专利摘要:
The present invention relates to compounds of formula (I), wherein the groups r1, r2 and r3 are as defined in claim 1, which have valuable pharmacological properties, particularly are sting modulators.
公开号:BR112019006512A2
申请号:R112019006512
申请日:2017-09-28
公开日:2019-10-01
发明作者:Fleck Martin;Carotta Sebastian;Oost Thorsten
申请人:Boehringer Ingelheim Int;
IPC主号:
专利说明:

CYCLIC DINUCLEOTIDE COMPOUNDS
FIELD OF THE INVENTION [001] The present invention relates to new cyclic dinucleotide compounds (CDNs) of formula I, including their pharmaceutically acceptable salts, which have the imidazopyridazinone non-purine nucleobase, a purine nucleobase and a 2 ', 5 non-canonical phosphorothioate and induce cytokine production. The present invention also relates to pharmaceutical compositions and combinations containing the compounds of the present invention, and their clinical use for the treatment of diseases associated or modulated by STING (interferon stimulating genes). In particular, the pharmaceutical compositions of the invention are suitable for the therapy of inflammation, allergic and autoimmune diseases, infectious diseases, cancer and as vaccine adjuvants.
BACKGROUND OF THE INVENTION [002] The function of the immune system is to protect the body from pathogens and malignant cells. However, viruses and cancer cells find ways to escape the immune system. Thus, the goal of immunotherapies is to initiate an antigen-specific immune response or reactivate a pre-existing response in certain cell types of the immune system against pathogenic invaders or cancer cells.
[003] The immune system consists of several specialized strains that can be grouped into two groups: the innate immune system and the adaptive immune system. In order for a successful immune reaction to occur, strains from both groups
Petition 870190091404, of 9/13/2019, p. 8/105
2/98 need to act in harmony. An important function of the innate immune system is to mount a rapid immune response against pathogens or malignant cells, which, unlike the adaptive system, is not antigen-specific and long-lasting. In addition to the direct death of pathogens or transformed cells, the innate immune system also activates and subsequently targets the adaptive immune system. Antigen-presenting cells, like dendritic cells, capture and present antigens in the form of a major peptide-histocompatibility complex (MHC) with T cells in lymphoid tissues. This antigen presentation, together with the secretion of certain cytokines, leads to the activation and differentiation of the antigen-specific CD4 and CD8 effector cells. The production of type I interferon (IFN) by antigen presenting cells, and other types of cells, is considered a key event in the activation of T cells, since the lack of type I IFN resulted in a reduced T cell dependent immune response. against viral infections or tumor cells (Zitvogel et al, Nature Reviews Immunology 15, 405 - 414, 2015). On the other hand, the presence of a type I IFN signature during cancer therapy is associated with an increase in the number of tumor infiltrating T cells and potentially favorable clinical results (Sistigu et al, Nature Medicine 20, 1301 - 1309, 2014 ).
[004] Recent studies in mice have shown that the efficient secretion of IFN type I in the tumor microenvironment and the induction of a T-cell-dependent immune response against cancer cells depends on the presence
Petition 870190091404, of 9/13/2019, p. 9/105
3/98 of the adapter protein interferon stimulating genes (STING, also known as Tmeml73, MPYS, MITA, ERIS) (Woo et al, Immunity 41, 5, 830 - 842, 2014; Corrales et al, Cell Reports 11, 1018 - 1030, 2015; Deng et al, Immunity 41, 5, 843 - 852, 2014). The importance of the presence of IFN type I was highlighted by the fact that the deletion of the STING resulted in a reduction in the levels of IFN type I in the tumor microenvironment and in a reduced antitumor effect in several models of tumor mice. On the other hand, specific STING activation resulted in an enhanced antigen-specific T cell immune response against cancer cells.
[005] STING belongs to the family of nucleic acid sensors and is the adapter for signaling cytosolic DNA. In its basal state, STING exists as a dimer with its N-terminal domain anchored in the ER and the C-terminal domain in the cytosol. Cyclic dinucleotides (CDNs), generated by the cyclic GMP-AMP synthase protein (cGAS) are the natural ligands of STING (Ablasser et al, Nature 498, 380 - 384, 2013). The binding of CDNs to STING induces conformational changes that allow the binding and activation of the kinase that binds to TANK (TBK1) and interferon regulatory factor 3 (IRF3) and the relocation of the ER to perinuclear endosomes (Liu et al, Science 347, Issue 6227, 2630-1 - 2630-14, 2015). The phosphorylation of the transcription factor IRF3 and NF-kB by TBK1 results in the expression of multiple cytokines, including IFN type I.
[006] Given the importance of type I IFN in various malignancies, including viral infections and cancer therapy, strategies that allow specific activation
Petition 870190091404, of 9/13/2019, p. 10/105
4/98 of STING are of therapeutic interest.
[007] WO 2014/189805 describes cyclic dinucleotide compounds that have two purine nucleobases and at least a 2 ', 5'-phosphodiester or non-canonical phosphorothioate moiety and induce STING-dependent cytokine production.
[008] WO 2015/185565 describes cyclic dinucleotide compounds that have two purine nucleobases, one or two cyclopentanes instead of ribose tetrahydrofuran rings and a non-canonical 2 ', 5'-phosphodiester moiety and modulate STING.
[009] WO 2016/120305 describes cyclic dinucleotide compounds that have two purine nucleobases, a ribose portion in which 2'-OH is replaced by a 2'-F portion and a 2 ', 5'-phosphodiester portion non-canonical, and modulate the STING.
[0010] US 2014/0329889, WO 2014/099824, WO 2015/017652, Cell 154, 748-762 (2013) and Molecular Cell 51, 226-235 (2013) describe the cyclic dinucleotide 2 ', 3' - cGAMP ([G (2 ', 5') pA (3 ', 5') p] cyclic) which has two portions of purine nucleobases, one portion being phosphodiester 3 ', 5' canonical and one portion 2 ', 5' non-canonical. Non-canonically bound 2'3'-cGAMP binds to human STING with greater affinity than canonically bound 3'3'-cGAMP or symmetrical bacterial c-di-GMP and induces the production of type I interferon.
[0011] WO 2014/093936 describes cyclic dinucleotide compounds that have two purine nucleobases and two portions of canonical 3 ', 5' phosphodiester or phosphorothioate and induce dependent cytokine production
Petition 870190091404, of 9/13/2019, p. 11/105
5/98 STING.
[0012] US 7,709,458 describes cyclic dinucleotide compounds that have two purine nucleobases and two portions of canonical 3 ', 5' phosphodiéser, and which can be used to inhibit cancer cell proliferation or to increase cell apoptosis cancerous, in particular, symmetrical bacterial CDN c-di-GMP.
[0013] US 7,592,326 describes immunostimulatory cyclic dinucleotide compounds that have two purine nucleobases and two canonical 3 ', 5' phosphodiester units, in particular, the symmetric bacterial CDN c-di-GMP.
[0014] WO 2016/096174 and WO 2016/145102 describe cyclic dinucleotide compounds that have two purine nucleobases and two portions of canonical 3 ', 5' phosphodiester or phosphorothioate and induce STING-dependent cytokine production.
[0015] Bioorg. Med. Chem. Lett. 18 (2008) 5631-5634 describes immunostimulatory mono- and bisphosphorothioate analogs of symmetrical bacterial cDNA-GMP. SUMMARY OF THE INVENTION [0016] In a first aspect, the invention provides cyclic dinucleotide compounds of formula I
Petition 870190091404, of 9/13/2019, p. 10/125
6/98 where
R 1 is selected of the group that It consists in H, F, -0- alkyl Ci -3 and OH, and R2 is H, or R 2 is - -CH 2 - and R 1 is -0-, forming together one bridge -CH 2 -0-
(blocked nucleic acid; LNA), and
R 3 is a purine nucleobase selected from the group consisting of purine, adenine, guanine, xanthine, hypoxanthine, linked through its N 9 nitrogen;
isoforms, tautomers, stereoisomers, metabolites, prodrugs, solvates, hydrates and their salts, particularly their physiologically acceptable salts with inorganic or organic bases, or combinations thereof.
[0017] In a further aspect, the invention provides new compounds of formula I, including their pharmaceutically acceptable salts, which induce the production of cytokines in a STING-dependent manner in vitro and / or in vivo, and have suitable pharmacological and pharmacokinetic properties for use in therapy, that is, for use as medicines.
[0018] In a further aspect, the invention provides new compounds of formula I, including their pharmaceutically acceptable salts, for use in the treatment of a disease or condition associated with or modulated by STING.
[0019] In a further aspect, the invention provides new compounds of formula I, or pharmaceutically acceptable salts thereof for the treatment of inflammation, allergic diseases or autoimmune diseases, for example, allergic rhinitis or asthma, for the treatment of diseases
Petition 870190091404, of 9/13/2019, p. 10/13
7/98 infectious or cancer, or for use as vaccine adjuvants.
[0020] In a further aspect, the invention provides a method of treating a disease or condition associated with or modulated by STING, in an individual comprising administering a therapeutically effective amount of a compound of formula I, or a pharmaceutically salt. acceptable to the individual.
[0021] In a further aspect, the invention provides a method of treating inflammation, allergic or autoimmune diseases, for example, allergic rhinitis or asthma, for the treatment of infectious diseases or cancer, in a patient needing it, comprising administering a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, to the patient.
[0022] In a further aspect, the invention provides pharmaceutical compositions comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and one or more of the pharmaceutically acceptable excipients.
[0023] In a further aspect, the invention provides the use of a compound of formula I, including its pharmaceutically acceptable salts, in the manufacture of a medicament for use in the treatment of a disease or condition in which STING modulation is beneficial or for use in the treatment of a disease or condition associated with or modulated by STING.
[0024] In a further aspect, the invention provides the use of a compound of formula I, or the pharmaceutically acceptable salts thereof, in the manufacture of
Petition 870190091404, of 9/13/2019, p. 10/145
8/98 a medicine for use in the treatment of inflammation, allergic diseases or autoimmune diseases, for example, allergic rhinitis or asthma, for the treatment of diseases
infectious or cancer, or to use i as adjuvants in vaccine.[0025] In one aspect Add there invention provides an combination what comprises a compound of formula I, or one
pharmaceutically acceptable salt thereof, and at least one therapeutically additional agent.
[0026] An additional object of the present invention is to provide a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one additional therapeutic agent and one or more of the pharmaceutically acceptable excipients.
[0027] In a further aspect, the invention provides a combination comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one additional therapeutically agent for use in therapy.
[0028] In a further aspect, the invention provides a combination comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one additional therapeutic agent for use in the treatment of a disease or condition in which the modulation of the STING is beneficial or for use in the treatment of a disease or condition associated with or modulated by STING.
[0029] In a further aspect, the invention provides a combination comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one additional therapeutic agent for use in the treatment of
Petition 870190091404, of 9/13/2019, p. 10/155
9/98 inflammation, allergic and autoimmune diseases, infectious diseases and cancer.
[0030] In a further aspect, the invention provides a method of treating a disease or condition in which STING modulation is beneficial, or a disease or condition associated with or modulated by STING, in a patient, which comprises administering to the patient a therapeutically effective amount of a combination comprising a
compound in formula I, or a salt pharmaceutically acceptable of same, and at least one agent therapeutic additional.[0031 ] In an additional aspect, The invention provides a
a method of treating inflammation, allergic and autoimmune diseases, infectious diseases and cancer in a patient, which comprises administering to the patient a therapeutically effective amount of a combination comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least an additional therapeutic agent.
[0032] In a further aspect, the invention provides a vaccine adjuvant comprising a compound of formula I, or a pharmaceutically acceptable salt thereof.
[0033] In a further aspect, the invention provides an immunogenic composition that comprises an antigen or antigen composition and a compound of formula I, or a pharmaceutically acceptable salt thereof.
[0034] In a further aspect, the invention provides an immunogenic composition comprising an antigen or antigen composition and a compound of formula I, or a pharmaceutically acceptable salt thereof, for use in the
Petition 870190091404, of 9/13/2019, p. 10/165
10/98 treatment or prevention of a disease.
[0035] In a further aspect, the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, for the manufacture of an immunogenic composition comprising an antigen or an antigen composition for the treatment or treatment. prevention of a disease.
[0036] In a further aspect, the invention provides a method of treating or preventing a disease, comprising administering to an individual human suffering from or susceptible to the disease, an immunogenic composition comprising an antigen or antigen composition and a compound of formula I, or a pharmaceutically acceptable salt thereof.
[0037] In a further aspect, the invention provides a vaccine composition that comprises an antigen or antigen composition and a compound of formula I, or a pharmaceutically acceptable salt thereof, for use in the treatment or prevention of a disease.
[0038] In a further aspect, the invention provides the use of a compound of formula I, or a pharmaceutically acceptable salt thereof, for the manufacture of a vaccine composition that comprises an antigen or an antigen composition for the treatment or the prevention of a disease.
[0039] In a further aspect, the invention provides a method of treating or preventing a disease that comprises administering to a human individual suffering from or susceptible to the disease, a vaccine composition comprising an antigen or antigen composition it is a
Petition 870190091404, of 9/13/2019, p. 10/175
11/98 compound of formula I, or a pharmaceutically acceptable salt thereof.
[0040] Other objects of the present invention will become apparent to the person skilled in the art from the description above and below, and from the examples.
[0041] The compounds of the present invention exhibit several advantages, such as a favorable binding affinity to human STING, favorable cellular activity, that is, in cells containing different human STING alleles, favorable stability in cellular assays and favorable pharmacokinetic properties (PK) .
DETAILED DESCRIPTION [0042] Unless otherwise stated, R 1 , R 2 and R 3 are defined as above and below. Some preferred meanings of groups and substituents of the compounds according to the invention will be provided below. Any and each of these definitions can be combined with each other.
R 1 and R 2 :
[0043] In a first modality, R 1 and R 2 are defined as mentioned above.
[0044] In another modality, R 1 and R 2 are both H. [0045] In still another modality, R 1 is F and R 2 is H. [0046] In still another mode R 1 is - -OH and R 2 is H. [0047] In still another modality, R 1 is -OCH3 and R 2 is H. [0048] In still another modality, R 1 is -0- and R 2 is -CH 2 forming together only one bridge -O-CH2-.
R 3 :
[0049] In a first modality, R 3 is defined as mentioned above.
[0050] In another modality, R 3 is purine, linked through
Petition 870190091404, of 9/13/2019, p. 10/185
12/98 of your nitrogen Ν 9 .
[0051]
In another modality
R 3 adenine bound through its nitrogen
N 9 .
[0052]
In yet another modality
R 3 guanma bound through its nitrogen
N 9 .
[0053]
In yet another modality
R 3 xanthine bound through its nitrogen
N 9 .
[0054]
In yet another modality
R 3 is hypoxanthine bound via its nitrogen N 9 · [0055] The following table represents other specified modalities 1-1 to 1-16 of the compounds of formula I:
Modality R R R is a purine nucleobase bound through its N 9 nitrogen, selected from the group consisting of 1-1 H H purine, adenine, guanine,xanthine, hypoxanthine 1-2 F H adenine 1-3 F H purine 1-4 F H guanine 1-5 F H xanthine 1-6 F H hypoxanthine 1-7 OH H adenine 1-8 OH H purine 1-9 OH H guanine 1-10 OH H xanthine 1-11 OH H hypoxanthine 1-12 R 1 is -0- and R 2 is -ch 2 forming a bridge -0-CH 2 together adenine
Petition 870190091404, of 9/13/2019, p. 10/195
13/98
1-13 R 1 is -0- and R 2 is -ch 2 forming a bridge -0-CH 2 together purine 1-14 R 1 is -0- and R 2 is -ch 2 forming a bridge -0-CH 2 together guanine 1-15 R 1 is -0- and R 2 is -ch 2 forming a bridge -0-CH 2 together xanthine 1-16 R 1 is -0- and R 2 is -ch 2 forming a bridge -0-CH 2 together hypoxanthine
[0056] A preferred substructure of the compounds according to the invention is shown in formula Ia,
Ia, where R 1 and R 2 , as well as their modalities, are defined as described above.
[0057] A preferred substructure of the compounds of
Petition 870190091404, of 9/13/2019, p. 10/20
14/98 according to the invention is shown in formula Ib,
where R 1 and R 2 , as well as their modalities, are defined as described above.
[0058] The following compounds according to the invention are particularly preferred:
Petition 870190091404, of 9/13/2019, p. 10/21
15/98
its tautomers and stereoisomers, its salts, particularly its physiologically acceptable salts with inorganic or organic bases, and its solvates or hydrates.
[0059] The compounds of the present invention have chiral phosphorus atoms with Rp or Sp configuration. All stereoisomers of the compounds of general formula I, Ia, Ib, la.l, Ia.2, Ia.3 and Ib.l, in the substantially pure form or as the necessary mixtures, are covered by the invention of the subject. Compounds of general formula I, Ia, Ib, la.l, Ia.2, Ia.3 and Ib.l, such as stereoisomers (Rp, Rp), (Rp, Sp), (Sp, Rp) or (Sp , Sp) substantially pure, are preferable, particularly the stereoisomer (Rp, Rp) substantially pure, that is, with both atoms
Petition 870190091404, of 9/13/2019, p. 10/22
16/98 of phosphorus with the Rp configuration.
[0060] The compounds according to the invention and their intermediates can be obtained using synthetic methods which are known to the person skilled in the art and are described in the organic synthesis literature. Preferably, the compounds are obtained analogously to the preparation methods explained in more detail below, and in particular as described in the experimental section. In some cases, the sequence adopted in the execution of the reaction schemes may vary. Variants of these reactions that are known to the person skilled in the art, but which are not described in detail here, can also be used. The general procedures for preparing the compounds according to the invention will become apparent to the person skilled in the art when studying the following schemes. The starting compounds are commercially available or can be prepared by methods that are described in the literature or in the present invention, or that can be prepared in an analogous or similar manner. Before the reaction is carried out, any corresponding functional groups in the compounds can be protected using conventional protecting groups. These protecting groups can be cleaved again at an appropriate stage within the reaction sequence using methods familiar to the person skilled in the art.
[0061] The cyclic dinucleotides disclosed in the present invention can be prepared as described in detail below, or by other methods known to those skilled in the art. It will be understood by a person normally skilled in the art that these schemes
Petition 870190091404, of 9/13/2019, p. 10/23
17/98 are in no way limiting, and that variations in details can be made without departing from the spirit of the present invention.
