CN118786133A - Renin modulating compositions and methods of use thereof - Google Patents

Renin modulating compositions and methods of use thereof Download PDF

Info

Publication number
CN118786133A
CN118786133A CN202280066569.0A CN202280066569A CN118786133A CN 118786133 A CN118786133 A CN 118786133A CN 202280066569 A CN202280066569 A CN 202280066569A CN 118786133 A CN118786133 A CN 118786133A
Authority
CN
China
Prior art keywords
compound
certain embodiments
modified oligonucleotide
disease
modified
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280066569.0A
Other languages
Chinese (zh)
Inventor
朱瑞
周智清
金伯利·富尔茨
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Adalx Pharmaceutical Co ltd
Original Assignee
Adalx Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Adalx Pharmaceutical Co ltd filed Critical Adalx Pharmaceutical Co ltd
Priority claimed from PCT/US2022/077389 external-priority patent/WO2023056446A1/en
Publication of CN118786133A publication Critical patent/CN118786133A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Some aspects of the present disclosure provide compounds, compositions, and methods for modulating the expression or activity of Angiotensinogen (AGT). In some aspects, the compounds, compositions and methods of the present disclosure can be used to reduce the expression of AGT mRNA in a cell or animal. In some aspects, the compounds, compositions and methods of the present disclosure can be used to reduce the expression of AGT proteins in cells or animals.

