Neuroactive steroids, compositions and uses thereof

文档序号:349886 发布日期:2021-12-07 浏览:29次 中文

阅读说明:本技术 神经活性类固醇、组合物和其用途 (Neuroactive steroids, compositions and uses thereof ) 是由 G.马丁内斯博特拉 B.L.哈里森 A.J.罗比肖 F.G.萨里图罗 R.J.赫尔 R.B 于 2014-08-22 设计创作,主要内容包括:本文描述了式(I)的神经活性类固醇:或其药学上可接受的盐;其中R~(1)、R~(2)、R~(5)、A和L如本文所定义。在某些实施方案中,这些化合物设想充当GABA调节剂。本发明还提供包含本发明化合物的药物组合物以及使用和治疗方法,例如,用于诱导镇静和/或麻醉。(Described herein are neuroactive steroids of formula (I): or a pharmaceutically acceptable salt thereof; wherein R 1 、R 2 、R 5 A and L are as defined herein. In certain embodiments, these compounds are contemplated to act as GABA modulators. The invention also provides pharmaceutical compositions comprising the compounds of the invention and methods of use and treatment, e.g., for inducing sedation and/or anesthesia.)

1. Use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating depression in a subject, wherein the compound of formula (I) is:

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3)-;

R1Is hydrogen or C1-C6An alkyl group;

R2is hydrogen or C1-C6An alkyl group;

each R3Is hydrogen;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

2. The use of claim 1, wherein R2Is C1-C6A haloalkyl group.

3. The use of claim 1, wherein the compound of formula (I) is a compound of formula (Ia):

or a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3)-;

R1Is hydrogen or C1-C6An alkyl group;

each R3Is hydrogen;

R5Is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

4. The use of claim 3, wherein the compound of formula (Ia) is a compound of formula (Ia-1):

or a pharmaceutically acceptable salt thereof.

5. Use according to claim 3, wherein A is benzotriazole, azabenzotriazole, diazabenzotriazole, benzopyrazole, azabenzopyrazole or diazabenzpyrazole.

6. The use of claim 3, wherein A is a substituted 5-membered nitrogen-containing heteroaryl or heterocyclyl.

7. The use of claim 6, wherein A is a substituted 5-membered nitrogen-containing heteroaryl or heterocyclyl group containing up to four nitrogen atoms.

8. The use of claim 7, wherein a is a substituted 5-membered nitrogen-containing heteroaryl or heterocyclyl group comprising 2,3, or 4 nitrogen atoms.

9. The use of claim 8, wherein a is a substituted pyrazole, triazole or tetrazole.

10. The use of claim 4, wherein the compound of formula (Ia-1) is a compound of formula (Ia-3):

or a pharmaceutically acceptable salt thereof,

wherein

R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcForm a ring together with the nitrogen atom to which it is bound; and is

n is 1,2 or 3.

11. The use of claim 10, wherein n is 1 or 2, and each R is4Independently is C1-C6Alkyl OR-C (O) ORa

12. The use of claim 11, wherein n is 1, and R is4Is methyl.

13. The use of claim 11, wherein RaIs C1-C6An alkyl group.

14. The use of claim 13, wherein RaIs ethyl.

15. The use of claim 10, wherein R1Is hydrogen or C1-C6Alkyl, and R4is-C (O) ORa

16. The use of claim 15, wherein R1Is C1-C6An alkyl group.

17. The use of claim 16, wherein R1Is methyl, ethyl or isopropyl.

18. The use of claim 16, wherein R1Is methyl and R4is-C (O) OEt.

19. The use of claim 10, wherein the compound is selected from the group consisting of:

or a pharmaceutically acceptable salt thereof.

20. Use of a compound of formula (Ib), wherein the compound of formula (Ib) is:

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3)-;

R1Is hydrogen or C1-C6An alkyl group;

R2is C1-C6An alkyl group;

each R3Is hydrogen;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

21. The use of claim 20, wherein R is2Is C1-C6A haloalkyl group.

22. The use of claim 20, wherein the compound of formula (Ib) is a compound of formula (Ib-1):

or a pharmaceutically acceptable salt thereof.

23. The use of claim 20, wherein the compound of formula (Ib) is a compound of formula (Ib-2):

or a pharmaceutically acceptable salt thereof.

24. The use of claim 22, wherein the compound of formula (Ib-1) is a compound of formula (Ib-3)

Or a pharmaceutically acceptable salt thereof,

wherein

R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcForm a ring together with the nitrogen atom to which it is bound; and is

n is 1,2 or 3.

25. The use of claim 23, wherein the compound of formula (Ib-2) is a compound of formula (Ib-4)

Or a pharmaceutically acceptable salt thereof,

wherein:

R4is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcForm a ring together with the nitrogen atom to which it is bound; and is

n is 1,2 or 3.

26. The use of claim 20, wherein a is a substituted 5-or 6-membered nitrogen-containing heteroaryl or heterocyclyl group containing up to four nitrogen atoms.

27. The use of claim 26, wherein a is a substituted 5-or 6-membered nitrogen-containing heteroaryl or heterocyclyl group comprising 1,2,3, or 4 nitrogen atoms.

28. The use of claim 20, wherein a is substituted morpholine or piperazine.

29. The use of claim 20, wherein a is benzotriazole, azabenzotriazole, diazabenzotriazole, benzopyrazole, azabenzopyrazole, or diazabenzpyrazole.

30. The use of claim 20, wherein a is a substituted pyrazole, triazole or tetrazole.

31. The use of claim 20, wherein R1Is C1-C6An alkyl group.

32. The use of claim 31, wherein R1Is methyl, ethyl or isopropyl.

33. The use of claim 32, wherein R1Is methyl.

34. The use of claim 20, wherein R2Is methyl.

35. The use of claim 24, wherein n is 1 or 2, and each R is4Independently of one another is cyano, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Raor-S (O)0-2Ra

36. The use of claim 35, wherein n is 1 or 2, and each R is4Independently is Br, Cl or F.

37. The use of claim 35, wherein n is 1 or 2, and each R is4is-OCH3

38. The use of claim 35, wherein n is 1, and R is4Is cyano.

39. The use of claim 35, wherein n is 1, and R is4Is C1-C6An alkyl group.

40. The use of claim 39, wherein R4Is methyl.

41. The use of claim 35, wherein n is 1 or 2, and each R is4is-C (O) Ra

42. The use of claim 41, wherein each RaIs C1-C6An alkyl group.

43. The use of claim 42, wherein each R isaIs methyl.

44. The use of claim 35, wherein n is 1, and R is4is-S (O)2Ra

45. The use of claim 44, wherein RaIs methyl.

46. The use of claim 24, wherein R1Is C1-C6Alkyl, n is 1, and R4is-C (O) Ra

47. The use of claim 46, wherein R1Is methyl and R4is-C (O) Me.

48. The use of claim 20, wherein R1Is methyl, ethyl or isopropyl.

49. The use of claim 48, wherein R1Is methyl.

50. The use of claim 25, wherein n is 1 or 2, and each R is4Independently is C1-C6Alkyl, -C (O) Raor-S (O)0-2Ra

51. The use of claim 50, wherein n is 1, and R4Is methyl.

52. The use of claim 50, wherein n is 1, and R4is-C (O) Me.

53. The use of claim 50, wherein n is 1, and R4is-S (O)2Me。

54. The use of claim 20, wherein the compound is selected from the group consisting of:

or a pharmaceutically acceptable salt thereof.

55. Use of a compound, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for treating depression in a subject, wherein the compound is selected from:

56. the use of any one of claims 1, 20, or 55, wherein the medicament is administered to a subject orally, subcutaneously, intravenously, or intramuscularly.

57. The use of claim 56, wherein the medicament is administered to a subject orally or intravenously.

58. The use of any one of claims 1, 20, or 55, wherein the depression is postpartum depression.

59. The use of any one of claims 1, 20 or 55, wherein the medicament is administered chronically.

60. The use of any one of claims 1, 20, or 55, wherein the subject is a mammal.

61. The use of claim 60, wherein the subject is a human.

62. The use of any one of claims 1, 20, or 55, wherein the medicament is administered in combination with another therapeutic agent.

Background

Brain excitability is defined as the level of arousal in an animal, i.e., a continuum ranging from coma to convulsion, and is regulated by a variety of neurotransmitters. In general, neurotransmitters are responsible for regulating the conduction of ions across neuronal membranes. At rest, the neuronal membrane has a potential (or membrane voltage) of about-70 mV, with the cell interior being negative relative to the cell exterior. The potential (voltage) is an ion (K) across a semi-permeable membrane of the neuron+、Na+、ClOrganic anion) balance. Neurotransmitters are stored in presynaptic vesicles and released under the influence of neuronal action potentials. When released into the synaptic cleft, excitatory chemical transmitters such as acetylcholine will cause depolarization of the membrane, e.g., a change in potential from-70 mV to-50 mV. This effect is mediated by postsynaptic nicotinic receptors that are stimulated by acetylcholine to increase Na+Membrane permeability of ions. Reduced membrane potential to synapseThe post-action potential form stimulates neuronal excitability.

In the case of the GABA Receptor Complex (GRC), the effect on brain excitability is mediated by GABA, a neurotransmitter. GABA has a profound effect on overall brain excitability, as up to 40% of the neurons in the brain utilize GABA as a neurotransmitter. GABA modulates the excitability of individual neurons by modulating the conduction of chloride ions across the neuronal membrane. GABA interacts with its recognition site on GRC to facilitate chloride ion flux to reduce the electrochemical gradient of GRC into the cell. This intracellular increase in anion levels causes hyperpolarization of the transmembrane potential, rendering the neuron less prone to excitatory input, i.e., reducing neuronal excitability. In other words, the higher the chloride ion concentration in the neuron, the lower the brain excitability and arousal level.

GRCs are well documented to be responsible for the mediation of anxiety, seizures, and sedation. Thus, GABA and drugs that act like or promote GABA action (e.g., therapeutically useful barbiturates and Benzodiazepines (BZ), e.g.) Through interaction with specific regulatory sites on GRCs, produce their therapeutically useful effects. Accumulating evidence has now shown that GRCs contain different sites for neuroactive steroids in addition to the benzodiazepine and barbiturate binding sites. See, e.g., Lan, N.C., et al, neurohem. Res. (1991)16: 347-.

Neuroactive steroids may occur endogenously. The most potent endogenous neuroactive steroids are 3 α -hydroxy-5-reductionpregn-20-one and 3 α -21-dihydroxy-5-reductionpregn-20-one, which are metabolites of the hormones steroid progesterone and deoxycorticosterone, respectively. The ability of these steroid metabolites to alter brain excitability was confirmed in 1986 (Majewska, M.D. et al, Science 232: 1004-.

The ovarian hormone progesterone and its metabolites have been shown to have profound effects on brain excitability (Backstrom, T. et al, Acta Obstet. Gynecol. Scan. Suppl.130:19-24 (1985); Pfaff, D.W and McEwen, B.S., Science 219: 808-. The levels of progesterone and its metabolites vary with the phase of the menstrual cycle. It is well documented that the levels of progesterone and its metabolites decrease before the onset of menstruation. Furthermore, the monthly recurrence of certain physical symptoms before the onset of menstruation is well documented. These symptoms that have become associated with Premenstrual Syndrome (PMS) include stress, anxiety and migraine (Dalton, k., Premenstrual Syndrome and prograsterone Therapy, 2 nd edition, Chicago Yerbook, Chicago (1984)). Subjects with PMS had a monthly recurrence of symptoms that existed pre-menstruation and did not exist post-menstruation.

In a similar manner, progesterone reduction is also temporally associated with increased frequency of seizure episodes in female epilepsy (i.e., menstrual epilepsy) (Laidlaw, j., Lancet, 1235-. A more direct correlation has been observed with respect to the reduction of progesterone metabolites (Rosciszewska et al, J.Neurol.Neurosurg.Psych.49:47-51 (1986)). In addition, the temporal incidence of seizure onset has been correlated with the incidence of symptoms of premenstrual syndrome in subjects with primary generalized petit epilepsy (Backstrom, t. et al, j. psychosm. obstet. gynaecol.2:8-20 (1983)). Steroidal deoxycorticosterones have been found to be effective in treating subjects with small seizures associated with their menstrual cycle (Aird, R.B. and Gordan, G., J.Amer.Med.Soc.145:715-719 (1951)).

In addition, the symptom associated with low progesterone levels is postpartum depression (PND). Shortly after delivery, progesterone levels are dramatically reduced, leading to the onset of PND. The symptoms of PND range from mild depression to psychosis requiring hospitalization. PND is also associated with severe anxiety and dysphoria. PND-related depression is not amenable to treatment by classical antidepressants, and women experiencing PND show an increased incidence of PMS (Dalton, k., Premenstrual Syndrome and progrestasterone Therapy, 2 nd edition, Chicago Yerbook, Chicago (1984)).

Collectively, these observations suggest a critical role for progesterone and deoxycorticosterone and more specifically their metabolites in the homeostatic regulation of brain excitability, manifested as seizures or an increase in symptoms associated with menstrual epilepsy, PMS and PND. The association between reduced levels of Progesterone and symptoms associated with PMS, PND and menstrual Epilepsy (Backstrom, T. et al, J Psychosom. Obstet. Gynaecol.2:8-20 (1983); Dalton, K., Premenstrual Syndrome and Progesterone Therapy, 2 nd edition, Chicago Yeast, Chicago (1984)) has prompted the use of Progesterone in its treatment (Mattson et al, "Meroxyprogesterone Therapy of cationic epiilepsy," Advances in Epilogy: XVth Epilpesy International Symposium, Raven Press, New York (1984), page 279 282; and Dalton, K., Syngnal moisture Therapy, fourth edition, York,2 nd edition (1984)). However, progesterone is not consistently effective in treating the aforementioned syndromes. For example, progesterone does not exhibit a dose-response relationship in the treatment of PMS (Maddocks et al, Obstet. Gynecol.154:573-581 (1986); Dennerstein et al, Brit. Med J290: 16-17 (1986)).

There is a need for new and improved neuroactive steroids which act as modulators of cerebral excitability, as well as agents for the prevention and treatment of CNS related diseases. The compounds, compositions and methods described herein are directed to this end.

Disclosure of Invention

Provided herein are C21 substituted neuroactive steroids designed, for example, to act as GABA modulators. In certain embodiments, these compounds are envisioned to be useful as therapeutic agents for inducing anesthesia and/or sedation in a subject. In some embodiments, these compounds are contemplated to be useful as therapeutic agents for treating CNS-related disorders (e.g., sleep disorders, mood disorders, schizophrenia spectrum disorders, spasticity, memory and/or cognitive disorders, movement disorders, personality disorders, autism spectrum disorders, pain, traumatic brain injury, vascular diseases, drug abuse disorders and/or withdrawal syndromes, or tinnitus) in a subject in need thereof (e.g., a subject with Rett syndrome, fragile X syndrome, or Angelman syndrome).

In one aspect, there is provided a compound of formula (I):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; r2Is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a single bond; and when saidWhen one of them is a double bond, R5Is absent.

The invention also provides pharmaceutical compositions comprising the compounds of the invention and methods of use and treatment, e.g., for inducing sedation and/or anesthesia, for the treatment of CNS-related disorders.

Steroids of formula (I), subgenera thereof, and pharmaceutically acceptable salts thereof are collectively referred to herein as "compounds of the invention".

In another aspect, there is provided a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient. In certain embodiments, the compounds of the present invention are provided in an effective amount in a pharmaceutical composition. In certain embodiments, the compounds of the present invention are provided in a therapeutically effective amount. In certain embodiments, the compounds of the present invention are provided in a prophylactically effective amount.

In certain embodiments, the compounds of the invention as described herein act as GABA modulators, e.g., affect GABA in a positive or negative mannerAA receptor. As by modulation of GABAAModulators of Central Nervous System (CNS) excitability mediated by the ability of the receptor, these compounds are expected to have CNS activity.

Thus, in another aspect, there is provided a method of treating a CNS related disorder in a subject in need thereof, comprising administering to the subject an effective amount of a compound of the invention. In certain embodiments, the CNS-related disorder is selected from the group consisting of sleep disorders, mood disorders, schizophrenia spectrum disorders, spasticity, memory and/or cognitive disorders, movement disorders, personality disorders, autism spectrum disorders, pain, traumatic brain injury, vascular disease, substance abuse disorders, and/or withdrawal syndrome, and tinnitus. In certain embodiments, the compound is administered orally, subcutaneously, intravenously, or intramuscularly. In certain embodiments, the compound is administered chronically. In certain embodiments, the compound is administered continuously, e.g., by continuous intravenous infusion.

Other objects and advantages will become apparent to those skilled in the art from consideration of the following detailed description, examples and claims.

Definition of

Chemical definition

The definitions of specific functional groups and chemical terms are described in more detail below. Chemical elements are identified according to the periodic table of elements, CAS edition, Handbook of Chemistry and Physics, 75 th edition, cover pages, and specific functional groups are generally defined as described herein. In addition, general principles of Organic Chemistry and specific functional moieties and reactivity are described in Thomas Sorrell, Organic Chemistry, University Science Books, Sausaltito, 1999; smith and March, March's Advanced Organic Chemistry, 5 th edition, John Wiley & Sons, Inc., New York, 2001; larock, Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989; and Carruther, Some model Methods of Organic Synthesis, 3 rd edition, Cambridge University Press, Cambridge, 1987.

The compounds described herein may contain one or more asymmetric centers and may therefore exist in various isomeric forms, such as enantiomers and/or diastereomers. For example, the compounds described herein may be in the form of individual enantiomers, diastereomers, or geometric isomers, or may be in the form of mixtures of stereoisomers (including racemic mixtures and mixtures enriched in one or more stereoisomers). The isomers may be isolated from the mixture by methods known to those skilled in the art, including chiral High Pressure Liquid Chromatography (HPLC) and the formation and crystallization of chiral salts; alternatively, preferred isomers may be prepared by asymmetric synthesis. See, e.g., Jacques et al, eneriomers, Racemates and solutions (Wiley Interscience, New York, 1981); wilen et al, Tetrahedron 33:2725 (1977); eliel, Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, tablets of solving Agents and Optical solutions, page 268 (E.L. Eliel, Ed., Univ.of Notre Dame Press, Notre Dame, IN 1972). The invention additionally encompasses compounds described herein in the form of individual isomers substantially free of other isomers and optionally in the form of mixtures of various isomers.

As used herein, a pure enantiomeric compound is substantially free of other enantiomers or stereoisomers of the compound (i.e., enantiomeric excess). In other words, the "S" form of the compound is substantially free of the "R" form of the compound and is thus in enantiomeric excess of the "R" form. The term "enantiomerically pure" or "pure enantiomer" means that the compound comprises more than 75 wt.%, more than 80 wt.%, more than 85 wt.%, more than 90 wt.%, more than 91 wt.%, more than 92 wt.%, more than 93 wt.%, more than 94 wt.%, more than 95 wt.%, more than 96 wt.%, more than 97 wt.%, more than 98 wt.%, more than 98.5 wt.%, more than 99 wt.%, more than 99.2 wt.%, more than 99.5 wt.%, more than 99.6 wt.%, more than 99.7 wt.%, more than 99.8 wt.%, or more than 99.9 wt.% of the enantiomer. In certain embodiments, the weight is based on the total weight of all enantiomers or stereoisomers of the compound.

In the compositions provided herein, enantiomerically pure compounds may be present with other active or inactive ingredients. For example, a pharmaceutical composition comprising an enantiomerically pure R-compound may comprise, for example, about 90% excipient and about 10% enantiomerically pure R-compound. In certain embodiments, the enantiomerically pure R-compounds in these compositions may, for example, comprise at least about 95% by weight of the R-compound and up to about 5% by weight of the S-compound, based on the total weight of the compound. For example, a pharmaceutical composition comprising an enantiomerically pure S-compound may comprise, for example, about 90% excipient and about 10% enantiomerically pure S-compound. In certain embodiments, the enantiomerically pure S-compounds in these compositions may, for example, comprise at least about 95% by weight of the S-compound and up to about 5% by weight of the R-compound, based on the total weight of the compound. In certain embodiments, the active ingredient may be formulated with little or no excipients or carriers.

The compounds described herein may also comprise one or more isotopic substitutions. For example, H may be in any isotopic form, including1H、2H (D or deuterium) and3h (T or tritium); c may be in any isotopic form, including12C、13C and14c; o is in any isotopic form, including16O and18o; and so on.

The articles "a" and "an" may be used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. For example, "an analog" refers to one analog or more than one analog.

When a range of values is recited, it is intended to include each value and sub-range within the recited range. For example, "C1-6Alkyl "is intended to cover C1、C2、C3、C4、C5、C6、C1-6、C1-5、C1-4、C1-3、C1-2、C2-6、C2-5、C2-4、C2-3、C3-6、C3-5、C3-4、C4-6、C4-5And C5-6An alkyl group.

The following terms are intended to have the meanings presented below with them and may be used to understand the description and intended scope of the present invention.

"alkyl" refers to a group of straight or branched chain saturated hydrocarbon groups having 1 to 20 carbon atoms ("C)1-20Alkyl "). In some embodiments, the alkyl group has 1 to 12 carbon atoms ("C)1-12Alkyl "). In some embodiments, the alkyl group has 1 to 8 carbon atoms ("C)1-8Alkyl "). In some embodiments, the alkyl group has 1 to 6 carbon atoms ("C)1-6Alkyl ", also referred to herein as" lower alkyl "). In some embodiments, the alkyl group has 1 to 5 carbon atoms ("C)1-5Alkyl "). In some embodiments, the alkyl group has 1 to 4 carbon atoms ("C)1-4Alkyl "). In some embodiments, the alkyl group has 1 to 3 carbon atoms ("C)1-3Alkyl "). In some embodiments, the alkyl group has 1 to 2 carbon atoms ("C)1-2Alkyl "). In some embodiments, the alkyl group has 1 carbon atom ("C)1Alkyl "). In some embodiments, the alkyl group has 2 to 6 carbon atoms ("C)2-6Alkyl "). C1-6Examples of alkyl groups include methyl (C)1) Ethyl (C)2) N-propyl (C)3) Isopropyl (C)3) N-butyl (C)4) Tert-butyl (C)4) Sec-butyl (C)4) Isobutyl (C)4) N-pentyl group (C)5) 3-pentyl radical (C)5) Pentyl group (C)5) Neopentyl (C)5) 3-methyl-2-butyl (C)5) Tert-amyl (C)5) And n-hexyl (C)6). Other examples of alkyl groups include n-heptyl (C)7) N-octyl (C)8) And the like. Unless otherwise indicated, each instance of an alkyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkyl") or substituted (a "substituted alkyl") with one or more substituents (e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent). In certain embodiments, the alkyl is unsubstituted C1-10The alkyl group (for example,-CH3). In certain embodiments, the alkyl is substituted C1-10An alkyl group. Common alkyl abbreviations include Me (-CH)3)、Et(-CH2CH3)、iPr(-CH(CH3)2)、nPr(-CH2CH2CH3)、n-Bu(-CH2CH2CH2CH3) Or i-Bu (-CH)2CH(CH3)2)。

"alkenyl" refers to a group of straight or branched chain hydrocarbon radicals having 2 to 20 carbon atoms, one or more carbon-carbon double bonds and no triple bonds ("C2-20Alkenyl "). In some embodiments, alkenyl groups have 2 to 10 carbon atoms ("C)2-10Alkenyl "). In some embodiments, alkenyl groups have 2 to 8 carbon atoms ("C)2-8Alkenyl "). In some embodiments, alkenyl groups have 2 to 6 carbon atoms ("C)2-6Alkenyl "). In some embodiments, alkenyl groups have 2 to 5 carbon atoms ("C)2-5Alkenyl "). In some embodiments, alkenyl groups have 2 to 4 carbon atoms ("C)2-4Alkenyl "). In some embodiments, alkenyl groups have 2 to 3 carbon atoms ("C)2-3Alkenyl "). In some embodiments, alkenyl has 2 carbon atoms ("C)2Alkenyl "). One or more carbon-carbon double bonds may be internal (e.g., in a 2-butenyl group) or terminal (e.g., in a 1-butenyl group). C2-4Examples of the alkenyl group include vinyl (C)2) 1-propenyl (C)3) 2-propenyl (C)3) 1-butenyl (C)4) 2-butenyl (C)4) Butadienyl radical (C)4) And the like. C2-6Examples of the alkenyl group include the foregoing C2-4Alkenyl and pentenyl (C)5) Pentadienyl (C)5) Hexenyl (C)6) And the like. Other examples of alkenyl groups include heptenyl (C)7) Octenyl (C)8) Octrienyl (C)8) And the like. Unless otherwise indicated, each instance of an alkenyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkenyl") or substituted (a "substituted alkenyl") with one or more substituents (e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent). In some embodiments of the present invention, the substrate is,said alkenyl being unsubstituted C2-10An alkenyl group. In certain embodiments, the alkenyl is substituted C2-10An alkenyl group.

"alkynyl" refers to a group ("C") having a straight or branched hydrocarbon group of 2 to 20 carbon atoms, one or more carbon-carbon triple bonds, and optionally one or more double bonds2-20Alkynyl "). In some embodiments, alkynyl groups have 2 to 10 carbon atoms ("C)2-10Alkynyl "). In some embodiments, alkynyl groups have 2 to 8 carbon atoms ("C)2-8Alkynyl "). In some embodiments, alkynyl has 2 to 6 carbon atoms ("C)2-6Alkynyl "). In some embodiments, alkynyl has 2 to 5 carbon atoms ("C)2-5Alkynyl "). In some embodiments, alkynyl groups have 2 to 4 carbon atoms ("C)2-4Alkynyl "). In some embodiments, alkynyl groups have 2 to 3 carbon atoms ("C)2-3Alkynyl "). In some embodiments, alkynyl has 2 carbon atoms ("C)2Alkynyl "). One or more carbon-carbon triple bonds may be internal (e.g., in 2-butynyl) or terminal (e.g., in 1-butynyl). C2-4Examples of alkynyl groups include, but are not limited to, ethynyl (C)2) 1-propynyl (C)3) 2-propynyl (C)3) 1-butynyl (C)4) 2-butynyl (C)4) And the like. C2-6Examples of the alkenyl group include the foregoing C2-4Alkynyl and pentynyl (C)5) Hexynyl (C)6) And the like. Other examples of alkynyl groups include heptynyl (C)7) (C) octynyl group8) And the like. Unless otherwise indicated, each instance of an alkynyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted alkynyl") or substituted (a "substituted alkynyl") with one or more substituents (e.g., 1 to 5 substituents, 1 to 3 substituents, or 1 substituent). In certain embodiments, the alkynyl is unsubstituted C2-10Alkynyl. In certain embodiments, the alkynyl is substituted C2-10Alkynyl.

"aryl" means a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n +2 aromatic ring system (e.g., bicyclic or tricyclic) providing 6 to 14 ring carbon atoms and 0 heteroatoms in the aromatic ring systemSharing 6, 10 or 14 pi electrons in a cyclic array) ("C)6-14Aryl "). In some embodiments, an aryl group has six ring carbon atoms ("C)6Aryl "; for example, phenyl). In some embodiments, an aryl group has ten ring carbon atoms ("C)10Aryl "; for example, naphthyl groups such as 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has fourteen ring carbon atoms ("C)14Aryl "; for example, an anthracene group). "aryl" also includes ring systems in which an aryl ring as defined above is fused to one or more carbocyclic or heterocyclic groups in which the linking group or point is on the aryl ring, and in these cases the number of carbon atoms continues to specify the number of carbon atoms in the aryl ring system. Aryl groups include, but are not limited to, phenyl, naphthyl, indenyl, and tetrahydronaphthyl. Unless otherwise indicated, each instance of an aryl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted aryl") or substituted (a "substituted aryl") with one or more substituents. In certain embodiments, the aryl is unsubstituted C6-14And (4) an aryl group. In certain embodiments, the aryl is substituted C6-14And (4) an aryl group.

In certain embodiments, aryl substituted with one or more groups is selected from halo, C1-C8Alkyl radical, C1-C8Haloalkyl, cyano, hydroxy, C1-C8Alkoxy and amino.

Representative examples of substituted aryl groups include the following

Wherein R is56And R57One of which may be hydrogen and R56And R57At least one of each is independently selected from C1-C8Alkyl radical, C1-C8Haloalkyl, 4-to 10-membered heterocyclyl, alkanoyl, C1-C8Alkoxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR58COR59、NR58SOR59、NR58SO2R59COOalkyl, COOaryl, CONR58R59、CONR58OR59、NR58R59、SO2NR58R59S-alkyl, SO2Alkyl, S aryl, SO2An aryl group; or R56And R57May be joined to form a cyclic ring of 5 to 8 atoms (saturated or unsaturated), optionally containing one or more heteroatoms selected from group N, O or S. R60And R61Independently of each other is hydrogen, C1-C8Alkyl radical, C1-C4Haloalkyl, C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl, substituted C6-C10Aryl, 5-10 membered heteroaryl or substituted 5-10 membered heteroaryl.

Other representative aryl groups having fused heterocyclic groups include the following:

wherein each W is selected from C (R)66)2、NR66O and S; and each Y is selected from carbonyl, NR66O and S; and R is66Independently of each other is hydrogen, C1-C8Alkyl radical, C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl and 5-10 membered heteroaryl.

Unless otherwise specified, "halo" or "halogen", alone or as part of another substituent, refers to a fluorine (F), chlorine (Cl), bromine (Br), or iodine (I) atom. The term "halide" by itself or as part of another substituent refers to a fluoride, chloride, bromide, or iodide atom. In certain embodiments, halo is fluoro or chloro.

"haloalkyl" and "haloalkoxy" can include alkyl and alkoxy structures substituted with one or more halo groups or with combinations thereof. For example, the terms "fluoroalkyl" and "fluoroalkoxy" include haloalkyl and haloalkoxy, respectively, wherein the halo is fluorine.

"heteroaryl" refers to a group of a 5-10 membered monocyclic or bicyclic 4n +2 aromatic ring system (e.g., sharing 6 or 10 pi electrons in a cyclic array) providing ring carbon atoms and 1-4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-10 membered heteroaryl"). In heteroaryl groups containing one or more nitrogen atoms, the point of attachment may be carbon or nitrogen depending on the valency allowed. Heteroaryl bicyclic ring systems may include one or more heteroatoms in one or both rings. "heteroaryl" includes ring systems in which a heteroaryl ring as defined above is fused to one or more carbocyclyl or heterocyclyl groups, wherein the point of attachment is on the heteroaryl ring, and in these cases the number of ring members continues to designate the number of ring members in the heteroaryl ring system. "heteroaryl" also includes ring systems in which a heteroaryl ring as defined above is fused with one or more aryl groups, where the point of attachment is on the aryl or heteroaryl ring, and in these cases the number of ring members designates the number of ring members in the fused (aryl/heteroaryl) ring system. Bicyclic heteroaryl groups, wherein one ring does not contain a heteroatom (e.g., indolyl, quinolinyl, carbazolyl, and the like), the point of attachment can be on either ring, i.e., a ring with a heteroatom (e.g., 2-indolyl) or a ring without a heteroatom (e.g., 5-indolyl).

In some embodiments, heteroaryl groups are 5-10 membered aromatic ring systems providing ring carbon atoms and 1-4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-10 membered heteroaryl"). In some embodiments, heteroaryl groups are 5-8 membered aromatic ring systems providing ring carbon atoms and 1-4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-8 membered heteroaryl"). In some embodiments, heteroaryl groups are 5-6 membered aromatic ring systems providing ring carbon atoms and 1-4 ring heteroatoms in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-6 membered heteroaryl"). In some embodiments, the 5-6 membered heteroaryl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1 ring heteroatom selected from nitrogen, oxygen, and sulfur. Unless otherwise indicated, each instance of heteroaryl is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heteroaryl") or substituted (a "substituted heteroaryl") with one or more substituents. In certain embodiments, the heteroaryl is an unsubstituted 5-14 membered heteroaryl. In certain embodiments, the heteroaryl is a substituted 5-14 membered heteroaryl.

Exemplary 5-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyrrolyl, furanyl, and thienyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, but are not limited to, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, but are not limited to, triazolyl, oxadiazolyl, and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, but are not limited to, tetrazolyl. Exemplary 6-membered heteroaryl groups containing one heteroatom include, but are not limited to, pyridinyl. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, but are not limited to, pyridazinyl, pyrimidinyl, and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, but are not limited to, triazinyl and tetrazinyl, respectively. Exemplary 7-membered heteroaryl groups containing one heteroatom include, but are not limited to, azepine, oxazepine and thiazepine. Exemplary 5, 6-bicyclic heteroaryls include, but are not limited to, indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothienyl, isobenzothienyl, benzofuranyl, benzoisothiofuranyl, benzimidazolyl, benzoxazolyl, benzoisoxazolyl, benzooxadiazolyl, benzothiazolyl, benzoisothiazolyl, benzothiadiazolyl, indolizinyl, and purinyl. Exemplary 6, 6-bicyclic heteroaryls include, but are not limited to, naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl.

Examples of representative heteroaryl groups include the following formulas:

wherein each Y is selected from carbonyl, N, NR65O and S; and R is65Independently of each other is hydrogen, C1-C8Alkyl radical, C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl and 5-10 membered heteroaryl.

"carbocyclyl" or "carbocycle" refers to a ring having from 3 to 10 ring carbon atoms in a non-aromatic ring system ("C)3-10Carbocyclyl ") and 0 heteroatom of a non-aromatic cyclic hydrocarbon group. In some embodiments, carbocyclyl has 3 to 8 ring carbon atoms ("C)3-8Carbocyclyl "). In some embodiments, carbocyclyl has 3 to 6 ring carbon atoms ("C)3-6Carbocyclyl "). In some embodiments, carbocyclyl has 3 to 6 ring carbon atoms ("C)3-6Carbocyclyl "). In some embodiments, carbocyclyl has 5 to 10 ring carbon atoms ("C)5-10Carbocyclyl "). Exemplary C3-6Carbocyclyl includes, but is not limited to, cyclopropyl (C)3) Cyclopropenyl group (C)3) Cyclobutyl (C)4) Cyclobutenyl radical (C)4) Cyclopentyl (C)5) Cyclopentenyl group (C)5) Cyclohexyl (C)6) Cyclohexenyl (C)6) Cyclohexadienyl (C)6) And the like. Exemplary C3-8Carbocyclyl includes, but is not limited to, C as described above3-6Carbocyclyl and cycloheptyl (C)7) Cycloheptenyl (C)7) Cycloheptadienyl (C)7) Cycloheptatrienyl (C)7) Cyclooctyl (C)8) Cyclooctenyl (C)8) Bicyclo [2.2.1]Heptyl (C)7) Bicyclo [2.2.2]Octyl radical (C)8) And the like. Exemplary C3-10Carbocyclyl includes, but is not limited to, C as described above3-8Carbocyclyl and cyclononyl (C)9) Cyclononenyl (C)9) Cyclodecyl (C)10) Cyclodecenyl (C)10) octahydro-1H-indenyl (C)9) Decahydronaphthyl (C)10) Spiro [4.5 ]]Decyl (C)10) And the like. As the foregoing examples illustrate, in certain embodiments, the carbocyclyl group is monocyclic ("monocyclic carbocyclyl") or contains a fused, bridged or spiro ringAre for example bicyclic systems ("bicyclic carbocyclyl") and may be saturated or may be partially unsaturated. "carbocyclyl" also includes ring systems in which a carbocyclic ring as defined above is fused to one or more aryl or heteroaryl groups, wherein the point of attachment is on the carbocyclic ring, and in these cases the number of carbons continues to specify the number of carbons in the carbocyclic ring system. Unless otherwise indicated, each instance of a carbocyclyl group is independently optionally substituted, i.e., unsubstituted (an "unsubstituted carbocyclyl") or substituted (a "substituted carbocyclyl") with one or more substituents. In certain embodiments, the carbocyclyl is unsubstituted C3-10A carbocyclic group. In certain embodiments, the carbocyclyl is substituted C3-10A carbocyclic group.

In some embodiments, "carbocyclyl" is a monocyclic saturated carbocyclyl ("C") having 3 to 10 ring carbon atoms3-10Cycloalkyl "). In some embodiments, cycloalkyl groups have 3 to 8 ring carbon atoms ("C)3-8Cycloalkyl "). In some embodiments, cycloalkyl groups have 3 to 6 ring carbon atoms ("C)3-6Cycloalkyl "). In some embodiments, cycloalkyl groups have 5 to 6 ring carbon atoms ("C)5-6Cycloalkyl "). In some embodiments, cycloalkyl groups have 5 to 10 ring carbon atoms ("C)5-10Cycloalkyl "). C5-6Examples of cycloalkyl include cyclopentyl (C)5) And cyclohexyl (C)5)。C3-6Examples of the cycloalkyl group include the aforementioned C5-6Cycloalkyl and cyclopropyl (C)3) And cyclobutyl (C)4)。C3-8Examples of the cycloalkyl group include the aforementioned C3-6Cycloalkyl and cycloheptyl (C)7) And cyclooctyl (C)8). Unless otherwise specified, each instance of a cycloalkyl group is independently unsubstituted (an "unsubstituted cycloalkyl") or substituted (a "substituted cycloalkyl") with one or more substituents. In certain embodiments, the cycloalkyl is unsubstituted C3-10A cycloalkyl group. In certain embodiments, said cycloalkyl is substituted C3-10A cycloalkyl group.