[0062] Cyclic dinucleotide compounds can be obtained by the methods described in Chem. Rev. 113, 7354-7401 (2013), Org. Lett., 12, 3269-3271 (2010), Tetrahedron 49, 1115-1132 (1993), WO 2014/189805, WO 2016/096174, WO 2015/185565, WO 2016/145102 or WO 2016/120305 and in the references cited in these documents.
[0063] The term protecting group, as used in the present invention, and unless otherwise defined, refers to a chemical functional group that is attached to an oxygen, nitrogen or phosphorus atom to prevent an additional reaction of that atom, or for other purposes. A wide variety of protection groups are known to those versed in the technique of organic synthesis, and are described, for example, in Protective Groups in Organic Synthesis by T.W. Greene and P.G.M. Wuts, third edition, 1999.
[0064] The compounds of formula (I) and their salts can be prepared by the methodology described below, further constituting aspects of this invention.
[0065] Persons skilled in the art will recognize that the phosphorothioate moieties in formula (I) may, individually, exist in the R (Rp) or S (Sp) configuration. The methodology described below can give rise to up to four diastereoisomers in relation to phosphorus atoms that can be separated by chromatographic methods known to the person skilled in the art, for example, by high pressure liquid chromatography with
Petition 870190091404, of 9/13/2019, p. 10/24
18/98 solvent systems and suitable columns at different stages of synthesis. In some cases, for example, when a sulfurization step proceeds in a diastereoselective way, the methodology described below can preferably only two diastereoisomers that can be separated by chromatographic methods known to the person skilled in the art, at different stages of the synthesis.
It is understood that substituents not explicitly specified in the preparation methods below encompass the definitions mentioned above in
Summary of the invention.
[0067] A compound of formula
where R4 optionally, RI are
OH can be prepared by deprotection of a compound of formula
where R 4 · 1 and, optionally, R 1 · 1 is oxygen with a suitable protecting group, such as tert-butyldimethylsilyl (TBS). For example, R 1 · 1 is H, F, O-alkyl or OTBS or, together with R 2
Petition 870190091404, of 9/13/2019, p. 10/25
19/98 forms a -CH2-O- bridge, and R 4 · 4 is OTBS. For example, a compound of formula (II) is dissolved in a suitable solvent, for example, in pyridine, treated with a mixture of triethylamine and triethylamine trihydrofluoride and stirred at a suitable temperature, for example, from 20 to 60 ° C, for an appropriate period of time, for example, 1 to 6 hours.
[0068] A compound of formula (II) can be prepared by deprotecting a compound of formula (III),
where R 3 · 1 denotes NH containing a suitable protecting group, such as benzoyl, and R 3 · 2 denotes H (protected adenine) or
R 3 · 1 denotes OH and R 3 · 2 · denotes NH containing a suitable protecting group, such as isobutyryl (protected guanine) or
R 3 · 1 denotes OH and R 3 · 2 · denotes H (hypoxanthine) or both R 3 · 1 and R 3 · 2 denote H (purine).
[0069] For example, a compound of formula (III) is dissolved in a suitable mixture, for example, methylamine or aqueous ammonia solution in methanol or ethanol, and stirred at a suitable temperature, for example, from 20 to 60 ° C , for an appropriate period of time, for example, 1 to hours.
[0070] A compound of formula (III) can be prepared by cyclization and subsequent sulfurization of a compound of
Petition 870190091404, of 9/13/2019, p. 10/26
20/98 formula (IV), where R 2 , R 3 · 1 , R 3 · 2 ·
R 1 1 and R 4 · 1 are defined as previously mentioned:
[0071] (IV)
For example, a compound of formula (IV) is dissolved in a suitable solvent, for example, pyridine and treated with a suitable coupling reagent, for example
2-oxide
2-chloro-5,5-dimethyl-1,3,2dioxaphosphorinane (DMOCP) or pivaloyl chloride or adamantoyl chloride, and stirred at a suitable temperature, for example, 20 ° C, for an appropriate period of time for example , from 0.1 to 2 hours. The cyclization reaction is suppressed by treatment with a suitable sulfurizing reagent, for example, 3H-1,2-benzodithiol-3-one or elemental sulfur, and stirred at a suitable temperature, for example, 20 ° C, for one adequate time, for example, from 0.1 to 2 hours.
[0072] A compound of formula (IV) can be prepared by coupling a compound of formula (V) with a compound of formula (VI), where R 2 , R 3 · 1 , R 3 · 2 ·, R 1 · 1 and R 4 · 1 are defined as mentioned above:
Petition 870190091404, of 9/13/2019, p. 10/275
21/98
[0073] For example, a compound of formula (VI) is dissolved in a suitable solvent, for example, acetonitrile, and is treated with a solution of a compound of formula (V) dissolved in a suitable solvent, for example, acetonitrile, optionally in the presence of a suitable coupling reagent, for example, tetrazole, Activator 42 ® (activator solution, containing 5- (3,5bis (trifluoromethyl) phenyl) -IH-tetrazole in acetonitrile), pyridinium dichloroacetate or pyridinium trifluoroacetate ( or mixtures of coupling reagents), and stirred at a suitable temperature, for example 20 2 C for a suitable period of time, for example 0.1 to 2 hours. The coupling reaction is suppressed by treatment with a suitable sulfurizing reagent, for example, 3 ((N, N-dimethylaminomethylidene) amino) -3H-1,2,4-dithiazole-3thione (DDTT) or phenylacetyl disulfide (PADS ) or 1,1H-1,2-benzodithiol-3-one 1,1 dioxide (Beaucage reagent) and stirred at a suitable temperature, for example, 20 ° C, for a suitable period of time, such as 0 , 1 to 2 hours. After the solvent has evaporated, the residue is dissolved in a suitable solvent, such as a mixture of dichloromethane and water, and treated with a suitable reagent, such as dichloroacetic acid, and stirred
Petition 870190091404, of 9/13/2019, p. 10/28
22/98 at a suitable temperature, for example, 20 ° C, for a suitable period of time, such as 0.1 to 2 hours. A solution containing the product (IV) is obtained by adding a suitable solvent, for example, pyridine, and by concentration, by evaporation.
[0074] A compound of formula (V) can be prepared by reacting a compound of formula (VII), where R 2 , R 3 · 1 , R 3 · 2 ·, Ri i θ R 4 · 1 are defined as mentioned before:
[0075] For example, a compound of formula (VII) is dissolved in a suitable mixture, for example, of water containing acetonitrile, and treated with pyridinium trifluoracetate, and stirred at a suitable temperature, for example, 20 ° C, for an appropriate period of time, such as 1 to 30 minutes. The tert-butylamine is added and the mixture is stirred at a suitable temperature, for example, 20 ° C, for a suitable period of time, such as 0.1 to 1 hour. The product is isolated by evaporation of the solvent, then it is dissolved in a suitable solvent, for example, in water containing dichloromethane, and treated with dichloroacetic acid and stirred at a suitable temperature, for example, 20 ° C, for a period adequate time, for example, from 0.1 to 1 hour. A concentrated solution of the product (V) in
Petition 870190091404, of 9/13/2019, p. 10/29
23/98 acetonitrile is obtained, for example, by the addition of pyridine followed by azeotropy of the mixture with acetonitrile.
[0076] A compound of formula (VI) can be prepared by reacting a compound of formula (VIII), where R 4 · 1 is defined as previously mentioned:
[0077] For example, after azeotroping with a suitable solvent, for example, acetonitrile, a compound of formula (VIII) is dissolved in a suitable solvent, for example, in dichloromethane, and reacted with phosphorylation reagent, for example, with 2 -cynoethyl Ν, Ν, Ν ', Ν'tetraisopropylphosphorodiamidite, in the presence of an activator, for example, IH-tetrazole, and stirred at an appropriate temperature, such as 20 2 C, for an appropriate period of time, for example, from 1 to 48 hours.
[0078] A compound of formula (VIII) can be prepared by reacting a compound of formula (IX):
[0079] For example, a compound of formula (IX) is dissolved in a suitable solvent, for example, pyridine, and reacted with a suitable silylation reagent, for
Petition 870190091404, of 9/13/2019, p. 10/30
24/98 example, with tert-butyldimethylsilyl chloride, in the presence of a suitable base, for example, imidazole, and stirred at a suitable temperature, for example, 20 ° C, for an appropriate period of time, for example, 1 to 48 hours. The 2 'and 3'-silylated regioisomers are isolated after purification in aqueous medium and can be separated, for example, by chromatography on silica gel with suitable solvent systems.
[0080] A compound of formula (IX) can be prepared by reacting a compound of formula (X):
[0081] For example, a compound of formula (X) is dissolved in a suitable solvent, for example, in pyridine, and reacted with 4,4'-dimethoxytrityl chloride, and stirred at a suitable temperature, for example, 20 ° C for a suitable period of time, such as 1 to 48 hours.
[0082] The compounds of general formula I, or their synthetic intermediates, can be separated into their diastereoisomers, as mentioned below. Diastereoisomeric mixtures of the compounds of general formula I can be separated into their diastereoisomers taking advantage of their different physico-chemical properties, using methods known per se, for example, by chromatography and / or by fractional crystallization.
[0083] As mentioned above, the compounds of formula I can be converted to salts, particularly for pharmaceutical use, in pharmaceutically acceptable salts.
Petition 870190091404, of 9/13/2019, p. 10/31
[0084] The compounds according to the invention can also be advantageously obtained using the methods described in the following examples, which can also be combined for this purpose with methods known to the person skilled in the literature.
Terms and Definitions [0085] The terms not specifically defined here must be given the meanings that would be given to them by a person skilled in the art in the light of revelation and context. However, as used in the specification, unless otherwise specified, the following terms have the indicated meaning and
following conventions are observed. [0086] The terms compound (s) in according to this invention, compound (s) of formula (I) , compound (s) of
invention and the like denote the compounds of formula (I) according to the present invention, including their tautomers, stereoisomers and mixtures thereof, and their salts, in particular, their pharmaceutically acceptable salts, and the solvates and hydrates of those compounds, including the solvates and hydrates of these tautomers, stereoisomers and salts thereof.
[0087] The terms treatment and treat cover both preventive, that is, prophylactic, and therapeutic treatment, that is, curative and / or palliative treatment. Thus, the terms treatment and treating include therapeutic treatment of patients who have already developed said condition, in particular, in a manifest way. Therapeutic treatment can be symptomatic treatment to alleviate the symptoms of the specific indication or the
Petition 870190091404, of 9/13/2019, p. 10/32
26/98 causal treatment, in order to revert or partially revert the conditions of the indication or to interrupt or slow the progression of the disease. Thus, the compositions and methods of the present invention can be used, for example, as therapeutic treatment over a period of time, as well as for chronic therapy. In addition, the terms treatment and treating include prophylactic treatment, that is, treatment of patients at risk of developing a condition mentioned above, thereby reducing said risk.
[0088] When this invention relates to patients requiring treatment, it mainly relates to treatment in mammals, and in particular in humans.
[0089] The term therapeutically effective amount means an amount of a compound of the present invention that (i) treats or prevents the specific disease or condition (o), (ii) alleviates, improves or eliminates one or more symptoms of the specific disease or condition (o) or (iii) prevents or delays the appearance of one or more symptoms of the disease or condition described in this document.
[0090] The terms modulated, modulating, or modulating, as used in the present invention, refer to the activation of the STING pathway with one or more compounds of the present invention, in this case, representing the STING agonists.
[0091] The terms mediated or mediating or mediates, as used in the present invention, unless otherwise indicated, refer to (i) treatment, including the prevention of the particular disease or condition, (ii) mitigation, improvement or elimination of one or more
Petition 870190091404, of 9/13/2019, p. 10/33
27/98 symptoms of the particular disease or condition, or (iii) preventing or delaying the onset of one or more symptoms of the particular disease or condition described herein.
[0092] In case a compound of the present invention is represented in the form of a chemical name and as a formula, in case of any discrepancy, the formula must prevail.
[0093] An asterisk can be used in subformulas to indicate the bond that is linked to the central molecule, as defined.
[0094] Unless specifically stated, throughout the specification and the appended claims, a given formula or chemical name must cover tautomers and all optical and geometric isomers, as well as stereoisomers (eg, enantiomers, diastereoisomers, isomers E / Z, etc.) and their racemates, as well as mixtures in different proportions of the separate enantiomers, mixtures of diastereoisomers or mixtures of any of the above forms, where such isomers and enantiomers exist, as well as salts, including their pharmaceutically acceptable salts and their solvates such as, for example, hydrates that include solvates of the free compounds or solvates of a salt of the compound.
[0095] The term substantially pure, as used in the present invention in relation to the compounds of formula I, refers to a diastereoisomer (Rp, Rp), (Rp, Sp), (Sp, Rp) or (Sp, Sp ) which is at least 75% pure in relation to the other possible diastereoisomers in relation to phosphorus atoms. In preferential terms, the
Petition 870190091404, of 9/13/2019, p. 10/34
The substantially pure compounds of formula I are at least 85% pure, at least 90% pure, at least 95% pure, at least 97% pure and at least 99% pure.
[0096] The phrase pharmaceutically acceptable is used in the present invention to refer to compounds, materials, compositions and / or dosage forms that are, within the scope of good medical judgment, suitable for use in contact with the tissues of humans and animals without excessive toxicity, irritation, allergic response, or other problem or complication, in proportion to a reasonable risk / benefit ratio.
[0097] As used in the present invention, pharmaceutically acceptable salts refer to derivatives of the disclosed compounds in which the parent compound is modified by making salts thereof with bases. The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains an acidic portion, by conventional chemical methods. In general, these salts can be prepared by reacting the free acid forms of these compounds with a sufficient amount of the appropriate base in water or in an organic solvent, such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile, or a mixture of the themselves. Alternatively, salts can be prepared by ion exchange, for example, by treating the aqueous solutions of the compounds of the invention (free acid or salt forms) with a cation exchanger.
Pharmacological activity [0098] The compounds according to the present invention exhibit favorable binding affinity for human STING. THE
Petition 870190091404, of 9/13/2019, p. 10/35
29/98 binding affinity can be, for example, determined by competition binding testing based on proximity scintillation testing (SPA), as described in Nat. Chem. Biol. 10, 1043-1048 (2014). Alternatively, the binding affinity can be, for example, determined by isothermal titration calorimetry (ITC), as described in Molecular Cell 51, 226-235 (2013). Alternatively, the binding affinity can be, for example, determined by surface Plasmon resonance (SPR), as described in WO 2016/145102. Alternatively, the binding affinity can be determined by Differential Scanning Fluorimetry (DSF), for example, as described in WO 2016/145102.
[0099] The compounds according to the present invention exhibit favorable cellular activity. Induction of cytokines in vitro can be measured in reporter cell lines, for example, in THP1 cells, in a manner similar to that described in WO 2016/096174. The compounds according to the present invention exhibit favorable cellular activity in cells that have different human STING alleles. Human STING exists in at least five known variants (WT, HAQ, REF / 232H, AQ and Q / 293 Q). To test the activity of different CDNs in human STING variants, KO THP1STING cells can be stably transduced with vectors that encode the different STING variants. In addition, in vitro cytokine induction can be measured in primary human PBMCs or in human dendritic cells.
[00100] The compounds according to the present invention exhibit favorable stability in in-cell assays
Petition 870190091404, of 9/13/2019, p. 36/105
30/98 in vitro, such as with THP1 cells, Calu-3 cells or human hepatocytes. In addition, the compounds according to the present invention exhibit favorable chemical stability in the solution and in the solid state.
[00101] In addition, the compounds according to the present invention exhibit favorable pharmacokinetic properties (PK). PK properties can be determined in preclinical animal species, for example, in mice, rats, hamsters, dogs, guinea pigs, mini-pigs, cynomologist monkeys and monkeys. The PK properties of a compound can be described, for example, by the following parameters: mean residence time (MRT), elimination half-life (t 1/2, that is, the time required for the concentration of the drug to reach half of its original value), volume of distribution (Vd, that is, the apparent volume in which the drug is distributed), area under the curve (AUC, that is, the whole number of the concentration-time curve after a single dose ), clearance (CL, that is, the volume of the drug's clean plasma per unit of time), as described in E. Kerns & L. Di (Drug-like properties: concepts, structure design and methods: from ADME to toxicity optimization , Elsevier, I to ed, 2008).
[00102] Certain compounds according to the present invention exhibit favorable in vivo pharmacological activity, for example, in mouse tumor models MC38, 4T1, Colon26 and EMT6 after intratumor or intravenous application.
[00103] Favorable binding affinity in combination with favorable cellular activity and / or favorable cellular stability and / or improved PK properties can
Petition 870190091404, of 9/13/2019, p. 37/105
31/98 enable lower doses for pharmacological efficacy. Lower doses have the advantages of lower drug load or drug burden (parent drug and its metabolites) for the patient, potentially causing less side effects and lower production costs for the drug product.
[00104] The binding of the compounds of the invention to human STING can be demonstrated using the following assay: SCAN DIFFERENTIAL FLUORIMETRY (DSF)
Materials:
384-well, thin-walled PGR Hard-Shell® plates (catalog η Ω HSP3805R, BIO-RAD)
Microseal® 'B' adhesive seals for PCR plates (catalog η Ω MSB-1001, BIO-RAD)
SYPRO orange solution in DMSO (SIGMA, catalog η Ω S5692500UL), 5000x concentration
Instrumentation: Reader: CFX384 system in real time (BioRad)
Pipetting robot: Hamilton Starlet
Assay buffer: 20 mM Tris, 150 mM NaCl, pH 7.5
Target protein: human STING (hSTING, residues 155-341, wild type sequence marked with His8 N-terminal and dividing site TEV, PM: 23601.5 Da)
Protein stock solution: c = 309 μΜ stock solution in assay buffer
Final test concentrations of test compounds: 100 μΜ, target protein 3 μΜ, 5x SYPR Orange
Test procedure:
1) The stock solutions of the compounds and their dilutions were prepared in assay buffer
Petition 870190091404, of 9/13/2019, p. 38/105
32/98
2) 5 μΐ of fluorescent dye stock solution (5000x SYPRO orange) were mixed with 50 μΐ of target protein (309 μΜ) and 945 μΐ of buffer.
3) 2 pL of this mixture of dye and protein (25x SYPRO orange and 15 μΜ of protein) was added to 8 μΐ of compound solution. The final volume was 10 pL.
4) Certain well positions were used as a negative control.
5) The plates were prepared for duplicate measurement and centrifuged for 2 min at 1000 g.