Description

Renin modulating compositions and methods of use thereof
Cross Reference to Related Applications
The present application claims the benefit of U.S. provisional application No.63/251,562 filed on 1-10-2021 and U.S. provisional application No.63/287,960 filed on 9-12-2021. The disclosure of each of the prior applications is considered to be part of the disclosure of the present application and is incorporated by reference in its entirety.
Background
Renin (angiotensinogen, AGT) (also known as SERPINA 8) is a member of the serine protease inhibitor (serpin) family. The encoded protein is an angiotensinogen precursor, which is expressed in large quantities in the liver and cleaved by the enzyme renin in response to reduced blood pressure. The product (angiotensin I) is then cleaved by angiotensin converting enzyme (angiotensin converting enzyme, ACE) to yield physiologically active angiotensin II. Angiotensin II is an active peptide of the renin-angiotensin-aldosterone system (renin-angeotenin-aldosterone system, RAAS). Angiotensin II interacts with receptors to mediate vasoconstriction, thirst, release of vasopressin and aldosterone, reabsorption of renal sodium, fibrosis, inflammation, angiogenesis, vascular aging and atherosclerosis. The release of aldosterone results in increased reabsorption of sodium and water by the kidneys, resulting in an increase in fluid volume in the body, which in turn can result in an increase in blood pressure. Thus, overstimulation or activity of the RAAS pathway can lead to hypertension (high blood pressure). High levels of angiotensin II are associated with chronic hypertension (systemic arterial hypertension, primary hypertension or hypertension), renal failure and cardiac fibrosis.
Hypertension is the most common risk factor for cardiovascular disease (CVD (cardiovascular disease); including coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation and peripheral arterial disease), chronic kidney disease (chronic KIDNEY DISEASE, CKD) and cognitive impairment, and is the major single contributor to death and disability worldwide (Forouzanfar et al., lancet,2016, 388:1659-1724). World health organization (World Health Organization) estimated that 12.8 million adults aged 30 to 79 years had hypertension worldwide. Less than half of this population is diagnosed and treated, and only about 20% of them are able to control their hypertension through medication, diet and lifestyle changes.
The american heart Association (AMERICAN HEART Association) defines refractory hypertension as uncontrolled Blood Pressure (BP) 130/80mmHg, although 3 antihypertensive drug types are used simultaneously, including calcium channel blockers, renin-angiotensin system blockers and thiazide diuretics, preferably chlorthalidone. Refractory hypertension can also be defined as treatment with ≡4 class antihypertensive drugs, regardless of BP. The global prevalence of refractory hypertension in the treated population is estimated to be about 14.7%. Current approved therapies for the treatment of hypertension have significant limitations. Drugs (e.g., ACE inhibitors and angiotensin receptor blockers) are the primary treatment for hypertension. Such drugs have limited ability to inhibit the RAAS pathway and have considerable adverse effects and contraindications in certain patient populations (Momoniat et al, CLEVELAND CLINIC Journal of Medicine,2019, 86:601-607).
Factors such as age and obesity predispose individuals to risk for refractory hypertension. In the case of aging and increasing overweight populations and lack of effective treatment, it is expected that the prevalence of hypertension, refractory hypertension and related diseases will continue to rise. Thus, there is a need to find an effective treatment for RAAS related diseases.
Summary of The Invention
The present disclosure provides compounds, compositions and methods for modulating the expression or activity of AGT. In certain embodiments, the compounds, compositions, and methods are useful for reducing the expression of AGT mRNA in a cell or animal. In certain embodiments, the compounds, compositions and methods are useful for reducing the amount of AGT protein in a cell or animal.
In certain embodiments, the animal has a RAAS-related disease, disorder, or condition. In certain embodiments, the disease, disorder or condition is hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. Certain compounds, compositions, and methods provided herein aim to alleviate RAAS-related diseases, disorders, or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in an animal. In certain embodiments, the compounds and compositions provided herein are highly potent and tolerable and inhibit AGT expression, which can be used to treat, prevent, ameliorate or slow the progression of: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
In certain embodiments, the compounds and compositions comprise one or more features effective for enhancing efficacy. In certain embodiments, the compounds and compositions comprise one or more features effective for improving tolerability. In certain embodiments, the compounds and compositions comprise one or more features effective for targeting the compounds or compositions to cells or tissues. In certain embodiments, the compounds and compositions are more potent, have a longer duration of action, or have a higher therapeutic value than the disclosed compounds.
Detailed Description
It is to be understood that both the foregoing summary and the following detailed description are exemplary and explanatory only and are not restrictive of the embodiments as claimed. 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 the present application, including but not limited to patents, patent applications, articles, books, treatises, and GenBank, NCBI, and other sequence reference records, are hereby expressly incorporated by reference herein for all portions of the documents discussed herein, and by the date of their filing date.
It will be appreciated that the sequence shown in each SEQ ID NO contained herein is independent of any modification of the sugar moiety, internucleoside linkage or nucleobase, even where shown in the context as a modified compound. Thus, the compounds defined by SEQ ID NOs may independently comprise one or more modifications to the sugar moiety, internucleoside linkage or nucleobase. The oligomeric compounds referred to by Compound Number (Compound Number) or Ref ID NO represent a combination of nucleobase sequences, chemical modifications and motifs.
As used herein, the singular includes the plural unless specifically stated otherwise. For example, a noun without a quantitative word modification as used herein refers to a grammatical object of one or more than one (i.e., at least one). For example, "an element" means one element or more than one element, such as a plurality of elements. As used herein, the use of "or" means "and/or" unless stated otherwise. Furthermore, the use of the term "include/comprise" and other forms (e.g., variations thereof) is not limiting and may be used interchangeably with the phrase "including but not limited to.
Definition of the definition
Unless otherwise indicated, the following terms have the following meanings:
"angiotensinogen" is used interchangeably with the term "AGT" and refers to any nucleic acid or protein of AGT. Exemplary nucleotide and amino acid sequences for AGT can be found, for example, in GenBank accession No. NM-000029.4 (incorporated herein by SEQ ID NO: 1), the complement of nucleotides 230702523 to 230745583 of NC-000001.11 (incorporated herein by SEQ ID NO: 2), NM-001382817.3 (incorporated herein by SEQ ID NO: 3), and nucleotides 5469 to 17068 of NG-008836.2 (incorporated herein by SEQ ID NO: 4). Additional examples of AGT sequences are readily available through publicly available databases (e.g., genBank, uniProt and OMIM). Additional information about AGT can be found in, for example, ncbi.nlm.nih.gov/gene/? term = AGT. AGT as used herein also refers to variations of the AGT gene, including the variants provided in the SNP database. Many sequence variations in the AGT gene have been identified and can be found, for example, in NCBI dbSNP and UniProt (see, e.g., ncbi.nl.gov/snp/. "AGT mRNA" means mRNA encoding AGT protein. AGT may be represented in uppercase or lowercase letters.
By "AGT specific inhibitor" is meant any agent capable of specifically inhibiting AGT RNA and/or AGT protein expression or activity at the molecular level. For example, AGT specific inhibitors include nucleic acids (including oligonucleotide compounds), peptides, antibodies, small molecules, and other agents capable of inhibiting the expression of AGT RNA and/or AGT proteins.
"2 '-O-methoxyethyl" or "2' -MOE" means 2'-O (CH 2)2-OCH3 modification. The 2' -O-methoxyethyl modified sugar is a modified sugar with 2'-O (CH 2)2-OCH3) instead of the 2' -OH group of the ribosyl ring.
"5 'Initiation site" means the nucleotide of the target nucleic acid or region that is aligned with the most 3' -nucleoside of the antisense oligonucleotide.
"3 'Termination site" means a nucleotide of a target nucleic acid or region that is aligned with the most 5' -nucleoside of an antisense oligonucleotide.
"About/about" means within + -10% of the value. For example, if the expression "a compound achieves an AGT inhibition of about 70%, it means that the AGT level is inhibited in the range of 60% to 80%. When "about" exists before a series of numbers or ranges, it is to be understood that "about/about" can modify each number in the series or range.
"Administration" and variations thereof refer to the route by which a compound or composition provided herein is introduced into an individual to achieve its intended function. For example, routes of administration that may be used include, but are not limited to, parenteral administration, such as subcutaneous, intravenous, or intramuscular injection or infusion.
"Amelioration" refers to an improvement or alleviation of at least one indicator, sign or symptom of a related disease, disorder or condition. In certain embodiments, improving comprises delaying or slowing the progression or severity of one or more indicators of a condition or disease. The progress or severity of an indicator may be determined by subjective or objective measurements known to those skilled in the art.
By "animal" is meant 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 oligonucleotide" or "antisense strand" means an oligonucleotide comprising a region complementary to a target nucleic acid (e.g., AGT RNA or a region thereof).
"Complementarity" with respect to an oligonucleotide means that when two nucleobase sequences are aligned in opposite directions, the nucleobase sequence of such oligonucleotide or one or more regions thereof is complementary to the nucleobase sequence of another oligonucleotide or nucleic acid or one or more regions thereof. Unless otherwise indicated, complementary nucleobases described herein are limited to the following pairing: adenine (A) and thymine (T), adenine (A) and uracil (U), and cytosine (C) and guanine (G). The complementary oligonucleotides and/or nucleic acids need not have nucleobase complementarity at each nucleoside and may contain one or more nucleobase mismatches. In contrast, "fully complementary" or "100% complementary" with respect to an oligonucleotide means that such an oligonucleotide has a nucleobase match at each nucleoside without any nucleobase mismatch.
By "composition" or "pharmaceutical composition" is meant a mixture of substances suitable for administration to an individual. For example, the composition may comprise one or more compounds or salts thereof in sterile aqueous solutions.
By "co-administration" is meant administration of two or more compounds in any manner wherein the pharmacological effects of both compounds are manifested in the patient. Co-administration does not require that the two compounds be administered in a single pharmaceutical composition, in the same dosage form, by the same route of administration, or simultaneously. The effect of both compounds does not require that they be simultaneously revealed. The effects need only overlap over a period of time and need not be co-extensive. Co-administration includes parallel or sequential administration of one or more compounds.
"Conjugate group" means a radical attached to an oligonucleotide. The conjugate group is optionally attached to the oligonucleotide via a conjugate linker. For example, a conjugate group may alter the distribution, targeting, or half-life of the compound into which it is incorporated. The conjugate group includes a targeting moiety.
"Conjugate linker" means a radical comprising at least one bond linking a linking moiety to an oligonucleotide.
"Identity" with respect to an oligonucleotide means that the nucleobase sequence of such oligonucleotide or one or more regions thereof matches the nucleobase sequence of another oligonucleotide or nucleic acid or one or more regions thereof. The identity of one oligonucleotide to another oligonucleotide or nucleic acid does not require a match for every nucleobase, and may comprise one or more different nucleobases. In contrast, "identical" or "100% identity" with respect to an oligonucleotide means that such an oligonucleotide has the same nucleobase as the other oligonucleotide or nucleic acid at each relative position along its length.
By "individual" is meant a human or non-human animal selected for treatment or therapy.
By "inhibiting expression or activity" with respect to a target nucleic acid or protein is meant reducing or blocking the expression or activity of such target relative to the expression or activity in an untreated or control sample, and does not necessarily mean complete elimination of the expression or activity.
As used herein, the term "internucleoside linkage" is a covalent linkage between adjacent nucleosides in an oligonucleotide. As used herein, "modified internucleoside linkage" means any internucleoside linkage other than phosphodiester internucleoside linkage. "phosphorothioate internucleoside linkages" are modified internucleoside linkages in which one non-bridging oxygen atom of the phosphodiester internucleoside linkage is replaced by a sulfur atom.
Representative internucleoside linkages having chiral centers include, but are not limited to, alkyl phosphonates and phosphorothioates. Modified oligonucleotides comprising internucleoside linkages having a chiral center can be prepared as a population of modified oligonucleotides comprising stereorandom internucleoside linkages, or as a population of modified oligonucleotides comprising phosphorothioate linkages of a particular stereochemical configuration, as described further below. Unless otherwise indicated, the chiral internucleoside linkages of the modified oligonucleotides described herein may be of a stereorandom or specific stereochemical configuration.
The compounds of the present disclosure may also contain non-natural proportions of atomic isotopes at one or more of the atoms making up such compounds. For example, the compounds may be radiolabeled with a radioisotope, such as tritium (3 H), iodine-125 (125 I), or carbon-14 (14 C), for example. All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present disclosure.
The term "isotopic variant" refers to such therapeutic agents (e.g., compounds and/or modified oligonucleotides disclosed herein) comprising a non-natural proportion of isotopes at one or more atoms constituting the therapeutic agent. In certain embodiments, an "isotopic variant" of a therapeutic agent comprises a non-natural proportion of one or more isotopes, including but not limited to hydrogen (H), deuterium (2 H), tritium (3 H), carbon-11 (11 C), carbon-12 (12 C), and combinations thereof, Carbon-13 (13 C), carbon-14 (14 C), nitrogen-13 (13 N), nitrogen-14 (14 N), Nitrogen-15 (15 N), oxygen-14 (14 O), oxygen-15 (15 O), oxygen-16 (16 O), Oxygen-17 (17 O), oxygen-18 (18 O), fluorine-17 (17 F), fluorine-18 (18 F), Phosphorus-31 (31 P), phosphorus-32 (32 P), phosphorus-33 (33 P), sulfur-32 (32 S), sulfur-33 (33 S), sulfur-34 (34 S), sulfur-35 (35 S), sulfur-36 (36 S), Chlorine-35 (35 Cl), chlorine-36 (36 Cl), chlorine-37 (37 Cl), bromine-79 (79 Br), Bromine-81 (81 Br), iodine 123 (123 I), iodine-125 (125 I), iodine-127 (127 I), Iodine-129 (129 I) and iodine-131 (131 I). In certain embodiments, an "isotopic variant" of a therapeutic agent comprises a non-natural proportion of one or more isotopes, including but not limited to hydrogen (H), deuterium (2 H), tritium (3 H), carbon-11 (11 C), carbon-12 (12 C), and combinations thereof, Carbon-13 (13 C), carbon-14 (14 C), nitrogen-13 (13 N), nitrogen-14 (14 N), Nitrogen-15 (15 N), oxygen-14 (14 O), oxygen-15 (15 O), oxygen-16 (16 O), Oxygen-17 (17 O), oxygen-18 (18 O), fluorine-17 (17 F), fluorine-18 (18 F), Phosphorus-31 (31 P), phosphorus-32 (32 P), phosphorus-33 (33 P), sulfur-32 (32 S), sulfur-33 (33 S), sulfur-34 (34 S), sulfur-35 (35 S), sulfur-36 (36 S), Chlorine-35 (35 Cl), chlorine-36 (36 Cl), chlorine-37 (37 Cl), bromine-79 (79 Br), Bromine-81 (81 Br), iodine 123 (123 I), iodine-125 (125 I), iodine-127 (127 I), Iodine-129 (129 I) and iodine-131 (131 I).
It will be appreciated that in a therapeutic agent (e.g., a compound and/or modified oligonucleotide disclosed herein), any hydrogen may be 2 H, or any carbon may be 13 C, for example, or any nitrogen may be 15 N, for example, or any oxygen may be 18 O, for example, as determined by the skilled artisan to be viable. In certain embodiments, an "isotopic variant" of a therapeutic agent comprises a non-natural proportion of deuterium (D).
"Mismatched" or "non-complementary" means that when a first oligonucleotide/nucleic acid and a second oligonucleotide/nucleic acid are aligned in an antiparallel orientation, the nucleobase of the first oligonucleotide or nucleic acid is non-complementary to the corresponding nucleobase of the second oligonucleotide or nucleic acid. For example, nucleobases, including but not limited to universal nucleobases, inosine, and hypoxanthine, can hybridize to at least one nucleobase, but still be mismatched or non-complementary to the nucleobase to which it hybridizes. As another example, when a first oligonucleotide and a second oligonucleotide are aligned in an antiparallel orientation, the nucleobase of the first oligonucleotide/nucleic acid that is unable to hybridize to the corresponding nucleobase of the second oligonucleotide/nucleic acid is a mismatched or non-complementary nucleobase.
By "modified oligonucleotide" is meant an oligonucleotide in which at least one sugar, nucleobase, or internucleoside linkage is modified.
"Modulation" refers to altering or modulating a characteristic of a cell, tissue, organ or organism. For example, modulating AGT RNA can mean increasing or decreasing the level of AGT RNA and/or AGT protein in a cell, tissue, organ or organism. "modulators" affect changes in cells, tissues, organs or organisms. For example, the AGT compound may be a modulator that reduces the amount of AGT RNA and/or AGT protein in a cell, tissue, organ or organism.
"Motif" means the pattern of unmodified and modified sugar moieties, nucleobases and/or internucleoside linkages in an oligonucleotide.
"Nucleic acid" refers to a molecule composed of monomeric nucleotides. Nucleic acids include, but are not limited to, ribonucleic acid (RNA), deoxyribonucleic acid (deoxyribonucleic acid, DNA), single-stranded nucleic acids, and double-stranded nucleic acids.
"Nucleobase" means a heterocyclic moiety capable of base pairing with another nucleic acid. As used herein, "naturally occurring nucleobases" are adenine (A), thymine (T), cytosine (C), uracil (U) and guanine (G). A "modified nucleobase" is a chemically modified naturally occurring nucleobase. A "universal base" or "universal nucleobase" is a nucleobase other than a naturally occurring nucleobase and a modified nucleobase, and is capable of pairing with any nucleobase.
"Nucleobase sequence" means the order of successive nucleobases in a nucleic acid or oligonucleotide independent of any sugar or internucleoside linkage.
"Nucleoside" means a compound comprising a nucleobase and a sugar moiety. The nucleobase and sugar moiety are each independently unmodified or modified. "modified nucleoside" means a nucleoside comprising a modified nucleobase and/or a modified sugar moiety. Modified nucleosides include abasic nucleosides lacking nucleobases.
By "oligomeric compound" is meant a compound comprising one or more oligonucleotides and optionally one or more additional features (e.g., a conjugate group or a terminal group). Examples of oligomeric compounds include single and double stranded compounds, such as oligonucleotides, antisense oligonucleotides, interfering RNA compounds (INTERFERING RNA compounds), microRNA targeting oligonucleotides, placeholder-based compounds (e.g., mRNA processing or translation blocking compounds, and splicing compounds). RNAi compounds include double-stranded compounds (e.g., short-INTERFERING RNA, SIRNA and double-stranded RNA (double-STRANDED RNA, DSRNA)) and single-stranded compounds (e.g., single-stranded siRNA (single-STRANDED SIRNA, SSRNA), single-stranded RNAi (single-STRANDED RNAI, SSRNAI), short-hairpin RNA (short HAIRPIN RNA, SHRNA) and microRNA mimics) that act at least in part through the RNA-induced silencing complex (RNA-induced silencing complex, RISC) pathway, resulting in sequence-specific degradation and/or sequestration (sequencing) of a target nucleic acid via a process known as RNA interference (RNA INTERFERENCE, RNAI). The term "RNAi compound" is intended to be equivalent to other terms used to describe nucleic acid compounds capable of mediating sequence-specific RNA interference, such as Interfering RNAs (iRNA), iRNA agents, RNAi agents, short interfering oligonucleotides, short interfering nucleic acids, short interfering modified oligonucleotides, chemically modified siRNA, and the like. In addition, the term "RNAi" means other terms equivalent to those used to describe sequence-specific RNA interference.
"Oligonucleotide" means a polymer of linked nucleosides independent of each other, each of which may be modified or unmodified.
The term "oligomeric duplex" means a duplex formed from two oligomeric compounds having complementary nucleobase sequences. Each oligomeric compound of an oligomeric duplex may be referred to as a "double-stranded oligomeric compound. The oligonucleotides of each oligomeric compound of an oligomeric duplex may include non-complementary protruding nucleosides. In some embodiments, the terms "double stranded oligomeric compound" and "modified oligonucleotide" are used interchangeably. In other embodiments, the terms "oligomeric duplex" and "compound" are used interchangeably.
By "parenteral administration" is meant administration by injection or infusion. Parenteral administration includes subcutaneous, intravenous, intramuscular, intraarterial, intraperitoneal or intracranial administration, e.g., intrathecal or intraventricular administration.
By "pharmaceutically acceptable carrier or diluent" is meant any substance suitable for administration to an individual. In certain embodiments, a pharmaceutically acceptable carrier or diluent facilitates administration and absorption of the compound to and by an individual, and may be included in the compositions of the present disclosure without producing significant adverse toxicological effects to the patient. Non-limiting examples of pharmaceutically acceptable excipients include water, naCl, normal saline solutions, and the like. For example, the pharmaceutically acceptable carrier may be a sterile aqueous solution, such as PBS or water for injection. Those skilled in the art will recognize that other pharmaceutical excipients may be used in the present disclosure.
By "pharmaceutically acceptable salt" is meant a physiologically and pharmacologically acceptable salt of a compound (e.g., an oligomeric compound or oligonucleotide), i.e., a salt that retains the desired biological activity of the parent compound without producing an undesirable toxicological effect thereon. As used herein, a pharmaceutically acceptable salt is any salt of a compound provided herein that retains its biological properties and is non-toxic or otherwise desirable for pharmaceutical use. Pharmaceutically acceptable salts of the therapeutic agents disclosed herein include salts prepared with relatively non-toxic acids or bases, depending on the particular substituents found in the compounds or modified oligonucleotides described herein.
When the compounds of the present disclosure contain relatively acidic functional groups, base addition salts can be obtained by contacting such compounds in neutral form with a sufficient amount of the desired base (whether pure or in a suitable inert solvent).
When the compounds of the present disclosure contain relatively basic functional groups, acid addition salts can be obtained by contacting such compounds in neutral form with a sufficient amount of the desired acid (whether pure or in a suitable inert solvent).
Thus, the compounds of the present disclosure may be present as salts, for example with pharmaceutically acceptable acids. Such salts may be derived from a variety of organic and inorganic counter ions known in the art. Such salts include, but are not limited to: (1) Acid addition salts with organic or inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, sulfamic acid, acetic acid, trifluoroacetic acid, trichloroacetic acid, propionic acid, caproic acid, cyclopentylpropionic acid, glycolic acid, glutaric acid, pyruvic acid, lactic acid, malonic acid, succinic acid, sorbic acid, ascorbic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, picric acid, cinnamic acid, mandelic acid, phthalic acid, lauric acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphoric acid, 4-methylbicyclo [2.2.2] -oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, t-butylacetic acid, lauryl sulfuric acid, gluconic acid, benzoic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, cyclohexylsulfamic acid, quinic acid, and the like; or (2) a salt formed when an acidic proton present in the parent compound undergoes the following (a) or (b): (a) Substituted by metal ions, such as alkali metal ions, alkaline earth metal ions or aluminum ions, or alkali metal or alkaline earth metal hydroxides, such as sodium, potassium, calcium, magnesium, aluminum, lithium, zinc and barium hydroxides, ammonia; (b) coordinates with: organic bases, such as aliphatic, alicyclic, or aromatic organic amines, e.g., ammonia, methyl amine, dimethyl amine, diethyl amine, methyl pyridine, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, lysine, arginine, ornithine, choline, N' -dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethyl amine, N-methylglucamine piperazine, tris (hydroxymethyl) -aminomethane, tetramethylammonium hydroxide, and the like (see, e.g., berge et al, "Pharmaceutical Salts," Journal of Pharmaceutical Science,1977, 66, 1-19).
Pharmaceutically acceptable salts also include, by way of example only and not limitation, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like, and when the compounds contain basic functional groups, salts of non-toxic organic or inorganic acids such as hydrohalides, such as hydrochloride and hydrobromide, sulfate, phosphate, sulfamate, nitrate, acetate, trifluoroacetate, trichloroacetate, propionate, hexanoate, cyclopentylpropionate, glycolate, glutarate, pyruvate, lactate, malonate, succinate, sorbate, ascorbate, malate, maleate, fumarate, tartrate, citrate, benzoate, 3- (4-hydroxybenzoyl) benzoate, picrate, cinnamate, mandelate, phthalate, laurate, methanesulfonate (methanesulfonate) (methanesulfonate), ethanesulfonate, 1, 2-ethane-disulfonate, 2-hydroxyethanesulfonate, benzenesulfonate (benzenesulfonate) (benzenesulfonate (besylate)), 4-chlorobenzenesulfonate, 2-naphthalenesulfonate, 4-toluenesulfonate, camphorate, camphorsulfonate, 4-methylbicyclo [2.2.2] -oct-2-ene-1-carboxylate, glucoheptonate, 3-phenylpropionate, trimethylacetate, t-butyl, lauryl sulfate, gluconate, benzoate, hydronaphthalene hydrochloride, salicylate, cyclohexyl sulfamate, aminosulfonate, muconate, and the like. In some embodiments, the pharmaceutically acceptable salts of the compounds and modified oligonucleotides disclosed herein are sodium or potassium salts. In some embodiments, the pharmaceutically acceptable salts of the compounds and modified oligonucleotides disclosed herein are sodium salts.
The neutral form of the compound is preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of the compound may differ from the various salt forms in certain physical properties, such as solubility in polar solvents. In some embodiments, the compounds of the present disclosure include both basic and acidic functionalities that allow the compounds to be converted into base addition salts or acid addition salts. The neutral form of the compound may be regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of a compound differs from the various salt forms in certain physical properties (e.g., solubility in polar solvents), but unless specifically indicated, the salts disclosed herein are equivalent to the parent form of the compound for purposes of the present disclosure.
By "agent" is meant a compound that provides a therapeutic benefit when administered to an individual.
"Phosphorothioate linkage" means a modified phosphate linkage in which one non-bridging oxygen atom is replaced with a sulfur atom.
"Moiety" means a defined number of consecutive (i.e., linked) nucleobases of a nucleic acid. In certain embodiments, a moiety is a defined number of consecutive nucleobases of a target nucleic acid. In certain embodiments, a moiety is a defined number of consecutive nucleobases of an oligonucleotide.
"Preventing" means delaying or preventing the onset, or progression of a disease, disorder, or condition over a period of time.
By "RNA interference compound" or "RNAi compound" is meant a compound that acts at least in part through the RNA-induced silencing complex (RISC) pathway or Ago2 (rather than through rnase H) to modulate a target nucleic acid and/or a protein encoded by a target nucleic acid. RNAi compounds include, but are not limited to, double stranded siRNA, single stranded siRNA, and microRNAs, including microRNA mimics.
"Sense oligonucleotide" or "sense strand" means the strand of a duplex compound that comprises a region that is substantially complementary to a region of the antisense strand of the compound.
"Specifically inhibit" with respect to a target nucleic acid or protein means reducing or blocking the expression or activity of the target nucleic acid or protein while minimizing or eliminating the effect on non-target nucleic acids or proteins.
"Subunit" with respect to an oligonucleotide means a nucleotide, nucleoside, nucleobase or sugar or modified nucleotide, nucleoside, nucleobase or sugar as provided herein.
"Target nucleic acid", "target RNA" and "nucleic acid target" all mean nucleic acids that can be targeted by the compounds described herein.
"Target region" means a portion of a target nucleic acid targeted by one or more compounds.
By "targeting moiety" is meant a conjugate group that provides enhanced affinity to a selected target (e.g., a molecule, cell or cell type, compartment (e.g., cell or organ compartment), tissue, organ, or body region) as compared to, for example, a compound that does not contain such moiety.
"Terminal group" means a chemical group or radical covalently attached to the end of an oligonucleotide.
