18.8 ISIS 463837 52 98 0.10 21.8 ISIS 464225 48 260 0.10 19.0

Example 13 Pharmacokinetic Measurement of Antisense Oligonucleotide in CD1 Mouse Liver

CD1 mice were treated with ISIS 463588, ISIS 463589, and ISIS 463690, and the oligonucleotide half-life as well as the elapsed time for oligonucleotide degradation and elimination from the liver was evaluated.

Treatment

Group of ten CD1 mice each were injected subcutaneously twice per week for 2 weeks (4 doses) with 50 mg/kg of ISIS 463588, ISIS 463589, or ISIS 463690. Groups of five mice each from each group were sacrificed 3 days and 56 days following the final dose. Livers were harvested for analysis.

Measurement of Oligonucleotide Concentration

The concentration of the full-length oligonucleotide as well as the total oligonucleotide concentration (including the degraded form) was measured. The method used is a modification of previously published methods (Leeds et al., 1996; Geary et al., 1999) which consist of a phenol-chloroform (liquid-liquid) extraction followed by a solid phase extraction. An internal standard (ISIS 355868, a 27-mer 2′-O-methoxyethyl modified phosphorothioate oligonucleotide, GCGTTTGCTCTTCTTCTTGCGTTTTTT, designated herein as SEQ ID NO: 323) was added prior to extraction. Tissue sample concentrations were calculated using calibration curves, with a lower limit of quantitation (LLOQ) of approximately 1.14 μg/g. Half-lives were then calculated using WinNonlin software (PHARSIGHT).

The results are presented in Table 16, expressed as μg/g liver tissue. The half-life of the ISIS oligonucleotides was calculated from these values and is also presented in Table 17. The half-life for each oligonucleotide was considered optimal.

TABLE 16 Oligonucleotide concentration of ISIS oligonucleotides in the liver of CD1 mice ISIS ISIS ISIS 463588 463589 463690 Day 3 157 168 196 Day 56 31 17 28

TABLE 17 Half-life of ISIS oligonucleotides in the liver of CD1 mice ISIS No Days 463588 22.4 463589 15.9 463690 18.7

Example 14 Measurement of Viscosity of ISIS Antisense Oligonucleotides Targeting Human FGFR4

The viscosity of the antisense oligonucleotides selected from in vivo studies described above was measured with the aim of screening out antisense oligonucleotides which have a viscosity more than 40 cP at a concentration of 165-185 mg/mL. Oligonucleotides having a viscosity greater than 40 cP would be too viscous to be administered to any subject.

ISIS oligonucleotides (32-35 mg) were weighed into a glass vial, 120 μL, of water was added and the antisense oligonucleotide was dissolved into solution by heating the vial at 50° C. Part of (75 μL) the pre-heated sample was pipetted to a micro-viscometer (Cambridge). The temperature of the micro-viscometer was set to 25° C. and the viscosity of the sample was measured. Another part (20 μL) of the pre-heated sample was pipetted into 10 mL of water for UV reading at 260 nM at 85° C. (Cary UV instrument). The results are presented in Table 18 and indicate that most of the antisense oligonucleotide solutions are optimal in their viscosity under the criterion stated above.

TABLE 18 Viscosity and concentration of ISIS antisense oligonucleotides targeting human FGFR4 ISIS Viscosity Concentration No. (cP) (mg/mL) 299005 44 174 463588 21 189 463589 17 174 463690 12 178 463691 9 194 463835 25 174 463837 8 181 464225 21 204

Example 15 Effect of ISIS Antisense Oligonucleotides Targeting Human FGFR4 in Cynomolgus Monkeys

Chinese cynomolgus monkeys were treated with ISIS antisense oligonucleotides selected from studies described in Examples 11-14. Antisense oligonucleotide efficacy and tolerability, as well as their pharmacokinetic profile in the liver and kidney, were evaluated. The human antisense oligonucleotides tested are also cross-reactive with the rhesus genomic sequence GENBANK Accession No NW001121000.1 truncated from nucleosides 3094000 to 3109000 (SEQ ID NO: 5). The greater the complementarity between the human oligonucleotide and the rhesus monkey sequence, the more likely the human oligonucleotide can cross-react with the rhesus monkey sequence. The start sites of each oligonucleotide to SEQ ID NO: 5 is presented in Table 19. “Target start site” indicates the 5′-most nucleotide to which the gapmer is targeted in the rhesus monkey gene sequence.

TABLE 19 Antisense oligonucleotides complementary to SEQ ID NO: 5 Target SEQ ID Start Site Sequence ISIS No Motif NO 4366 GGCACACTCAGCAGGACCCC 299005 5-10-5 7 4365 GCACACTCAGCAGGACCCCC 463588 5-10-5 16 4367 AGGCACACTCAGCAGGACCC 463589 5-10-5 17 5223 GCCAGGCGACTGCCCTCCTT 463690 5-10-5 45 5224 TGCCAGGCGACTGCCCTCCT 463691 5-10-5 46 6420 CGCTCTCCATCACGAGACTC 463835 5-10-5 70 6422 CACGCTCTCCATCACGAGAC 463837 5-10-5 72 12755 CTTCCAGCTTCTCTGGGCTC 464225 5-10-5 138