6) In the measurement, 160 cycles of 0.5 2 C were used (temperature ramp of 15 s / cycle, from 15 2 C to 95 -C).
Data analysis: the dissociation curves were processed in Bio-Rad's CFX manager. The peak type was defined as negative. In the case of examples 1.1, 2.1 and 4.1, at least two measurements of Tm were made.
The changes in Tm determined are shown in table 1.
Table 1: Changing the hSTING Tm
Example Changing the hSTING Tm [° C] 1, 1 16.1 1.2 12.0 2.1 23, 6 2.2 15, 0 3, 1 17, 0 3.2 9, 0 4, 1 13, 9 4.2 9, 0 ML-RR-CDA · 2Na + (compound 21 in WO 2014/189805) 12.0
[00105] The cellular activity of the compounds of the invention
Petition 870190091404, of 9/13/2019, p. 39/105
33/98 can be demonstrated using the following THP1 in vitro assay:
IN VITRO CYTOKIN INDUCTION [00106] The cytokine induction activities of the compounds according to the present invention have been demonstrated using a THP1 reporter cell line.
[00107] The activation of the STING protein expressed in cell lines results in an increase in the production of interferon. Through the stable integration of a reporter construct of SEAP (secreted embryonic alkaline phosphatase) inducible by interferon regulating factor (IRF), the functional interferon signaling pathway can be monitored. Using Invivogen's QUANTI-Blue ™ colorimetric enzyme assay and a suitable optical density (DO) reader, SEAP activity can be detected and quantified. This technique could be used to characterize the pharmacological modification of the STING protein.
[00108] SEAP activity measurements were performed on ISG THPl-Blue cells that stably express the human STING protein and the IRF-inducible SEAP reporter construct. The cells were grown for expansion in RPMI1640 medium with 10% fetal calf serum, 50 pg / ml penicillin-streptomycin, 100 pg / ml zeocin and 100 pg / ml normocin at 37 2 C, at 95% humidity and 5% CO 2 in an incubator. The cells ready for analysis were stored as frozen stocks. In preparation for the assay, the cells thawed in zeocin / normocin-free medium and were distributed on the assay plates with a density of 15,000 cells / 15 pL per well. The compounds were prepared by a serial dilution of 8
Petition 870190091404, of 9/13/2019, p. 40/105
34/98 or 16 points in 50% aqueous DMSO and with a final dilution step in medium to ensure a final concentration of 0.5% DMSO in the assay. 5 μΐ more compounds diluted 5 μΐ medium were added to the plates, followed by a period of 24 hours incubation at 37 2 C.
[00109] On the day of the test, 75 μΐ per well of Quanti-Blue reagent was added to all wells on the plate, and the plate was incubated for an additional 30 minutes at 37 2 C. The OD at 620 nm was measured in the Envision reader (PerkinElmer).
[00110] ECso values and Hill's coefficients were derived from four 8 or 16 point parametric nonlinear curve adjustments with the Megalab software (Boehringer Ingelheim) using the DO at 620 nM. The ECso values for examples 1.1, 2.1, 3.1 and 4.1 are the average of at least two measurements. See table 2.
Table 2: ECso values
Example ECso [μΜ] 1, 1 0.21 1.2 5, 8 2, 1 0.05 2.2 0.79 3, 1 0.17 3, 2 8.5 4, 1 0.19 4.2 6.2 ML-RR-CDA · 2Na + (compound 21 in WO 2014/189805) 0.65
[00111] In the THP1 test above, example 1.1 is more potent than the respective 1.2 diastereoisomeric example. An X-ray of example 1.1 in complex with the human STING indicates that both phosphorus atoms have the
Petition 870190091404, of 9/13/2019, p. 41/105
35/98 Rp configuration. Similarly, it is assumed that the respective more potent diastereoisomers of examples 2.1, 3.1 and 4.1 also have the Rp configuration on both phosphorus atoms.
[00112] Several unique nucleotide polymorphisms have been identified in the human STING gene, which can affect the response to cyclic dinucleotides. To determine the activity of the compounds of the invention, ISG THP1-BLUE reporter cell lines were generated expressing the different human STING variants. To do this, endogenous human STING was first excluded using the CRISPR / CAS9 system: ISG THPl-Blue cells were electroporated with CRISPR ALL-IN-ONE plasmids targeting the STING gene (acquired from Sigma that encodes the gRNA and GFP gene as the reporter gene for successful transduction). The GFP positive cells were then separated 24 hours after transfection and expanded. The cells were then dispersed in a semi-solid metocel medium to allow isolation of individual cell clones. The clones were then screened for cGAMP responsiveness using the Quanti-blue reporter assay. The unresponsive clones were subsequently analyzed for loss of STING by western blotting and sequencing of the STING locus.
[00113] For the overexpression of human STING variants, a confirmed KO clone of hSTING ISG THPl-Blue was transduced with individual retroviral plasmids (MSCVires-GFP-Blasti) encoding allele variants of hSTING (WT, HAQ, R232H, AQ and R293Q ). The transduced cells were separated for different levels of
Petition 870190091404, of 9/13/2019, p. 42/105
36/98 GFP fluorescence and STING allele expression were analyzed by western blot. Populations expressing the ectopic STING protein (WT, HAQ, R232H, AQ and R293Q) at levels comparable to the endogenous STING levels of the THP1Blue ISG progenitor cell lines were selected and used for CDN characterization. Examples of the present invention exhibit cellular activity in all five of the above variants.
[00114] The cellular stability of the compounds of the invention was measured as follows: the compound was dissolved in cell culture medium (MEM supplemented with 10% FCS, 1% non-essential amino acids and 1% pyruvate) at a concentration end of 10 μΜ and was incubated with the human lung epithelial cell line Calu-3 (60,000 cells / well in a 24-well plate) for up to 24h. Samples of cell culture supernatants were collected at 1, 6 and 24 hours and were quantified in LC-MS / MS.
[00115] The PK of the compounds of the invention in mice was determined using the following method:
MUSEUM PK
Animal experiment [00116] The compound was dissolved in a physiological NaCl solution and was administered intravenously to male C57BL / 6NRj mice at a dose of 10 pmol / kg. A blood sample of 20 pL was collected at 0.25, 0.5, 0.75, 1 and 2 h, using ETDA as an anticoagulant. Blood plasma was generated by centrifugation and stored at -20 ° C. The concentration of the compound was determined by LCMS / MS. The concentration-time relationships determined (mean of duplicate experiments) are shown in table 3.
Petition 870190091404, of 9/13/2019, p. 43/105
37/98
Table 3:
Example T =0.25 h T = 0.5 h T =0.75 h T = 1 h T = 2 h 1, 1 3080 nM 1200 nM 55 0 nM 24 9 nM 120 nM 2, 1 1970 nM 6 99 nM 401 nM 241 nM 7 8 nM 3, 1 2710 nM 1280 nM 5 95 nM 2 60 nM 111 nM
[00117] Accordingly, the present invention relates to compounds of formula I as a medicament.
[00118] Furthermore, the present invention relates to the use of a compound of general formula I or a pharmaceutical composition according to this invention for the treatment and / or prevention of diseases or conditions that can be influenced by the modulation of the STING in a patient or from diseases or conditions associated or modulated by STING in a patient. Preferably, the patient is a human being.
[00119] In yet another aspect, the present invention relates to a method for treating a disease or condition associated or modulated by STING in a mammal in need of such treatment, which includes the step of administering to the mammal, and preferably to a human being. human, a therapeutically effective amount of a compound or a pharmaceutical composition of the present invention.
[00120] Diseases or conditions that are associated with or that are modulated by, or may be influenced by, STING modulation include inflammation, allergic or autoimmune diseases, for example, allergic rhinitis or asthma, infectious diseases or cancer. In addition, due to their activity, the compounds of the present invention are suitable as vaccine adjuvants.
[00121] Autoimmune diseases include, but are not
Petition 870190091404, of 9/13/2019, p. 44/105
38/98 limit, systemic lupus erythematosus. Psoriasis, insulin dependent diabetes mellitus (IDDM), dermatomyositis and Sjogren's syndrome (SS).
[00122] Inflammation represents a group of vascular, cellular and neurological responses to trauma. Inflammation can be characterized as the movement of inflammatory cells, such as monocytes, neutrophils and granulocytes in tissues. This is generally associated with reduced endothelial barrier function and tissue edema. The inflammation can be classified as acute or chronic. Acute inflammation is the body's initial response to harmful stimuli and is achieved by the increased movement of plasma and leukocytes from the blood to the injured tissues. A cascade of biochemical events spreads and matures the inflammatory response, which involves the local vascular system, the immune system and several cells in the injured tissue. Prolonged inflammation, known as chronic inflammation, leads to a progressive change in the type of cells that are present at the site of the inflammation and is characterized by the simultaneous destruction and scarring of the inflammatory process tissue.
[00123] When it occurs as part of an immune response to infection or as an acute response to trauma, inflammation can be beneficial and is usually self-limiting. However, inflammation can be harmful in several conditions. This includes the production of excessive inflammation in response to infectious agents, which can lead to significant organ damage and death (for example, in the setting of sepsis). In addition, chronic inflammation is generally harmful and is at the root of numerous diseases
Petition 870190091404, of 9/13/2019, p. 45/105
39/98 chronic, causing serious and irreversible tissue damage. In these contexts, the immune response is often directed against the body's own tissues (autoimmunity), although chronic responses to foreign entities can also lead to observer damage to the tissues themselves. The aim of anti-inflammatory therapy is, therefore, to reduce this inflammation, inhibit autoimmunity, when present, and allow the progress of the physiological process or healing, and tissue repair.
[00124] The compounds of the invention can be used to treat inflammation of any tissue and organs in the body, including musculoskeletal inflammation, vascular inflammation, neural inflammation, inflammation of the digestive system, eye inflammation, inflammation of the reproductive system and other types of inflammation, as exemplified below.
[00125] Musculoskeletal inflammation refers to any inflammatory condition of the musculoskeletal system, particularly those conditions that affect skeletal joints, including joints of the hands, wrist, elbow, shoulder, jaw, spine, spine, neck, hip, knee, ankle and foot, and conditions that affect tissues connecting muscles to bones, such as tendons. Examples of musculoskeletal inflammations that can be treated with the compounds of the invention include arthritis (including, for example, osteoarthritis, rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, acute and chronic infectious arthritis, gout-associated arthritis and pseudogout and arthritis
Petition 870190091404, of 9/13/2019, p. 46/105
Juvenile idiopathic 40/98), tendinitis, synovitis, tenosynovitis, bursitis, fibrositis (fibromyalgia), epicondylitis, myositis and osteitis (including, for example, Paget's disease, pubic osteitis and fibrocystic osteitis). Eye inflammation refers to the inflammation of any structure in the eye, including the eyelids. Examples of ocular inflammation, which can be treated with the compounds of the invention, include blepharitis, blepharocalase, conjunctivitis, dacrioadenitis, keratitis, dry keratoconjunctivitis (dry eye), scleritis, trichiasis and uveitis. Examples of inflammation of the nervous system, which can be treated with the compounds of the invention, include encephalitis, Guillain-Barré syndrome, meningitis, neuromyotonia, narcolepsy, multiple sclerosis, myelitis and schizophrenia.
[00126] Examples of inflammation of the vasculature or the lymphatic system, which can be treated with the compounds of the invention, include arthrosclerosis, arthritis, phlebitis, vasculitis and lymphangitis.
[00127] Examples of inflammatory conditions of the digestive system, which can be treated with the compounds of the invention, include cholangitis, cholecystitis, enteritis, enterocolitis, gastritis, gastroenteritis, inflammatory bowel disease (such as Crohn's disease and ulcerative colitis), ileitis and proctitis.
[00128] Examples of inflammatory conditions of the reproductive system that can be treated with the compounds of the invention include cervicitis, chorioamnionitis, endometritis, epididymitis, omphalitis, oophoritis, orchitis, salpingitis, ovarian tube abscess, urethritis, vaginitis, vulvitis and vulvodynia.
Petition 870190091404, of 9/13/2019, p. 47/105
41/98 [00129] The agents can be used to treat autoimmune diseases having an inflammatory component. Such conditions include universal acute disseminated alopecia, Behcet's disease, Chagas' disease, chronic fatigue syndrome, dysautonomia, encephalomyelitis, ankylosing spondylitis, aplastic anemia, suppurative hydradenitis, autoimmune hepatitis, autoimmune oophoritis, celiac disease, Crohn's disease, diabetes mellitus type 1, giant cell arteritis, goodpasture syndrome, Grave's disease, Guillain-Barré syndrome, Hashimoto's disease, Henoch-Schönlein purpura, Kawasaki's disease, lupus erythematosus, microscopic colitis, microscopic polyarteritis, connective tissue disease, sclerosis multiple, myasthenia gravis, opsoclonia-myoclonia syndrome, optic neuritis, ord thyroiditis, pemphigus, polyarteritis nodosa, polymyalgia, rheumatoid arthritis, Reiter's syndrome, Sjogren's syndrome, temporal arteritis, Wegener's granulomatosis, autoimmune hemolytic anemia, autoimmune hemolytic type interstitial, lyme disease, morphea, psoriasis, sarcoidosis, scleroderma, colitis ulcerative and vitiligo.
[00130] The agents can be used to treat diseases of hypersensitivity mediated by T cells with an inflammatory component. Such conditions include contact hypersensitivity, contact dermatitis (including that due to poison ivy), hives, skin allergies, respiratory allergies (rhinitis, allergic rhinitis) and gluten-sensitive enteropathy (celiac disease).
[00131] Other inflammatory conditions that can be treated with the agents include, for example, appendicitis,
Petition 870190091404, of 9/13/2019, p. 48/105
42/98 dermatitis, dermatomyositis, endocarditis, fibrositis, gingivitis, glossitis, hepatitis, hidradenitis suppurativa, iritis, laryngitis, mastitis, myocarditis, nephritis, otitis, pancreatitis, parotitis, percarditis, peritonoitis, pharyngitis, pleuritis, pneumonitis, prostatitis and pyelonephritis stomatitis, transplant rejection (involving organs such as kidneys, liver, heart, lung, pancreas (eg islet cells), bone marrow, cornea, small intestine, skin allografts, skin homografts and heart valve xenografts, serum disease, and skin versus host disease), acute pancreatitis, chronic pancreatitis, acute respiratory distress syndrome, Sexary syndrome, congenital adrenal hyperplasia, non-suppurative thyroiditis, cancer-associated hypercalcemia, pemphigus, herpetiform bullous dermatitis, severe multiform erythema , exfoliative dermatitis, seborrheic dermatitis, seasonal or perennial allergic rhinitis, bronchial asthma, contact dermatitis, atopic dermatitis, drug hypersensitivity reactions, allergic conjunctivitis, gueratitis, ophthalmic herpes zoster, iritis and oiridocyclitis, chorioretinitis, optic neuritis, symptomatic sarcoidosis, guimotherapy against fulminant or disseminated pulmonary tuberculosis, idiopathic thrombocytopenia in adult, idiopathic thrombocytopenia, in idiopathic thrombocytopenia in idiopathic thrombocytopenia. adhered hemolytic (autoimmune), leukemia and lymphomas in adults, acute childhood leukemia, regional enteritis, autoimmune vasculitis, multiple sclerosis, chronic obstructive pulmonary disease, rejection of solid organ transplantation and sepsis. Preferred treatments include transplant rejection treatment, rheumatoid arthritis, arthritis
Petition 870190091404, of 9/13/2019, p. 49/105
43/98 psoriatic and multiple sclerosis. Type 1 diabetes, asthma, inflammatory bowel disease, systemic lupus erythematosus, psoriasis, chronic lung disease and inflammation that accompanies infectious conditions (for example, sepsis).
[00132] In one aspect, the disease or condition to be treated using the compounds of the invention is cancer. Examples of cancer diseases and conditions, where the compounds of formula I, or the pharmaceutically acceptable salts or solvates thereof, may have potentially beneficial antitumor effects include, but are not limited to, lung, bone, pancreas, skin, head cancers , neck, uterus, ovaries, 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 cells; soft tissue sarcoma; myxoma; rhabdomyoma; fibroma; lipoma; teratoma; cholangiocarcinoma; hepatoblastoma; angiosarcoma; hemangioma; hepatoma; fibrosarcoma; chondrosarcoma; myeloma; acute or chronic leukemia; lymphocytic lymphoma; primary CNS lymphoma; CNS neoplasms; tumors of the spine axis; squamous cell carcinomas; synovial sarcoma; malignant pleural mesothelioma; brain stem glioma; pituitary adenoma; bronchial adenoma; chondromatous hanlartoma; inesothelioma; Hodgkin's disease or a combination of one or more of the previous cancers.
[00133] Preferred cancers, which can be
Petition 870190091404, of 9/13/2019, p. 50/105
44/98 treated with the compounds according to the invention, are cancers of skin, lung, liver, colon, brain, breast, ovary, prostate, pancreas, kidney, stomach, head, neck and urothelial, as well as lymphoma and leukemia.
[00134] The new compounds can be used for the prevention, short or long-term treatment of the aforementioned diseases, and optionally also in combination with surgery, radiation therapy or other state of the art compounds such as cytostatic or cytotoxic substances , cell proliferation inhibitors, antiangiogenic substances, steroids or antibodies.
[00135] In their functions as adjuvants, in certain modalities, the present compounds and compositions can be used as adjuvants in a therapeutic or prophylactic strategy that uses vaccine (s). Thus, the substantially pure CDNs of the present invention, or the prodrugs or the pharmaceutically acceptable salts or salts thereof, can be used in conjunction with one or more selected vaccines to stimulate an immune response to one or more predetermined antigens. The substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof, can be supplied together with, or in addition to, such vaccines.
[00136] Such vaccines may comprise inactivated or attenuated bacteria or viruses comprising the antigens of interest, purified antigens, recombinantly manipulated live viral or bacterial release vectors
Petition 870190091404, of 9/13/2019, p. 51/105
45/98 to express and / or secrete antigens, antigen presenting cell vectors (APC) comprising cells that are loaded with the antigens or transected with a composition comprising a nucleic acid encoding the antigens, liposomal antigen delivery vehicles or naked nucleic acid vehicles that encode antigens. This list is not exhaustive. As an example, such vaccine (s) may also comprise an inactivated tumor cell that expresses and secretes one or more of the GM-CSF, CCL20, CCL3, IL-12p70, FLT-3 and cytokine ligands.