"Therapeutically effective amount" or "effective amount" means that amount of a compound, agent or composition that provides a therapeutic benefit to an individual. A "therapeutically effective amount" or "effective amount" is an amount sufficient for the compound to achieve the stated purpose (e.g., effect for which administration is effected, treat, prevent or ameliorate a disease or one or more symptoms of a disease or disorder) relative to the absence of the compound. An example of a "therapeutically effective amount" or "effective amount" is an amount sufficient to help treat, prevent, ameliorate, or alleviate one or more symptoms of a disease. "alleviating" of one or more symptoms (and grammatical equivalents of the phrase) means a reduction in the severity or frequency of one or more symptoms, or elimination of one or more symptoms. A "prophylactically effective amount" of a drug is an amount of such drug: will have the intended prophylactic effect, e.g., preventing or delaying the onset (or recurrence) of a lesion, disease, pathological condition or disorder, or reducing the likelihood of onset (or recurrence) of a lesion, disease, pathological condition or disorder, or symptoms thereof, when administered to a subject. The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent sufficient to provide a therapeutic benefit to an individual, e.g., to treat, prevent or ameliorate a disease or disorder or symptoms thereof, as described above. For example, a therapeutically effective amount will exhibit an increase or decrease of at least 5%, 10%, 15%, 20%, 25%, 40%, 50%, 60%, 75%, 80%, 90%, or at least 100% for a given parameter. Therapeutic effects may also be expressed as "fold" increases or decreases. For example, a therapeutically effective amount may have an effect of at least 1.2-fold, 1.5-fold, 2-fold, 5-fold, or more relative to a control.
The term "treatment" and variations thereof refers to any sign of successful treatment or amelioration of a lesion, disease, pathological condition, or disorder, including any objective or subjective parameter, such as alleviation; relief; weakening symptoms or making lesions, pathological conditions or disorders more tolerable to the patient; slowing the rate of degradation or decay; making the end point of degradation less debilitating; improving physical or mental health of the patient. Treatment or amelioration of symptoms can be based on objective or subjective parameters, including the results of physical examination. The term "treatment" and variations thereof may include the prevention of injury, pathological condition, disorder or disease. In some embodiments, the treatment is prophylaxis. In some embodiments, the treatment does not include prophylaxis.
As used herein, "treatment" or variations thereof (and as well understood in the art) also broadly includes any method for achieving a beneficial or desired result (including clinical results) in a subject's condition. Beneficial or desired clinical results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, preventing spread or spread of disease, slowing or slowing of disease progression, amelioration or palliation of the disease state, diminishment of disease recurrence, and remission, whether partial or total, and whether detectable or undetectable. In other words, "treatment" as used herein includes any cure, amelioration, or prevention of a disease. Treatment may prevent the occurrence of disease; inhibiting the spread of the disease; alleviating symptoms of the disease, completely or partially eliminating the underlying cause of the disease, shortening the duration of the disease, or a combination of these events.
As used herein, "treatment" and variations thereof include prophylactic treatment. The method of treatment comprises administering to the subject a therapeutically effective amount of a compound described herein. The administering step may consist of a single administration or may comprise a series of administrations. The length of the treatment period depends on a variety of factors, such as the severity of the condition, the age of the patient, the concentration of the compound, the activity of the composition used in the treatment, or a combination thereof. It will also be appreciated that the effective dosage of the agent for treatment or prevention may be increased or decreased during a particular treatment or prevention regimen. In some cases, long-term administration may be required. For example, the composition is administered to the subject in an amount and for a duration sufficient to treat the patient.
"Treating" refers to the administration of a compound or pharmaceutical composition to an animal to effect a change or amelioration of a disease, disorder, or condition in the animal.
Certain compounds of the present disclosure have an asymmetric carbon atom (optical or chiral center) or double bond; enantiomers, racemates, diastereomers, tautomers, geometric isomers, stereoisomeric forms, which may be defined in absolute stereochemistry as (R) -or (S) -or (D) -or (L) - (for amino acids, and individual isomers are all encompassed within the scope of the present disclosure. The compounds of the present disclosure do not include compounds known in the art that are too unstable to synthesize and/or isolate. The present disclosure is meant to include both racemic and optically pure forms of the compounds. Optically active (R) -and (S) -or (D) -and (L) -isomers can be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When a compound described herein contains an olefinic bond or other geometric asymmetric center, and unless specified otherwise, it is intended that the compound include both E and Z geometric isomers.
The term "isomer" as used herein refers to compounds having the same number and kind of atoms and thus the same molecular weight, but different structural arrangements or configurations of the atoms.
The term "tautomer" as used herein refers to one of two or more structural isomers that exist in equilibrium and are readily converted from one isomeric form to another.
It will be apparent to those skilled in the art that certain compounds of the present disclosure may exist in tautomeric forms, all such tautomeric forms of the compounds being within the scope of the present disclosure.
Unless otherwise indicated, structures described herein are also intended to include all stereochemical forms of the structure (i.e., R and S configurations for each asymmetric center). Thus, single stereochemical isomers, as well as mixtures of enantiomers and diastereomers, of the compounds of the invention are within the scope of the present disclosure.
As used herein, "chiral enriched population" means a plurality of molecules of the same molecular formula, wherein the number or percentage of molecules in a population containing a particular stereochemical configuration at a particular chiral center is greater than the number or percentage of molecules in a population that are expected to contain the same particular stereochemical configuration at the same particular chiral center when the particular chiral center is stereoirregular. A chiral enriched population of molecules having multiple chiral centers per molecule may comprise one or more stereogenic random chiral centers. In certain embodiments, the molecule is a modified oligonucleotide. In certain embodiments, the molecule is a compound comprising a modified oligonucleotide.
Unless otherwise indicated, structures described herein are also intended to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the structure of the present invention are within the scope of the present disclosure, except that hydrogen is replaced with deuterium or tritium, or carbon is replaced with 13 C or 14 C-enriched carbon.
As used herein, "stereogenic random chiral centers" means chiral centers having a random stereochemical configuration in the context of groups of molecules having the same formula. For example, in a population of molecules comprising a stereogenic chiral center, the number of molecules having the (S) configuration of the stereogenic chiral center may be, but need not be, the same as the number of molecules having the (R) configuration of the stereogenic chiral center. The stereochemical configuration of a chiral center is considered random when it is the result of a synthetic method that is not intended to control the stereochemical configuration. In certain embodiments, the stereorandom chiral center is a stereorandom phosphorothioate internucleoside linkage.
Certain embodiments
In certain aspects, the present disclosure relates to methods, compounds, and compositions for inhibiting AGT. In certain embodiments, AGT is specifically inhibited. In certain embodiments, AGT is specifically degraded. In certain embodiments, AGT expression is inhibited. In certain embodiments, AGT translation is inhibited. In certain embodiments, AGT activity is inhibited. In certain embodiments, AGT expression, translation, or activity is reduced by at least 10% relative to expression, translation, or activity in an untreated or control sample. For example, in certain embodiments, AGT expression, translation, or activity is reduced by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, 10% to 50%, 25% to 75%, 50% to 99%, or 75% to 99% relative to expression, translation, or activity in an untreated or control sample. In certain embodiments, AGT is reduced in expression, translation, or activity as measured by any suitable assay, including but not limited to an immunoassay, hybridization-based assay, or sequencing-based assay (e.g., RNA-Seq).
In certain aspects, the disclosure relates to compounds that target AGT nucleic acids. In certain embodiments, the AGT nucleic acid has the following: the sequence shown in GENBANK accession No. nm_000029.4 (incorporated herein by SEQ ID No. 1), the complement of nucleotides 230702523 to 230745583 of nc_000001.11 (incorporated herein by SEQ ID No. 2), nm_001382817.3 (incorporated herein by SEQ ID No. 3) and nucleotides 5469 to 17068 of ng_008836.2 (incorporated herein by SEQ ID No. 4).
In certain embodiments, the compound is an oligomeric compound. In certain embodiments, the compound is single-stranded. In certain embodiments, the compound is double-stranded.
Certain embodiments provide compounds comprising a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166.
Certain embodiments provide compounds comprising a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence.
Certain embodiments provide compounds comprising a modified oligonucleotide having a nucleotide sequence selected from the group consisting of SEQ ID NOs: 10 to 166 in nucleobase sequence the nucleobase sequence of either.
In certain embodiments, the modified oligonucleotide has a nucleotide sequence that matches SEQ ID NO:1 or 3, at least 80%, at least 85%, at least 90% or at least 95% complementary nucleobase sequence. In certain embodiments, the modified oligonucleotide comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase. In certain embodiments, the compound is double-stranded.
Certain embodiments provide a compound comprising: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide.
In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleobase sequence comprising at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any of the nucleobase sequences provided in tables 2 to 25, 42, 45, 50 and 51; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide.
Certain embodiments provide a compound comprising: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide.
Certain embodiments provide a compound comprising: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide.
In certain embodiments, the first modified oligonucleotide has a nucleobase sequence of: and SEQ ID NO:1 or 3 has at least 80%, at least 85%, at least 90% or at least 95% complementarity or identity. In certain embodiments, the first modified oligonucleotide having a nucleobase sequence hybridizes to SEQ ID NO:1 or 3 has at least 1, at least 2, at least 3 mismatches. In certain embodiments, the region of complementarity between the first strand and the second strand is 14 to 30 linked nucleosides in length. In certain embodiments, the region of complementarity between the first strand and the second strand is 14 to 23 linked nucleosides in length. In certain embodiments, the region of complementarity between the first strand and the second strand is 19 to 23 linked nucleosides in length. In certain embodiments, the region of complementarity between the first strand and the second strand is 21 to 23 linked nucleosides in length. In certain embodiments, the first modified oligonucleotide is fully complementary to the second modified oligonucleotide.
In certain embodiments, the first modified oligonucleotide of any of the foregoing compounds comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase. In certain embodiments, the second modified oligonucleotide of any of the foregoing compounds comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase. In certain embodiments, the modified internucleoside linkage is a phosphorothioate internucleoside linkage or a methylphosphonate internucleoside linkage. In certain embodiments, the phosphorothioate internucleoside linkage or methylphosphonate internucleoside linkage is at the 3 'end of the first modified oligonucleotide or the second modified oligonucleotide or the 5' end of the first modified oligonucleotide. In certain embodiments, the modified sugar comprises a modification selected from the group consisting of halogen, alkoxy, and bicyclic sugar. In certain embodiments, the modified sugar comprises a 2' -F modification. In certain embodiments, the modified sugar comprises a 2' -OMe modification. In certain embodiments, each nucleoside of the first modified oligonucleotide comprises a modified sugar. In certain embodiments, each nucleoside of the second modified oligonucleotide comprises a modified sugar. In certain embodiments, the modified sugar comprises a modification selected from the group consisting of: halogen, alkoxy, and bicyclic sugar, or combinations thereof. In certain embodiments, the modified sugar comprises a modification selected from the group consisting of: 2' -MOE, 2' -F and 2' -OMe, or a combination thereof. In certain embodiments, the first modified oligonucleotide comprises no more than ten 2' -F sugar modifications. In certain embodiments, the second modified oligonucleotide comprises no more than five 2' -F sugar modifications.
In certain embodiments, a compound of any of the preceding embodiments comprises a conjugate group. In certain embodiments, the conjugate group is attached to the 5' end of the modified oligonucleotide. In certain embodiments, the conjugate group is a targeting moiety. In certain embodiments, the targeting moiety comprises one or more galnacs. In certain embodiments, the modified oligonucleotide is a second modified oligonucleotide or a sense oligonucleotide. In certain embodiments, one or more galnacs are linked to the 2 'or 3' position of the ribosyl ring. In certain embodiments, one or more galnacs are linked to a 5' nucleoside of the modified oligonucleotide. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is selected from the following formula or a salt, solvate, or hydrate thereof, wherein R is a moiety of the modified oligonucleotide other than the 5' nucleoside:
In certain embodiments, R' is O. In certain embodiments, R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula I and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula I and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula II and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula II and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula III and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula III and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula IV and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula IV and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula V and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula V and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VI and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VI and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VII and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VII and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VIII and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VIII and R' is S.
Certain embodiments provide a compound comprising: a first modified oligonucleotide selected from any one of:
Ref ID NO:IA0297,IA0300,IA0301,IA0304,IA0305,IA0335-338,IA0343-359,IA0431432,IA0435,IA440446,IA0727-728,IA0500-501, And the group consisting of IA0868,
And a second modified oligonucleotide of 14 to 21 linked nucleosides in length that is fully complementary to the first modified oligonucleotide.
Certain embodiments provide compounds comprising a first modified oligonucleotide consisting of IA0443 and a second modified oligonucleotide consisting of IS 0505. In certain embodiments, the compound comprises a first modified oligonucleotide consisting of IA0445 and a second modified oligonucleotide consisting of IS 0509.
In certain embodiments, a compound of any of the preceding embodiments is in the form of a pharmaceutically acceptable salt. In certain embodiments, the pharmaceutically acceptable salt is a sodium salt. In certain embodiments, the pharmaceutically acceptable salt is a potassium salt.
Certain embodiments provide compositions comprising a compound of any of the preceding embodiments and a pharmaceutically acceptable carrier.
Certain embodiments provide compositions comprising a compound of any of the preceding embodiments for use in therapy.
Certain embodiments provide methods of treating, preventing, or ameliorating a disease, disorder, or condition associated with AGT in a subject, comprising administering to the subject a compound that targets AGT, thereby treating, preventing, or ameliorating the disease.
In certain embodiments, a compound or composition of any of the preceding embodiments is administered to a subject. In certain embodiments, the disease, disorder or condition is a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
In certain embodiments, administration of the compounds inhibits or reduces or ameliorates RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
In certain embodiments, a compound of any one of the preceding embodiments, or a composition comprising a compound, is administered to a subject in a therapeutically effective amount. In certain embodiments, a compound or a composition comprising a compound of any of the preceding embodiments is administered to a subject at a dosage level sufficient to deliver about 1 to 100mg/kg of subject body weight. In certain embodiments, a compound or a composition comprising a compound of any of the preceding embodiments is administered to a subject in a fixed dose of about 25mg to about 1,000 mg. In certain embodiments, the compound or composition is administered to an individual at up to the dose level or fixed dose one or more times a day.
In certain embodiments, a compound or a composition comprising a compound of any of the preceding embodiments is administered to an individual daily, weekly, monthly, quarterly, or yearly. In certain embodiments, the compound or composition comprising the compound of any of the preceding embodiments is administered to the subject from about once a quarter (i.e., once every three months) to about once a year. In certain embodiments, the compound of any one of the preceding embodiments or the composition comprising the compound is administered to the subject about once a quarter, about once every six months, or about once a year.
Certain embodiments provide a method of inhibiting AGT expression in a cell comprising contacting the cell with a compound that targets AGT, thereby inhibiting AGT expression in the cell. In certain embodiments, the cell is in the liver of the individual. In certain embodiments, the individual has or is at risk of having: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
Certain embodiments provide methods of reducing or inhibiting RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysm, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in an individual, comprising administering to the individual a compound that targets AGT, thereby reducing or inhibiting RAAS-related disease, disorder, or condition, or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysm, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in the individual. In certain embodiments, the individual has or is at risk of having: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound is an AGT-targeting compound. In certain embodiments, the compound is any one of the foregoing compounds. In certain embodiments, the compound or composition is administered parenterally.
Certain embodiments provide the use of an AGT-targeted compound for the treatment, prevention or amelioration of diseases, disorders or conditions associated with AGT. In certain embodiments, the disease, disorder or condition is a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound is an AGT-targeting compound. In certain embodiments, the compound is any one of the foregoing compounds.
Certain embodiments provide for the use of a compound that targets AGT in the manufacture of a medicament for treating, preventing or ameliorating a disease, disorder or condition associated with AGT. In certain embodiments, the disease, disorder or condition is a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound is an AGT-targeting compound. In certain embodiments, the compound is any one of the foregoing compounds.
Certain indications
In certain aspects, the present disclosure relates to methods of inhibiting AGT expression by administering an AGT-targeting compound, which are useful for treating, preventing, or ameliorating an AGT-related disorder in an individual. In certain embodiments, the compound may be an AGT-specific inhibitor. In certain embodiments, the compound may be an antisense oligonucleotide, an oligomeric compound, or an AGT-targeting oligonucleotide.
In certain aspects, the disclosure relates to treating, preventing or ameliorating a disease, disorder or condition associated with AGT. In certain embodiments, the AGT-related diseases, disorders or conditions that can be treated, prevented and/or ameliorated by the methods provided herein include RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, renal disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. Certain compounds provided herein relate to compounds and compositions that reduce the following in animals: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
In certain embodiments, a method of treating, preventing, or ameliorating a disease associated with AGT in a subject comprises administering to the subject a compound comprising an AGT-specific inhibitor, thereby treating, preventing, or ameliorating the disease. In certain embodiments, the individual is identified as having or at risk of having an AGT-related disease. In certain embodiments, the disease is a RAAS-related disease. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, a compound comprises a modified oligonucleotide of length (e.g., 14 to 30, e.g., 14 to 23 linked nucleosides) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, a compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In certain embodiments, the single-chain compound may be 14 to 30, 14 to 23, 14 to 20, 16 to 20, or 14 to 16 linked nucleosides in length. In certain embodiments, the single-chain compound may be 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 linked nucleosides in length. In certain embodiments, a duplex compound may comprise two oligonucleotides of the same or different lengths, as described elsewhere herein. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence. In certain embodiments, the compound is administered to the subject parenterally. In certain embodiments, administration of the compounds ameliorates, maintains (preserve) or prevents RAAS-related diseases, disorders or conditions, or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in an animal.
In certain embodiments, a method of treating, preventing or ameliorating a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in an animal comprises administering to the individual a compound comprising an AGT-specific inhibitor, thereby treating, preventing or ameliorating a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, a disease, Fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence. In certain embodiments, administration of the compounds improves, maintains or prevents RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in an animal. In certain embodiments, the individual is identified as having or at risk of having an AGT-related disease.
In certain embodiments, a method of inhibiting AGT expression in a subject having or at risk of having an AGT-related disease comprises administering to the subject a compound comprising an AGT-specific inhibitor, thereby inhibiting AGT expression in the subject. In certain embodiments, administration of the compound inhibits the expression of AGT in the liver. In certain embodiments, the disease is a RAAS-related disease. In certain embodiments, the individual has or is at risk of having: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO:85 and 134 in nucleobase sequence the nucleobase sequence of either. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 85 and 134 in nucleobase sequence the nucleobase sequence of either. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence. In certain embodiments, the compound is administered to the subject parenterally. In certain embodiments, administration of the compounds ameliorates, maintains or prevents RAAS-related diseases, disorders or conditions or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
In certain embodiments, the method of inhibiting AGT expression in a cell comprises contacting the cell with a compound comprising an AGT-specific inhibitor, thereby inhibiting AGT expression in the cell. In certain embodiments, the cell is a hepatocyte. In certain embodiments, the cells are in the liver. In certain embodiments, the cells are in the liver of an individual suffering from or at risk of suffering from: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. in certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 85 and 134 in nucleobase sequence the nucleobase sequence of either. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence.
In certain embodiments, a method of reducing or inhibiting RAAS-related diseases, disorders or conditions, or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in an individual suffering from or at risk of suffering from an AGT-related disease comprises administering to the individual a compound comprising an AGT-specific inhibitor, thereby reducing or inhibiting RAAS-related disease in the individual, A disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the individual has or is at risk of having: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence. in certain embodiments, the compound is administered to the subject parenterally. In certain embodiments, the individual is identified as having or at risk of having an AGT-related disease.
Certain embodiments relate to compounds comprising AGT-specific inhibitors for use in the treatment of diseases, disorders or conditions associated with AGT. In certain embodiments, the disease, disorder or condition is a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO:85 and 134 in nucleobase sequence the nucleobase sequence of either. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence. in certain embodiments, the compound is administered to the subject parenterally.
Certain embodiments relate to compounds comprising AGT-specific inhibitors for reducing or inhibiting the following: RAAS-related diseases, disorders or conditions or symptoms thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO:85 and 134 in nucleobase sequence the nucleobase sequence of either. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 85 and 134 in nucleobase sequence the nucleobase sequence of either. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. in certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 a nucleobase sequence of any one of the sequences; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence.
Certain embodiments relate to the use of a compound comprising an AGT-specific inhibitor in the manufacture or preparation of a medicament for the treatment of an AGT-related disease. Certain embodiments relate to the use of a compound comprising an AGT-specific inhibitor in the manufacture of a medicament for the treatment of a disease, disorder or condition associated with AGT. In certain embodiments, the disease, disorder, or condition is a RAAS-related disease, disorder, or condition, or a symptom thereof. In certain embodiments, the disease, disorder or condition is hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO:85 and 134 in nucleobase sequence the nucleobase sequence of either. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 85 and 134 in nucleobase sequence the nucleobase sequence of either. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence.
Certain embodiments relate to the use of a compound comprising an AGT-specific inhibitor in the manufacture or preparation of a medicament for reducing or inhibiting a RAAS-related disease, disorder, or condition, or symptom thereof, in an individual suffering from, or at risk of suffering from, an AGT-related RAAS-related disease, disorder, or condition, or symptom thereof. In certain embodiments, the RAAS-related disease, disorder or condition is hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. Certain embodiments relate to the use of a compound comprising an AGT-specific inhibitor in the manufacture of a medicament for the treatment of a disease, disorder or condition associated with AGT. In certain embodiments, the disease, disorder or condition is a RAAS-related disease, disorder or condition or symptom thereof, or hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment. In certain embodiments, the compound comprises an antisense oligonucleotide targeting AGT. In certain embodiments, the compound comprises an AGT-targeting oligonucleotide. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) and has a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166, a core of any one of nucleobase sequence of the base sequence. In certain embodiments, the compound comprises a modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a sequence comprising SEQ ID NO: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In certain embodiments, the compound comprises a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134. In any of the foregoing embodiments, the compound may be single-stranded or double-stranded. In any of the foregoing embodiments, the compound may be an antisense oligonucleotide or an oligomeric compound. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 or 167 to 327; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a nucleotide sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide (e.g., a modified oligonucleotide of 14 to 30, e.g., 14 to 23 linked nucleosides in length) having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: a nucleobase sequence of the nucleobase sequence of any one of 85 and 134; and a second modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 244 and 294, respectively one core nucleobase sequence of the base sequence.
In any of the foregoing methods or uses, the compound may be an oligomeric compound. In any of the foregoing methods or uses, the compound may be single-stranded or double-stranded. In any of the foregoing methods or uses, the compound may target AGT. In certain embodiments, the compound comprises or consists of a modified oligonucleotide. In certain embodiments, the compounds comprise one or more modified oligonucleotides. In certain embodiments, the compound comprises a first modified oligonucleotide and a second modified oligonucleotide. In certain embodiments, the modified oligonucleotide is 8 to 80 linked nucleosides in length, 10 to 30 linked nucleosides in length, 14 to 23 linked nucleosides in length, or 19 to 23 linked nucleosides in length. In certain embodiments, the modified oligonucleotide has a nucleobase sequence of: and SEQ ID NO:1 and 3 has at least 80%, at least 85%, at least 90%, at least 95% or 100% complementarity. In certain embodiments, the modified oligonucleotide comprises at least one modified internucleoside linkage, at least one modified sugar, and/or at least one modified nucleobase. In certain embodiments, the modified internucleoside linkage is a phosphorothioate internucleoside linkage. In certain embodiments, the modified sugar is a bicyclic sugar, 2' -MOE, 2' -F, or 2' -OMe. In certain embodiments, the modified nucleobase is a 5-methylcytosine. In any of the preceding embodiments, each modified oligonucleotide is independently 12 to 30, 14 to 25, 14 to 24, 14 to 23, 16 to 23, 17 to 23, 18 to 23, 19 to 22, or 19 to 20 linked nucleosides in length. In certain embodiments, the modified oligonucleotide having a nucleobase sequence hybridizes to SEQ ID NO:1 and 3 has at least 1, at least 2, at least 3 mismatches.