Treatment

This study was conducted at Charles River Laboratories, Nevada. Prior to the study, the monkeys were acclimated to their designated housing for at least 13 days before the start of dosing. The animals were confirmed to have at least one negative serum antibody test to simian retrovirus (SRV), as well as to other related viruses. Tuberculosis testing was also done. The animals were housed individually in stainless steel cages, as specified in the USDA Animal Welfare Act (9 CFR, Parts 1, 2, and 3). The monkeys were 2.5 to 8 years old and weighed between 2.5 and 4.0 kg. Eight groups of five randomly assigned male cynomolgus monkeys each were injected subcutaneously with ISIS oligonucleotide using a stainless steel dosing needle and syringe of appropriate size into any of six dosing sites, which were used on a rotational basis. These sites were one site each on the lateral portion of each thigh, and four separate sites on the back. The monkeys were dosed once every other day at a dose of 40 mg/kg for the first week (days 1, 3, and 5) as loading doses, and subsequently twice a week at a maintenance dose of 20 mg/kg (40 mg/kg/week) for weeks 2-13, with ISIS 299005, ISIS 463588, ISIS 463589, ISIS 463690, ISIS 463691, ISIS 463835, ISIS 463837, or ISIS 464225. A control group of 8 cynomolgus monkeys was injected with PBS subcutaneously once every other day for the first week (days 1, 3, and 5), and subsequently twice a week for weeks 2-13.

During the study period, the monkeys were observed twice daily for a sign of illness or distress. Veterinary care was available throughout the course of the study and animals were examined by the veterinary staff, as warranted for clinical signs or other changes. At the end of the study period, the animals were euthanized under deep anesthesia induced by ketamine and Beuthanasia-D®, followed by exsanguination. All organs were collected within 10 minutes of exsanguinations.

RNA analysis

Total RNA was extracted from liver and kidney tissue for real-time PCR analysis and FGFR4 mRNA levels were measured using human primer probe set RTS3232 and the rhesus primer probe set rhFGFR4_LTS00467 (forward sequence TCATCAACGGCAGCAGCTT, designated herein as SEQ ID NO: 333; reverse sequence TGAGCTATTGATGTCTGCAGTCTTC, designated herein as SEQ ID NO: 334; probe sequence CCGACGGCTTCCCCTATGTGCA, designated herein as SEQ ID NO: 335). Results are presented as percent inhibition of FGFR4, relative to PBS control, normalized to Cyclophilin expression levels and/or directly with RIBOGREEN®. As shown in Tables 20 and 21, treatment with ISIS antisense oligonucleotides resulted in significant reduction of FGFR4 mRNA in comparison to the PBS control.

TABLE 20 % Inhibition of FGFR4 mRNA in the cynomolgus monkey liver relative to the PBS control RTS3232/ rhFGFR4_LTS00467/ rhFGFR4_LTS00467/ ISIS No RTS3232/Ribogreen Cyclophilm RIBOGREEN Cyclophilin 299005 42 33 41 32 463588 71 72 68 68 463589 40 38 44 43 463690 64 67 58 61 463691 47 65 41 61 463835 61 51 50 37 463837 39 34 38 29 464225 65 64 61 60

TABLE 21 % Inhibition of FGFR4 mRNA in the cynomolgus monkey kidney relative to the PBS control rhFGFR4_LTS00467/ ISIS No RTS3232/Ribogreen RIBOGREEN 299005 60 52 463588 86 85 463589 77 71 463690 76 68 463691 75 63 463835 61 52 463837 54 49 464225 87 83

FGF19 and Leptin Levels

FGF19 has been known to reduce adiposity and improve insulin sensitivity in transgenic mice (Fu, L. et al., Endocrinology. 145: 2594-2603, 2004). FGF19 is also characterized as a high affinity ligand for FGFR4 (Xie, M.-H. et al., Cytokine. 11: 729-735, 1999). Leptin is a hormone which has been found to be present at very high levels in obese individuals compared to normal-weight individuals (Considine, R. V. et al., N. Engl. J. Med. 334: 292-295, 1996).

FGF19 mRNA levels were measured in ileum tissue samples by RT-PCR analysis, using the primer probe set rhFGF19_LTS00681 (forward sequence CCCCATGTGGGAATTGATCT, designated herein as SEQ ID NO: 336; reverse sequence CATGCCTGCTTCAGTCAGTTCT, designated herein as SEQ ID NO: 337; probe sequence TTTGCCCTTCCCAAACCCCTCCA, designated herein as SEQ ID NO: 338). The results are presented in Table 22, expressed as percent expression over the PBS control. The data indicates that treatment with any of the ISIS oligonucleotides enhanced the expression of FGF19.

The plasma samples of monkeys treated with ISIS 299005, ISIS 463588, ISIS 463589, and ISIS 463690 were assessed for FGF19 levels. The plasma samples of monkeys treated with ISIS 463588 and ISIS 463690 were assessed for leptin levels. Plasma levels of FGF19 were measured pre-dose and on days 23, 65 and 89 using an ELISA assay kit (R&D Systems). Plasma levels of leptin measured pre-dose and on days 58 and 93 using an ELISA assay kit (Alpco). Results are presented in Tables 23 and 24. The data indicates that treatment with any of the ISIS oligonucleotides increased FGF19 plasma levels and decreases leptin levels. In particular, treatment with ISIS 463588 caused the highest increase in FGF19 plasma levels compared to the PBS control as well as to the other experimental plasma samples. Treatment with ISIS 463588 caused the most significant decrease in leptin levels compared to the PBS control.