[00137] In a related aspect, the present invention relates to methods of inducing, stimulating or adjuvanting an immune response in an individual. These methods comprise administering the substantially pure CDNs of the present invention, or prodrugs or pharmaceutically acceptable salts thereof to the individual.
[00138] The dose range of the compounds of formula I applicable per day is normally 0.0001 to 10 mg, for example, 0.001 to 1 mg. Each dosage unit can conveniently contain from 0.0001 to 10 mg, for example, from 0.001 to 1 mg.
[00139] The effective therapeutically effective amount or therapeutic dosage will naturally depend on factors known to those skilled in the art, such as the age and weight of the patient, the route of application and the severity of the disease. In any case, the compound or composition will be administered in dosages and in a manner that allows a therapeutically effective amount to be administered based on the patient's unique condition.
Petition 870190091404, of 9/13/2019, p. 52/105
The compounds and compositions, including any combinations with one or more additional therapeutic agents, according to the invention, can be administered via the mucosa (e.g., oral, sublingual, vaginal, nasal, cervical, etc. .), intratumoral, peritumoral, transdermal, inhalation or parenteral (for example, by subcutaneous, intravenous, intramuscular, intraarterial, intradermal, intrathecal and epidural). In most cases, one of the intravenous, intratumoral, peritumoral or subcutaneous routes of administration is administered.
Pharmaceutical Compositions [00141] For the purposes of this disclosure, pharmaceutical compositions can be administered by a variety of means, including non-parenterally, parenterally, by inhalation spray, topically or rectally in formulations containing pharmaceutically acceptable carriers, adjuvants and vehicles. The intratumoral (directly in the tumor mass) or peritumoral (around the tumor mass) administration of the compounds of the present invention can directly activate the local infiltrating DC, directly promote the apoptosis of the tumor cells or sensitize the tumor cells to cytotoxic agents.
[00142] The pharmaceutical compositions of the disclosure may be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known technique, using those suitable dispersing or wetting agents and suspending agents, which have been
Petition 870190091404, of 9/13/2019, p. 53/105
47/98 mentioned above. The sterile injectable preparation can also be a sterile injectable solution or suspension in a parenterally acceptable non-toxic diluent or solvent, for example, as a solution in 1,3-butanediol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that can be used are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils can conventionally be used as a solvent or suspending medium. For this purpose, any soft fixed oil, including synthetic mono or diglycerides, can be used. In addition, fatty acids, such as oleic acid, can also be used in the preparation of injectables.
[00143] Formulations suitable for topical administration in the mouth include lozenges, comprising the active ingredient in a flavored base, usually sucrose and acacia or tragacanth; lozenges, comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia; and mouth rinses comprising the active ingredient in a suitable liquid carrier.
[00144] Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or spray formulations containing, in addition to the active ingredient, such vehicles, as is known in the art to be appropriate.
[00145] Formulations suitable for parental administration include aqueous and non-aqueous isotonic sterile injection solutions, which may contain antioxidants,
Petition 870190091404, of 9/13/2019, p. 54/105
48/98 buffers, bacteriostats and solutes, which make the formulation isotonic with the intended recipient's blood; and aqueous and non-aqueous sterile suspensions, which may include suspending agents and thickening agents. The formulations can be presented in sealed single-dose or multiple-dose containers, for example, as ampoules and vials, and can be stored in a lyophilized condition (freeze-dried), requiring only the addition of the sterile liquid carrier, for example, water for injection, just before use. Suspensions and
solutions injection can be prepared from powders, granules, and tablets sterile type previously described. Therapy combination [00146] The compounds of invention can be used alone or can be combined with excipients
pharmaceutically acceptable, in an amount sufficient to induce, modify or stimulate an appropriate immune response. The immune response can comprise, without limitation, a specific immune response, a non-specific immune response, an both specific and non-specific immune response, an innate response, a primary immune response, an adaptive immunity, a secondary immune response, an immune response of memory, an activation of the immune system cell, proliferation of the immune system, differentiation of immune cells and expression of cytokines. In certain embodiments, the compounds and compositions thereof described herein are administered together with one or more
Petition 870190091404, of 9/13/2019, p. 55/105
49/98 additional compositions, including vaccines designed to stimulate an immune response to one or more predetermined antigens; adjuvants; antagonists of the CTLA-4 and PD-1 pathway, lipids, liposomes, chemotherapeutic agents, immunomodulatory cell lines, etc.
The compounds and compositions thereof described in the present invention can be administered before, after and / or simultaneously with an additional therapeutic or prophylactic composition or modality. These include, without limitation, the co-stimulatory molecule B7, interleukin-2, interferon-g, GM-CSF, CTLA4 antagonists, OX-40 / OX-40 ligand, CD40 / CD40 ligand, sargramostim, levamisole, vaccinia virus, Calmette bacillus -Wine (BCG), liposomes, alum, Freund's complete or incomplete adjuvant, detoxified endotoxins, mineral oils, surfactants such as lipolecithin, pluronic polyols, polyanions, peptides and oil or hydrocarbon emulsions. Vehicles to induce a T cell immune response that preferentially stimulate a cytolytic T cell response versus an antibody response are preferred, although those that stimulate both types of response can be used as well. In cases where the agent is a polypeptide, the polypeptide itself or a polynucleotide that encodes the polypeptide can be administered. The vehicle can be a cell, such as an antigen presenting cell (ABC) or a dendritic cell. Antigen presenting cells include such cell types as macrophages, dendritic cells and B cells. Other professional antigen presenting cells include monocytes, Kupffer cells from the zone
Petition 870190091404, of 9/13/2019, p. 56/105
50/98 marginal, microglia, Langerhans cells, interdigitating dendritic cells, follicular dendritic cells and T cells. Optional antigen presenting cells can also be used. Examples of facultative antigen presenting cells include astrocytes, follicular, endothelial cells and fibroblasts. The vehicle can be a bacterial cell that is transformed to express the polypeptide or to deliver a polynucleotide that is subsequently expressed in the cells of the vaccinated individual. Adjuvants, such as aluminum hydroxide or aluminum phosphate, can be added to increase the vaccine's ability to trigger, enhance or prolong an immune response. Additional materials, such as cytokines, chemokines and bacterial nucleic acid sequences, such as CpG, a Toll ligand receptor (TLR) 9 antagonist, as well as additional agonists for TLR 2, TLR 4, TLR 5, TLR 7, TLR 8 and TLR9, including lipoprotein, LPS, monophosphoryl lipid A, lipoteic acid, imiquimod, resiquimod, and in addition, retinoic acid-inducible gene (RIG-I) agonists, such as poly I: C, used separately or in combination with described compositions, are also potential adjuvants. Other representative examples of adjuvants include the synthetic QS-21 adjuvant, comprising a homogeneous saponin purified from the bark of Quillaja saponaria and Corynebacterium parvum (McCune et al., Cancer, 1979; 43: 1619).
[00148] Methods for co-administration with an additional therapeutic agent are well known in the art
Petition 870190091404, of 9/13/2019, p. 57/105
.. 51/98 (Hardman, et al (eds) (2001) Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed, McGraw - Hill, New York, NY,.. Poole and Peterson (eds) (2001) Pharmacotherapeutics for Advanced Practice: A Practical Approach, Lippincott, Williams & Wilkins, Phila., PA; Chabner and Longo (eds.) (2001) Cancer Chemotherapy and Biotherapy, Lippincott, Williams & Wilkins, Phila., PA). Generally, co-administration or joint administration indicates the treatment of an individual with two or more agents, where the agents can be administered simultaneously or at different times. For example, such agents can be administered to a single individual as separate administrations, which can occur at essentially the same time or at different times, and which can be by the same route or by different routes of administration. These agents can be administered to a single individual in the same administration (for example, in the same formulation) in such a way that they are administered at the same time by the same route of administration.
[00149] Due to the adjuvant properties of the compounds of the present invention, their use can also be combined with other therapeutic modalities, including other vaccines, adjuvants, antigen, antibodies and immunomodulators. Examples are provided below.
Adjuvants [00150] In addition to the compounds of the present invention and their compositions described herein, the compositions or methods of the present invention may further comprise one or more additional substances which, due to their nature, may act to stimulate or otherwise use the system
Petition 870190091404, of 9/13/2019, p. 58/105
52/98 immune system to respond to cancer antigens present in the target tumor cell (s). Such adjuvants include, but are not limited to, lipids, liposomes, inactivated bacteria that induce innate immunity (eg, inactivated or attenuated Listeria monocytogenes), compositions that mediate innate immune activation via Toll-type receptors (TLRs) type receptors ( NOD) (NLRs), type (RIG) -I receptors based on the retinoic acid-inducible gene (RLRs), type C lectin receptors (CLRs) and / or pathogen-associated molecular patterns (PAMPS). Examples of PAMPs include lipoproteins, lipopolypeptides, peptidoglycans, zymosan, lipopolysaccharide, neisserial porins, flagellin, profilin, galactoceramide and muramyl dipeptide. Peptidoglycans, lipoproteins and lipoteic acids are components of the cell wall of gram-positive. Lipopolysaccharides are expressed by most bacteria, MPL being an example. Flagellin refers to the structural component of bacterial flagella that is secreted by pathogenic and commensal bacteria. Galactosylceramide is a natural killer T cell activator (NKT). The muramyl dipeptide is a portion of bioactive peptidoglycan common to all bacteria.
Immune Checkpoint Inhibitors
[00151] The compounds in present invention can to be used together with an inhibitor in Score in verification immunological , as an inhibitor in Score in verification immunological selected from the group consisting in
an antagonist of the CTLA-4 pathway, an antagonist of the PD-1 pathway, an antagonist of the Tim-3 pathway, an antagonist of the Vista pathway,
Petition 870190091404, of 9/13/2019, p. 59/105
53/98 an antagonist of the BTLA pathway, an antagonist of the LAG-3 pathway or an antagonist of the TIGIT pathway. In some embodiments, the immunological checkpoint inhibitor is selected from the group consisting of an anti-CTLA-4 antibody, an anti-PD-1 antibody, an anti-Tim-3 antibody, an anti-Vista antibody, an anti -BTLA, an anti-LAG-3 antibody or an anti-TIGIT antibody.
[00152] The compounds of the present invention can be used in conjunction with antagonists of the CTLA-4 pathway. In some embodiments, the combination is used to treat a solid tumor or a hematological malignancy. CTLA-4 is believed to be an important negative regulator of the adaptive immune response. Activated T cells suppress CTLA-4, which binds to CD80 and CD86 in antigen presenting cells with greater affinity than CD28, thereby inhibiting T-cell stimulation, IL-2 gene expression and proliferation of T cells. The anti-tumor effects of CTLA4 blockade have been observed in murine models of colon carcinoma, metastatic prostate cancer and metastatic melanoma. In some embodiments, the CTLA-4 pathway antagonist is an anti-CTLA-4 antibody molecule selected from the group consisting of tremelimumab and ipilimumab.
[00153] Ipilimumab (an CTLA-4 antibody, also known as MDX-010, CAS No. 477202-00-9) and tremelimumab (IgG2 monoclonal antibody formerly known as ticilimumab, CP-675206) are humanized monoclonal antibodies that bind to human CTLA4 and prevent its interaction with CD80 and CD86. Other negative immune regulators that may be the target of a
Petition 870190091404, of 9/13/2019, p. 60/105
54/98 Similar strategies include programmed cell death 1 (PD-1), B and T lymphocyte attenuator, transforming growth factor beta, interleukin-10 and vascular endothelial growth factor.
[00154] In some embodiments, the compounds of the present invention can be used in combination with an anti-CTLA-4 antibody and an anti-PD-1 antibody. In one embodiment, the combination includes an anti-PD-1 antibody molecule, for example, as described in the present invention, and an anti-CTLA-4 antibody, such as, for example, ipilimumab. Exemplary doses that can be used include a dose of an anti-PD-1 antibody molecule of about 1 to 10 mg / kg, for example, 3 mg / kg, and a dose of a CTLA-4 antibody, for example , of ipilimumab, of about 3 mg / kg.
[00155] The compounds of the present invention can be used in conjunction with antagonists of the CTLA-4 pathway. In some embodiments, the combination is used to treat a solid tumor or a hematological malignancy. PD-1 is another negative regulator of the adaptive immune response that is expressed in activated T cells. PD-1 binds to B7-H1 and B7-DC, and binding to PD-1 suppresses T cell activation. Antitumor effects have been demonstrated by blocking the PD-1 pathway. The molecules of the antiPD-1 antibodies (for example, Nivolumab (Opdivo®), pembrolizumab (Keytruda® and pidilizumab) and AMP-224 have been reported in the literature as being examples of the PD-1 pathway blockers that can find use in the present invention. In some embodiments, the PD-1 pathway antogonist is an anti-PD-1 antibody molecule selected from the group consisting of
Petition 870190091404, of 9/13/2019, p. 61/105
55/98 nivolumab, pembrolizumab or pidilizumab.
[00156] In some embodiments, the PD-1 pathway antagonist is an immunoadhesin (e.g., an immunoadhesin comprising an extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region (for example , to an Fc region of an immunoglobulin sequence.) In some embodiments, the PD-1 inhibitor is AMP-224 (B7-DCIg; Amplimmune; for example, disclosed in W02010 / 027827 and WO2011 / 066342) is a receptor PD-L2 Fc fusion solution that blocks the interaction between PD-1 and B7-H1.
[00157] In some embodiments, the PD-1 pathway antagonist is a PD-L1 or PD-L2 inhibitor. In some embodiments, the PD-L1 or PD-L2 inhibitor is an anti-PD-L1 antibody or an anti-PD-L2 antibody. In some embodiments, the anti-PD-LI inhibitor is chosen from YW243.55.S70, MPDL3280A, MEDI-4736, MSB-0010718C or MDX-1105. In some embodiments, the PD-L1 inhibitor is an anti-PD-Ll antibody MSB0010718C. MSB0010718C (also referred to as A09-246-2; Merck Serono) is a monoclonal antibody that binds to PDL1.
[00158] The compounds of the present invention can be used in conjunction with antagonists of the CTLA-4 pathway. In some embodiments, the combination is used to treat a solid tumor or a hematological malignancy. In some embodiments, the TIM-3 pathway antagonist is an anti-TIM-3 antibody. In some embodiments, the anti-TIM-3 antibody molecules are disclosed in US 2015/0218274, published on August 6, 2015, entitled Antibody Molecules to TIM-3 and Uses Thereof.
[00159] The compounds of the present invention can be
Petition 870190091404, of 9/13/2019, p. 62/105
56/98 used together with antagonists of the LAG-3 pathway. In some embodiments, the combination is used to treat a solid tumor or a hematological malignancy. In some embodiments, the antagonist of the LAG-3 pathway is an anti-LAG-3 antibody. In some embodiments, the anti-LAG-3 antibody molecules are disclosed in US 2015/0259420, filed on March 13, 2015 and titled Antibody Molecules to LAG-3 and Uses Thereof.
Amino acid catabolism [00160] The compounds of the present invention can be used together with an inhibitor of amino acid metabolism, such as an IDO or arginasel or arginase2 inhibitor, to antagonize the immune inhibitory effect of immune-suppressing immune cells, such as suppressor cells myeloid derivatives.
Purinergic signaling pathway [00161] The compounds of the present invention can be used in conjunction with purinergic signaling pathway inhibitors, such as CD39 and CD73 pathway antagonists or A2A / A2B receptor inhibitors.
Chemokines and chemokine receptors [00162] The compounds of the present invention can be used in conjunction with the chemokine or chemokine receptor antagonist to inhibit the recruitment of suppressor immune cells into the tumor microenvironment. For example, but not exclusively, the compounds of the present invention can be used in conjunction with the CCR2 or CCR5 antagonist to reduce the infiltration of myeloid suppressor cells and regulatory T cells.
T cell receptor agonists
Petition 870190091404, of 9/13/2019, p. 63/105
57/98 [00163] The compounds of the present invention can be used together with a T cell receptor agonist, such as a CD28 agonist, a 0X40 agonist, a GITR agonist, a CD137 agonist, a CD27 agonist or an HVEM agonist.
The compounds of the present invention can be used together with a CD27 agonist. Exemplary CD27 agonists include an anti-CD27 agonist antibody, for example, as described in PCT Publication No. WO 2012/004367.
[00165] The compounds of the present invention can be used together with a GITR agonist. In some embodiments, the combination is used to treat a solid tumor or a hematological malignancy. Exemplary GITR agonists include, for example, GITR fusion proteins and anti-GITR antibodies (for example, divalent anti-GITR antibodies).
TLR agonists [00166] The compounds of the present invention can be used together with a Toll-like receptor agonist. The term Toll-type receptor (or TLR), as used in the present invention, refers to a member of the Toll-type receptor protein family, or a fragment thereof, that detects a microbial product and / or initiates a adaptive immune response. In one embodiment, a TLR activates a dendritic cell (DC). Toll-type receptors (TLRs) are a family of pattern recognition receptors that were initially identified as sensors of the innate immune system that recognize microbial pathogens. The
Petition 870190091404, of 9/13/2019, p. 64/105
58/98
TLRs comprise a conserved membrane family that comprises molecules containing a leucine-rich repeat ectodomain, a transmembrane domain and an intracellular TIR domain (Toll / IL-IR). TLRs recognize distinct structures in microbes, often referred to as PAMPs (molecular patterns associated with a pathogen). Binding of the ligand to TLRs invokes a cascade of intracellular signaling pathways that induce the production of factors involved in inflammation and immunity.
[00167] TLR agonists known in the art and which find use in the present invention include, but are not limited to, the following:
Pam3Cys, a TLR-1/2 agonist;
CFA, a TLR-2 agonist;
MALP2, a TLR-2 agonist;
Pam2Cys, a TLR-2 agonist;
FSL-1, a TLR-2 agonist;
Hib-OMPC, a TLR-2 agonist;
Polyribosinic acid: polyribocytoid (Poly I: C), a TLR-3 agonist;
Polyadenosine-polyuridyl acid (poly AU), a TLR-3 agonist;
Polinosynic-polycytidyl acid stabilized with poly-L-lysine and carboxymethylcellulose (Hiltonol®), a TLR-3 agonist;
Monophosphoryl lipid A (MPL), a TLR-4 agonist;
LPS, a TLR-4 agonist;
bacterial flagellin, a TLR-5 agonist;
sialyl-Tn (STn), a carbohydrate associated with MUC1 mucin in several humans
Petition 870190091404, of 9/13/2019, p. 65/105
59/98 cancer cells and a TLR-4 agonist;
imiquimod, a TLR-7 agonist;
resiquimod, a TLR-7/8 agonist;
loxoribine, a TLR-7/8 agonist; and
Unmethylated CpG dinucleotide (CpG-ODN), a TLR-9 agonist.