In any of the foregoing methods or uses, the compound comprises a first modified oligonucleotide and a second modified oligonucleotide, wherein a region of complementarity exists between the first modified oligonucleotide and the second modified oligonucleotide. In certain embodiments, the length of the region of complementarity between the first oligonucleotide and the second oligonucleotide is 14 to 23, 19 to 23, or 21 to 23 linked nucleosides. In certain embodiments, the first modified oligonucleotide is fully complementary to the second modified oligonucleotide. In certain embodiments, the first modified oligonucleotide comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase. In certain embodiments, the second modified oligonucleotide comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase. In certain embodiments, the modified internucleoside linkage is a phosphorothioate internucleoside linkage or a methylphosphonate internucleoside linkage. In certain embodiments, the modified internucleoside linkage is at the 3 'end of the first modified oligonucleotide or the second modified oligonucleotide or the 5' end of the first modified oligonucleotide or the second modified oligonucleotide. In certain embodiments, the first modified oligonucleotide or the second modified oligonucleotide comprises one or more modified sugars. In certain embodiments, each nucleoside of the first modified oligonucleotide or the second modified oligonucleotide comprises a modified sugar. In certain embodiments, the modified sugar comprises a modification selected from the group consisting of halogen, alkoxy, and bicyclic sugar. In certain embodiments, the modified sugar comprises a modification selected from the group consisting of: 2' -MOE, 2' -F and 2' -OMe, or a combination thereof. In certain embodiments, the first modified oligonucleotide or the second modified oligonucleotide comprises no more than ten 2' -F sugar modifications. In certain embodiments, the first modified oligonucleotide or the second modified oligonucleotide comprises no more than five 2' -F sugar modifications.
In any of the foregoing methods or uses, the compound comprises a conjugate group. In certain embodiments, the conjugate group is attached to the 5' end of the modified oligonucleotide. In certain embodiments, the conjugate group is a targeting moiety. In certain embodiments, the targeting moiety comprises one or more galnacs. In certain embodiments, one or more galnacs are linked to the 2 'or 3' position of the ribosyl ring. In certain embodiments, one or more galnacs are linked to a 5' nucleoside of the modified oligonucleotide. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is selected from formulas I-VIII or a salt, solvate or hydrate thereof, wherein R is a modified oligonucleotide other than a 5' nucleoside. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula I and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula I and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula II and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula II and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula III and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula III and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula IV and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula IV and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula V and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula V and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VI and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VI and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VII and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VII and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VIII and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VIH and R' is S.
In any of the foregoing methods or uses, the compound comprises: a first modified oligonucleotide selected from any one of:
Ref ID NO:IA0297,IA0300,IA0301,IA0304,IA0305,IA0335-338,IA0343-359,IA0431432,IA0435,IA440-446,IA0727-728,IA0500-501, And IA0868;
And a second modified oligonucleotide of 14 to 23 linked nucleosides in length that is fully complementary to the first modified oligonucleotide. In certain embodiments, the compound comprises a peptide as Ref ID NO: the first modified oligonucleotide of IA0443 and is Ref ID NO: a second modified oligonucleotide of IS 0505. In certain embodiments, the compound comprises a peptide as Ref ID NO: the first modified oligonucleotide of IA0445 and a nucleotide sequence of Ref ID NO: a second modified oligonucleotide of IS 0509. In certain embodiments, the compound is in the form of a pharmaceutically acceptable salt. In certain embodiments, the pharmaceutically acceptable salt is a sodium salt. In certain embodiments, the pharmaceutically acceptable salt is a potassium salt. In certain embodiments, a composition comprises a compound of any of the preceding embodiments and a pharmaceutically acceptable carrier.
In any of the foregoing methods or uses, the compound of any of the foregoing embodiments or the composition comprising the compound is administered to the subject in a therapeutically effective amount. In certain embodiments, a compound or a composition comprising a compound of any of the preceding embodiments is administered to a subject at a dosage level sufficient to deliver about 1 to 100mg/kg of subject body weight. In certain embodiments, a compound or a composition comprising a compound of any of the preceding embodiments is administered to a subject in a fixed dose of about 25mg to about 1,000 mg. In certain embodiments, the composition is administered to the individual at up to the dose level or a fixed dose one or more times a day.
In any of the foregoing methods or uses, the compound or composition comprising the compound of any of the foregoing embodiments is administered to the individual daily, weekly, monthly, quarterly, or yearly. In certain embodiments, the compound or composition comprising the compound of any of the preceding embodiments is administered to the subject from about once a quarter (i.e., once every three months) to about once a year. In certain embodiments, the compound of any one of the preceding embodiments or the composition comprising the compound is administered to the subject about once a quarter, about once every six months, or about once a year.
Certain compounds
In certain aspects, the disclosure relates to compounds comprising or consisting of oligomeric compounds. In certain embodiments, the oligomeric compound comprises a nucleobase sequence that is complementary to a nucleobase sequence of a target nucleic acid.
In certain aspects, the disclosure relates to compounds comprising or consisting of modified oligonucleotides. In certain embodiments, the modified oligonucleotide has a nucleobase sequence that is complementary to a nucleobase sequence of a target nucleic acid.
In certain aspects, the disclosure relates to compounds comprising or consisting of antisense oligonucleotides. In certain embodiments, the antisense oligonucleotide has a nucleobase sequence that is complementary to a nucleobase sequence of a target nucleic acid.
In certain aspects, the disclosure relates to compounds that are single-chain compounds. In certain embodiments, the single-chain compound comprises or consists of an oligomeric compound. In certain embodiments, such oligomeric compounds comprise or consist of an oligonucleotide and optionally a conjugation group. In certain embodiments, the oligonucleotide is a modified oligonucleotide. In certain embodiments, the oligonucleotide is an antisense oligonucleotide. In certain embodiments, the oligonucleotide or modified oligonucleotide of the single-stranded compound comprises a self-complementary nucleobase sequence.
In certain aspects, the disclosure relates to compounds that are double-chain compounds. In certain embodiments, the double-chain compound comprises or consists of an oligomeric compound. In certain embodiments, the duplex compound comprises a first oligonucleotide and a second oligonucleotide. In certain embodiments, the first oligonucleotide has a region complementary to the target nucleic acid and the second oligonucleotide has a region complementary to the first modified oligonucleotide. In certain embodiments, the duplex compound comprises a modified oligonucleotide. In certain embodiments, the modified oligonucleotide has a region complementary to the target nucleic acid. In certain embodiments, the double-stranded compound comprises a first modified oligonucleotide and a second modified oligonucleotide. In certain embodiments, the first modified oligonucleotide has a region complementary to the target nucleic acid and the second modified oligonucleotide has a region complementary to the first modified oligonucleotide. In certain embodiments, the oligonucleotide or modified oligonucleotide of the duplex compound is an RNA oligonucleotide. In such embodiments, the thymine nucleobase in a modified oligonucleotide is replaced with a uracil nucleobase.
In certain embodiments, the compounds described herein comprise a conjugate group. In certain embodiments, the first oligonucleotide or first modified oligonucleotide of the duplex compound comprises a conjugate group. In certain embodiments, the second oligonucleotide or second modified oligonucleotide of the duplex compound comprises a conjugate group. In certain embodiments, the first oligonucleotide or first modified oligonucleotide and the second oligonucleotide or second modified oligonucleotide of the duplex compound each comprise a conjugate group.
In certain embodiments, the compound is 14 to 30 linked nucleosides in length. In certain embodiments, the first oligonucleotide or first modified oligonucleotide of the duplex compound is 14 to 30 linked nucleosides in length. In certain embodiments, the second oligonucleotide or second modified oligonucleotide is 14 to 30 linked nucleosides in length. In certain embodiments, the oligonucleotide or modified oligonucleotide of the duplex compound is blunt-ended at one or both termini of the compound. In certain embodiments, the oligonucleotide or modified oligonucleotide of the duplex compound comprises a non-complementary protruding nucleoside at one or both termini of the compound.
In certain embodiments, the compound has a nucleotide sequence comprising SEQ ID NO:10 to 166, at least 14 consecutive nucleobases of any of the nucleobase sequences. In certain embodiments, one of the oligonucleotide or modified oligonucleotide of the duplex compound has a sequence comprising SEQ ID NO:10 to 166, at least 14 consecutive nucleobases of any of the nucleobase sequences.
Some examples of single and double stranded compounds include, but are not limited to, oligonucleotides, antisense oligonucleotides, siRNA, microRNA targeting oligonucleotides, placeholder-based compounds (e.g., mRNA processing or translation blocking compounds and splicing compounds), and single stranded RNAi compounds (e.g., small hairpin RNAs (SMALL HAIRPIN RNA, SHRNA), single stranded sirnas (ssrnas), and microrna mimics).
In certain embodiments, the compounds described herein have nucleobase sequences that, when written in the 5 'to 3' direction, comprise the reverse complement of the target region of the target nucleic acid to which they are targeted.
In certain embodiments, the compounds described herein comprise oligonucleotides of 12 to 30 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides of 12 to 23 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides of 14 to 30 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides of 14 to 23 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 15 to 30 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 15 to 23 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 16 to 30 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 16 to 23 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides ranging in length from 17 to 30 linked subunits. In certain embodiments, the compounds described herein comprise oligonucleotides ranging in length from 17 to 23 linked subunits. In certain embodiments, the compounds described herein comprise oligonucleotides of 18 to 30 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides of 18 to 23 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides ranging in length from 19 to 30 linked subunits. In certain embodiments, the compounds described herein comprise oligonucleotides of 19 to 23 linked subunits in length. In other words, such oligonucleotides are respectively 12 to 30 linked subunits, 12 to 23 linked subunits, 14 to 30 linked subunits, 14 to 23 linked subunits, 15 to 30 linked subunits, 15 to 23 linked subunits, 16 to 30 linked subunits, 16 to 23 linked subunits, 17 to 30 linked subunits, 17 to 23 linked subunits, 18 to 30 linked subunits, 18 to 23 linked subunits, 19 to 30 linked subunits or 19 to 23 linked subunits. In certain embodiments, the compounds described herein comprise oligonucleotides of length 14 linked subunits. In certain embodiments, the compounds described herein comprise oligonucleotides that are 16 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 17 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 18 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 19 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 20 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 21 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides of 22 linked subunits in length. In certain embodiments, the compounds described herein comprise oligonucleotides that are 23 linked subunits in length. In other embodiments, a compound described herein comprises an oligonucleotide of 8 to 80, 12 to 50, 13 to 30, 13 to 50, 14 to 30, 14 to 50, 15 to 30, 15 to 50, 16 to 30, 16 to 50, 17 to 30, 17 to 50, 18 to 23, 18 to 24, 18 to 25, 18 to 50, 19 to 23, 19 to 30, 19 to 50, 20 to 23, or 20 to 30 linked subunits. In certain such embodiments, a compound described herein comprises an oligonucleotide of length 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 linked subunits, or a range defined by any two of the above values.
In certain embodiments, the compounds may also comprise additional moieties, such as a conjugate group or a delivery moiety. In certain embodiments, such compounds are oligomeric compounds, and the additional moiety is attached to the oligonucleotide. In certain embodiments, the conjugate group is attached to a nucleoside of the oligonucleotide.
In certain embodiments, the compound may be shortened or truncated. For example, one or more subunits may be deleted from the 5 'end (5' truncation) or alternatively from the 3 'end (3' truncation) of the oligonucleotide.
In certain embodiments, the compound may be extended. For example, one or more subunits may be linked to the 3 'end or the 5' end of an oligonucleotide. In certain embodiments, at least one subunit (e.g.,
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,
Or more subunits) are attached to the 5' end of the oligonucleotide. In certain embodiments, at least one subunit (e.g.,
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,
Or more subunits) are attached to the 3' end of the oligonucleotide. In certain embodiments, at least one or more subunits may be linked to the 3 'end or the 5' end of an oligonucleotide of a duplex compound, resulting in a 3 'and/or 5' terminal overhang. In certain embodiments, at least one subunit (e.g.,
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,
Or more subunits) are attached to the 5' ends of two oligonucleotides of a duplex compound. In certain embodiments, at least one subunit (e.g.,
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,
Or more subunits) are attached to the 3' ends of two oligonucleotides of a duplex compound. In certain embodiments, the subunits are linked at the same end to both oligonucleotides of the duplex compound (e.g., the subunits are linked to the 3 'end of one of the oligonucleotides and the subunits are linked to the 5' end of the other oligonucleotide). In certain embodiments, when a subunit is attached to two oligonucleotides of a duplex compound at the same end, the number of subunits attached to each oligonucleotide may be the same or may be different. In certain embodiments, when a subunit is linked at the same end to two oligonucleotides of a duplex compound, the number of subunits linked to each oligonucleotide is the same. In certain embodiments, when a subunit is attached to two oligonucleotides of a duplex compound at the same end, the number of subunits attached to each oligonucleotide is different. The case where subunits are linked at the same end to two oligonucleotides of a duplex compound may occur at one or both ends of the duplex compound. In certain embodiments, the subunits attached to the 3 'and/or 5' ends are modified.
In certain embodiments, the compounds described herein are oligonucleotides. In certain embodiments, the compounds described herein are modified oligonucleotides. In certain embodiments, the compounds described herein are antisense oligonucleotides. In certain embodiments, the compounds described herein are oligomeric compounds. In certain embodiments, the compounds described herein are RNAi compounds. In certain embodiments, the compounds described herein are siRNA compounds.
In certain embodiments, the compounds described herein can comprise any oligonucleotide sequence that targets AGT described herein. In certain embodiments, the compound may be double-stranded.
In certain embodiments, the compound comprises an oligonucleotide having a nucleobase sequence comprising the sequence of SEQ ID NO: at least 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 consecutive nucleobase portions of any of the nucleobase sequences of 10 to 166. In certain embodiments, the compound comprises a second oligonucleotide.
In certain embodiments, the compound comprises a ribonucleotide, wherein for any of the sequences provided herein, the oligonucleotide has uracil (U) in place of thymine (T). In certain embodiments, the compound comprises a deoxyribonucleotide, wherein for any of the sequences provided herein, the oligonucleotide has thymine (T) in place of uracil (U).
Some mechanisms
In certain embodiments, the compounds described herein comprise or consist of modified oligonucleotides. In certain embodiments, the compounds described herein comprise or consist of antisense oligonucleotides. In certain embodiments, the compound comprises or consists of an oligomeric compound. In certain embodiments, the compounds described herein are capable of hybridizing to a target nucleic acid. In certain embodiments, the compounds described herein selectively affect one or more target nucleic acids. Such compounds comprise the nucleobase sequence: hybridization to one or more target nucleic acids to produce one or more desired activities, and not to one or more non-target nucleic acids, or not in such a way as to result in significant undesired activity.
In certain embodiments, hybridization of a compound described herein to a target nucleic acid results in recruitment of one or more proteins that cause cleavage of the target nucleic acid. For example, loading certain compounds or portions of compounds described herein into RNA-induced silencing complex (RISC) ultimately results in cleavage of the target nucleic acid. For example, certain compounds described herein result in cleavage of a target nucleic acid by Argonaute. The compound loaded into RISC is an RNAi compound. The RNAi compound can be double stranded (siRNA) or single stranded (ssRNA).
In certain embodiments, hybridization of a compound described herein to a target nucleic acid does not result in recruitment of proteins that cleave the target nucleic acid. In certain such embodiments, hybridization of a compound to a target nucleic acid results in an alteration in the splicing of the target nucleic acid. In certain embodiments, hybridization of a compound to a target nucleic acid results in inhibition of binding interactions between the target nucleic acid and a protein or other nucleic acid. In certain such embodiments, hybridization of a compound to a target nucleic acid results in an alteration in RNA processing. In certain such embodiments, hybridization of a compound to a target nucleic acid results in an alteration in translation of the target nucleic acid.
The activity of the compound resulting from hybridization with the target nucleic acid can be directly or indirectly observed. In certain embodiments, the observation or detection of activity involves observing or detecting a change in the amount of a target nucleic acid or protein encoded by such target nucleic acid, a change in the proportion of splice variants of the nucleic acid or protein, and/or a phenotypic change in a cell or animal.
Certain modifications
In certain aspects, the disclosure relates to compounds comprising or consisting of oligonucleotides. The oligonucleotides consist of linked nucleosides. In certain embodiments, the oligonucleotide may be unmodified RNA or DNA, or may be modified. In certain embodiments, the oligonucleotide is a modified oligonucleotide. In certain embodiments, the modified oligonucleotide comprises at least one modified sugar, modified nucleobase, or modified internucleoside linkage relative to the unmodified RNA or DNA. In certain embodiments, the oligonucleotide has a modified nucleoside. The modified nucleoside can comprise a modified sugar, a modified nucleobase, or both a modified sugar and a modified nucleobase. Modified oligonucleotides may also include terminal modifications, such as 5 '-terminal modifications and 3' -terminal modifications.
Sugar modification and motifs
In certain embodiments, the modified sugar is a substituted furanosyl sugar or a non-bicyclic modified sugar. In certain embodiments, the modified sugar is a bicyclic or tricyclic modified sugar. In certain embodiments, the modified sugar is a sugar substitute. The sugar substitute may comprise one or more of the substituents described herein.
In certain embodiments, the modified sugar is a substituted furanosyl or non-bicyclic modified sugar. In certain embodiments, the furanosyl sugar is a ribosyl sugar. In certain embodiments, the furanosyl sugar comprises one or more substituents, including but not limited to substituents at the 2', 3', 4', and 5' positions.
In certain embodiments, substituents at the 2' position include, but are not limited to, F and OCH 3 ("OMe", "O-methyl" or "methoxy"). In certain embodiments, substituents suitable for the 2' position of the non-bicyclic modified sugar include, but are not limited to, halogen, allyl, amino, azido 、SH、CN、OCN、CF3、OCF3、F、Cl、Br、SCH3、SOCH3、SO2CH3、ONO2、NO2、N3, and NH 2. In certain embodiments, substituents at the 2' position include, but are not limited to, O- (C 1-C10) alkoxy, alkoxyalkyl, O-alkyl, S-alkyl, N-alkyl, O-alkenyl, S-alkenyl, N-alkenyl, O-alkynyl, S-alkynyl, N-alkynyl, O-alkyl-O-alkyl, alkynyl, wherein alkyl, alkenyl and alkynyl groups may be substituted or unsubstituted C 1 to C 10 alkyl or C 2 to C 10 alkenyl and alkynyl groups. In certain embodiments, substituents at the 2' position include, but are not limited to, alkylaryl, arylalkyl, O-alkylaryl, and O-arylalkyl groups. In certain embodiments, these 2' substituents may be further substituted with one or more substituents independently selected from the group consisting of: hydroxy, alkoxy, carboxy, benzyl, phenyl, nitro (NO 2), thiol, thioalkoxy, thioalkyl, halogen, alkyl, aryl, alkenyl, and alkynyl. In certain embodiments, substituents at the 2' position include, but are not limited to O[(CH2)nO]mCH3、O(CH2)nOCH3、O(CH2)nCH3、O(CH2)nONH2、O(CH2)nNH2、O(CH2)nSCH3 and O (CH 2)nON[(CH2)nCH3) ]2, wherein n and m are independently from 1 to about 10. in certain embodiments, substituents at the 2' position include, but are not limited to OCH2CH2OCH3("MOE")、O(CH2)2ON(CH3)2("DMAOE")、O(CH2)2O(CH2)2N(CH3)2("DMAEOE") and OCH 2C(=O)-N(H)CH3 ("NMA").
In certain embodiments, substituents at the 4' position suitable for non-bicyclic modified sugars include, but are not limited to, alkoxy (e.g., methoxy), alkyl, and Manoharan et al, those described in WO 2015/106128. In certain embodiments, substituents at the 5' position suitable for non-bicyclic modified sugars include, but are not limited to, methyl ("Me") (R or S), vinyl, and methoxy. In certain embodiments, the substituents described herein at the 2', 4', and 5' positions may be added to other specific positions on the sugar. In certain embodiments, such substituents may be added to the 3 'position of a sugar on the 3' terminal nucleoside or the 5 'position of the 5' terminal nucleoside. In certain embodiments, the non-bicyclic modified sugar may comprise more than one non-bridging sugar substituent. In certain such embodiments, the non-bicyclic modified sugar substituents include, but are not limited to, 5'-Me-2' -F, 5'-Me-2' -OMe (including both R and S isomers). In certain embodiments, modified sugar substituents include Migawa et al, WO 2008/101157 and Rajeev et al, those described in US 2013/0203836.
In certain embodiments, the modified sugar is a bicyclic sugar. Bicyclic sugar is a modified sugar comprising two rings, wherein the second ring is formed by a bridge connecting two atoms in the first ring, forming a bicyclic structure. In certain embodiments, the bicyclic sugar comprises a bridging substituent bridging two atoms of the furanosyl ring to form a second ring. In certain embodiments, the bicyclic sugar does not comprise a furanosyl moiety. A "bicyclic nucleoside" ("BNA") is a nucleoside having a bicyclic sugar. In certain embodiments, the bicyclic sugar comprises a bridge between the 4 'and 2' furanose ring atoms. In certain embodiments, the bicyclic sugar comprises a bridge between the 5 'and 3' furanose ring atoms. In some such embodiments, the furanose ring is a ribose ring. In certain embodiments, 4 'to 2' bridging substituents include, but are not limited to 4'-CH2-2'、4'-(CH2)2-2'、4'-(CH2)3-2'、4'-CH2-O-2'("LNA")、4'-CH2-S-2'、4'-(CH2)2-O-2'("ENA")、4'-CH(CH3)-O-2'( when in the S configuration are "constrained ethyl" or "cEt"), 4'-CH2-O-CH 2-2'、4'-CH2-N(R)-2'、4'-CH(CH2OCH3) -O-2' ("constrained MOE" or "cMOE") and analogs thereof (e.g., U.S. Pat. No.7,399,845), 4'-C (CH 3)(CH3) -O-2' and analogs thereof (e.g., U.S. Pat. No.8,278,283), 4'-CH 2-N(OCH3) -2' and analogs thereof (e.g., U.S. Pat. No.8,278,425), 4'-CH 2-O-N(CH3) -2' (e.g., U.S. Pat. publication No. 2004/0171570), 4'-CH 2 -N (R) -O-2', Wherein R is H, C 1-C1 alkyl or a protecting group (e.g., U.S. patent No.7,427,672), 4'-CH 2-C(H)(CH3) -2' (e.g., chattopadhyaya el al, j. Org. Chem.,2009, 74, 118-134 and 4'-CH 2-C(=CH2) -2' and analogs thereof (e.g., U.S. Pat. No.8,278,426). The entire contents of each of the foregoing are hereby incorporated by reference. Additional representative U.S. patents and U.S. patent publications teaching the preparation of bicyclic nucleic acid nucleotides include, but are not limited to, the following:
US patent No.6,268,490;6,525,191;6,670,461;6,770,748;6,794,499;6,998,484;7,053,207;7,034,133;7,084,125;7,399,845;7,427,672;7,569,686;7,741,457;8,022,193;8,030,467;8,278,425;8,278,426;8,278,283;US 2008/0039618; and US 2009/0012281, US 2013/0190383; WO 2013/036868,
The entire contents of each of which are hereby incorporated by reference. Any 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, e.g., WO 99/14226). Unless otherwise indicated, bicyclic nucleosides specified herein are in the β -D configuration.
In certain embodiments, the modified sugar is a sugar substitute. In certain embodiments, the sugar substitute has an oxygen atom replaced, for example, with a sulfur, carbon, or nitrogen atom. In certain such embodiments, the sugar substitute may further comprise bridging and/or non-bridging substituents as described herein. In certain embodiments, the sugar substitute comprises a ring having in addition to 5 atoms. In certain such embodiments, the sugar substitute comprises a cyclobutyl moiety in place of the pentose sugar. In certain embodiments, the sugar substitute comprises a six-membered ring in place of the pentose glycosyl sugar. In certain embodiments, the sugar substitute comprises tetrahydropyran ("THP") in place of the pentose glycosyl sugar. In certain embodiments, the sugar substitute comprises morpholino in place of the pentose glycosyl sugar. Representative U.S. patents teaching the preparation of such modified sugar structures include, but are not limited to
In the us patent No.4,981,957;5,118,800;5,166,315;5,185,444;5,319,080;5,359,044;5,393,878;5,446,137;5,466,786;5,514,785;5,519,134;5,567,811;5,576,427;5,591,722;5,597,909;5,610,300;5,627,053;5,639,873;5,646,265;5,658,873;5,670,633;5,700,920;7,875,733;7,939,677,8,088,904;8,440,803; and 9,005,906,
The entire contents of each of the foregoing are hereby incorporated by reference.
In some embodiments, the sugar substitute comprises an acyclic moiety. In certain embodiments, the sugar substitute is an unlocking nucleic acid ("UNA (unlocked nucleic acid)"). UNA is an unlocked acyclic nucleic acid in which any bond of a sugar has been removed, forming an unlocked "sugar" residue. In one example, UNA also encompasses monomers in which the bond between C1 'to C4' is removed (i.e., covalent carbon-oxygen-carbon bonds between C1 'and C4' carbons). In another example, the C2'-C3' bond of the sugar (i.e., the covalent carbon-carbon bond between the C2 'and C3' carbons) has been removed. Representative U.S. publications that teach the preparation of UNA include, but are not limited to, U.S. patent No.8,314,227; and U.S. patent publication nos. 2013/0096289, 2013/0011922, and 2011/0313020, each of which is incorporated herein by reference in its entirety. In certain embodiments, the sugar substitutes comprise peptide nucleic acids ("PNA (peptide nucleic acid)"), acyclic butyl nucleic acids (see, e.g., kumar et al, org.biomol. Chem.,2013, 11, 5853-5865), and nucleosides and oligonucleotides described in US2013/130378, manoharan et al, the entire contents of which are incorporated herein by reference. Many other bicyclic and tricyclic sugars and sugar-substituted ring systems known in the art can be used for the modified nucleosides.
In certain aspects, the disclosure relates to compounds comprising at least one oligonucleotide, wherein the nucleosides of such oligonucleotides comprise one or more types of modified and/or unmodified sugars, arranged in a defined pattern or "sugar motif" along the oligonucleotide or region thereof. In certain instances, such sugar motifs include, but are not limited to, any of the sugar modification patterns described herein.
In certain embodiments, the oligonucleotide comprises a spacer sugar motif. The spacer oligonucleotide comprises or consists of: an area having two outer "wing" regions and a central or inner "spacer" region. The spacer and wing regions form a contiguous sequence of nucleosides wherein a majority of the nucleoside sugars of each wing are different from a majority of the nucleoside sugars of the spacer. In certain embodiments, the wing region comprises a majority of modified sugars and the gap comprises a majority of unmodified sugars. In certain embodiments, the spacer nucleoside is a deoxynucleoside. Compounds having a spacer sugar motif are described, for example, in U.S. patent 8,790,919, the entire contents of which are incorporated herein by reference.
In certain embodiments, one or both oligonucleotides of the duplex compound comprises a triplet sugar motif. An oligonucleotide with a triplet sugar motif comprises three identical sugar modifications on three consecutive nucleosides. In certain embodiments, the triplets are at or near the cleavage site of the oligonucleotide. In certain embodiments, the oligonucleotide of the duplex compound may comprise more than one triplet sugar motif. In certain embodiments, the same sugar modification of the triplet sugar motif is a 2' -F modification. Compounds having a triplet sugar motif are disclosed, for example, in U.S. patent 10,668,170, the entire contents of which are incorporated herein by reference.