TABLE 22 Ileum FGF19 mRNA levels in the cynomolgus monkey (% expression over the PBS control) % ISIS No expression 299005 688 463588 715 463589 545 463690 1032 463691 477 463835 445 463837 384 464225 370

TABLE 23 Plasma FGF19 levels in the cynomolgus monkey (pg/ml) Pre- dose Day 23 Day 65 Day 89 PBS 106 79 104 84 ISIS 299005 125 110 191 202 ISIS 463588 192 146 309 401 ISIS 463589 111 117 177 151 ISIS 463690 184 154 287 266

TABLE 24 Plasma leptin levels in the cynomolgus monkey (ng/ml) Pre- dose Day 58 Day 93 PBS 0.21 0.60 0.53 ISIS 463588 0.15 0.23 0.26 ISIS 463690 0.27 0.32 0.36

Tolerability Studies Liver Function

To evaluate the effect of ISIS oligonucleotides on hepatic function, blood samples were collected from all the study groups. The blood samples were collected via femoral venipuncture on day 58, 48 hrs post-dosing and processed for serum. Concentrations of various metabolites were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). Plasma concentrations of ALT and AST were measured and the results are presented in Table 25, expressed in IU/L. Bilirubin is also a liver function marker, was similarly measured and is presented in Table 25, expressed in mg/dL. The results indicate that treatment with ISIS 463588, as well as several other ISIS oligonucleotides, was well tolerated in terms of the liver function of the monkeys.

TABLE 25 ALT, AST, and Bilirubin in cynomolgus monkey plasma (on day 58) ALT AST Bilirubin (IU/L) (IU/L) (mg/dL) PBS 47.4 40.3 0.2 ISIS 299005 45.2 37.4 0.2 ISIS 463588 77.6 73.2 0.1 ISIS 463589 33.8 29.8 0.2 ISIS 463690 103.6 47.6 0.2 ISIS 463691 76.2 72.4 1.8 ISIS 463835 116.2 42.0 0.1 ISIS 463837 121.0 43.2 0.1 ISIS 464225 81.4 40.8 0.1

Kidney Function

To evaluate the effect of ISIS oligonucleotides on kidney function, blood samples were collected from all the study groups. The blood samples were collected via femoral venipuncture on day 58, 48 hrs post-dosing and processed for serum. Concentrations of BUN and creatinine were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). Results are presented in Table 26, expressed in mg/dL.

The results indicate that most of the ISIS oligonucleotides did not have any adverse effects on the kidney function. Specifically, treatment with ISIS 463588 was well tolerated in terms of the kidney function of the monkeys.

TABLE 26 Plasma BUN and creatinine levels (mg/dL) in cynomolgus monkeys on day 58 BUN Creatinine PBS 28.6 0.8 ISIS 299005 24.3 0.7 ISIS 463588 23.2 0.7 ISIS 463589 28.7 0.8 ISIS 463690 22.7 0.7 ISIS 463691 16.8 0.5 ISIS 463835 32.2 0.8 ISIS 463837 26.7 0.7 ISIS 464225 25.8 0.6

Analysis of Markers of Inflammation

To evaluate the effect of ISIS oligonucleotides on factors involved in inflammation, blood was collected from all available animals for C-reactive protein (CRP) and complement C3 analysis, as well as for measurement of cytokine and chemokine levels. The blood samples were collected via femoral venipuncture on day 93, 48 hrs post-dosing and processed for separately for serum and plasma. Serum CRP and plasma complement C3 was measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). The data is presented in Tables 27 and 28, expressed in mg/dL.

For cytokine level analyses, blood (1 mL each) was collected and then centrifuged 3,000 rpm for 10 min at 2-8° C. Plasma samples of mice treated with ISIS 463588, ISIS 463589, and ISIS 463690 were sent to Aushon Biosystems Inc. (Billerica, Mass.) for measurement of chemokine and cytokine levels. Levels of IL-6, MIP-1α, IL-8, MIP-1β, MCP-1, IL-1β, and RANTES were measured using the respective cross-reacting human antibodies and IFN-γ and IL-1β were measured using the respective primate antibodies. Measurements were taken pre-dose and on day 93. The results are presented in Tables 29-36.

The data indicate that most of the ISIS oligonucleotides were not pro-inflammatory. Specifically, treatment with ISIS 463588 was well tolerated in terms of being non-pro-inflammatory in the monkeys since there were no changes in CRP, a marker of inflammation.

TABLE 27 CRP (mg/dL) in cynomolgus monkeys Pre-dose Day 30 Day 58 Day 93 PBS 2.2 3.1 2.5 4.1 ISIS 299005 1.0 1.5 1.2 1.2 ISIS 463588 2.9 5.2 3.7 3.8 ISIS 463589 1.8 1.9 2.2 2.4 ISIS 463690 2.2 3.1 2.1 3.6 ISIS 463691 6.3 5.2 10.3 2.6 ISIS 463835 9.7 16.2 4.7 5.7 ISIS 463837 2.5 11.4 2.8 2.9 ISIS 464225 2.5 8.1 6.9 5.2

TABLE 28 Complement C3 (mg/dL) in cynomolgus monkeys Pre-dose Day 30 Day 58 Day 93 PBS 114.3 109.1 112.5 113.3 ISIS 299005 108.3 92.1 99.6 91.4 ISIS 463588 106.7 91.9 94.9 95.7 ISIS 463589 116.3 102.0 105.1 100.9 ISIS 463690 113.3 89.4 85.6 78.7 ISIS 463691 123.5 89.2 70.6 97.6 ISIS 463835 105.5 66.2 66.5 69.0 ISIS 463837 107.1 91.1 88.7 86.5 ISIS 464225 104.7 91.9 92.7 80.1

TABLE 29 IL-6 (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 1.0 1.0 ISIS 463588 0.4 0.9 ISIS 463589 0.5 2.8 ISIS 463690 1.2 10.2

TABLE 30 IL-8 (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 544 482 ISIS 463588 1255 1159 ISIS 463589 424 636 ISIS 463690 719 1344