[00168] Due to their adjuvant qualities, TLR agonists are preferably used together with other vaccines, adjuvants and / or immunomodulators, and can be combined in various combinations. Thus, in certain embodiments, mono or di-FCDN compounds that bind to STING and induce the activation of STING-dependent TBK1 and an inactivated tumor cell that expresses and secretes one or more cytokines that stimulate induction, recruitment and / or maturation dendritic cells, as described herein, can be administered together with one or more TLR agonists for therapeutic purposes.
Antibody Therapeutic Agents [00169] The compounds of the present invention can be used in conjunction with therapeutic antibodies. In some embodiments, the therapeutic antibody's mechanism of action is antibody-dependent cell-mediated cytotoxicity (ADCC). ADCC is a cell-mediated immune defense mechanism by which an effector cell of the immune system actively smooths a target cell, whose membrane surface antigens have been linked by specific antibodies. This is one of the mechanisms by which antibodies, as part of the humoral immune response, can act to limit and
Petition 870190091404, of 9/13/2019, p. 66/105
60/98 contain an infection. Classical ADCC is mediated by natural killer cells (NK); macrophages, neutrophils and eosinophils can also mediate ADCC. ADCC is an important mechanism of action of therapeutic monoclonal antibodies, including trastuzumab and rituximab, against tumors. The compounds of the present invention can act to potentiate ADCC.
[00170] The following is an exemplary list of antibodies that can be used together with the compounds of the present invention:
Muromonab-CD3, infliximab, Adalimumab, Omalizumab, Daclizumab, Rituximab, Ibritumomab, Tositumomab, Cetuximab, Trastuzumab, Alentuzumab, Lym-1 Ipilimumab, Vitaxina, Bevacizumab and Abciximabe.
[00171] Additional therapeutic antibodies that can be used in conjunction with the compounds of the present invention include a prolactin receptor inhibitor (PRLR), an HER3 inhibitor, an EGFR2 and / or EGFR4 inhibitor, an M-CSF inhibitor, an anti-APRIL antibody or an anti-SIRPa or anti-CD47 antibody.
Chemotherapeutic agents [00172] In the additional embodiments of the methods described herein, the compounds of the present invention are used together with chemotherapeutic agents (for example, small molecule pharmaceutical compounds). Thus, the methods further involve administering to the individual an effective amount of one or more chemotherapeutic agents as an additional treatment or combination treatment. In certain embodiments, the one or more chemotherapeutic agents is / are selected from the group that
Petition 870190091404, of 9/13/2019, p. 67/105
61/98 consists of abiraterone acetate, altretamine, anhydrous vinblastine, auristatin, bexarotene, bicalutamide, BMS 184476, 2,3,4,5,6-pentafluor-N- (3-fluor-4methoxyphenyl) benzenesulfonamide, bleomycin, N, N-dimethyl-Lvalyl-L-valyl-N-methyl-L-valyl-L-proliol-l-Lprolinatbutylamide, caquectin, cemadotine, chlorambucil, cyclophosphamide, 3 ', 4'-dide-hydro-4'-deoxy- 8'norvincaleucoblastine, docetaxol, docetaxel, cyclophosphamide, carboplatin, carmustine, cisplatin, cryptophycin, cyclophosphamide, cytarabine, dacarbazine (DTIC), dactinomycin, daunorubicin, decitabine dolastatin, hydroxyamide, hydroxyamide, hydroxyamide, hydroxyamide , ifosfamide, liarozole, lonidamine, lomustine (CCNU), enzalutamide, mecloretamine (nitrogen mustard), melphalan, mivobulin isethionate, rhizoxin, sertenef, streptozocin, mitomycin, methotrexate, taxanes, nilutamine, proclone; , estrogen phosphate amustine, tamoxifen, tasonermin, taxol, tretinoin, vinblastine, vincristine, vindesine sulfate and vinflunine.
[00173] In additional embodiments of the methods described in the present invention, the compounds of the present invention are used together with chemotherapeutic agents and / or additional agents to treat the indications, as described in the methods of the present invention. In some embodiments, the compounds of the present invention are used together with one or more agents selected from the group consisting of sotrastaurine, nilotinib, 5- (2,4-dihydroxy-5-isopropylphenyl) -N-ethyl-4- (4- ( morpholinomethyl)
Petition 870190091404, of 9/13/2019, p. 68/105
62/98 phenyl) isoxazol-3-carboxamide, dactolisib, 8- (6methoxypyridin-3-yl) -3-methyl-1- (4-piperazin-1-yl-3trifluoromethylphenyl) -1,3-dihydroimidazo [4 , 5-c] quinolin-2one, 3- (2,6-dichloro-3,5-dimethoxyphenyl) -1- (6 - ((4- (4ethylpiperazin-1-yl) phenyl) amino) pyrimidin-4-yl ) -1methylurea, buparlisib, 8- (2,6-difluor-3,5-dimethoxyphenyl) N- (4 - ((dimethylamino) methyl) -1H-imidazol-2-yl) quinoxaline-5carboxamide, (S) -Nl - (4-methyl-5- (2- (1,1,1-trifluor-2methylpropan-2-yl) pyridin-4-yl) thiazol-2-yl) pyrrolidine-1,2-dicarboxamide, (S) -1- (4-chlorophenyl) -7-isopropoxy-6-methoxy2- (4- (methyl - (((Ir, 4S) -4- (4-methyl-3-oxopiperazin-1-yl) cyclohexyl) methyl) amino) phenyl ) -1,2-dihydroisoquinolin-3 (4H) -one, deferasirox, letrozole, (4S, 5R) -3- (2'-amino-2-morpholino-4 '(trifluoromethyl) - [4,5' -bipyrimidin] -6-yl) -4- (hydroxymethyl) 5-methyloxazolidin-2-one, (S) -5- (5-chloro-1-methyl-2-oxo-1,2,2hydro-pyridin-3-yl ) -6- (4-chlorophenyl) -2- (2,4dimethoxypyrimidin-5-yl) -1-isopropyl-5,6-dihydropyrrolo [3,4-d] imidazol-4 (IH) -one, 4- ( (2 - (((lR, 2R) -2hydroxycyclohexyl) amino) be nzo [d] thiazol-6-yl) oxy) -Nomethylpicolinamide, imatinib mesylate, 2-fluor-N-methyl4- (7- (quinolin-6-ylmethyl) imidazo [1,2-b] [1,2, b, 4] triazin-2yl) benzamide, ruxolitinib, panobinostat, osilodrostat, (S) -N - ((S) -1-cyclohexyl-2 - ((S) -2- (4- (4-fluorobenzoyl) thiazol- 2-yl) pyrrolidin-1-yl) -2-oxoethyl) -2- (methylamino) propanamide, (S) -N - ((S) -1-cyclohexyl-2 - ((S) -2- ( 4- (4fluorbenzoyl) thiazol-2-yl) pyrrolidin-1-yl) -2-oxoethyl) -2 (methylamino) propanamide, sonidegib phosphate, ceritinib, 7-cyclopentyl-N, N-dimethyl-2 - ((5 - (piperazin-1-yl) pyridin-2yl) amino) -7H-pyrrolo [2,3-d] pyrimidine-6-carboxamide, N- (4 ((1R, 3S, 5S) -3-amino-5- methylcyclohexyl) pyridin-3-yl) -6 Petition 870190091404, of 9/13/2019, p. 69/105
63/98 (2,6-difluorfenyl) -5-fluorpicolinamide, 2- (2 ', 3-dimethyl [2,4'-bipyridin] -5-yl) -N- (5- (pyrazin-2-yl) pyridin-2-yl) acetamide, encouragefenib, 7-cyclopentyl-N, N-dimethyl-2 - ((5 ((IR, 6S) -9-methyl1-4-OXO-3,9-diazabicycles [4.2.1] -nonan-3-yl) pyridin-2-yl) amino) -7H-pyrrolo [2,3-d] pyrimidine-6carboxamide, binimetinib, midostaurine, everolimus, 1methyl-5 - ((2- (5- (trifluormethyl) -lH-imidazol-2-yl) pyridin-4yl) oxy) -N- (4- (trifluormethyl) phenyl) -IH-benzo [d] imidazole 2amine, pasireotide diaspartate, dovitinib, (R, E) -N- (7chloro-1- (1- (4- (dimethylamino) but-2-enoyl) azepan-3-yl) -lHbenzo [d] imidazol-2-yl) -2-methylisonicotinamide, N6- (2isopropoxy-5-methyl -4- (1-methylpiperidin-4-yl) phenyl) -N4- (2 (isopropylsulfonyl) -phenyl) -IH-pyrazolo [3,4-d] pyrimidine4,6-diamine, Ι, Ι-3- (4- (4 - ((5-chloro-4 - (((5-methylH-pyrazol-3-yl) amino) pyrimidin-2-yl) amino) -5-fluor-2methylphenyl) piperidin-1-yl) tiethane , 5-chloro-N2- (2-fluor-5methyl-4- (1- (tetrahydro-2H-pyran-4-yl) -piperidin-4yl) phenyl) -N4- (5-methyl-1H-pyrazole -3-yl) pyrimidine-2,4-day ine, 5-chloro-N2- (4- (1-ethylpiperidin-4-yl) -2-fluor-5methylphenyl) -N4- (5-methyl-1H-pyrazol-3-yl) pyrimidine-2,4diamine, valspodar and vatalanib succinate.
[00174] In other embodiments, the compounds of the present invention can be used together with a PKC inhibitor, a BCR-ABL inhibitor, an HSP90 inhibitor, a PI3K inhibitor and / or mTOR, an FGFR inhibitor, an inhibitor PI3K inhibitor, FGFR inhibitor, PI3K inhibitor, cytochrome P450 inhibitor (for example, CYP17 inhibitor), HDM2 inhibitor, aromatase inhibitor, p53 and / or p53 / Mdm2 interaction or a CSF-1R tyrosine kinase inhibitor.
Petition 870190091404, of 9/13/2019, p. 70/105
64/98 [00175] Suitable preparations include, for example, tablets, capsules, suppositories, solutions particularly solutions for injection (sc, iv, im) and infusion - elixirs, emulsions or dispersible powders. The content of the pharmaceutically active compound (s) should be in the range of 0.1 to 90% by weight, preferably 0.5 to 50% by weight of the composition as a whole, that is, in quantities sufficient to achieve the dosage range specified below. The specified doses can, if necessary, be administered several times a day.
[00176] The dosage for the combination partners mentioned above is generally 1/5 of the lowest dose normally recommended up to 1/1 of the dose normally recommended.
[00177] In another aspect, the present invention relates to a method for treating a disease or condition associated with, or modulated by, or which can be influenced by the modulation of STING in a patient, said method including the step of administering to the patient , preferably to a human being in need of such treatment, a therapeutically effective amount of a compound of the present invention together with a therapeutically effective amount of one or more additional therapeutic agents described above.
[00178] The use of the compound according to the invention, together with the additional therapeutic agent, can occur simultaneously or at staggered times.
[00179] The compound according to the invention and the one or more additional therapeutic agents can be present together in one formulation or separately in two
Petition 870190091404, of 9/13/2019, p. 71/105
65/98 identical or different formulations, for example, as a so-called kit of parts.
[00180] Consequently, in another aspect, this invention relates to a pharmaceutical composition comprising a compound according to the invention and one or more additional therapeutic agents described hereinbefore and hereinafter, optionally together with one or more vehicles and / or inert diluents.
[00181] Other features and advantages of the present invention will become apparent from the more detailed Examples below, which illustrate, by way of example, the principles of the invention.
Synthesis of compounds according to the present invention GENERAL TECHNICAL OBSERVATIONS
[00182] The terms temperature environment and temperature environmental are used indistinctly and designate a temperature of about 20 0 C, for example, in
at 25 2 C.
[00183] As a general rule, NMR spectra and / or mass spectra were obtained from the prepared compounds. Unless otherwise stated, all chromatographic operations were performed at room temperature. During the synthesis of cyclic dinucleotides, the evaporation of solvents was typically carried out by rotary evaporation under reduced pressure with water bath temperatures not exceeding 35 ° C. In addition, during the synthesis of cyclic dinucleotides, the reactions were carried out under nitrogen or argon.
[00184] Nuclear magnetic resonance (NMR) spectra: for 3 H spectra, chemical shifts
Petition 870190091404, of 9/13/2019, p. 72/105
66/98 were referenced to the solvent signal DMSO (2.50 ppm) or, for measurements in D2O, to DSS (4,4-dimethyl-4silapentane-l-sulfonic acid). The 31 P NMR spectra were indirectly referenced by comparing the frequencies
absolute of 3 H / 31 P (Bruker BioSpin GmbH, Software: TopSpin, au program: xsi). All 31 P NMR spectra were recorded with proton decoupling.
List of abbreviations:
ACN acetonitrile aq. aqueous ° C Celsius degree GIVES Diode array DBU diazabicycle [5.4.0] undec-7-eno Zip code (2-cyanoethyl) - (N, N-diisopropyl)] -phosphoramidite DCM dichloromethane DDTT 3 - ((N, N-dimethylaminomethylidene) amino) -3H-1,2,4-dithiazole-3-thione DIPEA diisopropylethylamine DMAP 4-dimethylaminopyridine DMF N, N-dimethylformamide DMOCP 2-chloro-5,5-dimethyl-2-oxo-1,2,2-dioxafosforinano DMT 4,4'-dimethoxytrityl ESI-MS Electrospray ionization mass spectrometry EtOAc ethyl acetate eq equivalent FC flash chromatography, SiO2 is used if no other details are given
Petition 870190091404, of 9/13/2019, p. 73/105
67/98
H hour HC1 Hydrochloric acid HATU [dimethylamino- (1,2,3-triazolo [4,5-b] pyridin-3-yloxy) -methylene] -dimethylammonium HPLC high performance liquid chromatography L liter LiHMDS lithium hexamethyldisilazide m / z mass: charge ratio MeOH methanol min minute mL milliliter MS mass spectrum n. d. not determined NH 4 0H NH3 solution in water Pd-PEPPSI-IPent ™ dichloro [1,3-bis (2,6-di-3-
pentylphenyl) imidazol-2-ylidene] (3-chloropyridyl) palladium (II) psi Pound per square inch OK room temperature (about 20 ° C) WITHOUT 2- (trimethylsilyl) ethoxymethyl Sun solvent TBS tert-butyl-dimethylsilyl TEA triethylamine TEAF triethylammonium formate
TF / TFA trifluoroacetic acid
TFAA trifluoroacetic acid anhydride THF tetrahydrofuran t R retention time in minutesAnalytical HPLC settings:
Petition 870190091404, of 9/13/2019, p. 74/105
68/98
Configuration A (gradient HPLC):
[00185] VWR / Hitachi: L-2130 pump; VWR / Hitachi:
automatic sampler L-2200; VWR / Hitachi: L-2350 column oven (adjusted to 30 ° C); VWR / Hitachi: L-2400 variable UV / Vis wavelength detector; EZChrom software version 3.3.1 SP1.
[00186] YMC * ODS-A GEL 12 nm (10 pm; 250 x 0.4 mm)) channel A = 20 mM TEAF (pH 6.8) in water; channel B = 100% acetonitrile, 20 mM TEAF (pH 6.8). Gradient: 0 min at 100% A; 30 min at 100% B; 40 min 100% B, 30 C 2; flow rate: 1.0 mL / min; UV 261 nm;
Configuration B (isocratic HPLC):
[00187] VWR / Hitachi: L-7100 pump; VWR / Hitachi:
L-7400 variable UV / Vis wavelength detector; VWR / Hitachi: D-7500 integrator.
[00188] Analytical HPLC (configuration C; YMC * GEL ODS-A 12 nm (10 pm; 250 x 0.4 mm)) 11% acetonitrile, 20 mM TEAF (pH 6.8) in water; flow rate: 1.0 mL / min; UV 264 nm;
LC-MS analysis:
[00189] HPLC system: VWR / Hitachi: pump L-2130; VWR / Hitachi: automatic sampler L-2200; VWR / Hitachi: L-2300 column oven; VWR / Hitachi: L-2450 diode array detector; Agilent: OpenLab [00190] MS System: Bruker Esquire LC 6000 spectrometer
System A
Column: Kromasil 100-5 Cs, 5 pm, 50 mm x 3 mm.
Flow rate: 0.4 mL / min, 35 ° C, UV detection range: 220-300 nm
Mass spectrum: recorded on a mass spectrometer
Petition 870190091404, of 9/13/2019, p. 75/105
69/98 using negative and positive electrospray ionization Solvents: A: acetonitrile
B: water
C: 20 mM NH4HCO3 (pH 7) in water
Gradient:
Time% of A% of B% of C
20 755
9505
9505
20 755
20 755 [00191] Sample preparation: the samples (2 μΐ - 10 μΐ) were dissolved in 175 μΐ of acetonitrile and 175 μΐ of water, with an injection volume of 2 μΐ to 10 μΐ.
System B
Column: ACE 3 AQ 110-3 Cis, 5 pm, 50 mm x 3 mm.