In certain embodiments, one or both oligonucleotides of the duplex compound comprise a tetrad sugar motif. An oligonucleotide with a tetrad sugar motif comprises four identical sugar modifications on four consecutive nucleosides. In certain embodiments, the quadruplet is at or near the cleavage site. In certain embodiments, the oligonucleotides of the duplex compound may comprise more than one tetrad sugar motif. In certain embodiments, the same sugar modification of the tetrad sugar motif is a 2' -F modification. For duplex compounds having duplex regions of 19 to 23 nucleotides in length, the cleavage sites of antisense oligonucleotides are typically at about 10, 11 and 12 positions from the 5' end. In certain embodiments, the tetrad sugar motif is at positions 8, 9, 10, 11 of the sense oligonucleotide, counting from the first nucleoside at the 5 'end of the sense oligonucleotide, or counting from the first paired nucleotide within the duplex region at the 5' end of the sense oligonucleotide; positions 9, 10, 11 and 12; positions 10, 11, 12 and 13; 11, 12, 13, 14; or at positions 12, 13, 14, 15. In certain embodiments, the tetrad sugar motif is at positions 8, 9, 10, 11 of the antisense oligonucleotide, counting from the first nucleoside at the 5 'end of the antisense oligonucleotide, or counting from the first paired nucleotide within the duplex region at the 5' end of the antisense oligonucleotide; positions 9, 10, 11 and 12; positions 10, 11, 12 and 13; 11, 12, 13, 14; or at positions 12, 13, 14, 15. The cleavage site may vary depending on the length of the duplex region of the duplex and the position of the quadruplex may be varied accordingly.
In certain embodiments, the oligonucleotide comprises an alternating sugar motif. In certain embodiments, one or both oligonucleotides of the duplex compound comprise alternating sugar motifs. An oligonucleotide having an alternating sugar motif comprises at least two different sugar modifications, wherein one or more consecutive nucleosides comprising a first sugar modification alternate with one or more consecutive nucleosides comprising a second sugar modification and one or more consecutive nucleosides comprising a third sugar modification, and the like. For example, if A, B and C each represent a type of modification of a nucleoside, the alternating motif may be
"Ababababababab.," "aabbaabbaabbaabba.," "aabaabaabaab.," "aaabaaabaaab.," "aaabbbaabbabbb.," or "abccabcabac.," and the like.
In certain embodiments, the alternating sugar motif repeats at least 2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 consecutive nucleobases along the oligonucleotide. In certain embodiments, the alternan motif consists of two different sugar modifications. In certain embodiments, the alternan motif comprises 2'-OMe and 2' -F sugar modifications.
In certain embodiments, each nucleoside of an oligonucleotide is independently modified by one or more sugar modifications provided herein. In certain embodiments, each oligonucleotide of the duplex compound independently has one or more of the motifs provided herein. In certain embodiments, the oligonucleotide comprising a sugar motif is fully modified in that each nucleoside except the nucleoside comprising the sugar motif comprises a sugar modification.
Nucleobase modification and motif
In certain embodiments, the compounds described herein comprise modified oligonucleotides. In certain embodiments, the modified oligonucleotide comprises one or more nucleosides that comprise a modified nucleobase. In certain embodiments, the modified oligonucleotide comprises one or more nucleobase-free nucleosides, referred to as abasic nucleosides.
In certain embodiments, the modified nucleobase is selected from the group consisting of: 5-substituted pyrimidines, 6-azapyrimidines, alkyl-or alkynyl-substituted pyrimidines, alkyl-substituted purines and N-2, N-6 and O-6-substituted purines. In certain embodiments, the modified nucleobase is selected from the group consisting of: 2-aminopropyladenine, 5-hydroxymethylcytosine, 5-methylcytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-N-methylguanine, 6-N-methyladenine, 2-propyladenine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-propynyl (C.ident.C-CH 3) uracil, 5-propynyl cytosine, 6-azouracil, 6-azocytosine, 6-azothymine, 5-ribosyl uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-sulfanyl, 8-hydroxy 8-aza and other 8-substituted purines, 5-halogeno, in particular 5-bromo, 5-trifluoromethyl, 5-halogeno-uracil and 5-halogeno-cytosine, 7-methylguanine, 7-methyladenine, 2-F-adenine, 2-aminoadenine, 7-deazaadenine, 7-deazaguanine, 3-deazaadenine, 6-N-benzoyladenine, 2-N-isobutyrylguanine, 4-N-benzoylcytosine, 4-N-benzoyluracil, 5-methyl 4-N-benzoylcytosine, 5-methyl 4-N-benzoyluracil, universal bases, hydrophobic bases, pan-dominant base (promiscuous base), enlarged-size base (size expanded base), and fluorinated base. Further modified nucleobases include tricyclic pyrimidines, such as 1, 3-diazaphenonesOxazin-2-one, 1, 3-diazaphenothiazin-2-one and 9- (2-aminoethoxy) -1, 3-diazaphenoneOxazin-2-one (G-clamp). Modified nucleobases can also include those in which the purine or pyrimidine base is replaced by other heterocycles (e.g., 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine, and 2-pyridone).
Other nucleobases include those disclosed in the following:
U.S. patent 3,687,808;Modified Nucleosides in Biochemistry,Biotechnology and Medicine,Herdewijn,P.ed.Wiley-VCH,2008;The Concise Encyclopedia Of Polymer Science And Engineering,, pages 858 to 859, ;Kroschwitz,J.L,Ed.,John Wiley&Sons,1990,858-859;Englisch et al.,Angewandte Chemie,International Edition,1991,30,613;Sanghvi,Y.S.,, chapter 15, DSRNA RESEARCH AND Applications, pages 289 to 302, ;Antisense Research and Applications,Crooke,S.T.and Lebleu,B.,Eds.,CRC Press,1993,273-288;Antisense Drug Technology,Crooke S.T.,Ed.,CRC Press,2008,163-166 and 442-443 (chapters 6 and 15),
Each of which is incorporated herein by reference.
Publications teaching the preparation of some of the above-mentioned modified nucleobases and other modified nucleobases include, but are not limited to, U.S. applications 2003/0158403 and 2003/0175906; U.S. patent
4,845,205;5,130,302;5,134,066;5,175,273;5,367,066;5,432,272;5,434,257;5,457,187;5,459,255;5,484,908;5,502,177;5,525,711;5,552,540;5,587,469;5,594,121;5,596,091;5,614,617;5,645,985;5,681,941;5,811,534;5,750,692;5,948,903;5,587,470;5,457,191;5,763,588;5,830,653;5,808,027;6,005,096.6,015,886;6,147,200;6,166,197;6,166,199;6,222,025;6,235,887;6,380,368;6,528,640;6,639,062;6,617,438;7,045,610;7,427,672; And 7,495,088, the number of which is set,
The entire contents of which are hereby incorporated by reference.
In certain embodiments, the compounds described herein comprise oligonucleotides. In certain embodiments, the oligonucleotides comprise modified and/or unmodified nucleobases arranged in a defined pattern or motif along the oligonucleotide or region thereof. In certain embodiments, each nucleobase is modified. In certain embodiments, none of the nucleobases is modified. In certain embodiments, each purine or each pyrimidine is modified. In certain embodiments, each adenine is modified. In certain embodiments, each guanine is modified. In certain embodiments, each thymine is modified. In certain embodiments, each uracil is modified. In certain embodiments, each cytosine is modified. In certain embodiments, some or all of the cytosine nucleobases in a modified oligonucleotide are 5-methylcytosine.
In certain embodiments, the modified oligonucleotide comprises a block of modified nucleobases. In certain such embodiments, the block is at the 3' end of the oligonucleotide. In certain embodiments, the block is within 3 nucleosides of the 3' terminus of the oligonucleotide. In certain embodiments, the block is at the 5' end of the oligonucleotide. In certain embodiments, the block is within 3 nucleosides of the 5' end of the oligonucleotide.
Internucleoside linkage modifications and motifs
The 3 'to 5' phosphodiester linkage is a naturally occurring internucleoside linkage in RNA and DNA. In certain embodiments, the compounds described herein have one or more modified, i.e., non-naturally occurring, internucleoside linkages. Certain non-naturally occurring internucleoside linkages can confer desirable properties, such as enhanced cellular uptake, enhanced affinity for a target nucleic acid, and improved stability in the presence of nucleases. Representative phosphorus-containing modified internucleoside linkages include, but are not limited to, phosphotriesters, alkylphosphonates (e.g., methylphosphonates), phosphinates, and phosphorothioates ("p=s") and phosphorodithioates ("HS-p=s"). Representative phosphorus-free internucleoside linkages include, but are not limited to, methyleneimino (-CH 2-N(CH3)-O-CH2), thiodiester, thiocarbamate (-O-C (=O (NH) -S-)), siloxane (-O-SiH 2 -O-)), and N, N' -dimethylhydrazine (-CH 2-N((CH3)-N((CH3) -). Methods for preparing phosphorus-containing and phosphorus-free internucleoside linkages are well known to those skilled in the art. Neutral internucleoside linkages include, but are not limited to, phosphotriesters, methylphosphonates, MMIs (3 '-CH 2-N(CH3) -O-5'), amide-3 (3 '-CH 2 -C (=O) -N (H) -5'), amide-4 (3 '-CH 2 -N (H) -C (=O) -5')), methylal (3 '-O-CH 2 -O-5'), methoxypropyl and thiomethylal (3 '-S-CH 2 -O-5'). Other neutral internucleoside linkages include nonionic linkages comprising: siloxanes (dialkylsiloxanes), carboxylates, carboxamides, sulfides, sulfonates and amides (see, e.g., :Carbohydrate Modifications in Antisense Research;Y.S.Sanghvi and P.D.Cook,Eds.,ACS Symposium Series 580; chapter III and chapter IV, 40-65). Other neutral internucleoside linkages include nonionic linkages comprising mixed N, O, S and CH 2 moieties.
In certain embodiments, the compounds provided herein comprise at least one modified internucleoside linkage. The modified internucleoside linkage can be placed at any position of the oligonucleotide. For duplex compounds, the modified internucleoside linkages can be placed within the sense oligonucleotide, the antisense oligonucleotide, or both oligonucleotides of the duplex compound.
In certain embodiments, internucleoside linkage modifications may occur on each nucleoside of the oligonucleotide. In certain embodiments, internucleoside linkage modifications may occur in an alternating pattern along the oligonucleotide. In certain embodiments, substantially each internucleoside linkage group is an internucleoside linkage phosphate (p=o). In certain embodiments, each internucleoside linkage group of the modified oligonucleotide is a phosphorothioate (p=s). In certain embodiments, each internucleoside linkage group of the modified oligonucleotide is independently selected from phosphorothioate and phosphate internucleoside linkages. In certain embodiments, the pattern of internucleoside linkage modifications on each oligonucleotide of the duplex is the same. In certain embodiments, the pattern of internucleoside linkage modifications on each oligonucleotide of the duplex is different. In certain embodiments, the double-chain compound comprises 6 to 8 modified internucleoside linkages. In certain embodiments, 6 to 8 modified internucleoside linkages are phosphorothioate internucleoside linkages or alkylphosphonate internucleoside linkages. In certain embodiments, the sense oligonucleotide comprises at least two modified internucleoside linkages at one or both of the 5 'end and the 3' end. In some such embodiments, the modified internucleoside linkage is a phosphorothioate internucleoside linkage or an alkylphosphonate internucleoside linkage. In certain embodiments, the antisense oligonucleotide comprises at least two modified internucleoside linkages at one or both of the 5 'end and the 3' end. In some such embodiments, the modified internucleoside linkage is a phosphorothioate internucleoside linkage or an alkylphosphonate internucleoside linkage.
In certain embodiments, the double-chain compound comprises an overhang region. In certain embodiments, the double-chain compound comprises phosphorothioate or alkylphosphate internucleoside linkage modifications in the region of the overhang. In certain embodiments, the double-stranded compound comprises phosphorothioate or alkylphosphonate internucleotide linkages linking an overhang nucleotide to a paired nucleotide adjacent to the overhang nucleotide. For example, there may be at least two phosphorothioate internucleoside linkages between the terminal three nucleosides, with two of the three nucleosides being the overhang nucleoside and the third being the partner nucleoside adjacent to the overhang nucleoside. These terminal three nucleosides can be at the 3 'end of the antisense oligonucleotide, the 3' end of the sense oligonucleotide, the 5 'end of the antisense oligonucleotide, or the 5' end of the antisense oligonucleotide.
In certain embodiments, the modified oligonucleotides comprise one or more internucleoside linkages having a chiral center. Representative chiral internucleoside linkages include, but are not limited to, alkyl phosphonates and phosphorothioates. Modified oligonucleotides comprising internucleoside linkages having a chiral center can be prepared as a population of modified oligonucleotides comprising stereoregular internucleoside linkages, or as a population of modified oligonucleotides comprising phosphorothioate linkages in a particular stereochemical configuration. In certain embodiments, the population of modified oligonucleotides comprises phosphorothioate internucleoside linkages, wherein all phosphorothioate internucleoside linkages are stereospecifically random. Such modified oligonucleotides can be produced using synthetic methods that result in random selection of the stereochemical configuration of each phosphorothioate linkage. As is well known to those skilled in the art, each individual phosphorothioate in each individual oligonucleotide molecule has a defined steric configuration. In certain embodiments, the population of modified oligonucleotides is enriched with modified oligonucleotides comprising one or more specific phosphorothioate internucleoside linkages in a specific, independently selected stereochemical configuration. In certain embodiments, a particular configuration of a particular phosphorothioate linkage is present in at least 65% of the molecules in the population. In certain embodiments, a particular configuration of a particular phosphorothioate linkage is present in at least 70% of the molecules in the population. In certain embodiments, a particular configuration of a particular phosphorothioate linkage is present in at least 80% of the molecules in the population. In certain embodiments, a particular configuration of a particular phosphorothioate linkage is present in at least 90% of the molecules in the population. In certain embodiments, a particular configuration of a particular phosphorothioate linkage is present in at least 99% of the molecules in the population. Such enriched populations of modified oligonucleotides can be generated using synthetic methods known in the art, for example, oka et al, JACS 125, 8307 (2003), wann et al nuc.acid.res.42, 13456 (2014), and methods described in WO 2017/015555. In certain embodiments, the population of modified oligonucleotides is enriched in modified oligonucleotides having at least one indicated phosphorothioate in the (Sp) configuration. In certain embodiments, the population of modified oligonucleotides is enriched in modified oligonucleotides having at least one phosphorothioate in the (Rp) configuration.
Conjugation group
In certain embodiments, the compounds described herein comprise or consist of: one or more oligonucleotides and optionally one or more conjugation groups. The conjugate group may be attached to one or both ends of the oligonucleotide, and/or at any internal position. In certain embodiments, the conjugate group is attached to the 3' end of the oligonucleotide. In certain embodiments, the conjugate group is attached to the 5' end of the oligonucleotide. In certain embodiments, the oligonucleotide is covalently linked to one or more conjugate groups.
In certain embodiments, the conjugate group is a terminal group attached to one or both ends of the oligonucleotide. In some such embodiments, the end group is attached to the 3' end of the oligonucleotide. In some such embodiments, the end group is attached to the 5' end of the oligonucleotide. In certain embodiments, end groups include, but are not limited to, capping groups, phosphate moieties, protecting groups, modified or unmodified nucleosides, and two or more independently modified or unmodified nucleosides, such as overhangs.
In certain embodiments, the conjugate group modifies one or more properties of the attached oligonucleotide, including, but not limited to, pharmacodynamics, pharmacokinetics, stability, activity, half-life, binding, absorption, tissue distribution, cell distribution, cellular uptake, charge and clearance. In certain embodiments, the conjugate group enhances the affinity of the compound for a selected target, such as a molecule, cell or cell type, compartment (e.g., cell or organ compartment), tissue, organ, or body region, for example, as compared to a compound in the absence of such conjugate group. In certain embodiments, the conjugate group imparts novel properties to the attached oligonucleotide, e.g., a fluorophore or reporter group that enables detection of the oligonucleotide.
In certain embodiments, conjugation groups include, but are not limited to, intercalators, reporter molecules, polyamines, polyamides, peptides, carbohydrates, vitamin moieties, polyethylene glycols, thioethers, polyethers, cholesterol, thiocholesterols, cholic acid moieties, folic acid, lipids, phospholipids, biotin, phenazine, phenanthridine, anthraquinone, adamantane, acridine, fluorescein, rhodamine (rhodomine), coumarin, fluorophores, and dyes.
In certain embodiments, the conjugate group comprises an active drug substance, e.g., aspirin (aspirin), warfarin (warfarin), phenylbutazone, ibuprofen (ibuprofen), suprofen (suprofen), fenbufen (fen-bufen), ketoprofen (ketoprofen), (S) - (+) -pranoprofen (pranoprofen), carprofen (carprofen), dansyl sarcosine, 2,3, 5-triiodobenzoic acid, fingolimod (fingolimod), flufenamic acid (flufenamic acid), folinic acid, benzothiadiazine, chlorothiazide, diaza(Diazepine), indo-methicin, barbiturate, cephalosporin, sulfanilamide, antidiabetic, antibacterial or antibiotic.
In certain embodiments, the conjugate group is a targeting moiety. In certain embodiments, the targeting moiety includes, but is not limited to, a lectin, glycoprotein, lipid, protein, peptide, peptidomimetic, receptor ligand, antibody, thyrotropin, melanotropin, surfactant protein a, carbohydrate derivative, modified carbohydrate, carbohydrate cluster, polysaccharide, modified polysaccharide or polysaccharide derivative, mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine (GalNAc), N-acetylglucosamine multivalent mannose, multivalent fucose, glycosylated polyamino acid, multivalent galactose, transferrin, bisphosphonate, polyglutamic acid, polyaspartic acid, lipid, cholesterol, steroid, bile acid, folic acid, vitamin B12, vitamin a, biotin or RGD peptide mimetic.
In certain embodiments, the conjugate groups may include, but are not limited to, the conjugate groups described in the following references: for example cholesterol (e.g., LETSINGER ET al., proc. Natl. Acid. Sci. USA,1989, 86: 6553-6556); cholic acid (e.g., manoharan et al., biorg. Med. Chem. Let.,1994, 4:1053-1060); thioethers, for example, hexyl-S-tritylthiol (e.g., ,Manoharan et al.,Ann.NY.Acad.Sci.,1992,660:306-309;Manoharan et al.,Biorg.Med.Chem.Let.,1993,3:2765-2770); thiocholesterol (e.g., oberhauser et al., nucleic acids res.,1992, 20:533-538); Aliphatic chains, for example, dodecanediol or undecyl residues (e.g., ,Saison-Behmoaras et al.,EMBO J,1991,10:1111-1118;Kabanov et al.,FEBS Lett.,1990,259:327-330;Svinarchuk et al.,Biochimie,1993,75:49-54); phospholipids, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1, 2-di-O-hexadecyl-rac-glycerol-3-H-phosphonate (e.g., ,Manoharan et al.,Tetrahedron Lett.,1995,36:3651-3654;Shea et al.,Nucl.Acids Res.,1990,18:3777-3783); polyamine or polyethylene glycol chains (e.g., manoharan et al., nucleosis & Nucleotides, 1995, 14: 969-973); Adamantaneacetic acid (e.g., manoharan et al., tetrahedron lett.,1995, 36:3651-3654); palm-based (e.g., mishra et al, biochim. Biophys. Acta,1995, 1264:229-237); octadecylamine or hexylamino-carbonyloxy cholesterol moiety (e.g., crooke et al.j. Pharmacol. Exp. Ter., 1996, 277:923-937); tocopherols (e.g., NISHINA ET al., molecular Therapy Nucleic Acids,2015,4, e220 and NISHINA ET al., molecular Therapy,2008, 16:734-740); GalNAc and other carbohydrates (e.g., ,Maier et al.,Bioconjugate Chemistry,2003,14,18-29;Rensen et al.,J.Med.Chem.2004,47,5798-5808;WO2009/073809 and U.S. Pat. nos. 8,106,022;8,450,467 and 8,828,957; and WO2014/179445; WO2014/179620 and U.S. Pat. nos. 9,127,276;9,181,549 and 10,844,379), each of which is incorporated herein by reference in its entirety.
The conjugate group may be attached to the oligonucleotide via a conjugate linker. In certain embodiments, the conjugate linker comprises a chain structure (e.g., a hydrocarbyl chain) or an oligomer of repeating units or a combination of such repeating units. In certain embodiments, the conjugate linker comprises one or more groups selected from alkyl, amino, oxo, amide, disulfide, polyethylene glycol, ether, thioether, and hydroxyamino groups. In certain embodiments, the conjugate linker comprises at least one phosphorus group. In certain embodiments, the conjugate linker comprises at least one phosphate group. In certain embodiments, the conjugate linker comprises at least one neutral linking group. In certain embodiments, conjugated linkers include, but are not limited to, pyrrolidine, 8-amino-3, 6-dioxaoctanoic Acid (ADO), succinimidyl 4- (N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC), and 6-aminocaproic acid (AHEX or AHA). Other conjugation linkers include, but are not limited to, substituted or unsubstituted C 1-C10 alkyl, substituted or unsubstituted C 2-C10 alkenyl, or substituted or unsubstituted C 2-C10 alkynyl, with a non-limiting list of preferred substituents including hydroxy, amino, alkoxy, carboxy, benzyl, phenyl, nitro, thiol, thioalkoxy, halogen, alkyl, aryl, alkenyl, and alkynyl. In certain embodiments, the conjugate linker comprises 1 to 10 linker-nucleosides. In certain embodiments, such linker-nucleosides can be modified or unmodified nucleosides. It is often desirable to cleave the linker-nucleoside from the compound after the compound reaches the target tissue. Thus, the linker-nucleosides herein can be linked to each other and to the remainder of the compound by cleavable linkages. In this context, linker-nucleosides are not considered to be part of an oligonucleotide. Thus, in embodiments in which the compound comprises an oligonucleotide consisting of a particular number or range of linked nucleosides and/or having a particular percentage of complementarity to the reference nucleic acid, and the compound further comprises a conjugate group comprising a linker-nucleoside containing conjugate linker, these linker-nucleosides are not counted in the length of the oligonucleotide and are not used to determine the percentage of complementarity of the oligonucleotide to the reference nucleic acid.
In certain embodiments, the conjugate groups and conjugate linkers and other modifications include, but are not limited to, those described in the following references :US 5,994,517;US 6,300,319;US 6,660,720;US 6,906,182;US 7,262,177;US 7,491,805;US 8,106,022;US 7,723,509;US 9,127,276;US 2006/0148740;US 2011/0123520;WO2013/033230;WO2012/037254,Biessen et al.,J.Med.Chem.1995,38,1846-1852;Lee et al.,Bioorganic&Medicinal Chemistry 2011,19,2494-2500;Rensen et al.,J.Biol.Chem.2001,276,37577-37584;Rensen et al.,J.Med.Chem.2004,47,5798-5808;Sliedregt et al.,J.Med.Chem.1999,42,609-618;Valentijn et al.,Tetrahedron,1997,53,759-770;Lee,Carhohydr Res,1978,67,509-514;Connolly et al.,J Biol Chem,1982,257,939-945;Pavia et al.,Int J Pep Protein Res,1983,22,539-548;Lee et al.,Biochem,1984,23,4255-4261;Lee et al.,Glycoconjugate J,1987,4,317-328;Toyokuni et al.,Tetrahedron Lett,1990,31,2673-2676;Biessen et al.,J Med Chem,1995,38,1538-1546;Valentijn et al.,Tetrahedron,1997,53,759-770;Kim et al.,Tetrahedron Lett,1997,38,3487-3490;Lee et al.,Bioconjug Chem,1997,8,762-765;Kato et al.,Glycohiol,2001,11,821-829;Rensen et al.,J Biol Chem,2001,276,37577-37584;Lee et al.,Methods Enzymol,2003,362,38-43;Westerlind et al.,Glycoconj J,2004,21,227-241;Lee et al.,Bioorg Med Chem Lett,2006,16(19),5132-5135;Maierhofer et al.,Bioorg Med Chem,2007,15,7661-7676;Khorev et al.,Bioorg Med Chem,2008,16,5216-5231;Lee et al.,Bioorg Med Chem,2011,19,2494-2500;Kornilova et al.,Analyt Biochem,2012,425,43-46;Pujol et al.,Angew Chemie Int Ed Engl,2012,51,7445-7448;Biessen et al.,J Med Chem,1995,38,1846-1852;Sliedregt et al.,J Med Chem,1999,42,609-618;Rensen et al.,J Med Chem,2004,47,5798-5808;Rensen et al.,Arterioscler Thromh Vase Biol,2006,26,169-175;van Rossenberg et al.,Gene Ther,2004,11,457-464;Sato et al.,JAm Chem Soc,2004,126,14013-14022;Lee et al.,J Org Chem,2012,77,7564-7571;Biessen et al.,FASEB J,2000,14,1784-1792;Rajur et al.,Bioconjug Chem,1997,8,935-940;Duff et al.,Methods Enzymol,2000,313,297-321;Maier et al.,Bioconjug Chem,2003,14,18-29;Jayaprakash et al.,Org Lett,2010,12,5410-5413;Manoharan,Antisense Nucleic Acid Drug Dev,2002,12,103-128;Merwin et al.,Bioconjug Chem,1994,5,612-620;Tomiya et al.,Bioorg Med Chem,2013,21,5275-5281;
International application WO1998/013381;WO2011/038356;WO1997/046098;W02008/098788;W02004/101619;WO2012/037254;WO2011/120053;WO2011/100131;WO2011/163121;WO2012/177947;W02013/033230;W02013/075035;WO2012/083185;WO2012/083046;W02009/082607;WO2009/134487;W02010/144740;W02010/148013;WO 1997/020563;W02010/088537;W02002/043771;W02010/129709;WO2012/068187;WO2009/126933;W02004/024757;WO2010/054406;WO2012/089352;WO2012/089602;WO2013/166121;WO2013/165816; US patent 4,751,219;7,582,744;8,552,163;8,137,695;6,908,903;6,383,812;7,262,177;6,525,031;5,994,517;6,660,720;6,300,319;7,723,509;8,106,022;7,491,805;7,491,805;8,541,548;8,344,125;8,313,772;8,349,308;8,450,467;8,501,930;8,158,601;7,262,177;6,906,182;6,620,916;8,435,491;8,404,862;7,851,615; published US patent application publication US2011/0097264;
US2011/0097265;US2013/0004427;US2003/0119724;US2011/0207799;
US2012/0035115;US2012/0230938;US2005/0164235;US2006/0183886;
US2012/0136042;US2012/0095075;US2013/0109817;US2006/0148740;
US2008/0206869;US2012/0165393;US2012/0101148;US2013/0121954;
US 2011/0123218; US 2003/0077182; US2008/0108801; and US2009/0203132;
Each of which is incorporated by reference herein in its entirety.
Certain targeting moieties
In certain embodiments, the compounds provided herein comprise a conjugate group. In certain embodiments, the oligonucleotides provided herein comprise a conjugate group. In certain embodiments, the conjugate group is a targeting moiety. In certain embodiments, the targeting moiety comprises one or more galnacs. In certain embodiments, one or more galnacs are linked to one or more positions on the furanose ring. In certain embodiments, one or more galnacs are linked to a 2 'or 3' position on the furanose ring. In certain embodiments, the furanose ring is a subunit of an oligonucleotide. In certain embodiments, the furanose ring is the 5' nucleoside sugar of the oligonucleotide. In certain embodiments, the furanose ring is the 5' nucleoside sugar of the sense oligonucleotide. In some embodiments of the present invention, in some embodiments, the compound or oligonucleotide comprises one or more compounds having the formula or a salt thereof subunits of solvates or hydrates:
Wherein:
r 1 is H, adenine, guanine, thymine, cytosine, uracil, carbocyclyl, heterocyclyl, aryl, heteroaryl, or nucleobase isostere;
R 2 is an oligonucleotide sequence;
l 1 is alkyl or alkyl-C (=o) -NH-alkyl;
l 2 is alkyl or alkyl-C (=o) -NH-alkyl;
L 3 is a bond, phosphodiester bond, phosphorothioate bond, triazole, tetrazole, amide, reverse amide, carbamate, carbonate, urea, O, S, S (=o), S (=o) 2, NH, substituted N group, alkyl, alkenyl, dienyl, alkynyl, heteroalkyl, phosphate;
R 3 is H, -c= (O) -NH- (CH 2CH2O)j -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc;
R 4 is H, -c= (O) -NH- (CH 2CH2O)k -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc;
R 5 is-c= (O) -NH- (CH 2CH2O)m -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc;
R 6 is-c= (O) -NH- (CH 2CH2O)n -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc;
W and Q are each independently O, NH, CH 2, or CH 2 O;
S 1 and S 2 are each independently C (R 7) or N, wherein each instance of R 7 is independently H, alkyl, heteroalkyl, or halogen;
j is an integer from 1 to 10, inclusive;
k is an integer from 1 to 10, inclusive;
m is an integer from 1 to 10, inclusive; and
N is an integer from 1 to 10, inclusive.
In certain embodiments, R 3、R4、R5 and R 6 are the same. In certain embodiments, R 3、R5 and R 6 are the same. In certain embodiments, R 3 or R 4 is H.
In certain embodiments, L 1 and L 2 are the same.
In certain embodiments, L 1 and L 2 are each independently alkyl; r 3 is H, -C= (O) -NH- (CH 2CH2O)j -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc, R 4 is H, -C= (O) -NH- (CH 2CH2 O) k-GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc, R 5 is-C= (O) -NH- (CH 2CH2O)m -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc, and R 6 is-C= (O) -NH- (CH 2CH2O)n -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc.
In certain embodiments, L 1 and L 2 are each independently alkyl-C (=o) -NH-alkyl; r 3 is H, -C= (O) -NH- (CH 2CH2O)j -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc, R 4 is H, -C= (O) -NH- (CH 2CH2O)k -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc, R 5 is-C= (O) -NH- (CH 2CH2O)m -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc, and R 6 is-C= (O) -NH- (CH 2CH2O)n -GalNAc or-C (=O) -NH-alkyl-NH-C (=O) -alkyl-O-GalNAc.
In certain embodiments, R 4 is H.
In certain embodiments, L 1 and L 2 are each independently alkyl; r 3 is-c= (O) -NH- (CH 2CH2O)j -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc; R 4 is H; R 5 is-c= (O) -NH- (CH 2CH2O)m -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc, and R 6 is-c= (O) -NH- (CH 2CH2O)n -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc.
In certain embodiments, L 1 and L 2 are each independently alkyl-C (=o) -NH-alkyl; r 3 is-c= (O) -NH- (CH 2CH2O)j -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc; R 4 is H; R 5 is-c= (O) -NH- (CH 2CH2O)m -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc, and R 6 is-c= (O) -NH- (CH 2CH2O)n -GalNAc or-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc.
In certain embodiments, R 3 is-c= (O) -NH- (CH 2CH2O)j-GalNAc;R4 is H, R 5 is-c= (O) -NH- (CH 2CH2O)m -GalNAc, and R 6 is-c= (O) -NH- (CH 2CH2O)n -GalNAc.
In certain embodiments, R 3 is-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc; r 4 is H; r 5 is-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc; and R 6 is-C (=o) -NH-alkyl-NH-C (=o) -alkyl-O-GalNAc.
In some embodiments of the present invention, in some embodiments, the compound or oligonucleotide comprises one or more compounds having the formula or a salt thereof subunits of solvates or hydrates:
Wherein:
R 9 is H, adenine, guanine, thymine, cytosine, or uracil, or is an adenine, guanine, thymine, cytosine, or uracil, each comprising a Protecting Group (PG), a modified nucleobase, an optionally substituted alkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted carbocyclyl, an optionally substituted heterocyclyl, or a nucleobase isostere;
L is a bond, phosphodiester bond, phosphorothioate bond, triazole, tetrazole, amide, anti-amide, carbamate, carbonate, urea, alkyl or heteroalkyl;
R 2 is an oligonucleotide sequence;
Y 1 is O, CH 2、CH2 O or optionally substituted NH;
Y 2 is O, CH 2、CH2 O or optionally substituted NH;
y 3 is CO, SO 2、P(O)O、CH2-O-C(O)、CH2-NH-C(O)、CH2-NH-SO2 or CH 2;
Y 4 is CO, SO 2、P(O)O、CH2-O-C(O)、CH2-NH-C(O)、CH2-NH-SO2 or CH 2;
n 2 is 0, 1,2,3, 4,5, or 6; and
N 1、n3、n4 and n 5 are each independently 0, 1,2,3,4, 5, 6, 7, 8, 9 or 10.
In some embodiments of the present invention, in some embodiments, the compound or oligonucleotide comprises one or more compounds having the formula or a salt thereof subunits of solvates or hydrates:
Wherein:
Each n is independently 1, 2, 3, 4, or 5;
each m is independently 0,1, 2,3, 4, 5, or 6;
Each o is independently 0,1, 2,3, 4, 5, or 6;
