专利摘要:
The present invention relates to compounds, compositions and methods for the treatment of viral infections, diseases, syndromes or disorders that are affected by sting modulation. such compounds are represented by the following formula i: formula (i) wherein r1a, r1b, r1c, b1, r2a, and r2b are as defined herein.
公开号:BR112019010606A2
申请号:R112019010606
申请日:2017-11-21
公开日:2019-09-17
发明作者:C Bignan Gilles;Wang Guangyi;patrick edwards James;Beigelman Leonid;Tianbao Lu;Richter Mark;Zhong Minghong;Connolly Peter;Kumar Thatikonda Santhosh;Emanuel Stuart;Laquerre Sylvie
申请人:Janssen Biotech Inc;
IPC主号:
专利说明:

Invention Patent Descriptive Report for CYCLIC DYNUCLEOTIDS AS STING AGONISTS.
CROSS REFERENCE TO RELATED APPLICATIONS [0001] This application claims priority to US Provisional Patent Application No. 62 / 426,350, filed on November 25, 2016; US Provisional Patent Application No. 62 / 502,983, filed on May 8, 2017; US Provisional Patent Application No. 62 / 555,232, deposited on September 7, 2017; that are incorporated here, by reference in their entirety.
FIELD OF THE INVENTION [0002] The present invention relates to new compounds that are STING agonists (Interferon Gene Stimulator) and are useful for the treatment of disorders that are affected by the modulation of the STING protein. The invention also relates to pharmaceutical compositions that comprise one or more of such compounds, processes for preparing such compounds and compositions, and use of such compounds or pharmaceutical compositions for the treatment of various diseases, syndromes and disorders. The invention may be involved in the activation of the downstream signaling pathway, additionally resulting in the activation of second messengers and growth factors, and in the production of interferon involved in innate and adaptive immunity. More particularly, the present invention relates to the use of such compounds or pharmaceutical compositions for the treatment of various infections, diseases, syndromes and disorders including, but not limited to, melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma and antiviral therapy.
BACKGROUND OF THE INVENTION [0003] STING (interferon gene stimulator), also known as TMEM173, MITA, MPYS, and ERIS, is a transmembrane receptor located within the cell and a sensor
Petition 870190060374, of 06/28/2019, p. 5/142
2/122 cytosolic nucleic acids (Zhong B, et al. The Adapter Protein MITA Links Virus-Sensing Receptors to IRF3 Transcription Factor Activation. Immunity. 2008. vol. 29: 538 (-550); Recent studies have revealed the biology of STING and its role in mobilizing an innate immune response resulting in robust antitumor activity in mouse models.The activation of the STING pathway results in the production of Type I interferons (mainly IFN-α and IFN-β) induced through the IRF3 pathway (interferon regulating factor 3.) IRF3 activation is believed to be mediated by TBK1 which recruits and phosphorylates IRF3, thus forming an IRF3 homodimer capable of entering the nucleus to transcribe type I interferon and other genes (Liu S , et al. Phosphorylation of innate immune adapter proteins MAVS, STING, and TRIF induces IRF3 activation Science. 2015: 26302637). TBK1 also activates the kappa light chain enhancer of the nuclear factor of the activated B cell pathway, which leads to the pro production of pro-inflammatory cytokines (IL-1a, IL-Ιβ, IL-2, IL-6, TNF-a, etc.), through the oncogenic transcription factor NF-κΒ. In addition, STING activates STAT6 (signal transducer and transcription activator 6) to induce (type Th2), increase (IL-12) or decrease (IL-10) the production of various cytokines, including chemokines CCL2, CCL20, and CCL26 (Chen H, et al. Activation of STAT6 by STING Is Critical for Antiviral Innate Immunity Cell. 2011, vol.14: 433-446). It has also been reported that direct STING phosphorylation in Ser366 after activation occurs via TBK1 (Corrales, L. et al Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity Cell Reports, 2015, vol.11 : 1-13; Konno, H. et al. Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling Cell, 2013, vol. 155: 688698).
[0004] The natural ligand that binds and activates cyclic STING (2 ', 3')
Petition 870190060374, of 06/28/2019, p. 6/142
3/122 guanosine monophosphate-adenosine monophosphate (2 ', 3'-cGAMP) and the enzyme responsible for its synthesis (cGAs, also known as C6orf150 or MB21D1) have been elucidated, providing an opportunity to modulate this pathway. cGAMP is a high affinity ligand for STING produced in mammalian cells, which serves as a second endogenous messenger to activate the STING pathway. It is a cyclic dinucleotide with a unique 2 ', 3' bond produced by cGAS in the presence of exogenous double-stranded DNA (for example, that released by invading bacteria, viruses or protozoa) or auto-DNA in mammals (Wu et al ., 2013; Sun, L. et al. Cyclic GMP-AMP Synthase Is a Cytosolic DNA Sensor That Activates the Type I Interferon Pathway Science, 2013, vol. 339: 786-791; Bhat N and Fitzgerald KA. Recognition of Cytosolic DNA by cGAS and other STING-dependent sensors. Eur J Immunol, March 2014; 44 (3): 634-40). The activation of STING can also occur through the binding of exogenous cyclic (3 ', 3) dinucleotides (c-di-GMP, c-diAMP and 3'3'-cGAMP) that are released by invading bacteria (Zhang X, et al Cyclic GMP-AMP Containing Mixed Phosphodiester Linkages Is An Endogenous High-Affinity Ligand for STING Molecular Cell, 2013, vol. 51: 226-235; Danilchanka, O and Mekalanos, JJ. Cyclic Dinucleotides and the Innate Immune Response Cell. 2013. vol. 154: 962-970).
[0005] Activation of the STING pathway triggers an immune response that results in the generation of specific killer T cells that can shrink tumors and provide long-lasting immunity so that they do not reappear. The impressive antitumor activity obtained with STING agonists in preclinical models has generated a high level of excitement for this target and small molecule compounds that can modulate the STING pathway have the potential to treat both cancer and reduce autoimmune diseases.
[0006] The activation of the STING pathway also contributes to an antiviral response. The loss of function response, whether at the cellular level
Petition 870190060374, of 06/28/2019, p. 7/142
4/122 or organism, demonstrates an inability to control viral load in the absence of STING. Activation of the STING pathway triggers an immune response that results in antiviral and proinflammatory cytokines that fight viruses and mobilize the innate and adaptive arms of the immune system. Finally, long-term immunity is developed against the pathogenic virus. The surprising antiviral activity obtained with STING agonists in preclinical models has generated a high level of excitement for this target and small molecule compounds that can modulate the STING pathway have the potential to treat chronic viral infections, such as hepatitis B.
[0007] Hepatitis B virus (HBV) infection is a major global health problem, affecting more than 5% of the world's population (over 350 million people worldwide and 1.25 million individuals in the USA ). Despite the availability of certain vaccines and therapies against HBV, the burden of chronic HBV infection remains a significant unmet worldwide medical problem due to suboptimal treatment options and sustained rates of new infections in most parts of the developing world. Current treatments are limited to only two classes of agents: alpha interferon and nucleoside analogues that act as viral polymerase inhibitors. However, none of these therapies offers a cure for the disease, and problems with drug resistance, poor efficacy and tolerability limit their impact. The low cure rates for HBV are attributed, at least in part, to the fact that complete suppression of viral production is difficult to achieve with a single antiviral agent. However, persistent suppression of HBV DNA slows the progression of liver disease and helps prevent hepatocellular carcinoma. Current therapy goals for HBV-infected patients are aimed at reducing serum HBV DNA levels to low or undetectable levels, and ultimately to reducing or preventing the development of
Petition 870190060374, of 06/28/2019, p. 8/142
5/122 cirrhosis and hepatocellular carcinoma. There is, therefore, a need in the art for therapeutic agents that can increase suppression of viral production and that can treat, improve, or prevent HBV infection. The administration of such therapeutic agents to an HBV-infected patient, either as monotherapy or in combination with other treatments for HBV or auxiliary treatments, can lead to a significant reduction in viral load, better prognosis, decreased disease progression and intensification of seroconversion rates.
[0008] The potential therapeutic benefits of enhancing innate and adaptive immunity make STING an attractive therapeutic target that demonstrates impressive activity on its own and can also be combined with other immunotherapies.
SUMMARY OF THE INVENTION [0009] The present invention relates to compounds of Formula (I)
Formula (I) where:
Ria is hydroxy or fluorine and Ric is hydrogen; or, Ria is -O- and Ric is CH 2 so that Ria and Ric are taken together with the atoms to which they are attached to form a 5-membered ring;
Rib is selected from the group consisting of hydroxy, thiol and BH 3 -;
Bi is selected from the group consisting of rings b1 and b2
Petition 870190060374, of 06/28/2019, p. 9/142
6/122 ο νη 2
ΟΤ lb 1
Ν ^ Ν ^ ΝΗ ;, n n> b1 and b2;
R2A is selected from the group consisting of hydroxy and methoxy;
R2B is selected from the group consisting of hydroxy, thiol and BH 3 -;
provided that the compound of Formula (I) is different from the salt of (1,, 6 í, 8 í, 9 í, 10 í, 15 í, 17 í, 18 í) -17- (2-Amino-6-oxo-6,9-di- hydro1 / 7-pu ri n-9-yl) -8- (6-ami n-9/7-pu ri n-9-yl) -9-fllu r-3,12,18-tri-hid roxy2,4,7,11,13,16-hexaoxa-3Ã 5 , 12Ã 5- diphosphatriciclo [13,2,1,0 6 , 10 ] octadecane3,12-dione, bis-ammonium;
or an enantiomer, diastereomer or pharmaceutically acceptable salt form thereof.
[0010] The present invention also provides a pharmaceutical composition comprising, consisting of and / or consisting essentially of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and / or a pharmaceutically acceptable diluent and a compound of formula (I) or a form of the pharmaceutically acceptable salt thereof.
[0011] Processes are also provided for the manufacture of a pharmaceutical composition comprising, consisting of and / or consisting essentially of mixing a compound of Formula (I) and a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and / or a diluent pharmaceutically acceptable.
[0012] The present invention further provides methods for treating or ameliorating a viral infection, disease, syndrome or condition in an individual, including a mammal, and / or human being in which the viral infection, disease, syndrome or condition is affected by
Petition 870190060374, of 06/28/2019, p. 10/142
7/122 STING agonism, with the use of a compound of Formula (I). [0013] The present invention further provides methods for treating or ameliorating a viral infection, disease, syndrome or condition in an individual, including a mammal, and / or human with the use of a compound of Formula (I).
[0014] The present invention further provides methods for treating or ameliorating a viral infection, disease, syndrome or condition in an individual, including a mammal, and / or human being in which the viral infection, disease, syndrome or condition is affected by STING's agonism, selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma, and hepatitis B, using a compound of Formula (I).
[0015] The present invention further provides methods for treating or ameliorating a viral infection, disease, syndrome or condition in an individual, including a mammal, and / or human being, selected from the group consisting of melanoma, colon cancer , breast cancer, prostate cancer, lung cancer, fibrosarcoma, and hepatitis B, with the use of a compound of Formula (I).
[0016] The present invention is also directed to the use of any of the compounds described in the present invention in the preparation of a medicament, the medicament being prepared to treat a viral infection, disease, syndrome, or condition that is affected by the agonism of STING, selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma, and hepatitis B, in an individual who needs it.
[0017] The present invention is also directed to the use of any of the compounds described in the present invention in the preparation of a medicament, the medicament being
Petition 870190060374, of 06/28/2019, p. 11/142
8/122 prepared to treat a viral infection, disease, syndrome, or condition, selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma, and hepatitis B, in an individual that needs the same. [0018] The present invention also relates to the preparation of substituted cyclic dinucleotide derivatives that act as selective STING agonists.
[0019] To exemplify the invention, methods are provided for treating a viral infection, disease, syndrome or condition modulated by STING selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma and hepatitis B, comprising administering to a subject in need thereof a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above.
[0020] To exemplify the invention, methods of treating a disease, syndrome or condition selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma, and hepatitis B are provided, comprising administering to a person in need thereof a therapeutically effective amount of any of the compounds or pharmaceutical compositions described above.
[0021] In another embodiment, the present invention is directed to a compound of Formula (I) for use in the treatment of a viral infection, disease, syndrome or condition affected by the STING agonism selected from the group consisting of melanoma, colon cancer , breast cancer, prostate cancer, lung cancer, fibrosarcoma and hepatitis B.
[0022] In another embodiment, the present invention relates to a composition that comprises a compound of Formula (I) for the
Petition 870190060374, of 06/28/2019, p. 12/142
9/122 treatment of a viral infection, disease, syndrome or condition selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma and hepatitis B.
DETAILED DESCRIPTION OF THE INVENTION [0023] With reference to the substituents, the term independently refers to the situation in which more than one substituent is possible, the substituents may be the same or different from each other.
[0024] The term alkyl, whether used alone or as part of a substituent group, refers to straight and branched carbon chains having 1 to 8 carbon atoms. Therefore, the designated numbers of carbon atoms (for example Ci-s) refer independently to the number of carbon atoms in an alkyl moiety or to the alkyl moiety of a larger alkyl-containing substituent. In substituent groups with multiple alkyl groups such as (C 1-6 alkyl-), the C 1-6 alkyl groups of the dialkylamino may be the same or different.
[0025] The term alkoxy refers to an -O-alkyl group, where the term alkyl is as defined above.
[0026] The terms alkenyl and alkynyl refer to straight and branched carbon chains that have 2 to 8 carbon atoms, where an alkenyl chain contains at least one double bond and an alkynyl chain contains at least one triple bond.
[0027] The term cycloalkyl refers to saturated or partially saturated, monocyclic or polycyclic hydrocarbon rings of 3 to 14 carbon atoms. Examples of such rings include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and adamantyl.
[0028] The term heterocyclyl refers to a non-aromatic monocyclic or bicyclic ring system that has 3 to 10 ring members that include at least 1 carbon atom and 1 to 4
Petition 870190060374, of 06/28/2019, p. 13/142
10/122 heteroatoms independently selected from N, O and S. The term heterocyclyl includes a 5- to 7-membered non-aromatic cyclic ring in which 1 to 2 members are N, or a 5- to 7-membered non-aromatic cyclic ring in which 0, 1 or 2 members are N and up to 2 members are O or S, and at least one member must be N, O or S; wherein, optionally, the ring contains 0 to 1 unsaturated bond and, optionally, when the ring is 6 or 7 members, it contains up to two unsaturated bonds. The ring members of carbon atoms that form a heterocycle ring can be either completely saturated or partially saturated.
[0029] The term heterocyclyl also includes two 5-membered monocyclic heterocycloalkyl groups bridged to form a bicyclic ring. These groups are not considered to be completely aromatic and are not referred to as heteroaryl groups. When a heterocycle is bicyclic, both rings of the heterocycle are non-aromatic and at least one of the rings contains a heteroatom ring member. Examples of heterocycle groups include, and are not limited to, pyrrolinyl (including 2/7-pyrrole, 2-pyrrolinyl or 3-pyrrolinyl), pyrrolidinyl, imidazolinyl, imidazolidinyl, pyrazolinyl, pyrazolidinyl, piperidinyl, morpholinyl, thiomorpholinyl and piperazin. Unless otherwise specified, the heterocycle is attached to its pendant group on any heteroatom or carbon atom that results in a stable structure.
[0030] The term aryl refers to an unsaturated, aromatic, monocyclic or bicyclic carbocyclic ring with 6 to 10 carbon members. Examples of aryl rings include phenyl and naphthalenyl.
[0031] The term heteroaryl refers to a bicyclic or monocyclic aromatic ring system that has 5 to 10 ring members, containing carbon atoms and 1 to 4 heteroatoms selected independently from the group consisting of N, O and S Included
Petition 870190060374, of 06/28/2019, p. 14/142
11/122 within the term heteroaryl are the 5- or 6-membered aromatic rings, where the ring consists of carbon atoms and has at least one heteroatom member. Suitable heteroatoms include nitrogen, oxygen and sulfur. In the case of 5-membered rings, the heteroaryl ring preferably contains a member of nitrogen, oxygen or sulfur and, in addition, up to three additional nitrogen atoms. In the case of 6-membered rings, the ring preferably contains one to three nitrogen atoms. For the case where the 6-membered ring has three nitrogen atoms, a maximum of two nitrogen atoms are adjacent. Examples of heteroaryl groups include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, pyridinyl, pyridazinyl, pyrimidinyl, benzazol, benzazol, indazol, benzazol, , benzoxazolyl, benzisoxazolyl, benzothiadiazolyl, benzotriazolyl, quinolinyl, isoquinolinyl and quinazolinyl. Unless otherwise specified, the heteroaryl is attached to its pendant group on any heteroatom or carbon atom that results in a stable structure.
[0032] The term halogen or halo refers to fluorine, chlorine, bromine and iodine atoms.
[0033] Whenever the term alkyl or aryl or any of its prefixed roots appears in a substituent name (for example, arylalkyl, alkylamino) the name is to be interpreted as including those limitations presented above for alkyl and aryl. The designated numbers of carbon atoms (for example, Ci-Ce) refer, independently, to the number of carbon atoms in an alkyl portion, an aryl portion or in an alkyl portion of a larger substituent in which the alkyl appears as its prefixed root. For alkyl and alkoxy substituents, the designated number of carbon atoms includes all independent members included in a
Petition 870190060374, of 06/28/2019, p. 15/142
12/122 certain specified range. For example, C1-6 alkyl would include methyl, ethyl, propyl, butyl, pentyl and hexyl individually as well as their subcombination (for example, C1-2, C1-3, C1-4, C1-5, C2-6, C3-6, C4-6, C5-6, C2-5, etc.).
[0034] In general, under the standard nomenclature rules used throughout the present disclosure, the terminal portion of the designated side chain is first described followed by the adjacent functionality related to the attachment point. Therefore, for example, a C1-6 alkyl-carbonyl substituent refers to a group of Formula:
I 11
- £ -C—— Cí-Cs aiqtiila [0035] The term R in a stereocenter designates that the stereocenter is purely of the -R configuration as defined in the art; similarly, the term S means that the stereocenter is purely of the -S configuration. As used herein, the terms * R or * S in a stereocenter are used to designate that the stereocenter has a clear, yet unknown, configuration. As used herein, the term RS refers to a stereocenter that is present as a mixture of the -R and -S configurations. Similarly, the terms * RS or * SR refer to a stereocenter that is present as a mixture of the configurations -Re -Se is of unknown configuration in relation to another stereocenter in the molecule.
[0036] Compounds containing an extracted stereocenter without a stereo bond designation consist of a mixture of two enantiomers. The compounds that contain two stereocenters both extracted without stereo bond designations consist of a mixture of four diastereomers. Compounds with two stereocenters both identified as RS and extracted with stereo bond designations consist of a mixture of two components with stereochemistry
Petition 870190060374, of 06/28/2019, p. 16/142
13/122 relative as extracted. Compounds with two stereocenters both identified as * RS and extracted with stereo bond designations consist of a mixture of two components with unknown relative stereochemistry. The unidentified stereo centers represented without stereo link designations consist of a mixture of the -R and -S configurations. For unidentified stereo centers represented with stereo link designations, absolute stereochemistry is as represented.
[0037] Except where otherwise specified, it is intended that the definition of any substituent or variable at a specific location in a molecule is independent of its definitions anywhere in that molecule. It is understood that the substituents and substitution patterns in the compounds of this invention can be selected by one skilled in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as the methods described herein .
[0038] The term individual, as used herein, refers to an animal, preferably a mammal, most preferably a human being, who has been the object of treatment, observation or experiment.
[0039] The term therapeutically effective amount refers to an amount of an active compound or pharmaceutical agent, including a compound of the present invention, that produces a biological or medical response in a tissue, animal or human system that is being sought by a researcher, veterinarian, medical doctor or other physician, including relief or partial relief from the symptoms of the disease, syndrome, condition, or disorder being treated.
[0040] The term composition refers to a product that includes
Petition 870190060374, of 06/28/2019, p. 17/142
14/122 specific ingredients in therapeutically effective amounts, as well as any product that results, directly or indirectly, from combinations of specific ingredients in the specified quantities.
[0041] The term STING agonist is intended to include a compound that interacts with STING by binding to it and that induces signal transduction characterized by the activation of molecules associated with the STING function. This includes targeting the phosphorylation of STING, IRF3 and / or NF-kB and could also include STAT6. Activation of the STING pathway results in increased production of type I interferons (mainly IFN-α and IFN-β) and in the expression of genes stimulated by interferon (Chen H, et al. Activation of STAT6 by STING Is Critical for Antiviral Innate Immunity. Cell. 2011, vol. 14: 433-446; and Liu SY, et al. Systematic identification of type I and type II interferon-induced antiviral factors. Proc. Natl. Acad. Sci. 2012: vol.109 4239 -4244).
[0042] The term modulated by STING is used to refer to a condition affected by STING directly or via the STING pathway, including, but not limited to, viral infections, diseases or conditions such as melanoma, colon cancer, cancer of breast, prostate cancer, lung cancer, fibrosarcoma and hepatitis B.
[0043] As used here, unless otherwise specified, the term STING modulated disorder means any viral infection, disease, disorder or condition characterized by the fact that at least one of its characteristic symptoms is relieved or eliminated by treatment with a STING agonist. Suitable examples include, but are not limited to, melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma and hepatitis B.
[0044] As used here, unless otherwise specified, the term affect or affected (when referring to a viral infection, disease, syndrome, condition or disorder that is affected by agonism
Petition 870190060374, of 06/28/2019, p. 18/142
15/122 of STING) includes a reduction in the frequency and / or severity of one or more symptoms or manifestations of said viral infection, disease, syndrome, condition or disorder; and / or includes preventing the development of one or more symptoms or manifestations of said viral infection, disease, syndrome, condition or disorder or the development of viral infection, disease, condition, syndrome or disorder.