Flow rate: 0.4 mL / min, 35 ° C, UV detection range: 220-300 nm
Mass spectrum: recorded on a mass spectrometer using negative and positive electrospray ionization
Solvents: A: acetonitrile B: waterC: NH4HCO3 20 mM (pH 7) in Water Gradient: Time % of A % of B % in Ç 0 2 93 520 60 35 523 95 0 524 2 93 530 2 93 5
Petition 870190091404, of 9/13/2019, p. 76/105
70/98
Supplierdevice: Agilent Description: Agilent 1200 with DA and MS detector Method name: SYSTEM C Column: Sunfire, 3 x 30 mm, 2.5 pm Column supplier: Waters Gradient / solvent Time [min] % Sol [H 2 O, 0.1% TFA] % Sun[acetonitrile] Flow rate(ml / min) Temp [ 2 C] 0.00 97 3 2.2 60 0.20 97 3 2.2 60 1.20 0 100 2.2 60 1.25 0 100 3 60 1.40 0 100 3 60
Device vendor: Agilent Description : Agilent 1200 with DA and MS detector Method name: SYSTEM D Column: XBridge C18, 3 x 30 mm, 2.5 pm Column supplier: Waters Gradient / solventTime [min] % Sun[H2O, NH30.1%] % Sun[acetonitrile] Flow rate(ml / min) Temp [ 2 C] 0, 00 97 3 2.2 60 0.20 97 3 2.2 60 1.20 0 100 2.2 60 1.25 0 100 3 60 1.40 0 100 3 60
Agilent Supplier
Petition 870190091404, of 9/13/2019, p. 77/105
71/98
device:Description: Agilent 1200 with DA detector andMS
Method name: System E Column: Stable Bond, 3 x 30 mm, 1.8 pm Column supplier: Agilent Gradient / solvent Time [min] % Sol [H 2 O, 0.1% TFA] % Sun[acetonitrile] Flow rate(ml / min) Temp [ 2 C] 0, 00 97 3 2.2 60 0.20 97 3 2.2 60 1.20 0 100 2.2 60 1.25 0 100 3 60 1.40 0 100 3 60
Supplierdevice: Agilent Description: Agilent 1200 with DA detector andMS Method name: System F Column: Nucleodur SiOH, 3 x 30 mm, 5.0 pm Column supplier: Macherey-Nagel Gradient / solventTime [min] % DCM Sun % Sun fromMeOH Flow rate(ml / min) Temp [ 2 C] 0, 00 100 0 2.5 30 0.60 100 0 2.5 30 1.80 95 5 2.5 30 2.60 75 25 2.5 30 3.00 50 50 2.5 30 3, 10 0 100 2.5 30 3.50 0 100 2.5 30
Petition 870190091404, of 9/13/2019, p. 78/105
72/98
3.55 100 0 2.5 30 3.80 100 0 2.5 30 5.00 100 0 3.0 30 5.20 100 0 3.0 30
SUMMARY OF INTERMEDIARIES
INTERMEDIATE 1.1
Imidazopyridazinon-p-D-ribofuranoside d- (β-ρribofuranosyl) imidazo [4,5-d] pyridazin-4 (5H) -one)
[00192] The title compound was prepared as described in J. Chem. Soc. Perkin Trans. 1989, 1769-1774.
INTERMEDIATE 1.2
[00193] Imidazopyridazinon- [3-D-ribofuranoside (INTERMEDIARY 1.1, 4.00 g, 14.9 mmol) was azeotroped with anhydrous pyridine (3 x 20 mL), dried under vacuum and dissolved in anhydrous pyridine (25 mL) . To this solution was added a solution of 4,4'-dimethoxytrityl chloride (5.05 g, 14.9 mmol) in anhydrous pyridine (15 ml) and the reaction mixture was stirred for 1 h at room temperature. The reaction mixture was evaporated under reduced pressure and the resulting residue was purified by means of preparative reverse phase HPLC (X-Bridge C18, acetonitrile / water / NHs).
Petition 870190091404, of 9/13/2019, p. 79/105
73/98
LC-MS (system D):
t Re t = 0.82 min; ESI-MS: 571 [M + H] +
INTERMEDIATE 1.3 ~ a and INTERMEDIATE 1.3 ~ b
5'-DMT-2'-TBS-imidazopyridazinon-p-D-ribofuranoside (INTERMEDIATE 1.3-a) and 5'-DMT-3'-TBS-imidazopyridazinon-β-
INTERMEDIATE 1.3 ~ a
INTERMEDIATE 1.3 ~ b [00194] The 5 '-DMT-imidazopyridazinon- [3-Dribofuranoside (INTERMEDIATE 1.2, 5.30 g, 9.29 mmol) was azeotroped with anhydrous pyridine (3 x 30 mL), dried under vacuum and dissolved in anhydrous pyridine (20 ml). To this solution, imidazole (1.90 g, 27.9 mmol) and tert-butylchlorodimethylsilane (1.54 g, 10.2 mmol) were added and the reaction mixture was stirred for 6 h at room temperature. The reaction mixture was partitioned between dichloromethane and water. The organic layer was separated and the aqueous layer was extracted with dichloromethane. The combined organic extracts were washed with brine, dried on hydrophobic frit and evaporated under reduced pressure. The resulting residue was purified by medium pressure column chromatography (silica gel, gradient of 5 to 20% acetone in dichloromethane).
LC-MS (F system):
INTERMEDIATE 1.3-a: t Ret = 1.57 min; ESI-MS: 685 [M + H] +
Petition 870190091404, of 9/13/2019, p. 80/105
74/98
INTERMEDIATE 1.3-b: t Re t = 1.67 min; ESI-MS: 685 [M + H] +
INTERMEDIATE 1.4
[00195] 5 '-DMT-3' -TBS-imidazopyridazinon- [3-Dribofuranoside (INTERMEDIATE 1.3-b, 1.75 g, 2.56 mmol) was azeotroped with anhydrous acetonitrile (3 x 25 mL), dried in vacuo and dissolved in anhydrous dichloromethane (45 ml). To this solution were added 2-cyanoethyl Ν, Ν, Ν ', Ν'tetraisopropylphosphorodiamidite (1.62 mL, 5.12 mmol) and tetrazole (5.69 mL of a 0.5 M acetonitrile solution, 2.85 mmol ) and the reaction mixture was stirred at room temperature for 4 h. The reaction mixture was diluted with dichloromethane and washed with aqueous sodium hydrogen carbonate solution. The organic layer was separated and the aqueous layer was extracted with dichloromethane. The combined organic extracts were dried using a hydrophobic frit and evaporated under reduced pressure. The resulting residue was purified by medium pressure column chromatography (silica gel (deactivated with triethylamine in dichloromethane), gradient from 20 to 100% ethyl acetate (3% triethylamine) in cyclohexane). The product was obtained as a mixture of diastereoisomers.
LC-MS (system D):
tnet = 1.33 min;
ESI-MS: 885 [M + H] +
Petition 870190091404, of 9/13/2019, p. 81/105
75/98
31 P NMR (162 MHz, 303 K, CDCl 3 ) δ 150.9 and 149.7 ppm.
INTERMEDIATE 1.5 '-OH-2' -TBS-3 '-Hf phosphonate-N 6 -Bz-adenosine / = N .NHBz
HO
The HOH Z [00196] N 6 -Bz-5'-DMT-2'-TBS-3'-CEP-adenosine (obtained from ChemGenes, 0.890 g, 0.90 mmol) was dissolved in acetonitrile (15 mL) and water (0.033 mL, 1.83 mmol, 2 eq.) At room temperature. Pyridinium trifluoracetate (0.210 g, 1.09 mmol, 1.2 eq.) Was added and the reaction mixture was stirred at room temperature for 10 minutes. Terc-butylamine (10 ml, 95.7 mmol) was added and the reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was evaporated under reduced pressure, redissolved in anhydrous acetonitrile (25 ml) and evaporated under reduced pressure to produce a white to colorless foam. The residue was dissolved in dichloromethane (25 ml) and water (0.162 ml, 9 mmol, 10 eq.). Dichloroacetic acid (0.670 ml, 8.12 mmol, 9 eq.) In dichloromethane (25 ml) was added, and the resulting orange solution was stirred at room temperature for 10 minutes. Pyridine (1.31 ml, 16.23 mmol, 18 eq.) Was added and the reaction mixture was stirred at room temperature for 5 minutes.
[00197] The LC-MS analysis of the raw material confirmed the presence of INTERMEDIARY 1.5.
LC-MS (system A):
tnet = 3.10 min;
ESI-MS:
550 [M + H]
Petition 870190091404, of 9/13/2019, p. 82/105
76/98 [00198] The bottle was capped, carefully sealed and stored at + 2 ° C for 16 hours. The solvents were evaporated under reduced pressure and the residue was azeotroped with anhydrous acetonitrile (4 x 15 ml). During the last evaporation procedure, the solution was concentrated to approximately 5 ml of the final azeotrope. The anhydrous solution resulting from intermediate 1.5 was used immediately in the next sequence of reactions. INTERMEDIATE 1.6
______ linear______5 '-OH-3' -TBS-imidazopyridazinon-β-Ρribofuranoside (2 5 ') - cyanoethylphosphorothioate-2'-TBS-3'H-phosphonate-N 6 -Bz-adenosine dimer
HN
HO HO '
NHBz [00199] 5'-DMT-3'-TBS-2'-CEP-imidazopyrideζίηοη-β-Dribofuranoside (INTERMEDIATE 1.4, 1.340 g, 1.51 mmol, 1.7 eq.) Was azeotroped with anhydrous acetonitrile (4x10 mL ). During the last evaporation procedure, the solution was concentrated to approximately 3 ml of the final azeotrope. The resulting solution was added to 5'-OH-2'-TBS-3'-Hphosphonate-N 6 -Bz-adenosine (INTERMEDIATE 1.5) and dissolved in approximately 5 ml of anhydrous acetonitrile (theoretical amount of material: 0.495 g, 0.90 mmol) at room temperature. The reaction mixture was stirred at room temperature for 30 minutes. ((N, Ndimethylaminomethylidene) amino) -3 / -1, 2,4-dithiazoline-3-thione (DDTT) (0.203 g, 0.99 mmol, 1.1 eq.) Was added and the reaction mixture was stirred at room temperature
Petition 870190091404, of 9/13/2019, p. 83/105
77/98 for 30 minutes. The volatile compounds were evaporated under reduced pressure and the residue was dissolved in dichloromethane (25 ml) and water (0.162 ml, 9 mmol, 10 eq.). Dichloroacetic acid (1.340 ml, 16.24 mmol, 18 eq.) In dichloromethane (25 ml) was added, and the orange solution was stirred at room temperature for 20 minutes. Pyridine (10 ml) was added and the reaction mixture was stirred at room temperature for 5 minutes.
[00200] The LC-MS analysis of the raw material confirmed the presence of INTERMEDIARY 1.6 as a mixture of diastereoisomers.
LC-MS (system A):
INTERMEDIATE 1.6-a: t Re t = 7.36 min; INTERMEDIATE 1.6b: tnet = 7.57 min;
ESI-MS: 1063 [M + H] + for each diastereoisomer.
[00201] The bottle was capped, carefully sealed and stored at 2 ° C for 16 hours. The mixture was evaporated under reduced pressure and the residue was coevaporated with anhydrous pyridine (2 x 20 ml) under reduced pressure. An additional 40 ml portion of anhydrous pyridine was added and the residue was concentrated under reduced pressure to approximately 20 ml of the total volume. The anhydrous solution resulting from INTERMEDIATE 1.6 was used immediately in the next sequence of reactions.
INTERMEDIATE 1.7
_____ cyclic ____________3 '-TBS-imidazopyridazinon-β-Ρribofuranoside- (2'> 5 ') -cynoethyl-phosphorothioate-2'-TBS-N 6 Bz-adenosine- (3'> 5 ') - phosphorothioate dimer
Petition 870190091404, of 9/13/2019, p. 84/105
78/98
SO (CH 2 ) 2 CN o> - °
NHBz
TBS
HN
N = ·
I / OTBS ° <A 0 Á SH [00202] 2-oxide
2-chloro-5,5-dimethyl-1,3,2dioxaphosphorinane (DMOCP) (0.581 g, 3.15 mmo1, 3.5 eq.) '-OH-3' -TBS-imidazopyridazinon- [3-D ribofuranoid- (2 '^ 5') - CE-PS-2'-TBS-3'-H-phosphonate-N 6 -Bz crude adenosine (INTERMEDIATE 1.6) (theoretical amount of the desired material in the crude preparation: 0.957 g, 0.90 mmol) in anhydrous pyridine in a total volume of approximately 20 ml. The resulting mixture was stirred at room temperature for 20 minutes. Water (0.570 mL, 31.6 mmol, 35.1 eq.) And 3H-1,2-benzodithiol-3-one (0.230 g, 1.37 mmol, 1.5 eq.) Were added and stirring continued at room temperature. After 20 minutes, the reaction mixture was poured into a solution of sodium hydrogen carbonate (4,500 g, 53.6 mmol) in 150 ml of water and was stirred at room temperature for 5 minutes, followed by the addition of a mixture of ethyl acetate / methyl tert-butyl ether (150 ml, 1: 1). The organic phase was separated and the aqueous phase was further extracted twice with ethyl acetate / methyl tert-butyl ether (2 x 75 ml, 1: 1). The combined organic phases were dried over anhydrous magnesium sulfate, followed by evaporation of solvents under reduced pressure and a final coevaporation with 100 ml of anhydrous toluene. The raw material was purified by preparative flash chromatography (160 g of silica gel, gradient from 0 to 12.5% MeOH in dichloromethane) to produce 0.78 g of INTERMEDIATE 1.7
Petition 870190091404, of 9/13/2019, p. 85/105
79/98 enriched in purity (mixture of diastereoisomers).
LC-MS (system A):
INTERMEDIATE 1.7-a: t Re t = 7.56 min; INTERMEDIATE 1.7b: tRet = 8.18 min;
INTERMEDIATE 1.7-c: t Re t = 9.02 min; INTERMEDIATE 1.7d: t Re t = 10.17 min;
ESI-MS: 1077 [M + H] + for each diastereoisomer.
INTERMEDIATE 1.8
_____ cyclic _____ _______ 3 '-TBS-imidazopyridazinon-β-ibribofuranoside- (2' ^ 5 ') - phosphorothioate-2'-TBS-adenosine- (3'
absolute ethanol to 3'-TBS-imidazopyridazinon-p-Dribofuranoside- (2 '^ 5') - cyanoethyl-phosphorothioate-2'-TBS-N 6 Bz-adenosine- (3 '- ^ 5') - phosphorothioate enriched in purity (INTERMEDIATE 1.7; 0.780 g) and the resulting solution was stirred at room temperature for 4 hours. All volatiles were evaporated under reduced pressure and further dried under vacuum, yielding 0.758 g of crude INTERMEDIATE 1.8 (mixture of diastereoisomers), which was directly used in the next reaction.
LC-MS (system A):
INTERMEDIATE 1.8-a: t Re t = 1.47 min; INTERMEDIATE 1.8b: t Re t = 3.6 0 min;
INTERMEDIATE 1.8-c: t Re t = 3.90 min; INTERMEDIATE 1.8 d: t Re t = 5.53 min;
Petition 870190091404, of 9/13/2019, p. 86/105
80/98
ESI-MS: 920 [M + H] + for each diastereoisomer.
INTERMEDIATE 2.1 '-OH-2' -F-3 '-H-phosphonate-N 6 -Bz-2' -deoxyadenosine
[0 02 04] N 6 -benzoyl-5'-DMT-2'-F-2'-deoxyadenosine-3'CEP (obtained from Alfa Aesar) (1.05 g, 1.20 mmol) was dissolved in acetonitrile ( 6 mL) and water (0.043 mL, 2.40 mmol, 2 eq) at room temperature. Pyridinium trifluoracetate (278 mg, 1.44 mmol, 1.2 eq.) Was added and the reaction mixture was stirred at room temperature for 5 minutes. Then, tert-butylamine (6.0 mL, 57.1 mmol) was added and the reaction mixture was stirred at room temperature for 15 minutes. The reaction mixture was evaporated in vacuo, redissolved (2x) in anhydrous acetonitrile (12 ml) and again evaporated under vacuum to produce a white to colorless foam. The residue was dissolved in dichloromethane (14.4 ml) and water (0.22 ml, 12.0 mmol, 10 eq.). Dichloroacetic acid in dichloromethane (6%, 14.4 ml) was added, and the resulting orange solution was stirred at room temperature for 10 minutes. Pyridine (1.64 ml, 20.3 mmol, 17 eq.) Was added and the reaction mixture was evaporated in vacuo and azeotroped with anhydrous acetonitrile (3 x 11 ml). Finally, the remaining crude product was dried under high vacuum for an additional 30 min and used without further purification.
[00205] The LC-MS analysis of the raw material confirmed the presence of INTERMEDIARY 2.1.
LC-MS (system E):
Petition 870190091404, of 9/13/2019, p. 87/105
81/98 t Re t = 0.64 min ESI-MS: 438 [M + H] + .
INTERMEDIATE 2.2
Dimero______linear______5 '-OH-3' -TBS-imidazopyridazinon-β-Ρribofuranoside (2 '^ 5') - cyanoethylphosphorothioate-2'-F-3'-Hphosphonate-N 6 -Bz-2 '-deoxyadenosine
[00206] 5 '-DMT-3' - TBS-2 '-CEP-imidazopyridazinon- [3-Dribofuranoside (INTERMEDIATE 1.4, 1.38 g, 1.56 mmol, 1.0 eq.) Was dissolved in anhydrous acetonitrile ( 10 mL) and was azeotropically evaporated in vacuo. This operation was repeated three more times, leaving about 5 ml of azeotrope in the flask in the last evaporation. 10 pieces of molecular sieve (3 Â) were added and the resulting mixture was added to 5'-OH-2'-F-3'-H-phosphonate-N 6 Bz-2'-deoxyadenosine (INTERMEDIATE 2.1) dissolved in approximately 3 mL of anhydrous acetonitrile (theoretical amount of the desired material: 523 mg, 1.20 mmol) at room temperature. The reaction mixture was stirred at room temperature for 5 minutes. ((N, N-dimethylaminomethylidene) amino) -3 / 1-1,2,4-dithiazoline-3-thione (DDTT) (275 mg, 1.34 mmol, 0.9 eq.) Was added and the reaction mixture it was stirred at room temperature for 30 minutes. The volatile compounds were evaporated in vacuo and
Petition 870190091404, of 9/13/2019, p. 88/105
82/98 the residue was dissolved in dichloromethane (20 ml) and water (0.216 ml, 12 mmol, 7.7 eq.). Dichloroacetic acid in dichloromethane (6%, 19.2 ml) was added, and the resulting orange solution was stirred at room temperature for 10 minutes. After this period, pyridine (12 ml) was added and the reaction mixture was evaporated in vacuo. Finally, the remaining crude product was dried under high vacuum for an additional 30 min and used without further purification and characterization.