TABLE 31 MIP-1α (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 7.6 8.9 ISIS 463588 8.9 10.8 ISIS 463589 7.9 11.2 ISIS 463690 13.8 18.9

TABLE 32 MIP-1β (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 249 229 ISIS 463588 219 211 ISIS 463589 175 196 ISIS 463690 362 478

TABLE 33 MCP-1 (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 200 275 ISIS 463588 420 496 ISIS 463589 343 363 ISIS 463690 441 709

TABLE 34 IFN-γ (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 22.1 25.9 ISIS 463588 1.6 1.5 ISIS 463589 10.8 12.3 ISIS 463690 20.8 17.4

TABLE 35 IL-1β (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 0.09 0.28 ISIS 463588 0.07 0.08 ISIS 463589 0.13 0.06 ISIS 463690 0.20 0.36

TABLE 36 RANTES (pg/mL) in cynomolgus monkeys Pre-dose Day 93 PBS 43339 48967 ISIS 463588 45962 51326 ISIS 463589 38382 30985 ISIS 463690 37330 29209

Hematology

To evaluate any effect of ISIS oligonucleotides in cynomolgus monkeys on hematologic parameters, approximately 1.3 mL of blood was collected on day 93 from each of the available study animals in tubes containing K2-EDTA. Samples were analyzed for red blood cell (RBC) count, white blood cells (WBC) count, individual white blood cell counts, such as that of monocytes, neutrophils, lymphocytes, as well as for platelet count, hemoglobin content and hematocrit, using an ADVIA120 hematology analyzer (Bayer, USA). The data is presented in Tables 37 and 38.

The data indicate that most of the ISIS oligonucleotides did not have any adverse effects on the any hematologic parameters. Specifically, treatment with ISIS 463588 was well tolerated in terms of the hematologic parameters of the monkeys.

TABLE 37 Blood cells in cynomolgus monkeys RBC Platelets WBC Neutro- Lympho- Mono- (×106/ (×103/ (×103/ phils cytes cytes μL) μL) μL) (/μL) (/μL) (/μL) PBS 6.1 426 13.8 3244 9637 483 ISIS 299005 6.2 348 13.8 3395 9378 549 ISIS 463588 6.4 331 11.7 3081 7741 387 ISIS 463589 5.7 360 12.3 3590 8037 413 ISIS 463690 6.1 430 13.1 2592 9451 571 ISIS 463691 5.3 494 17.5 7511 8534 1144 ISIS 463835 5.5 558 12.7 3129 8374 664 ISIS 463837 5.8 480 13.3 3145 9025 566 ISIS 464225 5.9 429 13.6 2994 9349 762

TABLE 38 Hematologic parameters in cynomolgus monkeys Hemoglobin HCT (g/dL) (%) PBS 14.2 45.5 ISIS 299005 13.7 44.0 ISIS 463588 13.9 45.4 ISIS 463589 13.3 41.9 ISIS 463690 13.8 44.7 ISIS 463691 12.7 40.8 ISIS 463835 12.3 40.0 ISIS 463837 12.8 41.7 ISIS 464225 13.1 42.8

Pharmacokinetic Studies Measurement of Oligonucleotide Concentration

The concentration of the full-length oligonucleotide as well as the total oligonucleotide concentration (including the degraded form) was measured. The method used is a modification of previously published methods (Leeds et al., 1996; Geary et al., 1999) which consist of a phenol-chloroform (liquid-liquid) extraction followed by a solid phase extraction. An internal standard (ISIS 355868, a 27-mer 2′-O-methoxyethyl modified phosphorothioate oligonucleotide, GCGTTTGCTCTTCTTCTTGCGTTTTTT, designated herein as SEQ ID NO: 323) was added prior to extraction. Tissue sample concentrations were calculated using calibration curves, with a lower limit of quantitation (LLOQ) of approximately 1.14 μg/g. The results are presented in Table 39, expressed as μg/g tissue. The ratio of the concentrations in the kidney versus the liver was calculated and presented in Table 39. Treatment with ISIS oligonucleotides did not result in any abnormality in the ratio.

TABLE 39 Full-length oligonucleotide concentration (μg/g) in the liver and kidney of cynomolgus monkey ISIS No Kidney Liver Kidney/Liver 463588 1717 1033 1.7 463589 1663 1227 1.4 463690 1395 1226 1.1

Overall, the results of the study indicate that ISIS 463588 is a potent and tolerable antisense oligonucleotide for treatment of metabolic diseases, such as diabetes, obesity, insulin resistance, and insulin deficiency.

Example 16 In Vivo Effect of Antisense Inhibition of Murine FGFR4 in Diet-Induced Obesity (DIO) Mice with Caloric Restriction

DIO mice are C57BL/6 mice fed a high fat diet starting from 6 weeks of age and are a standard model used for assays related to studying the effect of therapeutic agents on lowering adiposity and improving insulin sensitivity. The antisense oligonucleotide, ISIS 393250, a 5-10-5 MOE gapmer, having a sequence of 5′-GCCACATTTCCTTCCAGCTG-3 (SEQ ID NO: 324), and with a target start site of 337 on murine FGFR4 mRNA (GENBANK Accession No. BC033313.1 (SEQ ID NO: 6) was used in this assay. The effect of ISIS 393250 on a DIO model under caloric restriction was evaluated.

Treatment

Male 6 week-old C57BL/6 mice (Jackson Laboratories) were fed with 58 kcal % high-fat diet (Research diet D12330) ad lib for 4 months to induce obesity. The mice were divided into 4 groups based on body weight and body fat content. The first group of mice was treated with 25 mg/kg ISIS 393250 administered subcutaneously twice weekly for 6 weeks. The second group of mice was treated with 25 mg/kg control oligonucleotide, ISIS 141923 (CCTTCCCTGAAGGTTCCTCC (SEQ ID NO: 325), 5-10-5 MOE gapmer with no known murine target), administered subcutaneously twice weekly for 6 weeks. Two control groups of mice were treated with PBS administered subcutaneously twice weekly for 6 weeks. After two weeks of treatment, the oligonucleotide-treated mice and one of the PBS control group mice were subjected to caloric restriction by providing 95% of the amount of food consumed daily by the FGFR4 ASO-treated mice during the first two weeks of treatment. The second PBS control group continued to be fed ad libitum with the same amount of food as in the first two weeks of treatment.

Weekly body weights were measured and body compositions were monitored at different time point with an Echo MRI Body Composition Analyzer. The mice were euthanized after 6 weeks of treatment.

RNA Analysis

RNA was extracted from the liver for RT-PCR analysis of murine FGFR4 expression. The primer probe set mFGFR4_LTS00702 (forward sequence CCCTGAGGCCAGATACACAGATAT, designated herein as SEQ ID NO: 339; reverse sequence ACGGATGACTTGCCGATGATA, designated herein as SEQ ID NO: 340; probe sequence CTCACTGGTTCTGCTTGTGCTCCTGCT, designated herein as SEQ ID NO: 341) was used for analysis. The results indicated that treatment with ISIS 393250 reduced murine FGFR4 levels by 76%.

Body Weight and Body Composition Analysis

Weekly body weights were measured and are presented in Table 40. Body fat content data is presented in Table 41, expressed as percent of the corresponding body weight. Lean body mass is presented in Table 42, expressed in grams. White adipose tissue weight was measured after euthanizing the mice and is presented in Table 43, expressed in grams. The data indicates calorie restriction significantly lowered body weight and total body fat content. Treatment with ISIS 393250 further lowered both body weight and fat content, but had no effect on body lean mass. Treatment with ISIS 141923 had no effect. Hence, antisense inhibition of FGFR4 expression has a beneficial effect on body weight and body fat content in subjects suffering from obesity.

TABLE 40 Weekly body weights (g) Calorie- restrict- Pre- Week Week Week Week Week Week ed dose 1 2 3 4 5 6 PBS No 51.7 51.1 52.1 52.6 53.1 50.6 53.4 PBS Yes 50.3 50.4 51.1 50.0 47.6 46.0 45.4 ISIS 141923 Yes 49.5 50.1 50.9 49.8 48.1 46.3 45.2 ISIS 393250 Yes 50.5 50.9 50.9 48.9 46.7 44.4 42.1

TABLE 41 Body fat content (% body weight) Calorie- restricted Pre-dose Week 2 Week 4 Week 6 PBS No 39.5 39.8 40 40 PBS Yes 38.7 40.2 39.1 38.3 ISIS 141923 Yes 38.9 39.1 38.1 37.7 ISIS 393250 Yes 39.4 38.6 35.3 31.3

TABLE 42 Lean body mass (g) Calorie- restricted Pre-dose Week 2 Week 4 Week 6 PBS No 27.8 28.2 28.2 28.3 PBS Yes 27.3 27.5 25.7 24.5 ISIS 141923 Yes 26.7 27.3 26 24.9 ISIS 393250 Yes 27.1 27.8 25.9 24.8

TABLE 43 White adipose tissue weight (g) Calorie- restricted Epididymal Peri-renal PBS No 2.5 1.1 PBS Yes 2.2 0.9 ISIS 141923 Yes 2.2 0.9 ISIS 393250 Yes 1.8 0.7

Metabolic Rate and Locomotor Activity Analysis

The metabolic rate was assessed by measuring the oxygen consumption and heat production of the mice. Both parameters were measured with an indirect calorimetry system (Oxymax system, Columbus Instruments). Locomotor activity was also assessed with the same instrument. Metabolic rate and locomotor activity was assessed both in darkness, when the mice are typically more active, and in light. The results are presented in Tables 44-46. The results indicate that calorie restriction reduced whole body oxygen consumption. Treatment with ISIS 393250 prevented this decrease in oxygen consumption without affecting locomotor activity. Hence, antisense inhibition of FGFR4 expression in obese subjects with a calorie-restricted diet would be beneficial as it would prevent any decline in metabolic rate in the subject.

TABLE 44 O2 consumption (mL/kg lean tissue/hr) Calorie- restricted dark light PBS No 4275 3327 PBS Yes 4085 3259 ISIS 141923 Yes 4094 3258 ISIS 393250 Yes 4268 3359

TABLE 45 Heat production (kcal/kg lean tissue/hr) Calorie- restricted dark light PBS No 19.8 15 PBS Yes 19.1 15.1 ISIS 141923 Yes 19.1 15.1 ISIS 393250 Yes 19.7 15.7

TABLE 46 Locomotor activity (events/min) Calorie- restricted dark light PBS No 16.4 1.9 PBS Yes 21.9 3.1 ISIS 141923 Yes 16.1 2.5 ISIS 393250 Yes 16.2 3

Example 17 In Vivo Effect of Antisense Inhibition of Murine FGFR4 in Diet-Induced Obesity (DIO) Mice with Caloric Restriction

The effect of ISIS 446259 (TCCATTTCCTCAGAGGCCTC (SEQ ID NO: 326), 5-10-5 MOE gapmer, with a target start site of 407 on GENBANK Accession No. BC033313.1 (SEQ ID NO: 6)) on DIO mice under caloric restriction was evaluated.