"heterocyclyl" or "heterocycle" refers to a group of a3 to 10 membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon ("3-10 membered heterocyclyl"). In heterocyclic groups containing one or more nitrogen atoms, the point of attachment may be carbon or nitrogen depending on the valency allowed. A heterocyclyl group can be a monocyclic ("monocyclic heterocyclyl") or a fused, bridged or spiro ring system such as a bicyclic system ("bicyclic heterocyclyl"), and can be saturated or can be partially unsaturated. Heterocyclyl bicyclic ring systems may include one or more heteroatoms in one or both rings. "heterocyclyl" also includes ring systems in which a heterocyclyl ring as defined above is fused to one or more carbocycles, where the point of attachment is on a carbocyclyl or heterocyclyl ring, or ring systems in which a heterocyclyl ring as defined above is fused to one or more aryl or heteroaryl groups, where the point of attachment is on a heterocyclyl ring, and in these cases the number of ring members continues to specify the number of ring members in the heterocyclyl ring system. Unless otherwise indicated, each instance of a heterocyclyl is independently optionally substituted, i.e., unsubstituted (an "unsubstituted heterocyclyl") or substituted (a "substituted heterocyclyl") with one or more substituents. In certain embodiments, the heterocyclyl is an unsubstituted 3-10 membered heterocyclyl. In certain embodiments, the heterocyclyl is a substituted 3-10 membered heterocyclyl.

In some embodiments, a heterocyclyl is a 5-10 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon ("5-10 membered heterocyclyl"). In some embodiments, heterocyclyl is a 5-8 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-8 membered heterocyclyl"). In some embodiments, heterocyclyl is a 5-6 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur ("5-6 membered heterocyclyl"). In some embodiments, the 5-6 membered heterocyclyl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heterocyclyl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heterocyclyl has one ring heteroatom selected from nitrogen, oxygen, and sulfur.

Exemplary 3-membered heterocyclic groups containing one heteroatom include, but are not limited to, aziridinyl, oxiranyl, thietanyl. Exemplary 4-membered heterocyclic groups containing one heteroatom include, but are not limited to, azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclic groups containing one heteroatom include, but are not limited to, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2, 5-dione. Exemplary 5-membered heterocyclic groups containing two heteroatoms include, but are not limited to, dioxolanyl, oxathiolanyl (oxathiolanyl), dithiofuranyl (disulphuranyl), and oxazolidin-2-one. Exemplary 5-membered heterocyclic groups containing three heteroatoms include, but are not limited to, triazolinyl, oxadiazolinyl, and thiadiazolinyl. Exemplary 6-membered heterocyclic groups containing one heteroatom include, but are not limited to, piperidinyl, tetrahydropyranyl, dihydropyridinyl, and thioalkyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, but are not limited to, piperazinyl, morpholinyl, dithianyl, dioxanyl. Exemplary 6-membered heterocyclic groups containing two heteroatoms include, but are not limited to, triazinanyl (triazanyl). Exemplary 7-membered heterocyclic groups containing one heteroatom include, but are not limited to, azepanyl, oxepinyl, and thiepanyl. Exemplary 8-membered heterocyclic groups containing one heteroatom include, but are not limited to, azocyclooctyl, oxocyclooctyl, and thiacyclooctyl. And C6Exemplary aryl ring fused 5-membered heterocyclic groups (also referred to herein as 5, 6-bicyclic heterocycles) include, but are not limited to, indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolonyl, and the like. Exemplary 6-membered heterocyclic groups fused to the aryl ring (also referred to herein as 6, 6-bicyclic heterocycles) include, but are not limited to, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.

Specific examples of heterocyclyl groups are shown in the following illustrative examples:

wherein each W is selected from CR67、C(R67)2、NR67O and S; and each Y is selected from NR67O and S; and R is67Independently of each other is hydrogen, C1-C8Alkyl radical, C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl and 5-10 membered heteroaryl. These heterocyclyl rings may be optionally substituted with one or more groups selected from acyl, acylamino, acyloxy, alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl (e.g., amido), aminocarbonylamino, aminosulfonyl, sulfonylamino, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, halogen, hydroxy, keto, nitro, thiol, -S-alkyl, -S-aryl, -S (O) -alkyl, -S (O) -aryl, -S (O)2-alkyl and-S (O)2-an aryl group. Substituents include carbonyl or thiocarbonyl groups which provide, for example, lactam and urea derivatives.

"acyl" refers to the group-C (O) R20Wherein R is20Is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl as defined herein. "alkanoyl" is acyl, wherein R is20Are groups other than hydrogen. Representative acyl groups include, but are not limited to, formyl (-CHO), acetyl (-C (═ O) CH3) Cyclohexylcarbonyl group, cyclohexylmethylcarbonyl group, benzoyl group (-C (═ O) Ph), benzylcarbonyl group (-C (═ O) CH2Ph)、--C(O)-C1-C8Alkyl, -C (O) - (CH)2)t(C6-C10Aryl), -C (O) - (CH)2)t(5-10 membered heteroaryl), -C (O) - (CH)2)t(C3-C10Cycloalkyl) and-C (O) - (CH)2)t(4-10 membered heterocyclyl), wherein t is an integer of 0 to 4. In certain embodiments, R21Is C substituted by halogen or hydroxy1-C8An alkyl group; or C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of said groups being unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy.

"acylamino" refers to the group-NR22C(O)R23Wherein R is22And R23Each occurrence of (a) is independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, as defined herein, or R22Is an amino protecting group. Exemplary "acylamino" groups include, but are not limited to, formylamino, acetylamino, cyclohexylcarbonylamino, cyclohexylmethyl-carbonylamino, benzoylamino and benzylcarbonylamino. A specific exemplary "acylamino" is-NR24C(O)-C1-C8Alkyl, -NR24C(O)-(CH2)t(C6-C10Aryl), -NR-24C(O)-(CH2)t(5-10 membered heteroaryl), -NR24C(O)-(CH2)t(C3-C10Cycloalkyl) and-NR24C(O)-(CH2)t(4-10 membered heterocyclyl), wherein t is an integer of 0 to 4, and each R24Independently represent hydrogen or C1-C8An alkyl group. In certain embodiments, R25Is H, C substituted by halogen or hydroxy1-C8An alkyl group; c3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of said groups being unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy substitution; and R is26Is H, C substituted by halogen or hydroxy1-C8An alkyl group; c3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of said groups being unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy substitution; with the proviso that R25And R26Is not H.

"acyloxy" refers to the group-OC (O) R27Wherein R is27Is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl as defined herein. Representative examples include, but are not limited to, formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl and benzylcarbonyl. In certain embodiments, R28Is C substituted by halogen or hydroxy1-C8An alkyl group; c3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of said groups being unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy.

"alkoxy" means a group-OR29Wherein R is29Is substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl. Specific alkoxy groups are methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy and 1, 2-dimethylbutoxy. Particular alkoxy groups are lower alkoxy groups, i.e., having 1 to 6 carbon atoms. Other specific alkoxy groups have 1 to 4 carbon atoms.

In certain embodiments, R29Is a group having 1 or more substituents, for example 1 to 5 substituents, and particularly 1 to 3 substituents, particularly 1 substituent, selected from amino, substituted amino, C6-C10Aryl, aryloxy, carboxy, cyano, C3-C10Cycloalkyl, 4-10 membered heterocyclyl, halogen, 5-10 membered heteroaryl, hydroxy, nitro, thioalkoxy, thioaryloxy, thiol, alkyl-S (O) -, aryl-S (O) -, alkyl-S (O)2And aryl-S (O)2-. Exemplary "substituted alkoxy" groups include, but are not limited to, -O- (CH)2)t(C6-C10Aryl), -O- (CH)2)t(5-10 membered heteroaryl), -O- (CH)2)t(C3-C10Cycloalkyl) and-O- (CH)2)t(4-10 membered heterocyclyl), wherein t is an integer from 0 to 4 and any aryl, heteroaryl, cycloalkyl or heterocyclyl present may itself be unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy. A specific exemplary 'substituted alkoxy' is-OCF3、-OCH2CF3、-OCH2Ph、-OCH2-cyclopropyl, -OCH2CH2OH and-OCH2CH2NMe2

"amino" refers to the group-NH2

"substituted amino" refers to the formula-N (R)38)2Wherein R is38Is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or an amino protecting group, wherein R is38Is not hydrogen. In certain embodiments, each R is38Independently selected from hydrogen, C1-C8Alkyl radical, C3-C8Alkenyl radical, C3-C8Alkynyl, C6-C10Aryl, 5-10 membered heteroaryl, 4-10 membered heterocyclyl or C3-C10A cycloalkyl group; or C substituted by halogen or hydroxy1-C8An alkyl group; c substituted by halogen or hydroxy3-C8An alkenyl group; c substituted by halogen or hydroxy3-C8Alkynyl, or- (CH)2)t(C6-C10Aryl), - (CH)2)t(5-10 membered heteroaryl), - (CH)2)t(C3-C10Cycloalkyl) or- (CH)2)t(4-10 membered heterocyclyl), wherein t is an integer from 0 to 8, each of said groups being unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy substitution; or two R38The groups join to form an alkylene group.

Exemplary "substituted amino" groups include, but are not limited to, -NR39-C1-C8Alkyl, -NR39-(CH2)t(C6-C10Aryl), -NR-39-(CH2)t(5-to 10-membered heteroaromatic group)Radical), -NR39-(CH2)t(C3-C10Cycloalkyl) and-NR39-(CH2)t(4-10 membered heterocyclyl), wherein t is an integer from 0 to 4, e.g. 1 or 2, each R39Independently represent hydrogen or C1-C8An alkyl group; and any alkyl groups present may themselves be substituted by halo, substituted or unsubstituted amino, or hydroxy; and any aryl, heteroaryl, cycloalkyl or heterocyclyl groups present may themselves be unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy. For the avoidance of doubt, the term 'substituted amino' includes the groups alkylamino, substituted alkylamino, alkylarylamino, substituted alkylarylamino, arylamino, substituted arylamino, dialkylamino and substituted dialkylamino as defined below. Substituted amino groups encompass mono-substituted amino and di-substituted amino groups.

"azido" refers to the group-N3

"carbamoyl" or "amido" refers to the group-C (O) NH2

"substituted carbamoyl" or "substituted amido" refers to the group-C (O) N (R)62)2Wherein each R is62Independently is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or an amino protecting group, wherein R is62Is not hydrogen. In certain embodiments, R62Selected from H, C1-C8Alkyl radical, C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl and 5-10 membered heteroaryl; or C substituted by halogen or hydroxy1-C8An alkyl group; or C3-C10Cycloalkyl, 4-10 membered heterocyclyl, C6-C10Aryl or 5-10 membered heteroaryl, each of said groups being unsubstituted C1-C4Alkyl, halo, unsubstituted C1-C4Alkoxy, unsubstituted C1-C4Haloalkyl, unsubstituted C1-C4Hydroxyalkyl or unsubstituted C1-C4Haloalkoxy or hydroxy substitution; with the proviso that at least one R62Is not H.

"carboxy" refers to the group-C (O) OH.

"cyano" refers to the group-CN.

"hydroxy" refers to the group-OH.

"Nitro" means the radical-NO2

"vinyl" refers to substituted or unsubstituted- (C ═ C) -. "ethylene" means substituted or unsubstituted- (C-C) -. "ethynyl" means- (C.ident.C) -.

"Nitrogen-containing heterocyclyl" group refers to a 4-to 7-membered non-aromatic cyclic group containing at least one nitrogen atom, such as, but not limited to, morpholine, piperidine (e.g., 2-piperidinyl, 3-piperidinyl, and 4-piperidinyl), pyrrolidine (e.g., 2-pyrrolidinyl and 3-pyrrolidinyl), azetidine, pyrrolidone, imidazoline, imidazolidinone, 2-pyrazoline, pyrazolidine, piperazine, and N-alkylpiperazines such as N-methylpiperazine. Specific examples include azetidines, piperidones, and piperazinones.

Alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl groups as defined herein are optionally substituted (e.g., "substituted" or "unsubstituted" alkyl, "substituted" or "unsubstituted" alkenyl, "substituted" or "unsubstituted" alkynyl, "substituted" or "unsubstituted" carbocyclyl, "substituted" or "unsubstituted" heterocyclyl, "substituted" or "unsubstituted" aryl or "substituted" or "unsubstituted" heteroaryl). In general, the term "substituted" whether preceded by the term "optionally," or not, means that at least one hydrogen present on the group (e.g., a carbon or nitrogen atom) is replaced with a permissible substituent (e.g., a substituent that, upon substitution, results in a stable compound (e.g., a compound that does not spontaneously undergo transformation such as rearrangement, cyclization, elimination, or other reaction). Unless otherwise specified, a "substituted" group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, the substituents at each position are the same or different. The term "substituted" is intended to include the replacement of the organic compound by all permissible substituents, any of which described herein results in the formation of a stable compound. The present invention encompasses any and all of these combinations to obtain stable compounds. For purposes of the present invention, a heteroatom such as nitrogen may have a hydrogen substituent and/or any suitable substituent as described herein that satisfies the valence of the heteroatom and results in the formation of a stable moiety.

Exemplary carbon atom substituents include, but are not limited to, halogen, -CN, -NO2、-N3、-SO2H、-SO3H、-OH、-ORaa、-ON(Rbb)2、-N(Rbb)2、-N(Rbb)3 +X-、-N(ORcc)Rbb、-SH、-SRaa、-SSRcc、-C(=O)Raa、-CO2H、-CHO、-C(ORcc)2、-CO2Raa、-OC(=O)Raa、-OCO2Raa、-C(=O)N(Rbb)2、-OC(=O)N(Rbb)2、-NRbbC(=O)Raa、-NRbbCO2Raa、-NRbbC(=O)N(Rbb)2、-C(=NRbb)Raa、-C(=NRbb)ORaa、-OC(=NRbb)Raa、-OC(=NRbb)ORaa、-C(=NRbb)N(Rbb)2、-OC(=NRbb)N(Rbb)2、-NRbbC(=NRbb)N(Rbb)2、-C(=O)NRbbSO2Raa、-NRbbSO2Raa、-SO2N(Rbb)2、-SO2Raa、-SO2ORaa、-OSO2Raa、-S(=O)Raa、-OS(=O)Raa、-Si(Raa)3、-OSi(Raa)3、-C(=S)N(Rbb)2、-C(=O)SRaa、-C(=S)SRaa、-SC(=S)SRaa、-SC(=O)SRaa、-OC(=O)SRaa、-SC(=O)ORaa、-SC(=O)Raa、-P(=O)2Raa、-OP(=O)2Raa、-P(=O)(Raa)2、-OP(=O)(Raa)2、-OP(=O)(ORcc)2、-P(=O)2N(Rbb)2、-OP(=O)2N(Rbb)2、-P(=O)(NRbb)2、-OP(=O)(NRbb)2、-NRbbP(=O)(ORcc)2、-NRbbP(=O)(NRbb)2、-P(Rcc)2、-P(Rcc)3、-OP(Rcc)2、-OP(Rcc)3、-B(Raa)2、-B(ORcc)2、-BRaa(ORcc)、C1-10Alkyl radical, C1-10Perhaloalkyl, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Carbocyclyl, 3-14 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RddSubstituted by groups;

Raaeach occurrence of (A) is independently selected from C1-10Alkyl radical, C1-10Perhaloalkyl, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Carbocyclyl, 3-14 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl, or two RaaThe groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring in which each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substitutedIs surrounded by 0, 1,2,3, 4 or 5RddSubstituted by groups;

Rbbeach occurrence of (A) is independently selected from hydrogen, -OH, -ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)2、C1-10Alkyl radical, C1-10Perhaloalkyl, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Carbocyclyl, 3-14 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl, or two RbbThe groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RddSubstituted by groups;

Rcceach instance of (A) is independently selected from hydrogen, C1-10Alkyl radical, C1-10Perhaloalkyl, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Carbocyclyl, 3-14 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl, or two RccThe groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RddSubstituted by groups;

Rddeach occurrence of (A) is independently selected from halogen, -CN, -NO2、-N3、-SO2H、-SO3H、-OH、-ORee、-ON(Rff)2、-N(Rff)2、-N(Rff)3 +X-、-N(ORee)Rff、-SH、-SRee、-SSRee、-C(=O)Ree、-CO2H、-CO2Ree、-OC(=O)Ree、-OCO2Ree、-C(=O)N(Rff)2、-OC(=O)N(Rff)2、-NRffC(=O)Ree、-NRffCO2Ree、-NRffC(=O)N(Rff)2、-C(=NRff)ORee、-OC(=NRff)Ree、-OC(=NRff)ORee、-C(=NRff)N(Rff)2、-OC(=NRff)N(Rff)2、-NRffC(=NRff)N(Rff)2、-NRffSO2Ree、-SO2N(Rff)2、-SO2Ree、-SO2ORee、-OSO2Ree、-S(=O)Ree、-Si(Ree)3、-OSi(Ree)3、-C(=S)N(Rff)2、-C(=O)SRee、-C(=S)SRee、-SC(=S)SRee、-P(=O)2Ree、-P(=O)(Ree)2、-OP(=O)(Ree)2、-OP(=O)(ORee)2、C1-6Alkyl radical, C1-6Perhaloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Carbocyclyl, 3-10 membered heterocyclyl, C6-10Aryl, 5-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RggSubstituted by groups;

Reeeach occurrence of (A) is independently selected from C1-6Alkyl radical, C1-6Perhaloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Carbocyclyl, C6-10Aryl, 3-10 membered heterocyclyl and 3-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3.4 or 5RggSubstituted by groups;

Rffeach instance of (A) is independently selected from hydrogen, C1-6Alkyl radical, C1-6Perhaloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Carbocyclyl, 3-10 membered heterocyclyl, C6-10Aryl and 5-10 membered heteroaryl, or two RffThe groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RggSubstituted by groups; and is

RggEach occurrence of (A) is independently halogen, -CN, -NO2、-N3、-SO2H、-SO3H、-OH、-OC1-6Alkyl, -ON (C)1-6Alkyl radical)2、-N(C1-6Alkyl radical)2、-N(C1-6Alkyl radical)3 +X-、-NH(C1-6Alkyl radical)2 +X-、-NH2(C1-6Alkyl radical)+X-、-NH3 +X-、-N(OC1-6Alkyl) (C1-6Alkyl), -N (OH) (C)1-6Alkyl), -NH (OH), -SH, -SC1-6Alkyl, -SS (C)1-6Alkyl), -C (═ O) (C)1-6Alkyl), -CO2H、-CO2(C1-6Alkyl), -OC (═ O) (C)1-6Alkyl), -OCO2(C1-6Alkyl), -C (═ O) NH2、-C(=O)N(C1-6Alkyl radical)2、-OC(=O)NH(C1-6Alkyl), -NHC (═ O) (C)1-6Alkyl), -N (C)1-6Alkyl) C (═ O) (C)1-6Alkyl), -NHCO2(C1-6Alkyl), -NHC (═ O) N (C)1-6Alkyl radical)2、-NHC(=O)NH(C1-6Alkyl), -NHC (═ O) NH2、-C(=NH)O(C1-6Alkyl), -OC (═ NH) (C)1-6Alkyl), -OC (═ NH) OC1-6Alkyl, -C (═ NH) N (C)1-6Alkyl radical)2、-C(=NH)NH(C1-6Alkyl), -C (═ NH) NH2、-OC(=NH)N(C1-6Alkyl radical)2、-OC(NH)NH(C1-6Alkyl), -OC (NH) NH2、-NHC(NH)N(C1-6Alkyl radical)2、-NHC(=NH)NH2、-NHSO2(C1-6Alkyl), -SO2N(C1-6Alkyl radical)2、-SO2NH(C1-6Alkyl), -SO2NH2、-SO2C1-6Alkyl, -SO2OC1-6Alkyl, -OSO2C1-6Alkyl, -SOC1-6Alkyl, -Si (C)1-6Alkyl radical)3、-OSi(C1-6Alkyl radical)3、-C(=S)N(C1-6Alkyl radical)2、C(=S)NH(C1-6Alkyl), C (═ S) NH2、-C(=O)S(C1-6Alkyl), -C (═ S) SC1-6Alkyl, -SC (═ S) SC1-6Alkyl, -P (═ O)2(C1-6Alkyl), -P (═ O) (C)1-6Alkyl radical)2、-OP(=O)(C1-6Alkyl radical)2、-OP(=O)(OC1-6Alkyl radical)2、C1-6Alkyl radical, C1-6Perhaloalkyl, C2-6Alkenyl radical, C2-6Alkynyl, C3-10Carbocyclyl, C6-10Aryl, 3-10 membered heterocyclyl, 5-10 membered heteroaryl; wherein X-Is a counter ion.

"counterions" or "anionic counterions" are negatively charged groups associated with cationic quaternary amino groups to maintain electronic neutrality. Exemplary counterions include halide ions (e.g., F)-、Cl-、Br-、I-)、NO3 -、ClO4 -、OH-、H2PO4 -、HSO4 -Sulfonate ions (e.g., methanesulfonate, trifluoromethanesulfonate, p-toluenesulfonate, benzenesulfonate, 10-camphorsulfonate, naphthalene-2-sulfonate, naphthalene-1-sulfonic acid-5-sulfonate, ethane-1-sulfonic acid-2-sulfonate, etc.), and carboxylate ions (e.g., acetate, propionate, benzoate, glycerate, lactate, tartrate, glycolate, etc.).

The nitrogen atoms may be substituted or unsubstituted as allowed by valence and include primary, secondary, tertiary and quaternary nitrogen atoms.Exemplary nitrogen atom substituents include, but are not limited to, hydrogen, -OH, -ORaa、-N(Rcc)2、-CN、-C(=O)Raa、-C(=O)N(Rcc)2、-CO2Raa、-SO2Raa、-C(=NRbb)Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)2N(Rcc)2、-P(=O)(NRcc)2、C1-10Alkyl radical, C1-10Perhaloalkyl, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Carbocyclyl, 3-14 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl, or two R attached to a nitrogen atomccThe groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RddIs substituted by radicals, and wherein Raa、Rbb、RccAnd RddAs defined above.

In certain embodiments, the substituent present on the nitrogen atom is an amino protecting group (also referred to herein as a nitrogen protecting group). Amino protecting groups include, but are not limited to, -OH, -ORaa、-N(Rcc)2、-C(=O)Raa、-C(=O)ORaa、-C(=O)N(Rcc)2、-S(=O)2Raa、-C(=NRcc)Raa、-C(=NRcc)ORaa、-C(=NRcc)N(Rcc)2、-SO2N(Rcc)2、-SO2Rcc、-SO2ORcc、-SORaa、-C(=S)N(Rcc)2、-C(=O)SRcc、-C(=S)SRcc、C1-10Alkyl radical, C2-10Alkenyl radical, C2-10Alkynyl, C3-10Carbocyclyl, 3-14 membered heterocyclyl, C6-14Aryl and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl and heteroaryl is independently substituted with 0, 1,2,3, 4 or 5RddIs substituted by radicals, and wherein Raa、Rbb、RccAnd RddAs defined herein. Amino Protecting Groups are well known in the art and include Protecting Groups in Organic Synthesis, t.w.greene and p.g.m.wuts, 3 rd edition, John Wiley, incorporated herein by reference&Sons,1999, detailed in those described in detail.

Exemplary amino protecting groups include, but are not limited to, amide groups (e.g., -C (═ O) Raa) Including, but not limited to, formamide and acetamide; carbamate group (e.g., -C (═ O) ORaa) Including, but not limited to, 9-fluorenylmethyl carbamate (Fmoc), tert-butyl carbamate (BOC), and benzyl carbamate (Cbz); sulfonamide groups (e.g., -S (═ O)2Raa) Including, but not limited to, p-toluenesulfonamide (Ts), methanesulfonamide (Ms) and N- [2- (trimethylsilyl) ethoxy]Methylamine (SEM).

In certain embodiments, the substituent present on the oxygen atom is an oxygen protecting group (also referred to as a hydroxyl protecting group). Oxygen protecting groups include, but are not limited to, -Raa、-N(Rbb)2、-C(=O)SRaa、-C(=O)Raa、-CO2Raa、-C(=O)N(Rbb)2、-C(=NRbb)Raa、-C(=NRbb)ORaa、-C(=NRbb)N(Rbb)2、-S(=O)Raa、-SO2Raa、-Si(Raa)3、-P(Rcc)2、-P(Rcc)3、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)(ORcc)2、-P(=O)2N(Rbb)2and-P (═ O) (NR)bb)2Wherein R isaa、RbbAnd RccAs defined hereinAnd (5) defining. Oxygen Protecting Groups are well known in the art and include Protecting Groups in Organic Synthesis, t.w.greene and p.g.m.wuts, 3 rd edition, John Wiley, incorporated herein by reference&Sons,1999, detailed in those described in detail.

Exemplary oxygen protecting groups include, but are not limited to, methyl, methoxymethyl (MOM), 2-methoxyethoxymethyl (MEM), benzyl (Bn), Triisopropylsilyl (TIPS), tert-butyldimethylsilyl (TBDMS), tert-butylmethoxyphenylsilyl (TBMPS), mesylate (methanesulfonate/mesylate), and tosylate (Ts).

In certain embodiments, the substituent present on the sulfur atom is a sulfur protecting group (also referred to as a thiol protecting group). Sulfur protecting groups include, but are not limited to, -Raa、-N(Rbb)2、-C(=O)SRaa、-C(=O)Raa、-CO2Raa、-C(=O)N(Rbb)2、-C(=NRbb)Raa、-C(=NRbb)ORaa、-C(=NRbb)N(Rbb)2、-S(=O)Raa、-SO2Raa、-Si(Raa)3、-P(Rcc)2、-P(Rcc)3、-P(=O)2Raa、-P(=O)(Raa)2、-P(=O)(ORcc)2、-P(=O)2N(Rbb)2and-P (═ O) (NR)bb)2Wherein R isaa、RbbAnd RccAs defined herein. Sulfur Protecting Groups are well known in the art and include Protecting Groups in Organic Synthesis, T.W.Greene and P.G.M.Wuts, 3 rd edition, John Wiley, incorporated herein by reference&Sons,1999, detailed in those described in detail.

These and other exemplary substituents are described in more detail in the detailed description, examples, and claims. The present invention is not intended to be limited in any way by the above-described exemplary list of substituents.

Other definitions

As used herein, the term "modulation" refers to inhibition or enhancement of GABA receptor function. A "modulator" (e.g., modulator compound) can be, for example, an agonist, partial agonist, antagonist, or partial antagonist of the GABA receptor.

"pharmaceutically acceptable" means having been or can be approved by a regulatory agency of the federal or a state government or a corresponding agency in a country other than the united states, or listed in the U.S. pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.

"pharmaceutically acceptable salt" refers to a salt of a compound of the invention that is pharmaceutically acceptable and has the desired pharmacological activity of the parent compound. In particular, these salts are non-toxic and may be inorganic or organic acid addition salts and base addition salts. In particular, these salts include: (1) acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1, 2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo [2.2.2] -oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, laurylsulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) when the acidic proton present in the parent compound is replaced by a metal ion, such as an alkali metal ion, alkaline earth ion, or aluminum ion; or a salt formed when coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, N-methylglucamine, or the like. By way of example only, salts also include sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functional group, is a salt of a non-toxic organic or inorganic acid, such as hydrochloride, hydrobromide, tartrate, methanesulfonate, acetate, maleate, oxalate, etc. The term "pharmaceutically acceptable cation" refers to an acceptable cationic counterion of the acidic functionality. Examples of such cations are sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium cations, and the like. See, e.g., Berge et al, J.pharm.Sci. (1977)66 (1: 1-79.

"solvate" refers to a form of a compound (also referred to as a "hydrate") that associates with a solvent or water, typically by a solvolysis reaction. Such physical association includes hydrogen bonding. Conventional solvents include water, ethanol, acetic acid, and the like. The compounds of the invention may be prepared, for example, in crystalline form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and also includes stoichiometric and non-stoichiometric solvates. In certain cases, such as when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid, the solvate will be able to separate. "solvates" encompasses both solution phase and isolatable solvates. Representative solvates include hydrates, ethanolates, and methanolates.

As used herein, the term "isotopic variant" refers to compounds in which one or more of the atoms comprising such compound contains an unnatural proportion of an isotope. For example, an "isotopic variant" of a compound can contain one or more non-radioactive isotopes, such as deuterium (g), (b), (c), (d), and (d)2H or D), carbon-13 (13C) Nitrogen-15 (15N), and the like. It is understood that in compounds that undergo such isotopic substitution, the following atoms, when present, can be varied such that, for example, any hydrogen can be2H/D, any carbon can be13C, or any nitrogen may be15N, and the presence and placement of these atoms can be determined within the skill of the art. Likewise, the invention may include the preparation of isotopic variants with radioisotopes, for example, in cases where the resulting compounds are useful in drug and/or substrate tissue distribution studies. Radioisotope tritium (i.e. tritium3H) And carbon-14 (i.e.14C) It is particularly useful for this purpose in view of its ease of incorporation and ready detection means. In addition, positron emitting isotopes such as11C、18F、15O and13n-substituted and will be useful in Positron Emission Tomography (PET) studies to study substrate receptor occupancyThe compound of (1). All isotopic variations of the compounds provided herein, whether radioactive or not, are intended to be encompassed within the scope of the present invention.

"stereoisomers": further, it is understood that compounds having the same molecular formula but differing in the nature or order of bonding of their atoms or the arrangement of their atoms in space are referred to as "isomers". Isomers whose atoms are arranged differently in space are referred to as "stereoisomers". Stereoisomers that are non-mirror images of each other are referred to as "diastereomers" and stereoisomers that are non-overlapping mirror images of each other are referred to as "enantiomers". When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible. Enantiomers can be characterized by the absolute configuration of their asymmetric centers and are described by the R-and S-sequence rules of Cahn and Prelog, or by: where the molecule rotates the plane of polarized light and is designated dextrorotatory or levorotatory (i.e., the (+) or (-) -isomer, respectively). The chiral compounds may exist as individual enantiomers or as mixtures thereof. Mixtures containing equal proportions of enantiomers are referred to as "racemic mixtures".

"tautomer" refers to a compound that is a tautomeric form of a particular compound structure and the shifts of hydrogen atoms and electrons are altered. Thus, both structures can be in equilibrium by the movement of pi electrons and atoms (usually H). For example, enols and ketones are tautomers, as they can be rapidly interconverted by treatment with acid or base. Another example of tautomerism is the acid-and nitro-forms of phenylnitromethane, which are likewise formed by treatment with an acid or a base. Tautomeric forms may be relevant for achieving optimal chemical reactivity and biological activity of the compound of interest.

A "subject" contemplated for administration includes, but is not limited to, a human (i.e., a male or female of any age group, e.g., a pediatric subject (e.g., an infant, a child, an adolescent) or an adult subject (e.g., a young adult, a middle-aged adult, or an elderly adult)) and/or a non-human animal, e.g., a mammal such as a primate (e.g., a cynomolgus monkey, a rhesus monkey), a cow, a pig, a horse, a sheep, a goat, a rodent, a cat, and/or a dog. In certain embodiments, the subject is a human. In certain embodiments, the subject is a non-human animal. The terms "human," "patient," and "subject" are used interchangeably herein.

Diseases, disorders, and conditions are used interchangeably herein.

As used herein, and unless otherwise indicated, the term "treating" encompasses actions that occur when a subject is afflicted with a specified disease, disorder, or condition, which reduce the severity of the disease, disorder, or condition, or delay or slow the progression of the disease, disorder, or condition ("therapeutic treatment"), and actions that occur before the subject begins to be afflicted with the specified disease, disorder, or condition ("prophylactic treatment").

Generally, an "effective amount" of a compound is an amount sufficient to elicit a desired biological response, e.g., to treat a CNS-related disorder, sufficient to induce anesthesia or sedation. As will be appreciated by those of ordinary skill in the art, the effective amount of a compound of the present invention may vary depending on factors such as: the desired biological endpoint, the pharmacokinetics of the compound, the disease being treated, the mode of administration, and the age, weight, health, and condition of the subject. An effective amount encompasses both therapeutic and prophylactic treatment.

As used herein, and unless otherwise indicated, a "therapeutically effective amount" of a compound is an amount sufficient to provide a therapeutic benefit in the treatment of a disease, disorder, or condition, or to delay or minimize one or more symptoms associated with the disease, disorder, or condition. A therapeutically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment of a disease, disorder, or condition. The term "therapeutically effective amount" can encompass an amount that improves overall treatment, reduces or avoids symptoms or causes of a disease or condition, or enhances the therapeutic efficacy of another therapeutic agent.

As used herein, and unless otherwise indicated, a "prophylactically effective amount" of a compound is an amount sufficient to prevent a disease, disorder, or condition, or one or more symptoms associated with the disease, disorder, or condition, or to prevent relapse thereof. A prophylactically effective amount of a compound refers to the amount of a therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in the treatment of a disease, disorder, or condition. The term "prophylactically effective amount" can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent.

Detailed Description

As generally described herein, the present invention provides C21 substituted neuroactive steroids designed, for example, to act as GABA modulators. In certain embodiments, these compounds are envisioned to be useful as therapeutic agents for inducing anesthesia and/or sedation in a subject. In certain embodiments, these compounds are contemplated to be useful as therapeutic agents for the treatment of CNS-related disorders.

Compound (I)

In one aspect, there is provided a compound of formula (I):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; r2Is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a single bond; and when saidWhen one of them is a double bond, R5Is absent.

In one aspect, there is provided a compound of formula (Ia):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a single bond; and when saidWhen one of them is a double bond, R5Is absent.

In some embodiments, the compound has formula (Ia-1):

in some embodiments, the compound has formula (Ia-2):

in some embodiments, a is monocyclic or bicyclic.

In some embodiments, a is monocyclic. In some aspects of these embodiments, a is attached through nitrogen. In some embodiments, a is heteroaryl. In some aspects of these embodiments, the heteroaryl group comprises up to five nitrogen atoms. In some embodiments, a is a5 membered heteroaryl or heterocyclyl. In some embodiments, a is a5 membered heteroaryl or heterocyclyl group containing up to four nitrogen atoms. In some embodiments, a is a5 membered heteroaryl or heterocyclyl group comprising 2,3, or 4 nitrogen atoms. In some embodiments, a is pyrazole, triazole, or tetrazole. In some embodiments, a is unsubstituted pyrazole, triazole, or tetrazole. In some embodiments, a is In some embodiments, a is unsubstituted triazole. In some embodiments, a is

In some embodiments, a is bicyclic. In some aspects of these embodiments, a is attached through nitrogen. In some embodiments, a is heteroaryl. In some aspects of these embodiments, the heteroaryl group comprises up to five nitrogen atoms. In some embodiments, the heteroaryl group comprises at least two nitrogen atoms. In some embodiments, a is heteroaryl comprising up to four nitrogen atoms. In some embodiments, a is heteroaryl comprising up to three nitrogen atoms. In some embodiments, a is heteroaryl comprising 2,3, or 4 nitrogen atoms. In some embodiments, the heteroaryl is benzotriazole, azabenzotriazole, diazabenzotriazole, benzopyrazole, azabenzopyrazole, or diazabenzpyrazole.

In some embodiments, the compound has formula (Ia-3):

wherein R is4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc) (ii) a Each RaIs hydrogen or C1-C6An alkyl group; each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and n is 0, 1,2 or 3.

In some embodiments, the compound has formula (Ia-4):

wherein R is4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc) (ii) a Each RaIs hydrogen or C1-C6An alkyl group; each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and n is 0, 1,2 or 3.

In some embodiments, n is 0.