INTERMEDIATE 2.3
_____ cyclic ____ de_____3 '-TBS-imidazopyridazinon-β-Ρ ribofuranoside- (2' ^ 5 ') - cyanoethyl-phosphorothioate-2'-FN 6 -Bz2'-deoxyadenosine- (3' ^ 5 ') dimer - phosphorothioate
[00207] The crude intermediate 2.2 (maximum theoretical amount of desired material: 1.48 g, 1.56 mmol) was dissolved in 36 ml of anhydrous pyridine and reduced to approximately 20 ml under vacuum. 2-chloro-5,5-dimethyl-1,3,2-dioxaphosphorinane 2-oxide (DMOCP) (775 mg, 4.20 mmol, 2.7 eq.) Was added, and the resulting mixture was stirred at room temperature for 5 minutes. Water (0.75 mL, 41.3 mmol, 26.5 eq.) And 3H-1,2-benzodithiol-3-one (0.302 g, 1.80 mmol, 1.15 eq.) Were added and stirring continued at room temperature. After 5 minutes, the mixture
Petition 870190091404, of 9/13/2019, p. 89/105
Reaction 83/98 was poured into a solution of sodium hydrogen carbonate (4.00 g, 47.6 mmol) in 140 ml of water and was stirred at room temperature for 5 minutes, followed by the addition of a mixture of ethyl acetate / methyl tert-butyl ether (140 ml, 1: 1). The organic phase was separated and the aqueous phase was further extracted with ethyl acetate / methyl tert-butyl ether (1: 1). The organic phases were combined and the solvent was removed in vacuo.
[00208] The remaining residue was dissolved in a minimum volume of dichloromethane and purified by preparative flash chromatography (silica gel, DCM / MeOH: 100/0 -½ 80/20). The fractions were analyzed by HPLC-MS. The product containing fractions were combined and the solvent was removed under vacuum to produce 900 mg of a mixture of diastereoisomers.
[00209] The LC-MS analysis of the material confirmed the presence of INTERMEDIARIES 2.3-a / b / c / d.
LC-MS (system E):
INTERMEDIATE 2.3-a: t Re t = 0.95 min; INTERMEDIATE 2.3b: tnet = 0.98 min;
INTERMEDIATE 2.3-c: t Re t = 1.00 min; INTERMEDIATE 2.3d: t Re t = 1.03 min;
ESI-MS: 965 [M + H] + for each diastereoisomer.
INTERMEDIATE 2.4
_____ cyclic ____________3 '-TBS-imidazopyridazinon-β-Ρribofuranoside- (2________' -> _______ 5 ') -phosphorothioate-2' -F-2 'deoxyadenosine- (3' 5 ') - phosphorothioate dimer
Petition 870190091404, of 9/13/2019, p. 90/105
84/98
[00210] To 10 mL of methanol and 10 mL of aqueous ammonia solution (30 - 33%) were added 300 mg (maximum theoretical amount of desired material: 0.31 mmol) of INTERMEDIATE 2.3. The resulting mixture was stirred at 50 2 C for 15 h. The reaction mixture was cooled to room temperature and nitrogen was bubbled through the mixture for 30 min. The solvent was removed in vacuo, the mixture was redissolved in anhydrous acetonitrile (30 ml) and evaporated again in vacuo. The residue was triturated with anhydrous acetonitrile, filtered, washed with 5 ml of ACN and dried overnight at room temperature. The crude product was dissolved in DMF and purified by preparative HPLC (X-Bridge C-18; acetonitrile / HgO / NHs). Fractions containing the product were collected and the solvent was removed by lyophilization. By this method, all four diastereoisomers could be separated.
[00211] The LC-MS analysis of the material confirmed the presence of INTERMEDIARIES 2.3-a / b / c / d.
LC-MS (system C):
INTERMEDIATE 2.4-a: t Re t = 1.04 min; INTERMEDIATE 2.4b: t Re t = 1.10 min;
INTERMEDIATE 2.4-c: t Re t = 1.13 min; INTERMEDIATE 2.4d: t Re t = 1.15 min;
Petition 870190091404, of 9/13/2019, p. 91/105
85/98
ESI-MS: 808 [M + H] + for each diasteroisomer.
INTERMEDIATE 3.1 '-OH-3' -H-phosphonate-N 6 -Bz-LNA-adenine
[00212] INTERMEDIARY 3.1 was prepared in a similar way to INTERMEDIARY 2.1 using LNA-A amidite (EQ0063-1000, obtained from Exiqon) as starting material.
[00213] The LC-MS analysis of the raw material confirmed the presence of INTERMEDIARY 3.1.
LC-MS (system E):
t Re t = 0.63 min; ESI-MS: 448 [M + H] +
INTERMEDIATE 3.2
Dimero______linear______5 '-OH-3' -TBS-imidazopyridazinon-β-Ρribofuranoside- (2 '^ 5') - cyanoethylphosphorothioate-3'-Hphosphonate-N 6 -Bz-LNA-adenine
[00214] INTERMEDIARY 3.2 was prepared in a similar way to INTERMEDIATE 2.2 using INTERMEDIATE 3.1 and INTERMEDIARY 1.4 as starting materials.
INTERMEDIATE 3.3
Dimero_____ciclico____de_____3 '-TBS-imidazopyridazinon-β-Ρribofuranoside- (2' ^ 5 ') - cyanoethylphosphorothioate- N 6 -Bz-LNA
Petition 870190091404, of 9/13/2019, p. 92/105
86/98 adenine (3 '^ 5') - phosphorothioate
[00215] INTERMEDIATE 3.3 was prepared in a similar way to INTERMEDIATE 2.3 using INTERMEDIATE 3.2 as starting material.
[00216] The LC-MS analysis of the material confirmed the presence of INTERMEDIARIES 3.3-a / b / c / d.
LC-MS (system E):
INTERMEDIATE 3.3-a: t Re t = 0.94 min; INTERMEDIATE 3.3b: tRet = 0.98 min;
INTERMEDIATE 3.3-c: t Re t = 0.99 min; INTERMEDIATE 3. θά: t Re t = 1.03 min;
ESI-MS: 975 [M + H] + for each diasteroisomer. INTERMEDIATE 3.4
_____ cyclic ____________3 '-TBS-imidazopyridazinon-β-Ρ ribofuranoside- (2' ^ 5 ') - phosphorothioate-LNA-adenine (3'5') - phosphorothioate dimer
Petition 870190091404, of 9/13/2019, p. 93/105
87/98 [00217] Intermediate 3.4 was prepared in a similar way to INTERMEDIATE 2.4 using INTERMEDIATE 3.3 as starting material.
[00218] The LC-MS analysis of the material confirmed the presence of INTERMEDIARIES 3.4-a / b / c / d.
LC-MS (system C):
INTERMEDIATE 3.4-a: t Re t = 0.81 min; INTERMEDIATE 3.4b: tRet = 0.89 min;
INTERMEDIATE 3.4-c: t Re t = 0.91 min; INTERMEDIATE 3.4d: t Re t = 0.99 min;
ESI-MS: 818 [M + H] + for each diasteroisomer.
INTERMEDIATE 4.1
5'-OH-2'-TBS-3'-H-phosphonatopurine-p-D-ribofuranoside
[00219] INTERMEDIATE 4.1 was prepared in a similar way to INTERMEDIATE 1.5 using 5'-DMT-2'-TBS-3'-CEPpurina- [3-D-ribofuranoside as starting material, which can be prepared as described in Fu et al. Biochemistry 1993, 32, 10629 - 10637. Unlike the
INTERMEDIATE 1.5, this intermediate was not stored overnight, but used immediately in the next sequence of reactions.
[00220] The LC-MS analysis of the raw material confirmed the presence of INTERMEDIARY 4.1.
LC-MS (system B):
t Re t = 8.56 min; ESI-MS: 431 [M + H] +
INTERMEDIATE 4.2
______ linear______5 '-OH-3' -TBS-imidazopyridazinon-β-Ρ dimer
Petition 870190091404, of 9/13/2019, p. 94/105
88/98 ribofuranoside- (2 '^ 5') - cyanoethylphosphorothioate-2'-TBD-3'-H phosphonatopurine-pD-ribofuranoside and O (CH 2 ) 2 CN
[00221] INTERMEDIATE 4.2 was prepared in a similar way to INTERMEDIATE 1.6 using INTERMEDIATE 1.4 and
INTERMEDIATE 4.1 described above as starting materials.
[00222] The LC-MS analysis of the raw material confirmed the presence of INTERMEDIATE 4.2 as a mixture of diastereoisomers.
LC-MS (system B):
INTERMEDIATE 4.2-a: t Re t = 14.86 min; INTERMEDIATE 4.2b: tRet = 15.01 min;
ESI-MS: 944 [M + H] + for each diastereoisomer. INTERMEDIATE 4.3
_____ cyclic ____________3 1 -TBS-imidazopyridazinon-β-Ρ ribofuranoside (2 '^ 5 *) - cyanoethylphosphorothioate-2'-TBS-purine
-β-D-ribofuranoside (3 '^ 5') - phosphorothioate and O (CH 2 ) 2 CN
[00223] INTERMEDIATE 4.3 (as a mixture of diastereoisomers) was prepared in a similar way to
INTERMEDIATE 1.7 using INTERMEDIATE 4.2 as starting material.
Petition 870190091404, of 9/13/2019, p. 95/105
89/98
LC-MS (system B):
INTERMEDIATE 4.3-a: t Re t = 15.76 min; INTERMEDIATE 4.3b: tnet = 16.61 min;
INTERMEDIATE 4.3-c: t Re t = 17.68 min; INTERMEDIATE 4.3d: tnet = 19.26 min;
ESI-MS: 958 [M + H] + for each diastereoisomer.
INTERMEDIATE 4.4
____ cyclic ____ de____3 '-TBS-imidazopyridazinon-β-Ρribofuranoside (2' ^ 5 ') - phosphorothioate-2'-TBS-purine-β-Ρribofuranoside (3' ^ 5 ') - phosphorothioate dimer
O,
HN
X
[00224] INTERMEDIATE 4.4 (as a mixture of diastereoisomers) was prepared in a similar way to INTERMEDIATE 1.8 using INTERMEDIATE 4.3 as starting material.
LC-MS (system B):
INTERMEDIATE 4.4-a: t Re t = 10.31 min; INTERMEDIATE 4.4b: tnet = 11.82 min;
INTERMEDIATE 4.4-c: t Re t = 12.26 min; INTERMEDIATE 4.4d: tnet = 14.47 min;
ESI-MS: 905 [M + H] + for each diastereoisomer.
SUMMARY OF COMPOUNDS ACCORDING TO THE PRESENT INVENTION [00225] General observation: The following pairs of compounds are diastereoisomers and differ in relation to the configuration of at least one phosphorus atom, respectively:
EXAMPLE 1.1 and EXAMPLE 1.2;
Petition 870190091404, of 9/13/2019, p. 96/105
90/98
EXAMPLE 2.1 and EXAMPLE 2.2;
EXAMPLE 3.1 and EXAMPLE 3.2;
EXAMPLE 4.1 and EXAMPLE 4.2.
EXAMPLE 1.1 and EXAMPLE 1.2 (Cyclic Imidazopyridazinon-p-D-ribofuranoside- (2'-5 ') phosphorothioate-adenosine- (3' -5 ') - phosphorothioate)
HS s / ° "P / 0-
HN
[00226] 30 ml of anhydrous pyridine and 15 ml of anhydrous triethylamine were added to 3'-TBSimidazopyridazinon- [3-D-ribofuranoside (2 '^ 5') -phosphorothioate
-2'-TBS-adenosine- (3 '^ 5') - crude phosphorothioate (INTERMEDIATE 1.8; 0.76 g). The resulting solution was concentrated under reduced pressure to approximately 5 ml of total volume, followed by the simultaneous addition of triethylamine trihydrofluoride (3.590 ml, 21.7 mmol) and 11 ml of anhydrous triethylamine. This solution was stirred at 50 ° C for 3.5 hours. After cooling to room temperature, the reaction was quenched with methoxytrimethylsilane (10 ml, 72.4 mmol) and then stirred at room temperature to consume any excess HF. After 30 minutes, all volatile components were evaporated under reduced pressure, followed by a final coevaporation with 50 ml of anhydrous toluene under reduced pressure. The residue was further dried in vacuo to produce the crude mixture containing Example 1.1 and Example 1.2.
[00227] 65 ml of water were added and the resulting suspension was placed in an ultrasound bath at
Petition 870190091404, of 9/13/2019, p. 97/105
91/98 room temperature. After 15 minutes, this suspension was poured into 60 ml of chloroform and the organic phase was separated. This extraction was repeated two more times with chloroform. The combined organic phases were extracted with 50 ml of water and the aqueous phase containing the combined product was filtered with a 0.45 pm Rotilabo®CME syringe filter (outside diameter: 33 mm) to remove particulate components. The product solution was diluted with water to 500 ml and applied to an anion exchange column of Q Sepharose ™ Fast Flow (40 to 165 pm; 125 x 35 mm; ~ 120 ml) form O 'previously regenerated with sodium chloride 2 M and washed with water. The column was washed with water (2 column volumes), followed by a gradient of 0 to 1 M triethylammonium bicarbonate buffer (TEAB, pH 7) in 25 column volumes (detection wavelength = 254 nm) . EXAMPLE 1.1 and EXAMPLE 1.2 eluted as a mixture of isomers with TEAB ~ 0.6 M. The product-containing fractions were carefully concentrated to a final volume of approximately 10 ml under reduced pressure.
[00228] The separation of EXAMPLE 1.1 (fourth elution) and EXAMPLE 1.2 (third elution) was carried out by repeated purifications by semi-prepared reverse phase HPLC. The product solution was applied to a YMC * GEL ODS-A 12 nm (10 pm; 250 x 16 mm; ~ 50 ml) column, previously equilibrated with 4% acetonitrile, 20 mM triethylammonium formate (LEAF, pH 6, 8) in water. Elution was performed with a 4%, 6% and 20% acetonitrile gradient in stages, in 20 mM TEAF (pH 6.8) in water.
[00229] Preparation of EXAMPLE 1.1, sodium salt (fourth diastereoisomer to elute).
Petition 870190091404, of 9/13/2019, p. 98/105
92/98 [00230] The desalination of the TEA salt from EXAMPLE 1.1 was performed by liquid chromatography under preparative reverse phase medium pressure (MPLC). The product solution (~ 40 mL) was applied to a Merck LiChroprep®RP-l8 column (15 - 25 pm; 450 x 25 mm; ~ 220 mL), previously equilibrated with water. The column was washed with water to remove excess TEAF buffer. Thereafter, 2% propanol 2% in water was used to elute the desalted EXAMPLE 1.1. The product-containing fractions were partially concentrated under reduced pressure and subsequently applied to a SP Sepharose ™ Fast Flow cation exchange column (45 - 165 pm; 125 x 35 mm; ~ 120 mL) Na + form, previously regenerated with sodium chloride 2 M and washed with water. The column was washed with water until no more UV absorbance was detected (detection wavelength 254 nm). The product-containing fractions were carefully evaporated under reduced pressure and were further dried under vacuum to produce EXAMPLE 1.1 as disodium salt.
HPLC (configuration A): t Re t =
8.56 min;
ESI-MS: 692 [M + H]
3 H NMR (400 MHz
318 K, 500 pL (CD 3 ) 2 SO + 30 pL D 2 O) δ 8.70
2H)
1H)
1H), 8.63 (s
8.4 Hz, 1H)
4.95 (dd
4.33
4.21
1H), 8.34
5.91 (d
7.5, 4.5
1H), 8.16 (s, 1H), 6.07
7.5 Hz
Hz, 1H) (m, 3H), 4.05 - 3.93 (d
1H)
4.44
5.07 - 4.99 (d
1H)
4.4
Hz
3.92 - 3.77
2H) ppm.
31 P NMR (162
MHz
D 2 O): δ 52.4 (s, IP)
55.3 (s
IP) ppm.
Preparation of
EXAMPLE 1.2, eluting diastereoisomeric sodium salt)
Petition 870190091404, of 9/13/2019, p. 99/105
93/98 [00231] The desalination and the exchange of TEA for sodium of EXAMPLE 1.2, the salt of TEA was carried out in a manner similar to that described for EXAMPLE 1.1, salt of TEA.
HPLC (configuration A): t Ret = 7.75 min;
ESI- MS: 692 [M + H] + NMR (400 MHz, 318 K, 500 pL (CD3) 2SO + 30 pL D2O) δ 8.72 (s, 1H), 8.59 (s, 1H), 8.33 (s, 1H), 8, 15 (s, 1H), 6.04 (d, J = 8.4 Hz, 1H), 5.88 (d, J = 8.2 Hz, 1H), 5, 25 (dd, J = 8.5, 4.4 Hz, 1H), 5.08 (ddd, J = 11.2 , 8.4, 4 , 5 Hz, 1H), 4.92 (dd, . J = 8.2, 4.3 Hz, 1H), 4.37 - 4.27 (m, 1H), 4, 27 - 4.21 (m, 3H), 4.08 (dd, J = 11.0, 4.2 ' Hz, 1H), 3.82 - 3.77 (m, 1H), 3.67 - 3.60 (m , 1H) ppm. NMR 31p ( 162 MHz, D 2 O): δ 55, 8 (s, IP), 57.0 (s, IP) ppm •
EXAMPLE 2.1 and EXAMPLE 2.2 (imidazopyridazinon-p-D-ribofuranoside- (2 '-5') phosphorothioate-2'-F-2'-deoxyadenosine- (3 '-5') - cyclic phosphorothioate
[00232] 16 mg (19 pmol) of INTERMEDIATE 2,4-d was added to 1 ml of anhydrous pyridine and 4 ml of anhydrous acetonitrile, and the solvent was azeotropically evaporated in vacuo. The residue was resuspended twice in 10 ml of anhydrous acetonitrile and evaporated again azeotropically under vacuum. 80.7 pL (1.0 mmol) of pyridine
Petition 870190091404, of 9/13/2019, p. 100/105
94/98 anhydrous and 168 μΐ (1.2 pmol) TEA were added to the residue before 103 pL (0.63 mmol) TEA * 3HF was carefully added via syringe. The resulting mixture was stirred at 50 2 C for 90 min. After cooling to room temperature, the reaction was quenched by adding 20 ml of an aqueous solution of triethylammonium carbonate (cone. = 1 mol / L). The resulting mixture was stirred for an additional 15 min at room temperature. The mixture was carefully loaded into a Sep Pak C18 © cartridge from Water (5 g of material C18, preconditioned with the first 25 ml of acetonitrile and then with 25 ml of water) and washed with 60 ml of water. Then, the product was eluted from the cartridge using 100 mL of a mixture of acetonitrile / triethylammonium acetate / water (prepared by adding 1 mL of an aqueous solution of triethylammonium acetate (cone = 1 mol / L) to 100 mL of water and 25 mL of acetonitrile). Fractions containing the product were combined and the solvent was removed by lyophilization. The resulting product was further purified by HPLC-prep (Atlantis C18; 20 mM aq. NH4OAc / acetonitrile = 98/2 80/20). After the solvent was removed by lyophilization, the product was dissolved in 2 ml of water and poured into a spin Bio-Rad column (filled with 250 mg of BT AG 50W-2 resin, 100-200 mesh hydrogen form, conditioned with 3 ml of 1 M aqueous NaOH solution and then washed with 6 ml of water resulting in pH ~ 7)) and eluted with 12 ml of water. Fractions containing the product were combined and the solvent was removed by lyophilization.
EXAMPLE 2.1
Petition 870190091404, of 9/13/2019, p. 101/105
95/98
LC-MS (system C):
tRet = 0.61 min; ESI-MS: 694 [M + H] +
3 H NMR (400 MHz, 303 K, D 2 O) δ ppm 8.73 (s, 1 H), 8.53 (s,
H), 8.24 (s, 1 H), 7.96 (s, 1 H), 6.40 (d, J = 15.9 Hz, 1
H), 6.26 (d, J = 8.6 Hz, 1 H), 5.55 (dd, J = 51, l, 3.8 Hz, 1
H), 4.83 - 4.99 (m, 2 H), 4.75 (d, J = 4.2 Hz, 1 H), 4.65 (ddd, J = 12.2, 9.0 , 2.2 Hz, 1 H), 4.53 - 4.59 (m, 2 H), 4.43 -4.48 (m, 1 H), 4.16 -  4.24 (m, 2 H)31 P NMR (162 MHz, D 2 O): δ 55.7 (s, IP), 52.2 (s, IP) ppm.
EXAMPLE 2.2 [00233] EXAMPLE 2.2 was prepared in a similar way to EXAMPLE 2.1, using INTERMEDIATE 2.4-c as starting material.
LC-MS (system C):
tRet = 0.30 min; ESI-MS: 6 94 [M + H] + 3 H NMR (400 MHz, 303K, D 2 O) δ ppm 8.80 (s, 1 H), 8.55 (s, 1 H), 8.24 (s, 1 H) , 8.24 (s, 1 H) , 6.44 (d, J = 15.0 Hz, 1 H), 6.28 (d, J = 8.5 Hz :, 1 H) , 5.54 _ [ 5.73 (m, 1 H), 4.99-  5, 10 (m, 2 H), 4.64 - 4.70 (m, 1 H) , 4.58 (d, J = 4.4 Hz, 1 H), 4.52 - 4.57 (m, 2 H), 4.37 - 4 .45 (m, 1 H), 4.22 (ddd, J = 12.3, 3.5, 1.1 Hz, 1 H), 4, 08 (ddd, J = ll, 7, 3,6, 1, 7 Hz, 1 H)31 P NMR (162 MHz, D 2 O): δ 55.5 (s, IP), 55.9 (s, IP) ppm.
EXAMPLE 3.1 and EXAMPLE 3.2
(Imidazopyridazinon-p-D-ribofuranoside (2 '-5') -phosphorothioate-LNA-adenine- (3 '-5') -phosphorothioate) sodium salt
Petition 870190091404, of 9/13/2019, p. 102/105
96/98
[00234] EXAMPLES 3.1 and 3.2 were prepared in a similar way to EXAMPLES 2.1 and 2.2, using INTERMEDIATE 3.4-d (for EXAMPLE 3-1) and 3.4-c (for EXAMPLE 3-2) as starting material, respectively. EXAMPLE 3.1
LC-MS (system E):
t Ret = 0.37 min; ESI-MS: 704 [M + H] +
1H NMR (400 MHz, 303 K, D 2 O) δ ppm 8.79 (s, 1 H), 8.53 (s, 1 H), 8.22 (s, 1 H), 7.95 (s , 1 H), 6.26 (d, J = 8.4 Hz, 1
H) , 6, 14 (s, 1 H), 5.08 (s, 1 H), 4.97 (ddd, J = = 9.6, 8.4, 4.4 Hz, 1 H) , 4.81 (d, J = 5.4 Hz, 1 H) , 4.82 - 4 .70 (m, 3 H) , 4.55 (d, J = 2.0 Hz, 1 H), 4.35 (dd, J = ll, 9, 2.7 Hz, 1 H) , 4.25 - 4, 16 (m, 3 H), 4.08 (d, J = 8 , 5 Hz, 1 H) 31 P NMR (162 MHz, D2O) : δ 56 , 0 (s, IP), 53.0 (s, IP) ppm.
EXAMPLE 3.2:
LC-MS (system E):
t Re t = 0.29 min; ESI-MS: 704 [M + H] +
1H NMR (400 MHz, 303 K, D 2 O) δ ppm 8.85 (s, 1 H), 8.56 (s,
H), 8.23 (s, 1 H), 8.15 (s, 1 H), 6.26 (d, J = 8.4 Hz, 1
H) , 6.16 (s, 1 H), 5.20 (s, 1 H), 5 , 00 (ddd, J = 13.0, 8.4, 4.5 Hz, 1 H) , 4.87 (d, J = 5.0 Hz, 1 H) , 4.83 - 4.73 (m, 2 H) , 4.62 (d, J = 4.5 Hz, 1 H), 4.54 - 4 , 52 (m, 1 H), 4.47 (dd, J = ll, 3, 5.0 Hz, 1 H), 4.27 - 4, 20 (m, 2 H), 4.12 (dd,
Petition 870190091404, of 9/13/2019, p. 103/105
97/98
J = 11, 3, 3.1 Hz, 1 H), 4.10 (d, J = 8.5 Hz, 1 H).
31 P NMR (162 MHz, D 2 O): δ 57.3 (s, IP), 55.8 (s, IP) ppm.
EXAMPLE 4.1 AND EXAMPLE 4.2
(Imidazopyridazinon-p-D-ribofuranoside (2 '^ 5') - phosphorothioatopurine-p-D-ribofuranoside- (3'5 ') - phosphorothioate) sodium salt
O, o
„OH sr, o.
OH
O-P = O
SH [00235] EXAMPLES 4.1 and 4.2 were prepared in a similar way to EXAMPLES 1.1 and 1.2 using INTERMEDIATE 4.4-d and INTERMEDIARY 4.4-c as starting material. In addition to the procedure described for EXAMPLES 1.1 and 1.2, before starting the reaction, the starting material was once azeotroped using a 2: 1 mixture of anhydrous pyridine and anhydrous triethylamine.
EXAMPLE 4.1:
HPLC (configuration B): tRet = 10.22 min;
ESI-MS: 677 [M + H] +
3 H NMR (400 MHz, 318 K, 500 pL (CD 3 ) 2 SO + 30 pL D 2 O) δ 9.16 (s, 1H), 8.95 (s, 1H), 8.82 (s, 1H), 8.68 (s, 1H), 8.62 (s, 1H), 6.06 (d, J = 8.3 Hz, 1H), 6.06 (d, J = 7.4 Hz, 1H), 5.11 - 4.98 (m, 3H), 4.44 (d, J = 4.4 Hz, 1H), 4.34 - 4.25 (m, 3H), 4, 08 - 3 , 97 (m, 1H), 3.93 - 3.83 (m, 1H), 3.84 3.77 (m, 1H).
31 P NMR (162 MHz, D 2 O): δ 55.3 (s, IP), 52.6 (s, IP) ppm.
Petition 870190091404, of 9/13/2019, p. 104/105
98/98
EXAMPLE 4.2:
HPLC (configuration B): tRet = 5.50 min;
ESI-MS: 677 [M + H] +
3 H NMR (400 MHz, 318 K, 500 pL (CD 3 ) 2 SO + 30 pL D 2 O) δ 9.16
(s, 1H), 8.94 (s, 1H), 8.82 (s, 1H), 8 72 (s, 1H), 8.59 (s, 1H), 6.04 (d, J = 8.5 Hz, 1H), 6, 03 (d, J = 8.1 Hz, 1H), 5.30 (dd, J = 8.5 , 4.3 Hz, 1H), 5.09 (ddd, J = 11.3, 8.4, 4, 6 Hz, 1H), 5.01 (dd, J = 8.2 , 4.3 Hz, 1H), 4.45 - 4.32
(m, 1H), 4.32 - 4.19 (m, 3H), 4.13 (dd, J = 11.0, 4.1 Hz,
1H), 3.84 - 3.75 (m, 1H), 3.71 - 3.61 (m, 1H).
31 P NMR (162 MHz, D 2 O): δ 55.8 (s, IP), 57.8 (s, IP) ppm.
权利要求:
Claims (3)
[1]
1. Compound CHARACTERIZED by the fact that it has formula I

on what
R 1 is selected from the group consisting of H, F, -0 C1-3alkyl and OH, and
R 2 is H, or
R 2 is -CH2- and R 1 is -0-, forming a -CH2-O- bridge together, and
R 3 is a purine nucleobase selected from the group consisting of purine, adenine, guanine, xanthine, hypoxanthine, linked via its N 9 nitrogen, or a salt thereof.
[2]
2/3 comprise one or more compounds as defined in
any of claims 1 to 4, or one or more of
their pharmaceutically acceptable salts, optionally together with one or more inert vehicles and / or diluents.
6. Method for the treatment of diseases or conditions selected from inflammation, allergic diseases, autoimmune diseases, infectious diseases or cancer, in a patient who needs it, CHARACTERIZED by the fact that he understands to administer to the patient a compound as defined in any of the claims 1 to 4.
7. Vaccine, CHARACTERIZED by the fact that it comprises a compound as defined in any of claims 1 to 4.
8. Use of a compound, as defined in any
one of claims 1 to 4, characterized by the fact that it is as a vaccine adjuvant.
9. Compound, according to any one of claims 1 to 4, CHARACTERIZED by the fact that be for
use as a medicine.
10. Compound, according to any one of claims 1 to 4, CHARACTERIZED by the fact that be for
use in the treatment of selected diseases or conditions of
inflammation, allergic diseases, autoimmune diseases diseases
infectious diseases and cancer.
11. Pharmaceutical composition, CHARACTERIZED by the fact that it comprises one or more compounds as defined in any one of claims 1 to 4, and one or more additional therapeutic agents, optionally together with one or more inert vehicles and / or diluents.
12. Pharmaceutical composition, according to with the
Petition 870190091396, of 9/13/2019, p. 11/10
2. Stereoisomer (Sp, Sp), (Rp, Rp), (Sp, Rp) or (Rp, Sp) substantially pure, CHARACTERIZED by the fact that it is a compound as defined in claim 1, or a salt thereof.
3. Stereoisomer (Rp, Rp) substantially pure, CHARACTERIZED by the fact that it is a compound as defined in claim 1, or a salt thereof.
4. Pharmaceutically acceptable salt, CHARACTERIZED by the fact that it is a compound as defined in any one of claims 1 to 3.
5. Pharmaceutical composition, CHARACTERIZED by the fact that
Petition 870190091396, of 9/13/2019, p. 9/11
[3]
3/3 claim 11, CHARACTERIZED in that it comprises a compound as defined in any one of claims 1 to 4 and one or more additional therapeutic agents.
类似技术:
公开号 | 公开日 | 专利标题
BR112019006512A2|2019-10-01|cyclic dinucleotide compounds
US10392419B2|2019-08-27|Modified cyclic dinucleotide compounds
JP2021098721A|2021-07-01|Substituted quinazoline compounds and methods for use thereof
EP3507367A1|2019-07-10|Locked nucleic acid cyclic dinucleotide compounds and uses thereof
JP2018009017A|2018-01-18|Condensed heterocyclic aromatic pyrrolidinone as syk inhibitor
WO2019185476A1|2019-10-03|Modified cyclic dinucleotide compounds
BR112020008839A2|2020-10-20|modulators of the integrated stress pathway
CA3082351A1|2019-05-16|Sting modulator compounds, and methods of making and using
JP2020535168A|2020-12-03|Salts of pyrorotriazine derivatives useful as TAM inhibitors
EP3774834A1|2021-02-17|Cyclic dinucleotide compounds containing 2-aza-hypoxanthine or 6h-pytazolo[1,5-d][1,2,4]triazin-7-one as sting agonists
BR102017021041A2|2019-04-16|CYCLIC DINUCLEOTIDE COMPOUNDS
同族专利:
公开号 | 公开日
EP3519420A1|2019-08-07|
EP3519420B1|2020-11-11|
US20180092937A1|2018-04-05|
MA46325A|2019-08-07|
EA037030B1|2021-01-28|
CA3038860A1|2018-04-05|
JP2018536695A|2018-12-13|
JOP20190066A1|2017-06-16|
HUE053819T2|2021-07-28|
UA124347C2|2021-09-01|
CO2019003178A2|2019-04-12|
MX2019003586A|2019-06-10|
LT3519420T|2021-01-25|
IL265642D0|2019-05-30|
AR109777A1|2019-01-23|
EA201990785A1|2019-10-31|
AU2017333933A1|2019-03-14|
JP6509445B2|2019-05-08|
TW201829442A|2018-08-16|
MA46325B1|2021-02-26|
ES2848724T3|2021-08-11|
PT3519420T|2021-02-02|
DK3519420T3|2021-01-25|
HRP20210027T1|2021-03-05|
SI3519420T1|2021-03-31|
WO2018060323A1|2018-04-05|
PE20191503A1|2019-10-22|
KR20190057131A|2019-05-27|
IL265642A|2022-01-01|
US10537590B2|2020-01-21|
CL2019000775A1|2019-06-14|
CN109843903A|2019-06-04|
AU2017333933B2|2022-01-06|
PH12019500706A1|2019-11-25|
RS61423B1|2021-03-31|
PL3519420T3|2021-08-16|
PL3519420T4|2021-08-16|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题

US7592326B2|2004-03-15|2009-09-22|Karaolis David K R|Method for stimulating the immune, inflammatory or neuroprotective response|
JP4931798B2|2004-03-15|2012-05-16|ケイ.アール. カラオリス,デイヴィッド|Methods for immune, inflammatory or neuroprotective responses|
CN102203125A|2008-08-25|2011-09-28|安普利穆尼股份有限公司|Pd-1 antagonists and methods of use thereof|
JP2013512251A|2009-11-24|2013-04-11|アンプリミューン、インコーポレーテッド|Simultaneous inhibition of PD-L1 / PD-L2|
EP2591001B1|2010-07-09|2021-11-17|Aduro Biotech Holdings, Europe B.V.|Agonistic antibody to cd27|
KR20150095668A|2012-12-13|2015-08-21|아두로 바이오테크, 인코포레이티드|Compositions comprising cyclic purine dinucleotides having defined stereochemistries and methods for their preparation and use|
PT2934598T|2012-12-19|2018-07-24|Univ Texas|Pharmaceutical targeting of a mammalian cyclic di-nucleotide signaling pathway|
JP2016518140A|2013-05-03|2016-06-23|ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア|Method for inducing cyclic dinucleotide of type I interferon|
SG11201508273RA|2013-05-18|2015-12-30|Aduro Biotech Inc|Compositions and methods for activating "stimulator of interferon gene"-dependent signalling|
WO2015017652A1|2013-07-31|2015-02-05|Memorial Sloan-Kettering Cancer Center|Sting crystals and modulators|
JOP20200096A1|2014-01-31|2017-06-16|Children’S Medical Center Corp|Antibody molecules to tim-3 and uses thereof|
UY36032A|2014-03-14|2015-10-30|Novartis Ag|ANTIBODY MOLECULES THAT JOIN LAG-3 AND USES OF THE SAME|
CA2950033A1|2014-06-04|2015-12-10|Glaxosmithkline Intellectual Property Development Limited|Cyclic di-nucleotides as modulators of sting|
WO2016096174A1|2014-12-16|2016-06-23|Invivogen|Fluorinated cyclic dinucleotides for cytokine induction|
GB201501462D0|2015-01-29|2015-03-18|Glaxosmithkline Ip Dev Ltd|Novel compounds|
WO2016145102A1|2015-03-10|2016-09-15|Aduro Biotech, Inc.|Compositions and methods for activating "stimulator of interferon gene" -dependent signalling|
JOP20200224A1|2015-08-13|2017-06-16|Merck Sharp & Dohme|Cyclic di-nucleotide compounds as sting agonists|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|
WO2018190719A2|2017-04-13|2018-10-18|Aduro Biotech Holdings, Europe B.V.|Anti-sirp alpha antibodies|
WO2019084060A1|2017-10-24|2019-05-02|Silverback Therapeutics, Inc.|Conjugates and methods of use thereof for selective delivery of immune-modulatory agents|
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|
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|
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|
CA3101368A1|2018-05-25|2019-11-28|Incyte Corporation|Tricyclic heterocyclic compounds as sting activators|
US11008344B2|2018-07-31|2021-05-18|Incyte Corporation|Tricyclic heteroaryl compounds as STING activators|
US10875872B2|2018-07-31|2020-12-29|Incyte Corporation|Heteroaryl amide compounds as sting activators|
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|
EP3848054A1|2018-09-06|2021-07-14|Daiichi Sankyo Company, Limited|Novel cyclic dinucleotide derivative and antibody-drug conjugate thereof|
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|
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|
WO2020178770A1|2019-03-07|2020-09-10|Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I.|3'3'-cyclic dinucleotides and prodrugs thereof|
WO2020214663A1|2019-04-17|2020-10-22|Gilead Sciences, Inc.|Solid forms of a toll-like receptor modulator|
EP3956329A1|2019-04-17|2022-02-23|Gilead Sciences, Inc.|Solid forms of a toll-like receptor modulator|
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|
US20210363273A1|2020-05-19|2021-11-25|Boehringer Ingelheim International Gmbh|Binding molecules for the treatment of cancer|
WO2022006327A1|2020-07-01|2022-01-06|Silverback Therapeutics, Inc.|Anti-asgr1 antibody conjugates and uses thereof|
WO2022031894A1|2020-08-07|2022-02-10|Gilead Sciences, Inc.|Prodrugs of phosphonamide nucleotide analogues and their pharmaceutical use|
WO2022032191A1|2020-08-07|2022-02-10|Tambo, Inc.|Trans-cyclooctene bioorthogonal agents and uses in cancer and immunotherapy|
法律状态:
2021-06-08| B15K| Others concerning applications: alteration of classification|Free format text: AS CLASSIFICACOES ANTERIORES ERAM: C07H 21/02 , C07H 19/23 , A61K 31/7076 , A61P 37/00 Ipc: A61K 31/7076 (2006.01), C07H 19/23 (2006.01), C07H |
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
EP16191919|2016-09-30|
PCT/EP2017/074608|WO2018060323A1|2016-09-30|2017-09-28|Cyclic dinucleotide compounds|
[返回顶部]