L 1、L2 and L 3 are each independently absent, C (=o) or C (=o) NH;
Each Y 1 is independently O, CH (R a)、S、S(=O)、S(=O)2, NH, substituted N group) 、NHC(=O)、C(=O)NH、P(=O)2-O-、P(=O)(=S)-O、P(=S)2-O、-O-P(=O)2-O-、-O-P(=O)(=S)-O-、-O-P(=S)2-O-、-O-P(=O)2-、-O-P(=O)(=S)-、-O-P(=S)2-;
Each Y 2 is independently O, CH (R b)、S、S(=O)、S(=O)2, NH, substituted N group) 、NHC(=O)、C(=O)NH、P(=O)2-O-、P(=O)(=S)-O、P(=S)2-O、-O-P(=O)2-O-、-O-P(=O)(=S)-O-、-O-P(=S)2-O-、-O-P(=O)2-、-O-P(=O)(=S)-、-O-P(=S)2-;
Het 1、Het2 and Het 3 are each independently optionally substituted heteroaryl or optionally substituted heterocyclyl;
R 1 is an oligonucleotide sequence linked by a bond, phosphodiester bond, phosphorothioate bond, triazole, tetrazole, amide, reverse amide, carbamate, carbonate, urea, alkyl or heteroalkyl;
r 5、R6 and R 7 are each independently
R 9 is an optionally substituted heterocyclyl;
Each R a is independently H, alkyl, halogen, OR c, OR SR c;
Each R b is independently H, alkyl, halogen, OR c, OR SR c; and
Each R c is independently H or alkyl.
In certain embodiments, the subunit is selected from formulas I-VIII or a salt, solvate, or hydrate thereof, wherein R is a modified oligonucleotide other than a 5' nucleoside. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula I and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula I and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula II and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula II and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula III and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula III and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula IV and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula IV and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula V and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula V and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VI and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is of formula VI and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VII and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VII and R' is S. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VIII and R' is O. In certain embodiments, the 5 'nucleoside of the modified oligonucleotide is formula VIII and R' is S.
Target nucleic acid and target region
In certain embodiments, the compounds described herein comprise or consist of an oligonucleotide comprising a region complementary to a target nucleic acid. In certain embodiments, the target nucleic acid is an endogenous RNA molecule. In certain embodiments, the target nucleic acid encodes a protein. In certain embodiments, the target nucleic acid is non-coding. In certain such embodiments, the target nucleic acid is selected from the group consisting of mRNA and pre-mRNA, including intronic regions, exonic regions, and untranslated regions. In certain embodiments, the target RNA is mRNA. In certain embodiments, the target nucleic acid is a pre-mRNA. In certain such embodiments, the target region is entirely within an exon. In certain such embodiments, the target region is entirely within an intron. In certain embodiments, the target region spans an intron/exon junction. In certain embodiments, the target region is at least 50% within an intron.
In certain embodiments, the compounds disclosed herein hybridize to AGT nucleic acids. The most common hybridization mechanism involves hydrogen bonding between complementary nucleobases of nucleic acid molecules. Hybridization can occur under different conditions. Hybridization conditions are sequence dependent and are determined by the nature and composition of the nucleic acid molecule to be hybridized. Methods for determining whether a sequence specifically hybridizes to a target nucleic acid are well known in the art. In certain embodiments, the compounds provided herein specifically hybridize to AGT nucleic acids.
The nucleotide sequences encoding AGT include, but are not limited to, the following: GENBANK accession No. nm_000029.4 (incorporated herein as SEQ ID No. 1), the complement of nucleotides 230702523 to 230745583 of nc_000001.11 (incorporated herein as SEQ ID No. 2), nm_001382817.3 (incorporated herein as SEQ ID No. 3), and nucleotides 5469 to 17068 of ng_008836.2 (incorporated herein as SEQ ID No. 4).
Complementarity and method of detecting complementary
The oligonucleotides provided herein may have a defined percentage of complementarity to a particular nucleic acid, target region, oligonucleotide, or portion thereof. Non-complementary nucleobases are tolerated provided that the oligonucleotide remains capable of specifically hybridizing to the nucleic acid, oligonucleotide or portion thereof. In certain embodiments, an oligonucleotide or a particular portion thereof provided herein is at least or up to 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% complementary to a target nucleic acid, target region, oligonucleotide or a particular portion thereof. In certain embodiments, an oligonucleotide or a particular portion thereof provided herein is 70% to 75%, 75% to 80%, 80% to 85%, 85% to 90%, 90% to 95%, 95% to 100%, or any value in between these ranges, complementary to a target nucleic acid, a target region, an oligonucleotide or a particular portion thereof. The percent complementarity of an oligonucleotide to a target nucleic acid, target region, oligonucleotide, or specific portion thereof may be determined using conventional methods. For example, an oligonucleotide in which 18 of the 20 nucleobases of the oligonucleotide are complementary to the target region and therefore specifically hybridise would represent 90% complementarity. In this example, the remaining non-complementary nucleobases can be clustered or interspersed with complementary nucleobases and need not be linked to each other or to complementary nucleobases. Thus, an oligonucleotide of 18 nucleobases in length has four non-complementary nucleobases flanked by two regions of complete complementarity to the target nucleic acid, with a total complementarity to the target nucleic acid of 77.8%. The percent complementarity of an oligonucleotide to a region of a target nucleic acid, target region, oligonucleotide, or a particular portion thereof can be routinely determined using the BLAST program (basic local ALIGNMENT SEARCH tool) known in the art. In certain embodiments, an oligonucleotide or a particular portion thereof described herein is fully complementary (i.e., 100% complementary) to a target nucleic acid, target region, oligonucleotide or a particular portion thereof. For example, the oligonucleotide may be fully complementary to the target nucleic acid, target region, oligonucleotide, or a particular portion thereof. "fully complementary" as used herein means that each nucleobase of an oligonucleotide is complementary to a corresponding nucleobase of a target nucleic acid, target region, oligonucleotide or a particular portion thereof. For example, a 20 nucleobase oligonucleotide is fully complementary to a 400 nucleobase long target sequence, so long as there are corresponding 20 nucleobase moieties in the target nucleic acid that are fully complementary to the compound. "fully complementary" may also be used to refer to a particular portion of a first and/or second nucleic acid. For example, a 20 nucleobase moiety in a 30 nucleobase oligonucleotide may be "fully complementary" to a 20 nucleobase region in a 400 nucleobase long target sequence. If the target sequence has a corresponding 20 nucleobase moiety in which each nucleobase is complementary to 20 nucleobase moieties in the compound, then 20 nucleobase moieties in a 30 nucleobase compound are fully complementary to the target sequence. Meanwhile, the entire 30 nucleobase compound may or may not be fully complementary to the target sequence, depending on whether the remaining 10 nucleobases of the compound are also complementary to the target sequence. In certain embodiments, an oligonucleotide described herein comprises one or more mismatched nucleobases relative to a target nucleic acid, target region, oligonucleotide, or particular portion thereof. In certain embodiments, an oligonucleotide of length of up to 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23 nucleobases described herein comprises no more than 4, no more than 3, no more than 2, or no more than 1 non-complementary nucleobases relative to a target nucleic acid or a particular portion thereof. in certain embodiments, an oligonucleotide of length of up to 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleobases described herein comprises 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 nucleobases relative to a target nucleic acid, target region, oligonucleotide, or a particular portion thereof. In certain embodiments, the mismatch is at position 1,2, 3,4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 of the 5' end of the oligonucleotide. In certain embodiments, the mismatch is at position 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12, 13, or 14 of the 3' end of the oligonucleotide. In certain embodiments, the mismatch forms a wobble base pair with a corresponding nucleobase on the target nucleic acid. For example, in certain embodiments, the mismatches form a wobble base pair selected from the group consisting of: hypoxanthine (inosine nucleobase) and uracil (I: U base pairs); guanine and uracil (G: U base pairs); hypoxanthine and adenine (I: A base pairs); and hypoxanthine and cytosine (I: C base pairs). Thus, in certain embodiments, the mismatched nucleobases on an oligonucleotide comprise hypoxanthine, guanine, or uracil.
In certain embodiments, the oligonucleotides described herein may be complementary to a portion of a nucleic acid. As used herein, "moiety" refers to a defined number of consecutive nucleobases within a nucleic acid region. "part" may also refer to a defined number of consecutive nucleobases of an oligonucleotide. In certain embodiments, the oligonucleotide is complementary to at least 8 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 9 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 10 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 11 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 12 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 13 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 14 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 15 nucleobase moieties in the nucleic acid. In certain embodiments, the oligonucleotide is complementary to at least 16 nucleobase moieties in the nucleic acid. Oligonucleotides that are complementary to at least 9,10, 17, 18, 19, 20, 21, 22, 23 or more nucleobase moieties in a nucleic acid, or a range defined by any two of these values, are also contemplated. In certain embodiments, the oligonucleotide is an antisense oligonucleotide. In certain embodiments, the portion of the antisense oligonucleotide is compared to the equal length portion of the target nucleic acid. In certain embodiments, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleobase moieties are compared to an equal length moiety of a target nucleic acid. In certain embodiments, the oligonucleotide is a sense oligonucleotide. In certain embodiments, the portion of the sense oligonucleotide is compared to the equal length portion of the antisense oligonucleotide. In certain embodiments, 8,9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleobase moieties in a sense oligonucleotide are compared to the equivalent length of an antisense oligonucleotide.
Identity of
The oligonucleotides provided herein may also have a defined percent identity to a particular nucleic acid, target region, oligonucleotide, or particular portion thereof. As used herein, an oligonucleotide is identical to a sequence disclosed herein if it has the same nucleobase pairing ability. For example, DNA containing thymidine in place of uracil in the disclosed RNA sequences would be considered identical to the RNA sequences, as both uracil and thymidine pair with adenine. Shortened and lengthened forms of the compounds described herein are also contemplated as well as compounds having different bases relative to the compounds provided herein. The different bases may be adjacent to each other or dispersed throughout the compound. The percent identity of an oligonucleotide is calculated based on the number of bases having the same base pairing relative to the sequence to which it is compared. In certain embodiments, an oligonucleotide or portion thereof described herein has or has at least 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identity to one or more of the nucleic acids, oligonucleotides, or portions thereof disclosed herein. In certain embodiments, an oligonucleotide described herein has about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity, or any percentage between such values, to a particular nucleic acid or oligonucleotide or portion thereof.
In certain embodiments, an oligonucleotide may have one or more mismatched nucleobases. In certain such embodiments, the mismatch is at position 1,2,3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 of the 5' end of the oligonucleotide. In certain such embodiments, the mismatch is at position 1,2,3, 4,5, 6, 7, 8, 9, 10, 11, or 12, 13, or 14 of the 3' end of the oligonucleotide. In certain embodiments, the portion of the oligonucleotide is compared to the equal length portion of the target nucleic acid. In certain embodiments, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleobase moieties are compared to an equal length moiety of a target nucleic acid. In certain embodiments, the oligonucleotide is a sense oligonucleotide. In certain embodiments, the portion of the sense oligonucleotide is compared to the equal length portion of the target nucleic acid. In certain embodiments, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleobase moieties are compared to an equal length moiety of a target nucleic acid.
Pharmaceutical composition and formulation
The compounds described herein may be mixed with pharmaceutically active or inert substances for the preparation of pharmaceutical compositions or formulations. The compositions and methods used to formulate pharmaceutical compositions depend on a number of criteria including, but not limited to, the route of administration, the extent of the disease or the dose to be administered. Certain embodiments provide pharmaceutical compositions comprising one or more compounds or salts thereof. In certain embodiments, the compound is an antisense oligonucleotide. In certain embodiments, the compound is an oligomeric compound. In some embodiments of the present invention, in some embodiments, the compound comprises one or more modified oligonucleotides or consist of one or more modified oligonucleotides. In certain such embodiments, the pharmaceutical compositions comprise one or more compounds and a suitable pharmaceutically acceptable diluent or carrier. In certain embodiments, the pharmaceutical composition comprises one or more compounds and a sterile saline solution. In certain embodiments, such pharmaceutical compositions consist of one compound and a sterile saline solution. In certain embodiments, the sterile saline is pharmaceutical grade saline. In certain embodiments, the pharmaceutical composition comprises one or more compounds and sterile water. In certain embodiments, the pharmaceutical composition consists of one compound and sterile water. In certain embodiments, the sterile water is pharmaceutical grade water. In certain embodiments, the pharmaceutical composition comprises one or more compounds and phosphate-buffered saline (PBS). In certain embodiments, the pharmaceutical composition consists of one compound and sterile PBS. In certain embodiments, the sterile PBS is a pharmaceutical grade PBS.
The AGT-targeting compounds described herein can be used in pharmaceutical compositions by combining the compounds with a suitable pharmaceutically acceptable diluent or carrier. In certain embodiments, the pharmaceutically acceptable diluent is water, such as sterile water suitable for injection. Thus, in one embodiment, employed in the methods described herein is a pharmaceutical composition comprising an AGT-targeting compound and a pharmaceutically acceptable diluent. In certain embodiments, the pharmaceutically acceptable diluent is water. In some embodiments of the present invention, in some embodiments, the compounds comprise one or more modified oligonucleotides provided herein or consist of one or more modified oligonucleotides provided herein.
Pharmaceutical compositions comprising the compounds provided herein encompass any pharmaceutically acceptable salt, ester, or salt of such an ester, or any other oligonucleotide capable of providing (directly or indirectly) a biologically active metabolite or residue thereof upon administration to an animal (including a human). In certain embodiments, the compound is an antisense oligonucleotide. In certain embodiments, the compound is an oligomeric compound. In some embodiments of the present invention, in some embodiments, the compound comprises one or more modified oligonucleotides or consist of one or more modified oligonucleotides. Thus, for example, the present disclosure also relates to pharmaceutically acceptable salts of compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other biological equivalents. Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts. Prodrugs may comprise additional nucleosides incorporated at one or both termini of the compound that are cleaved in the body by endogenous nucleases to form the active compound. In certain embodiments, the compound or composition further comprises a pharmaceutically acceptable carrier or diluent.
Examples
The following examples describe methods for identifying lead compounds that target AGT. Certain compounds are believed to have high potency and tolerability.
The following examples are merely illustrative of the compounds described herein and are not intended to be limiting. The following examples and related sequence listings attached to this document may identify the sequence as "RNA" or "DNA"; however, as disclosed herein, these sequences may be modified with any combination of chemical modifications. Those skilled in the art will readily appreciate that in some cases, designating a sequence as "RNA" or "DNA" is arbitrary. For example, an oligonucleotide comprising a nucleoside comprising a 2' -OH sugar moiety and a thymine base can be described as a DNA with a modified sugar (2 ' -OH for the natural 2' -H of DNA) or an RNA with a modified base (methylated uracil for the natural uracil of RNA). Thus, the nucleic acid sequences provided herein, including but not limited to the nucleic acid sequences in the sequence listing, are intended to encompass nucleic acids containing any combination of natural or modified RNAs and/or DNAs, including but not limited to such nucleic acids having modified nucleobases.
Each reference cited in the present application is incorporated herein by reference in its entirety.
TABLE 1
Chemical nomenclature
Example 1 inhibition of AGT in HepG2 cells
HepG2 cells (ATCC Cat#HB-8065) were seeded at 10,000 cells/well in 96 well plates in antibiotic free medium. The next day, the test AGT compound was diluted to 1.0 μm (stock solution 10 μm). Transfection mixtures were prepared according to the instructions of the Dharmacon Cat#T-2004-0 transfection reagent of Dharmacon 4. The prepared transfection mixture was incubated for 20 min at room temperature. During this incubation, the medium in the 96-well plate was replaced with 80 μl of antibiotic-free medium. Mu.l of transfection mixture was then added to each well at a final concentration of 10nM (tested in triplicate). AGT siRNA SMARTpool (Dharacon Cat#L-010988-00-0005) was used as a positive control. The cells were then incubated in 5% CO 2 at 37℃for 24 hours.
Cell lysis was performed according To the instructions of the Cells-To-CT 1 Step TaqMan kit. After cell lysis, 96-well plates were placed on ice while preparing qRT-PCR reactions. Mu.l of cell lysate was added to a reaction mixture (0.2 mL) containing 5. Mu.l of TaqMan 1-Step qRT-PCR mixture, 1. Mu.l of AGT (FAM) TaqMan gene expression assay (Hs 01586213 _m1), 1. Mu.l of GAPDH (VIC) TaqMan gene expression assay (Hs 02786624 _g1) and 11. Mu.l of RT-PCR grade nuclease-free water in a MicroAmp optical 96-well plate. qPCR was performed using QuantStudio qPCR machine with the following cycles: 50℃for 1 minute, 95℃for 20 seconds, and 40 cycles at 95℃for 15 seconds and 60℃for 1 minute. The results are expressed as percentage inhibition of AGT in the table below relative to untreated control cells. Unless otherwise indicated in the separate chemical tables below, the compounds are unmodified. Abbreviations for chemical modifications are provided in table 1 above. Ref ID NO: IA and IS in (a) represent the antisense and sense strands of the compound, respectively.
TABLE 2
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 3 Table 3
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 4 Table 4
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 5
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 6
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 7
Chemical compound chemistry
TABLE 8
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 9
Chemical compound chemistry
Table 10
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 11
Chemical compound chemistry
Table 12
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 13
Chemical compound chemistry
TABLE 14
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 15
Chemical compound chemistry
Table 16
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 17
Chemical compound chemistry
TABLE 18
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
TABLE 19
Chemical compound chemistry
Table 20
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
Table 21
Chemical compound chemistry
Table 22
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
Table 23
Chemical compound chemistry
Table 24
Targeting SEQ ID NO:1 inhibition of AGT mRNA by double-chain Compounds
Table 25
Chemical compound chemistry
Example 2: use of compounds targeting human AGT in cynomolgus monkeys
The compounds of interest identified in the in vitro gene expression screen were evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 6 to 8.5 years old and the body weight was 3.7 to 6.8kg. 3 cynomolgus monkeys in each of the 2 groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide (1 monkey) or two 3mg/kg subcutaneous doses of oligonucleotide (5 monkeys). The first dose was on study day 1 and the second dose was on day 17. During the study period, monkeys were observed daily for signs of disease or distress. Animals were bled for serum collection and analysis on day-6 and day 1 (prior to dosing), day 4, day 8, day 15, day 25, day 32, day 39, day 53 and day 60. The described protocol was approved by the Institutional animal care and Use Committee (Institutional ANIMAL CARE AND Use Committee, IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (day 1 prior to dosing) and as group mean for each compound.
Table 26
Average AGT inhibition
Example 3: use of compounds targeting human AGT in cynomolgus monkeys
The compounds of interest identified in the in vitro gene expression screen were evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 3.5 to 4 years, and the body weight was 3.1 to 4.6kg. On study day 1, 3 cynomolgus monkeys in each of the three groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide. During the study period, monkeys were observed daily for signs of disease or distress. Animals were bled for serum analysis on day-6 and day 1 (prior to dosing), day 4, day 8, day 15, day 22, day 29, day 36 and day 43. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (day 1 prior to dosing) and as group mean for each compound.
Table 27
Average AGT inhibition
Compounds of formula (I) Day 15 Day 22
RD1036 12 0
RD1046 15 0
Table 28
Average AGT inhibition
Example 4: use of compounds targeting human AGT in cynomolgus monkeys
The compounds of interest identified in the in vitro gene expression screen were evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 2.6 to 5.3 years old and the body weight was 2.2 to 4.4kg. On study day 1,2 cynomolgus monkeys in each of the 8 groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide. Animals were bled for serum analysis on day-6 and day 1 (prior to dosing), day 4, day 8, day 15, day 22, day 29, day 36 and day 43. One group (RD 1354) was injected with the second dose of 3mg/kg on day 43 and the study lasted for a total of 92 days. These animals were then exsanguinated on days 50, 57, 64, 71, 78, 85 and 92. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (day 1 prior to dosing) and as group mean for each compound. Clinical chemistry was performed on day 43 or day 92 (RD 1354 group). There was no related effect of the test article on body weight, and all serum chemistries were within the reference range.
TABLE 29 average AGT inhibition
Compounds of formula (I) Day 15
RD1324 0
RD1355 0
RD1356 16
RD1377 15
RD1378 25
RD1379 7
RD1380 24
Table 30
Average AGT inhibition
Table 31
Body weight (kg)
Compounds of formula (I) Day 1 Day 35 Day 92
RD1324 Cyno#1 2.6 3.0
RD1324 Cyno#2 3.4 3.4
RD1354 Cyno#1 3.2 3.4 3.3
RD1354 Cyno#2 2.7 2.8 2.8
RD1355 Cyno#1 2.4 2.8
RD1355 Cyno#2 2.6 2.8
RD1356 Cyno#1 3.2 3.4
RD1356 Cyno#2 3.1 3.2
RD1377 Cyno#1 3.3 4.0
RD1377 Cyno#2 2.6 3.1
RD1378 Cyno#1 2.5 2.8
RD1378 Cyno#2 2.7 2.8
RD1379 Cyno#1 3.1 3.2
RD1379 Cyno#2 4.4 4.8
RD1380 Cyno#1 2.3 2.4
RD 1380 Cyno#2 2.4 2.4
Table 32
Liver function marker
Table 33
Renal function markers
Example 5: use of compounds targeting human AGT in cynomolgus monkeys
The compounds of interest identified in the in vitro gene expression screen were evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 2.3 to 3.1 years old and the body weight was 1.8 to 2.4kg. On study days 1 and 22, 2 cynomolgus monkeys per group 8 were injected with 3mg/kg of the subcutaneous dose of oligonucleotide. All animals were bled for serum analysis on day-6 and day 1 (prior to dosing), day 4, day 8, day 14, day 22, day 29, day 36, day 43, day 50 and day 57. Four of the groups (RD 1614, RD1615, RD1623, and RD 1625) remained for an additional 35 days (92 days total) in the study, and were subsequently exsanguinated on days 64, 71, 78, 85, and 92. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (day 1 prior to dosing) and as group mean for each compound. Clinical chemistry was performed on days-6 and 57 (RD 1613, RD1622, RD1624 and RD 1626) or 92 (RD 1614, RD1615, RD1623 and RD 1625). There was no related effect of the test article on body weight, and all serum chemistry values were generally within the reference range.
Watch 34
Average AGT inhibition
Compounds of formula (I) Day 14 Day 22
RD1613 42 51
RD1614 66 70
RD1615 81 88
RD1622 39 44
RD1624 50 48
RD1626 14 36
Table 35
Average AGT inhibition
Table 36
Body weight (kg)
Compounds of formula (I) Day 1 Day 57 Day 92
RD1613 Cyno#1 2.1 2.3
RD1613 Cyno#2 2.2 2.4
RD1614 Cyno#1 2.6 2.8 2.8
RD1614 Cyno#2 1.9 2.1 1.9
RD1615 Cyno#1 2.1 2.6 2.1
RD1615 Cyno#2 2.1 2.4 2.1
RD1622 Cyno#1 1.8 2.0
RD1622 Cyno#2 2.1 2.3
RD1623 Cyno#1 2.4 2.6 2.5
RD1623 Cyno#2 2.1 2.3 2.1
RD1624 Cyno#1 2.1 2.2
RD1624 Cyno#2 1.7 1.9
RD1625 Cyno#1 2.0 2.2 1.9
RD1625 Cyno#2 2.1 2.3 2.1
RD1626 Cyno#1 2.1 2.3
RD1626 Cyno#2 1.7 1.0
Table 37
Liver function marker
Table 38
Liver function marker
Table 39
Renal function markers
Example 6: use of compounds targeting human AGT in cynomolgus monkeys
The compound of interest was evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 3.86 to 4.28 years old and the body weight was 2.98 to 3.92kg. On study day 1, 2 cynomolgus monkeys in each of the two groups were injected with a single 5mg/kg subcutaneous dose of oligonucleotide. All animals were bled for serum analysis on day-8 and day 1 (prior to dosing), day 4, day 8, day 15, day 22, day 27, day 36, day 43, day 50 and day 57. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (day 1 prior to dosing) and as group mean for each compound. There was no related effect of the test article on body weight.
Table 40
Average AGT inhibition
Table 41
Body weight (kg)
Compounds of formula (I) Day 1 Day 57
RD1623 Cyno#1 3.76 3.83
RD1623 Cyno#2 2.98 3.15
RD1623 Cyno#3 3.92 4.54
RD1625 Cyno#1 3.40 3.44
RD1625 Cyno#2 3.22 3.32
RD1625 Cyno#3 2.98 3.16
Example 7: use of compounds targeting human AGT in cynomolgus monkeys
The compound of interest was evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 3 to 5.6 years old and the body weight was 2.8 to 3.4kg. On study day 1, 2 cynomolgus monkeys in each of the two groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide. All animals were bled for serum analysis on day-6 as well as day 1 (prior to dosing), day 7, day 14, day 21, day 28, day 36 and day 42. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (day 1 prior to dosing) and as group mean for each compound. There was no related effect of the test article on body weight.
Table 42
Chemical compound chemistry
Table 43
Average AGT inhibition
Table 44
Body weight (kg)
Compounds of formula (I) Day 1 Day 42
RD2052 Cyno#1 3.0 3.4
RD2052 Cyno#2 2.8 2.9
RD2053 Cyno#1 3.1 3.4
RD2053 Cyno#2 3.4 3.4
Example 8: role of modified oligonucleotides targeting human AGT in cynomolgus monkeys
The compound of interest was evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 2.9 to 6.1 years old and the body weight was 2.4 to 3.5kg. On study day 1,2 cynomolgus monkeys in each of the three groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide. All animals were bled for serum analysis on day-6 as well as day 1 (prior to dosing), day 4, day 8, day 15, day 22, day 29, day 36, day 43, day 50, day 57 and day 64. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (mean of day-6 and day 1 prior to dosing) and as group mean for each compound. Clinical chemistry was performed on day-6 and day 64. There was no related effect of the test article on body weight, and all serum chemistry values were generally within the reference range.
Table 45
Chemical compound chemistry
Watch 46
Average AGT inhibition
Table 47
Body weight (kg)
Compounds of formula (I) Day 1 Day 63
RD2263 Cyno#1 3.5 3.4
RD2263 Cyno#2 3.0 3.1
RD2264 Cyno#1 3.0 2.8
RD2264 Cyno#2 3.0 3.2
RD2265 Cyno#1 2.6 2.7
RD2265 Cyno#2 2.4 2.5
Table 48
Liver function marker
Table 49
Renal function markers
Example 9: role of modified oligonucleotides targeting human AGT in cynomolgus monkeys
The compound of interest was evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 2.9 to 6.1 years old and the body weight was 2.4 to 3.5kg. On study day 1,2 cynomolgus monkeys in each of the two groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide. All animals were scheduled for exsanguination for serum analysis on day-6 and day 1 (prior to dosing), day 8, day 15, day 22, day 29, day 36, day 43, day 50, day 57 and day 64. The study ended after day 29. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (mean of day-6 and day 1 prior to dosing) and as group mean for each compound. Clinical chemistry was scheduled to proceed on day-6 and day 64.
Table 50
Chemical compound chemistry
Table 51
Average AGT inhibition
Example 10: role of modified oligonucleotides targeting human AGT in cynomolgus monkeys
The compound of interest was evaluated in cynomolgus monkeys. The monkeys were kept isolated prior to the study during which time the animals were observed daily for general health. The age of the monkeys was 2.9 to 6.1 years old and the body weight was 2.4 to 3.5kg. On study day 1,2 cynomolgus monkeys in each of the two groups were injected with a single 3mg/kg subcutaneous dose of oligonucleotide. All animals were scheduled to be bled for serum analysis on day-6 as well as day 1 (prior to dosing), day 4, day 8, day 15, day 22, day 29, day 36, day 43, day 50, day 57 and day 64. The study ended after day 43. The described protocol was approved by the Institutional Animal Care and Use Committee (IACUC). Circulating AGT levels were quantified using ELISA specific for human angiotensinogen (and cross-reactive with cynomolgus monkey) according to the manufacturer's protocol (IBL us # 27412). AGT inhibition data are expressed as a percentage of baseline values (mean of day-6 and day 1 prior to dosing) and as group mean for each compound. Clinical chemistry was scheduled to proceed on day-6 and day 64.
Watch 52
Chemical compound chemistry
Table 53
Average AGT inhibition