Treatment

Male 6 week-old C57BL/6 mice (Jackson Laboratories) were fed with 58 kcal % high-fat diet Research diet D12330) ad lib for 3.5 months to induce obesity. The mice were divided into 4 groups based on body weight and body fat content. The first group of mice was treated with 25 mg/kg ISIS 446259 administered subcutaneously twice weekly for 8 weeks. The second group of mice was treated with 25 mg/kg control oligonucleotide, ISIS 141923 administered subcutaneously twice weekly for 8 weeks. Two control groups of mice were treated with PBS administered subcutaneously twice weekly for 8 weeks. After two weeks of treatment, the oligonucleotide-treated mice and one of the PBS control group mice were subjected to caloric restriction by providing 90% of the amount of food consumed daily by the FGFR4 ASO-treated mice during the first two weeks of treatment. The second PBS control group continued to be fed ad libitum with the same amount of food as in the first two weeks of treatment.

Weekly body weights were measured and body compositions were monitored at different time point with an Echo MRI Body Composition Analyzer. The mice were euthanized after 8 weeks of treatment.

RNA Analysis

RNA was extracted from the liver for RT-PCR analysis of murine FGFR4 expression. The primer probe set mFGFR4 LTS00702 was used to analyze mRNA levels. The results indicated that treatment with ISIS 446259 reduced murine FGFR4 levels by 83%.

Body Weight and Body Composition Analysis

Weekly body weights were measured and are presented in Table 47. Body fat content data is presented in Table 48, expressed as percent of the corresponding body weight. Lean body mass was presented in Table 49, expressed in grams. The data indicates calorie restriction significantly lowered body weight and total body fat content. Treatment with ISIS 446259 further lowered both body weight and fat content, but had no effect on body lean mass. Treatment with ISIS 141923 had no further effect. Hence, antisense inhibition of FGFR4 expression has a beneficial effect on body weight and body fat content in subjects suffering from obesity in addition to effects seen by caloric restriction alone.

TABLE 47 Weekly body weights (g) Calorie- Week Week Week Week Week restricted 0 2 4 6 8 PBS No 48.7 49.9 51 53.4 52.5 PBS Yes 49.7 50.7 46.9 46.9 46.2 ISIS 141923 Yes 49.6 50.5 46.9 46.1 44.5 ISIS 446259 Yes 49.4 49.4 45.2 43.8 39.7

TABLE 48 Body fat content (% body weight) Calorie- restricted Week 0 Week 2 Week 5 Week 8 PBS No 41 43 43 42 PBS Yes 41 43 41 39 ISIS 141923 Yes 40 40 37 35 ISIS 446259 Yes 41 41 35 30

TABLE 49 Lean body mass (g) Calorie- restricted Week 0 Week 2 Week 5 Week 8 PBS No 26 24 26 27 PBS Yes 26 25 25 25 ISIS 141923 Yes 27 26 25 26 ISIS 446259 Yes 26 25 25 25

Plasma Lipid Analysis

To evaluate the effect of ISIS oligonucleotides on cholesterol and triglyceride metabolism, plasma levels of each were measured at the end of the treatment period. The mice were euthanized and blood was collected via cardiac puncture. The lipid levels were measured using an automated clinical chemistry analyzer (Hitachi Olympus AU400e, Melville, N.Y.). Results are presented in Table 50, expressed as mg/dL. The results indicate that treatment with ISIS 446259 reduced both cholesterol and triglyceride levels in the mice. Therefore, antisense inhibition of FGFR4 had a beneficial effect on the lipid profile and may be used to reduce adiposity in obese subjects.

TABLE 50 Cholesterol and lipid levels (mg/dL) Calorie- restricted Cholesterol Triglycerides PBS No 270 137 PBS Yes 240 128 ISIS 141923 Yes 222 113 ISIS 446259 Yes 181 84

Example 18 In Vivo Effect of Antisense Inhibition of Murine FGFR4 on FGF15 Levels in DIO Mice

The effect of ISIS 393250 and ISIS 446259 on FGF15 levels in DIO mice was evaluated.

Treatment

Male 6 week-old C57BL/6 mice (Jackson Laboratories) were fed with 58 kcal % high-fat diet Research diet D12330) ad lib for 3.5 months to induce obesity. A group of C57BL/6 mice were fed normal Purina mouse chow and served as the naïve control. The DIO mice were divided into groups based on body weight and body fat content. The first group of DIO mice was treated with 25 mg/kg ISIS 393250 administered subcutaneously twice weekly for 4 weeks. The second group of DIO mice was treated with 25 mg/kg ISIS 446259 administered subcutaneously twice weekly for 4 weeks. The third group of DIO mice was treated with 25 mg/kg control oligonucleotide, ISIS 141923 administered subcutaneously twice weekly for 4 weeks. A control group of DIO mice was treated with PBS administered subcutaneously twice weekly for 4 weeks. The mice were euthanized after 4 weeks of treatment.

FGF15 Levels

FGF15 is the rodent equivalent of FGF19 (Wright, T. J. et al., Dev. Biol. 269: 264-275, 2004), and is therefore important for the reduction of adiposity and improvement of insulin sensitivity in mice.

RNA was extracted from liver and ileum. Liver RNA was analyzed by RT-PCR analysis for FGFR4 mRNA levels using primer probe set mFGFR4_LTS00702. Ileum RNA was analyzed by RT-PCR analysis for FGF15 levels using primer probe set mFgf15_LTS00635 (forward sequence GACCAAAACGAACGAAATTTGTT, designated herein as SEQ ID NO: 342; reverse sequence ACGTCCTTGATGGCAATCG, designated herein as SEQ ID NO: 343; probe sequence AATTCCGCGCGGTCGCTCTG, designated herein as SEQ ID NO: 344). The results are presented in Table 51 and demonstrate that treatment with either antisense oligonucleotide significantly decreases FGFR4 mRNA levels and also significantly enhances FGF15 expression levels.

Plasma samples of the mice group were also analyzed at weeks 2 and 4 for FGF15 protein levels with ELISA using an anti-FGF15 antibody (Santa Cruz Biotechnology Inc). The results are presented in Table 52 and demonstrate that antisense inhibition of FGFR4 results in enhanced plasma levels of FGF15.

TABLE 51 FGFR4 and FGF15 mRNA levels relative to control Liver Ileum FGFR4 (% FGF15 (% ISIS No inhibition) expression) 141923 14 92 393250 96 1117 446259 94 707 C57BL/6 control 0 25

TABLE 52 FGF15 plasma levels at week 2 and 4 (ng/ml) Week 2 Week 4 PBS 0.13 0.13 ISIS 141923 0.12 0.14 ISIS 393250 0.69 0.96 ISIS 446259 0.18 0.25 C57BL/6 control 0.1 0.12

Example 19 In Vivo Effect of Antisense Inhibition of Murine FGFR4 on FGF15 Levels in C57BL/6 Mice

The effect of ISIS 393250 on FGF15 levels in C57BL/6 mice was evaluated.

Treatment

Male 6 week-old C57BL/6 mice (Jackson Laboratories) were fed normal Purina mouse chow. The mice were randomly divided into 3 groups. The first group of mice was treated with 50 mg/kg ISIS 393250 administered subcutaneously twice weekly for 5.5 weeks. The second group of mice was treated with 50 mg/kg control oligonucleotide, ISIS 141923 administered subcutaneously twice weekly for 5.5 weeks. A control group of mice was treated with PBS administered subcutaneously twice weekly for 5.5 weeks.

FGFR4 Levels

RNA was extracted from liver and RNA was analyzed by RT-PCR analysis for FGFR4 mRNA levels using primer probe set mFGFR4_LTS00702. The results are presented in Table 53 and demonstrate that treatment with ISIS 393250 significantly decreases FGFR4 mRNA levels

TABLE 53 FGFR4 mRNA inhibition levels (%) ISIS No % 141923 0 393250 79

FGF15 Levels

Plasma samples of the mice group were analyzed for FGF15 protein levels using with ELISA using an anti-FGF15 antibody (Santa Cruz Biotechnology Inc). The results are presented in Table 54 and demonstrate that antisense inhibition of FGFR4 results in enhanced plasma levels of FGF15.

TABLE 54 FGF15 plasma levels at day 16 ng/mL PBS 0.07 ISIS 141923 0.08 ISIS 393250 0.28

Example 20 In Vivo Effect of Antisense Inhibition of Murine FGFR4 on FGF15 Levels in Ob/Ob Mice

Leptin is a hormone produced by fat that regulates appetite. Deficiency of this hormone in both humans and in non-human animals, leads to obesity. ob/ob mice have a mutation in the leptin gene which results in obesity and hyperglycemia. As such, these mice are a useful model for the investigation of obesity and diabetes and related conditions provided herein. These mice models are also useful for testing compounds, compositions and methods designed to treat, prevent or ameliorate such conditions.

In accordance with the present invention, the effects of antisense inhibition of FGFR4 were investigated in the ob/ob mouse model of obesity. Male 12 week old ob/ob (C57Bl/6J-Lepob/Lepob) mice were purchased from Jackson Laboratories (Bar Harbor, Me.) and used for the current study.

Treatment

The mice were divided into groups based on body weight and body fat content. The first group of mice was treated with 25 mg/kg ISIS 393250 administered subcutaneously twice weekly for 14 weeks. The second group of mice was treated with 25 mg/kg control oligonucleotide, ISIS 141923 administered subcutaneously twice weekly for 14 weeks. A control group of mice was treated with PBS administered subcutaneously twice weekly for 14 weeks.

FGFR4 Levels

RNA was extracted from liver and RNA was analyzed by RT-PCR analysis for FGFR4 mRNA levels using primer probe set mFGFR4_LTS00702. The results are presented in Table 55 and demonstrate that treatment with ISIS 393250 significantly decreases FGFR4 mRNA levels

TABLE 55 FGFR4 mRNA inhibition levels (%) ISIS No % 141923 0 393250 89

FGF15 Levels

Plasma samples of the mice group were analyzed for FGF15 protein levels using with ELISA using an anti-FGF15 antibody (Santa Cruz Biotechnology Inc). The results are presented in Table 56 and demonstrate that antisense inhibition of FGFR4 results in enhanced plasma levels of FGF15.

TABLE 56 FGF15 plasma levels at week 4 and 8 (ng/mL) Week 4 Week 8 PBS 0.5 1.2 ISIS 141923 0.8 0.5 ISIS 393250 4.2 4.2

Example 21 Effect of Antisense Inhibition of Murine FGFR4 in Monkey Primary Hepatocytes

The effect of antisense inhibition of FGFR4 with ISIS 299004 on fatty acid oxidation in monkey hepatocytes was evaluated. AICAR was used as a positive control.

Treatment

Primary hepatocytes purchased from APL/Lovelace In Vitro Enterprises and cultured in William E medium. The cells were seeded at a density of 1 million cells per 25 ml flask. After 4-5 hrs of culture, the cells were treated with 30 nM of ISIS 299004 or 1000 μM AICAR for 18 hrs. A control set of cells was treated with PBS. FGFR4 levels were measured using the primer probe set cynoFGFR4_MGB_LTS00689 (forward sequence GCACCAGGGATGAGCTTGAC, designated herein as SEQ ID NO: 348; reverse sequence CCAAGTCTCCCACTTTCCAGTT, designated herein as SEQ ID NO: 349; probe sequence AAGAGCCTGACTCCAGT, designated herein as SEQ ID NO: 350). Treatment with ISIS 299004 reduced FGFR4 levels by 83%.

For evaluation of fatty acid oxidation, the cells were placed in low glucose media containing 1-14cOleic acid and BSA, and the culture flasks were capped with a rubber stopper containing a hanging reservoir bucket. The cells were then incubated at 37° C. under 5% CO2 for 1.5 hrs. Following incubation, 200 μl of 1M hyamine hydroxide (a 14CO2 trapping agent) was added to the reservoir bucket and 1 ml of 10% perchloric acid solution was added to the cells. The flasks were transferred to a 37° C. shaking incubator for 40 min. Upon completion of the incubation, the hanging bucket reservoir containing the hyamine hydroxide was separated from the flask and placed in scintillation fluid overnight, and read in the scintillation counter the next day. Bradford-based protein measurements were conducted on an equal number of primary monkey hepatocytes, by using the DC™ Biorad protein assay kit (Bearden, J. Biochem. Biophys. Acta. 533: 525. 1978). The values obtained from the protein readout was used for normalization of the CO2 production counted by the scintillation counter. The results are presented in Table 57 and indicate that antisense inhibition of FGFR4 increased fatty acid oxidation in primary hepatocytes. Five independent fatty acid oxidation experiments were conducted, which demonstrated a similar trend on the results.

TABLE 57 CO2 production (% of the control) CO2 ISIS 141923 +2 ISIS 299004 +48 AICAR +44

Claims

1. A compound comprising a modified oligonucleotide consisting of 12 to 30 linked nucleosides, wherein the linked nucleosides comprise at least 8 contiguous nucleobases of a sequence recited in SEQ ID NOs: 17, 45, 46, 70, 72, or 138.

2-7. (canceled)

8. The compound of claim 1 wherein the modified oligonucleotide is a single-stranded oligonucleotide.

9. (canceled)

10. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is at least 95% complementary to SEQ ID NO 1 or 2.

11. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is 100% complementary to SEQ ID NO 1 or 2.

12. The compound of claim 1 wherein at least one internucleoside linkage is a modified internucleoside linkage.

13. The compound of claim 12, wherein each internucleoside linkage is a phosphorothioate internucleoside linkage.

14. The compound of claim 1, wherein at least one nucleoside of the modified oligonucleotide comprises a modified sugar.

15. The compound of claim 14, wherein the at least one modified sugar is a bicyclic sugar.

16. The compound of claim 15, wherein each of the at least one bicyclic sugar comprises a 4′-CH2-N(R)—O-2′ bridge wherein R is, independently, H, C1-C12 alkyl, or a protecting group.

17. The compound of claim 15, wherein each of the at least one bicyclic sugar comprises a 4′-CH(CH3)-O-2′ bridge.

18. The compound of claim 14, wherein at least one modified sugar comprises a 2′-O-methoxyethyl group.

19. (canceled)

20. (canceled)

21. The compound of claim 1, wherein at least one nucleoside comprises a modified nucleobase.

22. The compound of claim 21, wherein the modified nucleobase is a 5-methylcytosine.

23. The compound of claim 1, wherein the modified oligonucleotide comprises:

a gap segment consisting of linked deoxynucleosides;
a 5′ wing segment consisting of linked nucleosides; and
a 3′ wing segment consisting of linked nucleosides;
wherein the gap segment is positioned between the 5′ wing segment and the 3′ wing segment and wherein each nucleoside of each wing segment comprises a modified sugar.

24. The compound of claim 23, wherein the modified oligonucleotide comprises:

a gap segment consisting of ten linked deoxynucleosides;
a 5′ wing segment consisting of five linked nucleosides; and
a 3′ wing segment consisting of five linked nucleosides;
wherein the gap segment is positioned between the 5′ wing segment and the 3′ wing segment, wherein each nucleoside of each wing segment comprises a 2′-O-methoxyethyl sugar; and wherein each internucleoside linkage is a phosphorothioate linkage.

25. (canceled)

26. (canceled)

27. The compound of claim 1, wherein the modified oligonucleotide consists of 20 linked nucleosides.

28. A composition comprising the compound of claim 1 or salt thereof and at least one of a pharmaceutically acceptable carrier or diluent.

29-46. (canceled)

47. A method for reducing or preventing a metabolic disease comprising administering to a human a therapeutically effective amount of a compound of claim 1, thereby reducing or preventing a metabolic disease.

48. The method of claim 47, wherein the metabolic disease is obesity.

49. The method of claim 47, comprising co-administering the compound or composition and a second agent.

50-91. (canceled)

Patent History
Publication number: 20150184164
Type: Application
Filed: Dec 4, 2014
Publication Date: Jul 2, 2015
Applicant: ISIS PHARMACEUTICALS, INC. (Carlsbad, CA)
Inventors: Sanjay Bhanot (Carlsbad, CA), Xing-Xian Yu (San Diego, CA), Michael L. McCaleb (La Jolla, CA)
Application Number: 14/561,080
Classifications
International Classification: C12N 15/113 (20060101); A61K 31/713 (20060101);