In some embodiments, n is 1. In some aspects of these embodiments, R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)ORaor-S (O)0-2Ra. In some embodiments, n is 1 and R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)ORaor-S (O)0-2Ra. In some aspects of these embodiments, R4Is a halo group (e.g., F, Cl, Br). In some embodiments, R4Is F. In some embodiments, R4Is Cl. In some embodiments, R4Is Br. In some embodiments, R4Is C1-C6Alkoxy (e.g., -OCH)3、-OCH2CH3). In some embodiments, R4Is cyano. In some embodiments, R4is-C (O) RaOR-C (O) ORa. In some aspects of these embodiments, RaIs C1-C6Alkyl (e.g., -CH)3、-CH2CH3). In some embodiments, R4is-S (O)0-2Ra. In some embodiments, R4is-S (O)2RaAnd R isaIs C1-C6An alkyl group. In some aspects of these embodiments, Rais-CH3. In some embodiments, R4is-S (O)2CH3. In some embodiments, R4Is C1-C6Alkyl (e.g., -CH)3、-CH2CH3)。

In some embodiments, n is 2. In some aspects of these embodiments, R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)ORaor-S (O)0-2Ra. In some embodiments, n is 2 and R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)ORaor-S (O)0-2Ra. In some aspects of these embodiments, R4Is a halo group (e.g., F, Cl, Br). In some embodiments, R4Is F. In some embodiments, R4Is Cl. In some embodiments, R4Is Br. In some embodiments, R4Is C1-C6Alkoxy (e.g., -OCH)3、-OCH2CH3). In some embodiments, R4Is cyano. In some embodiments, R4is-C (O) RaOR-C (O) ORa. In some aspects of these embodiments, RaIs C1-C6Alkyl (e.g., -CH)3、-CH2CH3). In some embodiments, R4is-S (O)0-2Ra. In some embodiments, R4is-S (O)2RaAnd R isaIs C1-C6An alkyl group. In some aspects of these embodiments, Rais-CH3. In some embodiments, R4Is C1-C6Alkyl (e.g., -CH)3、-CH2CH3)。

In some embodiments, n is 1 or 2, and R is4Is C1-C6Alkyl OR C (O) ORa. In some embodimentsIn the scheme, the compound R4Is methyl. In some embodiments, the compound RaIs C1-C6An alkyl group. In some embodiments, RaIs ethyl.

In some embodiments, R1Is hydrogen or C1-C6Alkyl, and R4is-C (O) ORa

In some embodiments, R1Is C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl. In some embodiments, the compound R1Is C1-C6An alkyl group. In some embodiments, the compound R1Is methyl, ethyl or isopropyl.

In some embodiments, R1Is methyl and R4is-C (O) OEt.

In some embodiments, the compound is selected from:

in one aspect, there is provided a compound of formula (Ib):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; r2Is C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a single bond; and when saidWhen one of them is a double bond, R5Is absent.

In some embodiments, the compound has formula (Ib-1):

in some embodiments, the compound has formula (Ib-2):

in some embodiments, the compound has formula (Ib-3)

Wherein R is4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc) (ii) a Each RaIs hydrogen or C1-C6An alkyl group; each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and n is 0, 1,2 or 3.

In some embodiments, the compound has formula (Ib-4)

Wherein R is4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc) (ii) a Each RaIs hydrogen or C1-C6An alkyl group; each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and n is 0, 1,2 or 3.

In some embodiments, a is monocyclic.

In some embodiments, a is bicyclic.

In some embodiments, a is attached through nitrogen.

In some embodiments, a is a 5-or 6-membered heteroaryl or heterocyclyl. In some embodiments, a is a 5-or 6-membered heteroaryl or heterocyclyl group containing up to four nitrogen atoms. In some embodiments, a is a 5-or 6-membered heteroaryl or heterocyclyl group comprising 1,2,3, or 4 nitrogen atoms.

In some embodiments, a is heterocyclyl. In some embodiments, a is morpholine or piperazine. In some embodiments, a is

In some embodiments, a is heteroaryl. In some aspects of these embodiments, the heteroaryl group comprises up to five nitrogen atoms. In some embodiments, the heteroaryl is benzotriazole, azabenzotriazole, diazabenzotriazole, benzopyrazole, azabenzopyrazole, or diazabenzpyrazole. In some embodiments, a is

In some embodiments, the heteroaryl is 5 membered.

In some embodiments, a comprises up to four nitrogen atoms. In some embodiments, a comprises 2,3, or 4 nitrogen atoms. In some embodiments, a is pyrazole, triazole, or tetrazole. In some embodiments, a is unsubstituted pyrazole, triazole, or tetrazole. In some embodiments, a isIn some embodiments, a is unsubstituted triazole. In some embodiments, a is

In some embodiments, R1Is C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl. In some embodiments, R1Is C1-C6An alkyl group. In some embodiments, R1Is methyl, ethyl or isopropyl. In some embodiments, R1Is methyl. In some embodiments, R1Is ethyl.

In some embodiments, R2Is C1-C6An alkyl group. In some embodiments, R2Is methyl. In some embodiments, R2Is C1-C6A haloalkyl group. In some embodiments, R2is-CF3

In some embodiments, n is 0.

In some embodiments, n is 1 or 2, and R is4Is cyano, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Raor-S (O)0-2Ra

In some embodiments, R4Is Br, Cl or F. In some embodiments, R4Is F. In some embodiments, R4Is Cl. In some embodiments, R4Is Br. In some embodiments, R4is-OCH3. In some embodiments, R4is-OCF3. In some embodiments, R4Is cyano. In some embodiments, R4Is C1-C6An alkyl group. In some embodiments, R4Is methyl. In some embodiments, R4is-C (O) Ra. In some embodiments, RaIs C1-C6An alkyl group. In some embodiments, RaIs methyl. In some embodiments, R4is-CF3. In some embodiments, R4is-S (O)2Ra. In some embodiments, RaIs methyl.

In some embodiments, R1Is C1-C6Alkyl and R4is-C (O) Ra. In some embodiments, R1Is methyl and R4is-C (O) Me.

In some embodiments, n is 2, and R is4Is cyano, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Raor-S (O)0-2Ra. In some embodiments, n is 2, one R4Is F and one R4Is F. In some embodiments, n is 2, one R4Is F and one R4Is Cl. In some embodiments, n is 2, one R4is-OCH3And one R4is-OCH3

In some embodiments, n is 0 or 1; r1Is hydrogen or C1-C6An alkyl group; and R is2Is methyl.

In some embodiments, R1Is methyl, ethyl or isopropyl. In some embodiments, R1Is methyl.

In some embodiments, R4Is C1-C6Alkyl, -C (O) Raor-S (O)0-2Ra. In some embodiments, R4Is methyl. In some embodiments, R4is-C (O) Me. In some embodiments, R4is-S (O)2Me. In some embodiments, R4Is cyano.

In some embodiments, the compound is selected from:

in one aspect, there is provided a pharmaceutical composition comprising a compound of any one of the preceding claims and a pharmaceutically acceptable excipient.

In one aspect, there is provided a method of inducing sedation and/or anesthesia in a subject comprising administering to the subject an effective amount of a compound of formula (I):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; r2Is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a single bond; and when saidWhen one of them is a double bond, R5Is absent.

In one aspect, there is provided a method of administering to a subject in need thereof an effective amount of a compound, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of a compound as described herein (e.g., a compound of formula (I), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ib-1), (Ib-2), (Ib-3), or (Ib-4)), wherein the subject experiences sedation and/or anesthesia within two hours of administration.

In some embodiments, the subject experiences sedation and/or anesthesia within one hour of administration.

In some embodiments, the subject is immediately subjected to sedation and/or anesthesia.

In some embodiments, the compound is administered by intravenous administration.

In some embodiments, the compound is administered chronically.

In some embodiments, the subject is a mammal. In some embodiments, the subject is a human.

In some embodiments, the compound is administered in combination with another therapeutic agent.

In another aspect, there is provided a method for treating tics in a subject, comprising administering to the subject an effective amount of a compound of formula (I):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; r2Is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a sheetA key; and when saidWhen one of them is a double bond, R5Is absent.

In one aspect, there is provided a method for treating epilepsy or status epilepticus in a subject, the method comprising administering to the subject an effective amount of a compound of formula (I):

a pharmaceutically acceptable salt thereof, wherein: a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group; l is-C (R)3)(R3) -, -O-, -S-or-NR3-;R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl; r2Is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group; each R3Independently is hydrogen or C1-C6An alkyl group; r5Is absent or hydrogen; and isRepresents a single or double bond, whereinWhen one of them is a double bond, the other isIs a single bond; and when saidWhen one of them is a double bond, R5Is absent.

In one aspect, there is provided a method for treating a disorder associated with GABA function in a subject in need thereof, said method comprising administering to said subject a therapeutically effective amount of a compound, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of one of the compounds as described herein (e.g., a compound of one of formulae (I), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ib-1), (Ib-2), (Ib-3), or (Ib-4)).

In one aspect, there is provided a method for treating a CNS related disorder in a subject in need thereof, comprising administering to the subject an effective amount of a compound as described herein, for example a compound of formula (I), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ib-1), (Ib-2), (Ib-3) or (Ib-4), or a pharmaceutically acceptable salt thereof. In some embodiments, the CNS-related disorder is a sleep disorder, a mood disorder, a schizophrenia spectrum disorder, a spasticity, a memory and/or cognitive disorder, a movement disorder, a personality disorder, an autism spectrum disorder, pain, traumatic brain injury, a vascular disease, a substance abuse disorder and/or withdrawal syndrome, or tinnitus. In some embodiments, the subject is a subject with Rett syndrome, fragile X syndrome, or angleman syndrome.

In one aspect, there is provided a kit comprising a solid composition comprising a compound as described herein, for example, a compound of formula (I), (Ia-1), (Ia-2), (Ia-3), (Ia-4), (Ib-1), (Ib-2), (Ib-3) or (Ib-4), and a sterile diluent.

Pharmaceutical composition

In one aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention (also referred to as an "active ingredient") and a pharmaceutically acceptable excipient. In certain embodiments, the pharmaceutical composition comprises an effective amount of an active ingredient. In certain embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an active ingredient. In certain embodiments, the pharmaceutical composition comprises a prophylactically effective amount of the active ingredient.

The pharmaceutical compositions provided herein can be administered by a variety of routes, including, but not limited to, oral (enteral) administration, parenteral (by injection), rectal administration, transdermal administration, intradermal administration, intrathecal administration, Subcutaneous (SC) administration, Intravenous (IV) administration, Intramuscular (IM) administration, and intranasal administration.

In general, the compounds provided herein are administered in an effective amount. The amount of compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight and response of the individual patient, the severity of the patient's symptoms, and the like.

When used to prevent the onset of a CNS disorder, the compounds provided herein will generally be administered to a subject at risk of developing the condition, at the dosage levels described above, under the recommendation and supervision of a physician. Subjects at risk of developing a particular condition typically include those with a family history of the condition, or those that have been identified by genetic testing or screening as particularly susceptible to developing the condition.

The pharmaceutical compositions provided herein can also be administered chronically ("chronic administration"). By long-term administration is meant administration of the compound or pharmaceutical composition thereof over an extended period of time, e.g., over 3 months, 6 months, 1 year, 2 years, 3 years, 5 years, etc., or may continue indefinitely, e.g., for the remainder of the subject's life. In certain embodiments, chronic administration is intended to provide a constant level of the compound in the blood, e.g., within a therapeutic window over an extended period of time.

The pharmaceutical compositions of the present invention can additionally be delivered using a variety of methods of administration. For example, in certain embodiments, the pharmaceutical composition may be provided as a bolus injection, e.g., to increase the concentration of the compound in the blood to an effective level. The placement of the bolus dose depends on the systemic level of the desired active ingredient throughout the body, e.g. intramuscular or subcutaneous bolus doses allow a slow release of the active ingredient, while boluses delivered directly to the vein (e.g. by IV drip) allow a faster delivery which quickly increases the concentration of the active ingredient in the blood to an effective level. In other embodiments, the pharmaceutical composition may be administered in the form of a continuous infusion (e.g., by IV drip) to provide maintenance of a steady state concentration of the active ingredient in the subject. Furthermore, in other embodiments, the pharmaceutical composition may be administered first in a bolus dose, followed by continuous infusion.

Compositions for oral administration may take the form of bulk liquid solutions or suspensions, or bulk powders. More often, however, the compositions are presented in unit dosage form to facilitate accurate administration. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampoules or syringes of liquid composition or, in the case of solid compositions, pills, tablets, capsules and the like. In these compositions, the compound is typically a minor component (about 0.1 to about 50% or preferably about 1 to about 40% by weight), the remainder being various vehicles or excipients and processing aids that assist in forming the desired dosage form.

Under oral administration, one to five and especially two to four and usually three oral doses per day are representative regimens. Using these modes of administration, each dose provides about 0.01 to about 20mg/kg of a compound provided herein, with preferred doses each providing about 0.1 to about 10mg/kg and particularly about 1 to about 5 mg/kg.

Transdermal dosages are typically selected to provide similar or lower blood levels than achieved using injectable dosages, typically in amounts ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.

The injectable dosage levels range from about 0.1 mg/kg/hr to at least 20 mg/kg/hr, both from about 1 to about 120 hours and especially from 24 to 96 hours. A pre-loaded bolus of about 0.1mg/kg to about 10mg/kg or more may also be administered to achieve sufficient steady state levels. For 40 to 80kg human patients, the maximum total dose is expected to be no more than about 5 grams per day.

Liquid forms suitable for oral administration may include suitable aqueous or non-aqueous vehicles with buffers, suspending and dispersing agents, coloring agents, flavoring agents, and the like. Solid forms may include, for example, any of the following ingredients or compounds of similar nature: binders, such as microcrystalline cellulose, tragacanth or gelatin; excipients, such as starch or lactose; disintegrants, for example alginic acid, Primogel or corn starch; lubricants, such as magnesium stearate; glidants, such as colloidal silicon dioxide; sweetening agents, such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.

Injectable compositions are typically based on injectable sterile saline or phosphate buffered saline or other injectable excipients known in the art. As before, the active compounds in these compositions are generally minor components, generally about 0.05 to 10% by weight, the remainder being injectable excipients and the like.

Transdermal compositions are typically formulated as topical ointments or creams containing the active ingredient. When formulated as an ointment, the active ingredient will typically be combined with a paraffin or water-miscible ointment base. Alternatively, the active ingredient may be formulated as a cream with, for example, an oil-in-water cream base. These transdermal formulations are well known in the art and typically include additional ingredients to enhance skin penetration of the stability of the active ingredient or formulation. All such known transdermal formulations and ingredients are included within the scope provided herein.

The compounds provided herein can also be administered via a transdermal device. Thus, transdermal administration can be achieved using a reservoir or patch of the porous membrane type or solid matrix type.

The above components for orally administrable, injectable or topically administrable compositions are merely representative. Other materials and processing techniques are described in Remington's Pharmaceutical Sciences, 17 th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, section 8, which is incorporated herein by reference.

The compounds of the present invention may also be administered in sustained release form or from a sustained release drug delivery system. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.

The invention also relates to pharmaceutically acceptable acid addition salts of the compounds of the invention. Acids useful for preparing pharmaceutically acceptable salts are those which form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions such as hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, p-toluenesulfonate and the like.

In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention and a pharmaceutically acceptable excipient, e.g., a composition suitable for injection, e.g., for Intravenous (IV) administration.

Pharmaceutically acceptable excipients include any and all diluents or other liquid vehicles, dispersing or suspending aids, surfactants, isotonic agents, preservatives, lubricants and the like as appropriate for the particular dosage form desired (e.g., injection). General considerations in the formulation and/or manufacture of Pharmaceutical compositions medicaments can be found, for example, in Remington's Pharmaceutical Sciences, sixteenth edition, e.w. martin (Mack Publishing co., Easton, Pa., 1980); and Remington, The Science and Practice of Pharmacy, 21 st edition (Lippincott Williams & Wilkins, 2005).

For example, injectable preparations, such as sterile injectable aqueous suspensions, can be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. Exemplary excipients that may be used include, but are not limited to, water, sterile or phosphate buffered saline, or ringer's solution.

In certain embodiments, the pharmaceutical composition further comprises a cyclodextrin derivative. The most common cyclodextrins are α -, β -and γ -cyclodextrins consisting of 6, 7 and 8 α -1, 4-linked glucose units, respectively, optionally containing one or more substituents on the linked sugar moiety, including (but not limited to) substituted or unsubstituted methylation, hydroxyalkylation, acylation and sulfoalkyl ether substitution. In certain embodiments, the cyclodextrin is a sulfoalkyl ether β -cyclodextrin, e.g., sulfobutyl ether β -cyclodextrin, also known as sulfobutyl ether β -cyclodextrinSee, e.g., U.S.5,376,645. In certain embodiments, the composition comprises hexapropyl- β -cyclodextrin. In a more specific embodiment, the composition comprises hexapropyl- β -cyclodextrin (10-50% in water).

The injectable compositions can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In general, the compounds provided herein are administered in an effective amount. The amount of compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, response, severity of the patient's symptoms, and the like, of the individual patient.

The compositions are presented in unit dosage form to facilitate accurate administration. The term "unit dosage form" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include pre-filled, pre-measured ampoules or syringes of the liquid composition. In these compositions, the compound is typically a minor component (about 0.1% to about 50% or preferably about 1% to about 40% by weight), with the balance being various vehicles or carriers and processing aids that assist in forming the desired dosage form.

The compounds provided herein can be administered as the only active agent, or they can be administered in combination with other active agents. In one aspect, the invention provides a combination of a compound of the invention and another pharmacologically active agent. The combined administration can be carried out by any technique apparent to those skilled in the art including, for example, separate, sequential, simultaneous and alternating administration.

Although the description of the pharmaceutical compositions provided herein is primarily directed to pharmaceutical compositions suitable for administration to humans, it will be understood by those skilled in the art that these compositions are generally suitable for administration to all kinds of animals. Modifications of pharmaceutical compositions suitable for administration to humans to render the compositions suitable for administration to various animals well understood, and veterinary pharmacologists of ordinary skill can design and/or perform such modifications using ordinary experimentation. General considerations in The formulation and/or manufacture of pharmaceutical compositions can be found, for example, in Remington, The Science and Practice of Pharmacy, 21 st edition, Lippincott Williams & Wilkins, 2005.

Methods of use and treatment

As generally described herein, the present invention is directed to C21 substituted neuroactive steroids designed, for example, to act as GABA modulators. In certain embodiments, these compounds are envisioned to be useful as therapeutic agents for inducing anesthesia and/or sedation in a subject. In some embodiments, these compounds are contemplated to be useful as therapeutic agents for treating CNS-related disorders (e.g., sleep disorders, mood disorders, schizophrenia spectrum disorders, spasticity, memory and/or cognitive disorders, movement disorders, personality disorders, autism spectrum disorders, pain, traumatic brain injury, vascular diseases, substance abuse disorders and/or withdrawal syndromes, or tinnitus) in a subject in need thereof (e.g., a subject with Rett syndrome, fragile X syndrome, or anglman syndrome).

Accordingly, in one aspect, the present invention provides a method of inducing sedation and/or anesthesia in a subject comprising administering to the subject an effective amount of a compound of the present invention or a composition thereof. In certain embodiments, the compound is administered by intravenous administration.

Earlier studies (see, e.g., Gee et al, European Journal of Pharmacology,136:419-423(1987)) demonstrated that certain 3 α -hydroxylated steroids were orders of magnitude more potent than others reported as GABA Receptor Complex (GRC) modulators (see, e.g., Majewska et al, Science 232:1004-1007 (1986); Harrison et al, J Pharmacol. Exp. Ther.241:346-353 (1987)). Majewska et al and Harrison et al teach that 3 α -hydroxylated-5-reducing steroids can only have a much lower level of effectiveness. In vitro and in vivo experimental data have now demonstrated that the high potency of these steroids makes them therapeutically effective for modulating brain excitability via GRC (see, e.g., Gee et al, European Journal of Pharmacology,136:419-423 (1987); Wieland et al, Psychopharmacology 118(l):65-71 (1995)).

Various synthetic steroids have also been formulated as neuroactive steroids. See, e.g., U.S. patent 5,232,917, which discloses neuroactive steroid compounds useful in a therapeutically beneficial manner for the treatment of stress, anxiety, insomnia, tic disorders, and mood disorders, which are suitable for GRC active agents, such as depression. Furthermore, it has been previously demonstrated that these steroids interact at unique sites on GRC that are distinct from other known interaction sites (e.g., Barbiturates, Benzodiazepines, and GABA) where therapeutic benefit has previously been elicited for stress, Anxiety, sleep, mood, and tic Disorders (see, e.g., Gee, K.W. and Yamamura, H.I., "Benzodiazepines and Barbiturates: Drugs for The Treatment of The response of Anxiety, Insomnia and Seizeders," Central New Neworvous System Disorders, Horvell eds., Marcel-Dekker, New York (1985), pp. 123-; Lloyd, K.G., and Moelli, P.L., "Psychozelogyne of The drug 147; Loloyd, K.G., and Moelli, P.L.," Psychogel of medicine, J. 183, J. Psychogel, P.183, P.P.J. J. P. J. P. 900, P. J. P. J. P. J. P. J. P. 900, P. J. P. J. P. 3, P. A. 3, D. A. A., P. A. A., D. A. A., D. No. A., D. A., D. No. D. A., D.. These compounds are desirable for their duration, potency and oral activity (as well as other forms of administration).

The compounds of the invention as described herein are generally designed to modulate GABA function and, therefore, act as neuroactive steroids for the treatment and prevention of CNS-related conditions in a subject. Modulation as used herein refers to inhibition or enhancement of GABA receptor function. Thus, the compounds and pharmaceutical compositions provided herein are useful as therapeutic agents for the prevention and/or treatment of CNS conditions in mammals (including human and non-human mammals). Accordingly, and as previously stated, the invention includes within its scope and extends to such methods of treatment, as well as to compounds for use in such methods, and to the use of such compounds for the preparation of medicaments useful in such methods.

Exemplary CNS conditions related to GABA modulation include, but are not limited to, sleep disorders [ e.g., insomnia ], mood disorders [ e.g., depression, dysthymic disorders (e.g., mild depression), bipolar disorder (e.g., I and/or II), anxiety disorders (e.g., Generalized Anxiety Disorder (GAD), social anxiety disorder), stress, post-traumatic stress disorder (PTSD), obsessive compulsive disorder (e.g., Obsessive Compulsive Disorder (OCD)) ], schizophrenia spectrum disorders [ e.g., schizophrenia, schizoaffective disorder ], spasticity [ e.g., epilepsy (e.g., Status Epilepticus (SE)), tics ], memory and/or cognitive disorders [ e.g., attention disorders (e.g., Attention Deficit Hyperactivity Disorder (ADHD)), dementia (e.g., alzheimer-type dementia, lewy body dementia, vascular dementia) ], dementia, Dyskinesia [ e.g., huntington's disease, parkinson's disease ], personality disorder [ e.g., antisocial personality disorder, obsessive-compulsive personality disorder ], Autism Spectrum Disorder (ASD) [ e.g., autism of monogenic origin such as synaptopathy, e.g., Rett syndrome, fragile X syndrome, angler's syndrome ], pain [ e.g., neuropathic pain, injury-related pain syndrome, acute pain, chronic pain ], Traumatic Brain Injury (TBI), vascular disease [ e.g., stroke, ischemia, vascular malformations ], substance abuse disorders and/or withdrawal syndromes [ e.g., opiates, cocaine and/or alcohol addiction ] and tinnitus.

In another aspect, there is provided a combination of a compound of the invention and another pharmacologically active agent. The compounds provided herein can be administered as the sole active agent or they can be administered in combination with other agents. The combined administration can be carried out by any technique apparent to those skilled in the art including, for example, separate, sequential, simultaneous and alternating administration.

In another aspect, there is provided a method of treating or preventing brain excitability in a subject susceptible to or suffering from a condition associated with brain excitability, comprising administering to the subject an effective amount of a compound of the invention.

In another aspect, there is provided a method of treating or preventing stress or anxiety in a subject comprising administering to a subject in need of such treatment an effective amount of a compound of the present invention or a composition thereof.

In another aspect, there is provided a method of reducing or preventing tic activity in a subject comprising administering to a subject in need of such treatment an effective amount of a compound of the invention.

In another aspect, there is provided a method of reducing or preventing insomnia in a subject, comprising administering to a subject in need of such treatment an effective amount of a compound of the present invention or a composition thereof.

In another aspect, there is provided a method of inducing sleep and substantially maintaining the level of REM sleep present in normal sleep, wherein substantial rebound insomnia is not induced, comprising administering an effective amount of a compound of the present invention.

In another aspect, there is provided a method of reducing or preventing PMS or PND in a subject comprising administering to a subject in need of such treatment an effective amount of a compound of the invention.

In another aspect, there is provided a method of treating or preventing a mood disorder in a subject comprising administering to a subject in need of such treatment an effective amount of a compound of the invention. In certain embodiments, the mood disorder is depression.

In another aspect, there is provided a method of inducing anesthesia in a subject comprising administering to the subject an effective amount of a compound of the invention.

In another aspect, there is provided a method of cognitive enhancement or treating memory disorders by administering to the subject a therapeutically effective amount of a compound of the invention. In certain embodiments, the disorder is alzheimer's disease. In certain embodiments, the disorder is Rett syndrome.

In another aspect, there is provided a method of treating attention disorders by administering to the subject a therapeutically effective amount of a compound of the invention. In certain embodiments, the attention disorder is ADHD.

In certain embodiments, the compound is administered chronically to the subject. In certain embodiments, the compound is administered to the subject orally, subcutaneously, intramuscularly, or intravenously.

Anesthesia/sedation

Anesthesia is a state of memory loss, analgesia, hyporesponsiveness, loss of skeletal muscle reflexes, decreased stress response, or both of these simultaneous pharmacologically induced and reversible states. These effects can be obtained from a single drug alone providing the appropriate combination of effects or occasionally with a combination of drugs (e.g., hypnotics, sedatives, paralytics, analgesics) to achieve a very specific combination of results. Anesthesia allows patients to perform surgery and other procedures without suffering and pain that they would otherwise experience.

Sedation is the reduction of irritability or agitation by administration of pharmacological agents, typically to facilitate a medical or diagnostic procedure.

Sedation and analgesia includes the continuum of states of consciousness ranging from minimal sedation (anxiolysis) to general anesthesia.

Minimal sedation is also known as anxiolysis. Minimal sedation is a drug induced state during which the patient responds normally to oral commands. Cognitive function and coordination may be impaired. Ventilation and cardiovascular function are generally unaffected.

Moderate sedation/analgesia (conscious sedation) is a drug-induced loss of consciousness during which the patient intentionally responds to verbal nomenclature, either alone or with mild tactile stimulation. No intervention is usually required to maintain the airway patency. Spontaneous breathing is usually sufficient. Cardiovascular function is normally maintained.

Deep sedation/analgesia is a drug-induced decrease in consciousness during which the patient does not wake easily, but responds intentionally (not a reflex from the retraction of the painful stimulus) after repeated or painful stimuli. The independent ventilation function may be impaired and the patient may need assistance to maintain the airway patent. Spontaneous breathing may be inadequate. Cardiovascular function is normally maintained.

General anesthesia is a drug-induced loss of consciousness during which the patient cannot wake up even for painful stimuli. The ability to maintain independent ventilation functions is often compromised and assistance is often required to maintain airway patency. Positive pressure ventilation may be required due to reduced spontaneous breathing or decreased neuromuscular function caused by drugs. Cardiovascular function may be impaired.

Sedation in the Intensive Care Unit (ICU) results in a decrease in the patient's environmental awareness and its response to external stimuli. It can play a role in the care of critically ill patients and encompasses a wide variety of symptom controls, which will vary from patient to patient and in different individuals throughout their course of disease. Severe sedation in intensive care has been used to promote tracheal intubation tolerance and ventilator synchronization, often with neuromuscular blockers.

In some embodiments, sedation (e.g., long-term sedation, continuous sedation) is induced and maintained in the ICU for an extended period of time (e.g., 1 day, 2 days, 3 days, 5 days, 1 week, 2 weeks, 3 weeks, 1 month, 2 months). Long-term sedatives may have a long duration of action. Sedatives in ICU may have a short elimination half-life.

Procedural sedation and analgesia (also known as conscious sedation) is a technique in which a sedative or dissociating agent is administered in the presence or absence of an analgesic to induce a state that tolerates unpleasant procedures in a subject while maintaining cardiopulmonary function.

Anxiety disorder

Anxiety disorder is a comprehensive term covering several different forms of abnormal and pathological fear and anxiety. Current diagnostic criteria for psychosis identify a wide variety of anxiety disorders.

Generalized anxiety disorder is a common chronic disorder characterized by persistent anxiety that is not focused on any one object or situation. Those suffering from generalized anxiety disorder experience non-specific persistent fear and apprehension and become overly concerned with everyday matters. Generalized anxiety disorder is the most common anxiety disorder affecting the elderly.

In panic, a person suffers from brief episodes of intense fear and apprehension, often marked by tremors, confusion, dizziness, nausea, dyspnea. These panic attacks (defined by APA as fear or discomfort that suddenly occurs and peaks in less than ten minutes) can last for several hours and can be triggered by stress, fear, or even exercise; but the specific reasons are not always obvious. In addition to recurrent and unexpected panic attacks, the diagnosis of panic disorder also requires that the attacks have long-term consequences: the potential impact of worrying episodes, the persistent fear of future episodes, or significant changes in episode-related behavior. Thus, those suffering from panic disorder experience symptoms even beyond a particular panic attack. Often, panic patients notice normal heartbeat changes, causing them to feel that their heart is having problems or they will have another panic attack. In some cases, increased awareness of body functions occurs during panic attacks (oversensing), where any perceived physiological changes are interpreted as a potentially life-threatening disease (i.e., an extremely suspicious condition).

Obsessive-compulsive disorder is a class of anxiety disorders primarily characterized by repetitive compulsions (painful, persistent, and invasive thoughts or images) and impulses (compelling to perform specific actions or rituals). The OCD thinking pattern can be likened to a vague one, as it relates to beliefs in causal relationships that do not exist in reality. Usually the process is completely illogical; for example, a particular pattern of walking forces may be used to mitigate the success of an impending injury. Moreover, in many cases, the forcing is entirely inexplicable, just forcing the completion of the ceremony triggered by the nervousness. In a few cases, OCD patients may only experience findings without significant obtrusiveness; a much smaller number of patients only experience duress.

A single largest category of anxiety disorders is phobias, which includes all cases in which fear and anxiety are triggered by a particular stimulus or situation. Patients often expect dire consequences by encountering objects of their fear, which may be anything from an animal to a location to bodily fluids.

Post-traumatic stress disorder or PTSD is an anxiety disorder resulting from a traumatic experience. Post-traumatic stress can result from extreme conditions, such as war, violence, hostage situations, or even serious accidents. It may also result from long term (chronic) exposure to severe stressors, such as soldiers who endure personal combat but cannot cope with continuous combat. Common symptoms include flashback, avoidance behavior, and depression.

Neurodegenerative diseases and disorders

The term "neurodegenerative disease" includes diseases and disorders associated with progressive loss of structure or function of neurons or neuronal death. Neurodegenerative diseases and disorders include, but are not limited to, alzheimer's disease (including symptoms associated with mild, moderate, or severe cognitive impairment); amyotrophic Lateral Sclerosis (ALS); hypoxic and ischemic injury; ataxia and spasticity (including for the treatment and prevention of convulsions caused by schizoaffective disorders or by drugs used to treat schizophrenia); benign amnesia; cerebral edema; cerebellar ataxia, including McLeod echinocytosis syndrome (MLS); closed head injury; coma; contusion injuries (e.g., spinal cord injury and head injury); dementia, including multi-infarct dementia and senile dementia; disturbance of consciousness; down syndrome; drug-or drug-induced parkinson's disease (e.g. acute akathisia, acute dystonia, parkinson's disease or tardive dyskinesia, neuroleptic malignant syndrome or drug-induced postural tremor); epilepsy; fragile X syndrome; gilles de la Tourette syndrome; head trauma; hearing impairment and loss; huntington's disease; lennox syndrome; movement disorders caused by levodopa; mental retardation; dyskinesias including akinesia and akinesia (rigidity) syndromes (including basal ganglia calcification, corticobasal degeneration, multiple system atrophy, parkinson's disease-ALS dementia complex, parkinson's disease, postencephalitic parkinson's disease and progressive supranuclear palsy); muscle spasms and conditions associated with muscle spasms or weakness, including chorea (e.g., benign hereditary chorea, drug-induced chorea, hemiballism, huntington's disease, acanthocytosis, Sydenham's chorea, and symptomatic chorea), movement disorders (including twitching such as complex twitching, simple twitching, and symptomatic twitching), myoclonus (including systemic myoclonus and focal myoclonus), tremors (e.g., resting tremor, postural tremor, and intention tremor), and dystonias (including longitudinal dystonia, dystonia writer's cramp, hemiplegic dystonia, paroxysmal dystonia, and focal dystonia such as blepharospasm, oromandibular dystonia, and spasmodic dysarthria and torticollis); neuronal damage including eye damage, retinopathy or macular degeneration of the eye; neurotoxic damage following cerebral stroke, thromboembolic stroke, hemorrhagic stroke, cerebral ischemia, cerebral vasospasm, hypoglycemia, amnesia, hypoxia, perinatal asphyxia, and cardiac arrest; parkinson's disease; twitching; status of epilepsy persistence; stroke; tinnitus; tuberous sclerosis, and neurodegenerative diseases caused by viral infection (e.g., caused by Acquired Immune Deficiency Syndrome (AIDS) and encephalopathy). Neurodegenerative disorders also include, but are not limited to, neurotoxic injury following cerebral stroke, thromboembolic stroke, hemorrhagic stroke, cerebral ischemia, cerebral vasospasm, hypoglycemia, amnesia, hypoxia, anoxia, perinatal asphyxia, and cardiac arrest. The method of treating or preventing a neurodegenerative disease further comprises treating or preventing loss of neuronal function characteristic of a neurodegenerative disorder.

Epilepsy

Epilepsy is a disorder of the brain characterized by recurrent convulsions over time. Epilepsy types may include, but are not limited to, generalized epilepsy, such as childhood absence epilepsy, juvenile myoclonic epilepsy, epilepsy with grand mal tics on waking, West syndrome, Lennox-Gastaut syndrome; partial epilepsy, e.g., temporal lobe epilepsy, frontal lobe epilepsy, benign focal epilepsy in children.

Status Epilepticus (SE)

Status Epilepticus (SE) may include, for example, convulsive status epilepticus, e.g., early status epilepticus, established status epilepticus, refractory status epilepticus, super-refractory status epilepticus; non-convulsive status epilepticus, e.g., generalized status epilepticus, complex partial status epilepticus; generalized periodic epileptiform discharges; and periodic unilateral epileptiform discharges. Convulsive status epilepticus is characterized by the presence of convulsive status epileptic tics, and may include early status epilepticus, established status epilepticus, refractory status epilepticus, super-refractory status epilepticus. Early status epilepticus is treated with first-line therapy. It has been established that status epilepticus is characterized by convulsions of status epilepticus that persist despite treatment with first line therapy, and a second line therapy is administered. Refractory status epilepticus is characterized by persistent status epilepticus tics despite treatment with first-line and second-line therapies, and is typically administered with general anesthesia. Ultra-refractory status epilepticus is characterized by status epilepticus tics that persist despite treatment with first-line therapy, second-line therapy, and general anesthesia for 24 hours or more.

Nonconvulsive status epilepticus may include, for example, focal nonconvulsive status epilepticus, e.g., complex partial nonconvulsive status epilepticus, simple partial nonconvulsive status epilepticus, subtle nonconvulsive status epilepticus; generalized nonconvulsive status epilepticus, e.g., tardive nonconvulsive status epilepticus, atypical nonconvulsive status epilepticus, or typical nonconvulsive status epilepticus.

The compositions described herein may also be administered as a prophylactic agent to a subject prior to the onset of tics with: CNS disorders, e.g., traumatic brain injury; status epilepticus, e.g., convulsive status epilepticus, e.g., early status epilepticus, established status epilepticus, refractory status epilepticus, super-refractory status epilepticus; non-convulsive status epilepticus, e.g., generalized status epilepticus, complex partial status epilepticus; generalized periodic epileptiform discharges; and periodic unilateral epileptiform discharges.

Twitching of the stomach

Tics are physical consequences or behavioral changes that occur following episodes of abnormal electrical activity in the brain. The term "twitch" is often used interchangeably with "spasm". When a person's body is shaken rapidly and uncontrollably, i.e. cramps. During spasticity, a person's muscles contract and relax repeatedly.

Tics are divided into two broad categories based on the type of behavior and brain activity: systemic and partial (also known as local or focal). Classifying seizure types helps physicians diagnose whether a patient has epilepsy.

Systemic tics are produced by electrical impulses from the entire brain, while partial tics are produced (at least initially) by electrical impulses in a relatively small part of the brain. The part of the brain that produces tics is sometimes referred to as the lesion.

There are six types of systemic tics. The most common and severe and therefore the best known is a generalized spasm, also known as a seizure. In this type of twitch, the patient loses consciousness and often collapses. Loss of consciousness is followed by a general physical stiffness (the "tonic" phase known as tics) lasting 30 to 60 seconds, followed by a violent jolt ("clonic" phase) lasting 30 to 60 seconds, after which the patient goes to deep sleep ("post-seizure" or post-tic phase). During a large seizure, injuries and accidents, such as tongue biting and urinary incontinence, may occur.

Absence tics cause a brief loss of consciousness (for only a few seconds), with little or no symptoms. Patients (most commonly children) often discontinue activity and stare at emotionally. These twitches begin and end abruptly and may occur several times a day. Patients are generally unaware that they have twitches, but they may realize "lost time".

Myoclonic tics consist of occasional tics, usually on both sides of the body. Patients sometimes refer to tics as brief shocks. When severe, these twitches can lead to a fall or involuntary throw.

Clonic tics are repetitive, rhythmic spasms that involve both sides of the body.

Tetanic tics are characterized by stiffness of the muscle.

Dystonic tics consist of a sudden and widespread loss of muscle tone, especially in the arms and legs, which often leads to falls.

Tics described herein may include epileptic tics; acute recurrent convulsions; cluster tics; successive twitching; uninterrupted convulsions; long-term convulsions; recurrent tics; status epilepticus tics, e.g., refractory spastic status epilepticus, non-spastic status epilepticus tics; refractory tics; myoclonic tics; tetanic tic; tonic-clonic tics; simple partial tics; complex partial tics; secondary systemic convulsions; atypical absence tic; loss of spirit and convulsions; dystonic tics; benign Rolandic zone tics; febrile convulsions; emotional convulsions; focal convulsions; dementia tic; generalized paroxysmal tic; infantile spasms; jacksonian tics; large scale bilateral myoclonic tic tics; multifocal convulsions; seizures in neonates; twitching at night; twitching occipital lobe; post-traumatic tic; micro twitching; sylvan tics; twitching of the visual reflex; or withdrawal of tics.

Equivalents and ranges

In the claims, articles such as "a" and "the" may refer to one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include "or" between one or more members of the group are deemed satisfactory if one, more than one, or all of the group members are present in, used in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The invention includes embodiments in which exactly one member of the group is present in, used in, or otherwise relevant to a given product or process. The invention includes embodiments in which more than one, or all of the group members are present in, used in, or otherwise relevant to a given product or process.

Furthermore, this disclosure encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, and descriptive terms from one or more of the listed claims are introduced into another claim. For example, any claim that depends from another claim may be amended to include one or more limitations that exist in any other claim that depends from the same base claim. Where elements are presented in a list (e.g., in markush group format), each subgroup of the elements is also disclosed, and any element can be removed from the group. It should be understood that, in general, where the invention or aspects of the invention are referred to as comprising particular elements and/or features, certain embodiments of the invention or aspects of the invention consist of, or consist essentially of, those elements and/or features. For the purpose of simplicity, those embodiments are not explicitly set forth in these terms herein. It is further noted that the terms "comprising" and "containing" are intended to be open-ended and allow for the inclusion of additional elements or steps. Where ranges are given, the endpoints are inclusive. Furthermore, unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges in different embodiments of the invention can assume any specific value or subrange within the range, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.

This application refers to various issued patents, published patent applications, journal articles and other publications, all of which are incorporated herein by reference. In the event of a conflict between any of the incorporated references and this specification, the present specification shall control. In addition, any particular embodiment of the invention that falls within the prior art may be explicitly excluded from any one or more claims. Because these embodiments are considered to be known to those of ordinary skill in the art, they may be excluded even if the exclusion is not explicitly set forth herein. Any particular embodiment of the invention may be excluded from any claim for any reason, whether or not related to the presence of prior art.

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments described herein. The scope of the embodiments described herein is not intended to be limited by the foregoing description, but is instead set forth in the following claims. It will be understood by those of ordinary skill in the art that various changes and modifications may be made to the present specification without departing from the spirit or scope of the present invention as defined in the following claims.

Examples

In order that the invention described herein may be more fully understood, the following examples are set forth. The synthetic and biological examples described in this application are provided to illustrate the compounds, pharmaceutical compositions and methods provided herein and should not be construed as limiting the scope thereof in any way.

Materials and methods

The compounds provided herein can be prepared from readily available starting materials using the following general methods and procedures. It is to be understood that where typical or preferred process conditions (i.e., reaction temperatures, times, molar ratios of reactants, solvents, pressures, etc.) are given, other process conditions may also be used unless otherwise specified. Optimal reaction conditions may vary with the particular reactants or solvents used, but those skilled in the art can determine such conditions by routine optimization.

In addition, as will be apparent to those skilled in the art, conventional protecting groups may be required to prevent certain functional groups from undergoing undesirable reactions. The selection of protecting groups suitable for particular functional groups and conditions suitable for protection and deprotection are well known in the art. For example, numerous Protecting groups, as well as their introduction and removal, are described in t.w.greene and p.g.m.wuts, Protecting groups in Organic Synthesis, second edition, Wiley, New York,1991, and references cited therein.

The compounds provided herein can be isolated and purified by known standard procedures. These procedures include, but are not limited to, recrystallization, column chromatography, HPLC, or Supercritical Fluid Chromatography (SFC). The following schemes are presented in detail to prepare representative heteroaryl and heterocyclyl groups that have been listed herein. The compounds provided herein can be prepared by those skilled in the art of organic synthesis from known or commercially available starting materials and reagents. Exemplary chiral columns useful in the separation/purification of enantiomers/diastereomers provided herein include (but are not limited to)AD-10、OB、OB-H、OD、OD-H、OF、OG、OJ andOK。

herein reported1H-NMR (e.g., with respect to an intermediate) can be part of a complete NMR spectrum of a compound (e.g., a compound described herein). For example, reported1H NMR can exclude regions between about 1 and about 2.5ppm delta (ppm).

Exemplary general procedure for preparative HPLC: column: waters RBridge preparation 10. mu. m C18, 19X 250 mm. Mobile phase: acetonitrile, water (NH)4HCO3) (30L of water, 24g of NH)4HCO3,30mL NH3.H2O). Flow rate: 25 mL/min.

Exemplary general procedure for analytical HPLC: mobile phase: a: water (10mM NH)4HCO3) And B: gradient of acetonitrile: 5% -95% B in 1.6 or 2 minutes. Flow rate: 1.8 or 2 mL/min; column: xbridge C18, 4.6X 50mm, 3.5 μm at 45 ℃.

Synthesis method

Example 1 general procedure a: preparation of A/B-trans skeleton

Step 1. preparation of compound a 2. Finely ground potassium hydroxide (28.0g, 165mmol) was added to a solution of commercially available 19-hydroxyandrost-4-ene-3, 17-dione (A1, 50.0g, 165mmol) in dry 1, 2-dimethoxyethane (500mL) at 0 ℃ under nitrogen, after which methyl sulfate (43.7g, 208mmol) was added in portions. The mixture was slowly warmed to room temperature and stirred for a total of 18 hours, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was diluted with water (500mL) and extracted with ethyl acetate (3X 200 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (100mL), dried over anhydrous sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (2:1) to give a2(26.8g, 50%) as a yellow solid.

Step 2. preparation of compound a 3. Triethyl orthoformate (6.2mL, 37mmol) and p-toluenesulfonic acid (400mg, 9.3mmol) were added to a solution of compound a2(9.9g, 31.0mmol) in anhydrous 1, 4-dioxane (40mL) and anhydrous ethanol (30mL) at room temperature under nitrogen and the mixture was stirred for 1.5 hours when TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated the reaction was complete. The mixture was diluted with saturated aqueous sodium bicarbonate (100mL), poured into water (300mL) and extracted with ethyl acetate (3X 100 mL). The combined organic extraction solvents were removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (2:1) to give compound a3(7.0g, 66%) as a white solid.

Step 3. preparation of compound a 4. A mixture of compound a3(7.0g, 20.3) and palladium on carbon (3.0g, 10 wt%) in anhydrous ethyl acetate (200mL) was shaken at room temperature under an atmosphere of hydrogen (1 atm) for 1 hour, at which time TLC analysis of the mixture (2:1 hexane/ethyl acetate) indicated that the reaction was complete. The atmosphere was switched to nitrogen and the mixture was filtered under reduced pressure through a pad of Celite, washing the filter cake with ethyl acetate (50 mL). The filtrate solvent was treated with 10% aqueous hydrochloric acid (100mL) and the biphasic mixture was stirred for 30 minutes. The mixture was extracted with ethyl acetate (2 × 100mL) and the combined organic extracts were washed sequentially with saturated aqueous sodium bicarbonate and saturated aqueous sodium chloride (50mL each), dried over anhydrous sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (4:1) to give compound a4(3.9g, 60%) as a colorless oil.

Step 4. preparation of compound a 5. Sodium hydride (1.7g, 45mmol, 60% in mineral oil) was added portionwise to a solution of trimethyl sulphoxide iodide (9.1g, 45mmol) in anhydrous dimethyl sulphoxide (100mL) at room temperature under nitrogen and the mixture was stirred for 1 hour after which a solution of compound a4(9.5g, 29.8mmol) in anhydrous dimethyl sulphoxide (100mL) was added. The resulting mixture was stirred at room temperature for 12 hours, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was diluted with water (500mL) and extracted with methyl tert-butyl ether (2X 300 mL). The combined organic extracts were washed with water (2 × 300mL), dried over anhydrous magnesium sulfate and filtered. The solvent was removed under reduced pressure to give compound a5 as a colorless oil, which was used in the next step without further purification (7.5g, 76%).

Step 5. preparation of Compound A6. Lithium aluminum hydride (67mL, 67mmol, 1M tetrahydrofuran solution) was added to a solution of crude compound a5(7.5g, 22.2mmol) in anhydrous tetrahydrofuran (5mL) at 0 ℃ under nitrogen, after which the mixture was slowly warmed to room temperature and stirred for a total of 2 hours, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was carefully treated with water (10mL) and saturated aqueous sodium chloride (30mL) in that order and extracted with ethyl acetate (3X 50 mL). The combined organic extracts were dried over anhydrous magnesium sulfate, filtered and the solvent removed under reduced pressure to give compound a6 as a colorless oil which was used in the next step without further purification (5.5g, 74%): LCMS M/z 319[ M + H-H ]2O]+

Step 6. preparation of compound a 7. Piromazine chlorochromate (4.0g, 19mmol) was added in portions to a solution of crude compound A6(4.2g, 12.5mmol) in dry dichloromethane (100mL) at 0 ℃ under nitrogen. The mixture was slowly warmed to room temperature and stirred for a total of 3 hours, at which timeTLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated the reaction was complete. The solids were removed by filtration and the filtrate solvent was removed under reduced pressure. The residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (7:3) to give compound a7(2.1g, 50%) as a light yellow solid: LCMS M/z 317[ M + H-H ]2O]+

Step 7. preparation of Compound A8. Potassium tert-butoxide (4.3g, 38mmol) was added to a mixture of ethyltriphenylphosphonium bromide (14.2g, 38mmol) in anhydrous tetrahydrofuran (30mL) at room temperature under nitrogen, after which the mixture was heated to 80 ℃ and stirred for 1 hour. A solution of compound A7(3.1g, 9.3mmol) in dry tetrahydrofuran (10mL) was added, after which stirring was continued at 80 ℃ for 2 hours, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The cooled mixture was diluted with water (30mL) and saturated aqueous sodium chloride (20mL) and extracted with ethyl acetate (2X 100 mL). The combined organic extraction solvents were removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (7:3) to give compound A8(2.0g, 66%) as an off-white solid: LCMS M/z 329[ M + H-H ]2O]+

Step 8. preparation of Compound A9. Borane-tetrahydrofuran complex (20.0mL, 20mmol, 1M tetrahydrofuran solution) was added to a solution of compound A8(2.0g, 5.8mmol) in anhydrous tetrahydrofuran (15mL) under nitrogen at 0 ℃, after which the mixture was slowly warmed to room temperature and stirred for a total of 1 hour. The mixture was cooled in an ice bath and a 10% aqueous sodium hydroxide solution (12mL) was slowly added followed by 30% aqueous hydrogen peroxide solution (12 mL). The resulting mixture was warmed to room temperature and stirred for 1 hour, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was extracted with dichloromethane (2 × 100mL) and the combined organic extracts were washed with saturated aqueous sodium chloride (25mL), dried over sodium sulfate and filtered. The solvent was removed under reduced pressure to give crude compound a9 as a white solid which was used in the next step without further purification (2.5g, > 99%).

Step 9. preparation of Compound A10. Piromazine chlorochromate (2.4g, 11mmol) was added in portions to a solution of crude compound A9(2.5g, 6.9mmol) in dry dichloromethane (30mL) at 0 ℃ under nitrogen. The mixture was slowly warmed to room temperature and stirred for a total of 2 hours, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The solids were removed by filtration and the filtrate solvent was removed under reduced pressure. The residue was purified by column chromatography on silica gel eluting with heptane/ethyl acetate (7:3) to give a10(1.5g, 61%) as an off white solid.

Step 10. preparation of Compound A11. Hydrogen bromide (3 drops, 48% in water) was added to a solution of a10(1.4g, 3.9mmol) in dry methanol (150mL) under nitrogen at room temperature in the dark, after which bromine (0.4mL, 7.7mmol) was added. The mixture was stirred for 1 hour, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was poured into ice water (100mL), treated with saturated aqueous sodium bicarbonate (30mL) and extracted with ethyl acetate (2X 60 mL). The combined organic extracts were washed with saturated aqueous sodium bicarbonate (4 × 100mL) and saturated aqueous sodium chloride (50mL), dried over magnesium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (1:1) to give compound a11(1.2g, 71%) as a colorless semi-solid: LCMS M/z 441[ M + H ]]+

Example 2 general procedure a: preparation of A/B-trans skeleton

Step 1. preparation of compound a 12. Prepared according to general procedure a, step 1 from a1(10.0g, 33mmol) and ethyl sulfate (17.3mL, 132mmol) and purified by silica gel column chromatography to give compound a12(4.6g, 42%) as a yellow oil.

Step 2. preparation of compound a 13. Prepared according to general procedure a, step 2, from compound a12(4.6g, 14mmol) to give crude compound a13 as a yellow oil, which was used in the next step without further purification.

Step 3. preparation of compound a 14. Prepared according to general procedure a, step 3 from crude compound a13, purified by silica gel column chromatography to give compound a14(1.5g, 31%) as a yellow oil.

Step 4. preparation of compound a 15. Prepared according to general procedure a, step 4, from compound a14(1.7g, 5.1mmol) to give crude compound a15 as a yellow oil, which was used in the next step without further purification.

Step 5. preparation of Compound A16. Prepared according to general procedure a, step 5 from crude compound a15 to give crude compound a16 as a yellow oil, which was used in the next step without further purification.

Step 6. preparation of compound a 17. Prepared according to general procedure a, step 6 from crude compound a16 and purified by silica gel column chromatography to give compound a17(751mg, 40%) as an off-white solid.

Step 7. preparation of Compound A18. Prepared according to general procedure a, step 7 from compound a17(750mg, 2.2mmol) and purified by silica gel column chromatography to give compound a18(757mg, 97%) as a colorless oil.

Step 8. preparation of Compound A19. Prepared according to general procedure a, step 8, from compound a18(757mg, 2.1mmol) to give crude compound a19 as a yellow oil, which was used in the next step without further purification.

Step 9. preparation of A20. Prepared according to general procedure a, step 9 from crude compound a19, purified by silica gel column chromatography to give a20(515mg, 65%) as a white solid: mp106-107 ℃;1HNMR(500MHz,CDCl3)δ3.51(d,J=16.5Hz,1H),3.43-3.36(m,3H),2.53(t,J=5.0Hz,1H),2.18-1.96(m,6H),1.74-0.92(m,25H),0.84-0.82(m,1H),0.62(s,3H)ppm;ESI MS m/z 359[M+H-H2O]+

step 10. preparation of A21. Hydrogen bromide (10 drops, 48% in water) was added to a solution of A20(490mg, 1.30mmol) in dry methanol (40mL) under nitrogen at room temperature in the darkTo the solution, bromine (235mg, 13.0mmol) was added thereafter. The mixture was stirred for 1 hour, at which time TLC analysis of the mixture (7:3 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was poured into ice water (100mL), treated with saturated aqueous sodium bicarbonate (30mL) and extracted with ethyl acetate (2X 60 mL). The combined organic extracts were washed with saturated aqueous sodium bicarbonate (4 × 100mL) and saturated aqueous sodium chloride (50mL), dried over magnesium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with heptane/ethyl acetate (1:1) to give compound a21(468mg, 79%) as a white solid. LCMS M/z 437[ M + H-H2O]+

Example 3. general procedure B: preparation of A/B-trans skeleton C-21 analogue

Reacting 5-chloro-1H-benzo [ d ] at room temperature under nitrogen][1,2,3]Triazole (470mg, 3.06mmol) and potassium carbonate (704mg, 5.1mmol) were added to a solution of compound a11(225mg, 0.51mmol) in anhydrous tetrahydrofuran (20mL) and the mixture was stirred for 16 hours at which time TLC analysis of the mixture (2:1 hexane/ethyl acetate) indicated that the reaction was complete. The mixture was diluted with water (120mL) and extracted with ethyl acetate (3X 100 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (60mL), dried over sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was semi-purified by silica gel column chromatography eluting with hexane/ethyl acetate (3:1) to give a mixture of the three regioisomers. The residue was further purified by reverse phase preparative HPLC to give 35(150mg, 29%) as an off white solid: mp 205-;1H NMR(300MHz,CDCl3)δ7.87(dd,J=1.8,0.6Hz,1H),7.81(dd,J=9.0,0.6Hz,1H),7.34(dd,J=9.0,1.8Hz,1H),5.55(d,JAB=17.1Hz,1H),5.46(d,JAB=17.1Hz,1H),3.48(d,J=9.9Hz,1H),3.38(d,J=10.2Hz,1H),3.30(s,3H),2.66(t,J=8.7Hz,1H),2.30-2.18(m,1H),2.18-2.09(m,1H),2.09-2.00(m,1H),1.82-1.38(m,11H),1.38-1.06(m,10H),1.06-0.92(m,1H),0.92-0.80(m,1H),0.76(s,3H)ppm;ESI MS m/z 514[M+H]+

further elution afforded 36(86mg, 17%) as an off-white solid: mp 97-101 ℃;1H NMR(300MHz,CDCl3)δ8.03-7.97(m,1H),7.36-7.31(m,2H),5.42(d,JAB=18.3Hz,1H),5.34(d,JAB=18.0Hz,1H),3.49(d,J=9.9Hz,1H),3.38(d,J=9.9Hz,1H),3.31(s,3H),2.72(t,J=8.7Hz,1H),2.30-2.10(m,2H),2.10-2.00(m,1H),1.85-1.40(m,11H),1.39-0.80(m,12H),0.75(s,3H)ppm;ESI MS m/z 5140[M+H]+

further elution afforded 37(112mg, 21%) as an off-white solid: mp 106-;1H NMR(300MHz,CDCl3)δ8.06(d,J=1.2Hz,1H),7.45(dd,J=9.0,1.5Hz,1H),7.27(d,J=8.7Hz,1H),5.41(s,2H),3.49(d,J=9.9Hz,1H),3.38(d,J=10.2Hz,1H),3.30(s,3H),2.71(t,J=8.7Hz,1H),2.29-2.00(m,3H),1.83-1.44(m,11H),1.44-0.82(m,12H),0.74(s,3H)ppm;ESI MS m/z 514[M+H]+

example 4. preparation of compound 8.

Prepared according to general procedure B from compound a11(50mg, 0.114mmol) and N-methylpiperazine (227mg, 2.27mmol) and purified by reverse phase preparative HPLC to give compound 8(36.6mg, 70%) as a white solid: mp 136-;1HNMR(500MHz,CDCl3)δ3.46(d,J=10.0Hz,1H),3.36(d,J=10.0Hz,1H),3.28(s,3H),3.17(s,2H),2.60-2.51(m,8H),2.30(s,3H),2.18-2.14(m,1H),2.02(dt,J=13.0,3.5Hz,1H),1.88(dt,J=12.0,3.5Hz,1H),1.71-1.46(m,10H),1.34-1.72(m,11H),0.98-0.84(m,1H),0.84-0.80(m,1H),0.64(s,3H)ppm;ESI MS m/z 461[M+H]+

example 5 preparation of compounds 3 and 1.

According toGeneral procedure B was prepared from compound a11(300mg, 0.67mmol) and 1H-1,2, 3-triazole (188mg, 2.71mmol), and purified by reverse phase preparative HPLC to give compound 3(36.6mg, 70%) as a white solid: mp 72-74 ℃;1HNMR(300MHz,CDCl3)δ7.75(d,J=1.2Hz,1H),7.63(d,J=1.2Hz,1H),5.20(q,J=18.0Hz,2H),3.46(d,J=9.9Hz,1H),3.37(d,J=10.2Hz,1H),2.65(t,J=9.0Hz,1H),2.09-2.01(m,1H),1.78-1.68(m,4H),1.60-1.49(m,13H),1.46-1.26(m,9H),1.23-0.87(m,2H),0.63(s,3H)ppm;ESI MS m/z 430[M+H]+

further elution afforded compound 1 as an off-white solid (110mg, 38%): mp 157-150 ℃;1HNMRδ7.67(s,2H),5.23(q,J=17.0Hz,2H),3.47(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.28(s,3H),2.57(t,J=9.5Hz,1H),2.24-2.17(m,1H),2.09-2.01(m,2H),1.75-1.67(m,4H),1.62-1.47(m,10H),1.38-1.23(m,3H),1.23-1.08(m,4H),1.02-0.92(m,1H),0.86-0.81(m,1H),0.73(s,3H)ppm;ESI MS m/z 430[M+H]+

example 6. preparation of compound 13.

Prepared according to general procedure B from compound a11(25mg, 0.057mmol) and 5-chlorotriazole (106mg, 1.14mmol) and purified by reverse phase preparative HPLC to give compound 13(16.8mg, 65%) as a white solid: mp 141-142 ℃;1HNMR(500MHz,CDCl3)δ7.85(s,1H),7.80(s,1H),4.95(dd,J=62.5,17.5Hz,2H),3.47(dd,J=10.0Hz,1H),3.37(dd,J=10.0Hz,1H),3.28(s,3H),2.60(t,J=9.0Hz,1H),2.23-2.20(m,1H),2.05-2.01(m,2H),1.76-1.69(m,4H),1.63-1.49(m,7H),1.47-1.10(m,10H),0.99-0.97(m,1H),0.87-0.85(m,1H),0.69(s,3H)ppm;ESI MS m/z 436[M+H-H2O]+

example 7. preparation of compounds 14 and 15.

Prepared according to general procedure B from compound a11(60mg, 0.14mmol) and tetrazole (57mg, 0.81mmol) by semi-purification via silica gel column chromatography and reverse phase preparative HPLC to give compound 15(6mg, 10%) as an off-white solid: mp 88-91 ℃;1H NMR(500MHz,CDCl3)δ8.73(s,1H),5.30(d,JAB=18.5Hz,1H),5.17(d,JAB=18.5Hz,1H),3.47(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.29(s,3H),2.66(t,J=9.0Hz,1H),2.28-2.20(m,1H),2.07-2.00(m,2H),1.82-1.69(m,4H),1.65-1.40(m,7H),1.35-1.09(m,10H),1.04-0.95(m,1H),0.92-0.83(m,1H),0.69(s,3H)ppm;ESI MS m/z 431[M+H]+

further elution afforded compound 14(7mg, 12%) as an off-white solid: mp72-75 deg.C;1H NMR(500MHz,CDCl3)δ8.56(s,1H),5.47(d,JAB=17.0Hz,1H),5.42(d,JAB=17.5Hz,1H),3.47(d,J=10.5Hz,1H),3.38(d,J=10.0Hz,1H),3.29(s,3H),2.64(t,J=9.0Hz,1H),2.27-2.19(m,1H),2.18-2.00(m,2H),1.80-1.68(m,4H),1.66-1.46(m,6H),1.44-1.37(m,1H),1.35-1.08(m,10H),1.04-0.94(m,1H),0.90-0.83(m,1H),0.74(s,3H)ppm;ESI MS m/z 431[M+H]+

example 8 preparation of compounds 16 and 17.

Prepared according to general procedure B from compound a11(215mg, 0.49mmol) and 5-methyl-1H-tetrazole (253mg, 2.92mmol), followed by silica gel column chromatography and reverse phase preparative HPLC semi-purification to give 16 as an off-white solid (56mg, 26%): mp 88-91 ℃;1H NMR(300MHz,CDCl3)δ5.13(d,JAB=18.0Hz,1H),5.06(d,JAB=18.0Hz,1H),3.48(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.29(s,3H),2.66(t,J=9.0Hz,1H),2.47(s,3H),2.27-2.15(m,1H),2.08-1.98(m,2H),1.85-1.38(m,11H),1.37-0.95(m,11H),0.91-0.83(m,1H),0.70(s,3H)ppm;ESI MS m/z 445[M+H]+

further eluting to obtain off-white solidBulk 17(95mg, 44%): mp 71-74 ℃;1H NMR(500MHz,CDCl3)δ5.37(d,JAB=17.0Hz,1H),5.32(d,JAB=17.5Hz,1H),3.47(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.29(s,3H),2.62(t,J=9.0Hz,1H),2.56(s,3H),2.26-2.18(m,1H),2.09-2.00(m,2H),1.80-1.68(m,4H),1.65-1.46(m,6H),1.43-1.08(m,11H),1.04-0.94(m,1H),0.90-0.82(m,1H),0.73(s,3H)ppm;ESI MS m/z 445[M+H]+

example 9. preparation of compound 18.

Prepared according to general procedure B from compound a11(21mg, 0.047mmol) and 4-chloro-1H-pyrazole (29mg, 0.28mmol) by silica gel column chromatography and reverse phase preparative HPLC semi-purification to give 18 as an off-white solid (10mg, 46%): mp 100-;1H NMR(500MHz,CDCl3)δ7.45(s,1H),7.40(s,1H),4.90(d,JAB=17.5Hz,1H),4.80(d,JAB=17.5Hz,1H),3.46(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.28(s,3H),3.13(s,3H),2.57(t,J=9.0Hz,1H),2.26-2.16(m,1H),2.07-1.98(m,2H),1.76-1.67(m,4H),1.63-1.46(m,6H),1.41-1.07(m,11H),1.03-0.92(m,1H),0.91-0.80(m,1H),0.69(s,3H)ppm;ESI MS m/z 463[M+H]+

example 10. preparation of compound 19.

Prepared according to general procedure B from compound a11(30mg, 0.06mmol) and 1H-pyrazole-3-carbonitrile (25mg, 0.03mmol), followed by silica gel column chromatography and preparative reverse phase HPLC to give 19 as an off-white solid (17mg, 56%): mp 115-120 ℃;1HNMR(300MHz,CDCl3)δ7.47(d,J=2.4Hz,1H),6.72(d,J=2.4Hz,1H),5.00(q,J=18.0Hz,2H),3.49(d,J=9.9Hz,1H),3.39(d,J=10.2Hz,1H),3.29(s,3H),2.65(t,J=9.0Hz,1H),2.09-2.01(m,1H),1.78-1.68(m,4H),1.60-1.49(m,10H),1.46-1.26(m,9H),1.23-0.87(m,2H),0.63(s,3H)ppm;ESI MS m/z436[M+H-H2O]+

example 11 preparation of compound 20.

Prepared according to general procedure B from compound a11(130mg, 0.29mmol) and 4-methyl-1H-pyrazole (247mg, 3.01mmol) with displacement to cesium carbonate (480mg, 1.5mmol) in anhydrous acetonitrile (8mL), followed by silica gel column chromatography and reverse phase preparative HPLC semi-purification to give 20(15mg, 11%) as an off-white solid: mp 67-71 ℃;1H NMR(500MHz,CDCl3)δ7.33(s,1H),7.16(s,1H),4.87(d,JAB=18.0Hz,1H),4.79(d,JAB=17.5Hz,1H),3.46(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.28(s,3H),3.13(s,3H),2.56(t,J=8.5Hz,1H),2.23-2.15(m,1H),2.09(s,3H),2.07-2.00(m,2H),1.75-1.65(m,4H),1.62-1.45(m,6H),1.40-1.08(m,11H),1.02-0.94(m,1H),0.89-0.82(m,1H),0.69(s,3H)ppm;ESI MS m/z 443[M+H]+

example 12 preparation of compound 21.

Prepared according to general procedure B from compound a11(50mg, 0.13mmol) and 2-fluoroimidazole hydrochloride (75mg, 0.61mmol) substituted with cesium carbonate (200mg, 0.62mmol) in anhydrous acetonitrile (4mL), followed by silica gel column chromatography and reverse phase preparative HPLC semi-purification to afford 21(35mg, 69%) as a yellow solid: mp 78-81 ℃;1H NMR(300MHz,CDCl3)δ6.91(d,J=1.5Hz,1H),6.64(s,J=1.5Hz,1H),5.54(d,JAB=18.3Hz,1H),4.41(d,JAB=18.0Hz,1H),3.48(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.30(s,3H),2.54(t,J=9.0Hz,1H),2.28-2.14(m,1H),2.09-1.98(m,2H),1.80-1.64(m,4H),1.64-1.46(m,6H),1.44-0.80(m,13H),0.69(s,3H)ppm;ESI MS m/z 470[M+H]+

example 13 preparation of compound 23.

Prepared according to general procedure B from compound a11(75mg, 0.17mmol) and 4-methylimidazole (279mg, 3.4mmol) by semi-purification via silica gel column chromatography and reverse phase preparative HPLC to give 23(18mg, 24%) as a white solid: mp 87-89 ℃;1H NMR(500MHz,CDCl3)δ7.40(s,1H),6.56(s,1H),4.63(dd,J=18.0,10.8Hz,2H),3.47(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.29(s,3H),2.60-2.54(m,1H),2.24-2.17(m,3H),2.08(s,1H),2.04-2.01(m,1H),1.96-1.93(m,1H),1.76-1.68(m,4H),1.63-1.46(m,6H),1.42-1.09(m,11H),1.02-0.93(m,1H),0.87-0.82(m,1H),0.68(s,3H)ppm;APCI MS m/z 443[M+H]+

example 14. preparation of compound 24.

Prepared according to general procedure B from compound a11(100mg, 0.23mmol) and 4- (methylsulfonyl) -1H-pyrazole (99mg, 0.68mmol) by semi-purification via silica gel column chromatography and reverse phase preparative HPLC to give 24 as a white solid (6mg, 5%): mp 90-92 ℃;1H NMR(500MHz,CDCl3)δ7.91(s,1H),7.86(s,1H),5.02(d,JAB=18.0Hz,1H),4.90(d,JAB=17.5Hz,1H),3.47(d,J=10.0Hz,1H),3.37(d,J=10.0Hz,1H),3.28(s,3H),3.13(s,3H),2.61(t,J=9.0Hz,1H),2.26-2.16(m,1H),2.07-2.00(m,2H),1.78-1.68(m,4H),1.64-1.46(m,6H),1.44-1.36(m,1H),1.35-1.08(m,10H),1.04-0.94(m,1H),0.89-0.82(m,1H),0.69(s,3H)ppm;APCI MS m/z 507[M+H]+

example 15 preparation of compounds 25 and 26.

Prepared according to general procedure B from compound a21(150mg, 0.33mmol) and 5-methyltetrazole (554mg, 6.6mmol) and purified by reverse phase preparative HPLC to give 25(43.6mg, 28%) as a white solid: mp 71-72 ℃;1HNMR(500MHz,CDCl3)δ5.34(dd,J=26.5,17.5Hz,2H),3.52(d,J=10.0Hz,1H),3.42-3.38(m,3H),2.62(t,J=9.0Hz,1H),2.56(s,3H),2.24-2.22(m,1H),2.08-2.03(m,2H),1.75-1.48(m,11H),1.39-1.08(m,13H),0.99-0.98(m,1H),0.86-0.85(m,1H),0.72(s,3H)ppm;ESI MS m/z 459[M+H]+

further elution afforded 26 as a white solid (14.9mg, 7%): mp 82-83 ℃;1HNMR(500MHz,CDCl3)δ5.09(dd,J=37.0,18.0Hz,2H),3.52(d,J=10.0Hz,1H),3.43-3.37(m,3H),2.65(t,J=9.0Hz,1H),2.47(s,3H),2.24-2.22(m,1H),2.06-2.03(m,2H),1.75-1.41(m,11H),1.33-1.09(m,13H),0.99-0.98(m,1H),0.87-0.86(m,1H),0.69(s,3H)ppm;ESI MS m/z 459[M+H]+

example 16. general procedure C: preparation of A/B-cis skeleton

Step 1. preparation of compound C2. A mixture of commercially available 19-hydroxyandrost-4-ene-3, 17-dione (A1, 13.6g, 45mmol) and palladium on carbon (3.2g, 10 wt%) in anhydrous tetrahydrofuran (150mL) was shaken at room temperature under an atmosphere of hydrogen (50psi) for 12 hours when TLC analysis of the mixture (2:1 hexane/ethyl acetate) indicated completion of the reaction. The atmosphere was exchanged for nitrogen and the mixture was filtered under reduced pressure through a pad of Celite, washing the filter cake with ethanol. The filtrate solvent was removed under reduced pressure to give C2 as a white solid which was used in the next step without further purification (13.0g, 95%): LCMS M/z 305[ M + H ]]+

Step 2. preparation of compound C3. Pyridine hydrochloride (750mg, 6.5mmol) was added to crude compound C2(15.0g, 49mmol) in ethylene glycol (65mL) and dry toluene (180mL) at room temperature under nitrogenIn the solution of (1). The mixture was heated at reflux for 12 hours and water was removed by Dean-Stark apparatus, at which time TLC analysis of the mixture (2:1 hexane/ethyl acetate) indicated completion of the reaction. The solvent was removed from the cooled mixture under reduced pressure and the residue was treated with saturated aqueous sodium chloride (50mL) and extracted with ethyl acetate (3 × 50 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (3 × 10mL), dried over anhydrous sodium sulfate and filtered. The solvent was removed under reduced pressure to give compound C3 as a colorless oil which was used in the next step without further purification (20.3g,>99%):1H NMR(300MHz,CDCl3)δ4.11-3.81(m,8H),3.60-3.54(m,1H),2.05-1.92(m,3H),1.81-163(m,4H),1.59-1.35(m,12H),1.28-1.12(m,5H),0.8(s,3H)ppm;LCMS m/z 393[M+H]+

step 3. preparation of compound C4. A solution of crude compound C3(20.3g, 49mmol) in anhydrous tetrahydrofuran (120mL) was added dropwise to a suspension of sodium hydride (7.9g, 197mmol, 60% in mineral oil) in anhydrous tetrahydrofuran (120mL) at 0 ℃ under nitrogen, after which the mixture was stirred at 0 ℃ for 30 min. Methyl iodide (15.3mL, 246mmol) was added dropwise, after which the mixture was heated to 35 ℃ and stirred for 3 hours, at which time TLC analysis of the mixture (3:1 hexane/ethyl acetate) indicated that the reaction was complete. The cooled mixture was treated with saturated ammonium chloride solution (100mL) and extracted with ethyl acetate (2X 50 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (2 × 20mL), dried over anhydrous sodium sulfate and filtered. The solvent was removed under reduced pressure to give crude compound C4 as a yellow oil which was used in the next step without further purification (25.6g,>99%):LCMS m/z 407[M+H]+

step 4. preparation of compound C5. A mixture of crude compound C4(25.5g, 49mmol) in tetrahydrofuran (150mL) and acetone (90mL) was treated with 2N HCl (123mL) at room temperature and the mixture was stirred for 16 hours, at which time TLC analysis of the mixture (2:1 hexane/ethyl acetate) indicated that the reaction was complete. The reaction mixture was adjusted to pH 8 by slow addition of saturated aqueous sodium bicarbonate and extracted with ethyl acetate (3 × 125 mL). The combined organic extracts were extracted with saturated aqueous sodium chloride (2 in a prepared form)20mL), dried over anhydrous sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with hexane/ethyl acetate (2:1) to give compound C5(10.6g, 67%) as a white solid:1H NMR(300MHz,CDCl3)δ3.62-3.59(m,1H),3.36-3.33(m,4H),2.67-2.63(m,1H),2.58-2.45(m,1H),2.42-2.27(m,3H),2.25-1.84(m,6H),1.71-1.23(m,11H),0.89(s,3H)ppm;LCMS m/z 319[M+H]+

step 5. preparation of compound C6. Iodine (84mg, 0.3mmol) was added to a solution of compound C5(10.6g, 33mmol) in dry methanol (200mL) under nitrogen at room temperature, after which the mixture was heated to 60 ℃ and stirred for 90 minutes, at which time TLC analysis of the mixture (2:1 hexane/ethyl acetate) indicated that the reaction was complete. The cooled mixture was treated with 1N sodium hydroxide solution (200mL) and extracted with hexane/ethyl acetate (3:1, 3X 100 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (2 × 25mL), dried over anhydrous sodium sulfate and filtered. The solvent was removed under reduced pressure to give compound C6 as a colorless oil which was used in the next step without further purification (13.8g,>99%);LCMS m/z 365[M+H]+

step 6. preparation of compound C7. Potassium tert-butoxide (11.2g, 100mmol) was added to a mixture of ethyltriphenylphosphonium bromide (36.9g, 100mmol) in anhydrous tetrahydrofuran (150mL) at room temperature under nitrogen, after which the mixture was heated to 60 ℃ and stirred for 4 hours. A solution of compound C6(13.8g, 33mmol) in dry tetrahydrofuran (100mL) was added, after which stirring was continued at 60 ℃ for 18 h. The cooled mixture was diluted with water (200mL) and hexane (100mL) and extracted with ethyl acetate (3X 100 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (2 × 25mL), treated with 2N HCl (100mL) and stirred at room temperature for 3 hours. The resulting mixture was washed with a saturated aqueous sodium bicarbonate solution and a saturated aqueous sodium chloride solution, dried over sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with hexane/ethyl acetate (9:1) to give compound C7(9.2g, 84%) as a colorless oil: LCMS M/z 331[ M + H ]]+

Step 7. preparation of compound C8. Bis (2, 6-di-tert-butyl-4-methylphenoxy) methylaluminium (40.6mL, 16mmol, 0.4M in toluene) was added in portions to a solution of compound C7(1.8g, 5.4mmol) in anhydrous toluene (20mL) at-78 ℃ under nitrogen, after which the mixture was stirred for 10 minutes. Methyl magnesium bromide (11.6mL, 16mmol, 1.4M in tetrahydrofuran/toluene) was added dropwise, after which the mixture was stirred at-78 ℃ for 1 hour. The mixture was warmed to ice bath temperature and treated slowly with 2N HCl (60mL), warmed to room temperature and extracted with ethyl acetate (3X 50 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (2 × 20mL), dried over sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was purified by silica gel column chromatography eluting with hexane/ethyl acetate (2:1) to give crude compound C8(1.5g, 91%) as a white semi-solid; LCMS M/z 347[ M + H ]]+

Step 8 preparation of compound C9. Borane-tetrahydrofuran complex (27.6mL, 27.6mmol, 1.0M tetrahydrofuran solution) was added to a solution of compound C8(2.4g, 6.9mmol) in anhydrous tetrahydrofuran (24mL) under nitrogen at 0 ℃, after which the mixture was slowly warmed to room temperature and stirred for a total of 4 hours. The mixture was cooled in an ice bath and a 10% aqueous sodium hydroxide solution (20mL) was slowly added followed by 30% aqueous hydrogen peroxide solution (20 mL). The resulting mixture was warmed to room temperature and stirred for 1 hour, then treated with saturated aqueous sodium chloride (100mL) and extracted with dichloromethane (3X 100 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (25mL), dried over sodium sulfate and filtered. The solvent was removed under reduced pressure to give crude compound C9 as a white solid which was used in the next step without further purification (2.7g,>99%);LCMS m/z365[M+H]+

step 9. preparation of C10. Piromodil chlorochromate (6.0g, 28mmol) was added in portions to a solution of compound C9(2.7g, 6.9mmol) in dichloromethane (100mL) at 0 ℃ under nitrogen, after which the mixture was slowly warmed to room temperature and stirred for a total of 16 hours. The solids were removed by filtration and the filtrate solvent was removed under reduced pressure. By silica gel column chromatography eluting with hexane/ethyl acetate (1:1)The residue was semi-purified, followed by further purification by reverse phase preparative HPLC to give C10(2.15g, 86%) as a white solid: mp 142-144 ℃;1HNMR(300MHz,CDCl3)δ3.55(d,J=9.0Hz,1H),3.33(s,3H),3.19(d,J=9.0Hz,1H),2.53(t,J=9.0Hz,1H),2.21-2.11(m,4H),2.08-1.87(m,3H),2.14-1.91(m,7H),1.77-1.36(m,16H),1.28(s,3H),1.26-1.07(m,2H),0.60(s,3H)ppm;LCMS m/z 345[M+H-H2O]+

step 10. preparation of compound C11. Hydrogen bromide (5 drops, 48% in water) was added to a solution of C10(2.15g, 5.9mmol) in dry methanol (150mL) under nitrogen at room temperature in the dark, after which bromine (0.6mL, 12mmol) was added and the mixture was stirred for 90 min. The mixture was poured into ice water (250mL) and treated sequentially with 2N sodium hydroxide solution (20mL) and saturated aqueous sodium bicarbonate solution (100 mL). The solid was collected under reduced pressure and purified by silica gel column chromatography eluting with hexanes/ethyl acetate (1:1) to give compound C11(1.4g, 53%) as a white solid: LCMS M/z 442[ M + H ]]+

Example 17 alternative preparation of intermediate C9.

Step 1. preparation of compound C2. To a solution of Pd/C (1g, 10% wet) in THF (10mL) was added a solution of A1(10g, 33.07mmol) in anhydrous THF (140mL) which was added to the mixture. After TLC showed complete consumption of starting material, the mixture was washed with CH2Cl2Filtered (300mL) and concentrated. The residue was purified by silica gel column chromatography (PE: EA ═ 8:1-4:1-2:1-1:1-EA) to give C2(8.3g, 82.43%) as a white solid.1H NMR(400MHz,CDCl3)δ(ppm)=3.96(d,J=8.0Hz,1H),3.69(d,J=8.0Hz,1H),2.69-2.65(m,1H),2.45-2.29(m,4H),2.12-1.69(m,8H),1.63-1.23(m,7H),0.88(s,3H)。

Step 2. preparation of compound C20. To a solution of compound C2(15g, 49.3mmol) in THF (150mL) at 0 deg.C was added KOH (8.4g, 149.7mmol) and Me2SO4(12.9g,10067 mol). The mixture was then warmed to 25 ℃ and stirred at the same temperature for 3 hours. TLC (PE: EA ═ 1:4) showed almost complete consumption of starting material. The mixture was quenched by the addition of 300mL of water. The resulting solution was extracted with EtOAc (200 mL. times.3). The combined organic layers were washed with saturated aqueous NaCl solution (50mL) and over anhydrous Na2SO4Drying and evaporation in vacuo afforded the crude product, which was purified by silica gel column chromatography (petroleum ether/ethyl acetate 4/1) to afford compound C20(9.5g, 60.5%) as a white solid.

Step 3. preparation of compound C21. To a solution of compound 2, 6-di-tert-butyl-4-methylphenol (4.15g, 18.84mmol) in toluene (8mL) at less than 25 deg.C was added AlMe dropwise3(4.7mL, 9.42mmol, 2M in toluene). The solution was stirred at room temperature for 1 hour. A solution of compound C20(1g, 3.14mmol) in toluene (3mL) was then added dropwise at-78 ℃. After stirring at the same temperature for 1 hour, MeMgBr (5.23mL, 15.7mmol, 3M in ether) was added dropwise at-78 ℃. The resulting solution was stirred at-78 ℃ to-50 ℃ for 3 hours. TLC (PE/EtOAc ═ 1/1) showed the reaction was complete. By saturation of NH at-78 deg.C4The reaction was quenched with aqueous Cl (200 mL). The resulting mixture was filtered through a celite pad and the pad was washed with EtOAc (100 mL). The combined organic layers were separated, washed with brine (100mL × 2) and concentrated in vacuo. The crude product was purified by silica column chromatography (petroleum ether/ethyl acetate 4/1) to give compound C21(1g, 95%) as a light yellow oil.

Step 4. preparation of compound C22. At 0 ℃ to PPh3To a solution of EtBr (42.17g, 113.6mmol) in THF (40mL) was added a solution of t-BuOK (12.75g, 113.6mmol) in THF (40 mL). After stirring at 60 ℃ for 1 hour, a solution of compound C21(7.6g, 22.72mmol) in THF (40mL) was added dropwise at 60 ℃. The reaction mixture was then stirred at 60 ℃ for 8 hours. TLC (PE/EtOAc ═ 3/1) showed the starting material still remaining. At 0 ℃ to PPh3To a solution of EtBr (42.17g, 113.6mmol) in THF (40mL) was added a solution of t-BuOK (12.75g, 113.6mmol) in THF (40 mL). After stirring at 60 ℃ for 1 hour, the solution was added to the reaction mixture. The reaction mixture was then stirred at 60 ℃For 8 hours. TLC (PE/EA ═ 3/1) showed that starting material still remained and the reaction was almost unchanged. The reaction mixture was filtered and the filtrate was concentrated in vacuo to remove most of the solvent. The residue was partitioned between EtOAc (300mL) and water (100 mL. times.3). The organic layer was washed with brine (100mL) and Na2SO4Dried and concentrated in vacuo. The crude product was purified by silica column (PE: EA ═ 5:1) to give compound C22(4.0g, 50.8%) as a light yellow oil.1H NMR(400MHz,CDCl3)δ5.15-5.09(m,1H),3.58(d,J=9.2Hz,1H),3.49(s,1H),3.33(s,3H),3.20(d,J=8.8Hz,1H),2.40-1.10(m,28H),0.85(s,3H)。

Step 5. preparation of compound C9. To a solution of compound C22(2.5g, 7.21mmol) in THF (30mL) at 0 deg.C was added BH dropwise3-Me2S solution (7.21mL, 32.88 mmol). The solution was stirred at 25 ℃ for 3 hours. TLC (PE/EtOAc ═ 1/1) showed the reaction was complete. After cooling to 0 ℃, NaOH solution (27.5mL, 3M) was added very slowly. After the addition was complete, H was added slowly2O2(15mL, 30%) and maintain the internal temperature below 10 ℃. The resulting solution was stirred at room temperature for 2 hours. The resulting solution was extracted with EtOAc (100 mL. times.3). The combined organic solution was saturated with Na2S2O3Aqueous solution (100mL), brine (100mL), washed over Na2SO4Drying and concentration in vacuo afforded the crude product, compound C23(2.5g, 95.15%) as a white solid. The crude product was used in the next step without further purification.

Example 18. general procedure E: preparation of A/B-cis skeleton C-21 analogue

Preparation of compounds 73, 74 and 75. Reacting 5-fluoro-1H-benzo [ d ] at room temperature under nitrogen][1,2,3]Triazole (112mg, 0.82mmol) and potassium carbonate (373mg, 2.7mmol) were added to a solution of compound C11(120mg, 0.27mmol) in anhydrous tetrahydrofuran (12mL) and the mixture was stirred for 16 h, at which time the mixture (2:1 hexane/ethyl acetate)) TLC analysis indicated the reaction was complete. The mixture was diluted with water (80mL) and extracted with ethyl acetate (3X 80 mL). The combined organic extracts were washed with saturated aqueous sodium chloride (2 × 25mL), dried over sodium sulfate and filtered. The solvent was removed under reduced pressure and the residue was semi-purified by silica gel column chromatography eluting with hexane/ethyl acetate (3:1) to give a mixture of the three regioisomers. The residue was further purified by reverse phase preparative HPLC to give 73(44mg, 33%) as a white solid: mp 82-84 ℃; 1H NMR (500MHz, CDCl)3)δ7.86(dd,J=9.0,4.5Hz,1H),7.46(dd,J=9.0,2.5Hz,1H),7.20(ddd,J=9.0,9.0,2.0Hz,1H),5.50(d,JAB=17.5Hz,1H),5.46(d,JAB=17.0Hz,1H),3.54(d,J=9.5Hz,1H),3.34(s,3H),3.21(d,J=9.0Hz,1H),2.64(t,J=9.0Hz,1H),2.27-2.18(m,1H),2.18-2.11(m,1H),1.96-1.88(m,2H),1.83-1.40(m,12H),1.39-1.10(m,10H),0.73(s,3H)ppm;ESI MS m/z 496[M-H]-

Further elution afforded 75(25mg, 18%) as a white solid: mp 205-;1H NMR(500MHz,CDCl3)δ7.72-7.68(m,1H),7.31-7.27(m,2H),5.43(d,JAB=18.0Hz,1H),5.36(d,JAB=18.0Hz,1H),3.54(d,J=9.0Hz,1H),3.34(s,3H),3.23(d,J=9.0Hz,1H),2.70(t,J=9.0Hz,1H),2.27-2.18(m,1H),2.18-2.12(m,1H),1.97-1.88(m,2H),1.84-1.72(m,3H),1.70-1.58(m,3H),1.57-1.43(m,6H),1.40-1.12(m,10H),0.71(s,3H)ppm;ESI MS m/z 498[M+H]+

further elution afforded 74(30mg, 22%) as an off-white solid: mp 195-197 ℃;1H NMR(500MHz,CDCl3)δ8.04(dd,J=9.0,4.5Hz,1H),7.15(dt,J=9.0,2.5Hz,1H),6.97(dd,J=7.5,2.0Hz,1H),5.40(d,JAB=18.0Hz,1H),5.33(d,JAB=18.0Hz,1H),3.54(d,J=9.0Hz,1H),3.34(s,3H),3.22(d,J=9.0Hz,1H),2.70(t,J=9.0Hz,1H),2.27-2.18(m,1H),2.18-2.12(m,1H),1.97-1.89(m,2H),1.84-1.72(m,3H),1.71-1.57(m,3H),1.57-1.42(m,6H),1.40-1.12(m,10H),0.72(s,3H)ppm;ESI MS m/z 498[M+H]+

example 19. preparation of compound 27.

Prepared according to general procedure E, step 2 from compound C11(60mg, 0.14mmol) and 1H-pyrazole-4-carbonitrile (63mg, 0.67mmol), purified by silica gel column chromatography to give compound 27 as an off-white solid (27.3mg, 44%): mp 176-;1HNMR(300MHz,CDCl3)δ7.83(d,J=12.3Hz,2H),4.95(q,J=18.3Hz,2H),3.53(d,J=9.0Hz,1H),3.33(s,3H),3.22(d,J=9.0Hz,1H),2.59(t,J=9.3Hz,1H),2.26-1.35(m,17H),1.31-1.08(m,9H),0.66(s,3H)ppm;ESI MS m/z 437[M+H-H2O]+

example 20. preparation of compound 28.

Prepared according to general procedure E, step 2, from compound C11(80mg, 0.18mmol) and 4-methyl-1H-pyrazole (45mg, 0.54mmol) with substitution of cesium carbonate (177mg, 0.54mmol) in anhydrous acetonitrile (6mL) at 65 ℃, purified by silica gel column chromatography to give compound 28(58mg, 73%) as a white solid: mp 158-160 ℃;1H NMR(500MHz,CDCl3)δ7.34(s,1H),7.16(s,1H),4.86(d,JAB=17.5Hz,1H),4.78(d,JAB=18.0Hz,1H),3.54(d,J=9.0Hz,1H),3.32(s,3H),3.19(d,J=9.0Hz,1H),2.54(t,J=9.5Hz,1H),2.23-2.13(m,1H),2.09(s,3H),2.07-2.02(m,1H),1.97-1.87(m,2H),1.80-1.35(m,12H),1.33-1.10(m,10H),0.66(s,3H)ppm;ESI MS m/z 443[M+H]+

example 21 preparation of compounds 29 and 30.

Prepared according to general procedure E, step 2, from compound C11(100mg, 0.23mmol) and 5-methyltetrazole (95mg, 1.13mmol) by semi-purification via silica gel column chromatography and reverse phase preparative HPLC to afford off-white29(12.2mg, 12%) as a colored solid: mp 90-92 ℃;1HNMR(300MHz,CDCl3)δ5.09(q,J=18.0Hz,2H),3.53(d,J=9.0Hz,1H),3.33(s,3H),3.22(d,J=9.0Hz,1H),2.65(t,J=9.0Hz,1H),2.47(s,3H),2.25-1.58(m,9H),1.55-1.14(m,17H),0.67(s,3H)ppm;ESI MS m/z 428[M+H-H2O]+

further elution afforded 30(13.4mg, 13%) as an off-white solid: mp 70-72 ℃;1HNMR(300MHz,CDCl3)δ5.34(s,2H),3.54(d,J=9.0Hz,1H),3.33(s,3H),3.20(d,J=9.0Hz,1H),2.64-2.57(m,4H),2.43-1.91(m,6H),1.81-1.10(m,20H),0.70(s,3H)ppm;ESI MS m/z 428[M+H-H2O]+

example 22 preparation of compounds 31 and 32.

Prepared according to general procedure E, step 2, from compound C11(82mg, 0.18mmol) and 1H-1,2, 3-triazole (75mg, 1.08mmol) by semi-purification via silica gel column chromatography and reverse phase preparative HPLC to give 32(17mg, 22%) as an off-white solid: mp 80-83 ℃;1H NMR(500MHz,CDCl3)δ7.79(s,1H),7.66(s,1H),5.26(d,JAB=18.0Hz,1H),5.13(d,JAB=18.0Hz,1H),3.53(d,J=9.0Hz,1H),3.33(s,3H),3.20(d,J=9.0Hz,1H),2.64(t,J=9.0Hz,1H),2.26-2.16(m,1H),2.12-2.06(m,1H),1.96-1.70(m,6H),1.66-1.42(m,6H),1.35-1.10(m,11H),0.90-0.83(m,1H),0.66(s,3H)ppm;ESI MS m/z 430[M+H]+

further elution afforded 31(12mg, 16%) as an off-white solid: mp 71-74 ℃;1H NMR(500MHz,CDCl3)δ7.68(s,2H),5.24(d,JAB=17.5Hz,1H),5.21(d,JAB=17.5Hz,1H),3.54(d,J=9.0Hz,1H),3.33(s,3H),3.19(d,J=9.0Hz,1H),2.56(t,J=9.0Hz,1H),2.24-2.15(m,1H),2.11-2.05(m,1H),1.96-1.88(m,2H),1.82-1.68(m,3H),1.66-1.40(m,6H),1.40-1.09(m,11H),0.90-0.83(m,1H),0.70(s,3H)ppm;ESI MS m/z 430[M+H]+

example 23 preparation of compound 33.

Prepared according to general procedure E, step 2 from compound C11(80mg, 0.18mmol) and 4- (methylsulfonyl) -1H-pyrazole (79mg, 0.54mmol) by semi-purification via silica gel column chromatography and reverse phase preparative HPLC to give 33(62mg, 69%) as a white solid: mp110-112 ℃;1H NMR(500MHz,CDCl3)δ7.92(s,1H),7.86(s,1H),5.00(d,JAB=18.0Hz,1H),4.90(d,JAB=17.5Hz,1H),3.53(d,J=9.0Hz,1H),3.33(s,3H),3.21(d,J=9.0Hz,1H),3.13(s,3H),2.60(t,J=9.0Hz,1H),2.25-2.15(m,1H),2.08-2.02(m,1H),1.96-1.88(m,2H),1.82-1.71(m,3H),1.67-1.56(m,3H),1.54-1.41(m,6H),1.39-1.13(m,10H),0.67(s,3H)ppm;ESI MS m/z 505[M-H]-

example 24. preparation of compound 34.

Prepared according to general procedure E, step 2 from compound C11(80mg, 0.18mmol) and 4-chloro-1H-pyrazole (45mg, 0.54mmol) and purified by silica gel column chromatography to give compound 34(58mg, 70%) as a white solid: mp 163-165 ℃;1H NMR(500MHz,CDCl3)δ7.45(s,1H),7.41(s,1H),4.99(d,JAB=17.5Hz,1H),4.80(d,JAB=17.5Hz,1H),3.53(d,J=9.0Hz,1H),3.33(s,3H),3.21(d,J=9.0Hz,1H),2.56(t,J=9.0Hz,1H),2.24-2.14(m,1H),2.07-2.01(m,1H),1.96-1.88(m,2H),1.80-1.69(m,3H),1.66-1.35(m,9H),1.34-1.10(m,10H),0.66(s,3H)ppm;ESI MS m/z 463[M+H]+

example 25 preparation of compound 38.

According to a general programSequence B from Compound A11(31mg, 0.071mmol) and 2H-pyrazolo [4,3-B]Preparation of pyridine (168mg, 1.41mmol) and purification by reverse phase preparative HPLC gave 38(8.7mg, 25%) as a white solid: mp 153-;1HNMR(500MHz,CDCl3)δ8.60(d,J=3.5Hz,1H),8.28(s,1H),7.59(d,J=8.5Hz,1H),7.29(dd,J=8.5,4.5Hz,1H),5.16(dd,J=29.0,18.0Hz,2H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.66(t,J=9.0Hz,1H),2.22-2.00(m,1H),2.13-2.11(m,1H),2.10-2.08(m,1H),1.79-1.40(m,11H),1.33-1.11(m,10H),0.99-0.97(m,1H),0.88-0.86(m,1H),0.73(s,3H)ppm;ESI MS m/z 480[M+H]+

example 26 preparation of compounds 39 and 40.

Prepared according to general procedure B from compound a11(21mg, 0.047mmol) and 1H-benzotriazole (33mg, 0.28mmol), followed by silica gel column chromatography and reverse phase preparative HPLC semi-purification to give 39(13mg, 58%) as an off-white solid: mp 78-80 ℃;1H NMR(500MHz,CDCl3)δ8.11-8.07(m,1H),7.51-7.46(m,1H),7.40-7.36(m,1H),7.33(d,J=7.5,1H),5.41(s,2H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.71(t,J=9.0Hz,1H),2.27-2.18(m,1H),2.17-2.10(m,1H),2.08-2.02(m,1H),1.82-1.68(m,4H),1.67-1.42(m,7H),1.35-1.09(m,10H),1.05-0.95(m,1H),0.91-0.83(m,1H),0.76(s,3H)ppm;ESI MS m/z 480[M+H]+

further elution afforded 40(7mg, 30%) as an off-white solid: mp 70-72 ℃;1H NMR(500MHz,CDCl3)δ7.90-7.85(m,2H),7.41-7.36(m,2H),5.54(d,JAB=17.0Hz,1H),5.48(d,JAB=17.0Hz,1H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.290(s,3H),2.63(t,J=9.0Hz,1H),2.28-2.20(m,1H),2.17-2.12(m,1H),2.07-2.01(m,1H),1.80-1.68(m,4H),1.67-1.46(m,6H),1.45-1.36(m,1H),1.35-1.08(m,10H),1.04-0.94(m,1H),0.90-0.82(m,1H),0.77(s,3H)ppm;ESI MS m/z 480[M+H]+

example 27 preparation of compounds 41, 42 and 43.

Prepared according to general procedure B from compound a11(100mg, 0.23mmol) and 5-fluorobenzotriazole (124mg, 0.91mmol) and purified by reverse phase preparative HPLC to give 43(39.2mg, 40%) as an off-white solid: mp 55-60 ℃;1HNMR(300MHz,CDCl3)δ7.86(dd,J=4.8,4.5Hz,1H),7.46(dd,J=9.0,2.1Hz,1H),7.23-7.16(m,1H),5.49(q,J=18.0Hz,2H),3.48(d,J=9.9Hz,1H),3.38(d,J=9.9Hz,1H),3.37(s,3H),2.66(t,J=8.7Hz,1H),2.29-2.01(m,3H),1.79-1.50(m,15H),1.45-1.05(m,6H),1.01-0.82(m,2H),0.76(s,3H)ppm;ESI MS m/z 498[M+H]+

further elution afforded 41(21.2mg, 34%) as an off-white solid: mp 65-70 ℃;1HNMR(300MHz,CDCl3)δ8.03(dd,J=9.0,4.5Hz,1H),7.18-7.12(m,1H),6.99-6.58(m,1H),5.37(q,J=18.3Hz,2H),3.47(d,J=10.2Hz,1H),3.38(d,J=10.2Hz,1H),3.31(s,3H),2.72(t,J=9.0Hz,1H),2.28-2.02(m,3H),1.79-1.33(m,13H),1.29-0.84(m,10H),0.75(s,3H)ppm;ESI MS m/z 498[M+H]+

further elution afforded 42(21.2mg, 34%) as an off-white solid: mp 60-65 ℃;1HNMR(300MHz,CDCl3)δ7.73(d,J=9.9Hz,1H),7.35-7.21(m,2H),5.40(q,J=18.3Hz,2H),3.49(d,J=9.9Hz,1H),3.37(d,J=10.2Hz,1H),3.30(s,3H),2.72(t,J=9.0Hz,1H),2.28-2.03(m,3H),1.82-1.46(m,9H),1.37-1.16(m,12H),1.06-0.84(m,2H),0.74(s,3H)ppm;ESI MS m/z 498[M+H]+

example 28 preparation of compounds 44 and 45.

General procedure B from Compound A11(50mg, 0.11mmol) and 1H-pyrazolo [3,4-c]Preparation of pyridine (67mg, 0.57mmol) by reverse phase preparation of HPLPurification of C afforded 44(7.5mg, 14%) as an off-white solid: mp 160-162 ℃;1HNMR(300MHz,CDCl3)δ9.26(s,1H),8.17(d,J=6.0Hz,1H),7.97(d,J=0.9Hz,1H),7.53(dd,J=6.0,1.2Hz,1H),5.27(q,J=18.0Hz,2H),3.46(d,J=9.9Hz,2H),3.37(d,J=10.2Hz,2H),2.68(t,J=9.3Hz,1H),2.30-2.19(m,1H),1.18-2.01(m,3H),1.79-1.64(m,5H),1.61-1.45(m,7H),1.39-1.08(m,8H),1.05-0.84(m,3H),0.74(s,3H)ppm;ESI MS m/z 480[M+H]+

further elution afforded 45 as an off-white solid (14.2mg, 26%): mp 92-94 ℃;1H NMR(300MHz,CDCl3)δ8.80(s,1H),8.34(d,J=5.4Hz,1H),8.09(d,J=0.6Hz,1H),7.64(dd,J=5.7,1.2Hz,1H),5.25(q,J=18.0Hz,2H),3.48(d,J=9.9Hz,2H),3.38(d,J=9.9Hz,2H),3.31(s,3H),2.70(t,J=8.7Hz,1H),2.27-1.98(m,3H),1.81-1.44(m,10H),1.34-1.12(m,9H),1.10-0.81(m,2H),0.74(s,3H)ppm;ESI MS m/z 480[M+H]+

example 29 preparation of compound 46.

Prepared according to general procedure B from compound a11(50mg, 0.114mmol) and benzimidazole (268mg, 2.3mmol) and purified by reverse phase preparative HPLC to give 46(36.5mg, 67%) as a white solid: mp 104-;1HNMR(500MHz,CDCl3)δ7.99(s,1H),7.86-7.82(m,1H),7.33-7.29(m,2H),7.20-7.18(m,1H),4.93(dd,J=24.0,18.5Hz,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.66(t,J=9.0Hz,1H),2.24-2.22(m,1H),2.07-2.05(m,1H),1.80-1.42(m,11H),1.34-1.11(m,11H),0.99-0.98(m,1H),0.88-0.86(m,1H),0.73(s,3H)ppm;ESI MS m/z 479[M+H]+

example 30 preparation of compounds 47, 48 and 49.

According to the general procedurePrepared from compound a11(100mg, 0.227mmol) and 4-fluorobenzotriazole (311mg, 2.27mmol), purification by reverse phase preparative HPLC gave 49(22.5mg, 20%) as a white solid: mp 125-126 ℃;1HNMR(500MHz,CDCl3)δ7.67(d,J=8.5Hz,1H),7.32-7.30(m,1H),7.03(dd,J=10.5,7.5Hz,1H),5.54(dd,J=28.0,17.0Hz,2H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.67(t,J=9.0Hz,1H),2.24-2.22(m,1H),2.15-2.13(m,1H),2.05-2.03(m,1H),1.78-1.50(m,9H),1.42-1.40(m,1H),1.34-1.12(m,11H),0.99-0.98(m,1H),0.87-0.86(m,1H),0.77(s,3H)ppm;ESI MS m/z 498[M+H]+

further elution afforded 47 as a white solid (7.3mg, 6%): mp 83-84 ℃;1HNMR(500MHz,CDCl3)δ7.86(d,J=8.0Hz,1H),7.29-7.27(m,1H),7.13(dd,J=10.5,7.5Hz,1H),5.54(s,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.70(t,J=9.0Hz,1H),2.24-2.22(m,1H),2.15-2.13(m,1H),2.06-2.04(m,1H),1.79-1.73(m,4H),1.63-1.43(m,7H),1.34-1.13(m,10H),1.02-1.00(m,1H),0.89-0.87(m,1H),0.75(s,3H)ppm;ESI MS m/z 498[M+H]+

further elution afforded 48(26.3mg, 23%) as a white solid: mp 129-130 ℃;1HNMR(500MHz,CDCl3)δ7.42(td,J=8.0,4.5Hz,1H),7.10(d,J=8.5Hz,1H),7.04(dd,J=10.0,7.5Hz,1H),5.42(s,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.70(t,J=9.0Hz,1H),2.23-2.21(m,1H),2.15-2.13(m,1H),2.06-2.04(m,1H),1.78-1.46(m,10H),1.34-1.12(m,11H),1.01-0.99(m,1H),0.89-0.88(m,1H),0.74(s,3H)ppm;ESI MS m/z 498[M+H]+

example 31 preparation of compounds 50 and 51.

Prepared according to general procedure B from compound a11(140mg, 0.32mmol) and 5-methoxybenzotriazole (132mg, 0.89mmol), purified by reverse phase preparative HPLC to give 50 as a white solid (12.9mg, 8%): mp 165-166 ℃;1HNMR(500MHz,CDCl3)δ7.72(d,J=10.0Hz,1H),7.07-7.05(m,2H),5.43(dd,J=29.5,17.0Hz,2H),3.87(s,3H),3.47(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.29(s,3H),2.63(t,J=9.0Hz,1H),2.24-2.22(m,1H),2.14-2.12(m,1H),2.05-2.04(m,1H),1.77-1.69(m,4H),1.62-1.10(m,17H),0.99-0.98(m,1H),0.86-0.84(m,1H),0.76(s,3H)ppm;ESI MS m/z 510[M+H]+

further elution afforded 51(11.8mg, 7%) as a white solid: mp 106-;1HNMR(500MHz,CDCl3)δ7.93(d,J=9.0Hz,1H),7.02(dd,J=9.0,2.0Hz,1H),6.61(d,J=2.0Hz,1H),5.33(d,J=3.5Hz,2H),3.88-3.86(m,3H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.70(t,J=9.0Hz,1H),2.23-2.21(m,1H),2.15-2.13(m,1H),2.06-2.04(m,1H),1.76-1.70(m,4H),1.64-1.44(m,6H),1.34-1.13(m,11H),0.99-0.98(m,1H),0.89-0.87(m,1H),0.75(s,3H)ppm;ESI MS m/z 510[M+H]+

example 32 preparation of compounds 52, 53 and 54.

Prepared according to general procedure B from compound a11(100mg, 0.23mmol) and 4, 5-difluorobenzotriazole (352mg, 2.3mmol) and purified by reverse phase preparative HPLC to give 54 as a white solid (46.0mg, 32%): mp 86-87 ℃;1HNMR(500MHz,CDCl3)δ7.63(ddd,J=9.0,3.5,1.0Hz,1H),7.30-7.28(m,1H),5.53(dd,J=31.5,17.0Hz,2H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.65(t,J=9.0Hz,1H),2.24-2.22(m,1H),2.14-2.12(m,1H),2.06-2.04(m,1H),1.78-1.73(m,4H),1.63-1.41(m,7H),1.34-1.12(m,10H),1.03-1.00(m,1H),0.88-0.86(m,1H),0.77(s,3H)ppm;ESI MS m/z 498[M+H-H2O]+

further elution afforded 52(19.3mg, 13%) as a white solid: mp 82-83 ℃;1HNMR(500MHz,CDCl3)δ7.80(ddd,J=9.0,3.5,1.0Hz,1H),7.23-7.21(m,1H),5.52(s,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.70(t,J=9.0Hz,1H),2.23-2.21(m,1H),2.14-2.12(m,1H),2.07-2.05(m,1H),1.79-1.72(m,4H),1.64-1.44(m,7H),1.35-1.13(m,10H),1.02-1.01(m,1H),0.89-0.88(m,1H),0.75(s,3H)ppm;ESI MS m/z 516[M+H]+

further elution afforded 53(34.3mg, 24%) as a white solid: mp 144-145 ℃;1HNMR(500MHz,CDCl3)δ7.38-7.37(m,1H),7.04(ddd,J=9.0,3.0,1.0Hz,1H),5.41(dd,J=22.5,18.0Hz,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.71(t,J=9.0Hz,1H),2.24-7.22(m,1H),2.12.14-2.12(m,1H),2.07-2.05(m,1H),1.78-1.71(m,4H),1.64-1.47(m,7H),1.34-1.13(m,10H),1.02-1.01(m,1H),0.89-0.88(m,1H),0.74(s,3H)ppm;ESI MS m/z 516[M+H]+

example 33 preparation of compounds 55 and 56.

From compound A11(125mg, 0.28mmol) and 4, 6-difluoro-1H-benzo [ d ] according to general procedure B][1,2,3]Preparation of triazole (219mg, 1.60mmol), semi-purified by silica gel column chromatography and reverse phase preparative HPLC in that order, to give 55(28mg, 19%) as an off-white solid: mp182-186 ℃;1H NMR(300MHz,CDCl3)δ7.30(dd,J=8.1,1.5Hz,1H),6.90(ddd,J=9.9,9.9,2.1Hz,1H),5.55(d,JAB=17.1Hz,1H),5.47(d,JAB=17.1Hz,1H),3.48(d,J=9.9Hz,1H),3.37(d,J=10.2Hz,1H),3.30(s,3H),2.67(t,J=8.7Hz,1H),2.30-2.17(m,1H),2.17-2.09(m,1H),2.09-1.99(m,1H),1.83-1.66(m,4H),1.66-1.40(m,7H),1.40-0.81(m,12H),0.76(s,3H)ppm;ESI MS m/z 516[M+H]+

further elution afforded 56(19mg, 13%) as an off-white solid: mp 96-100 ℃;1H NMR(300MHz,CDCl3)δ6.87(ddd,J=9.6,9.6,1.8Hz,1H),6.79(ddd,J=7.5,2.1,0.6Hz,1H),5.41(d,JAB=18.0Hz,1H),5.34(d,JAB=18.3Hz,1H),3.49(d,J=9.9Hz,1H),3.38(d,J=9.9Hz,1H),3.31(s,3H),2.72(t,J=8.7Hz,1H),2.30-2.00(m,3H),1.85-1.41(m,11H),1.38-0.82(m,12H),0.74(s,3H)ppm;SI MS m/z 516[M+H]+

example 34 preparation of compounds 57 and 58.

Prepared according to general procedure B from compound a11(100mg, 0.23mmol) and 4-methoxybenzotriazole (675mg, 4.5mmol), purified by reverse phase preparative HPLC to give 58(27mg, 23%) as a light brown solid: mp 78-80 ℃;1H NMR(500MHz,CDCl3)δ7.39(t,J=8.0Hz,1H),6.86(d,J=8.1Hz,1H),6.70(d,J=7.8Hz,1H),5.37(dd,J=18.0,2.7Hz,2H),4.12(s,3H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.1Hz,1H),3.30(s,3H),2.68(t,J=9.0Hz,1H),2.24-2.18(m,1H),2.14-2.09(m,1H),2.06-2.02(m,1H),1.76-1.69(m,4H),1.64-1.55(m,3H),1.54-1.37(m,4H),1.36-1.19(m,10H),1.18-1.08(m,2H),1.04-0.93(m,1H),0.90-0.84(m,2H),0.75(s,3H)ppm;APCI MS m/z 510[M+H]+

further elution afforded 57(33mg, 29%) as an off-white solid: mp 200-202 ℃;1H NMR(500MHz,CDCl3)δ7.63(d,J=8.4Hz,1H),7.24(d,J=8.3Hz,1H),6.76(d,J=7.7Hz,1H),5.58(dd,J=17.9,9.3Hz,2H),3.89(s,3H),3.49(d,J=10.0Hz,1H),3.39(d,J=10.0Hz,1H),3.30(s,3H),2.67(t,J=8.9Hz,1H),2.25-2.18(m,1H),2.15-2.11(m,1H),2.06-2.02(m,1H),1.73-1.67(m,3H),1.65-1.47(m,7H),1.42-1.11(m,12H),1.04-0.95(m,1H),0.89-10.84(m,1H),0.75(s,3H)ppm;APCI MS m/z 510[M+H]+

example 35 preparation of compounds 59, 60 and 61.

Prepared according to general procedure B from compound a11(150mg, 0.34mmol) and 4-chlorobenzotriazole (156mg, 1.02mmol), followed by silica gel column chromatography and reverse phase preparative HPLC semi-purification to afford 61 as a light brown solid (64mg, 37%): mp 170-172 ℃;1H NMR(500MHz,CDCl3)δ7.79(dd,J=8.5,0.5Hz,1H),7.40(dd,J=7.0,0.5Hz,1H),7.32(dd,J=8.5,7.5Hz,1H),5.57(d,JAB=17.0Hz,1H),5.53(d,JAB=17.0Hz,1H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.66(t,J=9.0Hz,1H),2.28-2.18(m,1H),2.17-2.10(m,1H),2.07-2.01(m,1H),1.81-1.68(m,4H),1.66-1.46(m,6H),1.44-1.36(m,1H),1.35-1.08(m,10H),1.04-0.94(m,1H),0.90-0.83(m,1H),0.77(s,3H)ppm;ESI MS m/z 514[M+H]+

further elution afforded 59(8mg, 4%) as a light brown solid: mp 162-164 ℃;1H NMR(500MHz,CDCl3)δ7.99(d,J=8.5Hz,1H),7.43(d,J=7.5,1H),7.29(t,J=7.5,1H),5.71(s,2H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.72(t,J=9.0Hz,1H),2.27-2.10(m,2H),2.08-2.02(m,1H),1.85-1.37(m,11H),1.35-1.09(m,10H),1.05-0.95(m,1H),0.91-0.83(m,1H),0.76(s,3H)ppm;APCI MS m/z 514[M+H]+

further elution afforded 60(32mg, 18%) as a light brown solid: mp 105-;1H NMR(500MHz,CDCl3)δ7.43-7.36(m,2H),7.23(dd,J=7.5,1.5Hz,1H),5.42(s,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.71(t,J=9.0Hz,1H),2.27-2.18(m,1H),2.18-2.10(m,1H),2.08-2.01(m,1H),1.82-1.68(m,4H),1.67-1.42(m,7H),1.35-1.09(m,10H),1.05-0.95(m,1H),0.91-0.84(m,1H),0.75(s,3H)ppm;ESI MS m/z 514[M+H]+

example 36 preparation of compounds 62, 63 and 64.

From compound A11(100mg, 0.23mmol) and 4, 5-dimethoxy-1H-benzo [ d ] according to general procedure B][1,2,3]Preparation of triazole (101mg, 0.57mmol), semi-purified by silica gel column chromatography and reverse phase preparative HPLC in that order, to give 64(32mg, 26%) as a light yellow solid: mp 188-190 ℃;1H NMR(500MHz,CDCl3)δ7.50(d,J=9.0Hz,1H),7.22(d,J=9.5Hz,1H),5.49(d,JAB=17.0Hz,1H),5.43(d,JAB=17.0Hz,1H),4.27(s,3H),3.95(s,3H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.29(s,3H),2.63(t,J=8.5Hz,1H),2.27-2.17(m,1H),2.16-2.10(m,1H),2.07-2.01(m,1H),1.79-1.68(m,4H),1.65-1.46(m,7H),1.42-1.08(m,10H),1.03-0.93(m,1H),0.89-0.81(m,1H),0.76(s,3H)ppm;APCI MS m/z 540[M+H]+

further elution afforded 63(19mg, 15%) as a white solid: mp 88-90 ℃;1H NMR(500MHz,CDCl3)δ7.24(d,J=9.0Hz,1H),6.82(d,J=9.0Hz,1H),5.36(d,JAB=18.0Hz,1H),5.32(d,JAB=18.0Hz,1H),4.57(s,3H),3.93(s,3H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.69(t,J=9.0Hz,1H),2.26-2.17(m,1H),2.16-2.10(m,1H),2.08-2.01(m,1H),1.80-1.68(m,4H),1.66-1.41(m,7H),1.36-1.09(m,10H),1.04-0.94(m,1H),0.90-0.83(m,1H),0.74(s,3H)ppm;APCI MS m/z 540[M+H]+

further elution afforded 62(13mg, 11%) as a white solid: mp 110-;1H NMR(500MHz,CDCl3)δ7.75(br s,1H),7.11(d,J=7.5Hz,1H),5.54(d,JAB=18.0Hz,1H),5.50(d,JAB=18.0Hz,1H),3.96(s,3H),3.90(s,3H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.69(t,J=8.5Hz,1H),2.28-2.01(m,3H),1.83-1.68(m,4H),1.65-1.47(m,6H),1.44-1.37(m,1H),1.36-1.08(m,10H),1.04-0.94(m,1H),0.90-0.83(m,1H),0.76(s,3H)ppm;APCI MS m/z 540[M+H]+

example 37 preparation of compounds 65 and 66.

From compound A11(125mg, 0.28mmol) and 4, 6-dimethoxy-1H-benzo [ d ] according to general procedure B][1,2,3]Preparation of triazole (127mg, 0.71mmol) and purification by silica gel column chromatography gave 65(42mg, 28%) as a light yellow solid: mp 106-108 ℃;1H NMR(500MHz,CDCl3)δ6.67(d,J=2.0Hz,1H),6.33(d,J=2.0Hz,1H),5.43(d,JAB=17.0Hz,1H),5.38(d,JAB=17.0Hz,1H),3.98(s,3H),3.96(s,3H),3.47(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.29(s,3H),2.61(t,J=9.0Hz,1H),2.26-2.16(m,1H),2.13-2.07(m,1H),2.07-2.00(m,1H),1.77-1.67(m,4H),1.63-1.46(m,7H),1.39-1.08(m,10H),1.02-0.93(m,1H),0.88-0.80(m,1H),0.75(s,3H)ppm;APCI MS m/z 540[M+H]+

further elution afforded 66(52mg, 34%) as an off-white solid: mp 110-;1H NMR(500MHz,CDCl3)δ6.34(d,J=2.0Hz,1H),6.16(d,J=2.0Hz,1H),5.28(s,2H),4.06(s,3H),3.84(s,3H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.29(s,3H),2.66(t,J=9.0Hz,1H),2.25-2.16(m,1H),2.13-2.08(m,1H),2.07-2.01(m,1H),1.79-1.67(m,4H),1.64-1.46(m,6H),1.45-1.37(m,1H),1.35-1.08(m,10H),1.03-0.93(m,1H),0.89-0.82(m,1H),0.74(s,3H)ppm;APCI MS m/z 540[M+H]+

example 38 preparation of compound 67.

General procedure B from Compound A11(200mg, 0.45mmol) and 6-bromo-1H-pyrazolo [4,3-B]Preparation of pyridine (450mg, 2.27mmol), semi-purified sequentially by silica gel column chromatography and reverse phase preparative HPLC, to give 67 as an off-white solid (43mg, 17%): mp 102-105 ℃;1H NMR(300MHz,CDCl3)δ8.60(d,J=1.5Hz,1H),8.23(s,1H),7.75(s,1H),5.16(d,JAB=18.0Hz,1H),5.06(d,JAB=18.0Hz,1H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.68(t,J=9.3Hz,1H),2.30-1.93(m,3H),1.92-1.38(m,11H),1.37-0.80(m,12H),0.73(s,3H)ppm;ESI MS m/z 558[M+H]+

example 39 preparation of compounds 68 and 69.

From compound a11(86mg, 0.19mmol) and 6-chloro-1H-pyrazolo [4,3-b]preparation of pyridine (570mg, 3.7mmol) and purification by reverse phase preparative HPLC gave 68(12mg, 12%) as a white solid: mp 88-90 ℃;1H NMR(500MHz,CDCl3)δ8.59(s,1H),8.51(s,1H),8.34(s,1H),5.40-5.22(m,2H),3.48(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.67(t,J=8.6Hz,1H),2.27-2.21(m,1H),2.11-1.86(m,2H),1.85-1.41(m,11H),1.34-1.09(m,11H),1.04-0.93(m,1H),0.91-0.81(m,1H),0.73(s,3H)ppm;APCI MS m/z 514[M+H]+

further elution afforded 69(40mg, 40%) as a white solid: mp 115-117 ℃;1H NMR(500MHz,CDCl3)δ8.58(s,1H),8.40(s,1H),7.77(s,1H),5.18(dd,J=18.0,32.1Hz,2H),3.49(d,J=10.0Hz,1H),3.38(d,J=10.0Hz,1H),3.30(s,3H),2.69(t,J=8.9Hz,1H),2.24-2.18(m,1H),2.14-2.10(m,1H),2.07-2.03(m,1H),1.80-1.68(m,4H),1.67-1.56(m,3H),1.55-1.42(m,4H),1.39-1.11(m,10H),1.05-0.98(m,1H),0.91-0.85(m,1H),0.73(s,3H)ppm;APCI MS m/z 514[M+H]+

example 40 preparation of compounds 70, 71 and 72.

Prepared according to general procedure B from compound a21(200mg, 0.44mmol) and 5-chlorobenzotriazole (1.35g, 8.8mmol) and purified by reverse phase preparative HPLC to give 72 as a white solid (12.6mg, 5%): mp 76-77 ℃;1HNMR(500MHz,CDCl3)δ7.87(d,J=1.5Hz,1H),7.81(d,J=9.0Hz,1H),7.34(dd,J=9.0,1.5Hz,1H),5.50(dd,J=32.5,17.0Hz,2H),3.52(d,J=9.5Hz,1H),3.43-3.38(m,3H),2.65(t,J=9.0Hz,1H),2.25-2.23(m,1H),2.14-2.13(m,1H),2.06-2.04(m,1H),1.77-1.48(m,10H),1.41-1.38(m,1H),1.33-1.11(m,13H),1.02-1.00(m,1H),0.88-0.86(m,1H),0.75(s,3H)ppm;ESI MS m/z 528[M+H]+

further elution afforded 70(17.1mg, 7%) as a white solid: mp 69-70 ℃;1HNMR(500MHz,CDCl3)δ8.00(d,J=9.0Hz,1H),7.35-7.33(m,2H),5.37(dd,J=40.5,18.0Hz,2H),3.53(d,J=9.5Hz,1H),3.43-3.40(m,3H),2.72(t,J=9.0Hz,1H),2.24-2.23(m,1H),2.16-2.14(m,1H),2.08-2.06(m,1H),1.79-1.42(m,11H),1.37-1.13(m,11H),1.01-0.99(m,1H),0.88(t,J=7.0Hz,3H),0.74(s,3H)ppm;ESI MS m/z 528[M+H]+

further elution afforded 71 as a white solid (23.6mg, 10%): mp 82-83 ℃;1HNMR(500MHz,CDCl3)δ8.06(d,J=1.5Hz,1H),7.45(dd,J=9.0,1.5Hz,1H),7.28(d,J=9.0Hz,1H),5.40(dd,J=29.5,18.0Hz,2H),3.53(d,J=10.0Hz,1H),3.42-3.39(m,3H),2.71(t,J=9.0Hz,1H),2.24-2.23(m,1H),2.16-2.14(m,1H),2.08-2.05(m,1H),1.77-1.48(m,11H),1.33-1.13(m,11H),1.02-0.99(m,1H),0.88(t,J=7.0Hz,3H),0.74(s,3H)ppm;ESI MS m/z 528[M+H]+

EXAMPLE 41 preparation of Compound 76.

Prepared according to general procedure E, step 2, from compound C11(120mg, 0.27mmol) and 5-chlorobenzimidazole (125mg, 0.82mmol), followed by silica gel column chromatography and reverse phase preparative HPLC semi-purification to afford 76(19mg, 14%) as a white solid: mp 85-87 ℃;1H NMR(300MHz,CDCl3)δ7.88-7.80(m,2H),7.35(d,J=9.1Hz,1H),5.49(s,2H),3.55(d,J=9.0Hz,1H),3.34(s,3H),3.22(d,J=9.0Hz,1H),2.64(t,J=8.5Hz,1H),2.27-2.12(m,2H),1.94-1.85(m,2H),1.84-1.36(m,15H),1.35-1.14(m,10H),0.73(s,3H)ppm;APCI MS m/z 514[M+H]+

example 42 preparation of Compounds 77, 78 and 79.

From compound C11(120mg, 0.27mmol) and 4, 5-difluoro-1H-benzo [ d ] according to general procedure E, step 2][1,2,3]Preparation of triazole (126mg, 0.82mmol), semi-purified by silica gel column chromatography and reverse phase preparative HPLC in sequence, to give 79(62mg, 44%) as a white solid:mp 103-105℃;1H NMR(500MHz,CDCl3)δ7.64(ddd,J=9.0,3.5,1.0Hz,1H),7.32-7.26(m,1H),5.54(d,JAB=17.0Hz,1H),5.49(d,JAB=17.0Hz,1H),3.54(d,J=9.0Hz,1H),3.34(s,3H),3.22(d,J=9.0Hz,1H),2.65(t,J=9.0Hz,1H),2.27-2.18(m,1H),2.17-2.11(m,1H),1.97-1.88(m,2H),1.84-1.73(m,3H),1.68-1.40(m,9H),1.40-1.11(m,10H),0.74(s,3H)ppm;ESI MS m/z 514[M-H]-

further elution afforded 77(20mg, 14%) as a white solid: mp 98-100 ℃;1H NMR(500MHz,CDCl3)δ7.81(ddd,J=9.0,4.0,1.0Hz,1H),7.24-7.19(m,1H),5.54(d,JAB=18.5Hz,1H),5.50(d,JAB=18.5Hz,1H),3.55(d,J=9.0Hz,1H),3.34(s,3H),3.22(d,J=9.0Hz,1H),2.71(t,J=9.0Hz,1H),2.27-2.17(m,1H),2.17-2.10(m,1H),1.98-1.89(m,2H),1.88-1.72(m,3H),1.70-1.42(m,9H),1.40-1.10(m,10H),0.72(s,3H)ppm;ESI MS m/z 514[M-H]-

further elution afforded 78(39mg, 28%) as an off-white solid: mp 100-;1H NMR(500MHz,CDCl3)δ7.41-7.34(m,1H),7.05(ddd,J=9.0,3.0,1.0Hz,1H),5.44(d,JAB=18.0Hz,1H),5.36(d,JAB=18.0Hz,1H),3.53(d,J=9.0Hz,1H),3.34(s,3H),3.23(d,J=9.0Hz,1H),2.70(t,J=9.0Hz,1H),2.26-2.17(m,1H),2.17-2.11(m,1H),1.98-1.88(m,2H),1.84-1.73(m,3H),1.69-1.42(m,9H),1.41-1.12(m,10H),0.71(s,3H)ppm;ESI MS m/z 514[M-H]-

EXAMPLE 43 preparation of Compound 80.

Prepared according to general procedure E, step 2, from compound C11(100mg, 0.23mmol) and benzimidazole (80mg, 0.68mmol) and purified by reverse phase preparative HPLC to give 80(68mg, 63%) as a white solid: mp 130-;1H NMR(300MHz,CDCl3)δ8.74(b s,1H),7.91-7.87(m,1H),7.41-7.38(m,2H),7.25-7.22(m,1H),5.16(dd,J=18.3,5.8Hz,2H),3.54(d,J=9.1Hz,1H),3.34(s,3H),3.23(d,J=9.1Hz,1H),2.71(t,J=8.9Hz,1H),2.29-2.08(m,2H),1.94-1.09(m,26H),0.72(s,3H)ppm;APCI MS m/z 479[M+H]+

example 44 preparation of compounds 81, 82 and 83.

From compound C11(120mg, 0.27mmol) and 4-fluoro-1H-benzo [ d ] according to general procedure E, step 2][1,2,3]Preparation of triazole (112mg, 0.82mmol) and purification by reverse phase preparative HPLC gave 82(40mg, 30%) as an off-white solid: mp 199-;1H NMR(500MHz,CDCl3)δ7.45-7.40(m,1H),7.11(d,J=8.3Hz,1H),7.06-7.02(m,1H),5.41(dd,J=18.0,9.2Hz,2H),3.54(d,J=9.0Hz,1H),3.34(s,3H),3.23(d,J=9.0Hz,1H),2.70(t,J=8.9Hz,1H),2.27-2.13(m,2H),1.97-1.89(m,2H),1.83-1.68(m,5H),1.67-1.43(m,9H),1.41-1.12(m,9H),0.72(s,3H)ppm;APCI MS m/z 498[M+H]+

further elution afforded 81(20mg, 15%) as a white solid: mp 98-100 ℃;1H NMR(500MHz,CDCl3)δ7.86(d,J=8.4Hz,1H),7.31-7.26(m,1H),7.15-7.11(m,1H),5.43(dd,J=18.2,0.95Hz,2H),3.55(d,J=9.0Hz,1H),3.34(s,3H),3.22(d,J=9.0Hz,1H),2.70(t,J=9.0Hz,1H),2.25-2.12(m,2H),1.97-1.75(m,6H),1.70-1.44(m,9H),1.39-1.13(m,10H),0.72(s,3H)ppm;APCI MS m/z 498[M+H]+

further elution afforded 83(48mg, 36%) as an off-white solid: mp 172-174 ℃;1H NMR(500MHz,CDCl3)δ7.67(d,J=8.6Hz,1H),7.35-7.30(m,1H),7.06-7.02(m,1H),5.53(dd,J=17.0,2.6Hz,2H),3.55(d,J=9.0Hz,1H),3.34(s,3H),3.21(d,J=9.0Hz,1H),2.65(t,J=9.0Hz,1H),2.26-2.13(m,3H),1.96-1.89(m,2H),1.83-1.71(m,3H),1.67-1.40(m,9H),1.37-1.11(m,9H),0.74(s,3H)ppm;APCI MS m/z 498[M+H]+

EXAMPLE 45 preparation of Compound 4.

To a solution of Compound A11(40mg, 0.09mmol) in THF (2mL) was added morpholine (390mg, 4.5mmol) and K2CO3(120mg, 0.9 mmol). The resulting solution was stirred at room temperature overnight. LCMS then indicated reaction completion. The reaction was diluted with EtOAc (40mL) and washed with brine (15 mL. times.2). The organic layer was washed with Na2SO4Dried, filtered and concentrated. The residue was purified by preparative HPLC to give compound 4(18mg, 45%) as a white solid. Compound 4:1H NMR:(500MHz,CDCl3),δ(ppm),3.79-3.77(m,4H),3.48(AB,1H,J=10Hz),3.38(AB,1H,J=10Hz),3.30(s,3H),3.22(s,2H),2.58(t,1H,J=9.3Hz),2.5(s,4H),1.25(s,3H),0.66(s,3H)。

example 46 preparation of compound 2.

To a solution of Compound A11(40mg, 0.09mmol) in THF (2mL) was added 1H-pyrazole (300mg, 4.5mmol) and K2CO3(120mg, 0.9 mmol). The resulting solution was stirred at room temperature overnight. LCMS then indicated reaction completion. The reaction was diluted with EtOAc (40mL) and washed with brine (15 mL. times.3). The organic layer was washed with Na2SO4Dried, filtered and concentrated. The residue was purified by preparative HPLC to give 2 as a white solid (16mg, 40%). Compound 2:1H NMR:(500MHz,CDCl3),δ(ppm),7.57(d,1H,J=1Hz),7.43(d,1H,J=1.5Hz),6.35(s,1H),4.98(AB,1H,J=17.5Hz),4.90(AB,1H,J=18Hz),3.48(AB,1H,J=10.5Hz),3.39(AB,1H,J=9.5Hz),3.31(s,3H),2.60(t,1H,J=8.8Hz),1.25(s,3H),0.72(s,3H)。

example 47. preparation of Compound 5.

Mixing compound A11(30mg, 0.07mmol),K2CO3(50mg) and 1- (piperazin-1-yl) ethanone (200mg) were dissolved in THF (3mL) and stirred at room temperature overnight. The solvent was removed in vacuo and the residue was purified by preparative HPLC to give compound 5(6mg, 20%) as a white solid. Compound 5:1H NMR:(400MHz,CDCl3),δ(ppm),3.68-3.66(m,2H),3.51(t,2H,J=5Hz),3.45(AB,1H,J=10Hz),3.37(AB,1H,J=10Hz),3.28(s,3H),3.21(s,2H),2.52-2.44(m,5H),2.08(s,3H),1.23(s,3H),0.64(s,3H)。

example 48 preparation of Compound B11.

Step 1. preparation of compound B2. Compound B1(10.0g, 33mmol) was dissolved in 100mL THF. Dihydropyran (25ml, 270mmol) and PPTS (4.16, 16mmol) were added and the resulting reaction mixture was stirred vigorously at room temperature for 15 hours. After concentration under reduced pressure, the reaction mixture was dissolved in EtOAc (500mL), washed with water (300mL) and brine (300mL), dried over sodium sulfate and concentrated under reduced pressure, and the residue was purified by silica gel chromatography (eluent: petroleum ether/EtOAc: 10/1-3/1) to give 12.52g of compound B2 (97.65%). Compound B2: LC-MS M/z 409.0[ M + Na ]]+

Step 2. preparation of compound B3. Lithium metal (3.0g, 0.4mmol) was added to concentrated ammonia (500ml) in a three-necked flask at-70 ℃. A solution of compound B2(5.0g, 13mmol) and tert-BuOH (0.95g, 13mmol) in dry tetrahydrofuran (100mL) was then added dropwise and stirred for 0.8 h. Ammonium chloride (30.0g) was added to quench the reaction and the ammonia was evaporated overnight. The residue was extracted with EtOAc (300 mL). The organic layer was washed with saturated NaCl solution (2X 200mL) and Na2SO4Dried and concentrated under reduced pressure and the residue purified by silica gel chromatography (eluent: petroleum ether/EtOAc: 10/1-2/1) to give 2.0g of compound 3 (39.60%). Compound B3: LC-MS, M/z 413.3[ M + Na ]]+

Step 3. preparation of compound B4. Mixing Me with water3SOI (16.9g, 76.80mmol) was dissolved in 80mL DMSOAnd NaH (1.84g, 76.80mmol) was added. The mixture was stirred at room temperature for 1 hour, then compound B3(6.0g, 15.36mmol) dissolved in 60mL DMSO was added. The solution was stirred at room temperature overnight. Water (10mL) was then added to the reaction mixture. The aqueous reaction mixture was extracted with EtOAc (300 mL. times.3). Subjecting the extract to Na2SO4Drying, filtering and concentrating. Crude compound B4 was used directly in the next step without further purification.

Step 4. preparation of compound B5. Crude compound B4 was slowly added to LiAlH at 0 ℃4(1.75g, 51mmol) in 100ml of anhydrous THF. The mixture was stirred at room temperature for 2 hours, and then 2.1g of a 15% aqueous NaOH solution was slowly added to quench the reaction. The reaction mixture was extracted with EtOAc (200 mL. times.3). The organic layer was MgSO4Dried, filtered and concentrated. Crude compound B5 was used directly in the next step without further purification.

Step 5. preparation of compound B6. Crude compound B5 was dissolved in 100ml of anhydrous CH2Cl2And 4.0g PCC is added at 0 ℃. The mixture was then stirred at room temperature for 6 hours. The reaction mixture was then filtered, concentrated and purified by flash chromatography on silica gel eluting with 10/1-3/1 petroleum ether ethyl acetate 10/1-3/1 to give compound B6, 3.10g (50.89%, three step yield).

Step 6. preparation of compound B7. In N2To a suspension of ethyltriphenylphosphonium bromide (14.20g, 38.3mmol) in anhydrous THF (40mL) under an atmosphere was added KOtBu (4.30g, 38.3 mmol). The mixture was heated at reflux for 1 hour, at which time the mixture turned bright orange. Compound B6(3.1g, 7.66mmol) in anhydrous THF (25mL) was then added to the above refluxing solution and stirred at reflux overnight. After cooling to room temperature, the solution was poured into brine (100 mL). The aqueous solution was extracted with ethyl acetate (100 mL. times.3). The extract was washed with brine (30 mL. times.2) and Na2SO4Drying, filtration, concentration and purification by silica gel column chromatography (petrol ether/EtOAC: 10/1 to 4/1) gave compound b72.2g (68.97%) as a white solid. In addition, C-3 isomer (0.30g, 9.63%) was obtained.

Step 7. preparation of compound B8. To a solution of compound B7(3g, 7.2mmol) in anhydrous THF (20mL) was added borane-tetrahydrofuran complex (29mL of 1.0M THF solution) and the reaction mixture was stirred at ambient temperature for 1 hour. Aqueous 10% NaOH (20mL) was added slowly. The mixture was cooled in ice and 30% H was added slowly2O2Aqueous solution (20 mL). The mixture was stirred at ambient temperature for 1 hour, then CH was used2Cl2(3X 100 mL). To merge CH2Cl2The extract is extracted with 10% Na2S2O3The aqueous solution (50mL) was washed and used directly in the next step without further purification.

Step 8. preparation of compound B9. Combined CH of Compound B8 of the last step2Cl2The extract was used without further purification. 3.5g PCC was added at 0 ℃. The mixture was then stirred at room temperature for 6 hours. The mixture was filtered, concentrated and purified by flash chromatography on silica gel eluting with 12/1-7/1 (petroleum ether: ethyl acetate) to give 1.28g of compound B9 (41.23%, two steps). Compound B9: LC-MS M/z 455.3[ M + Na ]]+1H NMR (500MHz, CDCl3) δ (ppm) 4.57 and 4.53(1H, t, J ═ 3.5Hz),3.96 and 3.87(1H, AB, J ═ 11.0 Hz), 3.82(1H, t, J ═ 9.5Hz),3.56 to 3.53(1H, m),3.44 and 3.27(1H, AB, J ═ 10.5Hz),2.53(1H, t, J ═ 9.0Hz),2.12 and 2.11(3H, s),1.22 and 1.21(3H, s),0.64 and 0.61(1H, s).

Step 9 preparation of compound B10. Compound B9(1.28g, 2.96mmol) was dissolved in 50mL anhydrous MeOH and 100mg PTSA was added. The reaction mixture was stirred at room temperature overnight. The reaction mixture was then concentrated under reduced pressure. This product mixture was isolated by flash chromatography on silica gel eluted with 8/1-2/1 (petroleum ether: EtOAc) to give 674mg of Compound B10 (65.32%). Compound B10: LC-MS (sodium chloride-sodium chloride) with M/z being 331.3[ M-H2O + H]+,m/z=349.2[M+H]+1H NMR(500MHz,CDCl3)δ(ppm):3.89(1H,AB,J=12.0Hz),3.72(1H,AB,J=12.0Hz),2.53(1H,t,J=9.0Hz),2.11(3H,s),1.22(3H,s),0.64(3H,s)。13C NMR(125.77MHz,CDCl3)δ(ppm):209.78,69.68,63.80,60.12,57.07,54.39,44.36,42.04,41.18,39.62,39.31,36.05,35.39,31.85,31.68,31.54,28.04,27.91,24.39,22.86,22.75,13.72。

Step 10. preparation of compound B11. Compound B10(50mg, 0.14mmol) was dissolved in 5mL anhydrous MeOH and 3 drops of Br were added2And 2 drops of aqueous HBr. The reaction mixture was stirred at room temperature for 3 hours, then treated with triethylamine at 0 ℃, concentrated under reduced pressure and used directly in the next step without further purification. Compound B11: LC-MS (liquid chromatography-mass spectrometry) with m/z being 410.1&411.2[M-H2O+H]+

Example 49 preparation of compound 9.

Crude Compound B11 was used as received, 8mL THF and 100mg K were added2CO30.5ml morpholine. The reaction mixture was stirred at room temperature overnight. The solution was diluted with ethyl acetate (100 mL). The resulting solution was washed with brine (100mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by reverse phase preparative HPLC to give 25mg (41.18%, two steps, from 50mg compound B10) of product 9 as a white solid. Compound 9:1H NMR(500MHz,CDCl3)δ(ppm):3.89(1H,dd,J=4.0Hz,J=11.5Hz),3.76(4H,t,J=4.5Hz),3.72(1H,dd,J=3.0Hz,J=11.0Hz),3.19(2H,s),2.58(1H,t,J=9.5Hz),2.45-2.55(4H,m),2.20~2.15(1H,m),2.07~2.04(1H,m),1.92~1.89(1H,m),1.23(3H,s),0.67(3H,s)。

example 50 preparation of compounds 12 and 10.

Crude compound B11 was used as received and 8mL of anhydrous THF and 100mg K were added2CO30.5ml of 1H-1,2, 3-triazole. The reaction mixture was stirred at room temperature overnight. The solution was diluted with EtOAc (100 mL). The resulting solution was washed with brine (100mL), dried over sodium sulfate and concentrated in vacuo. Purification of the residue by reverse phase preparative HPLCThe remainder, 10mg of compound 12 as a white solid and 19mg of compound 10 were obtained.

Compound 12:1H NMR(500MHz,CDCl3)δ(ppm):7.76(1H,s),7.64(1H,s),5.24(1H,AB,J=17.5Hz),5.16(1H,AB,J=18.0Hz),3.89(1H,AB,J=11.5Hz),3.73(1H,AB,J=11.5Hz),2.65(1H,t,J=9.0Hz),2.25~2.19(1H,m),2.10~2.05(2H,m),1.23(3H,s),0.71(3H,s)。

compound 10:1H NMR(500MHz,CDCl3)δ(ppm):7.68(2H,s),5.25(1H,AB,J=17.5Hz),5.22(1H,AB,J=17.5Hz)3.89(1H,AB,J=11.5Hz),3.73(1H,AB,J=11.5Hz),2.58(1H,t,J=8.5Hz),2.24~2.20(1H,m),2.11~2.04(21H,m),1.23(3H,s),0.75(3H,s)。

example 51 preparation of compound 11.

Crude compound B11 was used as received, 8mL anhydrous THF and 100mg K were added2CO30.5ml of pyrazole. The reaction mixture was stirred at room temperature overnight. The solution was diluted with ethyl acetate (100 mL). The resulting solution was washed with brine (100mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by reverse phase preparative HPLC to give 30mg of compound 11 as a white solid. Compound 11:1H NMR(500MHz,CDCl3)δ(ppm):7.54(1H,d,J=1.0Hz),7.40(1H,dt,J=2.0Hz),6.33(1H,t,J=1.5Hz),4.94(1H,AB,J=17.5Hz),4.90(1H,AB,J=17.0Hz),3.88(1H,AB,J=11.5Hz),3.73(1H,AB,J=12.0Hz),2.58(1H,t,J=8.5Hz)2.23~2.17(1H,m),2.07~2.05(2H,m),1.23(3H,s),0.72(3H,s)。

example 52. preparation of compound 6.

Crude compound B11 was used as received, 8mL anhydrous THF and 100mg K were added2CO30.5ml of 1- (piperazin-1-yl) ethanone. The reaction mixture was stirred at room temperature overnight. Then using ethyl acetateThe solution was diluted (100 mL). The resulting solution was washed with brine (100mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by reverse phase preparative HPLC to give 29mg (43.64%, two steps, from 50mg compound B10) of compound 6 as a white solid. Compound 6:1H NMR(500MHz,CDCl3)δ(ppm):3.89(1H,AB,J=11.5Hz),3.72(1H,AB,J=11.5Hz),3.68~3.65(2H,m),3.51(2H,t,J=5.0Hz)3.21(3H,s),2.55(1H,t,J=9.5Hz),2.45(3H,t,J=5.0Hz),1.23(3H,s),0.67(3H,s)

example 53 preparation of compound 7.

59mg (0.12mmol) of crude compound B11 were dissolved in 8mL THF and 100mg (0.77mmol) K was added2CO3100mg (0.61mmol) of 1- (methylsulfonyl) piperazine. The reaction mixture was stirred at room temperature overnight. The solution was diluted with ethyl acetate (100 mL). The resulting solution was washed with brine (100mL), dried over sodium sulfate and concentrated in vacuo. The residue was purified by reverse phase preparative HPLC to give 8mg (0.02mmol, 10.9%) of product 7 as a white solid. Compound 7:1H NMR(500MHz,CDCl3)δ(ppm):3.89(1H,AB,J=11.5Hz),3.72(1H,AB,J=12Hz),3.30(4H,t,J=5.0Hz),3.25(2H,s),2.78(3H,s)2.61(4H,t,J=5.0Hz),2.52(1H,t,J=8.5Hz),2.13~2.01(1H,m),1.23(3H,s),0.67(3H,s)。

example 54 preparation of Compound E15.

Step 1. preparation of E2. To a solution of E1(250.0g, 0.83mol) in pyridine (1L) was added Ac dropwise at 19 deg.C2O (168.8g, 1.65 mol). After the addition was complete, the reaction mixture was stirred at 19 ℃ overnight. TLC (petroleum ether: ethyl acetate 1:1) showed the reaction was complete. The reaction mixture was then concentrated in vacuo, the residue poured into water and extracted with dichloromethane (3X 500mL), the organic phase was washed with waterLayer was washed with 2N HCl (200mL), saturated NaHCO3(300mL), washed with brine and dried over anhydrous sodium sulfate, filtered, and concentrated to give the crude target product E2 as a brown oil (283.7g, 99.7%).1H NMR(E2):(400MHz,CDCl3)δ5.95-5.90(m,1H),4.70-4.64(m,1H),4.20-4.13(m,1H),2.68-2.55(m,1H),2.54-2.30(m,5H),2.15-1.90(m,6H),1.90-1.75(m,4H),1.63-1.38(m,2H),1.33-1.07(m,4H),0.90(s,3H)。

Step 2. preparation of E3. To a solution of E2(250.0g, 0.73mol) in 1, 4-dioxane (700mL) and EtOH (467mL) at 29 deg.C was added CH (OEt)3(227.2g, 1.53mol) and p-TsOH (2.8g, 14.60 mmol). After the addition was complete, the reaction mixture was stirred at 29 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate 3:1) showed the reaction was complete. The reaction mixture was then washed with saturated NaHCO3Quench (300mL) and pour into water, extract with EtOAc (3 × 500mL), wash the organic layer with brine and dry over anhydrous sodium sulfate, filter, concentrate, and recrystallize from petroleum ether ethyl acetate 10:1 to give the target product E3(155.8g, 57.6%) as a white solid.1H NMR(E3):(400MHz,CDCl3)δ5.42-5.38(m,1H),5.15-5.10(m,1H),4.46-4.50(m,1H),4.32-4.25(m,1H),4..05-3.98(m,1H),3.85-3.70(m,2H),2.53-2.42(m,1H),2.37-2.20(m,3H),2.18-1.91(m,7H),1.90-1.72(m,3H),1.62-1.48(m,2H),1.40-1.20(m,6H),1.20-1.12(m,1H),0.90(s,3H)。

Step 3. preparation of E4. In N2To a solution of E3(30.0g, 80.54mmol) in EtOAc (400mL) was added Pd/C (1.5g, 50% water) next. The reaction mixture was degassed under vacuum and washed with H2And (5) purifying for several times. The reaction mixture was then brought to 15 ℃ under H2Stirred under atmosphere for 1 hour. It was then filtered and the filtrate was stirred at 15 ℃, 10% HCl (100mL) was added and the reaction mixture was stirred at 15 ℃ for 1 hour. TLC (petroleum ether: ethyl acetate 3:1) showed the reaction was complete. The reaction mixture was poured into water and extracted with EtOAc (3X 200mL), and the organic layer was extracted with saturated NaHCO3Washed with brine and dried over anhydrous sodium sulfate, filtered, concentrated and purified by silica gel column (petroleum ether: ethyl acetate 10:1-3:1) to give the product E4(33.0g, yield: 59.1%) as a colorless oil.1H NMR(E4):(400MHz,CDCl3)δ4.75-4.50(m,1H),4.45-4.38(m,1H),2.55-2.35(m,5H),2.31-2.20(m,1H),2.15-2.05(m,4H),2.05-1.65(m,6H),1.60-1.15(m,7H),1.15-1.00(m,1H),0.95-0.85(m,4H)。

And 4. preparation of E5. To a solution of E4(31.0g, 89.48mmol) and ethane-1, 2-diol (50mL) in toluene (200mL) at 16 deg.C was added a catalytic amount of pyridine HCl (0.3g, 2.60 mmol). After the addition was complete, the reaction mixture was heated to reflux and water was removed through a Dean-Stark trap for 18 hours. TLC (petroleum ether: ethyl acetate 3:1) showed the reaction was complete. The reaction mixture was then cooled to 16 ℃, poured into water and extracted with EtOAc (3 × 100mL), the organic layer was washed with brine and dried over anhydrous sodium sulfate, filtered, and concentrated to give crude product E5(36.9g, yield: 94.8%) as a white solid, which was used in the next step.1H NMR(E5):(400MHz,CDCl3)δ4.40-4.30(m,2H),4.20-4.10(m,2H),4.00-3.80(m,8H),2.20-1.90(m,5H),1.85-1.05(m,20H),1.05-0.75(m,5H)。

Step 5. preparation of E6. To a solution of E5(55.0g, 126.56mmol) in THF (200mL) and MeOH (50mL) at 20 deg.C was added 4N LiOH (94.9mL, 379.69 mmol). After the addition was complete, the reaction mixture was stirred at 20 ℃ for 18 hours. TLC (petroleum ether: ethyl acetate 3:1) showed the reaction was complete. The reaction mixture was then poured into water and extracted with EtOAc (3 × 200mL), the organic layer was washed with brine and dried over anhydrous sodium sulfate, filtered, and concentrated to give crude product E6(46.3g, yield: 93.1%) as a white solid.1H NMR(E6):(400MHz,CDCl3)δ4.00-3.75(m,10H),2.25-2.15(m,1H),2.05-1.90(m,1H),1.85-1.60(m,8H),1.60-1.40(m,7H),1.35-1.00(m,5H),1.00-0.75(m,5H)。

And 6. preparation of E7. At 25 ℃ under N2To a suspension of 60% NaH (9.6g, 0.24mol) in anhydrous THF (100mL) was added dropwise a solution of compound E6(46.3g, 0.12mol) in anhydrous THF (200 mL). The mixture was stirred for 30 minutes, then MeI (51.1g, 0.36mol) was added dropwise at 25 ℃. After the addition was complete, the reaction mixture was stirred at 45 ℃ for 4 hours. TLC (petroleum ether: ethyl acetate 3:1) showed the reaction was complete. The reaction was cooled to room temperature and saturated NH added4Quenched with Cl (200mL), poured into water, extracted with EtOAc (3 × 200mL), and the organic layer washed with brine and dried over anhydrous sodium sulfate, filtered, and concentrated to give crude product E7(50.0g, crude) as a yellow solid.1H NMR(E7):(400MHz,CDCl3)δ3.95-3.80(m,8H),3.55-3.40(m,2H),3.28(s,3H),2.18-2.10(m,1H),2.01-1.95(m,1H),1.81-1.57(m,8H),1.46-1.37(m,8H),1.29-1.18(m,4H),1.02-0.85(m,7H)。

Step 7. preparation of E8. To a solution of compound E7(50.0g, 0.12mol) in THF (200mL) and acetone (40mL) was added 2N aqueous HCl (40 mL). After the addition was complete, the reaction mixture was stirred at 25 ℃ for 18 hours. TLC (petroleum ether: ethyl acetate 3:1) indicated the reaction was complete. The reaction mixture was then poured into water, extracted with EtOAc (3X 200mL), and the organic layer was extracted with saturated NaHCO3Washed with brine and dried over anhydrous sodium sulfate, filtered and concentrated to give crude product E8(41.0g, crude) as a yellow solid which was used directly in the next step.1H NMR(E8):(400MHz,CDCl3)δ3.73-3.58(m,2H),3.34(s,3H),2.55-2.40(m,3H),2.42-2.30(m,1H),2.20-1.65(m,8H),1.60-1.20(m,10H),1.10-0.75(m,7H)。

Step 8. preparation of E9. At-78 ℃ under N2To a solution of E8(40.0g, 125.7mmol) in dry THF (500mL) was added K-selectride (151mL, 150.8mmol, 1M in THF) dropwise. After the addition was complete, the reaction mixture was stirred at-78 ℃ for 3 hours. TLC (petroleum ether: ethyl acetate 3:1) indicated the reaction was complete. Using 30% H at-78 deg.C2O2(17.1g, 150.5mmol) the reaction mixture was quenched slowly and then poured into saturated NH4In Cl, extract with EtOAc (3X 100 mL). The organic phase was saturated with Na2S2O3Washed with brine, dried over sodium sulfate and evaporated to give the crude product. The crude product was purified by washing with PE: EtOAc 10:1 to give the title product E9(23.5g, 58%) as a white solid.1H NMR(E9):(400MHz,CDCl3)δ4.10(m,1H),3.52(d,1H),3.42(d,1H),3.30(s,3H),2.50-2.40(m,1H),2.16-2.03(m,1H),1.98-1.90(m,2H),1.86-1.64(m,10H),1.55-1.47(m,3H),1.32-1.17(m,6H),1.12-0.95(m,1H),0.88-0.81(m,4H)。

Step 9. preparation of E10. In N2At 0 ℃ to Ph3To a suspension of PEtBr (25.97g, 70mmol) in anhydrous THF (100mL) was added dropwise a solution of t-BuOK (7.70g, 70mmol) in anhydrous THF (50 mL). The mixture was stirred at room temperature for 1.5 hours. A solution of E9(2.8g, 8.75mmol) in THF (30mL) was then added dropwise and the resulting mixture was stirred at 60 ℃ for 12 hours. TLC (petroleum ether: ethyl acetate ═ 3:1) indicated complete consumption of starting material. The reaction mixture is saturated with NH4Aqueous Cl (100mL) was quenched and extracted with EtOAc (50 mL. times.2). The combined organic phases are passed over Na2SO4Dried and concentrated in vacuo. The residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate 40:1) to give E10(1.8g, 62%) as a white powder.1H NMR(E10):(400MHz,CDCl3)δ5.15-5.05(m,1H),4.13-4.05(m,1H),3.55-3.35(m,2H),3.30(s,3H),2.45-1.90(m,4H),1.80-1.35(m,13H),1.30-0.95(m,8H),0.90(s,3H),0.85-0.70(m,1H)。

Step 10. preparation of E11. To a solution of E10(1.8g, 5.41mmol) in DMF (20mL) was added imidazole (737mg, 10.82mmol) and TBSCl (1.22g, 8.12 mmol). The mixture was stirred at room temperature overnight. TLC (petroleum ether: ethyl acetate ═ 3:1) showed complete consumption of starting material. The reaction was diluted with EtOAc (20mL) and washed with brine (20 mL). The organic layer was dried over anhydrous sodium sulfate and concentrated under vacuum. The crude product was purified by silica gel chromatography eluting with petroleum ether on silica gel to give E11(2.33g, 96%) as a white solid.1H NMR(E11):(400MHz,CDCl3)δ5.11-5.09(m,1H),4.01-4.00(m,1H),3.50-3.39(m,2H),3.29(s,3H),2.40-2.30(m,1H),2.25-2.10(m,2H),1.90-1.82(m,1H),1.53-1.40(m,5H),1.30-0.95(m,8H),0.89-0.88(m,9H),0.86-0.75(m,2H),0.02-0.01(m,6H)。

Step 11. preparation of E12. To a solution of 9-BBN (81mL, 40.84mmol) was added THF (20mL) containing E11(2.33g, 5.06 mmol). The mixture was stirred at 60 ℃ for 16 hours. The mixture was then cooled to room temperature and 10% aqueous NaOH (40mL) and H were added dropwise2O2(20 mL). After stirring for 1 hour, the mixture was washed with Na2S2O3Aqueous quench and extraction with EtOAc (100mL). The organic layers were combined and dried over anhydrous sodium sulfate. The organic phase was concentrated under vacuum. The crude product was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate 40:1) to give E12(2.3g, 90%) as a white solid.1H NMR(E12):(400MHz,CDCl3)δ4.00-3.99(m,1H),3.71-3.65(m,1H),3.49-3.38(m,2H),3.28(s,3H),2.42-2.39(m,1H),1.89-1.82(m,5H),1.75-1.46(m,17H),1.38-1.00(m,14H),0.89(s,9H),0.68(s,3H),0.00(m,6H)。

Step 12. preparation of E13. To E12(2.1g, 4.52mmol) in CH2Cl2(20mL) was added Dess-Martin (3.8g, 9.04 mmol). The mixture was stirred at 12 ℃ for 5 hours. TLC (petroleum ether: ethyl acetate ═ 3:1) showed complete consumption of starting material. With Na2S2O3/NaHCO3The mixture was quenched with a solution of a mixture (3:1, 12g) in water (50 mL). The mixture was extracted with EtOAc (200 mL). The organic layer was dried over anhydrous sodium sulfate. The organic phase was concentrated in vacuo to give E13(2.3g, crude) as a white solid.1H NMR(E13):(400MHz,CDCl3)4.00-3.99(m,1H),3.49-3.38(m,2H),3.27(s,3H),2.41-2.39(m,2H),2.20-2.12(m,1H),2.11(s,3H),2.00-1.95(m,1H),1.89-1.84(m,3H),1.75-1.45(m,14H),1.42-0.92(m,11H),0.89(s,9H),0.87-0.82(m,1H),0.62(s,3H),0.00(m,6H)。

Step 13. preparation of E14. To a solution of E13(230mg, 0.48mmol) in CH2Cl2To the solution in (6mL) was added TFA (1 mL). The mixture was stirred at 15 ℃ for 30 minutes. TLC (petroleum ether: ethyl acetate ═ 3:1) showed complete consumption of starting material. The reaction is treated with NaHCO3Aqueous solution (10mL) was quenched and extracted with EtOAc (30 mL. times.2). The organic layer was dried over anhydrous sodium sulfate. The organic phase was concentrated under vacuum. The crude product was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate ═ 10:1) to give E14(166mg, 95%) as a white solid.1H NMR(E14):(400MHz,CDCl3)4.12-4.11(m,1H),3.52-3.42(m,2H),3.30(s,3H),2.58-2.53(m,1H),2.20-2.16(m,1H),2.13(s,3H),2.06-1.97(m,2H),1.75-1.60(m,8H),1.59-1.10(m,11H),1.03-0.92(m,1H),0.90-0.82(m,1H),0.63(s,3H)。

Step 14. preparation of E15. To E14(2g, 5.21mmol) in MeOH (25mL)HBr (5 drops) and Br were added to the solution of (1)2(8 mL). The mixture was stirred at room temperature for 4 hours. LC-MS showed the starting material was consumed. Reacting with H2O (20mL) was diluted and extracted with EtOAc (50 mL). The organic layer was dried over anhydrous sodium sulfate. The organic phase was concentrated under vacuum. The crude product was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate ═ 10:1) to give E15(1g, 41%) as a white solid.1H-NMR showed the presence of 70% E15 and 30% E14.1H NMR(E15):(400MHz,CDCl3)4.11-4.10(m,1H),3.94-3.88(m,2H),3.49-3.39(m,2H),3.28(s,3H),2.84-2.79(m,1H),2.20-2.15(m,1H),2.04-1.87(m,2H),1.74-1.60(m,8H),1.57-1.16(m,12H),1.03-0.76(m,2H),0.64(s,3H)。

Example 55 preparation of compounds 90 and 91.

To K2CO3(952mg, 6.9mmol) to a solution in DMF (20mL) was added 2H-1,2, 3-triazole (969mg, 13.8 mmol). The mixture was stirred at room temperature for 30 min, then a solution of E15(1g, 2.3mmol) in DMF (20mL) was added. The mixture was stirred at room temperature overnight. TLC (petroleum ether: ethyl acetate ═ 1:1) showed complete consumption of starting material. The reaction was diluted with EtOAc (50mL) and washed with brine (50 mL). The organic layer was dried over anhydrous sodium sulfate. The organic phase was concentrated under vacuum. The crude product was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate ═ 5:1) to give 90(204mg, 21%) and 91(437mg, 45%) as white powders.1H NMR(90):(400MHz,CDCl3)7.70(s,2H),5.31-5.20(m,2H),4.13-4.12(m,1H),3.52-3.43(m,2H),3.31(s,3H),2.63-2.58(m,1H),2.24-2.22(m,1H),2.12-1.97(m,2H),1.78-1.62(m,9H),1.54-1.10(m,10H),1.09-0.96(m,1H),0.91-0.84(m,1H),0.75(s,3H)。1H NMR(91):(400MHz,CDCl3)7.75(s,1H),7.63(s,1H),5.27-5.11(m,2H),4.13-4.10(m,1H),3.50-3.40(m,2H),3.28(s,3H),2.67-2.64(m,1H),2.26-2.18(m,1H),2.08-1.92(m,2H),1.74-1.25(m,15H),1.19-0.87(m,8H),0.69(s,3H)。

EXAMPLE 56 preparation of Compound 92.

Mixing E15(200mg, 0.45mmol) and K2CO3A mixture of (188mg, 1.36mmol), ethyl 1H-pyrazole-3-carboxylate (317mg, 2.27mmol) and DMF (2mL) was stirred at 25 ℃ for 12H. TLC showed the reaction was complete. The mixture was diluted with EtOAc (20mL), washed with brine (30 mL. times.3), and the organic layer was washed with anhydrous Na2SO4Dried and then concentrated to give the crude product. Purification by column chromatography (petroleum ether: ethyl acetate 4:1) gave 92(96mg, 44.4%) as a white solid.1H NMR(92):(400MHz,CDCl3)δ7.42(d,J=2.4Hz,1H),6.86(d,J=2.4Hz,1H),5.05-4.95(m,2H),4.39(q,J=7.2Hz,2H),4.20-4.10(m,1H),3.49-3.39(m,2H),3.28(s,3H),2.60-2.55(m,1H),2.22-1.91(m,3H),1.75-1.50(m,6H),1.45-1.10(m,15H),1.05-0.92(m,1H),0.90-0.84(m,1H),0.70(s,3H)。

Example 57 preparation of compound 93.

E15(200mg, 0.47mmol) in CH at 29 deg.C3CN (15mL), then K was added to the mixture at 29 deg.C2CO3(194.01g, 1.4mmol) and 4-methyl-1H-pyrazole (192.09mg, 2.34 mmol). The solution was stirred at 60 ℃ for 10 hours. After TLC showed complete consumption of starting material, the mixture was then concentrated. By CH2Cl2The mixture was extracted (30mL) and NaCl (aq) (20 mL. times.2). The combined organic layers were passed over Na2SO4Dried and concentrated to give the crude product. The product was purified by silica gel column chromatography eluting with (petroleum ether/ethyl acetate ═ 10:1 to 6:1) to give 93(94.4mg, yield: 46.81%).1H NMR(93):(400MHz,CDCl3)δ7.36(s,1H),7.18(s,1H),4.85(dd,J=34Hz,J=17.6Hz,2H),4.15-4.10(m,1H),3.47(dd,J=28.8Hz,J=10Hz,2H),3.31(s,3H),2.62-2.55(m,1H),2.28-2.18(m,1H),2.13-1.97(m,5H),1.78-1.64(m,8H),1.48-1.13(m,10H),1.04-0.84(m,2H),0.71(s,3H)。

Example 58 preparation of compounds 94, 95 and 96.

To a solution of E15(150mg, 0.35mmol) in DMF (6mL) at 28 ℃ was added Cs2CO3(343mg, 1.05mmol) and 4-methyl-1, 2, 3-triazole (145mg, 1.75 mmol). The reaction mixture was stirred at the same temperature for 6 hours. TLC showed complete consumption of starting material. The mixture was poured into water (20mL) and extracted with EtOAc (10 mL. times.2). The combined organic layers were washed with Na2SO4Dried and concentrated to give the crude product. The crude product was purified by preparative HPLC to give pure 94(35.8mg) and a mixture of 95 and 96 (20 mg). The mixture was then purified by SFC to give 95(3.9mg) and 96(5.6 mg). Total yield: 23.7 percent. The structures of the three targets were confirmed by NOE.

1H NMR(94):(400MHz,CDCl3)δ7.42(s,1H),5.19-5.08(m,2H),4.13-4.09(m,1H),3.53-3.38(m,2H),3.29(s,3H),2.61-2.54(m,1H),2.33(s,3H),2.26-2.15(m,1H),2.11-1.95(m,2H),1.78-1.61(m,9H),1.52-1.06(m,9H),1.04-0.81(m,2H),0.72(s,3H)。

1H NMR(95):(400MHz,CDCl3)δ7.50(s,1H),5.23-5.05(m,2H),4.15-4.06(m,1H),3.58-3.42(m,2H),3.31(s,3H),2.71-2.61(m,1H),2.33-2.14(m,6H),2.12-1.96(m,4H),1.34-1.18(m,10H),1.06-0.83(m,6H),0.70(s,3H)。

1H NMR(96):(400MHz,CDCl3)δ7.35(s,1H),5.22-5.02(m,2H),4.15-4.10(m,1H),3.56-3.39(m,2H),3.31(s,3H),2.67-2.61(m,1H),2.39(s,3H),2.29-1.88(m,10H),1.33-1.22(m,10H),1.02-0.97(m,1H),0.92-0.84(m,2H),0.70(s,3H)。

Preparation of examples 59.97 and 98.

To a solution of A11(360mg, 0.82mmol) in acetone (2.5mL) was added 5, 6-difluoro-2H-benzo [ d [ -d ] ]][1,2,3]Triazole (BB-1) (190mg, 1.23mmol) and K2CO3(230mg, 1.64 mmol). The mixture was stirred at 30 ℃ for 3 hours. TLC showed the reaction was complete. To the mixture was added water (2mL) and extracted with EtOAc (5 mL. times.2). The combined organic layers were concentrated in vacuo and purified by preparative HPLC to give 97(25mg, 6%) and 98(141mg, 33%) as white solids.

1H NMR(97):(400MHz,CDCl3)δ7.59(t,J=8.4Hz,2H),5.53-5.42(m,2H),3.48-3.36(m,2H),3.29(s,3H),2.65(t,J=8.4Hz,1H),2.27-2.02(m,3H),1.80-0.85(m,23H),0.75(s,3H)。LCMS(97):tR1.366 min, in 2 min chromatography, 10-80AB, purity 96.3%, MS ESI: with respect to C29H40F2N3O3Calculated value of [ M + H ]]+516, found 498([ M + H-18)]+)。

1H NMR(98):(400MHz,CDCl3)δ7.83(t,J=8.0Hz,1H),7.12(t,J=7.6Hz,1H),5.43-5.32(m,2H),3.50-3.37(m,2H),3.30(s,3H),2.80-2.68(m,1H),2.30-2.02(m,3H),1.80-0.85(m,23H),0.75(s,3H)。LCMS(98):tR1.317 min, in 2 min chromatography, 10-80AB, purity 99.6%, MS ESI: with respect to C29H40F2N3O3Calculated value of [ M + H ]]+516, found 516.

Scheme a. general procedure for the preparation of compounds 99-146.

EXAMPLE 76 preparation of Compounds 154 and 155.

To a solution of compound D6(300mg, 0.679mmol) in acetone (5mL) was added K2CO3(186mg, 1.35mmol) and 2H-pyrazolo [3,4-b]Pyrazine (120mg, 1.01 mmol). After stirring at 25 ℃ for 3h, LCMS showed the reaction was complete, one product (27%) and one product (21%). The reaction mixture was concentrated in vacuo to remove most of the solvent to give a residue. The residue was dissolved in EtOAc (50mL) and washed with water (50 mL). The reaction mixture was extracted with EtOAc (50 mL. times.3). The organic layers were combined and concentrated in vacuo to give the crude product. The crude product was purified by preparative hplc (fa) to give compound 154 as a white solid (4mg,1.22%) and compound 155(4mg, 1.22%) as a white solid.

1H NMR (154) (1.2% yield): (400MHz, CDCl)3)δ8.64(d,J=2.0Hz,1H),8.56(d,J=2.0Hz,1H),8.26(s,1H),5.40-5.19(m,2H),3.54(d,J=8.4Hz,1H),3.33(s,3H),3.20(d,J=9.2Hz,1H),2.68(t,J=8.8Hz,1H),2.30-1.20(m,26H),0.72(d,J=13.2Hz,3H)。LCMS tR1.155 min, in 2 min chromatography, 10-80AB, purity 96.2%, MS ESI: with respect to C28H41N4O3Calculated value of [ M + H ]]+481, found 481.

1H NMR (155) (1.2% yield): (400MHz, CDCl)3)δ8.59(d,J=2.4Hz,1H),8.44(d,J=2.0Hz,1H),8.34(s,1H),5.36-5.24(m,2H),3.55(d,J=9.2Hz,1H),3.34(s,3H),3.22(d,J=9.2Hz,1H),2.68(t,J=8.8Hz,1H),2.30-1.20(m,26H),0.71(s,3H)。LCMS tR0.854 min, in 1.5 min chromatography, 10-80AB, 96.8% purity, MS ESI: with respect to C28H41N4O3Calculated value of [ M + H ]]+481, found 463[ M + H-18]+

Example 77 preparation of compound 156.

1H NMR (156) (41% yield): (400MHz, CDCl)3)δ7.88(s,1H),7.83(s,1H),5.07-4.87(m,2H),3.55(d,J=9.0Hz,1H),3.35(s,3H),3.22(d,J=9.0Hz,1H),2.65-2.58(m,1H),2.28-2.15(m,1H),2.10-2.01(m,1H),1.97-1.91(m,2H),1.82-1.40(m,14H),1.35-1.09(m,8H),0.68(s,3H)。LCMS tR2.744 min, in 4 min chromatography, 10-80AB, purity 100.0%, MS ESI: with respect to C27H39N3O3Calculated value of Na [ M + Na ]]+477, measured 477.

EXAMPLE 78 preparation of Compound 147.

1H NMR (147) (yield 4%): (400MHz, CDCl)3)δ8.03(s,1H),7.68(d,J=8.5Hz,1H),7.22(s,1H),7.15(d,J=8.5Hz,1H),5.20-5.03(m,2H),3.57(d,J=9.0Hz,1H),3.36(s,3H),3.23(d,J=9.0Hz,1H),2.65(t,J=8.7Hz,1H),2.30-2.09(m,2H),1.99-1.91(m,2H),1.77-1.45(m,14H),1.34-1.14(m,8H),0.73(s,3H)。LCMS tR3.185 min, in 4 min chromatography, 10-80AB, purity 100.0%, MS ESI: with respect to C30H42ClN2O3Calculated value of [ M + H ]]+513, found value 513.

EXAMPLE 79 preparation of Compound 157.

1H NMR (157) (3.5% yield): (400MHz, CDCl)3)δ7.47-7.43(m,1H),7.14-7.06(m,2H),5.49-5.38(m,2H),3.55(t,J=9.2Hz,1H),3.36(d,J=4.0Hz,3H),3.25(d,J=8.8Hz,1H),2.73(s,1H),2.30-1.20(m,26H),0.74(s,3H)。LCMS tR0.931 min, in 1.5 min chromatography, 5-95AB, purity 96.8%, MS ESI: with respect to C29H41FN3O3Calculated value of [ M + H ]]+498, found 520[ M + Na]+

EXAMPLE 80 preparation of Compound 158.

1H NMR (158) (yield 18%): (400MHz, CDCl)3)δ7.50(d,J=2.5Hz,1H),6.75(d,J=2.5Hz,1H),5.08-4.90(m,2H),3.55(d,J=9.0Hz,1H),3.37-3.31(m,3H),3.22(d,J=9.0Hz,1H),2.64-2.57(m,1H),2.28-01(m,2H),1.99-1.91(m,2H),1.84-1.40(m,14H),1.39-1.08(m,8H),0.67(s,3H)。LCMS tR2.821 min, in 4 min chromatography, 10-80AB, purity 100.0%, MS ESI: aboutC27H39N3O3Calculated value of Na [ M + Na ]]+476, found 476.

Example 81 preparation of compound 159.

1H NMR (159) (yield: 13%): (400MHz, CDCl)3)δ7.48(s,1H),6.60(d,J=2.0Hz,1H),5.07-4.88(m,2H),3.56(d,J=9.0Hz,1H),3.35(s,3H),3.22(d,J=9.0Hz,1H),2.65-2.54(m,1H),2.28-2.15(m,1H),2.10-2.02(m,1H),1.99-1.90(m,2H),1.84-1.37(m,14H),1.36-1.07(m,8H),0.68(s,3H)。LCMS tR3.049 min, in 4 min chromatography, 10-80AB, purity 100.0%, MS ESI: with respect to C27H40F3N2O3Calculated value of [ M + H ]]+497, found 479[ M + H-H2O]+

Example 82. preparation of compound 160.

1H NMR (160) (11% yield): (400MHz, CDCl)3)δ8.59(s,1H),5.47(s,2H),3.56(d,J=9.0Hz,1H),3.35(s,3H),3.23(d,J=9.0Hz,1H),2.66(t,J=8.8Hz,1H),2.30-2.16(m,1H),2.15-2.05(m,1H),1.98-1.89(m,2H),1.84-1.41(m,14H),1.40-1.11(m,9H),0.72(s,3H)。LCMS tR2.667 min, in 4 min chromatography, 10-80AB, purity 100.0%, MS ESI: with respect to C24H39N4O3Calculated value of [ M + H ]]+431, found 413[ M + H-18]+

Example 83 preparation of compound F5.

Step 1. preparation of compound F1.To a solution of C2(2g, 6.28mmol) in THF (30mL) in a flask at 0 deg.C was added CsF (953mg, 6.28mmol) followed by dropwise addition of TMSCF3(1.33g, 9.42 mmol). The reaction was warmed to 25 ℃ and stirred for 2 hours. TLC (PE: EtOAc ═ 3:1) showed complete consumption of starting material. The reaction mixture was then treated with 2M aqueous HCl (10mL) and stirred for 6 hours. Then the reaction is reacted with H2O (30mL) was diluted and extracted with EtOAc (30 mL. times.2). The combined organic layers were washed with brine (20mL) and Na2SO4Drying and concentration gave the crude product which was purified by silica gel column (PE: EtOAc 50:1 to 10:1) to give product F1(1.1g, 45.0% yield) as a yellow oil.1H NMR(400MHz,CDCl3)δ3.49(d,J=8.0Hz,1H),3.32-3.22(m,4H),2.46-2.39(m,1H),2.10-1.71(m,8H),1.68-1.10(m,14H),0.85(s,3H)。

Step 2. preparation of compound F2. To a solution of ethyltriphenylphosphonium bromide (5.19g, 14.0mmol) in THF (30mL) was added t-BuOK (1.57g, 14.0 mmol). The reaction mixture was heated to 60 ℃ for 1 hour and F1(1.1g, 2.83mmol) was added to the mixture, which was stirred at 60 ℃ for a further 8 hours. TLC (PE: EtOAc ═ 3:1) showed the reaction was complete. The reaction mixture was cooled and then washed with H2O (30mL) was diluted and extracted with EtOAc (30 mL. times.2). The combined organic layers were washed with brine (20mL) and Na2SO4Dried and concentrated. The residue was purified by silica gel column (PE: EtOAc: 100:1 to 15:1) to give the product F2 as a yellow oil (1g, 88.6% yield).1H NMR(400MHz,CDCl3)δ5.15-5.02(m,1H),3.56(d,J=8.0Hz,1H),2.46-2.39(m,1H),3.34(s,3H),3.29(d,J=8.0Hz,1H),2.43-1.80(m,7H),1.58-1.10(m,19H),0.90(s,3H)。

Step 3. preparation of compound F3. At 0 ℃ under N2To a solution of F2(1g, 2.49mmol) in THF (15mL) was added BH dropwise with protection3-Me2S solution (2.48mL, 10M). The solution was stirred at 25 ℃ for 4 hours. TLC (PE/EtOAc ═ 3/1) showed the reaction was complete. After cooling to 0 ℃, NaOH solution (9.93mL, 3M) was added very slowly, releasing a large amount of gas. After the addition was complete, H was added slowly2O2(4.53mL, 33%) and internal temperatureMaintained below 10 ℃. The resulting solution was stirred at 25 ℃ for 1 hour. The resulting solution was extracted with EtOAc (20 mL. times.3). The combined organic solution was saturated with Na2S2O3Aqueous solution (20 mL. times.3), brine (20mL) and Na2SO4Dried and concentrated in vacuo to give the crude product (1g) as a yellow oil. The crude product was used in the next step without further purification.

Step 4. preparation of compound F4. A mixture of F3(1.0g, 2.38mmol), PCC (0.767g, 3.56mmol) and silica gel (0.843g, w/w ═ 1/1.1) in DCM (15mL) was stirred at 25 ℃ for 2 hours and the reaction mixture turned brown in color. TLC (PE/EtOAc ═ 3/1) showed the reaction was complete. The solution was filtered and the filter cake was washed with DCM (20 mL). The combined filtrates were concentrated in vacuo. The residue was purified by silica gel column eluting with PE: EtOAc 15:1 to 8:1 to give F5(800mg, 80.6%) as a white solid. MS ESI: with respect to C24H41O4Calculated value of [ M + H ]]+417, found 399([ M + H-18)]+)。1H NMR(400MHz,CDCl3)δ3.52(d,J=8.0Hz,1H),3.33(s,3H),3.28(d,J=8.0Hz,1H),2.58-2.52(m,1H),2.20-1.60(m,15H),1.53-1.10(m,11H),0.62(s,3H)。

Step 5. preparation of compound F5. To a solution of F4(0.5g, 1.20mmol) and catalytic amount of concentrated HBr (12.1mg, 40% in water) in MeOH (15mL) at 0 deg.C was added dibromo (230mg, 1.44mmol) dropwise. The reaction mixture was stirred at 25 ℃ for 1 hour. TLC (PE: EtOAc ═ 3:1) showed the reaction was complete. With saturated NaHCO3Quenching the reaction by using the aqueous solution and adjusting the pH value to 7-8. The reaction mixture was extracted with DCM (20 mL. times.2). The combined organic layers were washed with brine (20mL) and Na2SO4Dried and concentrated to give crude product F5(500mg) as a yellow oil.

Example 84. preparation of compounds 161 and 162.

To a solution of compound F5(150mg, 0.302mmol) in acetone (5mL)Adding K to the solution2CO3(62.6mg, 0.453mmol) and 4, 5-difluoro-2H-benzo [ d][1,2,3]Triazole (70.2mg, 0.453 mmol). After stirring at 25 ℃ for 3h, TLC (PE: EA ═ 3:1) showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated in vacuo to give the crude product (150 mg). The crude product was purified by preparative hplc (hcl) to give compound 162(18mg, 10.4%) and compound 161(31mg, 18%) as white solids.

1H NMR (161) (10.4% yield): (400MHz, CDCl)3)δ7.42-7.36(m,1H),7.09-7.06(m,1H),5.53-5.35(m,2H),3.50(t,J=9.2Hz,1H),3.34-3.29(m,4H),2.71(d,J=8.8Hz,1H),2.20-1.00(m,23H),0.74(s,3H)。LCMS tR1.350 min, in 2 min chromatography, 10-80AB, purity 100%, MS ESI: with respect to C29H37F5N3O3Calculated value of [ M + H ]]+570, found 570.

1H NMR (162) (18% yield): (400MHz, CDCl)3)δ7.68-7.64(m,1H),7.37-7.29(m,1H),5.59-5.49(m,2H),3.51(t,J=7.6Hz,1H),3.34-3.29(m,4H),2.69(d,J=8.8Hz,1H),2.24-1.14(m,23H),0.77(s,3H)。LCMS tR1.398 minutes, in 2 minutes chromatography, 10-80AB, 100% purity, MS ESI: with respect to C29H37F5N3O3Calculated value of [ M + H ]]+570, found 570.

Example 85 preparation of compounds 163, 164 and 165.

1H NMR (163) (11% yield): (400MHz, CDCl)3)δ7.73(t,J=5.6Hz,1H),7.09-7.06(m,2H),5.48-5.38(m,2H),3.88(s,3H),3.50(d,J=8.8Hz,1H),3.31(s,3H),3.28(d,J=9.2Hz,1H),2.62(t,J=8.8Hz,1H),2.17-1.11(m,23H),0.74(s,3H)。LCMS tR1.354 min, in 2 min chromatography, 10-80AB, 100% purity, MS ESI: with respect to C30H41F3N3O4Calculated value of [ M + H ]]+564, found 564.

1H NMR (164) (11% yield): (400MHz, CDCl)3)δ7.38(d,J=1.6Hz,1H),7.21(d,J=7.2Hz,1H),7.15(dd,J1=7.2Hz,J2=1.6Hz,1H),5.34(s,2H),3.89(s,3H),3.49(d,J=6.8Hz,1H),3.31(s,3H),3.28(d,J=6.8Hz,1H),2.66(t,J=7.2Hz,1H),2.35-1.10(m,23H),0.71(s,3H)。LCMS tR0.964 min, in 1.5 min chromatography, 5-95AB, 95% purity, MS ESI: with respect to C30H41F3N3O4Calculated value of [ M + H ]]+564, found 564.

1H NMR (165) (19% yield): (400MHz, CDCl)3)δ7.91(d,J=9.2Hz,1H),7.01(dd,J1=9.2Hz,J2=2.0Hz,1H),6.59(d,J=2.0Hz,1H),5.38-5.27(m,2H),3.85(s,3H),3.49(d,J=9.2Hz,1H),3.31(s,3H),3.28(d,J=9.2Hz,1H),2.67(t,J=8.4Hz,1H),2.43(brs,1H),2.30-1.05(m,23H),0.71(s,3H)。LCMS tR1.354 min, in 2 min chromatography, 10-80AB, 100% purity, MS ESI: with respect to C30H41F3N3O4Calculated value of [ M + H ]]+564, found 564.

EXAMPLE 86 preparation of Compound 166.

1H NMR (166) (10.5% yield): (400MHz, CDCl)3)δ7.73(t,J=5.6Hz,1H),7.09-7.06(m,2H),5.48-5.38(m,2H),3.88(s,3H),3.50(d,J=8.8Hz,1H),3.31(s,3H),3.28(d,J=9.2Hz,1H),2.62(t,J=8.8Hz,1H),2.17-1.11(m,22H),0.74(s,3H)。LCMS tR1.354 min, in 2 min chromatography, 10-80AB, 100% purity, MS ESI: with respect to C30H41F3N3O4Calculated value of [ M + H ]]+564, found 564.

Example 87 preparation of compounds 167, 168 and 169.

1H NMR (167) (1% yield): (400MHz, CDCl)3)δ7.89-7.83(m,2H),7.38(t,J=7.2Hz,1H),5.56-5.46(m,2H),3.52(d,J=9.2Hz,1H),3.34(s,1H),3.30(d,J=9.2Hz,1H),2.65(t,J=4.4Hz,1H),2.26-1.07(m,25H),0.76(s,3H)。LCMS tR1.424 min, in 2 min chromatography, 10-80AB, purity 99%, MS ESI: with respect to C29H38ClF3N3O3Calculated value of [ M + H ]]+568, found 568.

1H NMR (168) (6% yield): (400MHz, CDCl)3)δ8.01-7.99(m,1H),7.36-7.34(m,2H),5.43-5.31(m,2H),3.50(d,J=8.8Hz,1H),3.28-3.35(m,4H),2.70(t,J=8.8Hz,1H),2.24-1.13(m,23H),0.73(s,3H)。LCMS tR1.009 min, in 1.5 min chromatography, 5-95AB, 97% pure, MS ESI: with respect to C29H38ClF3N3O3Calculated value of [ M + H ]]+568, found 568.

1H NMR (169) (8% yield): (400MHz, CDCl)3)δ8.07(s,1H),7.45(t,J=7.2Hz,1H),7.29(s,1H),5.34-5.45(m,2H),3.49(d,J=8.8Hz,1H),3.28-3.32(m,4H),2.70(t,J=9.2Hz,1H),2.20-0.88(m,23H),0.71(s,3H)。LCMS tR0.995 min, in 1.5 min chromatography, 5-95AB, 99% purity, MS ESI: with respect to C29H38ClF3N3O3Calculated value of [ M + H ]]+568, found 568.

Example 88 preparation of Compound C19.

Step 1. preparation of compound C12. To C2(4g, 13.14mmol) in 15mL CH at 25 deg.C2Cl2To the solution of (1) was added 1H-imidazole (2.68g, 39.42mmol)And tert-butylchlorodimethylsilane (2.97g, 19.71mmol), and the reaction was stirred at 25 ℃ for 16 hours. Using 50mL CH2Cl2The reaction mixture was filtered and evaporated in vacuo. The residue was purified by silica gel column chromatography (PE: EtOAc ═ 50:1-30:1-20:1-15:1-10:1) to give C12(5g, 90.87% yield) as a white solid.1H NMR:(400MHz,CDCl3)δ3.81(d,J=8.0Hz,1H),3.58(d,J=8.0Hz,1H),2.62-2.55(m,1H),2.48-2.43(m,1H),2.40-2.28(m,3H),2.26-2.07(m,2H),1.87-1.86(m,1H),1.84-1.75(m,4H),1.56-1.27(m,10H),0.87(s,12H),0.042(s,6H)。

Step 2. preparation of compound C13. To a solution of C12(15.79g, 71.64mmol) in 30mL of toluene at 0 deg.C was added AlMe dropwise3Solution (17.91mL, 3 equiv). After 1 hour, (5R,8R,9S,10R,13S,14S) -10- (((tert-butyldimethylsilyl) oxy) methyl) -13-methyldodecahydro-1H-cyclopenta [ a ] is added dropwise at-78 deg.C]A solution of phenanthrene-3, 17(2H,4H) -dione (5g, 11.94mmol) in toluene (40mL) and the reaction mixture was stirred at-78 ℃ for 1 hour, then a solution of MeMgBr (11.94mL, 3 equivalents) was added dropwise to the mixture at-78 ℃ and it was stirred for a further 2 hours at-78 ℃. After TLC (PE: EtOAc ═ 3:1) showed complete consumption of starting material, the reaction mixture was washed with NH4Aqueous Cl (15mL), filtered and washed with 500mL EtOAc. The organic layer was extracted with 300mL EtOAc, washed with brine and concentrated. The residue was purified by silica gel column chromatography (PE: EA ═ 100-50:1-20:1-10:1-4:1) to give C13(5g, 96.3%) as a white solid.1H NMR(400MHz,CDCl3),δ3.76(d,J=8.0Hz,1H),3.40(d,J=8.0Hz,1H),2.44-2.39(m,1H),2.09-1.70(m,6H),1.61-1.18(m,19H),0.89(s,12H),0.04(s,6H)。

Step 3. preparation of compound C14. To a solution of C13(7g, 16.1mmol) in 70mL MeOH was added a solution of HBr (6.5g, 32.2mmol, 40% in water). The reaction mixture was stirred at 25 ℃ for 0.7 h. After TLC (PE: EtOAc ═ 3:1) showed complete consumption of starting material, the reaction mixture was washed with saturated NaHCO3Aqueous solution (200mL) was quenched and extracted with 500mL EtOAc, washed with brine (100mL) and concentrated to give product C14(5.6g, crude) as a white solid.

Step 4. preparation of compound C15. At 0 ℃ to PPh3To a solution of EtBr (51.8g, 140mmol) in THF (40mL) was added a solution of t-BuOK (15.7g, 140mmol) in THF (40 mL). After stirring at 60 ℃ for 1 hour, a solution of compound C14(9g, 28.0mmol) in THF (40mL) was added dropwise at 60 ℃. The reaction mixture was then stirred at the same temperature for 8 hours. TLC (PE/EtOAc ═ 3/1) showed the reaction was complete and the major product was found to be less polar. The reaction mixture was extracted three times with EtOAc (300 mL). The organic layer was washed with brine (100mL) and Na2SO4Dried and concentrated in vacuo to give the crude product. The crude product was purified by silica gel column (PE: EA ═ 5:1) to give compound C15(5.0g, 53.5%) as a light yellow oil.1H NMR(400MHz,CDCl3)δ5.15-5.05(m,1H),3.94(d,J=10.8Hz,1H),3.56(d,J=10.8Hz,1H),2.40-2.12(m,3H),2.01-1.71(m,3H),1.69-1.12(m,24H),0.85(s,3H)。

Step 5. preparation of compound C16. In N2To a solution of C15(500mg, 1.50mmol) in THF (15mL) in a flask was added NaH (171mg, 4.5mmol, 60% in oil) portionwise with protection. The reaction mixture was stirred for 10 minutes. Iodothane (701mg, 4.5mmol) was then added. The reaction mixture was heated and stirred at 50 ℃ for a further 2 hours. TLC (PE: EA ═ 3:1) showed the reaction was complete and found to have a lower polarity of the major product. Reacting with NH4Aqueous Cl (10mL) was quenched and extracted with EtOAc (20 mL. times.2). The combined organic layers were washed with aqueous NaCl (20mL) and Na2SO4Dried and then concentrated. The residue was purified by silica gel column chromatography (PE: EtOAc 15:1 to 8:1) to give the product C16(500mg, 91.9% yield) as a yellow oil.1H NMR(400MHz,CDCl3)δ5.12-5.09(m,1H),3.60(d,J=9.2Hz,1H),3.48-3.42(m,2H),3.23(d,J=9.2Hz,1H),2.38-2.12(m,3H),1.95-1.72(m,3H),1.65-1.10(m,26H),0.85(s,3H)。

Step 6. preparation of compound C17. To a solution of C16(500mg, 1.38mmol) in THF (15mL) at 0 deg.C was added BH dropwise3-Me2S solution (1.38mL, 10M). The solution was stirred at 25 ℃ for 4 hours. TLC (PE: EtOAc ═ 3:1) showed reactionAlmost complete and the major products with higher polarity were found. After cooling to 0 ℃, NaOH solution (5.5mL, 3M) was added very slowly. After the addition was complete, H was added slowly2O2(2.51mL, 33%) and the internal temperature was maintained below 10 ℃. The resulting solution was stirred at 25 ℃ for 2 hours. The resulting solution was extracted with EtOAc (20 mL. times.3). The combined organic solution was saturated with Na2S2O3Aqueous solution (30 mL. times.3), brine (30mL), washed with Na2SO4Dried and concentrated in vacuo to give the crude product as a yellow oil (500 mg). The crude product was used in the next step without further purification.

Step 7. preparation of compound C18. A suspension of C17(500mg, 1.32mmol), PCC (426mg, 1.98mmol) and silica gel (469mg, w/w ═ 1/1.1) in DCM (15mL) was stirred at 30 ℃ for 2 h. The reaction mixture turned brown in color. TLC (PE/EtOAc ═ 3/1) showed the reaction was complete and the major product was found to be less polar. The solution was filtered and the filter cake was washed with DCM (20 mL). The combined filtrates were concentrated in vacuo. The residue was purified by silica gel column chromatography eluting with PE/EtOAc 15/1 to 5/1 to give C18(400mg, 80.3%) as a white solid. MS ESI: with respect to C24H40O3Calculated value of [ M + H ]]+377, found 359([ M + H-18)]+)。1H NMR(400MHz,CDCl3)δ3.58(d,J=9.2Hz,1H),3.49-3.42(m,2H),3.24(d,J=9.2Hz,1H),2.56-2.51(m,1H),2.18-1.65(m,12H),1.60-1.10(m,19H),0.61(s,3H)。

Step 8 preparation of compound C19. To a solution of C18(400mg, 1.06mmol) and catalytic amount of concentrated HBr (10.7mg, 40% in water) in MeOH (15mL) at 0 deg.C was added dibromo (254mg, 1.59mmol) dropwise. The reaction mixture was stirred at 25 ℃ for 1 hour. TLC (PE: EtOAc ═ 3:1) showed the reaction was complete and the major product was found to be less polar. By saturated NaHCO3The aqueous solution quenches the reaction and the pH is adjusted to 7-8. The reaction mixture was extracted with DCM (20 mL. times.2). The combined organic layers were washed with brine (20mL) and Na2SO4Drying and concentration gave the crude product C19 as a yellow oil (400mg, 82.8% yield).

EXAMPLE 89 preparation of Compound 170.

To a solution of compound F5(150mg, 0.329mmol) in acetone (5mL) was added K2CO3(68.1mg, 0.493mmol) and 1H-pyrazole-4-carbonitrile (45.8mg, 0.493 mmol). After stirring at 25 ℃ for 3h, LCMS showed the reaction was complete. The reaction mixture was filtered, and the filtrate was concentrated in vacuo to give the crude product (150 mg). The crude product was purified by preparative hplc (hcl) to give the desired product 170(13mg, 8.41%) as a white solid.

1H NMR (170) (8.4% yield): (400MHz, CDCl)3)δ7.86(s,1H),7.81(s,1H),5.03-4.87(m,2H),3.54(d,J=9.2Hz,1H),3.68-3.41(m,2H),3.24(d,J=9.2Hz,1H),2.59(t,J=9.2Hz,1H),2.21-1.15(m,29H),0.65(s,3H)。LCMS tR0.949 min, in 1.5 min chromatography, 5-95AB, 98.6% purity, MS ESI: with respect to C28H42N3O3Calculated value of [ M + H ]]+467 found 450[ M + H-18]+

Example 90 alternative preparation of compound a 21.

Step 1. preparation of compound E16. To acetic acid ((5S,8R,9S,10R,13S,14S) -13-methyl-3, 17-dioxohexadecahydro-1H-cyclopenta [ a ]]To a solution of phenanthren-10-yl) methyl ester (E4, 5g, 14.4mmol) in THF (50mL) was added MeMgBr (15mL, 3M in ether, 450mmol) dropwise to control the internal temperature below-70 ℃. The mixture was then stirred at-78 ℃ for 1 hour. TLC showed the reaction was complete. To the mixture was added a solution of NH4Cl (6g) in water (30mL) and the internal temperature was raised to-20 ℃. The mixture was then warmed to 20 ℃. The organic layer was separated. The aqueous phase was extracted with EtOAc (50 mL). The combined organic layers were passed over Na2SO4DryingConcentrated in vacuo and purified by column chromatography (PE: EtOAc ═ 6:1 to 3:1) to give acetic acid ((3R,5S,8R,9S,10R,13S,14S) -3-hydroxy-3, 13-dimethyl-17-oxohexadecahydro-1H-cyclopenta [ a ] as a white solid]Phenanthren-10-yl) methyl ester (E16, 2.4g, 46%) and acetic acid ((3S,5S,8R,9S,10R,13S,14S) -3-hydroxy-3, 13-dimethyl-17-oxohexadecahydro-1H-cyclopenta [ a]Phenanthren-10-yl) methyl ester (1g, 19%).1H NMR(400MHz,CDCl3)δ4.29(d,J=12.1Hz,1H),4.13(d,J=12.1Hz,1H),2.48-2.35(m,1H),2.11-1.85(m,7H),1.85-1.59(m,6H),1.55-1.22(m,11H),1.09-0.74(m,7H)。

Step 2. preparation of compound E17. To a suspension of PPh3EtBr (4.61g, 12.4mmol) in THF (10mL) at 20 deg.C was added a solution of t-BuOK (1.86g, 16.6mmol) in THF (20 mL). The color of the suspension turned deep red. After stirring at 60 ℃ for 1 hour, acetic acid ((3R,5S,8R,9S,10R,13S,14S) -3-hydroxy-3, 13-dimethyl-17-oxohexadecahydro-1H-cyclopenta [ a ] was added dropwise at 60 ℃]A solution of phenanthren-10-yl) methyl ester (E16, 1.5g, 4.14mmol) in THF (20 mL). The reaction mixture was then stirred at 60 ℃ for 16 hours. TLC showed the reaction was complete. Adding NH to the reaction mixture4Cl (50mL, saturated aqueous solution). The color of the mixture turned light yellow. The organic layer was separated. The aqueous phase was extracted with EtOAc (50 mL). The combined organic layers were concentrated in vacuo and purified by silica gel column chromatography (PE: EtOAc: 10:1 to 4:1) to give (3R,5S,8S,9S,10R,13S,14S) -17-ethylene-10- (hydroxymethyl) -3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] as a white solid]Phenanthren-3-ol (E17, 1.0g, 72.6%).1H NMR(400MHz,CDCl3)δ5.19-5.08(m,1H),3.93(d,J=11.5Hz,1H),3.74(d,J=11.5Hz,1H),2.44-2.33(m,1H),2.32-2.13(m,2H),2.12-2.04(m,1H),1.87-1.71(m,2H),1.69-1.43(m,12H),1.38-1.08(m,11H),1.07-0.74(m,6H)

Step 3. preparation of compound a 18. To a solution of E17(0.8g, 2.4mmol) in THF (10mL) was added sodium hydride (475mg, 11.9mmol) and iodoethane (1.85g, 11.9mmol) in portions. The mixture was stirred at 50 ℃ for 12 hours. The reaction mixture was quenched with water and extracted with EtOAc (10 mL. times.2). The combined organic layers were passed over anhydrous Na2SO4Dried, filtered and concentrated. By silica gelThe residue was purified by column chromatography (PE: EA ═ 50:1) to give a18(0.5g, 57.5%) as a colorless oil.1H NMR(400MHz,CDCl3)δ5.13-5.08(m,1H),3.52(d,J=9.6Hz,1H),3.43-3.38(m,3H),2.38-2.32(m,1H),2.25-2.12(m,2H),2.06-2.01(m,1H),1.74-1.57(m,2H),1.56-1.39(m,6H),1.31-1.27(m,2H),1.24-1.16(m,11H),1.14-1.07(m,2H),1.05-0.91(m,2H),0.88(s,3H),0.87-0.74(m,3H)。

Step 4. preparation of compound a 19. To a solution of A18(0.5g, 1.38mmol) in THF (5mL) at 0 deg.C was added BH dropwise3-Me2S (0.69mL, 6.9 mmol). The solution was stirred at 30 ℃ for 2 hours. TLC (PE/EtOAc ═ 5/1) showed the reaction was complete. After cooling to 0 ℃, aqueous NaOH (5.51g, 10% in water) was added very slowly. After the addition was complete, H was added slowly2O2(1.56g, 30%) and the internal temperature was maintained below 10 ℃. The resulting solution was stirred at room temperature for 1 hour. A white solid formed. To the mixture was added EtOAc (5mL) and filtered. The filter cake was washed with EtOAc (5 mL). The combined organic layers were separated and washed with Na2S2O3(5mL, 20%, aq.) wash over Na2SO4Dried and concentrated in vacuo to give A19(0.4g, purity: 78%, yield: 59.7%) as a colorless oil, which was used without further purification. LCMS tR 1.085 min, in 2 min chromatography, 30-90AB, purity 77.6%, MS ESI: with respect to C24H42O3Calculated value of [ M + H ]]+379, found 361([ M + H-18)]+)。

Step 5. preparation of Compound A20. To a solution of A19(0.4g, 0.824mmol, purity: 78%) in dichloromethane (5mL) was added silica gel (1g) and PCC (0.885g, 4.11 mmol). The suspension was stirred at 30 ℃ for 16 hours. TLC (PE: EA ═ 5:1) showed complete consumption of the reaction. The reaction mixture was filtered, and the filtrate was concentrated. The residue was purified by silica gel column chromatography (PE: EA ═ 10:1) to give a20(0.2g, 64.4%) as a pale yellow oil.1H NMR(400MHz,CDCl3)δ3.50(d,J=10.0Hz,1H),3.42-3.37(m,3H),2.53(t,J=8.8Hz,1H),2.20-2.15(m,1H),2.11(s,3H),2.07-1.97(m,2H),1.73-1.64(m,4H),1.50-1.47(m2H),1.37-1.25(m,6H),1.21-1.14(m,9H),1.12-0.75(m,5H),0.61(s, 3H). LCMS tR ═ 1.124 min, in 2 min chromatography, 30-90AB, purity 100%, MS ESI: with respect to C24H40O3Calculated value of [ M + H ]]+377, found 359([ M + H-18)]+)。

Step 6. preparation of compound a 21. To a solution of A20(0.2g, 0.531mmol) in methanol (2mL) was added HBr (8.93mg, 0.053mmol, 48% in water) and Br2(127mg, 0.796 mmol). The mixture was stirred at 30 ℃ for 2 hours. With NaHCO3The aqueous solution quenched the reaction mixture to adjust the pH to about 8. The mixture was poured into water (10mL) and extracted with EtOAc (10 mL. times.2). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated to give a21(0.2g, 82.6%) as a light yellow solid, which was used without further purification. LCMS tR 1.184 min, in 2 min chromatography, 30-90AB, 100% purity, MS ESI: with respect to C24H39BrO3Calculated value of [ M + H ]]+455, found 437([ M + H-18)]+)。

EXAMPLE 91 preparation of Compound 171.

To a solution of A21(90mg, 0.197mmol) in acetone (2mL) was added potassium carbonate (67.9mg, 0.492mmol) and 1H-pyrazole-4-carbonitrile (27.4mg, 0.295 mmol). The suspension was stirred at 40 ℃ for 12 hours. The reaction mixture was cooled and filtered, and the filtrate was concentrated. The residue was purified by preparative HPLC to give 171(22mg, 23.8%) as a white solid.1H NMR(400MHz,CDCl3) δ 7.85(s,1H),7.81(s,1H),5.02(d, J ═ 18.0Hz,1H),4.89(m, J ═ 18.0Hz,1H),3.51(d, J ═ 9.2Hz,1H),3.42-3.37(m,3H),2.60(t, J ═ 9.2Hz,1H),2.25-2.17(m,1H),2.06-1.99(m,2H),1.75-1.69(m,4H),1.54-1.50(m,3H),1.46-0.81(m,19H),0.67(s, 3H). LCMS tR 1.109 min, in 2 min chromatography, 30-90AB, 100% purity, MS ESI: with respect to C28H41N3O3Calculated value of [ M + H ]]+46Found 490([ M + Na)]+)。

Example 92 preparation of compounds 172 and 173.

To a solution of 2-bromo-1- ((3R,5S,8S,9S,10R,13S,14S,17S) -10- (ethoxymethyl) -3-hydroxy-3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) ethanone (90mg, 0.197mmol) in acetone (2mL) were added potassium carbonate (67.9mg, 0.492mmol) and 4, 5-difluoro-2H-benzo [ d ] [1,2,3] triazole (45.7mg, 0.295 mmol). The mixture was stirred at 50 ℃ for 16 hours. The mixture was cooled, filtered and concentrated. The residue was purified by preparative HPLC to give 2- (4, 5-difluoro-2H-benzo [ d ] [1,2,3] triazol-2-yl) -1- ((3R,5S,8S,9S,10R,13S,14S,17S) -10- (ethoxymethyl) -3-hydroxy-3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) ethanone (172, 10mg, 9.28%, purity: 97%) as a light yellow solid and 2- (4, 5-difluoro-1H-benzo [ d ] [1,2,3] triazol-1-yl) -1- ((3R,5S,8S,9S,10R,13S as a light yellow solid, 14S,17S) -10- (ethoxymethyl) -3-hydroxy-3, 13-dimethylhexadecahydro-1H-cyclopenta [ a ] phenanthren-17-yl) ethanone (173, 15mg, 14.0%, purity: 98%).

1H NMR(172):(400MHz,CDCl3)δ7.63(dd,J=3.2Hz,8.8Hz,1H),7.31-7.24(m,2H),5.57-5.46(m,2H),3.53(d,J=9.6Hz,1H),3.43-3.40(m,3H),2.66(t,J=8.8Hz,1H),2.27-2.03(m,3H),1.79-1.69(m,4H),1.65-1.57(m,4H),1.32-1.26(m,6H),1.22-1.12(m,8H),1.09-0.82(m,4H),0.76(s,3H)。LCMS tR1.066 min, in 1.5 min chromatography, 5-95AB, 97% purity, MS ESI: with respect to C30H42F2N3O3Calculated value of [ M + H ]]+530, found 512([ M + H-18)]+)。

1H NMR(173):(400MHz,CDCl3)δ7.40-7.33(m,1H),7.05(d,J=7.2Hz,1H),5.46-5.35(m,2H),3.52(d,J=10.0Hz,1H),3.43-3.38(m,3H),2.71(t,J=8.4Hz,1H),2.24-2.03(m,3H),1.75-1.69(m,4H),1.62-1.53(m,4H),1.32-1.16(m,14H),1.13-0.83(m,4H),0.72(s,3H)。LCMS tR1.037 min, in 1.5 min chromatography, 5-95AB, 98% purity, MS ESI: with respect to C30H42F2N3O3Calculated value of [ M + H ]]+530, found 530([ M + H)]+)。

Measurement method

The compounds provided herein can be evaluated using various assays; examples of which are described below.

TBPS-bound steroid inhibition

TBPS binding assays using rat brain cortical membranes in the presence of 5. mu.M GABA have been described (Gee et al, J.Pharmacol.Exp.Ther.1987,241, 346-353; Hawkinson et al, mol.Pharmacol.1994,46, 977-985; Lewis, A.H et al, mol.Pharmacol.1989,35, 189-194).

Briefly, the cortex was removed rapidly after decapitation in Sprague-Dawley rats (200- & 250g) anesthetized with carbon dioxide. The cortex was homogenized in 10 volumes of ice-cold 0.32M sucrose using a glass/teflon homogenizer and centrifuged at 1500 × g for 10 minutes at 4 ℃. The resulting supernatant was centrifuged at 10,000 Xg for 20 minutes at 4 ℃ to obtain a P2 pellet. The P2 pellet was resuspended in 200mM NaCl/50mM Na-K phosphate pH 7.4 buffer and centrifuged at 10,000 Xg for 10 minutes at 4 ℃. This washing procedure was repeated twice and the pellet was resuspended in 10 volumes of buffer. With 3nM [ mu ] M GABA35S]Aliquots (100 μ L) of membrane suspension were incubated with 5 μ L aliquots (final 0.5%) of TBPS and test drug dissolved in dimethyl sulfoxide (DMSO). The incubation was brought to a final volume of 1.0mL using buffer. Nonspecific binding was determined in the presence of 2 μ M unlabeled TBPS and ranged from 15% to 25%. After 90 minutes incubation at room temperature, the assay was terminated by filtration through a glass fiber filter (Schleicher and Schuell No.32) using a cell harvester (Brandel) and washed three times with ice-cold buffer. Filter bound radioactivity was measured by liquid scintillation spectrometry. Nonlinear curve fitting of the total data for each drug averaged for each concentration was performed using prism (graphpad). If the sum of squares according to the F-test is significantly lower, the data is fitted to a partial rather than complete inhibition model. Similarly, if the sum of squares according to the F-test is significantly lower, the data is fitted to a two-component rather than a one-component inhibition model. Test Compound concentration (IC) that produces 50% inhibition of specific binding was determined for a separate experiment50) And maximum degree of inhibition (I)max) Where the same model was used for the total data and then the mean ± SEM of individual experiments was calculated. Tetrandrine served as a positive control for these studies as it has been demonstrated to potently inhibit TBPS binding.

Each compound is screened or can be screened to determine its activity as in vitro35S]Potential of TBPS binding modulators. These assays are according to or can be performed according to the procedures discussed above.

With respect to Table 1, "A" indicates IC50<10nM, "B" indicates IC5010nM to 50nM, "C" indicates IC50>50nM to 100nM, "D" indicates IC50>100nM to 500nM, and "E" indicates IC50>500nM。

Table 1.

Recombinant alpha1β2γ2And alpha4β3δGABAAPatch-clamp electrophysiology of receptors

Measurement of our GABA Using cellular electrophysiologyAPharmacological properties of receptor modulators in heterologous cell systems. Each compound was tested for sub-maximal agonist dose (GABA EC)202 μ M) affect the GABA-mediated current capacity. Alpha to GABA receptor via Lipofecatamine method1β2γ2Stable transfection of LTK cells with subunits and use of alpha4The β 3 δ subunit was transiently transfected into CHO cells. Cells were passaged at about 50-80% confluence and then seeded onto 35mm sterile culture plates containing 2ml of complete medium without antibiotics or antimycotics. The confluent clusters of cells are electrically coupled (Pritchett et al, Science,1988,242, 1306-. Because the response in remote cells is insufficiently clamped in voltage and because of uncertainty about the degree of coupling (Verdoorn et al, Neuron 1990,4,919-928), cells are cultured at a density that enables the recording of single cells (without visible connections to other cells).

Whole-cell currents were measured using the PatchMaster software with HEKA EPC-10 amplifiers or by using a high-throughput QPatch platform (Sophion). The bath solutions for all experiments contained (in mM): NaCl 137mM, KCl 4mM, CaCl2 1.8mM、MgCl21mM, HEPES 10mM, D-glucose 10mM, pH (NaOH) 7.4. In some cases, 0.005% cremophor was also added. Intracellular (pipette) solutions contain: KCl 130mM, MgCl21mM, Mg-ATP 5mM, HEPES 10mM, EGTA 5mM, pH 7.2. During the experiment, the cells and solution were maintained at room temperature (19 ℃ -30 ℃). For manual patch clamp recording, the cell culture dish was placed on the dish rack of the microscope and the bath solution was continuously perfused (1 ml/min). Giga-ohm sealing was formed between the patch electrodes and the cells (electrode resistance range: 2.5 M.OMEGA. -6.0 M.OMEGA.; seal resistance range:>1G Ω) the cell membrane across the electrode tip is ruptured to ensure electrical access to the cell interior (whole cell patch configuration). For experiments using the QPatch system, cells were transferred as a suspension to the QPatch system in bath solution and automated whole cell recordings were performed.

The cell voltage was clamped at a holding potential of-80 mV. For the assay of test items, GABA receptors were stimulated by 2 μ M GABA after sequential preincubation of increasing concentrations of test items. The duration of the pre-incubation was 30 seconds and the duration of GABA stimulation was 2 seconds. The test article was dissolved in DMSO to form a stock solution (10 mM). The test articles were diluted to 0.01, 0.1, 1 and 10 μ M in the bath solution. All concentrations of test article were tested on each cell. Relative hundredThe fractional enhancement is defined as responding to GABA EC in the presence of test substance20Divided by the peak amplitude in response to only GABA EC20The peak amplitude of (d), multiplied by 100.

TABLE 2 exemplary Compounds in GABAAElectrophysiological assessment at R.

With respect to Table 2. GABAA receptors α 1 β 2 γ 2 and α 4 β 3 δ% efficacy: "A" 10-100, "B" >100-500, "C" > 500; d indicates data is unavailable or undetermined.

GABA receptor potentiation

Two-electrode voltage clamp (TEVC) technique was used to study the concentration of 10. mu.M of compound on α expressed in oocytes from Xenopus laevis1β2γ2Or alpha4β3GABA composed of delta subunitsAThe influence of the receptor. GABA-induced currents were recorded from GABA receptor-expressing oocytes and the effect of the test item on the modulation of these currents was investigated.

Ovaries were collected from female Xenopus laevis that had been deeply anesthetized by cooling at 4 ℃ and immersion in tricaine mesylate (MS-222, concentration 150mg/L) in sodium bicarbonate (300 mg/L). Once anesthetized, animals were decapitated and spinal cord destroyed following the rules of the geneva animal rights. A small piece of the ovary was isolated for immediate preparation, while the rest was placed in a sterile Barth solution containing (in mM) NaCl 88, KCl 1, NaHCO at 4 deg.C3 2.4、HEPES 10、MgSO4.7H2O 0.82、Ca(NO3)2.4H2O 0.33、CaCl2.6H2O0.41 (pH 7.4) and supplemented with 20. mu.g/ml kanamycin, 100 units/ml penicillin and 100. mu.g/ml streptomycin. All recordings were performed at 18 ℃ and cells were incubated with a solution containing (in mM) NaCl 82.5, KCl 2.5, HEPES 5, CaCl2.2H2O、CaCl2.6H2Culture medium of O1 (pH 7.4) was superfused.

Plasmids containing the RNA of the human GABRA1/GABRB2/GABRG2 and GABRA4/GABRB3/GABRD genes were injected into oocytes using a proprietary automatic injection device (Hogg et al, J. Neurosci. methods, (2008)169: 65-75). These genes encode respectively for alpha1β2γ2And alpha4β3δGABAAA subunit. Receptor expression was assessed at least two days later using electrophysiology. With respect to alpha1β2γ2The ratio of RNA injections of (1: 1:1) and with respect to alpha4β3Delta is 5:1: 5. Electrophysiology recordings were performed using an automated process equipped with standard TEVC and data was captured and analyzed using proprietary data acquisition and analysis software running under Matlab (Mathworks Inc.). The membrane potential of the oocytes was maintained at-80 mV throughout the experiment. To explore the effect of the proprietary compounds, 10 μ M (. alpha.) was administered1β2γ2) Or 3. mu.M (. alpha.)4β3δ) GABA continued for 30 seconds to induce current. These concentrations approximate the EC of GABA at each receptor subtype50And (4) concentration. The oocytes were then re-exposed to GABA for 30 seconds again. 15 seconds after the start of GABA administration, the test article was co-administered at a concentration of 10. mu.M for 15 seconds. The enhancement of the peak current is evaluated. Data was filtered at 10Hz, captured at 100Hz and analyzed using proprietary data acquisition and analysis software running under Matlab (Mathworks Inc.). For statistical analysis, values were calculated using excel (microsoft) or Matlab (mathworks Inc.). To obtain mean measurements with standard deviation, all experiments were performed using at least three cells.

GABA was made as a concentrated stock solution in water (10)-1M) and then diluted in a recording medium to obtain the desired test concentration. Compounds were prepared as stock solutions in DMSO (10)-2M) and then diluted in a recording medium to obtain the desired test concentration. Residual DMSO did not exceed 1% concentration, which has been shown to have no effect on xenopus oocyte function.

TABLE 3 exemplary Compounds in GABAAElectrophysiological assessment at R.

With respect to Table 3. GABAA receptors α 1 β 2 γ 2 and α 4 β 3 δ% efficacy: a is 50-500, B is more than 500-1000, C is more than 1000-1500 and D is more than 1500-2000; "E" > 2000.

The invention comprises the following contents:

1. a compound of formula (I):

a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3) -, -O-, -S-or-NR3-;

R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl;

R2is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group;

each R3Independently is hydrogen or C1-C6An alkyl group;

R5is absent or hydrogen; and is

Represents a single or double bond, whichIn

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

2. A compound of formula (Ia):

a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3) -, -O-, -S-or-NR3-;

R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl;

each R3Independently is hydrogen or C1-C6An alkyl group;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

3. The compound of embodiment 2, wherein the compound is of formula (Ia-1):

4. the compound of embodiment 2, wherein the compound is of formula (Ia-2):

5. the compound of embodiment 2, wherein a is monocyclic.

6. A compound according to embodiment 5, wherein a is attached through the nitrogen.

7. A compound according to embodiment 5, wherein a is heteroaryl.

8. The compound of embodiment 7, wherein the heteroaryl comprises up to five nitrogen atoms.

9.A compound of embodiment 7 wherein the heteroaryl is benzotriazole, azabenzotriazole, diazabenzotriazole, benzopyrazole, azabenzopyrazole, or diazabenzpyrazole.

10. The compound of embodiment 5, wherein a is a5 membered heteroaryl or heterocyclyl.

11. The compound of embodiment 10, wherein a is a 5-membered heteroaryl or heterocyclyl containing up to four nitrogen atoms.

12. The compound of embodiment 11, wherein a is a5 membered heteroaryl or heterocyclyl comprising 2,3 or 4 nitrogen atoms.

13. A compound according to embodiment 12, wherein a is pyrazole, triazole or tetrazole.

14. The compound of embodiment 3, wherein the compound is of formula (Ia-3):

wherein

R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and is

n is 0, 1,2 or 3.

15. The compound of embodiment 3, wherein the compound is of formula (Ia-4):

wherein

R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and is

n is 0, 1,2 or 3.

16. A compound of embodiment 3 wherein n is 0.

17. The compound of embodiment 3 wherein n is 1 or 2, and R4Is C1-C6Alkyl OR C (O) ORa

18. The compound of embodiment 17, wherein R4Is methyl.

19. The compound of embodiment 17, wherein RaIs C1-C6An alkyl group.

20. The compound of embodiment 19 wherein RaIs ethyl.

21. A compound of embodiment 3 wherein R1Is hydrogen or C1-C6Alkyl, and R4is-C (O) ORa

22. The compound of embodiment 21, wherein R1Is C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl.

23. The compound of embodiment 22 wherein R1Is C1-C6An alkyl group.

24. The compound of embodiment 23 wherein R1Is methyl, ethyl or isopropyl.

25. Trueness ofThe compound of embodiment 21 wherein R1Is methyl and R4is-C (O) OEt.

26. The compound of embodiment 14, wherein the compound is selected from:

27. a compound of formula (Ib):

a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3) -, -O-, -S-or-NR3-;

R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl;

R2is C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group;

each R3Independently is hydrogen or C1-C6An alkyl group;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

28. The compound of embodiment 27, wherein the compound is of formula (Ib-1):

29. the compound of embodiment 27, wherein the compound is of formula (Ib-2):

30. the compound of embodiment 28, wherein the compound has formula (Ib-3)

Wherein

R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcWith nitrogen bound theretoThe atoms together form a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom); and is

n is 0, 1,2 or 3.

31. The compound of embodiment 29, wherein the compound has formula (Ib-4)

Wherein

R4Is cyano, nitro, hydroxy, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Ra、-C(O)N(Rb)(Rc)、-C(O)ORa、-N(Rb)(Rc)、-OC(O)N(Rb)(Rc)、-OC(O)ORa、-OC(O)Ra、-S(O)0-2Ra、-S(O)0-2ORaor-S (O)0-2N(Rb)(Rc);

Each RaIs hydrogen or C1-C6An alkyl group;

each RbAnd RcIndependently of each other is hydrogen, C1-C6Alkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl, or

RbAnd RcForm a ring (e.g., a 3-7 membered ring, e.g., a 5-7 membered ring; a ring containing at least one heteroatom such as nitrogen, oxygen, or sulfur atom) together with the nitrogen atom to which it is bound; and is

n is 0, 1,2 or 3.

32. A compound of embodiment 27 wherein a is monocyclic.

33. A compound of embodiment 27 wherein a is bicyclic.

34. A compound of embodiment 27 wherein a is attached through nitrogen.

35. The compound of embodiment 34, wherein a is 5-or 6-membered heteroaryl or heterocyclyl.

36. The compound of embodiment 35, wherein a is a 5-or 6-membered heteroaryl or heterocyclyl containing up to four nitrogen atoms.

37. The compound of embodiment 36, wherein a is a 5-or 6-membered heteroaryl or heterocyclyl comprising 1,2,3, or 4 nitrogen atoms.

38. A compound of embodiment 27 wherein a is heterocyclyl.

39. A compound of embodiment 38, wherein a is morpholine or piperazine.

40. A compound of embodiment 27 wherein a is heteroaryl.

41. The compound of embodiment 40, wherein the heteroaryl comprises up to five nitrogen atoms.

42. The compound of embodiment 40, wherein the heteroaryl is benzotriazole, azabenzotriazole, diazabenzotriazole, benzopyrazole, azabenzopyrazole, or diazabenzpyrazole.

43. The compound of embodiment 40, wherein the heteroaryl is 5-membered.

44. The compound of embodiment 43 wherein A comprises up to four nitrogen atoms.

45. A compound of embodiment 43 wherein a comprises 2,3 or 4 nitrogen atoms.

46. A compound of embodiment 40 wherein a is pyrazole, triazole or tetrazole.

47. The compound of embodiment 27, wherein R1Is C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl.

48. The compound of embodiment 47, wherein R1Is C1-C6An alkyl group.

49. The compound of embodiment 48, wherein R1Is methyl, ethyl or isopropyl.

50. The compound of embodiment 48, wherein R1Is methyl.

51. A compound of embodiment 27 which isIn R2Is methyl.

52. The compound of embodiment 27, wherein n is 0.

53. The compound of embodiment 27, wherein n is 1 or 2, and R4Is cyano, halo, C1-C6Alkyl radical, C1-C6Alkoxy, -C (O) Raor-S (O)0-2Ra

54. The compound of embodiment 53 wherein R4Is Br, Cl or F.

55. The compound of embodiment 53 wherein R4is-OCH3

56. The compound of embodiment 53 wherein R4Is cyano.

57. The compound of embodiment 53 wherein R4Is C1-C6An alkyl group.

58. The compound of embodiment 57 wherein R4Is methyl.

59. The compound of embodiment 53 wherein R4is-C (O) Ra

60. The compound of embodiment 59 wherein RaIs C1-C6An alkyl group.

61. The compound of embodiment 60, wherein RaIs methyl.

62. The compound of embodiment 53 wherein R4is-S (O)2Ra

63. The compound of embodiment 62, wherein RaIs methyl.

64. The compound of embodiment 27, wherein R1Is C1-C6Alkyl and R4is-C (O) Ra

65. The compound of embodiment 64 wherein R1Is methyl and R4is-C (O) Me.

66. The compound of embodiment 27, wherein n is 0 or 1; r1Is hydrogen or C1-C6An alkyl group; and R is2Is methyl.

67. The compound of embodiment 27, wherein R1Is methyl, ethyl or isopropyl.

68. The compound of embodiment 67 wherein R1Is methyl.

69. The compound of embodiment 27, wherein R4Is C1-C6Alkyl, -C (O) Raor-S (O)0-2Ra

70. The compound of embodiment 69, wherein R4Is methyl.

71. The compound of embodiment 69, wherein R4is-C (O) Me.

72. The compound of embodiment 69, wherein R4is-S (O)2Me。

73. The compound of embodiment 27, wherein said compound is selected from the group consisting of:

74. a pharmaceutical composition comprising a compound according to any one of the preceding embodiments and a pharmaceutically acceptable excipient.

75. A method of inducing sedation and/or anesthesia in a subject comprising administering to the subject an effective amount of a compound of formula (I):

a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3) -, -O-, -S-or-NR3-;

R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl;

R2is hydrogen, C1-C6Alkyl radical (example)E.g. C1-C6Haloalkyl) or C1-C6An alkoxy group;

each R3Independently is hydrogen or C1-C6An alkyl group;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

76. A method of administering to a subject in need thereof an effective amount of a compound, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of a compound of embodiment 1, wherein the subject experiences sedation and/or anesthesia within two hours of administration.

77. The method of embodiment 76, wherein the subject is undergoing sedation and/or anesthesia within one hour of administration.

78. The method of embodiment 76, wherein the subject is undergoing sedation and/or anesthesia immediately.

79. The method of embodiment 76, wherein said compound is administered by intravenous administration.

80. The method of embodiment 76, wherein the compound is administered chronically.

81. The method of embodiment 76, wherein the subject is a mammal.

82. The method of embodiment 81, wherein the subject is a human.

83. The method of embodiment 76, wherein the compound is administered in combination with another therapeutic agent.

84. A method for treating tics in a subject, comprising administering to the subject an effective amount of a compound of formula (I):

a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3) -, -O-, -S-or-NR3-;

R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl;

R2is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group;

each R3Independently is hydrogen or C1-C6An alkyl group;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

85. A method for treating epilepsy or status epilepticus in a subject, the method comprising administering to the subject an effective amount of a compound of formula (I):

a pharmaceutically acceptable salt thereof,

wherein:

a is an optionally substituted nitrogen-containing heteroaryl or heterocyclyl group;

l is-C (R)3)(R3) -, -O-, -S-or-NR3-;

R1Is hydrogen or C1-C6Alkyl radical, C1-C6Alkenyl radical, C1-C6Alkynyl, carbocyclyl or heterocyclyl;

R2is hydrogen, C1-C6Alkyl (e.g. C)1-C6Haloalkyl) or C1-C6An alkoxy group;

each R3Independently is hydrogen or C1-C6An alkyl group;

R5is absent or hydrogen; and is

Represents a single or double bond, in which

When in useWhen one of them is a double bond, the other isIs a single bond; and is

When saidWhen one of them is a double bond, R5Is absent.

86. A method for treating a disorder associated with GABA function in a subject in need thereof, said method comprising administering to said subject a therapeutically effective amount of a compound, a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of one of the compounds of embodiment 1.

87. A method for treating a CNS-related disorder in a subject in need thereof, comprising administering to the subject an effective amount of a compound of embodiment 1, or a pharmaceutically acceptable salt thereof.

88. The method of embodiment 87, wherein the CNS-related disorder is a sleep disorder, a mood disorder, a schizophrenia spectrum disorder, a spasticity, a memory and/or cognitive disorder, a movement disorder, a personality disorder, an autism spectrum disorder, pain, traumatic brain injury, a vascular disease, a substance abuse disorder and/or withdrawal syndrome, or tinnitus.

89. The method of embodiment 87, wherein the subject is a subject with Rett syndrome, fragile X syndrome, or angelman syndrome.

90. A kit comprising a solid composition comprising a compound of formula (I) as described in embodiment 1 and a sterile diluent.

158页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于过重女性患者的基于屈螺酮的避孕药

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!