Claims (66)

1. A compound comprising a modified oligonucleotide of 14 to 23 linked nucleosides in length having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20 consecutive nucleobases of any one of 10 to 166.
2. A compound comprising a modified oligonucleotide of 14 to 23 linked nucleosides in length having a sequence comprising SEQ ID NO:10 to 166, a core of any one of nucleobase sequence of the base sequence.
3. A compound comprising a modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 in nucleobase sequence the nucleobase sequence of either.
4. The compound of any one of claims 1 to 3, wherein the nucleobase sequence of the modified oligonucleotide hybridizes to SEQ ID NO:1 or 3, at least 80%, at least 85%, at least 90% or at least 95% complementary.
5. The compound of any one of claims 1 to 4, wherein the modified oligonucleotide comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase.
6. The compound of any one of claims 1 to 5, wherein the compound is double-stranded.
7. A compound comprising: a first modified oligonucleotide of 14 to 23 linked nucleosides in length having a sequence comprising SEQ ID NO: a nucleobase sequence of at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least 20, at least 21, at least 22, at least 23 consecutive nucleobases of any one of 10 to 166 and 167 to 327; and a second modified oligonucleotide of 14 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide.
8. A compound comprising: a first modified oligonucleotide of 14 to 23 linked nucleosides in length having a sequence comprising SEQ ID NO:10 to 166 or 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide of 14 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide.
9. A compound comprising: a first modified oligonucleotide having a sequence selected from the group consisting of SEQ ID NOs: 10 to 166 and 167 to 327 nucleobase sequence of the nucleobase sequence; and a second modified oligonucleotide 19 to 23 linked nucleosides in length having a region complementary to the first modified oligonucleotide.
10. The compound of any one of claims 7 to 9, wherein the nucleobase sequence of the first modified oligonucleotide hybridizes over its length to the nucleobase sequence of SEQ ID NO:1 or 3 has at least 80%, at least 85%, at least 90% or at least 95% complementarity or identity.
11. The compound of any one of claims 7 to 10, wherein the nucleobase sequence of the first modified oligonucleotide hybridizes to SEQ ID NO:1 or 3, said region having at least 1, at least 2, at least 3 mismatches with said first modified oligonucleotide.
12. The compound of any one of claims 7 to 11, wherein the length of the region of complementarity between the first modified oligonucleotide and the second modified oligonucleotide is 14 to 23 linked nucleosides.
13. The compound of any one of claims 7 to 11, wherein the length of the region of complementarity between the first modified oligonucleotide and the second modified oligonucleotide is 19 to 23 linked nucleosides.
14. The compound of any one of claims 7 to 11, wherein the length of the region of complementarity between the first modified oligonucleotide and the second modified oligonucleotide is 21 to 23 linked nucleosides.
15. The compound of any one of claims 7 to 11, wherein the first modified oligonucleotide is fully complementary to the second modified oligonucleotide.
16. The compound of any one of claims 7 to 15, wherein the first modified oligonucleotide comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase.
17. The compound of any one of claims 7 to 16, wherein the second modified oligonucleotide comprises at least one modification selected from the group consisting of a modified internucleoside linkage, a modified sugar, and a modified nucleobase.
18. The compound of any one of claims 5, 16 and 17, wherein the modified internucleoside linkage is a phosphorothioate internucleoside linkage or a methylphosphonate internucleoside linkage.
19. The compound of claim 18, wherein the phosphorothioate internucleoside linkage or methylphosphonate internucleoside linkage is at the 3 'end of the first or second modified oligonucleotide or at the 5' end of the first modified oligonucleotide.
20. The compound of any one of claims 5, 16 and 17, wherein the modified sugar comprises a modification selected from the group consisting of halogen, alkoxy, and bicyclic sugar.
21. The compound of claim 20, wherein the modified sugar comprises a 2' -F modification.
22. The compound of claim 20, wherein the modified sugar comprises a 2' -OMe modification.
23. The compound of any one of claims 7 to 15, wherein each nucleoside of the first modified oligonucleotide comprises a modified sugar.
24. The compound of any one of claims 7 to 15, wherein each nucleoside of the second modified oligonucleotide comprises a modified sugar.
25. The compound of claim 23 or 24, wherein the modified sugar comprises a modification selected from the group consisting of: halogen, alkoxy, and bicyclic sugar, or combinations thereof.
26. The compound of claim 25, wherein the modified sugar comprises a modification-LNA, cEt, 2'-MOE, 2' -F, 2'-OMe and 2' -deoxy, or a combination thereof, selected from the group consisting of.
27. The compound of claim 23, wherein the first modified oligonucleotide comprises no more than ten 2' -F sugar modifications.
28. The compound of claim 24, wherein the second modified oligonucleotide comprises no more than five 2' -F sugar modifications.
29. The compound of any one of the preceding claims, comprising a conjugate group.
30. The compound of claim 29, wherein the conjugate group is attached to the 5' end of the modified oligonucleotide.
31. The compound of claim 29 or 30, wherein the conjugate group comprises a targeting moiety.
32. The compound of claim 31, wherein the targeting moiety comprises one or more galnacs.
33. The compound of claim 32, wherein the modified oligonucleotide is the second modified oligonucleotide.
34. The compound of claim 32 or 33, wherein the one or more galnacs are linked to the 2' or 3' position of the ribosyl ring of the 5' nucleoside of the modified oligonucleotide.
35. The compound of claim 34, wherein the 5' nucleoside has the formula:
Wherein:
R 9 is H, adenine, guanine, thymine, cytosine, or uracil, or is adenine, guanine, thymine, cytosine, or uracil, each comprising a Protecting Group (PG), a modified nucleobase, an optionally substituted alkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted carbocyclyl, an optionally substituted heterocyclyl, or a nucleobase isostere;
L is a bond, phosphodiester bond, phosphorothioate bond, triazole, tetrazole, amide, anti-amide, carbamate, carbonate, urea, alkyl or heteroalkyl;
R 2 is the oligonucleotide sequence;
Y 1 is O, CH 2、CH2 O or optionally substituted NH;
Y 2 is O, CH 2、CH2 O or optionally substituted NH;
y 3 is CO, SO 2、P(O)O、CH2-O-C(O)、CH2-NH-C(O)、CH2-NH-SO2 or CH 2;
Y 4 is CO, SO 2、P(O)O、CH2-O-C(O)、CH2-NH-C(O)、CH2-NH-SO2 or CH 2;
n 2 is 0, 1,2,3, 4,5, or 6; and
N 1、n3、n4 and n 5 are each independently 0, 1,2,3,4, 5, 6, 7, 8, 9 or 10.
36. The compound of claim 34, wherein the 5' nucleoside is selected from any one of formulas I-VIII, and wherein R ' is S and R is part of the modified oligonucleotide other than the 5' nucleoside.
37. The compound of claim 34, wherein the 5' nucleoside is selected from any one of formulas I-VIII, and wherein R ' is O and R is part of the modified oligonucleotide other than the 5' nucleoside.
38. A compound comprising: a first modified oligonucleotide selected from any one of :Ref ID NO:IA0297,IA0300,IA0301,IA0304,IA0305,IA0335-338,IA0343-359,IA0431-432,IA0435,IA440-446,IA0727-728,IA0500-501, and IA0868; and a second modified oligonucleotide of 14 to 21 linked nucleosides in length that is fully complementary to the first modified oligonucleotide.
39. A compound comprising a peptide selected from the group consisting of Ref ID NO: a first modified oligonucleotide of IA0443 and IA0445, and a second oligonucleotide selected from Ref ID NO: second modified oligonucleotides of IS0505 and IS 0509.
40. The compound of any one of claims 1 to 39, wherein the compound is in the form of a pharmaceutically acceptable salt.
41. The compound of claim 40, wherein the pharmaceutically acceptable salt is a sodium salt.
42. The compound of claim 40, wherein the pharmaceutically acceptable salt is a potassium salt.
43. A composition comprising a compound of any one of claims 1 to 42 and a pharmaceutically acceptable carrier.
44. A composition comprising a compound of any one of the preceding claims for use in therapy.
45. A method of treating, preventing or ameliorating a disease, disorder or condition associated with AGT in a subject, comprising administering to the subject a compound that targets AGT, thereby treating, preventing or ameliorating the disease, disorder or condition.
46. A method of administering a compound of any one of claims 1 to 42 or a composition of claim 43 or 44 to an individual.
47. The method of claim 45 or 46, wherein the disease, disorder or condition is RAAS-related disease, disorder or condition or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
48. The method of any one of claims 45 to 47, wherein administration of the compound inhibits or reduces or ameliorates RAAS-related diseases, disorders or conditions or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
49. A method of inhibiting AGT expression in a cell comprising contacting the cell with a compound that targets AGT, thereby inhibiting AGT expression in the cell.
50. The method of claim 49, wherein the cell is in the liver of the subject.
51. The method of claim 50, wherein the individual has or is at risk of having: RAAS-related diseases, disorders or conditions or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
52. A method of reducing or inhibiting RAAS-related diseases, disorders or conditions, or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in a subject, comprising administering to the subject a compound that targets AGT, thereby reducing or inhibiting RAAS-related diseases, disorders, or conditions, or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment in the subject.
53. The method of claim 52, wherein the individual has or is at risk of having: RAAS-related diseases, disorders or conditions or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ damage (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
54. The method of any one of claims 45 to 53, wherein the compound is an AGT-targeting compound.
55. The method of any one of claims 45 to 54, wherein the compound is a compound of any one of claims 1 to 42 or a composition of claim 43 or 44.
56. The method of claim 55, wherein the compound or composition is administered parenterally.
57. Use of a compound targeting AGT for the treatment, prevention or amelioration of a disease, disorder or condition associated with AGT.
58. The use of claim 57, wherein the disease, disorder or condition is RAAS-related disease, disorder or condition or symptoms thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysms, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
59. The use of claim 57 or 58, wherein the compound is an AGT-targeting compound.
60. The use of any one of claims 57 to 59, wherein the compound is a compound of any one of claims 1 to 42 or a composition of claim 43 or 44.
61. Use of a compound targeting AGT for the manufacture of a medicament for the treatment, prevention or amelioration of a disease, disorder or condition associated with AGT.
62. The use of claim 61, wherein the disease is a RAAS-related disease, disorder or condition or symptom thereof, hypertension, refractory hypertension, fibrosis, kidney disease, chronic kidney disease, cardiovascular disease (e.g., coronary heart disease, heart failure, stroke, myocardial infarction, atrial fibrillation, aneurysm, and peripheral arterial disease), organ injury (e.g., heart, liver, or kidney), inflammatory bowel disease, or cognitive impairment.
63. The use of claim 61 or 62, wherein the compound is an AGT-targeting compound.
64. The use of any one of claims 61 to 63, wherein the compound is a compound of any one of claims 1 to 42 or a composition of claim 43 or 44.
65. The method or use of any one of the preceding claims, wherein the compound or composition is administered to the individual from about once every three months to about once a year.
66. The method or use of any one of the preceding claims, wherein the compound or composition is administered to the individual about once every three months, about once every six months, or about once every year.
CN202280066569.0A 2021-10-01 2022-09-30 Renin modulating compositions and methods of use thereof Pending CN118786133A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/251,562 2021-10-01
US202163287960P 2021-12-09 2021-12-09
US63/287,960 2021-12-09
PCT/US2022/077389 WO2023056446A1 (en) 2021-10-01 2022-09-30 Angiotensinogen-modulating compositions and methods of use thereof