[0045] The compounds of the present invention are useful in methods for treating or ameliorating a viral infection, disease, syndrome, condition or disorder that is affected by the STING agonism. These methods comprise, consist of and / or consist essentially of administration to an individual, including an animal, a mammal, and a human being in need of such treatment, relief and / or prevention, of a therapeutically effective amount of a compound of Formula ( I) as defined herein, or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof.
[0046] In particular, the compounds of Formula (I), or an enantiomer, a diastereomer, a solvate or a pharmaceutically acceptable salt form thereof, are useful for treating or alleviating diseases, syndromes, conditions or disorders such as melanoma, cancer colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma and hepatitis B.
[0047] More particularly, the compounds of Formula (I), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt form thereof are useful for treating or ameliorating melanoma, colon cancer, breast cancer, prostate cancer, cancer of lung, fibrosarcoma and hepatitis B, which comprises administering to a subject in need thereof a therapeutically effective amount of a compound of Formula (I), or an enantiomer, diastereomer, solvate or pharmaceutically acceptable salt thereof, as defined herein.
Petition 870190060374, of 06/28/2019, p. 19/142
16/122 [0048] Some modalities presented in the present invention refer to methods to improve and / or treat a viral infection, including infections caused by Hepadmarinhas, such as hepatitis B virus or HBV. The methods may include administering to an individual identified as suffering from a viral infection an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, or a pharmaceutical composition that includes one or more more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof.
[0049] Other modalities disclosed herein refer to a method for ameliorating and / or treating a viral infection, which may include bringing a cell infected with the virus into contact with an effective amount of one or more compounds described herein (for example, a compound of Formula (I), or a pharmaceutically acceptable salt form thereof), or a pharmaceutical composition that includes one or more compounds described herein, or a pharmaceutically acceptable salt thereof. Still other embodiments described herein refer to the use of one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, in the manufacture of a medicament to ameliorate and / or treat viral infection.
[0050] Still other embodiments described herein refer to one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, or a pharmaceutical composition that includes one or more compounds of Formula (I), or a form of pharmaceutically acceptable salt thereof, which can be used to ameliorate and / or treat a viral infection. Some embodiments disclosed herein refer to a method for inhibiting the replication of a virus, which may include bringing a cell infected with the virus into contact with an effective amount of one or more compounds of Formula (I), or a pharmaceutically salt form
Petition 870190060374, of 06/28/2019, p. 20/142
17/122 of the same, or a pharmaceutical composition that includes one or more compounds described herein, or a pharmaceutically acceptable salt thereof.
[0051] Other modalities described herein refer to the use of one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, in the manufacture of a medicament to inhibit the replication of a virus. Still other embodiments described herein refer to one or more compounds described herein, (for example, a compound of Formula (I), or a pharmaceutically acceptable salt form thereof), or a pharmaceutical composition that includes one or more compounds described herein. , or a pharmaceutically acceptable salt thereof, which can be used to inhibit the replication of a virus.
[0052] In some embodiments, the viral infection may be a viral infection with hepatitis B. Methods may include administering to an individual identified as having HBV an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, or a pharmaceutical composition that includes one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof.
[0053] Other modalities disclosed herein refer to a method to improve and / or treat viral infection which may include putting an HBV-infected cell in contact with an effective amount of one or more compounds of Formula (I), or a form of pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof. Still other embodiments described herein refer to the use of one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, in the manufacture of a medicament to improve and / or treat HBV.
Petition 870190060374, of 06/28/2019, p. 21/142
18/122 [0054] Still other embodiments described herein refer to one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, or a pharmaceutical composition that includes one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof, which can be used to improve and / or treat HBV. Some embodiments disclosed herein refer to a method for inhibiting HBV replication which may include bringing an infected cell with the virus into contact with an effective amount of one or more compounds of Formula (I), or a pharmaceutically acceptable salt form thereof , or a pharmaceutical composition that includes one or more compounds of Formula (I), or a pharmaceutically acceptable salt thereof.
[0055] Other modalities described herein refer to the use of one or more compounds of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament to inhibit HBV replication. Still other embodiments described herein refer to one or more compounds of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition that includes one or more compounds of Formula (I), or a pharmaceutically acceptable salt form of themselves, which can be used to inhibit HBV replication.
[0056] The embodiments of the present invention include a compound of Formula (I) as defined herein, or an enantiomer, diastereomer, solvate, or a pharmaceutically acceptable salt thereof, wherein the substituents selected from one or more of the variables herein defined (for example, Ria, Rib, Ric, Bi, R 2 a, R2b) are independently selected to be any individual substituent or any subset of substituents from those exemplified in the listing in Table 1, below.
Petition 870190060374, of 06/28/2019, p. 22/142
12/192
Table 1.
O O = pO — 1 N ^ iA N H 2 r l ± j V o 7 / VRic] --- / R2A 0 Bi --0— II R 2B o Formula (1) Compound No. Laugh Rib Rtc Bi R2A R2B 1 OCH 3 OH H b2 OH OH 2 OH OH H b2 OCH3 OH 3 OCH3 OH H b2 OCH3 OH 4 F (* R) -SH H b2 OH (* R) -SH 5 F (* S) -SH H b2 OH (* S) -SH 6 O- OH CHa to form a ring with Ria b2 OH OH 7 OH (* R) -BH 3 - H b2 OH (* R) -BH 3 - 8 OH (* S) -BH 3 - H b2 OH (* S) -BH 3 - 9 F (* R) -BH 3 - H b2 OCH3 (* R) -SH 10 F (* S) -BH 3 - H b2 OCH3 (* R) -SH 11 F (* R) -BH 3 - H b2 OCH3 (* R) -BH 3 - 12 F (* S) -BH 3 - H b2 OCH3 (* S) -BH 3 - 13 F (* R) -SH H b2 OCH3 (* R) -BH 3 - 14 F (* S) -SH H b2 OCH3 (* S) -BH 3 - 15 F (* R) -BH 3 H b2 OCH3 OH 16 F (* S) -BH 3 H b2 OCH3 OH 17 F OH H b2 OCH3 (* R) -BH 3 18 F OH H b2 OCH3 (* S) -BH 3 19 O- OH CHa to form a ring with Ria b2 OCH3 (* R) -BH 3
Petition 870190060374, of 06/28/2019, p. 23/142
12/20
O r ib <ΓΎ1 Η O = p — O— N N x N h 2 r ia ó Ud ZA R, C πη | o R 2A o Bj --O— II R 2 B 0 Formula (1) Compound No. Laugh Rib Rtc Bi R2A R2B 20 O- (* R) -SH CHs to form a ring with Ria b2 och 3 (* R) -BH 3 21 O- (* R) -BH 3 CHs to form a ring with Ria b2 och 3 (* R) -BH 3 22 O- (* R) -BH 3 CHs to form a ring with Ria b2 och 3 OH 23 O- (* R) -BH 3 CHs to form a ring with Ria b2 och 3 (* R) -SH
[0057]
One embodiment of the present invention relates to a compound of Formula (I)
Formula (I) where:
Ria is hydroxy or fluorine and Ric is hydrogen; or, Ria is -O- and
Petition 870190060374, of 06/28/2019, p. 24/142
12/21
Ric is CH 2 so that Ria and Ric are taken together with the atoms to which they are attached to form a 5-membered ring;
Rib is selected from the group consisting of hydroxy, thiol and BH 3 -;
Bi éb2
R 2 a is selected from the group consisting of hydroxy and methoxy;
R 2 b is selected from the group consisting of hydroxy, thiol and BH 3 -;
provided that the compound of Formula (I) is different from the salt of (1,, 6 í, 8 í, 9 í, 10 í, 15 í, 17 í, 18 í) -17- (2-Amino-6-oxo-6,9-di- hydro1 / -7-pu ri n-9-yl) -8- (6-ami no-9 / -7-pu ri n-9-i I) -9-f u ur-3,12,18 -trihydroxy2,4,7,11,13,16-hexaoxa-3À 5 , 12λ 5 diphosphatriciclo [13,2,1,0 6 , 10 ] octadecane-3,12-dione, bis-ammonium;
or an enantiomer, diastereomer or pharmaceutically acceptable salt form thereof.
[0058] In another embodiment, the present invention is directed to compounds of Formula (I) selected from compounds 1 to 23, the
nh 2 1
Petition 870190060374, of 06/28/2019, p. 25/142
12/22
Petition 870190060374, of 06/28/2019, p. 26/142
12/23
ΝΗ 2 6
Petition 870190060374, of 06/28/2019, p. 27/142
12/24
Petition 870190060374, of 06/28/2019, p. 28/142
12/25
νη 2 15
Petition 870190060374, of 06/28/2019, p. 29/142
12/26
Petition 870190060374, of 06/28/2019, p. 30/142
12/27
Petition 870190060374, of 06/28/2019, p. 31/142
12/28
or a pharmaceutically acceptable salt form thereof.
[0059] For use in medicaments, the salts of the compounds of Formula (I) refer to the non-toxic pharmaceutically acceptable salts. However, other salts can be useful in the preparation of compounds of Formula (I) or their pharmaceutically acceptable salt forms. Suitable pharmaceutically acceptable salts of compounds of Formula (I) include acid addition salts which can, for example, be formed by mixing a solution of the compound with a solution of a pharmaceutically acceptable acid, such as hydrochloric acid, sulfuric acid, fumaric acid , maleic acid, succinic acid, acetic acid, benzoic acid, citric acid, tartaric acid, carbonic acid or phosphoric acid. In addition, when the compounds of
Petition 870190060374, of 06/28/2019, p. 32/142
12/29
Formula (I) carries an acidic portion, its suitable pharmaceutically acceptable salts may include alkali metal salts, such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; and salts formed with suitable organic binders such as quaternary ammonium salts. Accordingly, representative pharmaceutically acceptable salts include acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, stolate, esolate, esolate, esate. gluceptate, gluconate, glutamate, glycollylsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxinaftoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylbromide, methylate, nitrate, methylate, nitrate, hydrate N-methylglucamine ammonium, oleate, pamoate (embonate), palmitate, pantothenate, phosphate / diphosphate, polygalacturonate, silicylate, stearate, sulfate, subacetate, succinate, tannate, tartrate, theoclate, tosylate, trietiodide and valerate.
Representative acids and bases that can be used in the preparation of pharmaceutically acceptable salts include acids including acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, (+) camphoric acid, camphorosulfonic acid, (+) - (1 S) -canphorus-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, cyclamic acid, dodecyl sulfuric acid , ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, formic acid, fumaric acid, galactic acid, gentisic acid, glycoheptonic acid, D-glyconic acid, D-glucoronic acid, L-glutamic acid, a-oxo-glutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, (+) L-lactic acid, (±) -DL-lactic acid, lactobionic acid, maleic acid o, acid (-)
Petition 870190060374, of 06/28/2019, p. 33/142
12/30
L-malic acid, malonic acid, (±) -DL-mandelic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene-1,5-disulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, oleic acid , orotic acid, oxalic acid, palmitic acid, pamoic acid, phosphoric acid, L-pyroglutamic acid, salicylic acid, 4-aminosalicylic acid, sebaic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+) - L -tartaric acid, thiocyanic acid, ptoluenesulfonic acid and undecylenic acid; and bases including ammonia, Larginine, benetamine, benzathine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2- (diethylamino) -ethanol, ethanolamine, ethylenediamine, N-methyl-glucamine, hydrabamino, 1H-imidazole, L-lysine , magnesium hydroxide, 4- (2-hydroxyethyl) -morpholine, piperazine, potassium hydroxide, 1- (2-hydroxyethyl) -pyrrolidine, sodium hydroxide, triethanolamine, tromethamine and zinc hydroxide.
[0061] The embodiments of the present invention include prodrugs of the compounds of Formula (I). In general, these prodrugs will be functional derivatives of compounds that are readily convertible in vivo into required compounds. Therefore, in the modalities of the treatment or prevention methods of the present invention, the term administer encompasses the treatment or prevention of various diseases, conditions, syndromes and disorders described with the specifically disclosed compound or with a compound that may not be specifically disclosed, but which converts to the specified compounds in vivo after administration to a patient. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in Design of Prodrugs, ed. H. Bundgaard, Elsevier, 1985.
[0062] When the compounds according to the modalities of this invention have at least one chiral center, they can also exist, consequently, as enantiomers. When compounds
Petition 870190060374, of 06/28/2019, p. 34/142
12/31 have two or more chiral centers, they must additionally exist as diastereomers. It is to be understood that all of these isomers and mixtures thereof are within the scope of the present invention. In addition, some of the crystalline forms of the compounds can exist as polymorphs and as such are included in the present invention. In addition, some of the compounds can form solvates with water (i.e., hydrates) or common organic solvents and such solvates are also within the scope of this invention. Those skilled in the art will understand that the term compound for use in the present invention should include the solvated compounds of Formula (I).
[0063] When the processes for preparing the compounds according to certain modalities of the invention give rise to the mixture of stereoisomers, these isomers can be separated by conventional techniques as by preparative chromatography. The compounds can be prepared racemically, or the individual enantiomers can be prepared by means of enantiospecific synthesis or by resolution. The compounds can, for example, be separated into their component enantiomers by standard techniques such as the formation of diastereomeric pairs by the formation of salt with an optically active acid such as (-) - di-ptoluoyl-d-tartaric acid and / or acid (+) - di-p-toluoyl-1-tartaric followed by fractional crystallization and regeneration of the free base. The compounds can also be resolved by forming diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds can be separated using a chiral HPLC column.
[0064] One embodiment of the present invention relates to a composition, including a pharmaceutical composition, which comprises, which consists of, and / or which essentially consists of
Petition 870190060374, of 06/28/2019, p. 35/142
32/122 one (+) - enantiomer of a compound of Formula (I), wherein said composition is substantially free of the (-) - isomer of said compound. In the present context, substantially exempt means less than about 25%, preferably less than about 10%, more preferably less than about 5%, even more preferably less than about 2% and more preferably, less than about 1% of the (-) - isomer calculated as follows% (+) enantiomer = ------------ ™ (+) - enantiomer) ---- -------- χ , θθ (mass (+) - enantiomer) + (mass (-) - enantiomer) [0065] Another embodiment of the present invention is a composition, including a pharmaceutical composition, which comprises, which consists of, and / or which essentially consists of (-) - enantiomer of a compound of Formula (I), said composition being substantially free of the (+) - isomer of said compound. In the present context, substantially free of means less than about 25%, preferably less than about 10%, more preferably less than about 5%, most preferably less than about 2% and, with even more preferably, less than about 1% of the (+) isomer calculated as follows (míW.Víí () 'esíknôôwroí i «ejsssitoia & íO .................... ........................; ......................... .......................................; .......... ....... () - eTianaòíTiero) i) * enaithiomer) [0066] During any of the processes for preparing the compounds of the various modalities of the present invention, it may be necessary and / or desirable to protect sensitive groups or reactive in any of the related molecules. This can be accomplished using conventional protective groups, such as those described in Protective Groups in Organic Chemistry, Second Edition, JFW
Petition 870190060374, of 06/28/2019, p. 36/142
12/33
McOmie, Plenum Press, 1973; T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991; and TW Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, Third Edition, John Wiley & Sons, 1999. Protective groups can be removed at a convenient subsequent stage using methods known in the art.
[0067] Although compounds of the modalities of the present invention (including their pharmaceutically acceptable salts and pharmaceutically acceptable solvates) can be administered alone, they will, in general, be administered in a mixture with a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and / or a pharmaceutically acceptable diluent selected according to the intended route of administration and standard pharmaceutical or veterinary practices. Therefore, the specific embodiments of the present invention relate to pharmaceutical and veterinary compositions comprising compounds of Formula (I) and at least one pharmaceutically acceptable carrier, pharmaceutically acceptable excipient, and / or pharmaceutically acceptable diluent.
[0068] As an example, in the pharmaceutical compositions of the modalities of the present invention, the compounds of Formula (I) can be mixed with any (any) binder (s), lubricant (s), suspending agent (s), coating (s), suitable solubilizing agent (s) and combinations thereof.
[0069] Solid oral dosage forms, such as tablets or capsules, that contain the compounds of the present invention can be administered in at least one dosage form at a time, as appropriate. It is also possible to administer the compounds in prolonged release formulations.
[0070] Additional oral forms in which the present compounds of
Petition 870190060374, of 06/28/2019, p. 37/142
The inventions can be administered include elixirs, solutions, syrups and suspensions; each optionally containing flavoring and coloring agents.
[0071] Alternatively, the compounds of Formula (I) can be administered by inhalation (intratracheal or intranasal) or in the form of a suppository or pessary, or they can be applied topically in the form of a lotion, solution, cream, ointment or talc. For example, they can be incorporated into a cream comprising, consisting of and / or consisting essentially of an aqueous emulsion of poly (ethylene glycols) or liquid paraffin. They can also be incorporated, at a concentration of between about 1% and about 10%, by weight, of the cream, in an ointment comprising, consisting of, and / or essentially consisting of a wax or soft paraffin base along with any stabilizers and preservatives as may be needed. An alternative means of administration includes transdermal administration using a dermal or transdermal patch.
[0072] The pharmaceutical compositions of the present invention (as well as the compounds of the present invention alone) can also be injected parenterally, for example, intracavernously, intravenously, intramuscularly, subcutaneously, intradermally or intrathecally. In that case, the compositions will also include at least one between a suitable vehicle, a suitable excipient and a suitable diluent.
[0073] For parenteral administration, the pharmaceutical compositions of the present invention are best used in the form of a sterile aqueous solution that may contain other substances, for example, sufficient salts and monosaccharides to constitute the isotonic solution with the blood.
[0074] In addition to the administration routes described above for the
Petition 870190060374, of 06/28/2019, p. 38/142
35/122 cancer treatment, pharmaceutical compositions can be adapted for administration by intratumoral or peritumoral injection. Activation of the immune system in this way to kill tumors in a remote location is commonly known as the abscopal effect and has been demonstrated in animals with multiple therapeutic modalities, (van der Jeught, et al., Oncotarget, 2015, 6 (3), 1359-1381). Another advantage of local or intratumoral or peritumoral administration is the ability to achieve equivalent efficacy in much smaller doses, thereby minimizing or eliminating adverse events that can be seen in much higher doses (Marabelle, A., et al. , Clinical Cancer Research, 2014, 20 (7), 1747-1756).
[0075] For buccal or sublingual administration the pharmaceutical compositions of the present invention can be administered in the form of tablets or lozenges, which can be formulated in a conventional manner.
[0076] By means of additional examples, pharmaceutical compositions, which contain at least one of the compounds of Formula (I) as the active ingredient, can be prepared by mixing the compound (s) with a pharmaceutically acceptable carrier, a pharmaceutically acceptable diluent, and / or a pharmaceutically acceptable excipient according to conventional pharmaceutical composition techniques. The vehicle, the excipient and the diluent can take a wide variety of forms depending on the desired route of administration (for example, oral, parenteral, etc.). Thus, for liquid oral preparations such as suspensions, syrups, elixirs and solutions, suitable vehicles, excipients and diluents include water, glycols, oils, alcohols, flavoring agents, preservatives, stabilizers, coloring agents and the like; for solid oral preparations such as powders, capsules and tablets, suitable carriers, excipients and diluents include starches, sugars, diluents,
Petition 870190060374, of 06/28/2019, p. 39/142
36/122 granulation, lubricants, binders, disintegrating agents and the like. Solid oral preparations can also optionally be coated with substances such as sugars or enterically coated in order to modulate the main absorption site and disintegration. For parenteral administration, the vehicle, excipient and diluent will, in general, include sterile water, and other ingredients can be added for the purpose of increasing the solubility and preservation of the composition. Injectable suspensions or solutions can also be prepared using aqueous vehicles together with suitable additives such as solubilizers and preservatives.
[0077] A therapeutically effective amount of a compound of Formula (I) or a pharmaceutical composition thereof includes a dose range of about 0.01 mg to about 3000 mg, or any specific amount or range thereof, in particular about 0.05 mg to about 1000 mg, or any specific amount or range of it, or, more particularly, from about 0.05 mg to about 250 mg or any specific amount or range of it, of the active ingredient in a about 1 to about 4 times a day for an average human being (70 kg); although it is apparent to the skilled person that the therapeutically effective amount for a compound of Formula (I) will vary, as will the diseases, syndromes, conditions and disorders being treated.
[0078] For oral administration, a pharmaceutical composition is preferably supplied in the form of tablets containing about 1.0, about 10, about 50, about 100, about 150, about 200, about 250 and about 500 milligrams of a compound of Formula (I).
[0079] Advantageously, a compound of Formula (I) can be administered in a single daily dose, or the total daily dosage can be administered in doses divided into two, three and four times a day.
Petition 870190060374, of 06/28/2019, p. 40/142
37/122 [0080] The ideal dosages of a compound of Formula (I) to be administered can be readily determined and will vary according to the particular compound used, the mode of administration, the strength of the preparation and the progress of viral infection, disease, syndrome, condition or disorder. In addition, factors associated with the particular individual being treated, including the individual's sex, age, weight, diet and time of administration, will result in the need to adjust the dosage in order to achieve an appropriate therapeutic level and the desired therapeutic effect. The dosages mentioned above are, therefore, exemplary of the average case. Naturally, there may be individual cases where greater or lesser dosage ranges are warranted and these are within the scope of the present invention.
[0081] The compounds of Formula (I) can be administered in any of the foregoing compositions and dosage regimens or by means of those compositions and dosing regimes established in the art when the use of a compound of Formula (I) is required for a individual who needs it.
[0082] As agonists of the STING protein, the compounds of Formula (I) are useful in methods for treating or preventing a viral infection, disease, syndrome, condition or disorder in an individual, including an animal, a mammal and a human being in whom viral infection, disease, syndrome, condition or disorder is affected by the modulation, including agonism, of the STING protein. Such methods comprise, consist and / or essentially consist of administering to an individual, including an animal, a mammal and a human in need of such treatment or prevention, a therapeutically effective amount of a compound, salt, or solvate of Formula (I) .
[0083] In one embodiment, the present invention is directed to a compound of Formula (I), or a pharmaceutically salt form
Petition 870190060374, of 06/28/2019, p. 41/142
38/122 acceptable, for use in the treatment of cancer, cancer diseases and conditions, or a viral infection.
[0084] Examples of cancer diseases and conditions for which the compounds of Formula (I), or pharmaceutically acceptable salts or solvates thereof, may have potentially beneficial antitumor effects include, but are not limited to, lung, bone, pancreas, skin, head, neck, uterus, ovary, stomach, colon, breast, esophagus, small intestine, intestine, endocrine system, thyroid gland, parathyroid gland, adrenal gland, urethra, prostate, penis, testicles, ureter, bladder, kidney or liver; rectal cancer; cancer of the anal region; carcinomas of the fallopian tubes, endometrium, cervix, vagina, vulva, renal pelvis, renal cell; soft tissue sarcoma; myxoma; rhabdomyoma; fibroma; lipoma; teratoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hemangioma; hepatoma; fibrosarcoma; chondrosarcoma; myeloma chronic or acute leukemia; lymphocytic lymphomas; primary CNS lymphoma; CNS neoplasms; tumors of the vertebral axis; squamous cell carcinomas; synovial sarcoma; malignant pleural mesothelioma; brain stem glioma; pituitary adenoma; bronchial adenoma; chondromatous hanlartoma; inesothelioma; Hodgkin's disease and a combination of one or more of the aforementioned cancers. Suitably, the present invention relates to a method for treating or reducing the severity of cancers selected from the group consisting of brain cancer (gliomas), glioblastomas, astrocytomas, glioblastoma multiforme, Bannayan-Zonana syndrome, Cowden's disease, Lhermitte-Duclos, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymomas, medulloblastoma, head and neck, kidney, liver, melanoma, ovary, pancreatic, adenocarcinoma, ductal madenocarcinoma, adenosquamous cell carcinoma, insulin cell carcinoma, glucagonma, procagonma , sarcoma, osteosarcoma, tumor of
Petition 870190060374, of 06/28/2019, p. 42/142
39/122 giant bone cells, thyroid, lymphoblastic T cell leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, capillary cell leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, chronic neutrophilic leukemia, acute lymphoblastic T cell leukemia, plasmacytoma, immunoblastic large cell leukemia, mantle cell leukemia, multiple myeloma, magacarioblastic leukemia, multiple myeloma, acute megakaryocytic leukemia, pro-myelocytic leukemia, erythroleukemia, malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoma, lymphoma Burkitt, follicular lymphoma, neuroblastoma, bladder cancer, urothelial cancer, vulvar cancer, cervical cancer, endometrial cancer, kidney cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharyngeal cancer, oral cancer, cancer of the mouth, GIST (gastrointestinal tumors stinal) and testicular cancer.
[0085] In another embodiment, the present invention is directed to a compound of Formula (I), or a pharmaceutically acceptable salt form thereof, for use in the treatment of a disorder affected by the STING agonism selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer, fibrosarcoma and hepatitis B.
The displayed compounds of Formula (I) may be useful in combination with one or more additional compounds useful to treat HBV infection. Such additional compounds may comprise other disclosed compounds and / or compounds known to treat, prevent or reduce the symptoms or effects of HBV infection. Such compounds include, but are not limited to, HBV polymerase inhibitors, interferons, viral entry inhibitors, viral maturation inhibitors, capsid assembly modulators described in the literature, reverse transcriptase inhibitors, immunomodulatory agents, TLR agonists and others agents with distinct or unknown mechanisms that affect
Petition 870190060374, of 06/28/2019, p. 43/142
40/122 the life cycle of HBV or affecting the consequences of HBV infection.
[0087] In non-limiting examples, the disclosed compounds can be used in combination with one or more drugs (or a salt thereof) selected from the group comprising:
HBV reverse transcriptase inhibitors, and DNA and RNA polymerase inhibitors including, but not limited to, lamivudine (3TC, Zeffix, Heptovir, Epivir, and Epivir-HBV), entecavir (Baraclude, Entavir), adefovir dipivoxil (Hepsara, Preveon, bis-POM PMEA), tenofovir disoproxil (Viread, TDF or PMPA);
interferons including, but not limited to, alpha interferon (IFN-α), interferon beta (IFN-β), lambda interferon (IFN-λ), and gamma interferon (IFN-γ);
viral entry inhibitors;
viral maturation inhibitors;
capsid assembly modulators, such as, but not limited to, BAY 41-4109;
reverse transcriptase inhibitors;
immunomodulatory agents such as TLR agonists; and agents of distinct or unknown mechanisms, such as, but not limited to, AT-61 ((E) -N- (1-chloro-3-oxo-1-phenyl-3- (piperidin-1yl) prop-1 - en-2-yl) benzamide), AT-130 ((E) -N- (1-bromo-1 - (2-methoxy-phenyl) -3oxo-3- (piperidin-1-yl) prop-1-en -2-yl) -4-nitrobenzamide), and the like.
[0088] In one embodiment, the additional therapeutic agent is an interferon. The term interferon or IFN refers to any member of the family of highly homologous species-specific species that inhibit viral replication and cell proliferation and modulate the immune response. For example, human interferons are grouped into three classes: Type I, which includes interferon-alpha (IFN-α), interferon
Petition 870190060374, of 06/28/2019, p. 44/142
41/122 beta (IFN-β), and interferon-omega (IFN-ω), Type II, which includes interferongama (IFN-γ), and Type III, which includes interferon-lambda (IFN-λ). Recombinant forms of interferons that have been developed and are commercially available are covered by the term interferon, as used here. Interferon subtypes, such as chemically modified or mutated interferons, are also covered by the term interferon, as used here. Chemically modified interferons can include pegylated interferons and glycosylated interferons. Examples of interferons also include, but are not limited to, interferon-alpha-2a, interferon-alpha-2b, interferon-alpha-n1, interferon-beta-1a, interferon-beta-1b, interferon-lambda-1, interferonlambda -2, and interferon-lambda-3. Examples of pegylated interferons include pegylated alpha-2a interferon and pegylated interferon alfa-2b.
[0089] Consequently, in one embodiment, the compounds of Formula (I) can be administered in combination with an interferon selected from the group consisting of interferon alpha (IFN-α), interferon beta (IFN-β), interferon lambda (IFN -λ), and gamma interferon (IFN-γ). In a specific embodiment, interferon is interferon-alpha-2a, interferonalfa-2b, or interferon-alpha-n1. In another specific embodiment, interferon-alpha-2a or interferon-alpha-2b is pegylated. In a preferred embodiment, interferon-alpha-2a is pegylated interferon-alpha-2a (PEGASYS). In another embodiment, the additional therapeutic agent is selected from among immune modulator or immune stimulating therapies, which include biological agents belonging to the interferon class.
[0090] In addition, the additional therapeutic agent may be an agent that disrupts the function of other essential viral proteins or host proteins necessary for HBV replication or persistence.
[0091] In another embodiment, the additional therapeutic agent is a
Petition 870190060374, of 06/28/2019, p. 45/142
42/122 antiviral agent that blocks viral entry or maturation or targets HBV polymerase as inhibitors of nucleotide or nucleoside or non-nucleotide or non-nucleoside polymerase. In an additional modality of combination therapy, the reverse transcriptase inhibitor or DNA or RNA polymerase inhibitor is Zidovudine, Didanosine, Zalcitabine, ddA, Stavudine, Lamivudine, Abacavir, Entricitabine, Entecavir, Apricitabine, Atevirapine, ribavirin, acyclovirine, acyclovirine, acyclovirine, acyclovirine, acyclovirine valacyclovir, ganciclovir, valganciclovir, Tenofovir, Adefovir, PM PA, cidofovir, Efavirenz, Nevirapine, Delavirdine or Etravirine.
[0092] In one embodiment, the additional therapeutic agent is an immunomodulatory agent that induces a limited natural immune response leading to the induction of immune responses against unrelated viruses. In other words, the immunomodulatory agent can affect the maturation of antigen presenting cells, the proliferation of Tea cells and release of cytokines (eg, IL-12, IL-18, IFN-alpha, -beta, and gamma and TNF-alpha among others), [0093] In another embodiment, the additional therapeutic agent is a TLR modulator or a TLR agonist, such as a TLR-7 agonist or TLR-9 agonist. In an additional modality of combination therapy, the TLR-7 agonist is selected from the group consisting of SM360320 (9-benzyl-8-hydroxy-2- (2-methoxy-ethoxy) adenine) and AZD 8848 (methyl [3 - ({[3- (6-amino-2-butoxy-8-oxo-7,8-dihydro-9H-purin-9-yl) propyl] [3 (4-morpholinyl) propyl] amino} methyl) phenyl] acetate).
[0094] In any of the methods provided herein, the method may additionally comprise administering to the individual at least one HBV vaccine, an HBV nucleoside inhibitor, an interferon or any combination thereof. In one embodiment, the HBV vaccine is at least one of RECOMBIVAX HB, ENGERIX-B, ELOVAC B, GENEVAC-B, or SHANVAC B.
[0095] In one embodiment, the methods described here
Petition 870190060374, of 06/28/2019, p. 46/142
43/122 further comprise the administration of at least one additional therapeutic agent selected from the group consisting of nucleotide / nucleoside analogs, entry inhibitors, fusion inhibitors and any combination of these or other antiviral mechanisms.
[0096] In another aspect, a method of treating an HBV infection in an individual in need is provided herein, which comprises reducing the viral load of HBV by administering to the individual a therapeutically effective amount of a compound disclosed alone or in combination with a reverse transcriptase inhibitor; and additionally administering to the individual a therapeutically effective amount of HBV vaccine. The reverse transcriptase inhibitor can be at least one of Zidovudine, Didanosine, Zalcitabine, ddA, Stavudine, Lamivudine, Abacavir, Entricitabine, Entecavir, Apricitabine, Atevirapine, ribavirin, acyclovir, fanciclovir, valacyclovir, valacyclovir, Valacyclovir, , cidofovir, Efavirenz, Nevirapine, Delavirdine or Etravirine.
[0097] In another aspect, a method of treating an HBV infection in an individual in need is provided herein, which comprises reducing the viral load of HBV by administering to the individual a therapeutically effective amount of a disclosed compound alone or in combination with an antisense oligonucleotide or RNA interference agent that targets HBV nucleic acids; and additionally administering to the individual a therapeutically effective amount of HBV vaccine. The antisense oligonucleotide or RNA interference agent has sufficient complementarity with the target HBV nucleic acids to inhibit the replication of the viral genome, the transcription of viral RNAs, or the translation of viral proteins.
[0098] In another modality, the revealed compound and the hair
Petition 870190060374, of 06/28/2019, p. 47/142
44/122 minus an additional therapeutic agent are co-formulated. In yet another embodiment, the disclosed compound and at least one additional therapeutic agent are co-administered. For any combination therapy described here, a synergistic effect can be calculated, for example, using suitable methods such as the Sigmoid-Emáx equation (Holford & Scheiner, 19981, Clin. Pharmacokinet. 6: 429453), Loewe's additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol. 114: 313-326) and the median effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul. 22: 27-55). Each equation mentioned above can be applied to experimental data to generate a corresponding graph that assists in the evaluation of the effects of the drug combination. The corresponding graphs associated with the equations mentioned above are the concentration-effect curve, the isobologram curve and the combination index curve, respectively.
[0099] In an embodiment of any of the methods of administering combination therapies provided in the present invention, the method may further comprise monitoring or detecting the individual's HBV viral load, the method being performed over a period of time including until the time when the HBV virus is undetectable.
[0100] The abbreviations used in the specification of the present invention, particularly in the diagrams and examples, are as follows:
ACN acetonitrile AcOH glacial acetic acid ADDP azodicarboxylic dipiperidide aq. aqueous Bn or Bzl benzila BINAP 2,2'-bis (diphenylphosphino) -1,1 '-binaftyl Boc tert-butyl oxycarbonyl
Petition 870190060374, of 06/28/2019, p. 48/142
45/122
cone. focused dba dibenzylideneacetone DBU 1,8-diazabicycle [5.4.0] undec-7-eno DCC Λ /, Λ / '- dicyclohexyl carbodiimide DCE 1,2-dichloroethane DCM dichloromethane DEAD diethyl azodicarboxylate DIBAL diisobutylaluminum hydride DIPEA or DIEA diisopropyl ethylamine DMA dimethylaniline DMAP 4-dimethylaminopyridine DME dimethoxyethane DMF A /, A / -dimethylformamide DMSO dimethyl sulfoxide DMT 4,4'-dimethoxytrityl DPPA diphenylphosphoryl azide dppf 1,1'-bis (diphenylphosphino) ferrocene AND THE ethyl acetate EDCI 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide ESI electrospray ionization EtOAc or EA ethyl acetate EtOH ethanol GCMS gas chromatography-mass spectrometry h or hr (s) hour or hours HEK human embryonic kidneys HPLC high performance liquid chromatography LAH lithium and aluminum hydride LDA lithium diisopropylamide LHMDS lithium bis (trimethylsilyl) amide MEK methyl ethyl ketone
Petition 870190060374, of 06/28/2019, p. 49/142
46/122
MeOH methanol MHz megahertz min minute or minutes MS mass spectrometry Ms methanesulfonyl NBS A / -bromosuccinimide NIS / -Iodosuccinimide NMM N-methylmorpholine NMP A / -methyl pyrrolidone NMR nuclear magnetic resonance PCC pyridinium chlorochromate PE Petroleum ether RP reverse phase ta or TA room temperature Tr retention time Sec second or seconds SEM-CI 2- (trimethylsilyl) ethoxymethyl chloride TBAF tetrabutyl ammonium fluoride TBDMS t-butyldimethylsilyl TBP tributyl phosphate TEA or Et 3 N Triethylamine TFA trifluoroacetic acid THF tetrahydrofuran TIPS triisopropylsilyl TLC thin layer chromatography TMS tetramethyl silane Ts 4-toluenesulfonyl
Petition 870190060374, of 06/28/2019, p. 50/142
47/122
Specific examples
Example 1
NHBz
N (i-Pr) 2
1d
1) TFA * Py water, MeCN RT, 2 min
2) t-BuNH 2
RT, 1D min
TMSCI, Py
1-methyl imidazole P (NiPr 2 ) (OCE) CI
DIPEA.THF
RT = TA MS = EM
Petition 870190060374, of 06/28/2019, p. 51/142
48/122
Compound 2 ammonium salt
Compound 2 sodium salt
Step 1: preparation of compound 1e [0101] To a solution of 3'-0-methyl-guanosine 1d (CAS 10300-27-
3, 1.0 g, 3.36 mmols) in pyridine (20 ml) was added by dropping tert-butylchlorodimethyl silane (3.2 ml, 25.2 mmols) at room temperature. After one hour, isobutyryl chloride (1.08 g, 10.1 mmols) was added by dropping at room temperature. The final mixture was stirred at room temperature for two hours. The mixture was quenched with water (30 ml) at 0 ° C and NH 4 OH (6 ml) was added by dropping at 0 ° C. After 10 minutes, the mixture was stirred at room temperature for 0.5 h. The mixture was concentrated. The crude product was purified by FCC (DCM: MeOH =
10: 1) to produce 1e (790 mg, 63.9%) as a white solid. NMR
H 1 (400 MHz, DMSO-de) 12.08 (s, 1H), 11.67 (s, 1H), 8.27 (s, 1H), 5.81 (d, J = 6.0 Hz, 1H), 5.51 (d, J = 6.0 Hz, 1H), 5.10 (t, J = 5.2 Hz, 1H),
Petition 870190060374, of 06/28/2019, p. 52/142
49/122
4.59-4.57 (m, 1H), 4.01-3.99 (m, 1H), 3.86-3.84 (m, 1H), 3.65 - 3.57 (m, 2H ), 3.41 (s, 3H), 3.17 (d, J = 5.2 Hz, 1H), 2.79-2.76 (m, 1H), 1.14 (s, 3H), 1 , 12 (s, 3H). ESI-MS: m / z 368.0 [M + 1] + .
Step 2: preparation of compound 1f [0102] A solution of compound 1e (790 mg, 2.15 mmol) and DMTrCI (0.765 g, 2.26 mmol) in pyridine (10 mL) was stirred at room temperature for one day to the other. DMTCI (0.765 g, 2.26 mmol) was added, and the mixture was stirred at room temperature for two hours. The mixture was abruptly cooled with water (10 ml) and extracted with DCM (10 ml x 4). The combined organic layer was dried with Na2SC> 4, filtered and the filtrate was concentrated. The residue was purified by flash chromatography (DCM: MeOH = 15: 1, ph = = 0.5) to produce compound 1f (1.28 g, 88.9%) as a light yellow solid. NMR H 1 (400 MHz, CDCh) 11.87 (s, 1H), 7.68 - 7.66 (m, 2H), 7.57 (d, J = 7.6 Hz, 2H), 7.44 (t, J = 9.2 Hz, 4H), 7.31 - 7.29 (m, 2H), 7.22 - 7.19 (m, 1H), 6.87 -6.81 (m, 4H ), 5.70 (d, J = 7.2 Hz, 1H), 5.30 -5.27 (m, 1H), 5.05 - 5.03 (m, 1H), 4.19 - 4, 18 (m, 1H), 4.09 - 4.07 (m, 1H), 3.78 (s, 3H), 3.77 (s, 3H), 3.58 - 3.55 (m, 1H) , 3.47 (s, 3H), 3.05 - 3.03 (m, 1H), 1.47 -1.40 (m, 1H), 0.85 (d, J = 6.8 Hz, 3H ), 0.55 (d, J = 6.8 Hz, 3H); ESI-MS: m / z = 670.2 [M + 1] + .
Step 3: preparation of compound 1q [0103] To a solution of compound 1f (1.28 g, 1.91 mmol) and DIPEA (741.0 mg, 5.73 mmol) in THF (5 mL) was added 3 - ((chlorine (diisopropylamino) phosphine) oxy) propanonitrile (1.36 g, 5.73 mmols) at room temperature. The reaction mixture was stirred at room temperature for one hour. The reaction mixture was quenched with MeOH. The mixture was extracted with EtOAc and the combined organic layers were washed with brine twice. The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by flash column chromatography (DCM: MeOH =
Petition 870190060374, of 06/28/2019, p. 53/142
50/122
10: 1, Η = 0.6) to produce compound 1g (1 g, 60.1%). NMR H 1 (400 MHz, CD3CN) 7.88 (d, J = 9.0 Hz, 1H), 7.48 - 7.41 (m, 2H), 7.35 - 7.24 (m, 7H) , 6.88 - 6.79 (m, 4H), 6.02 - 5.89 (m, 1H), 5.19 - 4.95 (m, 1H), 4.28 4.20 (m, 1H ), 4.07 - 4.04 (m, 1H), 3.78 (d, J = 1.6 Hz, 7H), 3.67 - 3.48 (m, 4H), 3.44 (d, J = 15.6 Hz, 3H), 3.33 (td, J = 2.8, 10.8 Hz, 1H), 2.72 - 2.65 (m, 1H), 2.61 -2.53 (m, 1H), 2.51 (t, J = 6.0 Hz, 1H), 1.26-1.24 (m, 4H), 1.18 - 1.12 (m, 12H), 0, 91 (d, J = 6.8 Hz, 3H); P 31 NMR (162 MHz, CD3CN) 150.90 (s, 1P), 150.81 (s, 1P), 13.80 (s, 1P); ESI-MS: m / z787.2 [M + 1] + .
Step 4: preparation of compound 1b [0104] To a solution of compound 1a (4.3 g, 4.51 mmols) and water (156.8 mg, 8.7 mmols) in dry CH 3 CN (16 mL) added pyridinium trifluoroacetate (1.0 g, 5.2 mmols) at room temperature. After 1 minute, t-butylamine (4 ml) was added. The resulting mixture was stirred at 15 ° C for 20 minutes. The mixture was concentrated for two hours to produce crude product 1b as a white solid (4.0 g). The crude product was used directly in the next step.
Step 5: preparation of compound 1c [0105] To a solution of compound 1 b (4.05 g, 4.35 mmol) and water (832.0 mg, 46.2 mmol) in DCM (40 mL) was added dichloroacetic acid (2.1 g, 16.3 mmol) at room temperature for 50 minutes. After 10 minutes, pyridine (730.6 mg, 9.24 mmol) was added. The mixture was concentrated and the residue was purified by flash column chromatography (CH2 Cl2: MeOH = 5: 1, R = 0.4) to yield compound 1c (2.45 g, 89.5%) as a white solid.
[0106] ESI-MS: m / z = 449.9 [M + 1] + .
Step 6: preparation of compound 1i [0107] A solution of compound 1c (300 mg, 0.48 mmol) and 4 Á molecular sieves in dry CH 3 CN (20 mL) was stirred at room temperature under N 2 for 10 minutes . 1 Himidazole perchlorate (1.5 g, 8.8 mmol) was added. After 10 minutes, 0
Petition 870190060374, of 06/28/2019, p. 54/142
51/122 compound 1 g (0.54 g, 0.62 mmol) in CH 3 CN (5 ml) was added. The mixture was stirred at room temperature for 50 minutes. Tert-butyl hydroperoxide (0.43 mL, 2.39 mmol) was added. The resulting mixture was stirred at room temperature for one hour and concentrated. The mixture was concentrated and the residue was purified by preparative HPLC (water (10 mM NH4HCO 3 ) -CH 3 CN) to produce compound 1i (168 mg, 34.1%) as a white solid. NMR H 1 (400 MHz, CD3OD) 8.89 (s, 1H), 8.79 (s, 1H), 8.36 (s, 1H), 8.16 (d, J = 7.5 Hz, 2H ), 7.73 - 7.67 (m, 1H), 7.62 (t, J = 7.0 Hz, 2H), 6.27 - 6.18 (m, 2H), 5.38 - 5, 30 (m, 1H), 4.81 (m, 2H), 4.44 (s, 1H), 4.29 (s, 1H), 4.26 - 4.15 (m, 3H), 3.92 - 3.85 (m, 1H), 3.75 (d, J = 12.4 Hz, 1H), 3.61 3.57 (m, 3H), 2.74 (td, J = 6.6, 13.1 Hz, 1H), 1.21 (dd, J = 6.8, 15.2 Hz, 6H), 0.85 (s, 9H), 0.12 (s, 3H), -0.04 (s, 3H); P 31 NMR (162 MHz, CD3OD) δ 3.61 (s, 1P), -1.68 (s, 1P); ESI-MS: m / z = 1033.2 [M + 1] + . Step 7: preparation of compound 1k [0108] To a solution of compound 1i (160 mg, 0.16 mmol) and 4 Á molecular sieves in pyridine (40 ml) was added DMOCP (87.0 mg, 0.47 mmol) at room temperature. The mixture was stirred at room temperature for one hour. sludge (199.4 mg, 0.79 mmol) and water (28.3 mg, 1.57 mmol) were added. After one hour, the mixture was filtered and then a saturated solution of Na 2 SO 3 was added by dripping until the color of the filtrate changed to pale yellow. The mixture was filtered and the filtrate was concentrated. The residue was purified by preparative HPLC (water (10 mM NH4HCO 3 ) -CH 3 CN from 23% to 53%) to produce the target product 1k (48 mg, 31.3%) as a white solid. ESI-MS: m / z 977.5 [M + 1] + .
Step 8: preparation of compound 11 [0109] Compound 1k (40.0 mg, 0.041 mmol) was treated with a solution of methylamine in EtOH (33%, 10 ml), was stirred at room temperature for one hour. The reaction mixture was concentrated to
Petition 870190060374, of 06/28/2019, p. 55/142
52/122 yield the crude compound 1 I (32.9 mg, 100%) which was used directly in the next step. ESI-MS: m / z803.4 [M + 1] + .
Step 9: preparation of compound 2 [0110] A solution of compound 1 I (32.86 mg, 0.041 mmol), Et 3 N (248.5 mg, 2.46 mmol) and triethylamine trihydrofluoride (198.0 mg, 1.2 mmol) in pyridine (5 ml) was stirred at 50 ° C for 5 h. The mixture was diluted with THF (10 ml) and isopropoxytriethyl silane (541.5 mg, 4.1 mmol) was added at room temperature for 1.5 h. The mixture was concentrated and the residue was purified by preparative HPLC (water (0.05% NH4OH v / v) -CH 3 CN from 0% to 15%) to produce target product 2 as its ammonium salt (6.7 mg) as a white solid. 1 1 H NMR (400 MHz, D2O) δ = 8.18-8.16 (d, J = 10 Hz, 2H), 7.74 (s, 1H), 6.06 (s, 1H) 5 , 79-5.77 (d, J = 8.8 Hz, 1H), 5.62-5.56 (m, 1H), 4.99-4.94 (m, 1H), 4.45 (m , 1H), 4.39-4.34 (m, 2H), 4.15 - 4.03 (m, 4H), 3.46 (s, 3H); NMRP 31 (162 MHz, D2O) -1.28 (s, 1P), -2.65 (s, 1 P); ESI-MS: m / z689.5 [M + 1] + .
Step 9: preparation of (Compound 2, Na salt) [0111] A volume of 3 ml of Dowex 50W x 8, 200 to 400 (form H) was added to a beaker (for 6.7 mg of compound 5 of salt ammonium) and washed with deionized water (2x). Then, 15% H2SO4 in deionized water (50 ml) was added to the resin, the mixture was stirred for 15 minutes, and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 CV), and then with deionized water until it was neutral. The resin was transferred back to the beaker, and 15% NaOH in aqueous solution (50 ml) was added, and the mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 CV) and then with water until it was neutral (at least 4 CV). Compound 5 (6.7 mg) was dissolved in deionized water (6.7 mg in 1 ml) and added to the top of the column, and
Petition 870190060374, of 06/28/2019, p. 56/142
53/122 eluted with deionized water. The compound was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce the Na salt of the target compound (6.4 mg, 94.2%) as a white foam. NMR H 1 (400 MHz, D2O) 8.16 (s, 1H), 8.13 (s, 1H), 7.74 (s, 1H), 6.04 (s, 1H), 5.78 (d , J = 8.8 Hz, 1H), 5.61 - 5.56 (m, 1H), 4.98 - 4.93 (m, 1H), 4.65 (s, 1H), 4.44 - 4.35 (m, 3H), 4.15 -4.05 (m, 4H), 3.46 (s, 3H); P 31 NMR (162 MHz, D2O) -1.26, -2.64; ESI-MS: m / z689.0 [M + 1] + .
Example 2
MS = MS
Petition 870190060374, of 06/28/2019, p. 57/142
54/122
Compound 1, ammonium salt
Compound 1, ammonium salt
Dowex-Na
Ο
NH 2
Compound 1, sodium salt
Step 1: preparation of compound 2b [0112] To a solution of DMT-2'-OMe-Bz-adenosine-EC phosphoramidite 2a (1.0 g, 1.13 mmol) and water (40.6 mg, 2.25 mmols) in dry CH 3 CN (4 ml) pyridinium trifluoroacetate (261.0 mg, 1.35 mmol) was added at 15 ° C. To the reaction mixture, t-butylamine (4 ml) was added. The mixture was concentrated to produce 1 g of crude compound 2b as a white solid, which was coevaporated with DCM (3x) and used directly for the next step. ESI-MS: m / z = 450.0 [M + 1] + . (DMT = 4,4'-dimethoxy trityl).
Step 2: preparation of compound 2c [0113] To a solution of compound 2b (930.0 mg, 1.12 mmol) and water (0.2 g, 11.2 mmol) in CH2 Cl2 (10 ml) was added acid dichloroacetic (0.51 g, 4.0 mmols) at 15 ° C for 0.5 h. After 10
Petition 870190060374, of 06/28/2019, p. 58/142
55/122 minutes, pyridine was added. The mixture was concentrated and the residue was purified by flash column chromatography (CH2 Cl2: MeOH = 5: 1, phit = 0.5) to produce compound 2c (400 mg, 0.76 mmol, 67.6% yield) like a white solid. P 31 NMR (400 MHz, DMSO-de) δ 0.05; ESI-MS: m / z = 450.0 (M + 1).
Step 3: preparation of compound 2f [0114] A solution of compound 2c (860.0 mg, 1.63 mmol) and 4 Á molecular sieves (1 g) in dry CH3CN (16 mL) was stirred at 15 ° C under N 2 for 10 minutes. 1/7-Imidazole perchlorate (5.16 g, 30.26 mmol) was added. After 10 minutes, DMT-3'-O-TBDMSG (iBu) -CE phosphoramidite 2d (2.05 g, 8.11 mmols) in anhydrous CH3CN (4 ml) was added. The mixture was stirred at room temperature for 50 minutes. A solution of tert-butyl hydroperoxide (TBHP, 1.48 mL, 8.14 mmol, 5.5% M in hexane) was added. The resulting mixture was stirred at 15 ° C for one hour. The mixture was concentrated and the residue was purified by preparative HPLC (water (10 mM NH4HCO3) -CH 3 CN) to produce compound 2f (600 mg, 0.58 mmol, 35.7% yield) as a white solid . NMR H 1 (400 MHz, CD3OD) δ 8.61 (d, J = 5.2 Hz, 1H), 8.41 - 8.30 (m, 1H), 8.24
- 8.17 (m, 1H), 8.04 - 7.90 (m, 2H), 7.61 - 7.49 (m, 2H), 7.48 - 7.40 (m, 2H), 6 , 11 - 6.03 (m, 1H), 6.02 - 5.98 (m, 1H), 5.36 - 5.12 (m, 1H), 4.55
- 4.43 (m, 2H), 4.41 - 4.32 (m, 1H), 4.30 - 4.19 (m, 2H), 4.13 - 4.02 (m, 1H), 3 , 99 - 3.85 (m, 2H), 3.75 - 3.65 (m, 1H), 3.63 - 3.53 (m, 1H), 3.37 (s, 3H), 2.70 - 2.69 (m, 1H), 2.60 - 2.52 (m, 2H), 1.10 - 1.03 (m, 6H), 0.82-0.77 (m, 9H), 0 , 04 - 0.00 (m, 6H); P 31 NMR (162 MHz, CD3OD) δ 3.17, 3.13, -2.58, -2.69; ESI-MS: m / z = 517.1 [M / 2 + 1] + and 1032.3 [M + 1] + .
Step 4: preparation of compound 2i + compound 2i [0115] To a solution of compound 2g (280.0 mg, 0.27 mmol) and molecular sieves of 4 Á (1 g) in pyridine (60 ml) were added 5 , 5
Petition 870190060374, of 06/28/2019, p. 59/142
56/122 dimethyl-2-oxo-2-chloro-1,3,2-dioxa-phosphinan (DMOCP, 150.2 mg, 0.81 mmol) at 16 ° C. The mixture was stirred at 16 ° C for one hour. Iodine (344.3 mg, 1.36 mmol) and water (48.9 mg, 2.71 mmol) were added. After one hour, the reaction was quenched with Na 2 SO 3 solution . The mixture was filtered and the filtrate was concentrated. The residue was purified by preparative HPLC (water (10 mM NH4HCO3) -CH 3 CN) to produce a mixture of compound 2i and compound 2j (170 mg, 0.17 mmol, 60.8% yield) as a solid White. ESI-MS: m / z = 1030.4 [M + H] + .
Step 5: preparation of compound 2i [0116] A mixed compound 2i and compound 2j (170 mg, 0.17 mmol) was treated with a solution of methylamine in EtOH (15 mL, 33%) and the resulting solution was stirred at 15 ° C for one hour. Crude product 2j (134.3 mg) was used directly in the next step. Step 6: preparation of compound 1 [0117] A solution of compound 2j (134.3 mg, crude), Et 3 N (1.0 g, 10.0 mmols) and triethylamine trihydrofluoride (Et 3 N-3HF , 807.4 mg, 5.00 mmols) in pyridine (10 ml) was stirred at 50 ° C for 5 h. The mixture was diluted with THF (10 ml) and isopropoxy trimethyl silane (2.2 g, 16.7 mmols) was added. After stirring at 15 ° C for one hour, the mixture was concentrated at 15 ° C and the residue was purified by preparative HPLC (water (0.05% v / v NH4OH) -CH 3 CN) to produce compound 1 as its ammonium salt (19.5 mg, 0.028 mmol) as a white solid after lyophilization. NMR H 1 (400 MHz, D2O) δ 8.26 (s, 1H), 8.15 (s, 1H), 7.78 (s, 1H), 6.116 (s, 1H), 5.87 (d, J = 8.4 Hz, 1H), 5.66 (s, 1H), 4.95 (s, 1H), 4.48 (d, J = 4.4 Hz, 1H), 4.24 (m, 5H ), 3.97 (d, J = 11.7 Hz, 1H), 3.83 (d, J = 12.0 Hz, 1H), 3.69 (s, 3H); P 31 NMR (162 MHz, D2O) δ -1.57, -3.38; ESIMS: m / z = 688.9 [M + H] + .
Preparation of compound 1, sodium salt [0118] Dowex 50W x 8, 200 to 400 (25 mL, form H) was added to
Petition 870190060374, of 06/28/2019, p. 60/142
57/122 a beaker and washed with deionized water (60 mL). Then, to the resin, 15% H2SO4 in deionized water was added, and the mixture was gently stirred for 5 minutes and decanted (50 ml). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 CV), and then with deionized water until it was neutral. The resin was transferred back to the beaker, 15% NaOH in deionized water solution was added, and the mixture was gently stirred for 5 minutes, and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in H 2 O (at least 4 CV) and then with deionized water until it was neutral. Compound 1, ammonium salt (16 mg) was dissolved in a minimum amount of deionized water, added to the top of the column, and eluted with deionized water. The appropriate fractions of UV-based CDN were pooled together and lyophilized to produce the sodium salt form of compound 1 (13.5 mg). NMR H 1 (400 MHz, D2O) <5 8.17 (s, 1H), 8.14 (s, 1H), 7.73 (s, 1H), 6.14 (s, 1H), 5.83 (d, J = 8.8 Hz, 1H), 5.58-5.52 (m, 1H), 5.01 (s, 1H), 4.98-4.90 (m, 1H), 4, 04-4.51 (m, 5H), 4.04 (d, J = 11.7 Hz, 1H), 3.78 (d, J = 12.0 Hz, 1H), 3.69 (s, 3H ); P 31 NMR (162 MHz, D 2 O) δ -1.63, -2.29; ESI-MS: m / z = 689 [M + H] +.
Example 3
Compound 6
RT = TA MS = EM
Petition 870190060374, of 06/28/2019, p. 61/142
58/122
N (iPr) 2
1) TFA-Py, water, MeCN RT, 2 min
2) f-BuNHp, RT, Orrin
Py
DCA (6% in CH 2 CI 2 ). CH 2 CI 2
RT, 25 min
Compound 6, ammonium salt
Compound 6 ammonium salt Compound 6 sodium salt
Step 1: preparation of compound 3a [0119] To a solution of DMT-3'-OTBDMS-G (iBu) -CE phosphoramidite compound 2d (1 g, 1.03 mmol) and water (37.1 mg, 2, 06 mmols) in dry CH 3 CN (4 ml), pyridinium trifluoroacetate (238.9 mg,
1.2 mmol) at room temperature. To the reaction mixture was added tert
Petition 870190060374, of 06/28/2019, p. 62/142
59/122 butylamine (4 mL). The resulting mixture was stirred at room temperature for 20 minutes. The mixture was concentrated to produce compound 3a (941.1 mg) as a white solid, which was coevaporated with DCM (3x) and used directly in the next step.
Step 2: preparation of compound 3b [0120] To a solution of compound 3a (941.1 mg, 1.03 mmol) and water (0.19 g, 10.0 mmols) in CH2 Cl2 (30 ml) was added dichloroacetic acid solution (0.47 g, 3.62 mmols, 6% in DCM) at room temperature for 0.5 h. Pyridine (0.163 g, 2.06 mmol) was added. After 10 minutes, the mixture was concentrated and the residue was purified by flash column chromatography (DCM: MeOH = 5: 1, ph = = 0.5) to produce 3c (515 mg, 0.84 mmol) as a white solid. ESI-MS m / z532.1 [M + 1] + .
Step 3: preparation of compound 3ea + compound 3eb [0121] A solution of compound 3b (500 mg, 0.82 mmol) and MS (molecular sieves) of 4 Á (0.5 g) in dry CH3CN (10 mL) was stirred at room temperature under N 2 for 3 minutes. 1 / - / - Imidazole perchlorate (IMP, 2.54 g, 15.1 mmol) was added. After 10 minutes, LNA-dA (Bz) -CE phosphoramidite, compound 3c (943 mg, 1.06 mmol) in CH3CN (5 mL) was added. The mixture was stirred at room temperature for 50 minutes. A solution of tert-butyl hydroperoxide (TBHP, 5.5 M in hexane, 0.74 mL, 4.09 mmols) was added. The resulting mixture was stirred at room temperature for one hour. The mixture was concentrated and the residue was purified by preparative HPLC (water (10 mM NH4HCO3) -ACN) to produce a mixture of compound 3ea and compound 3eb (135.7 mg, 0.132 mmol) as a white solid. The product mixture was used Step 4: preparation of compound 3q [0122] To a solution of compound 3ea and compound 3eb (135.7 mg, 0.132 mmol) and 4 Á (0.5 g) molecular sieves in pyridine ( 30
Petition 870190060374, of 06/28/2019, p. 63/142
60/122 mL) DMOCP (72.6 mg, 0.39 mmol) was added at 29 ° C. The mixture was stirred at 29 ° C for one hour. Iodine (166.3 mg, 0.66 mmol) and water (23.6 mg, 1.31 mmol) were added. After one hour, the reaction was quenched with Na2SO 3 solution. The mixture was filtered and the filtrate was concentrated. The residue was purified by preparative HPLC (water (10 mM NH4HCO3) -CH 3 CN from 35% to 65%) to produce compound 3g (22 mg, 0.021 mmol) as a white solid. ESI-MS m / z 514.5 [M / 2 + 1] + and 1029.3 [M + 1] + .
Step 5: preparation of compound 3h [0123] Compound 3g (22 mg, 0.021 mmol) was treated with a solution of methylamine in EtOH (33%, 10 mL) was stirred at room temperature for one hour. The reaction mixture was concentrated to produce the crude compound 3h, which was evaporated with pyridine (3x) and used directly in the next step.
Step 6: preparation of compound 6, ammonium salt [0124] A solution of compound 3h, Et 3 N (176.7 mg, 1.75 mmol) and triethylamine trihydrofluoride (Et 3 N-3HF, 140.7 mg, 0.87 mmol) in pyridine (10 ml) was stirred at 50 ° C for 5 h. The mixture was diluted with THF (10 mL) and isopropoxy trimethyl silane (384.9 mg, 2.91 mmol) was added at 15 ° C over one hour. The mixture was concentrated at room temperature and the residue was purified by preparative HPLC (NH4OH 0.05% v / v) -CH 3 CN from 0% to 15%) to produce compound 6 as its ammonium salt (6.1 mg, 0.009 mmol) as a white solid after lyophilization. NMR H 1 (400 MHz, D2O) 8.30 (s, 1H), 8.11 (s, 1H), 7.83 (brs, 1H), 6.19 (s, 1H), 5.96 (d , J = 6.8 Hz, 1H), 4.98 (s, 1H), 4.57 (s, 1H), 4.36 - 4.30 (m, 3H), 4.16 (d, J = 6.0 Hz, 3H), 4.04 (d, J = 7.6 Hz, 1H), 3.86 (d, J = 11.6 Hz, 2H); P 31 NMR (162 MHz, D2O) -1.73, -3.40; ESI-MS m / z687.0 [M + 1] + . Preparation of compound 6, sodium salt [0125] Dowex 50W x 8, 200 to 400 (2 ml, form H) was added to a beaker and washed with deionized water (15 ml). So, to the resin
Petition 870190060374, of 06/28/2019, p. 64/142
61/122 15% H2SO4 in deionized water was added, the mixture was gently stirred for 5 minutes and decanted (10 ml). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 CV), and then with deionized water until it was neutral. The resin was transferred back to the beaker, 15% NaOH in deionized water solution was added, and the mixture was gently stirred for 5 minutes, and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 CV) and then with deionized water until it was neutral. Compound 6, ammonium salt (3.5 mg) was dissolved in a minimum amount of deionized water, added to the top of the column, and eluted with deionized water. The appropriate fractions of UV-based CDN were pooled together and lyophilized to produce the sodium salt of compound 6 (3.02 mg). H 1 NMR (400 MHz, D2O): δ 8.11 (s, 1H), 7.88 (s, 1H), 7.74 (s, 1H), 6.02 (s, 1H), 5.85 (d, J = 8.4 Hz, 1H), 5.65-5.58 (s, 1H), 4.91 (d, J = 3.2 Hz, 1H), 4.83 (s, 1H) , 4.50 (d, J = 4.8 Hz, 1H), 4.35-4.28 (m, 1H), 4.26-4.19 (m, 2H), 4.13-4.01 (m, 3H), 3.90 (d, J = 8Hz, 1H), 3.76 (d, J = 12.4 Hz, 2H); P 31 NMR (162 MHz, D 2 O) δ-1.64, -1.91.
Example 4
Compound 3
RT = TA MS = EM
Petition 870190060374, of 06/28/2019, p. 65/142
62/122
NHBz
2) β-BuNH 2 , RT, 10 min
1) TFA «Py, water, MeCN RT, 2 min
N (i-Pr) 2
O s s y ci
(DMOCP) pyridine, 4A MS
4da, R = CH 2 CH 2 CN
4db, R = H
Compound 3
Dowex - Na
Compound 3 ammonium salt
Compound 3 sodium salt
Step 1: preparation of compound 4ba and compound 4bb [0126] A solution of compound 1c (300 mg, 0.57 mmol) and 4 Á molecular sieves (0.5 g) in dry CH 3 CN (10 mL) was stirred at 29 ° C under N2 for 3 minutes. 1 / - / - imidazole perchlorate (1.76 g, 10.5 mmols) was
Petition 870190060374, of 06/28/2019, p. 66/142
63/122 added. After 10 minutes, a solution of compound 1g (500 mg, 0.58 mmol) in CH 3 CN (10 ml) was added. The mixture was stirred at room temperature for 50 minutes and tert-butyl hydroperoxide (TBHP, 0.52 ml, 2.84 mmols) was added. The resulting mixture was stirred at 29 ° C for one hour. The mixture was concentrated and the residue was purified by preparative HPLC (water (10 mM NH4HCO 3 ) -CH 3 CN) to produce a mixture of compounds 4ba and 4bb (100 mg, 0.114 mmol) as a white solid. P 31 NMR (162 MHz, DMSO) -0.66, -2.44; ESI-MS: m / z932.3 (M + 1).
Step 2: preparation of compound 4da and compound 4db [0127] To a suspension of compound 4ba and compound 4bb (100.0 mg, 0.11 mmol) and molecular sieves of 4 Á (0.5 g) in pyridine ( 30 ml) DMOCP (59.4 mg, 0.32 mmol) was added at 28 ° C. The mixture was stirred at 28 ° C for one hour, sludge (136.2 mg, 0.54 mmol) and water (19.3 mg, 1.1 mmol) were added. After one hour, the reaction was quenched with Na2SO 3 solution. The mixture was filtered and the filtrate was concentrated. The residue was purified by preparative HPLC (water (10 mM NH4HCO 3 ) -ACN from 1% to 28%) to produce a mixture of compound 4da and 4bd (50 mg, 0.057 mmol) as a white solid. The product was used directly in the next step.
Step 3: preparation of compound 3, ammonium salt [0128] A mixture of compounds 4da and 4db (50 mg, 0.057 mmol) was treated with a solution of methylamine in EtOH (33%, 20 mL) and the mixture was stirred at room temperature for two hours. The reaction mixture was concentrated to produce crude compound 3, which was purified by preparative HPLC (water (NH4OH 0.05% v / v) -CH 3 CN from 0% to 10%) to produce 0 compound 3, salt of ammonium, as a white solid (16 mg, 0.023 mmol). P 31 NMR (162 MHz, D2O) -1.53, -3.41.
[0129] The product was further purified by HPLC
Petition 870190060374, of 06/28/2019, p. 67/142
64/122 preparative (water (NH4OH 0.05% v / v) -CH 3 CN from 0% to 10%) to produce compound 3 as its ammonium salt, as a white solid.
Step 4: preparation of compound 3, sodium salt [0130] The ammonium salt of compound 3 was dried under high vacuum to produce a white solid (12 mg). Dowex 50W x 8, 200 to 400 (form H, 3 ml) was added to a beaker (for 12 mg of compound 6) and washed with deionized water (2x). Then to the resin, 15% H2SO4 in deionized water (50 ml) was added, and the mixture was stirred for 15 minutes, and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 CV), and then with deionized water until it was neutral. The resin was transferred back to the beaker, and 15% NaOH in deionized water solution (50 ml) was added and the mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in deionized water (at least 4 CV) and then with deionized water until it was neutral (at least 4 CV). Compound 3 was dissolved in deionized water (12 mg in 1 ml), added to the top of the column, and eluted with deionized water. The converted sodium salt was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 3, sodium salt (7.4 mg, 0.010 mmol). 1 1 H NMR (400 MHz, D2O) óóppm 8.17 (s, 1H), 8.14 (s, 1H), 7.74 (s, 1H), 6.14 (s, 1H), 5, 79 (d, J = 8.8 Hz, 1H), 5.65 - 5.59 (m, 1H), 5.02-5.00 (m, 1H), 4.44 (s, 1H), 4 , 36 - 4.29 (m, 3H), 4.15 - 4.11 (m, 3H), 4.04 - 4.01 (m, 1H), 3.66 (s, 3H), 3.46 (s, 3H); P 31 NMR (162 MHz, D2O) -1.53, -2.62; ESI-MS m / z702.5 (M + 1).
Petition 870190060374, of 06/28/2019, p. 68/142
65/122
Example 5
Compound 4
RT = TA MS = EM
Compound 5
1) TFA-Py, water, MeCN
RT, 2 min
2) / -BuNH 2 . RT, 10 min
CAS # 136834-22-5
5a 5b 5c
IMP = Imidazole Perchlorate
4A MS, MeCN
I DDTT
4A MS, MeCN
5ha + 5hb
Petition 870190060374, of 06/28/2019, p. 69/142
66/122
Compound 4
Compound 5
O
nh 2
Dowex - Na
Compound 4 ammonium salt
Compound 4 sodium salt
O
nh 2
Dowex - Na
O
nh 2
Compound 5 ammonium salt
Step 1: preparation of compound 5b
Compound 5 sodium salt [0131] To a phosphoramidite solution of DMT-2'-F-dA (Bz) -CE 5a (2.20 g, 2.51 mmols) in CH 3 CN (12.0 ml_) added water (90.5 mg, 5.02 mmols, 2.0 eq) and pyridinium trifluoroacetate (582.1 mg, 3.01 mmols,
1.2 eq). The mixture was stirred at 25 ° C for 5 min. Then, tert-butylamine (12.0 ml) was added and the reaction mixture was stirred at 25 ° C for 15 min. The mixture was concentrated under reduced pressure to produce a foam, which was dissolved in CH 3 CN (10.0 ml) and concentrated again to produce compound 5b (1.69 g, 2.29 mmols, 91.0% yield) as a white foam. ESI-MS: m / z 740.2 [M + H] + .
Petition 870190060374, of 06/28/2019, p. 70/142
67/122
Step 2: preparation of compound 5c [0132] To a solution of compound 5b (1.69 g, 2.29 mmoles) in CH2 Cl2 (24.0 ml) was added water (411.8 mg, 21.9 mmols, 10.0 eq) and a solution of 2,2-dichloroacetic acid (6% in DCM, 24 mL) slowly. The mixture was stirred at 25 ° C for 0.5 h. The reaction was quenched with pyridine (2 mL) and the reaction mixture was concentrated to produce a residue, which was purified by column chromatography on silica gel (DCM / MeOH = 10/1 to 5/1) to produce 0 compound 5c (856 mg, 1.62 mmol, 70.9% yield) as a white foam.
Step 3: preparation of compound 5e [0133] A solution of compound 5c (380 mg, 0.70 mmol) in CH3CN (12.0 mL) was added to 4 Á molecular sieves (0.5 g), the resulting mixture it was stirred at 25 ° C for 10 min. 1 / - / - Imidazole perchlorate (IMP, 356.7 mg, 2.1 mmol, 3.0 eq) was added and the mixture was stirred for an additional 10 minutes before DMT-3'-O phosphoramidite -TBDMS-G (iBu) -CE 2d ((J. Am. Chem. Soc. 2001,123, 81658176), 811.6 mg, 0.84 mmol, 1.2 eq) be added. The mixture was stirred at 25 ° C for one hour to produce a solution of compound 5d (a solution in CH3CN), then N, N-dimethyl-N '- (5-sulfanilidene-1,2,4-dithiazol-3il) metanimidamide (DDTT, 715.7 mg, 3.49 mmol, 5 eq.) was added to the above reaction mixture at 25 ° C and stirred for one hour at the same temperature. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to produce a residue, which was purified by preparative HPLC (H2O-CH3CN) to produce compound 5e (130.0 mg, 0.125 mmol, 18.0% two-step yield) as a white solid. ESI-MS: m / z 1036.1 [M + H] + .
Step 4: preparation of compound 5f [0134] To a solution of compound 5e (130.0 mg, 0.125 mmol) in pyridine (24 mL) was added 2-chloro-5,5-dimethyl-2-oxide, 3.2
Petition 870190060374, of 06/28/2019, p. 71/142
68/122 dioxafosfinane (DMOCP, 69.5 mg, 0.38 mmol, 3.0 eq) at 25 ° C, the mixture was stirred at 25 ° C for one hour to produce a solution of compound 5f (127.7 mg , 0.125 mmol, 100% yield, a solution in pyridine) which was used in the next step without further purification.
Step 5: preparation of compound 5qa + compound 5qb [0135] To a solution of compound 5f (127.7 mg, 0.125 mmol) in pyridine was added 3H-benzo [c] [1,2] dithiol-3-one ( 211.1 mg, 1.26 mmol, 10 eq) at 25 ° C, and the resulting mixture was stirred at 25 ° C for one hour. The reaction mixture was filtered, and the filtrate was concentrated under reduced pressure to produce a residue, which was purified by preparative HPLC (water (0.225% formic acid) - CH 3 CN) to produce compound 5ga (22.0 mg, 0.021 mmol, 18.1% yield in two steps) as a white solid and the compound 5gb (55.0 mg, 0.052 mmol, 45.2% yield in two steps) as a white solid. ESI-MS: m / z 1050.2 [M + H] + , 525.8 [M / 2 + H] + (compound 5ga). ESIMS: m / z 1050.2 [M + H] + , 525.6 [M / 2 + H] + (5gb compound).
Step 6: preparation of compound 5hb [0136] Compound 5gb (55.0 mg, 0.052 mmol, 1.00 eq) was treated with a solution of methylamine (3.00 ml_, 35% in EtOH) and the resulting mixture was stirred at 25 ° C for 12 h. The reaction mixture was concentrated under reduced pressure to produce compound 5hb (39.0 mg, 0.047 mmol, 90.5% yield) which was used in the next step without further purification. ESI-MS: m / z823.1 [M + H] + .
Step 7: preparation of compound 4, ammonium salt [0137] To a solution of compound 5hb (44.0 mg, 0.053 mmol) in pyridine (13.0 ml_) was added Et 3 N (324.7 mg, 3 , 2 mmols, 60 eq) and triethylamine trihydrofluoride (258.6 mg, 1.6 mmol, 30 eq) at 25 ° C, and the mixture was stirred at 50 ° C for 5 hours, then isopropoxy trimethyl silane ( 707.4 mg, 5.3 mmols, 100 eq) was added at 15 ° C and stirred
Petition 870190060374, of 06/28/2019, p. 72/142
69/122 for one hour. The mixture was concentrated at 15 ° C and the residue was purified by preparative HPLC (water (NH4OH 0.05% v / v) -CH 3 CN) to produce compound 4 as its ammonium salt (6.0 mg, 0.008 mmol, 15.8% yield) as a white solid. NMR H 1 (400 MHz, D2O) δ 8.33 (s, 1H), 8.25 (s, 1H), 7.85 (s, 1H), 6.45 (d, J = 14.3 Hz, 1H), 5.92 (d, J = 8.5 Hz, 1H), 5.77 (s, 1H), 5.51 (s, 0.5H), 5.38 (s, 0.5H), 5.23 (d, J = 21.5 Hz, 1H), 4.53-4.42 (m, 5H), 4.10 (d, J = 11.3 Hz, 1H), 3.99 (d , J = 12.8 Hz, 1H); F 19 NMR (376 MHz, D2O) δ -122.94 (br, s, 1F); P 31 NMR (162 MHz, D 2 O) δ 55.99 (brs, 1 P), 51.19 (brs, 1P); ESI-MS: m / z 708.9 [M + H] + .
Step 6a: preparation of compound 5ha [0138] Compound 5ga (13.0 mg, 0.012 mmol, 1.00 eq) was treated with a solution of methylamine (1.00 mL, 35% in EtOH) and the solution was stirred at 25 ° C for 12 h. The reaction mixture was concentrated under reduced pressure to produce the compound 5ha (9.0 mg, 0.011 mmol, 88.4% yield) which was used in the next step without further purification. ESI-MS: m / z 823.3 [M + H] + .
Step 7a: preparation of compound 5, ammonium salt [0139] To a solution of compound 5ha (39.0 mg, 0.047 mmol) in pyridine (7.0 mL) was added Et 3 N (287.8 mg, 2 , 84 mmol, 60 eq) and triethylamine trihydrofluoride (229.2 mg, 1.42 mmol, 30 eq) at 25 ° C, the mixture was stirred at 50 ° C for 5 hours, then isopropoxy trimethyl silane (627.0 mg, 4.74 mmol, 100 eq) was added at 15 ° C and stirred for 1 hour. The mixture was concentrated at 15 ° C and the residue was purified by preparative HPLC (water (NH4OH 0.05% v / v) -CH 3 CN) to produce compound 5 as its ammonium salt (6.60 mg, 0.009 mmol, 19.6% yield) as a white solid. NMR H 1 (400 MHz, D 2 O) δ 8.54 (s, 1H), 8.25 (s, 1H), 7.84 (s, 1H), 6.46 (d, J = 13.8 Hz, 1H), 5.94 (d, J = 8.3 Hz, 1H), 5.81 - 5.75 (m, 1H), 5.54 (d, J = 2.8 Hz, 0.5H ), 5.41 (d, J = 3.0 Hz, 0.5H), 5.30-5.23 (m, 1H), 4.54-4.40 (m,
Petition 870190060374, of 06/28/2019, p. 73/142
70/122
5H), 4.09-4.04 (m, 2H); NMR F 19 (376 MHz, D2O) δδ -201.92 (brs, 1F); P 31 NMR (162 MHz, D2O) δδ55.97 (s, 1 P), 53.90 (brs, 1P); MS: m / z 708.9 [M + H] +
Step 8: preparation of compound 4, sodium salt [0140] Dowex 50W x 8, 200 to 400 (3 ml_, form H) was added to a beaker and washed with deionized water (15 ml_). Then, to the resin was added H 2 SC> 4 to 15% in deionized water, the mixture was gently stirred for 5 minutes and decanted (10 ml). The resin was transferred to a column with H 2 SC> 4 to 15% in deionized water and washed with H 2 SC> 4 to 15% (at least 4 CV), and then with deionized water until it was neutral. The resin was transferred back to the beaker, 15% NaOH in deionized water solution was added, and the mixture was gently stirred for 5 minutes, and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 CV) and then with deionized water until it was neutral. Compound 4, ammonium salt (6.9 mg) was dissolved in a minimum amount of deionized water, added to the top of the column, and eluted with deionized water. The appropriate fractions of UV-based CDN were pooled together and lyophilized to produce the sodium salt form of compound 4 (6.45 mg). NMR H 1 (400 MHz, D2O) δ 8.41 (s, 1H), 8.12 (s, 1H), 7.73 (s, 1H), 6.35 (d, J = 13.6 Hz, 1H), 5.83 (d, J = 8.4 Hz, 1H), 5.72-5.68 (m, 1H), 5.43 (d, J = 2.8 Hz, 0.5 H) , 5.30 (d, J = 2.8 Hz, 0.5 H), 5.18-5.10 (m, 1H), 4.65-4.29 (m, 5H), 4.05- 3.93 (m, 2H); F 19 NMR (376 MHz, D2O) δ -201.76; P 31 NMR (162 MHz, D 2 O) δ 55.88, 53.91.
Step 9: Preparation of compound 5, sodium salt [0141] Dowex 50W x 8, 200 to 400 (3 ml_, form H) was added to a beaker and washed with deionized water (15 ml_). Then, to the resin was added H 2 SC> 4 to 15% in deionized water, the mixture was gently stirred for 5 minutes and decanted (10 ml). The resin was
Petition 870190060374, of 06/28/2019, p. 74/142
71/122 transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 CV), and then with deionized water until it is neutral. The resin was transferred back to the beaker, 15% NaOH in deionized water solution was added, and the mixture was gently stirred for 5 minutes, and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 CV) and then with deionized water until it was neutral. Compound 5, ammonium salt (6.0 mg) was dissolved in a minimum amount of deionized water, added to the top of the column, and eluted with deionized water. The appropriate fractions of UV-based CDN were pooled together and lyophilized to produce the sodium salt form of compound 5 (5.12 mg). NMR H 1 (400 MHz, D2O) δ 8.15 (s, 1H), 8.12 (s, 1H), 7.72 (s, 1H), 6.34 (d, J = 14.4 Hz, 1H), 5.81 (d, J = 8 Hz, 1H), 5,655.58 (m, 1H), 5.45 (d, J = 3.2 Hz, 0.5H), 5.32 (d, J = 3.6 Hz, 0.5H), 5.205.05 (m, 1H), 4.65-4.29 (m, 5H), 3.90-4.04 (m, 2H); F 19 NMR (376 MHz, D2O) δ-201.92; P 31 NMR (162 MHz, D2O) δδ55.74, 53.23; MS: m / z709.00 [M + H] +.
Example 6
Compound 8
7
RT = TA MS = EM
Petition 870190060374, of 06/28/2019, p. 75/142
72/122
6a 6c 6d
RT = TA
RT = TA
6g 5h
3HF: TEA, TEA (1.0eq), DMSO, 50 C, 4 hr.
6i - Isomer 1
Isomer - Isomer
6j- Isomer 2
Dowex - Na
6i - Isomer 1
Compound 7
Isomer - Isomer
Compound = Compound
Petition 870190060374, of 06/28/2019, p. 76/142
73/122
Dowex - Na
nh 2
6j- Isomer 2
Compound 8
Isomer - Isomer
Compound = Compound
Step 1: preparation of compound 6d [0142] The nucleoside compound 6a (1.63 g, 2.12 mmol) was coevaporated with a mixture of anhydrous toluene: anhydrous acetonitrile (1: 1, v / v, 3 x 20 ml_) ), and then dissolved in anhydrous acetonitrile (50 ml) and phosphoramidite 6b (2.1 g, 2.12 mmol). Molecular sieve powder of 4 Á (4.0 g) was added to this mixture. The resulting heterogeneous mixture was bubbled with argon gas for 4 minutes. After stirring this mixture at room temperature for 30 minutes, tetrazole in 0.45 M acetonitrile (30 ml, 12.72 mmols) was added at room temperature. After stirring the reaction for 45 minutes, the reaction mixture was filtered and then washed with saturated aqueous NaHCOs (1 x 20 ml) and saturated aqueous NaCl (1 x 20 ml), dried with MgSCU, and the filtrate was evaporated to dryness to produce compound 6c, phosphite, which was used directly without further purification in the next step. [0143] Compound 6c, crude phosphite, was dissolved in anhydrous CH2 Cl2 (40 ml), then 4 Á molecular sieve powder (4.0 g) was added to this mixture. The resulting heterogeneous mixture was bubbled with argon gas for 4 minutes. After stirring this mixture at room temperature for 30 minutes, a solution of borane-dimethyl sulfide complex (2.0 M in THF, BH3-DMS, 3.49 ml_, 6.99 mmols) was added very slowly over 5 minutes at 0 ° C. After stirring the reaction for 20 minutes at room temperature, the reaction mixture is rapidly
Petition 870190060374, of 06/28/2019, p. 77/142
74/122 filtered, then diluted with EtOAc (120 ml), abruptly cooled with water (20 ml). The phases were separated and the organic phase was washed with water (1 x 20 ml), saturated aqueous NaCl (1 x 20 ml) and then the aqueous phase was extracted back with EtOAc (1 x 20 ml). The combined organic phases were evaporated to dryness, and the resulting crude material was purified by flash column chromatography on silica gel (0 to 85% EtOAc in Hexane, v / v) to produce 6d boranophosphate dimer (980 mg). ESI-MS: m / z 1670.30 [M + H] + .
Step 2: preparation of compound 6e [0144] Di-DMTr-boranophosphate 6d dimer (2.3 g, 1.37 mmol) was dissolved in 80% aqueous AcOH: CH 3 CN (3: 1, v / v, 13 mL). After stirring the reaction mixture for 16 h at 37 ° C, the mixture was diluted with EtOAc (70 ml) and then washed sequentially with saturated aqueous NaHCO 3 (3 x 20 ml) and aqueous saturated NaCl (1x15 ml). The organic phase was evaporated to dryness, resulting in a crude residue that was purified by flash column chromatography on silica gel (20 to 100% acetone in hexane, v / v) to produce the diolboranophosphate 6e dimer (0.9 g). ESI-MS: m / z 1066.45 [M + H] + .
Step 3: preparation of compound 6f [0145] The nucleoside diol compound 6e (0.55 g, 0.516 mmol) was coevaporated with a mixture of anhydrous toluene: anhydrous acetonitrile (1: 1, v / v, 3 x 20 mL) and then dissolved in anhydrous acetonitrile (20 mL) and 4 Á powder molecular sieves (1.0 g) were added to this mixture. The resulting heterogeneous mixture was bubbled with Argon gas for 4 minutes. After stirring this mixture at room temperature for 30 minutes, tetrazole in 0.45 M acetonitrile (7 mL, 3.09 mmol) was added at room temperature, and then, after stirring the reaction for 75 minutes, the mixture was filtered, and the filtrate was washed sequentially with saturated aqueous NaHCO 3 (1 x 20 ml) and saturated aqueous NaCl (1 x 20 ml), dried over MgSCU (stirred for 5 min and
Petition 870190060374, of 06/28/2019, p. 78/142
75/122 then filtered) and evaporated to dryness to produce compound 6f. The resulting mixture was used directly without further purification in the next step. Crude phosphite 6f was dissolved in anhydrous CH2 Cl2 (20 mL), then 4 Á molecular sieve powder (1.0 g) was added to this mixture. The resulting heterogeneous mixture was bubbled with argon gas for 4 minutes. After stirring this mixture at room temperature for 30 minutes, borane-dimethyl sulfide complex solution (2.0 M in THF, BH3-DMS, 0.93 mL, 1.84 mmol) was added very slowly over 5 minutes at 0 ° C. After stirring the reaction at room temperature, the reaction mixture is quickly filtered, then diluted with EtOAc (80 ml), and abruptly cooled with water (20 ml). The phases were partitioned and the organic phase was washed with saturated aqueous NaCl (1 x 20 ml) then, the aqueous phase was back extracted with EtOAc (1 x 20 ml). The combined organic phases were evaporated to dryness. The resulting crude material was purified by flash column chromatography on silica gel (0 to 10% MeOH in dichloromethane, v / v) to produce fully protected cyclic boranophosphate 6f (480 mg, 75% pure). ESI-MS: m / z 1179.73 [M + H] + .
Step 4: preparation of compound 6q [0146] 3'-Silyl-G (iBu) -2'-silyl-A (Bz) 2 ', 3'-cyclic 6f (437 mg, approximately 75% pure) dinucleotide-boranophosphate was dissolved in the aqueous ammonia: EtOH mixture (7 mL, 3: 1, v / v). After stirring the reaction mixture for 16 h at 50 ° C, the reaction mixture was concentrated to dryness and coevaporated with EtOH (2x10 ml) and toluene (2 x 20 ml). The resulting crude solid was washed with dichloromethane (40 ml) and the precipitate was collected by filtration to produce the cyclic dimer protected from di-TBS 6g (ESI-MS: m / z896.20 [MH]), which was used for the next reaction without any further purification.
[0147] To remove the TBS groups, the compound of the cyclic dimer 6g (350 mg, raw) was dissolved in anhydrous DMSO (5.5 ml) and, for this, it was
Petition 870190060374, of 06/28/2019, p. 79/142
76/122 Triethylamine trihydrofluoride (HF.3TEA, 2.8 mL and 0.6 mL trimethylamine) was added. After stirring the reaction mixture for 3.5 h at 50 ° C, it was neutralized with triethylamine and purified by preparative HPLC (Buffer A: 50 mM triethyl ammonium acetate in H 2 O; Buffer B: 50 mM triethyl ammonium acetate in CH 3 CN, gradient: 0 to 30% B over 30 minutes, flow rate 24 mL / minute) to produce two isomers of boranophosphate 6i (9 mg) and 6j (4 mg) as a triethyl ammonium salt as a white solid. ESI-MS: m / z 668.6 [MH] '.
Step 5: preparation of compound 7 and compound 8 as sodium salt. [0148] Dowex 50W x 8, 200 to 400 (3 ml, form H) was added to a beaker and washed with deionized water (20 ml). Then, to the resin was added H 2 SC> 4 to 15% in deionized water, the mixture was gently stirred for 5 minutes and decanted (10 ml). The resin was transferred to a column with H 2 SC> 4 to 15% in deionized water and washed with H 2 SC> 4 to 15% (at least 4 CV), and then with deionized water until it was neutral. The resin was transferred back to the beaker, 15% NaOH in deionized water solution was added, and the mixture was gently stirred for 5 minutes, and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 CV) and then with deionized water until it was neutral. The triethylammonium form of both 6i (9 mg) and 6j (4 mg) cyclic boranophosphate isomers was dissolved in a minimum amount of deionized water, added to the top of the column, and eluted with deionized water. The appropriate fractions of UV-based CDN were pooled together and lyophilized to produce the sodium salt form of compound 7 (8.2 mg) and compound 8 (3.3 mg), respectively.
[0149] Compound 7: NMR H 1 (400 MHz, D 2 O): δ 8.15 (s, 1H), 8.09 (s, 1H), 7.99 (s, 1H), 6.01 ( s, 1H), 5.93 (d, J = 8.7 Hz, 1H), 4.98-4.93 (m, 1H), 4.87-4.80 (m, 1H), 4.74 -4.60 (m, 1H), 4.42-4.37 (m, 2H), 4.31 (s, 1H), 4.24 -4.15 (m, 2H), 3.92-3 82 (m, 2H), 0.5 to -0.5 (very peak
Petition 870190060374, of 06/28/2019, p. 80/142
77/122 broad, 6H); P 31 NMR (162 MHz, D2O) 94.70 (very broad peak); ESIMS: m / z 668.6 [M-1] '.
[0150] Compound 8: NMR H 1 (400 MHz, D 2 O): δ 8.12 (s, 1H), 8.10 (s, 1H), 7.95 (s, 1H), 5.99 ( d, J = 2.4 Hz, 1H), 5.92 (d, J = 8.4 Hz, 1H), 5.22-5.15 (m, 1H), 4.83-4.76 (m , 1H), 4.75-4.71 (m, 1H), 4.50 (d, J = 4 Hz, 1H), 4.40-4.30 (m, 1H), 4.31 (s, 1H), 4.25-4.18 (m, 1H), 4.15-4.10 (m, 1H), 4.02-3.96 (m, 1H), 3.90-3.84 ( m, 1H), -0.2 to 0.9 (very broad peak, 6H); P 31 NMR (162 MHz, D2O) δ 93.75 (very broad peak); ESIMS: m / z: 668.7 [M-1] '.
Example 7
Compound 9
Compound 10 o
H 3 CO OH
1) TMSCI
2) i-PrCOCI
3) NH4OH
1d
1f
Petition 870190060374, of 06/28/2019, p. 81/142
78/122
7b
Ο
DMTrO ·
AND ODMTr
NH O
NHBz | O -CN s
Aq. to 80% AcOH,
C, ON
NHBz
NHBz
7d
I'tTX-CN s
7e
OCE
0.45 M Tetrazole in ACN, ACN, 4Â sieve powder
Aq. in
CH 2 CI 2 , bh 3 -dms
C, then RT, 20 min
7f
RT = TA
nh 2
7g (smaller) nh 2
7h (higher)
ΝΉNJ-l ·
Compound 10 (smallest)
Compound 9 (largest)
Petition 870190060374, of 06/28/2019, p. 82/142
79/122
Step 1: preparation of compound 7c [0151] The nucleoside compound 1f (2.3 g, 3.43 mmoles) was coevaporated with an anhydrous toluene / anhydrous acetonitrile mixture (1: 1, v / v, 3 x 50 mL ), and then dissolved in anhydrous acetonitrile (80 mL). Molecular sieves of 4 Á powder (4.0 g) and tetrazole in acetonitrile (61 mL, 0.45 M, 27.44 mmols) were added to the reaction mixture. The resulting heterogeneous mixture was purged with bubbling Ar (g) for 4 minutes. After stirring at room temperature for 10 minutes, a solution of amidite 7a (3.0 g, 3.43 mmols, ChemGenes Corp.) in anhydrous acetonitrile (15 ml) was added at room temperature. After stirring for one hour and 45 minutes at room temperature, the reaction mixture was diluted with ethyl acetate (250 ml), filtered, and the filtrate was washed with saturated aqueous NaHCOs and brine (1 x 40 ml) (1 x 40 mL). The filtrate was then dried (MgSCU), stirred for 5 minutes, filtered, and the filtrate was concentrated to dryness to yield phosphite 7b (ESI-MS: m / z 1444.45 [M + 1] + .).
[0152] The crude phosphite compound 7b was used in the next step without further purification. The crude compound 7b was dissolved in anhydrous pyridine (100 mL) and (E) -A /, A / -dimethyl- / V- (3-thioxo-3H-1,2,4-dithiazol-5yl) formimidamide (DDTT, 2.12 g, 10.29 mmols) was added at room temperature. After stirring for one hour at room temperature, the reaction mixture was diluted with ethyl acetate (250 ml), washed with sequentially saturated aqueous NaHCOs (1 x 40 ml) and brine (1 x 40 ml), and concentrated to dry. The aqueous layer was extracted with ethyl acetate (1 x 20 ml). The combined organic phases were concentrated to dryness under reduced pressure to produce a crude material that was purified by flash chromatography on silica gel (0 to 8% MeOH in CH2 Cl2, v / v) to produce di-DMTr-phosphorothioate 7c dimer ( 4.6 g, ~ 92%, approximately 90% purity). ESI-MS: m / z 1476.90 [M + H] + .
Petition 870190060374, of 06/28/2019, p. 83/142
80/122
Step 2: preparation of compound 7d [0153] The dimer 7c (4.5 g, 3.05 mmol) was dissolved in 80% AcOH: aqueous acetonitrile (90 mL, 8: 2, v / v). After stirring the reaction mixture for 20 h at 45 ° C, the mixture was diluted with ethyl acetate (400 ml) and washed sequentially with saturated aqueous NaHCOs (3 x 80 ml) and brine (1 x 50 ml). The aqueous layer was separated and extracted with ethyl acetate (1 x 20 ml). The combined organic phases were concentrated to dryness under reduced pressure and the resulting crude residue was purified by flash chromatography on silica gel (0 to 15% MeOH in CH2 Cl2, v / v) to produce the 7d dimer (1.65 g, 63 %). ESI-MS: m / z872.10 [M + H] + .
Step 3: preparation of compound 7f [0154] The dimeric compound 7d (275 mg, 0.315 mmol) was coevaporated with a mixture of anhydrous toluene / anhydrous acetonitrile (1: 1, v / v, 3 x 10 mL), and then , dissolved in anhydrous acetonitrile (20 mL, sonicated for 5 minutes for complete solubility of compound 7d). Molecular sieves of 4 Á powder (0.6 g) and tetrazole in acetonitrile (5.6 ml, 0.45 M, 2.52 mmols) were then added. The resulting heterogeneous mixture was purged with bubbling Ar ( g ) for 4 minutes. After stirring the mixture at room temperature for 10 minutes, 2-cyanoethyl Λ /, ΛΖ-diisopropyl chlorophosphoramidite (142 mg, 0.473 mmol, 1.5 eq) was added at room temperature in five portions over 20 minutes. After stirring for 90 minutes at room temperature, the reaction mixture was diluted with ethyl acetate (60 ml), filtered, and the filtrate was washed sequentially with saturated aqueous NaHCOs (1 x 20 ml) and brine (1 x 20 ml ). The filtrate was then dried (MgSO4) with stirring for 5 minutes, filtered, and the filtrate was concentrated to dryness under reduced pressure to yield compound 7e (ESI MS: m / z971.10 [M + 1] + .) . The resulting residue was used in the next step without further purification.
Petition 870190060374, of 06/28/2019, p. 84/142
81/122 [0155] The crude compound 7e was dissolved in anhydrous dichloromethane (25 ml), to which 4 4 powdered molecular sieves (0.5 g) were added. The resulting heterogeneous mixture was purged with bubbling Ar (g) for 4 minutes. Upon stirring the mixture at room temperature for 10 minutes, the mixture was cooled to 0 ° C. A solution of borane-dimethyl sulfide complex (2.0 M in THF, BH 3 -DMS, 550 pL, 3.5 eq) was added very slowly over 5 minutes at 0 ° C, and the reaction mixture was stirred at room temperature for 12 minutes. The mixture was then quickly filtered, diluted with ethyl acetate (80 ml), and quenched with water (20 ml). The organic phase was washed with brine (1 x 20 ml), and the aqueous layer was extracted with ethyl acetate (1x20 ml). The combined organic layers were concentrated to dryness under reduced pressure to produce a crude residue, which was purified by flash chromatography on silica gel (0 to 10% MeOH in dichloromethane, v / v) to produce a mixture of 7f diastereoisomers (130 mg, ~ 42% for two steps). ESI-MS: m / z 984.95 [M + H] +
Step 4: preparation of compound 7q and compound 7h [0156] The mixture of diastereoisomers 7f (130 mg) was dissolved in an aqueous mixture of ammonia / ethanol (7 mL, 3: 1, v / v). After stirring the reaction mixture for 18 h at 50 ° C, the reaction mixture was concentrated to dryness and coevaporated with ethanol (2 x 10 ml) and toluene (2 x 20 ml). The resulting crude solid was washed with dichloromethane (15 mL), collected by filtration and purified by preparative reverse phase HPLC (column: Synergi 4 μ, Hydro RP, 250 mm x 30 mm, Mobile Phase: Buffer A: triethyl ammonium acetate 50 mM in H 2 O; Buffer B: 50 mM triethyl ammonium acetate in CH 3 CN, gradient: 0 to 30% B over 30 minutes, flow rate of 24 mL / min) to produce a smaller boronophosphotioate isomer less than 7g ( 8.7 mg) and a second isomer of
Petition 870190060374, of 06/28/2019, p. 85/142
82/122 boranophosphothioate greater 7 h (13.1 mg) as salts of triethyl ammonium acetate (TEAA). ESI-MS: m / z 703.1 [M-1] ’.
Step 5: preparation of compound 9 and compound 10 [0157] Dowex 50W x 8, 200 to 400 (5 ml, form H) was added to a beaker and washed with deionized water (30 ml). Then, to the resin 15% H2SO4 in deionized water was added, the mixture was gently stirred for 5 minutes, then decanted (30 ml). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 CV), and then with deionized water until neutral pH 7 was reached. The resin was transferred back to the beaker, 15% NaOH in deionized water solution was added, the mixture was gently stirred for 5 minutes, then decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 CV), then with deionized water until neutral pH 7 was reached. Each isomer of boranophosphotopate 7 g (8.7 mg) and 7 h (13.1 mg) of TEAA salts was dissolved in a minimum amount of deionized water, added to the top of the column, and eluted with deionized water. Suitable fractions of compounds 9 and 10 were pooled together and lyophilized to produce compound 10 (7.8 mg) and compound 9 (12.4 mg) respectively as a sodium salt.
[0158] Compound 9 (major isomer): NMR H 1 (400 MHz, D2O): δ 8.07 (s, 1H), 7.97 (s, 1H), 7.86 (s, 1H), 6, 25 (d, J = 16.4 Hz, 1H), 5.86 (d, J = 8.4 Hz, 1H), 5.52 (d, J = 3.6 Hz, 0.5H), 5, 40 (d, J = 3.6 Hz, 0.5H) 5.16-5.19 (m, 1H), 4.83-4.89 (m, 1H), 4.41 - 4.45 (m , 2H), 4.25-4.32 (m, 2H), 4.06-4.17 (m, 2H), 3.88-3.94 (m, 1H), 3.46 (s, 3H ), -0.1 to 0.65 (very broad peak, 3H); P 31 NMR (162 MHz, D2O): δ 94-95 (very broad peak, boranophosphate), 52.59 (phosphorothiate); F 19 NMR (379 MHz, D 2 O): δ -201.7 (multiplet); ESI-MS: m / z: 703.1 [M-1] '.
[0159] Compound 10 (minor isomer): NMR H 1 (400 MHz, D2O): δ 8.18 (s, 1H), 8.13 (s, 1H), 8.07 (s, 1H), 6, 31 (d, J = 15.6 Hz, 1H), 5.91
Petition 870190060374, of 06/28/2019, p. 86/142
83/122 (d, J = 8.4 Hz, 1H), 5.65 (d, J = 2.8 Hz, 0.5H), 5.50 (d, J = 2.8 Hz, 0.5H ) 5.07-5.30 (m, 2H), 4.40-4.48 (m, 2H), 4.20-4.35 (m, 2H), 4.10-4.14 (m, 1H), 3.96-4.00 (m, 1H), 3.83-3.88 (m, 1H), 3.48 (s, 3H), 0.2 to 0.8 (very broad peak, 3H); P 31 NMR (162 MHz, D2O): δ 94-95 (very broad peak, boranophosphate), 57.79 (phosphorothiate); F 19 NMR (379 MHz, D2O): δ -202.4 (multiplet); ESI-MS: m / z: 703.1 [M-1] '.
Example 8
Compound 11
Compound 12
7b 8a
RT = TA
Petition 870190060374, of 06/28/2019, p. 87/142
84/122
Compound 11 ammonium salt Compound 12 sodium salt
Step 1: preparation of compound 8a [0160] To a solution of compound 7b (8 g, 5.538 mmols) in CH2 Cl2 (80 ml) was added 4 Á powdered molecular sieves (8 g) and the resulting heterogeneous mixture was bubbled with argon for 4 minutes. After stirring at room temperature for 30 minutes, a solution of borane-dimethyl sulfide complex (2.0 M in
THF, BH3-DMS, 9.138 ml, 18.277 mmoles) was added very slowly over 5 minutes at 0 ° C. After stirring the reaction for two hours at room temperature, the reaction mixture was quickly filtered, diluted with CH2 Cl2 (40 ml) and quenched with water (50 ml). The phases were separated and the organic layer was successively washed with water (1 x 50 ml_), brine (1 χ 50 ml_) and the aqueous layers were extracted again with CH2 Cl2 (1 χ 50 ml_). The combined organic layers were concentrated under reduced pressure until dry and the resulting crude material was purified by flash column chromatography on silica gel (petroleum ether / EtOAc = 1/0 ~ 0/1) to produce compound 8a (7, 5 g, 5.143 mmols) as a pale yellow oil.
Step 2: preparation of compound 8b [0161] Compound 8a (7.5 g, 5.143 mmols) was added to a
Petition 870190060374, of 06/28/2019, p. 88/142
85/122 CH 3 CN solution (20 ml_) in 80% aqueous acetic acid solution (60 ml_) (CH 3 CN / aqueous acetic acid solution = 1/3, 48 ml_ acetic acid, 12 ml H 2, 20 mL CH3CN). After stirring the reaction mixture overnight at 25 ° C, triethyl silane was added and the reaction was stirred at 25 ° C for an additional hour. The reaction mixture was filtered quickly, diluted with EtOAc (50 ml) and abruptly cooled with water (20 ml). The pH was adjusted to 7 to 8 with saturated aqueous NaHCO 3 solution. The phases were separated and the organic layer was washed successively with water (1 x 100 ml), brine (1 x 100 ml) and concentrated under reduced pressure until dry. The residue was purified by flash column chromatography on silica gel (petroleum / EtOAc = 0/1 then ChhCb / MeOH = 1/0 at 20/1) to produce compound 8b (6.5 g, 7.126 mmols) as a white solid. ESI-MS: m / z
854.1 [M + 1] + . NMR H 1 (400 MHz, DMSO-d 6 ) δ 12.07 (br d, J = 13.5 Hz, 1H), 11.56 (d, J = 9.7 Hz, 1H), 11.22 ( br d, J = 7.1 Hz, 1H), 8.72 (d, J =
3.1 Hz, 1H), 8.56 - 8.41 (m, 1H), 8.27 - 8.18 (m, 1H), 8.02 (br d, J = 6.2 Hz, 2H), 7 , 65 - 7.60 (m, 1H), 7.56 - 7.50 (m, 2H), 6.35 (br t, J = 19.0 Hz, 1H), 6.00 (d, J = 6.4 Hz, 1H), 5.95 (t, J = 6.5 Hz, 1H), 5.65 - 5.43 (m, 1H), 5.34 - 5.20 (m, 2H), 4.75 - 4.55 (m, 1H), 4.16 - 3.97 (m, 6H), 3.92 - 3.82 (m, 1H), 3.67-3.49 (m, 2H ), 3.14 (d, J = 5.1 Hz, 3H), 2.81 -2.66 (m, 3H), 1.08 (br d, J = 6.8 Hz, 6H), 0, 59 - -0.18 (m, 3H); P 31 NMR (162 MHz, DMSO-de) 115.80 (br s, 1P).
Step 3: preparation of compound 8c [0162] Compound 8b (1 g, 1.096 mmol) was coevaporated with CH 3 CN (3 x 20 ml) and dissolved in anhydrous CH 3 CN (44 ml). It was then added to 4 Á molecular powder sieves (1000 mg) and after stirring the mixture for 30 minutes, a solution of tetrazole in CH 3 CN (0.45 M, 14.6 ml, 6 eq) was added at room temperature. The resulting mixture was stirred for 15 minutes at room temperature. A solution of 3
Petition 870190060374, of 06/28/2019, p. 89/142
86/122 ((bis (diisopropylamino) phosphanyl) oxy) propanenitrile (495.647 mg, 1.644 mmol) in CH 3 CN (5.0 mL) for 20 minutes. After stirring the reaction mixture for one hour, an additional solution of tetrazole in CH 3 CN (0.45 M, 9.7 mL, 4 eq) was added to that mixture at room temperature. The reaction mixture was filtered and the filtrate extracted with EtOAc (3 x 400 ml). The combined organic layers were washed successively with aqueous NaHCO 3 (3 x 200 ml), brine (3 x 200 ml), dried with anhydrous Na 3 SO4, filtered and the filtrate was concentrated under reduced pressure. The residue was purified by flash column chromatography on silica gel (CH2 Cl2: MeOH 100: 0 to CH2 Cl2: MeOH 90: 10) to yield compound 8c (600 mg, 0.63 mmol) as a white solid.
Step 4: preparation of compound 8d [0163] To a solution of compound 8c (600 mg 0.63 mmol) in CH2 Cl2 (15 mL) was added a solution of 2 M dimethyl borane sulfide complex in THF (1039.276 pL, 2.079 mmols) very slowly over 5 minutes at 0 ° C. After stirring the reaction mixture at 0 ° C for 15 minutes, the reaction was quenched with MeOH (20 ml) at 0 ° C, then concentrated under reduced pressure. The reaction was repeated a second time using the same scale. The two crude batches were combined and purified by flash column chromatography on silica gel (gradient eluent: CH2 Cl2: MeOH 100: 0 to CH2 Cl2: MeOH 90: 10) to produce compound 8d (1 g, 1.035 mmol, combined batches ) as a white solid. ESI-MS: m / z = 966.4 [M + 1] + .
Step 5: preparation of compounds 11 and 12 [0164] Compound 8d (1.0 g, 1.035 mmol) was treated with a solution of MeNH 2 in EtOH (33%, 10 ml). After stirring at room temperature for 3 hours, the reaction mixture was concentrated under reduced pressure and the residue was purified by
Petition 870190060374, of 06/28/2019, p. 90/142
87/122 high performance preparative liquid chromatography (HPLC Prep condition: Column: Phenomenex Kinetex XB-C18 150 mm x 30 mm, 5 pm; Conditions: H2O (A) -CHsCN (B); Beginning B: 0; End B : 20; Flow: 25 mL / min). The pure fractions were collected and lyophilized until dry to produce the crude compound 11 (0.11 g, 0.160 mmol) and the crude compound 12 (0.14 g, 0.204 mmol) as white solids.
[0165] Crude compounds 11 and 12 were further purified by preparative high performance liquid chromatography (HPLC Prep conditions: Column: DuraShell 150 x 25 mm χ 5um; Condition: water (NH4HCO3 10 mM) (A) -CHsCN (B) ); Beginning B: 0; End B: 15; Flow: 35 mL / min). The pure fractions were collected and lyophilized until dry to produce compound 11 (0.08 g, 0.117 mmol) and compound 12 (0.04 g, 0.058 mmol), each as a white solid.
[0166] Compound 11: NMR H 1 (400 MHz, D 2 O) δ 8.21 (d, J = 13.2 Hz, 2H), 8.05 (s, 1H), 6.36 (d, J = 16.3 Hz, 1H), 5.93 (d, J = 8.2 Hz, 1H), 5.73 - 5.55 (m, 1H), 5.19 - 5.02 (m, 2H) , 4.55 - 4.48 (m, 2H), 4.34
- 4.24 (m, 2H), 4.16 (d, J = 4.4 Hz, 1H), 4.05 - 3.94 (m, 2H), 3.57 (s, 3H), 0, 76 - 0.13 (m, 3H), -0.32 (br s, 3H); ESI-MS: m / z = 686.9 [M + 1] + . NMR F 19 (376 MHz, D 2 O) -202.02 (td, J = 20.0, 50.3 Hz, 1F); P 31 NMR (162 MHz, D2O) oppm - 94.42 (br s, 1 P).
[0167] Compound 12: NMR dH 1 (400 MHz, D 2 O) ó8.31 (s, 1H), 8.23 (s, 1H), 7.86 (brs, 1H), 6.40 (br d , J = 15.4 Hz, 1H), 5.89 (br d, J = 8.6 Hz, 1H), 5.64 - 5.47 (m, 1H), 5.35 (br s, 1H) , 5.00 - 4.86 (m, 1H), 4.54
- 4.45 (m, 2H), 4.38 (br d, J = 11.7 Hz, 1H), 4.20 (br d, J = 17.0 Hz, 2H), 4.07 (br d , J = 8.2 Hz, 1H), 3.96 (br d, J = 10.1 Hz, 1H), 3.53 (s, 3H), 0.24 (br s, 6H); ESI-MS: m / z = 686.9 [M + 1] + . 19 F NMR (376 MHz, D2O) -200.77 - -202.57 (m, 1F); P 31 NMR (162 MHz, D2O) 99.68 - 84.67 (m, 1P).
Petition 870190060374, of 06/28/2019, p. 91/142
88/122
Step 6: preparation of the sodium salt of compound 11 and the sodium salt of compound 12 [0168] Compound 11, sodium salt. Dowex 50W x 8, 200 to 400 (form H, 50 g) was added to a beaker (for 45 mg of the compound
11) and washed with deionized water (2x), then added to the resin (15% H2SO4 in deionized water, 50 mL). The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 11 was dissolved in deionized water (50 mg in 40 ml), added to the top of the column, and eluted with deionized water. Compound 11 was eluted from the column in initial fractions as detected by TLC (UV). The mixture was lyophilized to produce the sodium salt of compound 11 (24.3 mg, 0.033 mmol) as a white solid. ESI-MS: m / z = 686.9 [M + 1] + . NMR H 1 (400 MHz, D2O) δ 7.81 (2 H, d, J = 3.6 Hz), 7.71 (1 H, s), 5.99 (1 H, d, J = 15, 6 Hz), 5.61 (1 H, d, J = 8.2 Hz), 5.16 5.32 (1 H, m), 4.65 - 4.84 (2 H, m), 4, 19 - 4.28 (2 H, m), 3.96 - 4.11 (2 H, m), 3.88 (1 H, d, J = 4.2 Hz), 3.69 - 3.80 (2 H, m), 3.31 (3 H, s), -1.07 - 0.45 (6 H, m); 19 F NMR (377 MHz, D2O) -202.06 (1 F, s); P 31 NMR (162 MHz, D 2 O) 94.39 (1 P, brs).
[0169] Compound 12, sodium salt. Dowex 50W x 8, 200 to 400 (form H, 50 g) was added to a beaker (for 50 mg of the compound
12) and washed with deionized water (2x), then added to the resin (15% H2SO4 in deionized water, 50 mL). The mixture was stirred
Petition 870190060374, of 06/28/2019, p. 92/142
89/122 for 15 minutes and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 12 was dissolved in deionized water (50 mg in 40 ml), added to the top of the column, and eluted with deionized water. Compound 12 was eluted from the column in initial fractions as detected by TLC (UV). The mixture was lyophilized to produce the sodium salt of compound 12 (43.3 mg, 0.059 mmol) as a white solid. ESI-MS: m / z = 686.9 [M + 1] + . NMR H 1 (400 MHz, D2O) δ 8.11 (s, 1 H), 8.06 (s, 1H), 7.94 (s, 1H), 6.28 (d, J = 16.06 Hz , 1H), 5.89 (d, J = 8.53 Hz, 1H), 5.44-5.60 (m, 1H), 5.27 (td, J = 8.97, 4.39 Hz, 1H), 4.86 - 5.00 (m, 1H), 4.46 - 4.55 (m, 2H), 4.37 (br d, J = 11.80 Hz, 1H), 4.19 - 4.28 (m, 1H), 4.19 - 4.28 (m, 1H), 4.15 (br dd, J = 11.67, 2.13 Hz, 1H), 4.02 (br d, J = 12.30 Hz, 1H), 3.54 (s, 3H), -0.01 - 0.75 (m, 6H); 19 F NMR (377 MHz, D2O) -201.59 (s, 1 F); P 31 NMR (162 MHz, D 2 O) 94.01 (br s, 1 P). Example 9
O o
14
Petition 870190060374, of 06/28/2019, p. 93/142
90/122
RT = ΤΑ
BzHN BzHN
8b 8c
O
BzHN
9b
9a Compound 13
nh 2 nh 2
Compound 13, ammonium salt Compound 13, sodium salt
Petition 870190060374, of 06/28/2019, p. 94/142
91/122
9b 9c
Compound 14, ammonium salt Compound 14, sodium salt
Step 1: preparation of compound 8a [0170] Compound 7b (10.46 g, 7.241 mmols) was dissolved in anhydrous CH2Cl2 (100 mL) and 4 Á powder molecular sieves (4.0 g) were added to this solution . The resulting heterogeneous mixture was degassed under reduced pressure and purged with Argon several times. After stirring this mixture at 25 ° C for 30 minutes, borane-dimethyl sulfide complex solution (2.0 M in THF, BH3-DMS, 11.948 mL, 23.897 mmols) was added very slowly over 5 minutes at 0 ° C . After stirring the reaction for 20 minutes at 25 ° C, the reaction mixture was filtered quickly, diluted with CH2 Cl2 (120 ml) and abruptly cooled with water (20 ml). The organic layer was washed successively with water (50 ml) and brine (50 ml). The aqueous layers were extracted again with EtOAc (50 ml). At
Petition 870190060374, of 06/28/2019, p. 95/142
92/122 combined organic layers were concentrated under reduced pressure until dry and the resulting crude material was purified by flash column chromatography on silica gel (CH 2 Cl2 / MeOH = 100/1 to 10/1) to produce compound 8a ( 11.1 g, 7.612 mmols) as a yellow solid.
Step 2: preparation of compound 8b [0171] Compound 8a (11.1 g, 7.612 mmols) was dissolved in 80% aqueous AcOH / CH 3 CN / Et 3 SiH (3/1/1, v / v, 200 mL). After stirring the mixture for 16 h at 37 ° C, the reaction mixture was diluted with EtOAc (500 ml) and then neutralized with saturated aqueous NaHCO 3 . The organic layer was washed successively with water (500 ml) and brine (2 x 250 ml), then dried with Na 2 SO4, filtered, and the filtrate was evaporated to dryness to produce a residue. The residue was purified by flash column chromatography on silica gel (CHECk / MeOH = 100/1 to 10/1) to yield compound 8b (3.6 g, 4.218 mmol) as a white solid. ESI-MS: m / z854.2 [M + H] +; F 19 NMR (376 MHz, CD3CN) δ -203.09 (s, 1F), -203.36 (s, 1F); P 31 NMR (162 MHz, CD3CN) δ 116.95-116.34 (m, 1P).
Step 3: preparation of compound 8c [0172] Compound 8b (1.5 g, 1.757 mmol) was coevaporated with a mixture of anhydrous toluene / CH 3 CN (1/1, v / v, 3 x 20 mL) to produce a white solid. The solid was then dissolved in CH 3 CN (80 ml) and 4 Á molecular powder sieves (2.0 g) were added to this solution. After stirring this mixture at 25 ° C for 30 minutes, a solution of tetrazole in CH 3 CN (0.45 M, 31.242 ml, 14.059 mmol) was added to the solution at 25 ° C. The reaction mixture was stirred for 20 minutes at 25 ° C. 2-Cyanoethoxybis- (N, N-diisopropylamino) phosphine (794.519 mg, 2.636 mmoles) was added to the solution over 20 minutes (in five portions). After stirring for 60 minutes, another portion of the tetrazole solution in CH 3 CN (0.45
Petition 870190060374, of 06/28/2019, p. 96/142
93/122
Μ, 10 mL) was added to this solution. After stirring for an additional hour, the reaction mixture was diluted with EtOAc (100 ml) and filtered. The organic layer was washed with saturated aqueous NaHCOs (50 ml), brine (50 ml), dried with MgSCU (after stirring for 5 minutes followed by filtration), and the filtrate was evaporated to dryness to yield compound 8c (1.76 g, raw) as a white solid. The resulting solid was used directly without further purification in the next step. Step 3 was repeated a second time using the same scale.
Step 4: preparation of compounds 9a and 9b [0173] To a solution of compound 8c (1.76 g, 1.848 mmol) in MeCN (30 ml) was added 3H-benzo 1,1-dioxide [c] [1 , 2] dithiol-3-one 1.85 g, 9.238 mmols) at 25 ° C. After stirring at 25 ° C for one hour, the reaction mixture was filtered, and the resulting cake was washed with CHLCk / MeOH (10/1, 20 ml x 3). The combined filtrates were concentrated under pressure to produce a residue. The residue was purified by flash column chromatography on silica gel (CHLCk / MeOH = 100/1 to 10/1) to produce compound 9a (652 mg) as a yellow foam and compound 9b (660 mg) as a foam Yellow.
[0174] Compound 9a was purified again by preparative reverse phase HPLC (column: Phenomenex Gemini C18 250x50 10 pm; mobile phase: water (NH4HCO310 mM) -ACN, Beginning B: 30; End B: 60; Flow: 25 mL / min gradient time: 15 min) to produce compound 9a (225 mg, 0.229 mmol) as a white solid. Compound 9b was purified again by preparative reverse phase HPLC (column: Waters Xbridge 150x25 5 pm; mobile phase: water (10 mM NH4HCO3) -ACN, Beginning B: 37; End B: 67; Flow: 25 mL / min of gradient time: 8 min) to produce 9b (351 mg, 0.356 mmol, 19.294% yield) as a white solid. ESI-MS: m / z 985.5 [M + H] +
Petition 870190060374, of 06/28/2019, p. 97/142
94/122
Step 5: Preparation of compound 13, ammonium salt [0175] Compound 9a (100 mg, 0.102 mmol) was treated with MeNH 2 (33% in EtOH, 5 ml) and stirred at 25 ° C for 12 hours. The reaction mixture was concentrated under reduced pressure to produce a residue. The residue was purified by preparative reverse phase HPLC (column: Agela Durashell C18 150x25 5 pm; mobile phase: water (NH4HCO310 mM) -ACN, Beginning B: 0, End B: 15%, flow rate: 35 ml / min, Time Gradient: 10 min) to produce compound 13, ammonium salt (56 mg, 0.08 mmol) as a white solid. ESI-MS: m / z 704.8 [M + H] + NMR H 1 (400 MHz, D2O) δ 8.30 (br, s, 1H), 7.93 (br s, 2H), 6.41 (br, d, J = 15.8 Hz, 1H), 5.98 - 5.68 (m, 2H), 5.57 (br, s, 1H), 5.24 - 4.95 (m, 1H ), 4.62 - 4.35 (m, 3H), 4.29 - 4.12 (m, 3H), 4.02 (br d, J = 9.3 Hz, 1H), 3.60 (s , 3H), -0.48 (br, s, 3H); F 19 NMR (376 MHz, D2O) -199.64-201.37 (m, 1F); NMR P 31 (162 MHz, D 2 O) 91.37 (s, 1P), 91.31 (s, 1 P), 90.52 (s, 1 P), 89.94 (s, 1P), 52 , 36 (s, 1P), 52.26 (s, 1P).
Step 6: preparation of compound 13, sodium salt [0176] Dowex 50W x 8, 200 to 400 (form H, 50 g) was added to a beaker (for 56 mg of compound 13) and washed with deionized water (2x) , then added to the resin (15% H2SO4 in deionized water, 50 mL). The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 12 was dissolved in deionized water (50 mg in 40 ml), added to the top of the column, and eluted with water
Petition 870190060374, of 06/28/2019, p. 98/142
95/122 deionized. Compound 12 was eluted from the column in initial fractions as detected by TLC (UV). Compound 13 was dissolved in deionized water (56 mg in 30 ml), added to the top of the column, and eluted with deionized water. The product was eluted from the column in the initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 13, sodium salt (45.4 mg, 0.064 mmol) as a white solid. NMR H 1 (400 MHz, D2O) δ 8.25 (s, 1H), 8.21 (s, 1H), 7.95 (s, 1H), 6.42 (d, J = 14.8 Hz, 1H), 5.91 - 5.84 (m, 1.5H), 5.76 (d, J = 3.8 Hz, 0.5H), 5.60 - 5.51 (m, 1H), 5 , 19 - 5.04 (m, 1H), 4.61 - 4.53 (m, 2H), 4.52 - 4.44 (m, 1H), 4.27 - 4.18 (m, 3H) , 4.07 (dd, J = 4.0, 12.0 Hz, 1H), 3.60 (s, 3H), 0.47 - -0.89 (m, 3H); F 19 NMR (377 MHz, D2O) -201.93 (s, 1F); P 31 NMR (162 MHz, D 2 O) δ 92.47 (br dd, J = 26.4, 73.4 Hz, 1P), 91.98 (s, 1P), 91.71 (s, 1P) , 91.24 (s, 1P), 91.19 (s, 1P), 91.10 (s, 1P), 90.97 (s, 1P), 52.78 (s, 1P), 52.64 ( s, 1P); ESI-MS: m / z704.8 [M + H] +
Step 7: preparation of compound 9c [0177] To a solution of compound 9b (311 mg, 0.316 mmol) in MeCN / EtOH (1 / 1.5 mL) was added tert-butylamine (5 mL). After stirring for two hours, the reaction mixture was concentrated under reduced pressure to produce a residue. The residue was purified by preparative reverse phase HPLC (column: Waters Xbridge 150x25 5 pm; mobile phase: water (NH4HCO310 mM) -ACN, Beginning B: 8; End B: 38; Flow: 25 mL / min of gradient time : 8 min) to produce compound 9c (243 mg, 0.277 mmol) as a white solid.
Step 8: preparation of compound 14, ammonium salt [0178] Compound 9c (243 mg, 0.277 mmol) was treated with MeNH 2 (33% in EtOH, 10 mL) and stirred at 25 ° C for 12 hours. The reaction mixture was then concentrated under reduced pressure to produce a residue. The residue was purified by preparative reverse phase HPLC (column: Agela Durashell C18 150x25 5 pm; mobile phase: water (10 mM NH4HCO3) -ACN, Beginning B: 0, End B: 15%, flow rate: 35
Petition 870190060374, of 06/28/2019, p. 99/142
96/122 ml / min, Gradient Time: 10 min) to produce compound 14, ammonium salt (125 mg, 0.177 mmol) as a white solid. 1 H NMR (400 MHz, D2O) δ 8.23 (d, J = 2.5 Hz, 2H), 8.01 (s, 1H), 6.43 (d, J = 15.1 Hz, 1H) , 5.95 - 5.76 (m, 2H), 5.56 (dt, J = 4.3, 8.9 Hz, 1H), 5.21 5.05 (m, 1H), 4.64 - 4.46 (m, 3H), 4.33 - 4.15 (m, 4H), 3.56 (s, 3H), 0.96 - -0.39 (m, 3H); 19 F NMR (377 MHz, D2O) δ -201.77 (s, 1F); P 31 NMR (162 MHz, D 2 O) δ 93.50 (s, 1P), 92.44 (s, 1P), 52.51 (s, 1P); ESI-MS: m / z 704.8 [M + H] +
Step 9: preparation of compound 14, sodium salt [0179] Dowex 50W x 8, 200 to 400 (form H, 50 g) was added to a beaker (for 125 mg of compound 14) and washed with deionized water (2x) , then added to the resin (15% H 2 SO4 in deionized water, 50 mL). The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to a column with 15% H 2 SO4 in deionized water and washed with 15% H 2 SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 14 was dissolved in deionized water (125 mg in 40 ml), added to the top of the column, and eluted with deionized water. The product was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 14, sodium salt (105.4 mg, 0.141 mmol) as a white solid. 1 H NMR (400 MHz, D2O) δ 8.21 (br, d, J = 9.0 Hz, 2H), 7.89 (s, 1H), 6.38 (d, J = 15.3 Hz, 1H), 5.89 - 5.72 (m, 2H), 5.67 (br, s, 1H), 5.19 5.02 (m, 1H), 4.62 - 4.42 (m, 3H ), 4.27 - 4.17 (m, 3H), 4.07 (br d, J = 9.8 Hz, 1H), 3.57 (s, 3H), 0.36 (br, s, 3H ); 19 F NMR (377 MHz, D2O) δ -201.25
Petition 870190060374, of 06/28/2019, p. 100/142
97/122 (s, 1F); P 31 NMR (162 MHz, D 2 0) δ 92.42 - 90.93 (m, 1P), 52.35 (s, 1P)
ESI-MS: m / z 704.8 [M + H] +
Example 10
O O
Compound 15 Compound 16
10c 10d 10e, R = OH and / or OCH 2 CH 2 CN
RT = TA
NH 4 OH / EtOH = 3/1
Petition 870190060374, of 06/28/2019, p. 101/142
98/122
Dowex- Na
Compound 15, sodium salt
Step 1: Preparation of compound 7b [0180] To a solution of compound 1f (5.0 g; 7.47 mmol) in acetonitrile (180 mL) was added 1 H-tetrazole (0.45 M, 132.7 mL ) at 25 ° C. After stirring the solution for 10 minutes at 25 ° C, a solution of compound 7a (6.87 g, 7.84 mmoles) in acetonitrile (7.84 20 ml) was added by dropping. After stirring for two hours at 25 ° C, the solution was used in the next step without further purification.
Step 2: Preparation of compound 10a [0181] To the previous solution of compound 7b (332.7 ml, 7.44 mmols) in acetonitrile was added tert-butyl hydroperoxide (3.35 g, 37.22 mmols) at 25 ° C ° C. After stirring at 25 ° C for 1.5 hours, the solution was diluted with EA (100 ml) and washed with saturated aqueous NaHCOs (2 x 100 ml) and brine (2 x 100 ml). The organic layer was dried successively with anhydrous NasSO4, filtered and the solvent evaporated under reduced pressure to produce compound 10a (10 g) as a white solid.
Step 3: Preparation of compound 10b [0182] To a solution of compound 10a (10 g, crude) in acetonitrile (50 ml) was added triethyl silane (40 ml) and 80% acetic acid in acetonitrile (200 ml) at 25 ° C. After stirring the solution at 50 ° C for 12 hours, the mixture was neutralized with saturated aqueous NaHCOs to pH 8. The mixture was diluted with EtOAc (500 ml) and the organic layer was washed successively with saturated aqueous NaHCOs solution
Petition 870190060374, of 06/28/2019, p. 102/142
99/122 (100 ml), brine (100 ml) and evaporated under reduced pressure until dry. The aqueous layer was extracted with EtOAc (2 x 200 ml) of EtOAc, and evaporated under reduced pressure until dry. The combined crude material was purified by flash column chromatography on silica gel (0 to 9% MeOH in CH2 Cl2, v / v) to yield compound 10b (5 g) as a white solid. ESI-MS: m / z 856.2 [M + H] + .
Step 4: Preparation of compound 10c [0183] To a solution of compound 10b (1.5 g, 1.4 mmol) in tetrahydrofuran (2 ml) and acetonitrile (75 ml) was added 1H-tetrazole (0.45 M , 15.58 ml, 7.01 mmols) at 25 ° C. A solution of 3 - ((bis (diisopropylamino) phosphine) oxy) propanonitrile (845.34 mg, 2.8 mmols) in acetonitrile (5 ml) was then added at 25 ° C. After stirring for 1.5 hours at 25 ° C, the solution was washed with saturated aqueous NaHCOs (50 ml), brine (50 ml) and evaporated under reduced pressure until dry to produce the compound 10c (1.8 g) as a yellow solid that was used in the next step without any further purification. ESI-MS: m / z
955.5 [M + H] + .
Preparation of compound 15
Step 5: Preparation of compounds 10d + 10e [0184] To a solution of compound 10c (1.5 g, 1.57 mmol) (100 mL) in DCM was added borane dimethyl sulfide (2.36 mL, 4, 71 mmols) 0 ° C for 2 minutes. After stirring the mixture at 25 ° C for 15 minutes, water (30 ml) was added. The resulting solution was filtered and the filtrate was concentrated under reduced pressure to produce a yellow solid. The solid was diluted with DCM (100 ml) and the organic layer was successively washed with water (2 x 100 ml), brine (3 x 100 ml) and concentrated under pressure to produce a residue. The crude solid was purified by flash column chromatography on silica gel (0 to 9% MeOH in CH2 Cl2, v / v) to produce a mixture of compounds 10d and 10e (500 mg, crude) as a yellow solid. ESI-MS: m / z 969.3 [M + H] +
Petition 870190060374, of 06/28/2019, p. 103/142
100/122
Step 6: Preparation of compound 15 [0185] A solution of compounds 10d and 10e (500 mg, crude) in a mixture of ethanol (14 ml) and NH4OH (42 ml) was stirred at 50 ° C for 12 hours. The solution was concentrated under pressure to produce a yellow solid. The yellow solid was purified by preparative reverse phase HPLC (Column: Synergi Polar-RP 100 x 30 5 μΜ; Condition: water (10 mM NH4HCO3) -ACN; Start B: 0, End B: 20; Gradient time (min ): 12; Flow rate (ml / min): 25) to produce compound 15 (110 mg) as a white solid. Compound 15 was purified for the second time by preparative reverse phase HPLC (Column: Phenomenex Kinetex XB-C18 150 mm x 30 mm, 5 μΜ; Condition: water (NH4HCO3 10 mM) -ACN; Beginning B: 0, End B: 5; Gradient time (min): 7; Flow rate (ml / min): 30) to produce compound 15, ammonium salt (45 mg) as a white solid. 1 H NMR (400 MHz, D2O) δ 8.31 (br, s, 1H), 8.15 (s, 1H), 7.94 (br, s, 1H), 6.43 (br, d, J = 16.3 Hz, 1H), 5.98 (br, d, J = 7.7 Hz, 1H), 5.67 - 5.47 (m, 1H), 5.28 (br, s, 1H) , 4.93 (br, s, 1H), 4.61 - 4.49 (m, 2H), 4.43 (br, d, J = 9.7 Hz, 1H), 4.26 (br, s , 2H), 4.18 (br, s, 1H), 4.05 (br s, 1H), 3.60 (s, 3H), 0.33 (br s, 3H). F 19 NMR (376 MHz, D2O) δ -201.82 (s, 1F). P 31 NMR (162 MHz, D 2 O) δ 96.09 (br, s, 1P), -2.40 (s, 1P). ESI-MS: m / z 688.9 [M + H] + .
Step 7: Preparation of compound 15, sodium salt [0186] Dowex 50W x 8, 200 to 400 (form H, 5 ml) was added to a beaker (for 45 mg of compound 15, ammonium salt) and washed with water deionized (2x), then added to the resin (15% H2SO4 in deionized water, 50 mL). The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a
Petition 870190060374, of 06/28/2019, p. 104/142
101/122 NaOH solution (15% NaOH in aqueous solution, 50 mL) was added. The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 15, ammonium salt, was dissolved in deionized water (45 mg in 5 ml), added to the top of the column, and eluted with deionized water. The product was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 15, sodium salt P1 (42.6 mg) as a white solid. 1 H NMR (400 MHz, D2O) δ 8.21 (s, 1H), 8.09 (s, 1H), 7.97 (s, 1H), 6.39 (br, d, J = 16.1 Hz, 1H), 6.00 (br, d, J = 8.3 Hz, 1H), 5.70 - 5.52 (m, 1H), 5.23 (dt, J = 4.5, 8, 3 Hz, 1H), 5,044.88 (m, 1H), 4.63-4.51 (m, 3H), 4.44 (br, d, J = 11.8 Hz, 1H), 4.31 4.19 (m, 3H), 4.06 (br, d, J = 10.8 Hz, 1H), 3.59 (s, 3H), 0.67-0.12 (m, 3H). F 19 NMR (376 MHz, D2O) δ -201.80 (s, 1F). P 31 NMR (162 MHz, D2O) δ 95.45 (s, 1P), -2.26 (s, 1P). ESI-MS: m / z688.8 [M + H] +
Preparation of compound 15 and compound 16
Step 5: Preparation of compounds 10d + 10e [0187] To a solution of compound 10c (2.3 g, 2.41 mmol) (100 mL) in DCM was added dimethyl borane sulfide (3.61 mL, 7, 23 mmols) 0 ° C for 2 minutes. After stirring the mixture at 25 ° C for 15 minutes, water (20 ml) was added. The resulting solution was filtered and the filtrate was concentrated under reduced pressure to produce a yellow solid. The solid was diluted with DCM (100 ml) and the organic layer was successively washed with water (2 x 100 ml), brine (3 x 100 ml) and concentrated under pressure to produce a yellow residue. The crude solid was purified by preparative reverse phase HPLC (Column: Agela Durashell C18 150 x 25 5 μΜ; Condition: water (NH4HCOs 10 mM) -ACN; Beginning B: 25, End B: 55; Gradient time (min) : 12; Flow (ml / min): 25) to produce a mixture of compounds 10d and 10e
Petition 870190060374, of 06/28/2019, p. 105/142
102/122 (65 mg) as a white solid. 1 H NMR (400 MHz, CD3OD) δ 8.81 8.71 (m, 1H), 8.50 (d, J = 6.2 Hz, 1H), 8.08 (br, d, J = 8, 4 Hz, 2H), 7.69 7.54 (m, 3H), 6.59 - 6.50 (m, 1H), 6.37 - 6.26 (m, 2H), 6.02 - 5, 86 (m, 1H), 5.26 - 5.04 (m, 2H), 4.77 - 4.68 (m, 1H), 4.63 - 4.54 (m, 3H), 4.43 ( br, d, J = 6.2 Hz, 2H), 4.34 - 4.27 (m, 2H), 3.86 - 3.69 (m, 2H), 2.98 - 2.90 (m, 3H), 2.71 (br, dd, J = 5.8, 13.1 Hz, 1H), 2.61 - 2.49 (m, 1H), 1.22 (td, J = 3.3, 6.7 Hz, 7H). ESI-MS: m / z969.3 [M + H] +
Step 6: Preparation of compound 15 and compound 16 [0188] A solution of compounds 10d and 10e (65 mg, crude) in a mixture of ethanol (4 ml) and NH4OH (12 ml) was stirred at 50 ° C for 12 hours. The solution was concentrated under pressure to produce a residue. The residue was purified by preparative reverse phase HPLC (column: Synergi Polar-RP 100 x 30 5 μΜ; Condition: water (NH4HCO310 mM) -ACN; Beginning B: 0, End B: 20; Gradient time (min): 12; Flow rate (ml / min): 25) to produce compound 15 (37 mg) and compound 16 (17 mg) as white solids.
[0189] Compound 15, ammonium salt: NMR H 1 (400 MHz, D2O) δ 8.41 (br, s, 1H), 8.23 (br, s, 1H), 7.98 (br, s, 1H), 6.46 (br, d, J = 16.3 Hz, 1H), 5.99 (br, s, 1H), 5.74 - 5.46 (m, 1H), 5.27 (br , s, 1H), 5.07-4.91 (m, 1H), 4.61 - 4.50 (m, 2H), 4.42 (br, s, 1H), 4.26 (br, s , 2H), 4.16 (br, s, 1H), 4.05 (br, s, 1H), 3.61 (s, 3H), 0.71 - 0.07 (m, 2H). 19 F NMR (376 MHz, D2O) δ -75.63 (s, 1F). P 31 NMR (162 MHz, D 2 O) δ 94.56 (s, 1 P), -3.69 (s, 1 P). ESI-MS: m / z 688.9 [M + H] + [0190] Compound 16, ammonium salt: 1 H NMR (400 MHz, D2O) δ 8.37 (br s, 1H), 8.24 ( br s, 1H), 7.85 (s, 1H), 6.42 (d, J = 14.1 Hz, 1H), 5.91 - 5.88 (m, 1H), 5.8 (br s , 1H), 5.51 (br s, 1H), 5.38 (br s, 1H), 5.32 5.20 (m, 1H), 4.56 - 4.48 (m, 2H), 4 , 45 - 4.36 (m, 1H), 4.28 - 4.17 (m, 3H), 4.09 (br d, J = 11.0 Hz, 1H), 3.59 (s, 3H) , 0.61 - 0.10 (m, 3H). 19 F NMR (376 MHz, D2 O) δ -202.9 (s, 1F). P 31 NMR (162 MHz, D 2 O) δ 96.67 (m, 1P), -3.02 (s, 1P). ESI-MS: m / z689.2 [M + H] +
Petition 870190060374, of 06/28/2019, p. 106/142
103/122
Step 7: Preparation of compound 15, sodium salt [0191] Dowex 50W x 8, 200 to 400 (form H, 5 ml) was added to a beaker (for 37 mg of compound 15, ammonium salt) and washed with water deionized (2x), then added to the resin (15% H2SO4 in deionized water, 50 mL). The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1x). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 15, ammonium salt, was dissolved in deionized water (37 mg in 5 ml), added to the top of the column, and eluted with deionized water. The product was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 15, sodium salt P1 (28.4 mg) as a white solid. 1 H NMR (400 MHz, D2O) δ 8.25 (s, 1H), 8.12 (s, 1H), 7.99 (s, 1H), 6.42 (br, d, J = 15.8 Hz, 1H), 6.02 (br, d, J = 8.3 Hz, 1H), 5.70 - 5.52 (m, 1H), 5.30 - 5.21 (m, 1H), 5 , 06 4.91 (m, 1H), 4.62 - 4.54 (m, 2H), 4.44 (br, d, J = 12.8 Hz, 1H), 4.30 (br, d, J = 4.3 Hz, 2H), 4.21 (br, d, J = 12.0 Hz, 1H), 4.06 (br, d, J = 11.8 Hz, 1H), 3.60 ( s, 3H), 0.71 - 0.09 (m, 3H). 19 F NMR (376 MHz, D2O) δ -75.62 (s, 1F), 201.89 (s, 1F). P 31 NMR (162 MHz, D 2 O) δ 96.08 (s, 1P), -2.24 (s, 1P). ESIMS: m / z 688.8 [M + H] +
Petition 870190060374, of 06/28/2019, p. 107/142
104/122
Example 11
nh 2 17 18
8b 11a
NH 4 OH, EtOH, 50 ° C
H 2 N h 2 n
Compound 17, sodium salt
Petition 870190060374, of 06/28/2019, p. 108/142
105/122
Ο
η 2 ν
Dowêx- Na
Compound 18, sodium salt
Step 1: preparation of (11 a) [0192] To a solution of 8b (100 mg, 0.11 mmol) in CH 3 CN / THF (1: 1, v / v, 4.4 ml_) was added sieves molecules of 4 Á (1 g) and 1Htetrazole in CH 3 CN (1.94 ml, 0.9 mmol). After stirring the mixture at 25 ° C for 0.5 hour, Ν, Ν, Ν ', Ν'-tetraisopropylphosphorodiamidite of 2-cyanoethyl (49.56 mg, 0.16 mmol) in CH 3 CN was added to the mixture. The mixture was stirred at 25 ° C for two hours and 1 H-tetrazole in CH 3 CN (0.49 ml, 0.22 mmol, 0.45M) was added to the mixture. After stirring the mixture at 25 ° C for 0.5 hour, a 0.5 M solution of 1 2 in THF: Py: H 2 O (8: 1: 1; V / V / V) (0.66 ml_ , 0.33 mmol) were added to the reaction. After stirring the mixture at 25 ° C for 2 h, a saturated aqueous solution of sodium thiosulfate (2 ml) was added; The resulting mixture was filtered and the filtrate was concentrated under reduced pressure until dry. The residue was purified by preparative reverse phase HPLC (Agela Durashell C18 150 x 25 5 μΜ; Condition: water (10 mM NH4HCO 3 ) -CAN A: water (10 mM NH4HCO 3 ) B: MeCN; Start B: 25% for B: 55%, Gradient Time (min) 12; 100% B, Retention Time (min) 2.2; Flow (ml / min) 25). The pure fractions were collected, the solvent was concentrated under reduced pressure and the aqueous layer was lyophilized until dry to produce 11a as a white solid (20 mg). ESI-MS: m / z 969.3 [M + 1] +.
Step 2: preparation of 17 and 18 [0193] To a solution of 11a (20 mg 0.017 mmol) in EtOH (1.5 ml) was added NH 3 .H 2 O (1.5 ml, 25%). After stirring the solution at 50 ° C for 3 days, the reaction mixture was filtered and the filtrate was concentrated
Petition 870190060374, of 06/28/2019, p. 109/142
106/122 under reduced pressure until dry. The residue was purified by preparative reverse phase HPLC (column: Syneri Polar-RP 100 x 30 5 μΜ Condition: water (NH4HCO310 mM) -MeCN A: water (NH4HCO310 mM) B: MeCN; Start B: 0% for B: 20%, Gradient Time (min) 12; 100% B, Retention Time (min) 2.2; Flow (ml / min) 25). The pure fractions were collected and the solvent was evaporated under reduced pressure to produce 17 (25 mg) and 18 (20 mg) as a white solid.
[0194] Analog 17, ammonium salt: ESI-MS: m / z 688.8 [M + 1] +. 1 H NMR (400 MHz, D 2 O) δδ8.26 (br s, 1 H) 8.18 (s, 1 H) 7.77 (br s, 1 H) 6.38 (br d, J = 14 , 31 Hz, 1 H) 5.77 (br d, J = 8.03 Hz, 1 H) 5.64 (br s, 1 H) 5.30 - 5.52 (m, 1 H) 4.95 - 5.13 (m, 1 H) 4.50 (br d, J = 9.03 Hz, 1 H)
4.34 - 4.44 (m, 2 H) 4.07 - 4.25 (m, 3 H) 3.99 (br d, J = 11.04 Hz, 1 H) 3.52 (s, 3 H) -0.92 - 0.05 (m, 3 H).
[0195] Analog 18, ammonium salt: ESI-MS: m / z 688.8 [M + 1] +. 1 H NMR (400 MHz, D 2 O) δ 8.29 (s, 1 H) 8.18 (s, 1 H) 7.75 (s, 1 H) 6.35 (d, J = 14.05 Hz, 1 H) 5.76 (s, 2 H) 5.30 - 5.52 (m, 1 H) 4.99 -5.20 (m, 1 H)
4.35 - 4.50 (m, 3 H) 4.09 - 4.22 (m, 3 H) 3.94 - 4.03 (m, 1 H) 3.47 (s, 3 H) 0.05 ( s, 3 H).
Step 3: preparation of 17, sodium salt [0196] Dowex 50W x 8, 200 to 400 (form H, 25 g) was added to a beaker (for 33 mg of compound 17) and washed with deionized water (2 x 10 mL), then added to the 15% H2SO4 resin in deionized water (80 mL). The mixture was stirred for 15 minutes and decanted (1x10 ml). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1 x 10 ml). The resin was transferred to the column and washed with 15% NaOH in water (at least
Petition 870190060374, of 06/28/2019, p. 110/142
107/122 column volumes) and then with water until it is neutral (at least 4 column volumes). Compound 17, ammonium salt, was dissolved in deionized water (33 mg in 5 ml), added to the top of the column, and eluted with deionized water. The product was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 17, sodium salt (12.4 mg) as a white solid. ESI-MS: m / z = 688.8 [M + 1] + . NMR H 1 (400 MHz, D 2 O) δ 8.08 - 8.05 (m, 1H), 7.97 (s, 1H), 7.37 -
7.36 (m, 1H), 6.41 - 6.33 (m, 1H), 5.88 (d, J = 8.0 Hz, 1H), 5.64 (d, J = 4.4 Hz , 1H), 5.44 - 5.27 (m, 2H), 4.48 (d, J = 2.4 Hz, 1H), 4.38 - 4.30 (m, 2H), 4.20 4 , 11 (m, 2H), 3.50 (s, 3H), 3.46 (d, J = 13.6 Hz, 1H), 3.22 - 3.18 (m, 1H); 19 F NMR (376 MHz, D2O) δ -196.87 (s, 1F). P 31 NMR (162 MHz, D2O) δ 7.80 (s, 1P), -1.22 (s, 1P).
Step 4: preparation of 18, sodium salt [0197] Dowex 50W x 8, 200 to 400 (form H, 15 g) was added to a beaker (for 30 mg of compound 18) and washed with deionized water (2x10 mL) , then added to the 15% H2SO4 resin in deionized water (80 mL). The mixture was stirred for 15 minutes and decanted (1x10 ml). The resin was transferred to a column with 15% H2SO4 in deionized water and washed with 15% H2SO4 (at least 4 column volumes), and then with deionized water until the resin was neutral. The resin was transferred back to the beaker, and a NaOH solution (15% NaOH in aqueous solution, 50 ml) was added. The mixture was stirred for 15 minutes and decanted (1 x 10 ml). The resin was transferred to the column and washed with 15% NaOH in water (at least 4 column volumes) and then with water until it was neutral (at least 4 column volumes). Compound 18, ammonium salt, was dissolved in deionized water (30 mg in 5 ml), added to the top of the column, and eluted with deionized water. The product was eluted in initial fractions as detected by TLC (UV). The product was lyophilized to produce compound 18, sodium salt
Petition 870190060374, of 06/28/2019, p. 111/142
108/122 (13.2 mg) as a white solid. ESI-MS: m / z = 688.8 [M + 1] + . NMR H 1 (400 MHz, D2O) δ 8.22 (s, 1 H) 8.14 (s, 1 H) 7.76 (s, 1 H) 6.34 (d, J = 14.05 Hz, 1 H) 5.77 (d, J = 8.28 Hz, 1 H) 5.60 - 5.69 (m, 1 H) 5.30 - 5.48 (m, 1 H) 4.94 - 5 , 11 (m, 1 H) 4.49 (br d, J = 9.29 Hz, 1 H) 4.35 - 4.45 (m, 2 H) 4.17-4.23 (m, 1 H ) 4.12 - 4.17 (m, 1 H) 4.09 (br d, J = 4.52 Hz, 1 H) 3.99 (br dd, J = 12.05, 4.52 Hz, 1 H) 3.52 (s, 3 H) -0.87 - 0.01 (m, 3 H). P 31 NMR (162 MHz, D2O) δ 92.38 (br s, 1 P) -1.31 (s, 1 P). 19 F NMR (376 MHz, D 2 O) δ -75.64 (s, 1 F) -202.91 (br s, 1 F).
[0198] By the method of Example 3, by substituting the appropriate reagents, the following compounds can be prepared by a person skilled in the art:
Compound No. Structure 19 0 OH /! * 1NH i <J O = P — O — IN N nh 2 0 ^ 0 pJ AoA OH À .N. n / rí V 1 - 0 —pC Νγλ / * g BH 3 nh 2 20 O j ™ O = p — O — 1 N ^ N ^ NHj 0 ^ 0 ΓΌ / OH Á rí Ίτ 1 - 0 —pC * bh 3 nh 2 21 o * bh 3 O = p — O — 1 N n N h 2 O ^ O PoA oh À rí Ίι 1 —o —- pC W * ii bh 3 nh 2
Petition 870190060374, of 06/28/2019, p. 112/142
109/122
Biological examples
In vitro tests
Biological example 1
SPA STING binding assay [0199] The human SPA STING binding assay measures the displacement of tritium marked with 2 ', 3'cGAMP (guanosine- (2' -> 5 ') monophosphate-adenosine- (3' -> 5 ') - cyclic monophosphate) to biotinylated STING protein. A soluble version of recombinant STING was expressed in E.coli which does not have the four transmembrane domains and contains residues 139 to 379 of Q86WV6 with R in position 232 (H232R). Based on the allele frequency of 58% of the population, H232R is considered to be wild type (Yi, et. Al., Single Nucleotide Polymorphisms of Human STING can affect innate immune response to cyclic dinucleotides PLOS ONE. 2013, 8 (10) , e77846). The STING construct has an N terminal of the HIS tag, followed by a
Petition 870190060374, of 06/28/2019, p. 113/142
110/122 divination of TEV protease and an AVI tag to allow biotinylation directed by biotin ligase BirA (Beckett et al., A minimal peptide substrate in biotin holoenzyme synthetase-catalyzed biotinylation. (1999) Protein Science 8, 921-929). The HIS tag is cleaved after purification and before biotinylation.
[0200] The assay was performed on 1536-well plates in a total volume of 8 μΙ_ per well by adding 8 nM of [ 3 H] -2'3'-cGAMP and 40 nM of biotin-STING protein in assay buffer [25 mM HEPES (Corning 25-060-C1) pH 7.5, mC NaC1150 (Sigma S5150), 0.5 mg / mL BSA (Gibco 15260-037), 0.001% Tween-20 (Sigma P7949), water molecular grade (Corning 46-000-CM)]. Test compounds (80 µl) were added with an acoustic dispenser (EDC Biosystems) in 100% DMSO to a final test concentration of 1% DMSO. The plates were centrifuged for 1 minute and incubated for 60 minutes at room temperature. Finally, streptavidin SPA (2 μΙ_) polystyrene capsules (PerkinElmer RPNQ0306) were added and the plates were sealed and centrifuged for 1 minute at room temperature. The plates were adapted to the dark for two hours and read in a ViewLux (Perkin Elmer) for 12 minutes per plate. A saturation binding curve for [ 3 H] -2'3'cGAMP showed a K d of 3.6 ± 0.3 nM for binding to STING, comparable to the values reported for the natural ligand (Zhang et al., Cyclic GMP-AMP containing mixed phosphodiester linkages is an endogenous high-affinity ligand for STING.) [0201] Other natural ligands including cyclic di-GMP also returned to the values in this assay within the expected range. The reference compound is cGAMP and the results are reported as percent inhibition and IC 50 values. The binding to the mouse STING used a construct similar to the one described above containing residues 138 to 378 of Q3TBT3.
Petition 870190060374, of 06/28/2019, p. 114/142
111/122
Full length human STING binding assay [0202] The human STING of residues 1 to 379 of Q86WV6 with an R in position 232 (H232R) with a 6HIS N-terminal tag followed by a FLAG tag, a protease dividing site TEV and an AVI tag for biotinylation was recombinantly expressed in HEK293-EXPI cells. The purified membranes were prepared from these cells and the expression of STING was confirmed and quantified by immunoblot. The membranes containing STING were combined with the test compound in a 384-well Greiner assay plate and incubated at room temperature for one hour in the same assay buffer used for the STING SPA binding assay. Then, [ 3 H] -2'3'-cGAMP was added and the plates were incubated for 30 minutes at room temperature. The reactions were transferred to a prewashed Pali 5073 filter plate and each well was washed 3 times with 50 µl of assay buffer. The filter plates were dried at 50 Q C for one hour. For each well, 10 pL of Microscint scintillation fluid was added and the plates were sealed and read in a TopCount (Perkin Elmer) for 1 minute per well.
STING SPR binding assay [0203] The compounds were analyzed on an S200 biacore SPR instrument (GE Healthcare). The truncated STING protein produced by E.coli was immobilized on a streptavidin S series chip through biotin capture (GE Healthcare # BR100531). The compounds were screened at 1: 2 dilutions from 100 µM to 0.195 µM in running buffer (10 mM HEPES, pH 7.4, 150 mM NaCI, 0.005% P20, 1 mM TECEP). The kinetic and affinity evaluations at steady state were performed using the 1: 1 binding model (STING was treated as a dimer). The test parameters were as follows: 60 seconds active, 300 seconds disabled for IFM compounds, cyclic di-GMP (60 s active / 60 s disabled),
Petition 870190060374, of 06/28/2019, p. 115/142
112/122 thiol isomer 1 (60 s active / 300 s deactivated) and cGAMP (60 s active / 1200 s deactivated) with a flow rate of 50 pL / min and data collection at 40 Hz at 25 ° C.
Human STING cell reporter assay [0204] Human STING pathway agonism is evaluated in THP1-ISG cells (Invivogen, cat # thp-isg) derived from human THP1 monocyte cell line by stable integration of a human construct SEAP reporter inducible by interferon regulating factor (IRF). THP1-Blue ISG cells express a secreted embryonic alkaline phosphatase reporter gene (SEAP) under the control of a minimal ISG54 promoter together with five interferon stimulated response elements (IFN). As a result, THP1 -Blue ISG cells allow control of IRF activation by determining SEAP activity. Levels of SEAP induced by IRF in the cell culture supernatant are readily evaluated with alkaline phosphatase detection medium, a SEAP detection reagent. These cells are resistant to Zeocina. 2'3'cGAMP was used as a positive control in the assay. To perform the assay, 60,000 cells were dispensed in 30 pL / well of a 384-well, white, opaque bottom plate, treated with tissue culture.
[0205] The test compounds were added in a volume of 10 pL (final concentration of 1% DMSO). The compounds are prepared in 100% DMSO initially, deposited in an intermediate dilution plate and then diluted in the medium before transfer. The assay was incubated for 24 hours at 37 ° C, 5% CO2 then the plates were centrifuged at 1200 rpm (120x g) for 5 minutes. After the final incubation, 90 µl of alkaline phosphatase detection medium substrate was added to each well of a new 384-well clear plate and 10 µl of the cell supernatant was transferred from the assay plate to the new detection medium plate.
Petition 870190060374, of 06/28/2019, p. 116/142
113/122 of alkaline phosphatase using a Biomek FX and mixed 4 times. The plates were incubated at room temperature for 20 minutes and then the absorbance at 655 nm was determined with Tecan Safire2. Mouse STING cell reporter assay [0206] The agonism of the mouse STING pathway is evaluated in RAW Lucia cells (Invivogen, cat # rawl-isg) derived from the mouse macrophage cell line RAW-264.7 by stable integration of a construct of Lucia's luciferase reporter inducible by interferon. RAW Lucia cells express a luciferase reporter gene secreted under the control of an ISG54 minimal promoter together with five interferon stimulated response (IFN) elements. As a result, RAW Lucia cells allow monitoring of IRF activation by determining luciferase activity. Levels of IRF-induced luciferase in the cell culture supernatant are readily evaluated with QUANTI-Luc ™, a luciferase detection reagent. These cells are resistant to Zeocina. 2'3'cGAMP is used as a positive control in this assay. To perform the assay, 100,000 cells were dispensed in 90 pL / well from a clear, flat, 96-well plate, treated with tissue culture. Test compounds were added in a volume of 10 pL. The assay was incubated for 24 hours at 37 ° C and 48 hours at 5% CO2. After incubation, 20 µl of the cell supernatant from the assay plate was transferred to a new 96-well white plate and 50 µl of QUANTI-Luc substrate was added. The plate was incubated, with shaking, at room temperature for 5 minutes then the luminescence was read on an EnVision 2104 equipment with an integration time of 0.1 s.
Human interferon-β induction assay [0207] THP1-Blue ISG cells are used to measure IFN-β secretion in the culture supernatant after activation of the STING pathway.
Petition 870190060374, of 06/28/2019, p. 117/142
114/122
To perform the assay, anti-IFN-β capture antibodies were coated on 96-well MultiArray plates (Mesoscale Discovery). After a one-hour incubation, the plates were washed and 50 µl of supernatant from the human STING cell reporter assay plates or IFN-β standards were mixed with 20 µl of Sulfotag-conjugated detection antibody on the coated plates. The plates were incubated, shaken for two hours, washed and the reading buffer was applied. Electrochemiluminescence was measured on the Sectorlmager equipment.
Evaluation of the STING cell signaling pathway [0208] The agonism of the STING pathway was measured in THP1 BLUE ISG cells as by the western blot of phospho-STING (S366), fosfoTBK1 (S172) AND phospho-IRF3 (S396). Briefly, 5 million cells in 90 µl of nucleofection buffer were mixed with 10 µl of test compounds. These mixtures were electroporated using the V001 program in an Amaxa Nucleofector equipment (Lonza). The cells were transferred to 12-well plates with fresh medium and allowed to recover for 5 hours at 37 ° C, 5% CO2. The cells were then washed in cold HBSS and lysed in RIPA buffer. The samples were normalized by total protein and diluted in ProteinSimple sample buffer or in LDS loading buffer. The samples were heat denatured at 95 Q C for 5 minutes, then PeggySue (ProteinSimple) was used to measure total and phospho-Sting Sting 0 IRF3 while NuPAGE (Invitrogen) was used to measure system TBK1. Data were analyzed using Compass or Licor Odyssey software, respectively.
In vivo STING activity [0209] For all studies, female Balb / c mice were obtained from Charles River Labs (Wilmington, MA, USA) and used when they were 6 to 8 weeks old and weighed approximately
Petition 870190060374, of 06/28/2019, p. 118/142
115/122
g. All animals were allowed to acclimate and recover from any transport-related stress for a minimum of 5 days before experimental use. Chlorinated water by reverse osmosis and irradiated food (Laboratory Autoclavable Rodent Diet 5010, Lab Diet) were provided ad libitum (at will), and the animals were kept on a 12-hour light and dark cycle. The cages and beds were autoclaved before use and changed weekly. All experiments were performed according to The Guide for the Care and Use of Laboratory Animals and as approved by the Institutional Animal Care and Use Committee of Janssen R & D, Spring House, PA, USA. Each experimental group contained 8 mice. The in vivo efficacy in a mouse CT26 tumor model was determined by implanting 500,000 CT26 colon carcinoma tumor cells subcutaneously into Balb / c mice and allowed the tumors to settle from 100 to 300 mm 3 . The compounds were injected intratumorally and formulated in phosphate buffered saline in a volume of 0.1 ml per injection. The mice were administered 0.05 mg every three days for a total of three doses. Efficacy was measured as the percentage of tumor growth inhibition (TGI) calculated by reducing the size of the treated tumor volume (T) in relation to the Control tumor volume (C) according to the following Formula: ((CT ) / (C)) * 100 when all control animals were still under study. Cures were defined as the number of animals without a measurable tumor detected in 10 times of tumor volume doubling (TVDT) after the last dose had been administered.
[0210] The resulting data are presented in Table 2.
Petition 870190060374, of 06/28/2019, p. 119/142
116/122
Table 2 ·
Compound No. hSTING SPAIC50(μΜ) * Human cell reporter EC50(μΜ) * Human SPRSTINGΚϋ (μΜ) ThermoFluorKD(μΜ) Human IFN-β(rating value) Activityin vivo(% TGI) In vivo activity (cures) 1 > 100 > 100 > 100 > 83.33 ND ND ND 2 <0.01 0.064 0.003 0.020 2205 87.1 2/8 3 > 88.25 > 10 ND > 66.67 ND ND ND 4 <0.01 0.09 0.002 0.001 2247 93.7 6/8 5 <0.01 0.16 0.008 0.084 2737 93.3 7/8 6 0.023 0.12 0.017 0.310 27 73.9 1/8 7 0.06 1.12 0.049 1,270 2260 94.3 5/8 8 0.035 0.11 0.042 0.510 2054 95.3 4/8 9 <0.01 0.64 0.000195 ND 2240 89.8 ND 10 0.012 0.54 0.00158 ND 1321 11 <0.01 0.49 3840 12 <0.01 0.22 3860 13 <0.01 0.59 2900 14 <0.01 0.45 4964 151.108172.88
ND-not done, human IFN-β classification value determined by the classification value determined by the cumulative IFN-β induction over the range of * IC50 and EC50 are averages of at least three values.
Biological Example 2
Primary human PBMC cytokine induction assay with STING [0211] Human STING pathway agonism is evaluated in primary human peripheral blood mononuclear cells (PBMC) derived from human whole blood. 1 pint (approximately 420 mL) of fresh donor blood (AllCells Inc., Alameda, CA) is placed on
Petition 870190060374, of 06/28/2019, p. 120/142
117/122 a Lymphocyte Separation Medium (1.077 to 1.080 g / ml, Corning, Manassas, VA, USA), and then centrifuged at 500 g for 20 minutes at room temperature without interruption. PBMC collected at the interface between the serum and the Lymphocyte Separation Medium are collected, washed and then counted. PBMCs are composed of subtypes of lymphocytes and monocytes, such as B cells, T cells, etc., and these subtypes have been characterized in the literature to express different levels of the STING protein. In response to STING agonists, such as 2'3'-cGAMP, these cells become activated and are induced to express a variety of antiviral and pro-inflammatory cytokines. In addition, through stimulation with STING agonists, these cells make positive regulation of activation markers. Levels of cytokine induction can be measured by a variety of methods including ELISA, Luminex and MSD. The levels of positive regulation of the activation marker can be measured by flow cytometry.
[0212] To perform the assay, 1,000,000 cells were dispensed in 96-well flat-bottom plates, treated with tissue culture of 225 pL / well. The test compounds were added in a volume of 25 µl at a concentration of 10x. Some compounds were solubilized in 100% DMSO and the final concentration of DMSO in the cultures receiving these compounds was 1%. The assay was incubated for 48 h at 37 ° C, 5% CO2. 200 µl of supernatants were harvested without disturbing the cells on the bottom of the plate, then they were frozen at -20 ° C until the Luminex measurement time. The tests with Luminex were performed with the use of G-CSF, IFNoc2, IFN ±, IL-1b, IL-6, IL-10, IL12 (p40), IL-12 (p70), TNFa together with MILLIPLEX MAP Human Cytokine / Chemokine Magnetic Bead Panel - Immunology Multiplex ΙΡΝβ1 assay and analyte kit with the MILLIPLEX MAP Human Cytokine / Chemokine Magnetic Bead Panel IV kit (EMD Millipore, Billerica, MA, USA), following the manufacturers' protocol. Cytokine induction was
Petition 870190060374, of 06/28/2019, p. 121/142
118/122 measured using a Luminex FlexMAP 3D® instrument (Luminex Corporation, Radnor, PA, USA). The analysis of collected Luminex data was performed using MILLIPLEX Analyst software (EMD Millipore). Suppression of HBV virus in PHH cells using conditioned media from primary human STEM-activated PBMCs [0213] Primary human hepatocytes can be infected with hepatitis B virus and during an established infection, will produce viral proteins such as HBsAg and HBeAg that can be detected by ELISA. Therapeutic treatment with compounds such as entecavir may suppress the reproduction of HBV, which can be measured by decreasing the production of viral protein. (No. of cells) 4x10 5 cells / well of primary human hepatocytes (BioReclamation, Westbury, NY, USA) were dispensed in 500 pL / well of 24-well plates treated with flat-bottom tissue culture. 24 h later, the cells were infected with 30 to 75 moi of HBV. The next day, PHH cells were washed 3x and fresh maintenance medium was added to the cells. Consequently, PBMCs were isolated as described above. To stimulate PBMC, 10,000,000 cells were dispensed into 24-well flat-bottom plates, treated with 400 pL / well tissue culture. The test compounds were added in a volume of 100 pL, then the cultures were incubated for 48 hours at 37 ° C, 5% CO2. Supernatants were collected. The cells were measured for activation of positive marker regulation with the use of flow cytometry. Briefly, the cells were stained with fluorescently labeled antibodies directed to CD56, CD19, CD3, CD8a, CD14, CD69, CD54, CD161, CD4 and CD80. The samples were analyzed on an Attune NxT flow cytometer (Thermo Fisher, Carlsbad, CA, USA) [0214] From the stimulated PBMC cultures, a portion of the supernatant was reserved for detection of cytokines by Luminex,
Petition 870190060374, of 06/28/2019, p. 122/142
119/122 as previously described. The rest of the supernatant was divided in half, and an aliquot was stored at 4 ° C for use on day 8 of the assay. The other aliquot of the supernatant was diluted 1: 1 with PHH 2X medium, then it was added to the PHH infected cells on day 4. After 96 hours, the spent medium was changed and the supernatant was added in a 1: 1 dilution with PHH 2X medium. At this point, an interim HBsAg measurement was performed using an HBsAg ELISA kit (Wantai Bio-pharm, Beijing, China). After 96 hours, the medium was collected and HBsAg was measured.
[0215] Table 3: Measurement (number of times) of cytokine induction in PBMC cultures stimulated with CDN compounds. The measurement (number of times) of the induction is calculated by measuring the cytokine concentrations induced after 48 hours in approximately 20 μΜ of compound and then dividing it by the baseline levels of cytokine production from cells incubated with PBS. The data are the average of several donors over three experiments, nt = not tested. Table 3 ·
Compound No. IL-6 IL-10 IFN-y IL-1b IFN-a TNFa IL-12p40 IL-12p70 G-CSF IFN-β 1 1.1 2.9 401.3 0.6 1.3 26.1 5.2 0.0 0.1 0.0 2 0.3 1.8 133.2 0.1 1.4 5.1 1.4 nt 0.0 nt 2 7.1 42.7 19.1 11.9 6.3 11.5 1.4 25.4 0.8 13.6 3 0.6 2.0 370.4 0.2 2.6 10.9 0.5 nt 0.0 nt 3 4.8 39.8 0.7 3.5 0.2 0.9 4.0 1.5 3.6 0.2 4 0.0 0.4 0.1 0.0 0.1 0.1 0.7 nt 0.0 nt 5 1.4 2.9 605.1 0.9 1.4 50.4 2.1 nt 0.1 nt 7 1.0 6.4 502.1 0.8 38.0 29.5 0.5 420.5 0.0 62.2 8 0.6 1.2 133.7 0.3 6.5 12.8 0.5 nt 0.0 nt 2'3'-cGAMP 2.5 5.9 17.4 1.4 6.8 5.6 1.0 5.5 0.4 16.0 PBS 1.0 1.0 1.0 1.0 1.0 1.0 1.0 1.0 1.0 1.0 DMSO 0.0 1.2 0.2 0.0 0.1 0.1 0.8 nt 0.0 nt
Petition 870190060374, of 06/28/2019, p. 123/142
120/122 [0216] Table 4: Measurement (number of times) of cytokine induction in PBMC cultures stimulated with higher concentrations of CDN compounds. The measurement (number of times) of the induction is calculated by measuring the cytokine concentrations induced after 48 hours at the indicated concentration of the compound, then dividing it by the baseline levels of cytokine production of cells incubated with PBS. The data are the average of several donors over three experiments, nt = not tested.
Table 4.
CompoundNo. Concentrate ofTop(pm) IL-6 IL-10 IFNy ΙΙ_-1β IFNa2 TNFa IL12 p40 IL12 p70 G-CSF IFNP1 1 111.1 0.7 0.4 2.0 1.7 0.9 3.4 0.7 35.2 1.1 nt 2 40 2523.6 61.0 3225.5 544.8 27.0 643.4 9.6 252.7 227.0 18.2 3 40 491.4 21.2 4.0 102.8 1.4 7.2 3.1 3.7 7.4 0.4 4 111.1 0.1 0.0 1.0 0.1 0.3 0.6 0.0 7.0 0.0 nt 5 111.1 0.2 0.0 3.1 2.2 0.5 4.4 0.1 34.5 0.3 nt 7 111.1 321.9 4.1 2088.8 113.2 1033.1 244.6 0.6 27.7 2.6 14.1 8 111.1 0.4 0.0 2.1 0.7 0.6 1.4 0.0 39.4 0.1 nt 9 40 5084.7 121.4 4776.7 5072.5 106.4 1003.1 26.6 640.0 775.6 22.1 10 40 2791.9 37.7 3104.9 342.6 41.1 555.4 25.5 274.0 58.4 20.7 14 40 2209.3 24.1 4760.1 288.8 44.7 811.7 22.9 574.2 295.5 18.7 13 40 2536.1 50.0 6065.9 445.5 39.5 881.8 32.0 686.4 246.2 16.6 2'3'-cGAMP 40 454.0 12.1 1919.1 251.2 27.8 117.1 1.8 17.1 14.1 13.5 DMSO0.5 0.3 0.4 0.6 0.5 0.4 0.5 0.6 1.2 nt
Petition 870190060374, of 06/28/2019, p. 124/142
121/122 [0217] Table 5. Conditioned medium from CDN-stimulated PBMCs can suppress the viral load of HBV-infected PHH cells. PBMCs were stimulated with the indicated CDN at 20, 4, 0.8 μΜ for 48 hours. The supernatants were mixed with fresh medium in a 1: 1 ratio, and then added to HBV-infected PHH cells. HBsAg production was measured 8 days later. The data are an average of two independent donors.
Table 5 ·
Compound No. EC50 (μΜ) 2 8.24E-04 3 88119.3 7 8.51 E-05
[0218] Table 6. CDN activates PBMC. PBMCs were stimulated with 20 μΜ of CDN for 48 hours. The cells were evaluated by flow cytometry for positive regulation of CD54 in monocytes. The measurement increase (fold) in the mean fluorescence intensity was calculated in relation to the levels in the resting cells. The data are an average of two independent donors.
Table 6 ·
Compound No. MFI 2 5.0 3 2.0 7 5.1 2'3'-cGAMP 4.5 PBS 1.0
Petition 870190060374, of 06/28/2019, p. 125/142
122/122 [0219] Although the aforementioned specification describes the principles of the present invention, with the examples provided for the purpose of illustration, it will be understood that the practice of the invention covers all the usual variations, adaptations and / or modifications, in accordance with with the scope of the following claims and their equivalents.
权利要求:
Claims (18)
[1]
1. A compound characterized by formula (I) being the

O
Formula (I) where:
Ria is hydroxy or fluoro and Ric is hydrogen; or, Ria is -O- and Rio is CH 2 so that Ria and Ric are taken together with the atoms to which they are attached to form a 5-membered ring;
Rib is selected from the group consisting of hydroxy, thiol and BH 3 -;
Bi is selected from the group consisting of rings b1 and b2

b1 e

R 2 a is selected from the group consisting of hydroxy and methoxy;
R 2 b is selected from the group consisting of hydroxy, thiol and BH 3 -;
provided that the compound of Formula (I) is different from the salt of (1,, 6 í, 8 í, 9 í, 10 í, 15 í, 17 í, 18 í) -17- (2-Amino-6-oxo-6,9-di- hydro1 / -7-pu ri n-9-yl) -8- (6-ami no-9 / -7-pu ri n-9-i I) -9-f I u oro-3,12,18 -Tri-hydroxyPetition 870190060374, of 06/28/2019, p. 127/142
[2]
11/11
2,4,7,11,13,16-hexaoxa-3À 5 , 12λ 5 diphosphatriciclo [13,2,1,0 6 , 1 °] octadecane-3,12-dione, bis-ammonium;
or an enantiomer, diastereomer or pharmaceutically acceptable salt form thereof.
2. Compound according to claim 1, characterized by the fact that Bi is b2

[3]
3. Compound characterized by being of formula (I)

Formula (I) selected from the group consisting of the

NH 2 2
Petition 870190060374, of 06/28/2019, p. 128/142
3/11



ΝΗ 2 6
Petition 870190060374, of 06/28/2019, p. 129/142
[4]
4/11



Petition 870190060374, of 06/28/2019, p. 130/142
[5]
5/11



Petition 870190060374, of 06/28/2019, p. 131/142
[6]
6/11

Petition 870190060374, of 06/28/2019, p. 132/142
[7]
7/11

Petition 870190060374, of 06/28/2019, p. 133/142
[8]
11/11



Petition 870190060374, of 06/28/2019, p. 134/142
[9]
9/11


or a pharmaceutically acceptable salt form thereof.
Pharmaceutical composition, characterized in that it comprises a compound as defined in any one of claims 1 to 3, and at least one of a pharmaceutically acceptable carrier, a pharmaceutically acceptable excipient and a pharmaceutically acceptable diluent.
5. Pharmaceutical composition, according to claim 4, characterized by the fact that the composition is a solid form for oral dosage.
6. Pharmaceutical composition according to claim 4, characterized by the fact that the composition is a syrup, an elixir or a suspension.
7. Pharmaceutical composition, characterized by the fact that it comprises a compound as defined in claim 3, and at least one among a pharmaceutically acceptable carrier, a
Petition 870190060374, of 06/28/2019, p. 135/142
[10]
10/11 pharmaceutically acceptable excipient, and a pharmaceutically acceptable diluent.
8. Method of treating a disease, syndrome or condition modulated by STING, characterized by the fact that it comprises administering to an individual who needs therapeutically effective amount of a compound as defined in claim 1.
9. Method of treatment of a disease, syndrome or condition, in which said disease, syndrome or condition is affected by the STING agonism, characterized by the fact that it comprises administering to an individual who needs it a therapeutically effective amount of the compound as defined in claim 1.
10. Method according to claim 9, characterized by the fact that said disease, syndrome or condition is cancer.
[11]
11. Method, according to claim 10, characterized by the fact that said cancer is selected from the group consisting of melanoma, colon cancer, breast cancer, prostate cancer, lung cancer and fibrosarcoma.
[12]
12. Method according to claim 9, characterized by the fact that said disease, syndrome, or condition is viral infection.
[13]
13. Method according to claim 11, characterized by the fact that the viral infection is hepatitis B.
[14]
14. Method of treatment of a disease, syndrome or condition selected from the group consisting of viral infection, melanoma, colon cancer, breast cancer, prostate cancer, lung cancer and fibrosarcoma, characterized by the fact that it comprises administering to a individual needing a therapeutically effective amount of the composition as defined in claim 4.
[15]
15. Use of a compound as defined in claim 1,
Petition 870190060374, of 06/28/2019, p. 136/142
11/11 characterized by the fact that it is for the preparation of a drug to treat a disease, syndrome or condition selected from the group consisting of viral infection, melanoma, colon cancer, breast cancer, prostate cancer, lung cancer and fibrosarcoma, in an individual who needs it.
[16]
16. Use of a compound as defined in claim 1, characterized by the fact that it is for use in a method to treat a disease, syndrome or condition selected from the group consisting of viral infection, melanoma, colon cancer, breast cancer, prostate cancer, lung cancer and fibrosarcoma, in an individual who needs it.
[17]
17. Method according to any one of claims 13 to 15, characterized by the fact that the viral infection is hepatitis B.
[18]
18. The method of claim 17, characterized in that it comprises administering to a subject who needs a therapeutically effective amount of a compound as defined in claim 3.
类似技术:
公开号 | 公开日 | 专利标题
AU2017365158B2|2022-02-17|Cyclic dinucleotides as sting agonists
US20190375782A1|2019-12-12|Cyclic dinucleotides as sting agonists
ES2891326T3|2022-01-27|Cyclic dinucleotides as STING agonists
BR112020010941A2|2020-11-17|cyclic dinucleotides as sting agonists
US20090274686A1|2009-11-05|Nucleoside phosphonate derivatives
JP6860585B2|2021-04-14|Incorporation by reference of acyclic antiviral priority application
BR112021000693A2|2021-04-13|CYCLIC DYNUCLEOTIDS AS STINGING AGONISTS
US11161864B2|2021-11-02|Sting agonists
BR112021010236A2|2021-08-24|Cyclobutyl nucleoside analogues as antivirals
同族专利:
公开号 | 公开日
JP2020500862A|2020-01-16|
MX2019006082A|2019-11-12|
CA3044693A1|2018-05-31|
AU2017365158B2|2022-02-17|
US20180162899A1|2018-06-14|
AU2017365158A1|2019-06-06|
CN110291096A|2019-09-27|
WO2018098203A1|2018-05-31|
KR20190085107A|2019-07-17|
TW201831192A|2018-09-01|
JOP20170188A1|2019-01-30|
EP3544989A1|2019-10-02|
UY37495A|2018-05-31|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

PT2934598T|2012-12-19|2018-07-24|Univ Texas|Pharmaceutical targeting of a mammalian cyclic di-nucleotide signaling pathway|
SG11201508273RA|2013-05-18|2015-12-30|Aduro Biotech Inc|Compositions and methods for activating "stimulator of interferon gene"-dependent signalling|
US9549944B2|2013-05-18|2017-01-24|Aduro Biotech, Inc.|Compositions and methods for inhibiting “stimulator of interferon gene”—dependent signalling|
GB201501462D0|2015-01-29|2015-03-18|Glaxosmithkline Ip Dev Ltd|Novel compounds|
JOP20200224A1|2015-08-13|2017-06-16|Merck Sharp & Dohme|Cyclic di-nucleotide compounds as sting agonists|
AU2016343993A1|2015-10-28|2018-05-10|Aduro Biotech, Inc.|Compositions and methods for activating "stimulator of interferon gene"-dependent signalling|
EP3322713B1|2015-12-03|2021-01-20|GlaxoSmithKline Intellectual Property Development Limited|Cyclic purine dinucleotides as modulators of sting|JP6980198B2|2016-03-18|2021-12-15|イミューン センサー リミテッド ライアビリティ カンパニー|Cyclic dinucleotide compound and usage|
US11098077B2|2016-07-05|2021-08-24|Chinook Therapeutics, Inc.|Locked nucleic acid cyclic dinucleotide compounds and uses thereof|
SG10201914029RA|2016-10-14|2020-03-30|Precision Biosciences Inc|Engineered meganucleases specific for recognition sequences in the hepatitis b virus genome|
JOP20170192A1|2016-12-01|2019-01-30|Takeda Pharmaceuticals Co|Cyclic dinucleotide|
US20200055883A1|2017-02-17|2020-02-20|Eisai R&D Management Co., Ltd.|Cyclic di-nucleotides derivative for the treatment of cancer|
WO2019084060A1|2017-10-24|2019-05-02|Silverback Therapeutics, Inc.|Conjugates and methods of use thereof for selective delivery of immune-modulatory agents|
US11203610B2|2017-12-20|2021-12-21|Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I.|2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein|
US10966999B2|2017-12-20|2021-04-06|Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I.|3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein|
KR20200124716A|2018-02-26|2020-11-03|길리애드 사이언시즈, 인코포레이티드|Substituted pyrrolysine compounds as inhibitors of HBV replication|
EP3774883A1|2018-04-05|2021-02-17|Gilead Sciences, Inc.|Antibodies and fragments thereof that bind hepatitis b virus protein x|
TW202005655A|2018-04-06|2020-02-01|捷克科學院有機化學與生物化學研究所|2'3'-cyclic dinucleotides|
TW202005654A|2018-04-06|2020-02-01|捷克科學院有機化學與生物化學研究所|2'2'-cyclic dinucleotides|
TW202010503A|2018-04-06|2020-03-16|捷克科學院有機化學與生物化學研究所|3’3’-cyclic dinucleotides|
TW201945388A|2018-04-12|2019-12-01|美商精密生物科學公司|Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis B virus genome|
TW202014193A|2018-05-03|2020-04-16|捷克科學院有機化學與生物化學研究所|2’3’-cyclic dinucleotides comprising carbocyclic nucleotide|
CN112512531A|2018-06-01|2021-03-16|卫材R&D管理有限公司|Methods for treatment of bladder cancer|
WO2020028097A1|2018-08-01|2020-02-06|Gilead Sciences, Inc.|Solid forms of -11--12--3,3-dimethyl-8-0x0-2,3,8,13b-tetrahydro-1h-pyrido[2,1-a]pyrrolo[1,2-c] phthalazine-7-c arboxylic acid|
JP2022500414A|2018-09-12|2022-01-04|シルバーバック セラピューティックス インコーポレイテッド|Methods and Compositions for Treating Diseases with Immunostimulatory Conjugates|
CN110938104A|2018-09-21|2020-03-31|上海迪诺医药科技有限公司|Cyclic dinucleotide analogs, pharmaceutical compositions and uses thereof|
US11110106B2|2018-10-29|2021-09-07|Venenum Biodesign, LLC|Sting agonists for treating bladder cancer and solid tumors|
AU2019371206A1|2018-10-29|2021-05-27|Venenum Biodesign, LLC|Novel sting agonists|
EP3873608A1|2018-10-31|2021-09-08|Gilead Sciences, Inc.|Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity|
CR20210215A|2018-10-31|2021-06-17|Gilead Sciences Inc|Substituted 6-azabenzimidazole compounds as hpk1 inhibitors|
CN111655712B|2018-12-29|2021-02-05|上海济煜医药科技有限公司|Compound as tumor immunity class and application thereof|
WO2020178770A1|2019-03-07|2020-09-10|Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I.|3'3'-cyclic dinucleotides and prodrugs thereof|
WO2020178769A1|2019-03-07|2020-09-10|Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I.|2'3'-cyclic dinucleotides and prodrugs thereof|
WO2020178768A1|2019-03-07|2020-09-10|Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I.|3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator|
WO2020214663A1|2019-04-17|2020-10-22|Gilead Sciences, Inc.|Solid forms of a toll-like receptor modulator|
CN109929894B|2019-04-17|2021-06-01|中国农业科学院兰州兽医研究所|Preparation and activity identification method of porcine second messenger molecule 2 '3' -cGAMP|
EP3956329A1|2019-04-17|2022-02-23|Gilead Sciences, Inc.|Solid forms of a toll-like receptor modulator|
WO2020227159A2|2019-05-03|2020-11-12|Flagship Pioneering Innovations V, Inc.|Methods of modulating immune activity|
EP3965891A1|2019-05-10|2022-03-16|Takeda Pharmaceutical Company Limited|Antibody drug conjugates|
WO2020237025A1|2019-05-23|2020-11-26|Gilead Sciences, Inc.|Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors|
CA3142513A1|2019-06-25|2020-12-30|Gilead Sciences, Inc.|Flt3l-fc fusion proteins and methods of use|
CA3145889A1|2019-07-05|2021-01-14|Tambo, Inc.|Trans-cyclooctene bioorthogonal agents and uses in cancer and immunotherapy|
WO2021034804A1|2019-08-19|2021-02-25|Gilead Sciences, Inc.|Pharmaceutical formulations of tenofovir alafenamide|
WO2021067181A1|2019-09-30|2021-04-08|Gilead Sciences, Inc.|Hbv vaccines and methods treating hbv|
WO2021067644A1|2019-10-01|2021-04-08|Silverback Therapeutics, Inc.|Combination therapy with immune stimulatory conjugates|
WO2021113765A1|2019-12-06|2021-06-10|Precision Biosciences, Inc.|Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome|
US11179473B2|2020-02-21|2021-11-23|Silverback Therapeutics, Inc.|Nectin-4 antibody conjugates and uses thereof|
WO2021188959A1|2020-03-20|2021-09-23|Gilead Sciences, Inc.|Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same|
WO2021228832A1|2020-05-11|2021-11-18|Erytech Pharma|Red cell extracellular vesiclescontaining cargoes and methods of use and production thereof|
WO2022006327A1|2020-07-01|2022-01-06|Silverback Therapeutics, Inc.|Anti-asgr1 antibody conjugates and uses thereof|
WO2022032191A1|2020-08-07|2022-02-10|Tambo, Inc.|Trans-cyclooctene bioorthogonal agents and uses in cancer and immunotherapy|
WO2022031894A1|2020-08-07|2022-02-10|Gilead Sciences, Inc.|Prodrugs of phosphonamide nucleotide analogues and their pharmaceutical use|
法律状态:
2021-06-15| B15K| Others concerning applications: alteration of classification|Free format text: AS CLASSIFICACOES ANTERIORES ERAM: C07H 21/02 , A61K 31/7084 , A61P 35/00 , A61P 31/12 Ipc: A61K 31/7084 (2006.01), C07H 21/02 (2006.01), A61P |
2021-10-05| B06W| Patent application suspended after preliminary examination (for patents with searches from other patent authorities) chapter 6.23 patent gazette]|
2021-10-05| B350| Update of information on the portal [chapter 15.35 patent gazette]|
优先权:
申请号 | 申请日 | 专利标题
US201662426350P| true| 2016-11-25|2016-11-25|
US201762502983P| true| 2017-05-08|2017-05-08|
US201762555232P| true| 2017-09-07|2017-09-07|
PCT/US2017/062901|WO2018098203A1|2016-11-25|2017-11-21|Cyclic dinucleotides as sting agonists|
[返回顶部]