Publications (1)

Publication Number Publication Date
CN118786133A true CN118786133A (en) 2024-10-15

Family

ID=92996043

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280066569.0A Pending CN118786133A (en) 2021-10-01 2022-09-30 Renin modulating compositions and methods of use thereof

Country Status (1)

Country Link
CN (1) CN118786133A (en)

Similar Documents

Publication Publication Date Title
JP6877414B2 (en) KRAS expression modulator
CN113444725B (en) Compositions and methods for inhibiting alpha-1 antitrypsin gene expression
JP2019500346A (en) Compositions and methods for the treatment of kidney disease
US20220042022A1 (en) Antisense oligonucleotides for modulating htra1 expression
KR20230012508A (en) Bifunctional Molecules and Methods of Their Use
CA3088522A1 (en) Modulators of dnm2 expression
CN118786133A (en) Renin modulating compositions and methods of use thereof
US7393950B2 (en) Antisense oligonucleotides targeted to human CDC45
CN118922540A (en) Prekallikrein modulation compositions and methods of use thereof
EP4408525A1 (en) Angiotensinogen-modulating compositions and methods of use thereof
TW202405171A (en) Angiotensinogen-modulating compositions and methods of use thereof
WO2020136125A2 (en) Antisense oligonucleotides targeting card9
US12042509B2 (en) Prekallikrein-modulating compositions and methods of use thereof
TW202340463A (en) Complement factor b-modulating compositions and methods of use thereof
US20080300212A1 (en) Treatment and prevention of hyperproliferative conditions in humans and antisense oligonucleotide inhibition of human replication-initiation proteins
JP2024541771A (en) COMPOSITIONS THAT MODULATE COMPLEMENT FACTOR B AND METHODS OF USE THEREOF
WO2024220930A2 (en) Mapt-modulating compositions and methods of use thereof
WO2020011745A2 (en) Antisense oligonucleotides targeting cers6
WO2020011744A2 (en) Antisense oligonucleotides targeting cers5
KR20240139057A (en) Composition and method for inhibiting expression of prekallikrein (PKK) protein
WO2020007772A1 (en) Antisense oligonucleotides targeting gbp-1
WO2020011653A1 (en) Antisense oligonucleotides targeting kynu
WO2020038971A1 (en) Antisense oligonucleotides targeting vcan
WO2020089260A1 (en) Antisense oligonucleotides targeting tia1

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination