Fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2H-pyrido [1,2-a ] pyrazine-7-carboxamides as HIV integrase inhibitors

文档序号:1894594 发布日期:2021-11-26 浏览:24次 中文

阅读说明:本技术 作为HIV整合酶抑制剂的稠合的9-羟基-1,8-二氧代-1,3,4,8-四氢-2H-吡啶并[1,2-a]吡嗪-7-甲酰胺 (Fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2H-pyrido [1,2-a ] pyrazine-7-carboxamides as HIV integrase inhibitors ) 是由 亚历山大·瓦西里耶维奇·伊瓦切恩科 尼古拉·菲利波维奇·萨夫丘克 安德烈·亚历山德罗维奇·伊瓦先 于 2020-03-05 设计创作,主要内容包括:本发明涉及新颖的化合物,其具有抗病毒活性,且更具体地,针对人免疫缺陷病毒(HIV)的整合酶的抑制活性。本发明的主题是新颖的通式1或2的稠合的9-羟基-1,8-二氧代-1,3,4,8-四氢-2Н-吡啶并[1,2-а]吡嗪-7-甲酰胺、及其立体异构体、药学上可接受的盐、溶剂合物和晶形或多晶形(I)和(II),其中:环А~(1)是任选地甲基取代的5-7元饱和杂环或二环杂环,环А~(2)是5-6元任选地甲基取代的饱和的或部分饱和的单环杂环,环А~(3)是5-6元单环饱和环烷烃和四氢-2H-吡喃,R是任选地被1、2或3个任选地相同的取代基取代的5-7元单环或二环杂环残基,所述杂环残基包含1-4个选自O、S和N的杂原子,但不包括(2S,5R,13aS)-8-羟基-7,9-二氧代-N-{[3-(三氟甲基)-吡啶-2-基]甲基}-2,3,4,5,7,9,13,13a-八氢-2,5-亚甲基吡啶并[1',2':4,5]吡嗪并[2,1-b][1,3]氧氮杂环庚三烯-10-甲酰胺(式A4)和(1R,4S,12aR)-N-[(3,5-二氟吡啶-2-基)甲基]-7-羟基-6,8-二氧代-1,2,3,4,6,8,12,12a-八氢-1,4-亚甲基二吡啶并[1,2-a:1',2'-d]吡嗪-9-甲酰胺(式A5)。(The present invention relates to novel compounds which are active againstViral activity, and more specifically, inhibitory activity against the integrase of Human Immunodeficiency Virus (HIV). Subject of the invention are novel fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2-non-pyrido [1,2-a ] of the general formula 1 or 2]Pyrazine-7-carboxamide, and stereoisomers, pharmaceutically acceptable salts, solvates, and crystalline or polymorphic forms (I) and (II) thereof, wherein: ring A 1 Is an optionally methyl-substituted 5-7 membered saturated or bicyclic heterocycle, cyclic A 2 Is a 5-6 membered optionally methyl substituted saturated or partially saturated monocyclic heterocycle, cycloA 3 Is a 5-6 membered monocyclic saturated cycloalkane and tetrahydro-2H-pyran, R is a 5-7 membered monocyclic or bicyclic heterocyclic residue optionally substituted with 1,2 or 3 optionally identical substituents, said heterocyclic residue comprising 1-4 heteroatoms selected from O, S and N, but excluding (2S,5R,13aS) -8-hydroxy-7, 9-dioxo-N- { [3- (trifluoromethyl) -pyridin-2-yl]Methyl } -2,3,4,5,7,9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazepine-10-carboxamides (formula A4) and (1R,4S,12aR) -N- [ (3, 5-difluoropyridin-2-yl) methyl]-7-hydroxy-6, 8-dioxo-1, 2,3,4,6,8,12,12 a-octahydro-1, 4-methylenedipyrido [1,2-a:1',2' -d]Pyrazine-9-carboxamides (formula A5).)

1. Fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2 n-pyrido [1,2-a ] pyrazine-7-carboxamide of general formulae 1 and 2, or a stereoisomer, pharmaceutically acceptable salt, solvate, crystalline form or polymorph thereof

Wherein

Ring A1Is an optionally methyl-substituted 5-7 membered saturated heterocyclic or heterobicyclic ring,

ring A2Is an optionally methyl-substituted 5-6 membered saturated or partially saturated monocyclic heterocycle,

ring A3Is 5-6 membered monocyclic saturated cycloalkane and tetrahydro-2H-pyran,

r is a 5-7 membered monocyclic or bicyclic heterocyclic residue optionally substituted by 1,2 or 3 optionally identical substituents, said heterocyclic residue containing 1-4 heteroatoms selected from the series O, S and N, but excluding (2S,5R,13aS) -8-hydroxy-7, 9-dioxo-N- { [3- (trifluoromethyl) -pyridin-2-yl ] methyl } -2,3,4,5,7,9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5] pyrazino [2,1-b ] [1,3] oxazepinyl-heptatriene-10-carboxamide (formula A4) and (1R,4S,12aR) -N- [ (3, 5-difluoropyridin-2-yl) methyl ] -7-hydroxy-6, 8-dioxo-1, 2,3,4,6,8,12,12 a-octahydro-1, 4-methylenebipyridino [1,2-a:1',2' -d ] pyrazine-9-carboxamide (formula A5).

2. The compound of claim 1, wherein the heterocyclic residue R is thienyl, furyl, pyrazolyl, isoxazolyl, thiazolyl, oxazolyl, imidazolyl, thiadiazolyl, [1,2,5] oxadiazolyl, [1,2,4] triazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, 1,2, 3-triazinyl, 1,2, 4-triazinyl, and 1,3, 5-triazinyl, imidazo [2,1-b ] thiazolyl, imidazo [2,1-b ] [1,3,4] thiadiazolyl, benzothienyl, benzofuranyl, indolyl, 1, 3-benzodioxol-5-yl, 2, 3-dihydro-1, 4-benzodioxinn-6-yl, oxadiazinyl, thiadiazolyl, and thiadiazolyl, 1, 3-benzothiazolyl, 1, 3-benzoxazolyl, benzimidazolyl, 1, 3-dihydro-2-oxobenzimidazolyl, 2,1, 3-benzothiadiazolyl, 2,1, 3-benzooxadiazolyl, quinolinyl, isoquinolinyl, imidazo [1,2-a ] pyridyl, 1,2, 4-triazolo [4,3-a ] pyridyl, imidazo [1,2-a ] pyrimidinyl, imidazo [1,2-a ] pyrazinyl, 1,2, 4-triazolo [4,3-b ] pyridazinyl, 4,5,6, 7-tetrahydrobenzothienyl, 5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyridyl, 1,4,5,6,7, 8-hexahydrocyclohepta [ c ] pyrazolyl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] thiazolyl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] isothiazol-3-yl, [1,2,4] triazolo [4,3-b ] pyridazin-3-yl.

3. The compound of claim 1 or 2, wherein the heterocyclic residue R is 2-thienyl, 2-furyl, 1 n-pyrazol-3-yl, 1 n-pyrazol-4-yl, 1 n-pyrazol-5-yl, isoxazol-4-yl, thiazol-2-yl, 1, 3-oxazol-2-yl, imidazol-2-yl, 1,2, 3-thiadiazol-5-yl, 1,2, 5-oxadiazol-3-yl, 1,2, 4-oxadiazol-5-yl, 1 nn-1, 2, 4-triazol-3-yl, 1 nn-1, 2,3, 4-tetrazol-5-yl, 2-pyridyl, etc, 3-pyridyl, 4-pyridyl, pyridazin-4-yl, pyrimidin-4-yl, pyrazin-2-yl, imidazo [2,1-b ] thiazol-6-yl, imidazo [2,1-b ] [1,3,4] thiadiazol-6-yl, benzothien-5-yl, benzofuran-2-yl, 1N-indol-5-yl, 1, 3-benzodioxol-5-yl, 2, 3-dihydro-1, 4-benzodioxin-6-yl, 1, 3-benzothiazol-2-yl, 1, 3-benzoxazol-2-yl, 1N-benzimidazol-2-yl, pyrimidine-4-yl, pyrazin-2-yl, imidazo [2,1-b ] thiazol-6-yl, 1N-indol-5-yl, 1, 3-benzodioxol-2-yl, 1N-benzodioxol-2-yl, 1, N-benzodioxol-yl, 2-yl, N-yl, 1, 2-yl, 2-benzodioxol, 2-yl, or a pharmaceutically acceptable salt thereof, 1, 3-dihydro-2-oxobenzimidazol-5-yl, 2,1, 3-benzothiadiazol-5-yl, 2,1, 3-benzoxadiazol-5-yl, quinolin-2-yl, quinolin-3-yl, quinolin-4-yl, quinolin-5-yl, quinolin-6-yl, quinolin-7-yl, quinolin-8-yl, 1-isoquinolyl, imidazo [1,2-a ] pyridin-3-yl, 1,2, 4-triazolo [4,3-a ] pyridin-3-yl, imidazo [1,2-a ] pyrimidin-2-yl, imidazo [1,2-a ] pyrazin-3-yl, quinoxalinyl-2-yl, quinoxalinyl-3-yl, 1,2, 4-triazolo [4,3-b ] pyridazin-3-yl, 4,5,6, 7-tetrahydrobenzothiophen-2-yl, 5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyridin-3-yl, 1,4,5,6,7, 8-hexahydrocyclohepta [ c ] pyrazol-3-yl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] [1,3] thiazol-2-yl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] isothiazol-3-yl, [1,2,4] triazolo [4,3-b ] pyridazin-3-yl.

4. A compound according to any one of claims 1,2 or 3, wherein the heterocyclic residue R is substituted with 1,2 or 3 substituents selected from C1-C3 alkyl groups or halogen atoms, preferably F, Cl and Br.

5. A compound according to any one of claims 1,2,3 or 4 of general formula 1.1, 1.2 or 1.3, a stereoisomer, pharmaceutically acceptable salt, solvate, crystalline form or polymorph thereof

Wherein R is as given above.

6. A compound according to claim 1 of general formula 2.1 or 2.2, a stereoisomer, pharmaceutically acceptable salt, solvate, crystalline form or polymorph thereof

Wherein R is as given above.

7. A compound according to claim 1 selected from the following series:

8. a process for the production of compounds of formula 1 or 2 according to claim 1 and stereoisomers thereof by in the presence of CO and Pd (PPh)3)4By interaction of the corresponding bromide of formula 3 or 4 with a heterocyclylmethylamine in dimethyl sulfoxide in the presence of elevated temperature, followed by debenzylation of the resulting compound 5 or 6

Wherein the A1、А2、А3R and Bn are as given above.

9. A process for the production of compounds of formula 1 or 2 and stereoisomers thereof by acylation of heterocyclylmethylamines with the corresponding acids of compounds of formula 7 or 8, followed by debenzylation of the resulting compounds of formula 5 or 6

Wherein the A1、А2、А3And Bn are as given above.

10. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound of general formula 1 or 2 according to any one of claims 1 to 7 and a pharmaceutically acceptable excipient.

11. The pharmaceutical composition of claim 10, containing one or more additional therapeutic agents.

12. The pharmaceutical composition according to claim 10, containing an anti-HIV agent as an additional therapeutic agent.

13. The pharmaceutical composition of claim 12, containing one or more additional therapeutic agents selected from the group consisting of: HIV protease inhibitors, HIV non-nucleoside reverse transcriptase inhibitors, HIV nucleoside or nucleotide reverse transcriptase inhibitors, HIV capsid assembly inhibitors, and combinations thereof.

14. The pharmaceutical composition according to any one of claims 11-13, containing a first additional therapeutic agent selected from abacavir sulfate, tenofovir disoproxil fumarate, tenofovir alafenamide, tenofovir cyclobutylalafenamide hemifumarate, and tenofovir cyclobutylalafenamide fumarate, elsufavirine, and VM-1500A, GS-CA1, and a second additional therapeutic agent selected from emtricitabine and lamivudine.

15. The pharmaceutical composition according to claim 11, containing one or more anti-HBV agent as an additional therapeutic agent.

16. The pharmaceutical composition of claim 11, comprising one or more anti-HCV agents as an additional therapeutic agent.

17. The pharmaceutical composition according to claim 11, containing one or more anti-HBV agent and one or more anti-HCV agent as additional therapeutic agents.

18. The pharmaceutical composition according to claim 10 in the form of a lyophilizate, which is obtained by: freeze-drying a nanosuspension of a pharmaceutically effective amount of a compound of formula 1 or 2 according to any one of claims 1 to 7, said nanosuspension having a particle size of 200-900nm, preferably 200nm, containing a pharmaceutically acceptable excipient.

19. A process for preparing a pharmaceutical composition according to claim 18, said process consisting of the steps of: wet particles of a compound of the general formula 1 or 2 according to any one of claims 1 to 7 with excipients and water are milled to obtain a particle size of 200 and 900nm, preferably 200nm, and the resulting suspension is subsequently lyophilized.

20. The method of claim 19, wherein the excipient is selected from the group consisting of mannitol, polysorbates, polyethylene glycols, poloxamers, mannitol, and sucrose.

21. A pharmaceutical nanosuspension to be used as an injectable medicament for long-term maintenance therapy of HIV infection, comprising the pharmaceutical composition according to claim 18, phosphate buffered saline and water for injection.

22. A method of preparing a pharmaceutical nanosuspension by mixing the pharmaceutical composition of claim 18, Phosphate Buffered Saline (PBS) having a pH of 6.8, and water for injection.

23. An injectable medicament for long-term maintenance therapy of HIV infection, comprising the pharmaceutical composition according to claim 18, phosphate buffered saline and water for injection.

24. A method of preparing an injectable drug, the method consisting of mixing the pharmaceutical composition of claim 18, Phosphate Buffered Saline (PBS) having a pH of 6.8, and water for injection.

25. A method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed individual by administering to said individual a therapeutically effective amount of a compound of general formula 1 or 2 according to any one of claims 1-7, or a pharmaceutical composition according to any one of claims 10-18, or a pharmaceutical nanosuspension according to claim 21, or an injectable medicament according to claim 23.

Technical Field

The present invention relates to a novel compound having antiviral activity, in particular, inhibitory activity against the integrase of Human Immunodeficiency Virus (HIV).

Background

HIV is a lentivirus (a subset of retroviruses) responsible for the indolent disease HIV infection [ Weiss R.A. how does HIV cause AIDS. science 1993,260(5112),1273-1279.Douek D.C., Roederer M., Koup R.A. emulsifying conjugates in the Immunopathogenesis of AID, Annu.Rev.Med.2009,60,471-84]. Human immunodeficiency virus was independently discovered in 1983 in two laboratories: one was discovered by the French Pasteur Institute (Pasteur Institute in France) by a research team led by Luc Montagnier, and the other by Robert Gallo of the National Cancer Institute (National Cancer Institute in the United States). The discovery of the first Isolation of a new retrovirus from tissues of patients with symptoms of AIDS was discussed in journal Science, 5/20 in 1983 [ Barr é -Sinoussi F. et al, Isolation of a T-lymphopic retroviruses from a tissue for Acquired Immune Deficiency Syndrome (AIDS). Science 1983,220(4599),868-871.Gallo R.C. et al, Isolation of human T-cell leukemia virus in Acquired Immune Deficiency Syndrome (AIDS). Science 1983,220(4599),865 867.]. In 2008, Luc Montagnier andBarre-Sinoussi becauseTheir "discovery of human immunodeficiency virus" shares the Nobel Prize for Physiology and Medicine (Nobel Prize for Physiology and Medicine).

HIV virus infects cells of the immune system that have CD4 receptors on their surface: t helper cells, monocytes, macrophages, langerhans cells, dendritic cells, microglia. This results in: the development of immunosuppressive and acquired immunodeficiency syndrome (AIDS); the patient's ability to protect himself from infection and tumor loss; the appearance of secondary opportunistic diseases atypical for persons with a normal immune status. Without treatment, the mean survival time after HIV infection was estimated to be 9-11 years depending on the HIV subtype [ https:// en.wikipedia.org/wiki/HIV ].

According to global statistics [ http:// www.lenoblspid.ru/news 24/popular/own _ news/1166], 3670 million people worldwide were HIV carriers, 210 million people were newly infected with HIV, and 110 million people died from AIDS-related diseases in 2015. Since the start of the epidemic, 7800 million people have been infected with HIV, of which 3500 million people have died of AIDS-related diseases.

HIV is transmitted between humans by the exchange of bodily fluids such as blood, semen, rectal and vaginal fluids, and breast milk. It does not spread through saliva.

HIV can be inhibited by a combination antiretroviral therapy (APT) involving three or more antiretroviral drugs with different mechanisms of action. APT does not cure HIV infection, but inhibits viral replication in humans and helps to strengthen the immune system and restore its ability to fight infection. The life expectancy of patients receiving APT with single and/or combination of two-component drugs can be extended to 70-80 years of age. [ http:// www.who.int/media/videos/fs 360/ru/].

Antiretroviral drugs (ARDs) are classified as Nucleoside Reverse Transcriptase Inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), fusion inhibitors, capsid inhibitors, Protease Inhibitors (PIs) and integrase inhibitors (INI).

Examples of one-component ARDs may be: elsufavirine, VM-1500A [ WO 2005/102989, RU 2389719, WO 2010/028968]Ribavirin, a pharmaceutical compositionForest [ https:// pupchem. ncbi. nlm. nih. gov/compound/Rilpivirine # section ═ Top; https:// pubchem. ncbi. nlm. nih. gov/compound/5270790# section ═ Top]And efavirenz [ https:// pubchem. ncbi. nlm. nih. gov/compound/efavirenz]Is NNRTI; lamivudine [ https:// pubchem. ncbi. nlm. nih. gov/compound/lamivudine # section ═ 2D-Structure]Emtricitabine [ https:// pubchem. ncbi. nlm. nih. gov/compound/Emtricitabine # section ═ 2D-Structure]And their pro-inhibitors (proinihibitors) [ RU 2659388]Is NRTI; tenofovir Disoproxil Fumarate (Tenofovir Disoproxil Fumarate)[https://pubchem.ncbi.nlm.nih.gov/compound/Tenofovir_Disoproxil_Fumarate]Tenofovir alafenamide hemifumarate (TAF, GS-7340, Vemlidy) [ https:// pubchem. ncbi. nlm. nih. gov/compound/71492247]Tenofovir Cyclobutylalafenamide (TCBA) and TCBA fumarate [ RU 2647576 ]]Is NtRTTI; elvitegravir [ https:// ubchem. ncbi. nlm. nih. gov/compound/Elvitegravir]Is INI; GS-CA1(GS-6207) [ WO 2018035359; http:// www.natap.org/2018/IDWeek/IDWeek _03.htm]Is a capsid inhibitor; and can be compared with sitostat (Cobicistat) [ https:// pubchem. ncbi. nlm. nih. gov/compound/Cobicistat](which does not exhibit antiviral activity, but rather a pharmacokinetic enhancer) is an inhibitor of cytochrome P4503A (CYP 3A).

In recent years, INI has attracted a great deal of attention as a drug for combined APT involving the simultaneous use of several ARGs with different mechanisms of action, in particular for long-term HIV suppression (antiretroviral therapy as long-lasting suppression, ATLAS).

In 2007, the U.S. Food and Drug Administration (FDA) approved the use of INI HIV as an APT drug [ https:// www.healthline.com/health/HIV-aids/integrin-inhibitors # HIV ].

The first INI drug registered for APT by the FDA (10 months 2007) was Latiravir (Raltegravir) developed by Merk [ https:// aidsinfo. nih. gov/news/803/FDA-imprves-the-first-integrin-inhibitor-Raltegravir-october-12-2007 ], which was patented by Shionogi in Europe [ EP1422218(2004) ]. Shionogi later obtained patents to cabozir (Cabotegravir) and doliteravir (Dolutegravir) [ WO 2006/116764, US 8129385(2012), US 8778943(2015), EP 3260457(2017) ].

Later, patents were obtained for Canavavir [ WO 2014/100323], INI A1, A2 [ EP 3196201] and INI A3 [ WO 2016161382 ].

Wherein A1, A2: q is an optionally substituted carbocyclic or heterocyclic ring; a and D are optionally substituted heterocycle; r3And R8Independently hydrogen, halogen, hydroxy, optionally substituted lower alkyl and other substituents; r14And RхIndependently hydrogen, optionally substituted lower alkyl and other substituents.

A3: a is optionally substituted 3-7 membered cycloalkyl or partially unsaturated heterocycle; a. the1Is a 5-7 membered saturated or optionally substituted 4-7 membered monocyclic heterocycle; r1 nIs optionally the same halogen or C1-C3 alkyl; n is 1-3; r2Is optionally the same H or C1-C4 alkyl.

At present, CAB, DTG and BIC are the most advanced HIV INI. Depending on the assay, the inhibitory activity of CAB is EC50=0.25-3.0nMa,b(ii) a For DTG, EC50=1.1-7.4nMa-c(ii) a And for BIC, EC50=1.0-7.5nMb,c[aHassounah S.A. et al AAC 2017,61(12), pii: e01695-17.bYoshinaga T. et al AAC 2015,59(1),397-406;https://aac.asm.org/content/aac/59/1/397full.pdf;https://aac.asm.org/content/61/12/e01695-17.long.cTsiang M. et al AAC 2016,60(12), 7086-; https:// aac. asm. org/content/60/12/7086]。

Tourethrivir (trade name: Tivicay) was approved by the FDA [ https:// www.drugs.com/history/tifacay. html ] in 8 months of 2013, and the FDA approved drug Juluca [ https:// www.drugs.com/history/Juluca. html ] in 11 months of 2017, which is a fixed combination of 52.6mg of Tourethriv's sodium salt and about 27.5mg of rilpivirine hydrochloride [ https:// www.gsksource.com/pharma/content/dam/Glaxo SmithKline/US/en/Presscribingg _ Information/Juluca/julucf/Juluca-PI-PIL. PDF ].

Bicagvir is the component part of the drug Bictarvi, which is a fixed combination (50mg Bicagvir, 200mg emtricitabine and 25mg tenofovir alafenamide) [ http:// www.gilead.com// media/files/pdfs/medianes/hiv/biktarvy/biktarvy _ pi. Bictarvi was approved by FDA in 2018 [ https:// patents.glgoo.top/www.google.com/searchq ═ biktarvy + FDA + apuval + date & rlz ═ 1C1GGRV _ enUS751US751& oq ═ biktarvy + FDA & aqs ═ chrome.2.0j69857j0l 2.208j0j0j4 & sourceid ═ chrome & ie ═ UTF-8 ].

Carboplus is being actively studied in the clinic in the form of oral tablets and as long-acting injectable nanosuspensions to be injected into muscles (known as Carboplus LA or CAB LA (LA means "long-acting") [ https:// aidsinfo. nih. gov/drugs/513/Cabotegravir/0/patient. Trezza C. et al. Formulation and pharmacological of Long-acting Cabotegravir. current. image. HIV AIDS.2015,10 (239-245. T.D. McPherson et al. Cabotegravir in the treatment and prediction of Human Immunodeficiency Virus Virus-1. Experin Optig drugs.2018, (27 4-420. C.D. D. filtration Virus 2018), expression of Human Immunodeficiency Virus strain, 2018, Vm. filtration, see 31. acquisition, see 31. fig. 251, 1. experiment, 1. sample of Human immunoglobulin drugs, 31, 1. filtration, 1. C.D. D. shows.

Carbotevir is currently under active investigation as a drug in clinical stage III ATLAS (NCT02951052), FLAIR (NCT02938520), ATLAS-2M (NCT03299049) and ACTG A5359(NCT03635788) [ https:// aidsinfo. nih. gov/drugs/513/cabotegravir/0/patient ].

The cabozvir and a1-A3 inhibitors have not been approved by the FDA.

Despite the results obtained in recent years in the development of INI drugs for use in combination with APT, the search for new drugs of this type with improved characteristics is still of interest.

The following sets forth definitions of various terms used to describe the present invention. These definitions apply to the terms used throughout the specification and claims, either individually or as part of a larger group, unless otherwise limited in specific instances.

The term "HIV infection" is a disease caused by retroviruses, leading to progressive immunodeficiency (AIDS) and characterized by an increase in the end-stage of opportunistic diseases.

The term "AIDS-associated complex (AAC)" refers to the early symptomatic stage of HIV. This stage is associated with the risk of opportunistic infections. The clinical presentation of AAC is accompanied by the appearance of systemic symptoms: fever, profuse night sweats, 10% or more weight loss, progressive weakness. Characteristic features include the appearance of dermatological symptoms, damage to the oral mucosa, recurrent herpetic infections and recurrent cutaneous mucocandidiasis. Upper respiratory tract diseases (sinusitis, bronchitis and pneumonia), inflammatory diseases of pelvic organs, cervical dysplasia and peripheral neuropathy are often involved.

The term "AIDS" refers to acquired immunodeficiency syndrome or a later symptomatic stage. The infectious process lasts for 7-10 years. In some cases, the disease progresses more rapidly and enters the end stage after 2-3 years. This stage is characterized by severe life-threatening infections and malignant neoplasms, which have a generalized form (generalized form). The damage to organs and systems in the patient is irreversible.

The term "halogen" or "halo" refers to fluorine, bromine, chlorine and iodine.

The term "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the specified event or circumstance occurs and instances where it does not.

The term "crystalline form" refers to a structure of matter characterized by the packing of constituent molecules into certain types of crystal lattices.

The term "polycrystalline" refers to a polycrystalline structure of a substance, i.e., a structure consisting of a plurality of small single crystals or crystallites of a certain crystalline form.

The term "solvate" refers to the product of adding a solvent to a dissolved substance. A particular example of a solvate is a hydrate (where water acts as a solvent). Solvates generally form in solution, but are often obtained (upon cooling of the solution, evaporation of the solvent, etc.) in the form of a crystalline phase known as crystalline solvates.

The term "stereoisomers" (stereoisomers) are chemical compounds that have the same structure, but differ in the spatial arrangement of the atoms. Stereoisomers that are sterically incompatible with each other as mirror images are referred to as enantiomers or optical isomers. The phenomenon of optical isomerism is a characteristic of compounds whose molecules have a chiral element, for example an asymmetric (chiral) carbon atom bonded to four different substituents. It was first discovered in 1848 by l.pasteur using tartaric acid as an example and is explained by the concept of j.x.van Hoff and j.a.le Belem at 1874 based on the tetrahedral configuration of the carbon atoms in saturated compounds. Molecules containing asymmetric carbon atoms can be represented in the form of two optical isomers that do not overlap spatially (i.e., they are related to each other as their mirror images). Such differences are only in that the opposite arrangement of mirror isomers of the same substituent at the chiral center are referred to as enantiomers. In general, enantiomers have different biological activities, and are also characterized by optical activity-the ability to affect plane polarized light (to rotate the plane of polarization). Enantiomers rotate the plane of polarization at the same angle but in opposite directions and are therefore called enantiomers. For compounds with nn chiral centers in the molecule, the number of possible stereoisomers is 2n2 n. However, for n.gtoreq.2, there are stereoisomers that are distinguished by portions of the intrinsic chiral element. Stereoisomers other than enantiomers are referred to as diastereomers.

The term "pharmaceutically acceptable salt" of a given compound means a salt of: which retains the biological effectiveness and properties of the compound and is not biologically or otherwise undesirable. Pharmaceutically acceptable base addition salts can be prepared from inorganic or organic bases. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines such as alkylamines, dialkylamines, trialkylamines, substituted alkylamines, di (substituted alkyl) amines, tri (substituted alkyl) amines, alkenylamines, dienylamines, trienylamines, substituted alkenylamines, and the like. Also included herein are amines: wherein two or three substituents together with the nitrogen atom of the amino group form a heterocyclyl or heteroaryl group.

Pharmaceutically acceptable acid addition salts may be prepared from inorganic acids. In this case, salts derived from inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.

The term "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. They are used in the therapeutic compositions unless any conventional media or agent is incompatible with the active ingredient. The composition may also contain additional active ingredients.

The term "active ingredient" (drug substance) denotes a physiologically active substance of synthetic or other (biotechnological, plant, animal, bactericidal, etc.) origin having pharmacological activity, which is the active ingredient in a pharmaceutical composition.

The term "drug" refers to a substance (or mixture of substances in a pharmaceutical composition) in the form of tablets, capsules, injections, ointments, and other finished dosage forms intended to restore, correct or alter physiological function in humans and animals, as well as for the treatment and prevention of diseases, diagnosis, anesthesia, contraception, cosmetology, and the like.

The term "nanosuspension" specifically refers to a solid-liquid system having a particle size of less than 1 micron. In recent years, pure pharmaceutical products have been used as nanoparticles. Water is often used as the dispersion medium, where the substance is largely insoluble and is administered to the subject in the form of a suspension of nanoparticles. Nanosuspensions are of great importance in medicine for substances that are poorly soluble in water (<5mg/l), but improve their biopharmaceutical properties (e.g. absorption, bioavailability) in the form of suspensions.

The term "pharmaceutical composition" denotes a composition comprising a compound of formula 1 or 2 and at least one component selected from the group consisting of: pharmaceutically acceptable and pharmacologically compatible fillers, solvents, diluents, carriers, excipients, distribution and delivery agents such as preservatives, stabilizers, fillers, disintegrants, wetting agents, emulsifiers, suspending agents, thickening agents, sweeteners, flavoring and antibacterial agents, fungicides, lubricants and extended delivery control agents, the selection and proportions of which depend on the nature and on the route and dosage of administration. Examples of suitable suspending agents are ethoxylated isostearyl alcohols, polyoxyethylene, sorbitol and sorbitol ethers, microcrystalline cellulose, aluminum metahydroxide (aluminum metahydroxide), bentonite, agar-agar and tragacanth, and mixtures thereof. Protection against microorganisms can be provided using various antibacterial and antifungal agents, such as parabens, chlorobutanol, sorbic acid, and the like. The compositions may also contain isotonic agents, such as sugars, sodium chloride and the like. Sustained action of the composition can be achieved using agents that slow the absorption of the active ingredient, for example, aluminum monostearate and gelatin. Examples of suitable carriers, solvents, diluents and delivery agents include water, ethanol, polyols and mixtures thereof, natural oils (such as olive oil) and organic esters for injection (such as ethyl oleate). Examples of fillers are lactose, milk sugar, sodium citrate, calcium carbonate, calcium phosphate, etc. Examples of disintegrating and partitioning agents are starch, alginic acid and its salts and silicates. Examples of lubricants are magnesium stearate, sodium lauryl sulfate, talc and high molecular weight polyethylene glycols. Pharmaceutical compositions for oral, sublingual, transdermal, intramuscular, intravenous, subcutaneous, topical or rectal administration of active ingredients, alone or in combination with another active ingredient, can be administered to animals and humans in standard administration forms, as a mixture with conventional pharmaceutical carriers. Suitable unit dosage forms include: oral forms such as tablets, gelatin capsules, pills, powders, granules, chewing gums and oral solutions or suspensions; sublingual and buccal administration forms; an aerosol formulation; an implant; topical, transdermal, subcutaneous, intramuscular, intravenous, intranasal, or intraocular administration forms; and rectal administration forms.

The term "prodrug" refers to a compound that: when administered in vivo, it is metabolized by one or more steps or processes, or otherwise converted to a biologically, pharmaceutically, or therapeutically active form of the compound. A prodrug refers, for example, to a compound that is chemically designed to release the parent drug efficiently after overcoming the biological barrier of oral delivery. To obtain prodrugs, the pharmaceutically active compounds are modified such that the active compounds are regenerated as a result of metabolic processes. The prodrug may be intended to alter the metabolic stability or delivery characteristics of the drug in order to mask side effects or toxicity, to improve the taste of the drug, or to alter other characteristics or properties of the drug. When pharmaceutically active compounds are known, one skilled in the art can develop prodrugs of such compounds based on knowledge of the pharmacodynamic processes and drug metabolism in vivo (see, e.g., Nogrady's (1985) Medicinal Chemistry: A Biochemical Approach, Oxford University Press, New York, p. 388) -392).

The term "inert filler" as used herein denotes such compounds: which are used in the manufacture of pharmaceutical compositions and are generally safe, non-toxic and neither biologically nor otherwise undesirable, and include excipients that are acceptable for veterinary use and pharmacologically acceptable for human use. The compounds of the invention may be administered alone, but are generally administered in admixture with one or more pharmaceutically acceptable excipients, diluents or carriers selected with regard to the intended route of administration and standard pharmaceutical practice.

The term "therapeutically effective amount" as used herein refers to the amount of a substance, prodrug, or drug necessary to alleviate the symptoms of a disease in a subject. The dosage of the substance, prodrug or drug will meet the individual requirements in each particular case. The dosage may vary widely depending on numerous factors, such as the severity of the disease to be treated, the age and general condition of the patient, other drugs used to treat the patient, the mode and route of administration, and the experience of the attending physician. For oral administration, the daily dose may vary from about 0.01 to 10g, including all values therebetween, in monotherapy and/or in combination therapy. A preferred daily dosage is about 0.1-7 g. Typically, treatment begins with a large initial "loading dose" to rapidly reduce or eliminate the virus, and continues to reduce the dose to a level sufficient to prevent an infection outbreak.

The term "subject" refers to a mammal, including, but not limited to, cattle, pigs, sheep, chickens, turkeys, buffalos, alpacas, ostriches, dogs, cats, and humans; preferably the subject is a human.

Disclosure of Invention

Subject of the present invention is a novel fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2H-pyrido [1,2-a ] pyrazine-7-carboxamide of general formula 1 or 2, or any stereoisomer, any pharmaceutically acceptable salt, any solvate or any crystalline form or polymorph thereof:

wherein:

ring A1Is an optionally methyl-substituted 5-7 membered saturated heterocyclic or heterobicyclic ring;

ring A2Is a 5-6 membered saturated or partially saturated monocyclic heterocycle optionally substituted with methyl;

ring A3Is a 5-6 membered monocyclic saturated cycloalkane and tetrahydro-2H-pyran;

r is a 5-7 membered monocyclic or bicyclic heterocyclic residue optionally substituted by 1,2 or 3 optionally identical substituents, said heterocyclic residue comprising 1-4 heteroatoms selected from the series O, S and N, but excluding (2S,5R,13aS) -8-hydroxy-7, 9-dioxo-N- { [3- (trifluoromethyl) pyridin-2-yl ] methyl } -2,3,4,5,7,9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5] pyrazino [2,1-b ] [1,3] oxazepinyl-heptatriene-10-carboxamide (formula A4) and (1R,4S,12aR) -N- [ (3, 5-difluoropyridin-2-yl) methyl ] -7-hydroxy-6, 8-dioxo-1, 2,3,4,6,8,12,12 a-octahydro-1, 4-methylene (methano) bipyridino [1,2-a:1',2' -d ] pyrazine-9-carboxamide (formula A5)

An optionally substituted monocyclic heteroaryl residue R is selected from the series consisting of: thienyl, furyl, pyrazolyl, isoxazolyl, thiazolyl, oxazolyl, imidazolyl, thiadiazolyl, [1,2,5] oxadiazolyl, [1,2,4] triazolyl, tetrazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, 1,2, 3-triazinyl, 1,2, 4-triazinyl and 1,3, 5-triazinyl, imidazo [2,1-b ] thiazolyl, imidazo [2,1-b ] [1,3,4] thiadiazolyl, benzothienyl, benzofuranyl, indolyl, 1, 3-benzodioxol-5-yl, 2, 3-dihydro-1, 4-benzodioxinl-6-yl, 1, 3-benzothiazolyl, 1, 3-benzoxazolyl, 1,2, 5-benzodioxol-yl, Benzimidazolyl, 1, 3-dihydro-2-oxobenzimidazolyl, 2,1, 3-benzothiadiazolyl, 2,1, 3-benzoxadiazolyl, quinolyl, isoquinolyl, imidazo [1,2-a ] pyridyl, 1,2, 4-triazolo [4,3-a ] pyridyl, imidazo [1,2-a ] pyrimidyl, imidazo [1,2-a ] pyrazinyl, 1,2, 4-triazolo [4,3-b ] pyridazinyl, 4,5,6, 7-tetrahydrobenzothiophenyl, 5,6,7, 8-tetrahydro- [1,2,4] triazolo [4,3-a ] pyridyl, 1,4,5,6,7, 8-hexahydrocyclohepta [ c ] pyrazolyl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] thiazolyl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] isothiazol-3-yl, [1,2,4] triazolo [4,3-6] pyridazin-3-yl.

Preferably, the optionally substituted monocyclic heteroaryl residue R is selected from the series consisting of: 2-thienyl, 2-furyl, 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl, isoxazol-4-yl, thiazol-2-yl, 1, 3-oxazol-2-yl, imidazol-2-yl, 1,2, 3-thiadiazol-5-yl, 1,2, 5-oxadiazol-3-yl, 1,2, 4-oxadiazol-5-yl, 1H-1,2, 4-triazol-3-yl, 1H-1,2,3, 4-tetrazol-5-yl, 2-pyridyl, 3-pyridyl, 4-pyridyl, pyridazin-4-yl, pyridazin-5-yl, and the like, Pyrimidin-4-yl, pyrazin-2-yl, imidazo [2,1-b ] thiazol-6-yl, imidazo [2,1-b ] [1,3,4] thiadiazol-6-yl, benzothien-5-yl, benzofuran-2-yl, 1H-indol-5-yl, 1, 3-benzodioxol-5-yl, 2, 3-dihydro-1, 4-benzodioxin-6-yl, 1, 3-benzothiazol-2-yl, 1, 3-benzoxazol-2-yl, 1H-benzimidazol-2-yl, 1, 3-dihydro-2-oxobenzimidazol-5-yl, 1H-benzimidazol-2-yl, 1, 3-dihydrobenzoxazol-5-yl, and pharmaceutically acceptable salts thereof, 2,1, 3-benzothiadiazol-5-yl, 2,1, 3-benzoxadiazol-5-yl, quinolin-2-yl, quinolin-3-yl, quinolin-4-yl, quinolin-5-yl, quinolin-6-yl, quinolin-7-yl, quinolin-8-yl, 1-isoquinolinyl, imidazo [1,2-a ] pyridin-3-yl, 1,2, 4-triazolo [4,3-a ] pyridin-3-yl, imidazo [1,2-a ] pyrimidin-2-yl, imidazo [1,2-a ] pyrazin-3-yl, 1,2, 4-triazolo [4,3-b ] pyridazin-3-yl, and mixtures thereof, 4,5,6, 7-tetrahydrobenzothien-2-yl, 5,6,7, 8-tetrahydro [1,2,4] triazolo [4,3-a ] pyridin-3-yl, 1,4,5,6,7, 8-hexahydrocyclohepta [ c ] pyrazol-3-yl; 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] [1,3] thiazol-2-yl, 5,6,7, 8-tetrahydro-4H-cyclohepta [ d ] isothiazol-3-yl, [1,2,4] triazolo [4,3-b ] pyridazin-3-yl.

Preferred substituents of the heteroaryl residue R are 1,2 or 3 independent substituents selected from the group consisting of lower C1-C3 alkyl and halogen atoms, preferably F, Cl and Br.

The invention relates to compounds of general formula 1.1, 1.2 or 1.3, wherein R is as defined above, ring A1Respectively are 4-methyl-1, 3-oxazolidine, 4-methyl-1, 3-oxazinane and (1R,5S) -2-oxa-4-azabicyclo [3.2.1]Octane and pharmaceutically acceptable salts or solvates thereof

Wherein R is as given above.

Subject of the present invention are fused 9-hydroxy-1, 8-dioxo-N- (pyridylmethyl) -1,3,4, 8-tetrahydro-2H-pyrido [1,2-a ] pyrazine-7-carboxamides of general formula 2.1 or 2.2, wherein R is as given above, or their stereoisomers, their pharmaceutically acceptable salts, their solvates, their crystalline or polymorphic forms or their nanocrystalline forms

Wherein R is as given above.

Subject of the present invention is a fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2H-pyrido [1,2-a ] pyrazine-7-carboxamide of formula 1 or formula 2, selected from the series comprising:

the subject of the present invention is the production (scheme 1) of fused 9-hydroxy-1, 8-dioxo-N- (pyridine) of the general formula 1 or 2Ylmethyl) -1,3,4, 8-tetrahydro-2H-pyrido [1,2-a]Process for the preparation of pyrazine-7-carboxamides and their stereoisomers by reacting them at elevated temperature in the presence of CO and Pd (PPh)3)4The corresponding bromide (3,4) and heterocyclylmethylamine are allowed to interact in dimethyl sulfoxide in the presence of a catalyst, and the resulting compound (5,6) is subsequently debenzylated to give the desired product (1,2)

Scheme 1

Wherein Bn, R, and A1And an A2As given above.

Another subject of the invention is a process for the production (scheme 2) of fused 9-hydroxy-1, 8-dioxo-N- (pyridylmethyl) -1,3,4, 8-tetrahydro-2-in-pyrido [1,2-a ] pyrazine-7-carboxamide of general formula (1 or 2) and its stereoisomers by acylation of the heterocyclylmethylamine with the corresponding acid (7 or 8) and subsequent debenzylation of the resulting product (5,6) to give (1,2)

Scheme 2

Where Bn is as given above.

The nanomolar inhibitory activity of the novel compounds of formula 1 or 2 (table 1) is comparable to that of the state-of-the-art integrase inhibitors CAB, DLG and BIC. Thus, compounds 1.1.7, 1.1.17, 1.1.19 and 1.1.48 had EC of 0.59nM, 0.24nM and 0.43nM, respectively50The value is obtained. This result is unexpected because it is known that substitution of the benzyl moiety with a heterocyclylmethyl moiety in BIC and its analogs results in a dramatic decrease in inhibitory activity. Specifically, only two inhibitors, a4 and a5, are known to contain a pyridin-2-ylmethyl moiety instead of a benzyl moiety [ WO 2014/100323]]. In fact, in BIC (Compound 42, EC)50Replacement of the benzyl moiety with a pyridin-2-ylmethyl moiety in WO 2014/100323 (2.5 nM) resulted in a4 inhibitor (compound)Substance 49, EC5033.3nM in WO 2014/100323), which is 13.3 times less active than BIC. For a pair of inhibitors A5 (Compound 49, EC)5017.8nM in WO 2014/100323) and a6 (compound 66, EC)509.4nM in WO 2014/100323), a similar pattern was observed. Substitution of the benzyl moiety with a pyridine-2-base moiety in a6 resulted in a5 with a nearly 2-fold decrease in activity.

Novel compounds of formula 1 or 2 possess ADME properties desirable as drug candidates.

Depending on their structure, their solubility in aqueous solution differs (table 2). For example, compound 1.1.48 has a solubility in water (0.00285 μ g/ml) that is nearly 3 times lower than the solubility of CAB (0.008 μ g/ml), which can be a significant advantage when 1.1.48 is used as the INI in chronic injection HIV therapy. Also, for example, the solubility of the sodium salt 1.1.20 of compound 1.1.19 (2.679 μ g/ml) and the solubility of INI 1.1.19 (0.269 μ g/ml) exceed that of CAB by several orders of magnitude, making them more promising when used as INI in oral HIV therapy. In particular, this was confirmed by comparing the pharmacokinetic parameters obtained from compound 1.1.19 and the prototype CAB orally and intravenously administered to rats under the same conditions (table 3). As can be seen from Table 3, the pharmacokinetic parameter AUC of Compound 1.1.4 in the case of intravenous injectionINF、AUClast、СmaxAnd (T)1/2Significantly higher than the corresponding value for CAB. Even greater differences (2-fold or more) in these parameters were observed when these compounds were administered orally to rats (table 3). In addition, the time to peak plasma concentration (T)1/216.5h) significantly higher value (T) than CAB1/218.3h) and the bioavailability of compound 1.1.19 (17.5%) was 2 times that of CAB (8.8%) calculated by the following formula: f ═ AUC (AUC)last PO/5·AUClast IV)·100。

The novel compounds of general formula 1 or 2 are stable in human and murine plasma (figure 2) and the human and murine liver S9 fraction (figure 3) and have a high binding to human and rat plasma proteins (table 4).

The subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or a compound of general formula (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or a polymorph thereof.

The subject of the present invention is a pharmaceutical composition containing a compound of general formula 1.1, 1.2, 1.3, 2.1 or 2.2 or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof and a pharmaceutically acceptable excipient.

The subject of the present invention is a pharmaceutical composition containing a compound selected from the group consisting of the compounds in the ranges 1.1.1-1.1.65, 1.2.1-1.2.4, 1.3.1-1.3.3, 2.1.1-2.1.3, 2.2.1-2.2.3 or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof and a pharmaceutically acceptable excipient.

The subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, and one or more additional therapeutic agents.

The subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, and one or more anti-HIV agents.

Subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, and one or more additional therapeutic agents selected from: HIV protease inhibitors, non-nucleoside HIV reverse transcriptase inhibitors, nucleoside or nucleotide HIV reverse transcriptase inhibitors, allosteric HIV integrase inhibitors, HIV capsid assembly inhibitors, and combinations thereof.

The subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, and a first additional therapeutic agent selected from abacavir sulfate, tenofovir disoproxil fumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutyl alafenamide hemifumarate, elsulfavirin, 1500-CA-1500A, GS-1, and a second additional therapeutic agent selected from emtricitabine and lamivudine.

The subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, and a first additional therapeutic agent selected from tenofovir cyclobutylalafenamide, tenofovir cyclobutylalafenamide fumarate or tenofovir cyclobutylalafenamide, tenofovir cyclobutylalafenamide fumarate and tenofovir cyclobutylalafenamide hemifumarate, tenofovir cyclobutylalafenamide hemifumarate.

The subject of the present invention is a pharmaceutical composition containing a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, and one or more anti-Hepatitis C (HCV) and/or anti-Hepatitis B (HBV) agents.

anti-HCV agents can include:

-prodrugs of nucleoside inhibitors of HCV NS5B, for example,(Sofosbuvir, PSI-7977, GS-7977) [ https:// www.accessdata.fda.gov/drug satda _ docs/label/2015/204671s004lbl]Or (S) -2- { (S) - [ (2R,3R,4R,5R) -5- (3, 4-dihydro-2, 4-dioxo-2H-pyrimidin-1-yl) -3-hydroxy-4-methyl-4-fluoro-tetrahydrofuran-2-ylmethoxy]-phenoxy-phosphorylamino } -propionic acid cyclobutyl ester [ patent RU 2644256(2018)];

HCV NS5A inhibitors, e.g., Daclatasvir (Daklinza) (Daclatatasvir, BMS790052) [ https:// www.accessdata.fda.gov/drug satda _ docs/label/2017/206843s006lbl.pdf ]; heapivir (AV-4025) [ Ivachthkenko, A.V. et al, Discovery of Novel high content pharmaceuticals C Virus NS5A Inhibitor (AV-4025), J.Med.chem.2014,57,7716-; obetavir (Ombitasvir) (ABT-267) [ https:// www.drugbank.ca/drugs/DB09296 ]; elbavar (Elbasvir) (MK-8742), [ https:// www.drugbank.ca/drugs/DB11574 ]; velpatasvir (Velpatasvir) (VEL, GS-5816), [ https:// www.drugbank.ca/drugs/DB11613 ]; perfermavir (Pibrentasvir) (ABT-530; ABT 530; ABT 530) [ https:// www.drugbank.ca/drugs/DB13878 ]; gezoprevir (Grazoprevir) (MK-5172) [ https:// www.drugbank.ca/drugs/DB1157]

-HCV NS3 inhibitors, e.g., Narapivir (Narlaprevir) (SCH 900518) [ https:// newdragrapprovervals.org/tag/Narlaprevir /)/];

HCV NS3/NS4 inhibitors, e.g., Olysio (Ciimipirvir (Simeprevir)) [ https:// www.accessdata.fda.gov/drug atfda _ docs/label/2013/205123s001lbl.pdf ]; voxilaprevir (Voxilaprevir) (GS-9857; GS 9857; GS9857) [ https:// www.drugbank.ca/drugs/DB12026 ];

combinations of the above HCV inhibitors [ V.Soriano et al. Treatment of hepatitis C with new fixed dose combinations. expert Opin Pharmacother.2017, 8 months; 18(12):1235-1242.]Including fixed combinations, e.g. EPCLU(Sofosbuvir and Wippatasvir) [ https:// www.accessdata.fda.gov/drug atfdda _ docs/label/2016/208341s000lbl];

Bidada (Zepatier) (Elmbavir/Gezopivir) [ https:// www.merck.com/product/usa/pi _ circulators/z/Zepatier/Zepatier _ pi];Viekira XRTM(Dacebuvir (dasabuvir), orbetavir, paritaprevir, and ritonavir) [ https:// www.rxabbvie.com/pdf/viekiraxr _ pi](ii) a Mavyret (glecaprevir)/perambuvir) [ https:// www.rxabbvie.com/pdf/Mavyret _ pi]And the like.

anti-HCV agents can include Baraclude (entecavir) [ https:// packageinserters. bms. com/pi/pi _ Baraclude. pdf ]; epivir (lamivudine) [ https:// www.ema.europa.eu/documents/product-information/EPivir-ear-product-information _ en.pdf ]; hepera (adefovir, adefovir dipivoxil) [ https:// www.ema.europa.eu/documents/product-information/hepera-ear-product-information _ en.pdf ]; tyzeka (Telbivudine) [ https:// www.accessdata.fda.gov/drug atfdda _ docs/label/2006/022011lbl.pdf ]; viread (tenofovir) [ https:// www.ema.europa.eu/documents/variation-report/video-h-c-419-ii-0120-ear-assessment-report-variation _ en.pdf ]; vemlidy (tenofovir alafenamide fumarate, TAF) [ https:// www.gilead.com// media/files/pdfs/medianes/liver-disease/vmidy _ pi. pdfla ═ en ]; fumaric acid (S) -2- [ [ [ (R) -2- (6-aminopurine-9-yl) -1-methyl-ethoxy ] methyl-phenoxy-phosphoryl ] amino ] -propionic acid cyclobutyl ester [ patent RU 2674576(2018) ], and the like.

Another subject of the present invention is a pharmaceutical composition in the form of a lyophilizate obtained by freeze-drying a nanosuspension of a compound of general formula 1 or 2, said nanosuspension having a particle size of from 200nm to 900nm, preferably 200nm, containing a pharmaceutically acceptable excipient.

The process for preparing a lyophilized pharmaceutical composition consists of the following steps: the wet particles of the compound of formula 1 or 2 with excipients and water are milled to obtain a particle size of 200-900nm, preferably 200nm, and the resulting suspension is subsequently lyophilized.

The excipients used in the lyophilized pharmaceutical composition are selected from mannitol, polysorbates, polyethylene glycols, poloxamers, mannitol, and sucrose.

The subject of the present invention is also an injectable drug for long-term supportive therapy of HIV infection, comprising a pharmaceutical composition containing a lyophilized compound of general formula 1 or 2, a phosphate buffered saline solution and water for injection.

The process for the preparation of the novel injectable drug consists of the following steps: mixing a pharmaceutical composition containing a lyophilized compound of formula 1 or 2, a Phosphate Buffered Saline (PBS) solution at pH 6.8, and water for injection.

Subject of the present invention is a method of treating HIV infection in an HIV-infected or HIV-exposed individual by administering to said individual a therapeutically effective amount of a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt or solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition or injectable medicament as disclosed herein.

Subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed individual by administering to said individual a therapeutically effective amount of a compound of general formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition or injectable medicament as disclosed herein, and further administering to said human a therapeutically effective amount of one or more additional therapeutic agents.

The subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed human by administering to said human a therapeutically effective amount of a compound of general formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition or injectable medicament disclosed herein, and further administering to said human a therapeutically effective amount of one or more additional therapeutic agents selected from the group consisting of: HIV protease inhibitors, non-nucleoside HIV reverse transcriptase inhibitors, nucleoside or nucleotide HIV reverse transcriptase inhibitors, allosteric HIV integrase inhibitors, HIV capsid assembly inhibitors, and combinations thereof.

In a particular embodiment, the method for preventing and treating HIV infection in an HIV-infected or HIV-exposed human involves administering to said human a therapeutically effective amount of a compound of formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition or injectable medicament disclosed herein, and further administering to said human a therapeutically effective amount of a non-nucleoside HIV reverse transcriptase inhibitor.

The subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed human by administering to said human a therapeutically effective amount of a compound of general formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition or injectable medicament disclosed herein, and further administering to said human a therapeutically effective amount of a first additional therapeutic agent selected from abacavir sulfate, tenofovir disoproxil fumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, or tenofovir cyclobutylalafenamide hemifumarate, elsufavirine, VM-1500A, GS-CA1, and the second additional therapeutic agent is selected from emtricitabine and lamivudine.

The subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed individual by administering to said individual a single dosage form for one administration, e.g. in a solid dosage form for oral administration of a compound of general formula (i) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, in combination with a first additional therapeutic agent selected from abacavir sulfate, tenofovir difurfuryl, tenofovir disoproxil fumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutyl alafenamide fumarate, tenofovir cyclobutyldelavamide hemifumarate, tenofovir disoproxil fumarate, tenofovir disoproxil fumarate, or a, elsufavirine, VM-1500A, GS-CA, and the second additional therapeutic agent is selected from emtricitabine and lamivudine.

Subject of the present invention is the use of a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, in antiretroviral therapy (APT), in combination antiretroviral therapy (APT) and as long-lasting inhibitory Antiretroviral Therapy (ATLAS) for the treatment of HIV infection in HIV infected or HIV exposed humans.

The subject of the present invention is the use of the pharmaceutical composition or injectable medicament disclosed herein in antiretroviral therapy (APT), in combination antiretroviral therapy (APT) and antiretroviral therapy as long-lasting inhibition (ATLAS) for the treatment of HIV infection in HIV infected or HIV exposed humans.

The subject of the present invention is the use of a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition or injectable medicament as disclosed herein, for pre-exposure prophylaxis before an individual is exposed to HIV, to prevent the spread of an HIV infection (in case the individual is exposed to the virus), and/or to prevent a persistent viral infection, and/or to prevent the onset of symptoms, and/or to prevent the appearance of detectable levels of the virus in the blood.

The subject of the present invention is a method for inhibiting HIV replication using a therapeutically effective amount of a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition or injectable medicament disclosed herein to affect HIV under conditions promoting HIV replication inhibition.

Subject of the present invention is the use of a compound of general formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a pharmaceutical composition or injectable medicament disclosed herein, for inhibiting the activity of HIV integrase.

Subject of the present invention is the use of a compound of general formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or of a pharmaceutical composition or injectable medicament disclosed herein, as an HIV inhibitor.

It will be appreciated that in any of the embodiments of the invention described above, a compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a compound of formula (1) or (2) described herein as described above, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereofSalt, solvate thereof, or A in crystalline form or polymorph thereof1、А2、A3Or any particular substituent of the group of R, may be independently combined with other embodiments and/or substituents of the compound of any one of formulas (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, to yield embodiments not specifically described above. In addition, if in a particular embodiment and/or in the claims is any particular A1、А2、A3Or R gives a list of substituents, it is to be understood that each individual substituent may be excluded from a particular embodiment and/or claim, and the remaining list of substituents will be within the scope of the embodiments disclosed herein.

A compound of general formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, may be administered to a subject in pure form or as part of a corresponding pharmaceutical composition or injectable medicament disclosed herein, according to embodiments disclosed herein, by any conventional means of administering agents of similar purpose.

According to embodiments disclosed herein, the pharmaceutical compositions disclosed herein may be prepared by combining a compound according to embodiments disclosed herein with suitable pharmaceutically acceptable excipients, and may be formulated in solid, semi-solid, or liquid gaseous forms (such as tablets, capsules, powders, granules, ointments, nanosuspensions, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols). Typical routes of administration for the pharmaceutical composition include, but are not limited to, oral, topical, transdermal, pulmonary, parenteral, sublingual, buccal, rectal, vaginal and intranasal.

Pharmaceutical compositions according to embodiments disclosed herein are formulated so as to provide bioavailability of the active ingredient within the composition to be administered to a subject. The composition is administered to a subject or patient in a dosage form containing one or more dosage units, where, for example, a tablet may be a dosage form containing one dosage unit and a container with a compound (in the form of an aerosol) according to embodiments disclosed herein may contain a number of dosage units.

The actual methods for preparing the dosage forms are known or will be apparent to those skilled in the art; see, for example, Remington, the Science and Practice of Pharmacy, 20 th edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will in any event comprise a therapeutically effective amount of a compound according to embodiments disclosed herein, or a pharmaceutically acceptable salt thereof, for treating the disease or disorder of interest according to the recommendations described herein.

In one embodiment of the invention, the pharmaceutical composition is an oral unit dosage form.

In another embodiment, the pharmaceutical composition is a solid oral unit dosage form.

In another embodiment, the pharmaceutical composition is a tablet.

In another embodiment, the pharmaceutical composition is a capsule.

In another embodiment, the pharmaceutical composition is a lyophilized nanosuspension placed in a pre-sterilized glass vial that is covered with a pre-sterilized stopper and tightly sealed.

The pharmaceutical compositions disclosed herein may be prepared by methods well known in the pharmaceutical art. For example, pharmaceutical compositions intended for administration by injection may be prepared by combining the compounds of the embodiments disclosed herein with sterile distilled water to form a solution or nanosuspension. To ensure homogeneity of the solution or nanosuspension, a surfactant may be added.

Surfactants are compounds which: which interacts non-covalently with and facilitates dissolution of a compound according to embodiments disclosed herein or homogeneity of a nanosuspension of the compound in an aqueous delivery system.

A compound of general formula (1) or (2) according to embodiments disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof (on its own or within a corresponding pharmaceutical composition) is administered to a subject or patient in a therapeutically effective amount that will vary with factors such as the activity of the particular compound used; metabolic stability and duration of action of the compound; the age, weight, general health, sex, and diet of the patient; mode and time of administration; the rate of excretion; a pharmaceutical composition; the severity of the particular disorder or condition, and the subject being treated.

In certain embodiments of the present invention, for the treatment or prevention of HIV infection in an HIV-infected or HIV-exposed human, a method is provided comprising administering to the human a therapeutically effective amount of a compound disclosed herein, or a stereoisomer thereof disclosed herein, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a pharmaceutical composition disclosed herein, in combination with a therapeutically effective amount of one or more (e.g., 1,2,3, 1 or 2, or 1-3) additional therapeutic agents (up to 3).

In certain embodiments of the present invention, methods for treating HIV infection as provided herein are presented, which involve, if necessary, administering to a patient a therapeutically effective amount of a compound as disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt or solvate thereof, or a crystalline form or polymorph thereof, in combination with a therapeutically effective amount of one or more (e.g., 1,2,3, 1 or 2, or 1-3) additional therapeutic agents suitable for treating HIV infection.

A compound disclosed herein (e.g., any compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof) can be combined with one or more additional therapeutic agents at any dose of the compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof (e.g., from 50mg to 1000mg of the compound).

In one embodiment of the invention disclosed herein, a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, is used in combination with one or more (e.g., 1,2,3, 1 or 2, or 1-3) additional therapeutic agents and pharmaceutically acceptable excipients.

In the above embodiments, the additional therapeutic agent may be an anti-HIV agent. For example, in certain embodiments of the invention, the additional therapeutic agent is selected from HIV protease inhibitors, non-nucleoside or non-nucleotide HIV reverse transcriptase inhibitors, nucleoside or nucleotide HIV reverse transcriptase inhibitors, HIV integrase inhibitors, non-catalytic (or allosteric) site HIV integrase inhibitors, HIV nucleoside or nucleotide reverse transcriptase inhibitors, HIV capsid assembly inhibitors, HIV penetration inhibitors (e.g., CCR5 inhibitors, gp41 inhibitors (t + e + fusion inhibitors) and CD4 attachment inhibitors, CXCR4 inhibitors, gpl20 inhibitors, G6PD and NADH oxidase inhibitors, anti-HIV vaccines, HIV maturation inhibitors, latency reversal agents (e.g., histone deacetylase inhibitors, proteasome inhibitors, protein kinase c (pkc) activators and BRD inhibitors), HIV capsid compounds ("capsid inhibitors", such as capsid polymerization inhibitors or capsid disrupting compounds, HIV p7 nucleocapsid protein inhibitors (NCp7), and HIV p24 capsid protein inhibitors), pharmacokinetic enhancers, immunological therapies (e.g., Pd-1 modulators, Pd-Ll modulators, toll-like receptor modulators, IL-15 agonists), HIV antibodies, bispecific antibodies, and "antibody-like" therapeutic proteins (e.g.,fab derivatives), including agents that act on HIV gpl20 or gp41, anti-HIV combinations, HIV p17 matrix protein inhibitors, IL-13 antagonists, peptidyl-prolyl cis/trans isomerase a modulators, protein disulfide isomerase inhibitors, C5a complement receptor antagonists, DNA methyltransferase inhibitors, HIV vif gene modulators, HIV-1 viral infectious agent inhibitors, TAT protein inhibitors, HIV-1Nef modulators, Nek tyrosine kinase modulators, mixed lineage kinase 3(MLK-3) inhibitors, HIV-1 splicing inhibitors, Rev protein inhibitors, integrin antagonists, nucleoprotein inhibitors, splicing factor modulators, protein 1 modulators containing COMM domains, HIV ribonuclease H inhibitors, late cyclin (retrocyclin) modulators, CDK-9 inhibitors.Formulations, dendritic cell specific ICAM-3-grasping non-integrin protein-1 inhibitors (cleaving transforming protein-1inhibitor), HIV GAG protein inhibitors, HIV POL protein inhibitors, complement factor H modulators, ubiquitin ligase inhibitors, deoxycytidine kinase inhibitors, cyclin dependent kinase inhibitors, pro-protein converting enzyme PC9 stimulators, ATP dependent RNA helicase DDX3X inhibitors, inhibitors of priming reverse transcriptase complexes, HIV gene therapy, PI3K inhibitors, compounds similar to those disclosed in WO 2013/006738, US 2013/0165489, WO 2013/091096a1, WO 2009/062285, US 20140221380, US 20140221378, WO 2010/130034, WO2013/159064, WO2012/145728, WO 2012/003497, WO 2014/100323, WO2012/145728, WO2013/159064, WO 2012/003498 and WO 2013/006792, and other drugs and combinations thereof for HIV therapy.

In certain embodiments, the compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, is prepared in the form of a tablet, which may optionally contain one or more other compounds for HIV treatment. In certain embodiments, the tablet may contain another active ingredient for the treatment of HIV, such as HIV protease inhibitors, non-nucleoside or non-nucleotide HIV reverse transcriptase inhibitors, nucleoside or nucleotide HIV reverse transcriptase inhibitors, HIV integrase inhibitors, non-catalytic (or allosteric) HIV integrase site inhibitors, HIV capsid assembly inhibitors, pharmacokinetic enhancers, and combinations thereof.

In certain embodiments of the invention, the tablet is suitable for 1 administration per day.

In certain embodiments, the additional therapeutic agent is:

-one or more combinations selected from:(efavirenz + tenofovir disoproxil fumarate + emtricitabine),(rilpivirine + tenofovir disoproxil fumarate + emtricitabine),(avivoravir + cobicistat + tenofovir disoproxil fumarate + emtricitabine), doriravir + abacavir sulfate + lamivudine,(Dotiravir + abacavir + lamivudine), lamivudine + Nevirapine + Zidovudine, Dotiravir + Ripivirine, atazanavir sulfate + Coxsita, atazanavir + Coxsita, Darunavir (darunavir) + Coxsita, efavirenz + Laevidine + Tenofovir disoproxil fumarate, Tenofovir alafenamide hemifumarate + Entricitabine + Coxsita + Emviravir, Tenofovir alafenamide hemifumarate + Entricitabine, Tenofovir alafenamide + Entricitabine + Rivasavine, Vacc-4x + Milodaricitabine, Tenamivir + Entricitabine, Tenofovir + Altricitabine + Rivastigmine, Vacuc-4 x + Milodavir (Nomidepsin + Entricitabine), Tenofovir + Altricitabine, Adriavir,(lopinavir + ritonavir), atazanavir sulfate + ritonavir,(zidovudine + lamivudine, AZT +3TC),(abacavir sulfate+ lamivudine, ABC +3TC),(abacavir sulfate + zidovudine + lamivudine, ABC + AZT +3TC),(tenofovir disoproxil fumarate + emtricitabine, TDF + FTC), tenofovir + lamivudine, lamivudine + tenofovir disoproxil fumarate, and lamivudine + tenofovir cyclobutyl alafenamide or fumarate or hemifumarate thereof;

-an HIV protease inhibitor selected from amprenavir, atazanavir, furametpyr (fosamprenavir), furametpyr calcium, indinavir sulfate, lopinavir, ritonavir, nelfinavir mesylate, saquinavir mesylate, tipranavir, brecanavir, darunavir, DG-17, TMB-657(PPL-100) and TMC-310911;

-non-nucleoside or non-nucleotide HIV reverse transcriptase inhibitors selected from delavirdine, delavirdine mesylate, nevirapine, etravirine (etravirine), dapivirin, doravilin, rilpivirine, efavirenz, KM-023, VM-1500, lentinan and AIC-292;

-nucleoside or nucleotide HIV reverse transcriptase inhibitors selected fromAnd(didanosine, ddl), zidovudine, emtricitabine, didanosine, stavudine, zalcitabine, lamivudine, censvudin, abacavir sulfate, amdoxovir (amdoxovir), elvucitabine (elvucitabin), alovudine, phosphazide, fozivudine tidoxil, aricitabine (apricitibine), amdoxovir, KP-1461, fosalvudine tidoxil, tenofovir disoproxil fumarate, tenofovir hemifumarateDipyridamole, tenofovir alafenamide hemifumarate, tenofovir alafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, elsufavirine, VM-1500A, adefovir dipivoxil, and festinavir;

-an HIV integrase inhibitor selected from curcumin, curcumin derivatives, chicoric acid derivatives, 3, 5-dicaffeoylquinic acid derivatives, aurintricarboxylic acid derivatives, caffeic acid phenethyl ester derivatives, tyrfostin derivatives, quercetin derivatives, latilatavir, elvitegravir, dortavir and cabotevir;

-HIV non-catalytic (or allosteric) site integrase inhibitors (NCINI) selected from CX-05168, CX-05045 and CX-14442;

-an HIV capsid assembly inhibitor selected from GS-CA 1;

-an HIV gp41 inhibitor selected from the group consisting of enfuvirtide, cidevitide (sifuvirtide) and ibufenade (albivitide);

-an HIV penetration inhibitor selected from the group consisting of cericiviroc;

-an HIV gpl20 inhibitor selected from Radha-108 (receptor) and BMS-663068;

-a CCR5 inhibitor selected from alavirol, viriviroc, maravirroc, ceriviroc, PRO-140, adaptavir (RAP-101), TBR-220(TAK-220), nifeviroc (TD-0232), TD-0680 and vmip (haiipipu);

-a CD4 attachment inhibitor selected from ibalizumab (ibalizumab);

-CXCR4 inhibitors selected from plerixafor, ALT-1188, vMIP and Haimipu;

-a pharmacokinetic enhancer selected from the group consisting of costat and ritonavir;

-immunological drugs selected from modulators of dermaVir, interleukin-7, praquinqueril (hydroxychloroquine), pre-interleukin (aldesleukin, IL-2), interferon alpha-2 b, interferon alpha-3 b, pegylated interferon alpha, interferon gamma, hydroxyurea, mycophenolic acid (MPA) and its ether derivative mycophenolmofetil (MMF), WF-10, ribavirin, IL-2, IL-12, polyethyleneimine Polymer (PEI), hepone, VGV-1, MOR-22, BMS-936559, toll-like receptors (tlrl, tlr2, tlr3, tlr4, tlr5, tlr6, tlr7, tlr8, tlr9, tlrlO, tlrl, tll 2 and rll 3), rin-toli and IR-103;

an anti-HIV vaccine selected from the group consisting of peptide vaccines, recombinant subunit-based vaccines, live vector vaccines, DNA vaccines, virus-like particle-based vaccines (pseudovirion-based vaccines), CD4 derivative-based peptide vaccines, vaccine combinations, rgpl20(AIDSVAX), ALVAC HIV (vCP1521)/AIDSVAX IN/E (gpl20) (RV144), Remune, ITV-1, Contre Vir, Ad5-ENVA-48, DCVax-001(CDX-2401), PEP-6409, Vacc-4x, Vacc-C5, VAC-3S, recombinant DNA adenovirus serotype 5(rAd5) -based multiclass vaccines, Pennvax-G, VRC-HIV MAB 060-00-cGX-101, vaccines Tat OYAVi, AB X-201, HIV-vax, Ad35, DNA 35-3/VSNAP 8651/RCGAX-51, Poly-ICLC adjuvant vaccine, Tatlmmune, GTU-MultiHIV (FIT-06), AGS-004, gpl40[ delta ] V2.TVl + MF-59, rVSVIN HIV-1Gag vaccine, SeV-Gag vaccine, AT-20, DNK-4, Ad35-GRIN/ENV, TBC-M4, HIVAX, HIV AX-2, NYVAC-HIV-PT1, NYVAC-HIV-PT4, DNA-HIV-PT123, rAAVl-PG9DP, GOVX-B11, GOVX-B21, ThV-01, TUTI-16, VGX-3300, TVI-HIV-1, Ad-4(Ad4-env Clade C + Ad4-mGag), EN41-UGR7C, EN41-FPA2, PreVaxtat, TL-01, SAV-001, AE-H, MYM-V101, CombiHIVvac, ADVAX, MYM-V201, MVA-CMDR, ETV-01, and DNA-Ad5 gag/pol/nef/nev (HVTN 505);

anti-HIV antibodies, bispecific antibodies and "antibody-like" therapeutic proteins (such asFab derivatives), including BMS-936559, TMB-360 and drugs acting on gpl20 or gp41 HIV, selected from baviximab (bavituximab), UB-421, C2F5, C2G12, C4E10, C2F5+ C2G12+ C4E10, 3-BNC-117, PGT145, PGT121, MDX010 (ipilimumab), VRC01, AT 32, 7B 2, 10E 8 and VRC 07;

-latency reversal agents selected from histone diacetyl esterase inhibitors such as romidepsin, vorinostat, panobinostat; proteasome inhibitors such as velcade; protein Kinase C (PKC) activators such as indolactam (indoctam), prostratin, ingenol B and DAT lactone, ionomycin, GSK-343, PMA, SAHA, inhibitors BRD4, IL-15, JQ1, disulfiram and amphotericin B;

-HIV p7 nucleocapsid protein inhibitor (NCp7) selected from azodicarbonamide;

-an HIV maturation inhibitor selected from BMS-955176 and GSK-2838232;

-PI3K inhibitor selected from idelalisib, AZD-8186, buparlisib, CLR-457, picrolisib (pictilib), neratinib (neratinib), rigosertib sodium, EN-3342, TGR-1202, aspergilli (alpelisib), daverinib (duvelisib), UCB-5857, taselisib, XL-765, gedatolisib, VS-5584, copanissib (copanisib), CAT orotate, perifoscin (perifossin), RG-7666, GSK-2636771, DS-7423, panculisib, GSK-2269557, GSK-2126458, CUDC-907, PQR-309, PQR-040093, larealisib, saralalisib-17232, SAR-1113-3978, mlsinp-3879, amslr-1126, sarrp-grin-3879, sarrp-1113, sarrp-387, SARs-1113, SARs-3976, SARs-3970, SARs-413, SARs-3970, sarp-3879, mlisib-413, SARs-3970, SARs-3, SARs-3970, SARs-3976, SARs-3, SARs-3976, SARs-3970, SARs-3976, SARs-p-3976, SARs-3, SARs-3970, SARs-3976, SARs-3, SARs-p-3970, SARs-3976, SARs-p-3976, SARs-3970, SARs-p-3, SARs-p-3976, SARs-p, SARs-p, SARs-p-3879, SARs-p, SARs-p, SARs-p, SARs-p, SARs-p;

compounds disclosed in WO 2004/096286, WO 2006/110157, WO 2006/015261, WO 2013/006738, US 2013/0165489, US 20140221380, US 20140221378, WO 2013/006792, WO 2009/062285, WO 2010/130034, WO 2013/091096a1, WO2013/159064, WO2012/145728, W02012/003497, W02014/100323, WO2012/145728, WO2013/159064 and WO 2012/003498;

-a further anti-HIV drug selected from BanLec, MK-8507, AG-1105, TR-452, MK-8591, REP 9, CYT-107, alisporivir, NOV-205, IND-02, methionine enkephalin, PGN-007, acetyl-pyran (acemannan), gamimune, purastine, 1, 5-dicaffeoylquinic acid, BIT-225, RPI-MN, VSSP, HiViral, 1 moko-phenico, SB-728-T, RPI-MN, VIR-576, HGTV-43, polka-1376, rHIV 7-shl-TAR-5 RZ, MazF-CCR gene therapy, akAickAide, acbkB-464, SCY-85, naltrexone, AAV-eCD4-Ig gene therapy, and aIg-1050040 (aag-040).

In certain embodiments of the invention, a compound of formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate or crystalline form or polymorph thereof disclosed herein is combined with 1,2,3,4 or more additional therapeutic agents.

The 1,2,3,4 or more additional therapeutic agents may be different therapeutic agents selected from one class of therapeutic agents or from different classes of therapeutic agents.

In a particular embodiment of the invention, a compound of formula (1) or (2) as disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with an HIV non-nucleoside reverse transcriptase inhibitor.

In a particular embodiment of the invention, a compound of formula (1) or (2) as disclosed herein, or a stereoisomer, or a pharmaceutically acceptable salt or solvate thereof, is combined with an HIV nucleoside or nucleotide reverse transcriptase inhibitor and an HIV non-nucleoside reverse transcriptase inhibitor.

In another specific embodiment of the invention, a compound of formula (1) or (2), as described herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with an HIV nucleoside or nucleotide reverse transcriptase inhibitor and an HIV protease inhibiting compound.

In another embodiment of the present invention, the compounds of formula (1) or (2) disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, are combined with HIV nucleoside or nucleotide reverse transcriptase inhibitors, HIV non-nucleoside reverse transcriptase inhibitors, and HIV protease inhibiting compounds.

In another embodiment of the present invention, a compound of formula (1) or (2) disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with an HIV nucleoside or nucleotide reverse transcriptase inhibitor, an HIV non-nucleoside reverse transcriptase inhibitor, an HIV capsid assembly inhibitor, and a pharmacokinetic enhancer.

In certain embodiments of the present invention, a compound of formula (1) or (2) disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with at least one HIV nucleoside reverse transcriptase inhibitor, integrase inhibitor and pharmacokinetic enhancer.

In another specific embodiment of the present invention, a compound of formula (1) or (2) disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, is combined with two HIV nucleoside or nucleotide reverse transcriptase inhibitors.

In a specific embodiment of the present invention, a compound of formula (1) or (2) disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with abacavir sulfate, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, elsufavirine, VM-1500A, or GS-CA 1.

In a particular embodiment of the invention, a compound of formula (1) or (2), as described herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with a first additional therapeutic agent and a second additional therapeutic agent, the first additional therapeutic agent is selected from abacavir sulfate, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, elsufavirine, VM-1500A, or GS-CA1, and the second additional therapeutic agent is selected from emtricitabine and lamivudine.

In a particular embodiment of the invention, a compound of formula (1) or (2) as disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with a first additional therapeutic agent selected from tenofovir, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, or tenofovir cyclobutylalafenamide fumarate, elsufavir, VM-1500A or GS-CA1, and a second additional therapeutic agent, emtricitabine.

In a specific embodiment of the present invention, a compound of formula (1) or (2) disclosed herein, or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with 5-30mg tenofovir, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir alafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, elsufavirine, VM-1500A or GS-CA1 and 200mg emtricitabine.

In certain embodiments of the invention disclosed herein, a compound of formula (1) or (2), or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with 5-10mg, 5-15mg, 5-20mg, 5-25mg, 25-30mg, 20-30mg, 15-30mg, or 10-30mg tenofovir disoproxil, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir alafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, elsufavirine, VM-1500A or GS-CA 1in combination with 200mg emtricitabine.

In a particular embodiment of the invention disclosed herein, a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with 10mg tenofovir, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, elsufavirine, VM-1500A or GS-CA1 and 200mg emtricitabine.

In a particular embodiment of the invention disclosed herein, a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, is combined with 25mg tenofovir, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, tenofovir cyclobutylalafenamide hemifumarate, elsufavirine, VM-1500A or GS-CA1 and 200mg emtricitabine.

In the above embodiments, the additional therapeutic agent may be the above anti-HBV and/or anti-HCV agent.

In certain embodiments of the invention disclosed herein, a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, or a pharmaceutical composition is combined with one or more additional therapeutic agents as described above, the composition components being administered simultaneously or sequentially. Sequential administration of the combination may involve two or more administrations.

In certain embodiments of the invention disclosed herein, a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, is combined with one or more additional therapeutic agents in a single dosage form to be administered to a patient simultaneously, e.g., in a solid dosage form for oral administration of fixed dose components.

In certain embodiments of the invention disclosed herein, a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, is administered with one or more additional therapeutic agents.

Co-administration of a compound disclosed herein and one or more additional therapeutic agents generally refers to the simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents to ensure the presence of a therapeutically effective amount of a compound disclosed herein and a therapeutically effective amount of one or more additional therapeutic agents in a patient.

Co-administration involves administering a unit dose of a compound disclosed herein before or after administering a unit dose of one or more additional therapeutic agents, e.g., within seconds, minutes, or hours before or after administering one or more additional therapeutic agents. For example, in certain embodiments, a unit dose of a compound disclosed herein is administered first, followed by administration of a unit dose of one or more additional therapeutic agents within seconds or minutes.

Alternatively, other embodiments involve first administering a unit dose of one or more additional therapeutic agents, followed by administration of a unit dose of a compound disclosed herein within seconds or minutes. In certain embodiments of the invention, a unit dose of a compound disclosed herein is administered first, followed by administration of a unit dose of one or more additional therapeutic agents several hours later (e.g., 1-12 hours later).

In other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of a compound disclosed herein after several hours (e.g., after 1-12 hours).

In another embodiment of the present invention, the compounds of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline or polymorphic form thereof, or a pharmaceutical composition disclosed herein, are proposed for use in HIV therapy (as a method of treatment) in an HIV-infected or HIV-exposed individual.

In another embodiment of the present invention, there is provided a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof or a pharmaceutical composition disclosed herein, for use in the treatment of an HIV infection in a human having or at risk of acquiring a specified infection, the treatment further involving administering to the individual one or more additional therapeutic agents.

In another embodiment of the present invention, the compound of formula (1) or (2) or a stereoisomer thereof, or a novel pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition disclosed herein is proposed for use in the treatment of HIV infection in an HIV-infected or HIV-exposed individual, wherein said treatment further comprises administering to said individual one or more additional therapeutic agents selected from the group consisting of: HIV protease inhibitors, HIV non-nucleoside reverse transcriptase inhibitors, HIV nucleoside or nucleotide reverse transcriptase inhibitors, allosteric HIV integrase inhibitors, HIV capsid assembly inhibitors, and combinations thereof.

In another embodiment of the present invention, the compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition disclosed herein, is proposed for use in the treatment of HIV infection in an HIV-infected or HIV-exposed individual, wherein said treatment further comprises administering to said individual a single dosage form for simultaneous administration (e.g. in the form of a solid dosage form for oral administration) to the patient of a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, or a crystalline form or polymorph thereof, or a pharmaceutical composition disclosed herein and a first additional therapeutic agent selected from abacavir sulfate, a pharmaceutically acceptable salt thereof, a polymorph thereof, or a pharmaceutical composition disclosed herein, and a first additional therapeutic agent selected from abacavir sulfate, a second additional therapeutic agent, a first additional therapeutic agent selected from the group consisting of abacavir sulfate, a second additional therapeutic agent, and a second additional therapeutic agent, Tenofovir, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate, tenofovir alafenamide hemifumarate, tenofovir cyclobutylalafenamide fumarate, elsulfavirine, VM-1500A or GS-CA1, and the second additional therapeutic agent is selected from the group consisting of emtricitabine and lamivudine.

The pharmaceutical compositions of the embodiments disclosed herein may be prepared as follows: a compound of formula (1) or (2) or a stereoisomer thereof, or a pharmaceutically acceptable salt thereof, or a solvate thereof, is combined with suitable pharmaceutically acceptable excipients to produce a medicament in solid, semi-solid, liquid, or gaseous form (such as tablets, capsules, powders, granules, ointments, solutions, nanosuspensions, suppositories, injections, inhalants, gels, microspheres, and aerosols). Typical routes of administration of the pharmaceutical composition include, but are not limited to, oral, topical, transdermal, inhalation, parenteral, sublingual, buccal, rectal, vaginal and intranasal routes.

Pharmaceutical compositions according to embodiments disclosed herein are formulated so as to provide bioavailability of the active ingredient within the composition to be administered to a subject. The composition is administered to a subject or patient in a dosage form containing one or more dosage units, where, for example, a tablet may be a dosage form containing one dosage unit and a container with a compound (in the form of an aerosol) according to embodiments disclosed herein may contain a number of dosage units. The actual methods for preparing the dosage forms are known or will be apparent to those skilled in the art; see, for example, Remington, the Science and Practice of Pharmacy, 20 th edition (Philadelphia College of Pharmacy and Science, 2000). The composition to be administered will in any event comprise a therapeutically effective amount of a compound according to embodiments disclosed herein recommended for the treatment of the disease or disorder of interest according to the description herein.

The pharmaceutical compositions disclosed herein may be prepared by methods well known in the pharmaceutical art. For example, pharmaceutical compositions intended for administration by injection may be prepared by combining the compounds of the embodiments disclosed herein with sterile distilled water to form a solution or nanosuspension. To ensure homogeneity of the solution or nanosuspension, a surfactant may be added. Surfactants are compounds which: which interacts non-covalently with and facilitates dissolution of a compound according to embodiments disclosed herein or homogeneity of a nanosuspension of the compound in an aqueous delivery system.

Subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed individual by administering to said individual a therapeutically effective amount of a compound of formula 1 or 2, or a pharmaceutical composition containing a compound of formula 1 or 2.

Subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed individual by orally administering to said individual a therapeutically effective amount of a compound of formula 1 or 2, or a pharmaceutical composition containing a compound of formula 1 or 2.

The subject of the present invention is a method for the prevention and treatment of HIV infection in an HIV-infected or HIV-exposed individual by injecting said individual with a therapeutically effective amount of a nanosuspension containing a compound of general formula (1) or a compound of general formula (2) or a stereoisomer thereof.

The following figures illustrate the invention:

FIG. 1 simulation Using Pauli technology(3S,11aR) -6-hydroxy-N- [ (2, 6-difluoro-3-pyridinyl) methyl]-3- Methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazole [3,2-a ]]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.19)X-ray diffraction pattern of the sample. Blue line indicates experimental XRD pattern; the red line is the calculated value; and the grey lines are the difference curves. The vertical lines indicate peaks caused by impurities.

Figure 2a stability of compounds of general formula 1.1 in human plasma.

Figure 2b stability of compounds of general formula 1.1 in rat plasma.

Figure 3a stability of compounds of general formula 1.1 in the S9 fraction of human liver.

Figure 3b stability of compounds of general formula 1.1 in rat liver S9 fraction.

Description of the preferred embodiments

The following examples illustrate the invention but are not to be construed as limiting.

Example 1. physicochemical study of the compounds of general formula 1 or 2.

General chemical procedures.All chemicals and solvents were used as received without further purification. The crude reaction mixture was concentrated under reduced pressure by removing the organic solvent on a rotary evaporator.

Nuclear magnetic resonance spectrum (NMR) isRecorded at room temperature (rt) using a Broker DPX-400 spectrometer using tetramethylsilane as an internal standard. Chemical shifts (δ) are given in parts per million (ppm) and the signal is presented as s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet) or brs (broad singlet).

High resolution mass spectrometry spectrum (HRMS) isObtained using an Orbitrap Elite mass spectrometer equipped with a HESI ion source (Thermo, Bremen, Germany).

High Performance Liquid Chromatography (HPLC).The purity of the final compound was determined using HPLC and reached over 98%. HPLC conditions for purity evaluation were as follows: shimadzu HPLC, Xbridge C18,4.6mm X250 mm (3.5 μm); a gradient of 0.1% TFA in 5% acetonitrile/water (a) and 0.1% TFA in acetonitrile (B); the flow rate is 0.5 ml/min; collecting for 20 min; and UV wavelengths 214 and 254 nm. The preparative HPLC system included two sets of Shimadzu LC-8A pumps, a Shimadzu SCL 10Avp controller, and a Shimadzu spdu SPD 10Avp detector. A10 μm, 250mm by 20mm Reprosil-Pur C18-AQ column was used. The mobile phase had a gradient of 0.1% TFA in water (a) and 0.1% TFA in acetonitrile (B). In using cationic electrospray [ M + H ]]+ LC/MS (LC/MS) was performed on a PE Sciex API 165 system and a Shimadzu HPLC system equipped with a Waters Xbridge C183.5 μm column (4.6 mm. times.150 mm).

The diastereoisomers were separated on a chiral HPLC Phenomenex Lux 5u Cellulose-4, AXIA F, 250X 30.00mm (flow: 25 ml/min; UV detector at 215 nm).

X-ray phase analysis.X-ray diffraction patterns were obtained on a Bruker D8 Advance Vario diffractometer equipped with a copper anode and a Ge (III) monochromator (CuK. alpha.) (III)1) And a position sensitive LynxEye detector in a transport setting. The emission angle ranges from 2 to 60 ° 2 θ and the emission step size is 0.02 ° 2 θ. Bruker Topas5 software [ Bruker TOPAS5 User Manual ] -Karlsruhe, Germany Bruker AXS GmbH,2015]And (6) carrying out analysis.

Example 2 general synthetic procedure for fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2 n-pyrido [1,2-a ] pyrazine-7-carboxamide (formula 1 or 2) according to scheme 1.

Heterocyclylmethylamine or its hydrochloride (0.75mmol), diisopropylamine (0.131ml,0.75 mmol; 0.75mmol per hydrochloride) and Pd (PPh)4(29mg,0.025mmol) of the corresponding fused 9-benzyloxy-7-bromo-1, 8-dioxo-1, 3,4, 8-tetrahydro-2-no-pyrido [1,2-a ] of formula 3 or 4]Pyrazine (0.5mmol) in DMSO (2 ml). The reaction mass was stirred under CO at 90 ℃ for 14 h. After the reaction was complete (LC-MS control), the reaction mass was evaporated in vacuo. The residue was dissolved in dichloromethane, washed with water, dried over sodium sulfate, evaporated on a rotary evaporator and column chromatographed on silica gel to give the corresponding fused 9-benzyloxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2-nin-pyrido [1,2-a]Pyrazine-7-carboxamide (formula 6 or 7).

a) The resulting compound of formula 6 or 7 (0.35mmol) was dissolved in a mixture of THF (18ml) and methanol (2ml), then 10% Pd/C (0.04g) was added and the mixture was stirred under a hydrogen atmosphere for 8 h. The reaction mass was passed through celite and the filtrate was evaporated. The residue was treated with ether, the precipitate was filtered off and dried in vacuo to yield the corresponding fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2-n-pyrido [1,2-a ] pyrazine-7-carboxamide (formula 1 or 2).

b) The compound of formula 6 or 7 (0.35mmol) was dissolved in 2ml trifluoroacetic acid and stirred at room temperature for 2 h. The solution was evaporated in vacuoThe residue was dissolved in chloroform and taken up with saturated NaHCO3Washing with Na solution2SO4Dried and evaporated in vacuo. The residue was treated with ether, the precipitate was filtered off and dried in vacuo to give the corresponding fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2-nin-pyrido [1,2-a ] -peptide]Pyrazine-7-carboxamides (formula 1 or 2).

c) The compound of formula 6 or 7 (0.35mmol) was dissolved in 1.5ml of N, N-dimethylacetamide, then 0.148g (3.5mmol) of LiCl was added, and the mixture was stirred at 80 ℃ for 3 h. After the reaction was complete (LC-MS control), the reaction mass was evaporated in vacuo. The product was separated by HPLC to yield the corresponding fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2N-pyrido [1,2-a ] pyrazine-7-carboxamide (formula 1 or 2), including

(3S,11aR) -6-hydroxy-3-methyl-5, 7-dioxo-N- (2-thienylmethyl) -2,3,5,7,11,11 a-hexa-kis Hydrogen [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.1):LC-MS(M+1)=376;1H NMR(DMSO-d6,400MHz)δ11.47(brs,1H),10.33(t,J=4.8Hz,1H),8.49(s,1H),7.40(dd,J1=8.8Hz,J2=4.8Hz,1H),7.03(d,J=2.8Hz,1H),6.96(dd,J1=4.8Hz,J2=3.6Hz,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.90(dd,J1=12.0Hz,J2=4.0Hz,1H),4.70(d,J=5.6Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=7.2Hz,1H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (5-methyl-2-furyl) methyl]-5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.3):LC-MS(ESI)374(M+H)+1H NMR(DMSO-d6,400MHz)δ11.46(brs,1H),10.20(t,J=5.2Hz,1H),8.46(s,1H),6.15(d,J=2.4Hz,1H),5.99(d,J=2.4Hz,1H),5.39(dd,J1=9.6Hz,J2=4.0Hz,1H),4.89(dd,J1=12.0Hz,J2=4.0Hz,1H),4.46(d,J=5.2Hz,2H),4.39(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.00(t,J=11.0Hz,1H),3.66(dd,J1=8.4Hz,J2=7.2Hz,1H),2.23(s,3H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-N- [ (3, 5-dimethyl-NEN-pyrazol-4-yl) methyl]-3-methyl-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.4):LC-MS(ESI)388(M+H)+1H NMR(DMSO-d6,400MHz)δ12.04(brs,1H),11.42(brs,1H),9.92(t,J=4.8Hz,1H),8.46(s,1H),5.38(dd,J1=10.0Hz,J2=4.0Hz,1H),4.88(dd,J1=12.0Hz,J2=4.0Hz,1H),4.38(dd,J1=8.0Hz,J2=7.2Hz,1H),4.29(m,1H),4.26(d,J=5.2Hz,2H),4.00(t,J=11.2Hz,1H),3.66(dd,J1=8.0Hz,J2=7.2Hz,1H),1.33(d,J=6.4Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (1,3, 5-trimethyl-1N-pyrazol-4-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.5):LC-MS(ESI)402(M+H)+1H NMR(DMSO-d6,400MHz)δ11.40(brs,1H),9.92(t,J=4.8Hz,1H),8.46(s,1H),5.38(dd,J1=9.6Hz,J2=3.6Hz,1H),4.88(dd,J1=12.0Hz,J2=3.6Hz,1H),4.39(t,J=7.4Hz,1H),4.29(m,1H),4.25(d,J=4.8Hz,2H),4.00(t,J=11.0Hz,1H),3.66(t,J=7.4Hz,1H),3.32(s,3H),2.19(s,3H),2.08(s,3H),1.33(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-N- [ (1-ethyl-3, 5-dimethyl-1N-pyrazol-4-yl) methyl]3-methyl-5, 7- Dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.6):LC-MS(ESI)416(M+H)+1H NMR(DMSO-d6,400MHz)δ11.40(brs,1H),9.92(t,J=5.2Hz,1H),8.47(s,1H),5.38(dd,J1=10.0Hz,J2=4.0Hz,1H),4.88(dd,J1=12.0Hz,J2=4.0Hz,1H),4.39(dd,J1=8.4Hz,J2=6.8Hz,1H),4.28(m,1H),4.25(d,J=5.6Hz,2H),4.00(t,J=11.2Hz,1H),3.94(q,J=7.2Hz,2H),3.66(dd,J1=8.4Hz,J2=6.4Hz,1H),2.20(s,3H),2.09(s,3H),1.33(d,J=6.0Hz,3H),1.24(t,J=7.2Hz,3H);

(3S,11aR) -6-hydroxy-N- [ (4, 5-dichloro-1-methyl-1N-pyrazol-3-yl) methyl]3-methyl-5, 7-bis Oxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.7): LC-MS(ESI)443(M+H)+1H NMR(DMSO-d6,400MHz)δ11.44(brs,1H),10.28(t,J=5.6Hz,1H),8.46(s,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.89(dd,J1=12.0Hz,J2=4.0Hz,1H),4.50(d,J=5.6Hz,2H),4.39(dd,J1=8.4Hz,J2=6.8Hz,1H),4.29(sxt,J=6.4Hz,1H),4.01(t,J=11.2Hz,1H),3.80(s,3H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- (thiazol-2-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.8):LC-MS(ESI)377(M+H)+1H NMR(DMSO-d6,400MHz)δ11.50(brs,1H),10.58(t,J=6.0Hz,1H),8.49(s,1H),7.74(d,J=3.2Hz,1H),7.62(d,J=3.2Hz,1H),5.40(dd,J1=10.0Hz,J2=4.0Hz,1H),4.90(dd,J1=12.0Hz,J2=4.0Hz,1H),4.84(d,J=6.0Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.30(m,1H),4.01(dd,J1=12.0Hz,J2=10.0Hz,1H),3.67(dd,J1=8.4Hz,J2=6.4Hz,1H),1.35(d,J=6.4Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (4-methyl-1, 2, 5-oxadiazol-3-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.12): LC-MS(ESI)376(M+H)+1H NMR(DMSO-d6,400MHz)δ11.51(brs,1H),10.45(t,J=5.6Hz,1H),8.47(s,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.89(dd,J1=12.0Hz,J2=4.0Hz,1H),4.74(d,J=5.6Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.00(t,J=11.0Hz,1H),3.67(dd,J1=8.0Hz,J2=7.2Hz,1H),2.37(s,3H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (4-methyl-4N-1, 2, 4-triazol-3-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.14): LC-MS(ESI)375(M+H)+1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.41(t,J=5.2Hz,1H),8.48(s,1H),8.40(s,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.89(dd,J1=11.6Hz,J2=3.6Hz,1H),4.69(d,J=5.2Hz,2H),4.39(t,J=7.6Hz,1H),4.29(m,1H),4.01(t,J=11.2Hz,1H),3.66(m,1H),3.65(s,3H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- (pyridin-2-ylmethyl) -5, 7-dioxo-2, 3,5,7,11, 11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.15):LC-MS(ESI)371(M+H)+1H NMR(DMSO-d6,400MHz)δ11.44(brs,1H),10.52(t,J=5.6Hz,1H),8.53(d,J=4.0Hz,1H),8.48(s,1H),7.76(t,J=7.6Hz,1H),7.33(d,J=8.0Hz,1H),7.28(m,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.89(dd,J1=12.0Hz,J2=3.6Hz,1H),4.64(d,J=5.6Hz,2H),4.40(t,J=7.6Hz,1H),4.30(m,1H),4.01(t,J=11.2Hz,1H),3.66(dd,J1=8.0Hz,J2=6.8Hz,1H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (2-methylpyrimidin-4-yl) methyl]-5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.26):LC-MS(M+1)=386;1H NMR(DMSO-d6,400MHz)δ11.50(brs,1H),10.50(t,J=6.0Hz,1H),8.61(d,J=5.0Hz,1H),8.46(s,1H),7.16(d,J=5.0Hz,1H),5.40(dd,J1=10.0Hz,J2=3.2Hz,1H),4.89(dd,J1=12.0Hz,J2=3.2Hz,1H),4.59(d,J=6.0Hz,2H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.30(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=6.4Hz,1H),2.60(s,3H),1.35(d,J=6.0Hz,3H);

(3S,11aR) -N- (1-benzothien-5-ylmethyl) -6-hydroxy-3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.31) LC-MS (ESI)426(M + H)+1H NMR(DMSO-d6,400MHz)δ11.47(brs,1H),10.38(t,J=6.0Hz,1H),8.50(s,1H),7.96(d,J=8.4Hz,1H),7.80(s,1H),7.76(d,J=9.4Hz,1H),7.43(d,J=9.4Hz,1H),7.33(dd,J1=8.4Hz,J2=0.6Hz,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.90(dd,J1=12.0Hz,J2=4.0Hz,1H),4.66(d,J=5.6Hz,2H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.30(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=6.8Hz,1H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -N- (1-benzofuran-2-ylmethyl) -6-hydroxy-3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.32), LC-MS (ESI)410(M + H)+1H NMR(DMSO-d6,400MHz)δ11.50(brs,1H),10.41(t,J=5.6Hz,1H),8.50(s,1H),7.59(d,J=7.6Hz,1H),7.54(d,J=7.6Hz,1H),7.27(t,J=7.6Hz,1H),7.22(t,J=7.2Hz,1H),6.75(s,1H),5.40(dd,J1=10.0Hz,J2=3.6Hz,1H),4.91(dd,J1=12.0Hz,J2=3.6Hz,1H),4.72(d,J=5.6Hz,2H),4.40(t,J=7.6Hz,1H),4.28(sxt,J=6.0Hz,1H),4.02(t,J=11.0Hz,1H),3.67(t,J=7.6Hz,1H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (1-methyl-1N-indol-5-yl) methyl]-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.33):LC-MS(ESI)423(M+H)+1H NMR(DMSO-d6,400MHz)δ11.43(brs,1H),10.27(t,J=6.0Hz,1H),8.50(s,1H),7.48(s,1H),7.39(d,J=8.8Hz,1H),7.30(d,J=2.8Hz,1H),7.12(dd,J1=8.8Hz,J2=0.8Hz,1H),6.37(dd,J1=2.8Hz,J2=0.4Hz,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.90(dd,J1=12.0Hz,J2=4.0Hz,1H),4.59(d,J=5.6Hz,2H),4.39(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.01(t,J=11.2Hz,1H),3.77(s,3H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -N- (1, 3-benzodioxol-5-ylmethyl) -6-hydroxy-3-methyl-5, 7-dioxo- 2,3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.34):LC-MS(ESI)414(M+H)+1H NMR(DMSO-d6,400MHz)δ11.45(brs,1H),10.24(t,J=5.8Hz,1H),8.48(s,1H),6.86(m,2H),6.79(m,1H),5.98(s,2H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.89(dd,J1=12.4Hz,J2=4.0Hz,1H),4.43(d,J=6.0Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=6.0Hz,1H),1.34(d,J=6.4Hz,3H)。

(3S,11aR) -N- [ (6-bromo-1, 3-benzodioxol-5-yl) methyl]-6-hydroxy-3-methyl-5, 7- Dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.35):LC-MS(ESI)493(M+H)+1H NMR(DMSO-d6,400MHz)δ11.44(brs,1H),10.30(m,1H),8.46(d,J=2.4Hz,1H),7.23(d,J=2.4Hz,1H),6.93(d,J=2.4Hz,1H),6.05(d,J=2.4Hz,2H),5.38(m,1H),4.88(m,1H),4.46(m,2H),4.39(m,1H),4.29(m,1H),4.00(m,1H),3.66(m,1H),1.34(dd,J1=6.4Hz,J2=2.8Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (7-methyl-2, 3-dihydro-1, 4-benzodioxin-6- Radical) methyl]-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8- Formamide (1.1.36):LC-MS(ESI)434(M+H)+1H NMR(DMSO-d6,400MHz)δ11.43(brs,1H),10.14(t,J=5.6Hz,1H),8.47(s,1H),6.73(s,1H),6.68(s,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.89(dd,J1=12.0Hz,J2=3.6Hz,1H),4.39(m,3H),4.29(m,1H),4.18(s,4H),4.00(t,J=11.2Hz,1H),3.66(dd,J1=8.0Hz,J2=7.2Hz,1H),2.17(s,3H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -N- (1, 3-benzothiazol-2-ylmethyl) -6-hydroxy-3-methyl-5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.37):LC-MS(ESI)427(M+H)+1H NMR(DMSO-d6,400MHz)δ11.54(brs,1H),10.69(t,J=5.6Hz,1H),8.50(s,1H),8.04(d,J=8.0Hz,1H),7.95(d,J=8.0Hz,1H),7.50(t,J=7.2Hz,1H),7.41(t,J=7.2Hz,1H),5.40(dd,J1=9.6Hz,J2=3.6Hz,1H),4.97(d,J=5.6Hz,2H),4.90(dd,J1=12.0Hz,J2=3.6Hz,1H),4.40(t,J=7.6Hz,1H),4.30(m,1H),4.02(t,J=11.2Hz,1H),3.67(t,J=7.6Hz,1H),1.35(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (5-chloro-1, 3-benzoxazol-2-yl) methyl]-5, 7-dioxo- 2,3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.38):LC-MS(ESI)445(M+H)+1H NMR(DMSO-d6,400MHz)δ11.52(brs,1H),10.54(t,J=5.6Hz,1H),8.47(s,1H),7.83(d,J=2.0Hz,1H),7.75(d,J=8.8Hz,1H),7.42(dd,J1=8.8Hz,J2=2.0Hz,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.88(m,3H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.30(m,1H),4.01(dd,J1=11.6Hz,J2=6.4Hz,1H),3.67(dd,J1=8.4Hz,J2=6.4Hz,1H),1.35(d,J=6.4Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (6-chloro-1, 3-benzoxazol-2-yl) methyl]-5, 7-dioxo- 2,3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1, 2]-d]Pyrazine-8-carboxamide (1.1.39):LC-MS(ESI)445(M+H)+1H NMR(DMSO-d6,400MHz)δ11.52(brs,1H),10.54(t,J=5.4Hz,1H),8.47(s,1H),7.92(d,J=1.2Hz,1H),7.72(d,J=8.4Hz,1H),7.41(dd,J1=8.4Hz,J2=1.6Hz,1H),5.40(dd,J1=10.0Hz,J2=4.0Hz,1H),4.89(m,1H),4.87(d,J=6.0Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.30(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.0Hz,J2=6.8Hz,1H),1.35(d,J=6.4Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (5-methyl-1N-benzimidazol-2-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.40): LC-MS(ESI)424(M+H)+1H NMR(DMSO-d6,400MHz)δ12.19(s,1H),11.47(brs,1H),10.48(s,1H),8.47(s,1H),7.32(m,2H),6.96(m,1H),5.40(m,1H),4.90(m,1H),4.72(m,2H),4.40(m,1H),4.30(m,1H),4.02(m,1H),3.67(m,1H),2.39(s,3H),1.35(d,J=4.4Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (5-fluoro-1N-benzimidazol-2-yl) methyl]-5, 7-dioxo- 2,3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.41):LC-MS(ESI)428(M+H)+1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.51(brs,1H),8.47(s,1H),7.52(m,1H),7.33(d,J=8.4Hz,1H),7.03(t,J=8.4Hz,1H),5.40(m,1H),4.90(m,1H),4.77(brs,2H),4.40(m,1H),4.30(m,1H),4.02(t,J=10.8Hz,1H),3.67(m,1H),1.35(d,J=5.6Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (1-methyl-6-chlorobenzimidazol-2-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.43): LC-MS(ESI)458(M+H)+1H NMR(DMSO-d6,400MHz)δ11.47(brs,1H),10.58(t,J=5.2Hz,1H),8.50(s,1H),7.65(d,J=1.2Hz,1H),7.58(d,J=8.4Hz,1H),7.27(dd,J1=8.4Hz,J2=1.2Hz,1H),5.40(dd,J1=9.6Hz,J2=3.2Hz,1H),4.90(m,1H),4.85(d,J=5.2Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.30(m,1H),4.02(t,J=11.0Hz,1H),3.81(s,3H),3.67(dd,J1=8.4Hz,J2=7.2Hz,1H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- { [ 1-isopropyl-1H-benzimidazol-2-yl]Methyl } -5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.44): LC-MS(ESI)452(M+H)+1H NMR(DMSO-d6,400MHz)δ11.47(brs,1H),10.57(t,J=5.2Hz,1H),8.52(s,1H),7.70(d,J=8.0Hz,1H),7.61(d,J=8.0Hz,1H),7.18(m,2H),5.40(dd,J1=10.0Hz,J2=4.0Hz,1H),4.91(dd,J1=12.0Hz,J2=4.0Hz,1H),4.86(d,J=5.2Hz,2H),4.83(m,1H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.29(m,1H),4.02(dd,J1=12.0Hz,J2=10.0Hz,1H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),1.56(d,J=6.8Hz,6H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-N- [ (2-oxo-2, 3-dihydro-1N-benzimidazol-5-yl) methyl]-3-methyl- 5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.46):LC-MS(ESI)426(M+H)+1H NMR(DMSO-d6,400MHz)δ11.46(brs,1H),10.56(s,2H),10.26(t,J=5.6Hz,1H),8.48(s,1H),6.88(m,3H),5.39(m,1H),4.90(m,1H),4.49(d,J=5.6Hz,2H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.29(m,1H),4.01(t,J=11.0Hz,1H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -N- (2,1, 3-benzoxadiazol-5-ylmethyl) -6-hydroxy-3-methyl-5, 7-dioxo-2, 3, 5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.47):LC-MS(ESI)412(M+H)+1H NMR(DMSO-d6,400MHz)δ11.51(brs,1H),10.46(t,J=6.0Hz,1H),8.49(s,1H),8.03(d,J=9.4Hz,1H),7.78(s,1H),7.57(d,J=9.4Hz,1H),5.40(dd,J1=9.8Hz,J2=3.4Hz,1H),4.90(dd,J1=12.0Hz,J2=3.4Hz,1H),4.68(d,J=5.6Hz,2H),4.40(t,J=7.6Hz,1H),4.30(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=6.8Hz,1H),1.35(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- (quinolin-2-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]-oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.49):LC-MS(ESI)421(M+H)+1H NMR(DMSO-d6,400MHz)δ11.44(brs,1H),10.30(t,J=5.6Hz,1H),8.50(s,1H),7.45(s,1H),7.42(d,J=8.0Hz,1H),7.16(dd,J1=8.0Hz,J2=0.8Hz,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.90(m,1H),4.61(d,J=5.6Hz,2H),4.39(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(sxt,J=6.4Hz,1H),4.01(t,J=11.2Hz,1H),3.70(s,3H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),2.50(s,3H),1.34(d,J=6.4Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- (quinolin-3-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.50):LC-MS(ESI)421(M+H)+1H NMR(DMSO-d6,400MHz)δ11.50(brs,1H),10.47(t,J=6.0Hz,1H),8.91(d,J=2.0Hz,1H),8.50(s,1H),8.24(s,1H),8.02(d,J=8.0Hz,1H),7.96(d,J=7.6Hz,1H),7.74(dt,J1=8.0Hz,J2=1.2Hz,1H),7.61(m,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.89(dd,J1=12.0Hz,J2=4.0Hz,1H),4.76(d,J=6.0Hz,2H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.30(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=6.8Hz,1H),1.35(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- (2-methylquinolin-4-ylmethyl) -5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyridine compoundAnd [1,2-d ]]Pyrazine-8-carboxamide (1.1.51):LC-MS(ESI)435(M+H)+1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.47(t,J=5.6Hz,1H),8.52(s,1H),8.14(d,J=8.4Hz,1H),7.95(d,J=8.4Hz,1H),7.73(t,J=7.6Hz,1H),7.57(t,J=7.2Hz,1H),7.28(s,1H),5.40(dd,J1=10.0Hz,J2=4.0Hz,1H),5.03(d,J=5.6Hz,2H),4.91(dd,J1=12.0Hz,J2=4.0Hz,1H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.30(m,1H),4.02(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=6.8Hz,1H),2.61(s,3H),1.34(d,J=6.4Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- (quinolin-5-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.52):LC-MS(ESI)421(M+H)+1H NMR(DMSO-d6,400MHz)δ11.44(s,1H),10.42(t,J=6.0Hz,1H),8.92(dd,J1=8.0Hz,J2=1.2Hz,1H),8.61(d,J=8.0Hz,1H),8.52(s,1H),7.97(d,J=8.4Hz,1H),7.73(t,J=8.2Hz,1H),7.58(m,2H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),5.03(d,J=6.0Hz,2H),4.91(dd,J1=12.0Hz,J2=4.0Hz,1H),4.39(dd,J1=8.4Hz,J2=6.8Hz,1H),4.29(sxt,J=6.4Hz,1H),4.01(t,J=11.2Hz,1H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),1.33(d,J=6

(3S,11aR) -6-hydroxy-3-methyl-N- (quinolin-8-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.54):LC-MS(ESI)421(M+H)+1H NMR(DMSO-d6,400MHz)δ11.41(brs,1H),10.49(t,J=5.6Hz,1H),8.99(dd,J1=4.0Hz,J2=1.6Hz,1H),8.47(s,1H),8.40(dd,J1=8.0Hz,J2=1.2Hz,1H),7.91(d,J=8.0Hz,1H),7.67(d,J=6.8Hz,1H),7.58(m,2H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),5.13(d,J=5.6Hz,2H),4.88(dd,J1=12.0Hz,J2=4.0Hz,1H),4.39(dd,J1=8.0Hz,J2=7.6Hz,1H),4.29(m,1H),4.00(t,J=11.2Hz,1H),3.66(dd,J1=8.0Hz,J2=6.8Hz,1H),1.34(d,J=6.0Hz,3H).

(3S,11aR) -6-hydroxy-3-methyl-N- (5,6,7, 8-tetrahydro- [1,2, 4)]Triazolo [4,3-a]Pyridin-3-yl Methyl) -5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a]Pyrido [1,2-d]pyrazine-8-A Amide (1.1.61):LC-MS(ESI)415(M+H)+1H NMR(DMSO-d6,400MHz)δ11.48(brs,1H),10.38(t,J=5.6Hz,1H),8.48(s,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.89(dd,J1=12.0Hz,J2=3.6Hz,1H),4.64(d,J=5.6Hz,2H),4.39(dd,J1=8.0Hz,J2=7.2Hz,1H),4.29(sxt,J=6.0Hz,1H),4.01(t,J=11.2Hz,1H),3.93(t,J=6.0Hz,2H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),2.79(t,J=6.4Hz,2H),1.88(m,2H),1.79(m,2H),1.34(d,J=6.0Hz,3H).

(3S,11aR) -6-hydroxy-N- (1,4,5,6,7, 8-hexahydrocyclohepta [ c)]Pyrazol-3-ylmethyl) -3-carboxylic acid methyl ester Yl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.62):LC-MS(ESI)428(M+H)+1H NMR(DMSO-d6,400MHz)δ12.13(brs,1H),11.43(brs,1H),10.07(t,J=5.2Hz,1H),8.47(s,1H),5.40(dd,J1=10.0Hz,J2=3.6Hz,1H),4.89(dd,J1=12.0Hz,J2=3.6Hz,1H),4.39(m,3H),4.29(m,1H),4.00(t,J=11.2Hz,1H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),2.63(m,2H),2.45(m,2H),1.75(m,2H),1.55(m,4H),1.34(d,J=6.0Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-5, 7-dioxo-N- (5,6,7, 8-tetrahydro-4H-cyclohepta-e [ d ]] [1,3]Thiazol-2-ylmethyl) -2,3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8- Formamide (1.1.63):LC-MS(ESI)445(M+H)+1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.47(m,1H),8.48(s,1H),5.39(m,1H),4.90(m,1H),4.67(m,2H),4.40(m,1H),4.30(m,1H),4.01(m,1H),3.67(m,1H),2.82(m,2H),2.73(m,2H),1.78(m,2H),1.60(m,4H),1.34(dd,J1=6.4Hz,J2=2.8Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- [ (1, 2-dimethyl-1N-benzimidazol-5-yl) methyl]-5, 7-bis Oxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.64): LC-MS(ESI)438(M+H)+1H NMR(DMSO-d6,400MHz)δ11.44(brs,1H),10.30(t,J=5.6Hz,1H),8.50(s,1H),7.45(s,1H),7.42(d,J=8.0Hz,1H),7.16(dd,J1=8.0Hz,J2=0.8Hz,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.90(m,1H),4.61(d,J=5.6Hz,2H),4.39(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(sxt,J=6.4Hz,1H),4.01(t,J=11.2Hz,1H),3.70(s,3H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),2.50(s,3H),1.34(d,J=6.4Hz,3H)。

(3S,11aR) -6-hydroxy-3-methyl-N- (quinolin-4-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.65):LC-MS(ESI)421(M+H)+1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.50(t,J=6.0Hz,1H),8.85(d,J=4.0Hz,1H),8.52(s,1H),8.21(d,J=8.8Hz,1H),8.06(d,J=8.0Hz,1H),7.79(t,J=6.8Hz,1H),7.66(t,J=7.6Hz,1H),7.39(d,J=4.0Hz,1H),5.40(dd,J1=10.0Hz,J2=3.6Hz,1H),5.08(d,J=6.0Hz,2H),4.91(dd,J1=12.0Hz,J2=3.6Hz,1H),4.40(dd,J1=8.0Hz,J2=7.2Hz,1H),4.29(m,1H),4.02(t,J=11.2Hz,1H),3.67(dd,J1=8.0Hz,J2=6.8Hz,1H),1.33(d,J=6.0Hz,3H)。

(3aS,6R,13aS) -9-hydroxy-N- [ (2, 6-difluoropyridin-3-yl) methyl]-6-methyl-8, 10-dioxo-1, 2,3,5,6,8,10,13 a-octahydrocyclopenta [ b ] s][1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-11-carboxamides (2.1.1): LC-MS (ESI)447(M + H)+1H NMR(DMSO-d6,400MHz)δ12.28(s,1H),10.39(t,J=6.0Hz,1H),8.41(s,1H),8.08(q,J=8.4Hz,1H),7.14(dd,J1=8.4Hz,J2=2.0Hz,1H),4.55(m,2H),4.36(m,3H),3.76(dd,J1=8.0Hz,J2=6.0Hz,1H),2.33(m,1H),2.14(m,1H),1.77(m,3H),1.38(d,J=6.0Hz,3H)。

(3aS,6R,13aS) -9-hydroxy-N- [ (3, 5-difluoropyridin-4-yl) methyl]-6-methyl-8, 10-dioxo- 1,2,3,5,6,8,10,13 a-octahydrocyclopenta [ b ] s][1,3]Oxazolo [3,2-a]Pyrido [1,2-d]pyrazine-11-A Amide (2.1.2):LC-MS(ESI)447(M+H)+1H NMR(DMSO-d6,400MHz)δ12.28(s,1H),10.48(t,J=6.0Hz,1H),8.50(s,2H),8.38(s,1H),4.66(m,2H),4.34(m,3H),3.75(dd,J1=8.0Hz,J2=6.0Hz,1H),2.30(m,1H),2.12(m,1H),1.76(m,3H),1.36(d,J=6.0Hz,3H)。

(3aS,6R,13aS) -9-hydroxy-6-methyl-N- [ (2,4, 6-trifluoropyridin-3-yl) methyl]-8, 10-dioxo Substituted-1, 2,3,5,6,8,10,13 a-octahydrocyclopenta [ b ] s][1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine- 11-carboxamide (2.1.3):LC-MS(ESI)465(M+H)+1H NMR(DMSO-d6,400MHz)δ12.28(s,1H),10.42(t,J=5.6Hz,1H),8.38(s,1H),7.34(d,J=8.0Hz,1H),4.54(m,2H),4.35(m,3H),3.75(m,1H),2.31(m,1H),2.12(m,1H),1.76(m,3H),1.36(d,J=6.0Hz,3H)。

(3aS,6S,13aS) -9-hydroxy-N- [ (2, 6-difluoropyridin-3-yl) methyl]-6-methyl-8, 10-dioxo- 1,2,3,5,6,8,10,13 a-octahydrocyclopenta [ b ] s][1,3]Oxazolo [3,2-a]Pyrido [1,2-d]pyrazine-11-A Amide (2.2.1):LC-MS(ESI)447(M+H)+1H NMR(DMSO-d6,400MHz)δ12.29(s,1H),10.40(t,J=5.8Hz,1H),8.41(s,1H),8.08(q,J=8.4Hz,1H),7.15(dd,J1=8.4Hz,J2=2.0Hz,1H),4.55(m,2H),4.35(m,3H),3.75(dd,J1=8.0Hz,J2=6.4Hz,1H),2.33(m,1H),2.13(m,1H),1.76(m,3H),1.37(d,J=6.4Hz,3H)。

(3aS,6S,13aS) -9-hydroxy-N- [ (3, 5-difluoropyridin-4-yl) methyl]-6-methyl-8, 10-dioxo- 1,2,3,5,6,8,10,13 a-octahydrocyclopenta [ b ] s][1,3]Oxazolo [3,2-a]Pyrido [1,2-d]pyrazine-11-A Amide (2.2.2):LC-MS(ESI)447(M+H)+1H NMR(DMSO-d6,400MHz)δ12.46(s,1H),10.47(t,J=5.8Hz,1H),8.49(s,2H),8.42(s,1H),5.43(dd,J1=9.2Hz,J2=4.0Hz,1H),5.09(brs,1H),4.66(m,3H),4.60(brs,1H),4.01(dd,J1=12.4Hz,J2=9.6Hz,1H),1.94(brs,4H),1.84(d,J=12.0Hz,1H),1.57(m,1H)。

(3aS,6S,13aS) -9-hydroxy-6-methyl-N- [ (2,4, 6-trifluoropyridin-3-yl) methyl]-8, 10-dioxo Substituted-1, 2,3,5,6,8,10,13 a-octahydrocyclopenta [ b ] s][1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine- 11-carboxamide (2.2.3):LC-MS(ESI)46е(M+H)+1H NMR(DMSO-d6,400MHz)δ12.28(s,1H),10.42(t,J=5.8Hz,1H),8.38(s,1H),7.34(d,J=8.4Hz,1H),4.54(m,2H),4.34(m,3H),3.75(dd,J1=8.0Hz,J2=6.0Hz,1H),2.30(m,1H),2.12(m,1H),1.76(m,3H),1.36(d,J=6.0Hz,3H)。

example 3 general synthetic procedure for fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2N-pyrido [1,2-a ] pyrazine-7-carboxamide of general formula 1.1.

Preparation according to scheme 3(3S,11aR) -6-hydroxy-N- [ (2, 6-difluoropyridin-3-yl) methyl]-3-methyl-5, 7- Dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.19)

Scheme 3. synthesis of compound 1.1.19.

The reaction product of ((3S,11aR) -6- (benzyloxy) -8-bromo-3-methyl-2, 3,11,11 a-tetrahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-5, 7-dione (3.1) (3g,7.4mmol) was dissolved in a mixture of THF (250ml) and methanol (150 ml). The reaction mixture was transferred to an autoclave and triethylamine (1.3ml,8.9mmol) and Pd (dppf) Cl were added2(0.05g,0.07 mmol). Pump and method of operating the sameCO to 15atm was added and the reaction mixture was heated to 100 ℃ for 3 days. After the reaction was complete (LC-MS control), the reaction mass was filtered through celite, evaporated in vacuo, dissolved in dichloromethane, washed with water, dried over sodium sulfate and evaporated on a rotary evaporator. The residue was subjected to column chromatography on silica gel with dichloromethane-methanol (60:1) as eluent to give 1.8g (63%) of (3S,11aR) -6- (benzyloxy) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1]-oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxylic acid methyl ester (3.2):1H NMR(DMSO-d6,400MHz)δ8.39(s,1H),7.53(m,2H),7.34(m,3H),5.32(dd,J1=10.0Hz,J2=3.6Hz,1H),5.04,5.17(ABq,JAB=10.4Hz,2H),4.67(dd,J1=12.0Hz,J2=3.2Hz,1H),4.29(m,2H),3.94(dd,J1=12.0Hz,J2=10.0Hz,1H),3.63(dd,J1=8.0Hz,J2=6.4Hz,1H),1.27(d,J=6.0Hz)。

reacting LiOH with H2A solution of O (0.33g,7.8mmol) in 5ml of water is added to a solution of dimethyl ether 3.2(1.5g,3.9mmol) in 15ml of dioxane. The mixture was then stirred at 50 ℃ for 3 h. The dioxane was evaporated on a rotary evaporator. The residue was diluted with water and acidified to pH 3 with 10% sulfuric acid. The precipitate was filtered off, washed with water and dried in vacuo to give 1g (69%) of (3S,11aR) -6- (benzyloxy) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxylic acid (3.3):1H NMR(DMSO-d6,400MHz)δ15.45(brs,1H),8.75(s,1H),7.54(m,2H),7.36(m,3H),5.38(dd,J1=10.0Hz,J2=3.2Hz,1H),5.15,5.25(ABq,JAB=10.4Hz,2H),4.87(dd,J1=12.0Hz,J2=3.2Hz,1H),4.32(m,2H),4.13(t,J=11.0Hz,1H),3.66(dd,J1=7.6Hz,J2=6.4Hz,1H),1.28(d,J=6.0Hz)。

triethylamine (1.2ml,8.1mmol) was added to a solution of the acid 3.3(1g,2.7mmol) in 20ml of dichloromethane. The reaction mixture was stirred at room temperature for 15min and TBTU (0.82g,3.3mmol) was added. The mixture was stirred for an additional 15min, then [ (2, 6-difluoropyridin-3-yl) methyl ] was added]Amine (0.47g,3.3mmol), andthe mixture was stirred at room temperature for 14 h. The reaction mass was washed with 10-% aqueous potassium carbonate solution and water. The organic layer was dried over sodium sulfate and evaporated on a rotary evaporator. Diethyl ether was added to the residue and the resulting precipitate was filtered off and dried in vacuo. The product was used further without additional purification. Yield 0.94g (70%) of (3S,11aR) -6- (benzyloxy) -3-methyl-5, 7-dioxo-N- [ (2, 6-difluoropyridin-3-yl) methyl]-2,3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (3.4.19):1H NMR(DMSO-d6,400MHz)δ10.45(t,J=5.9Hz,1H),8.56(s,1H),8.13-8.00(m,1H),7.57-7.52(m,2H),7.40-7.29(m,3H),7.15(dd,J1=8.1Hz,J2=2.3Hz,1H),5.35(dd,J1=10.0Hz,J2=3.7Hz,1H),5.21(d,J=10.6Hz,1H),5.07(d,J=10.6Hz,1H),4.79(dd,J1=12.2Hz,J2=3.7Hz,1H),4.56(d,J=5.9Hz,2H),4.38-4.22(m,2H),4.08-3.98(m,1H),3.64(dd,J1=8.1Hz,J2=6.4Hz,1H),1.27(d,J=6.1Hz,3H)。

compound 3.4.19(0.9g,1.8mmol) was dissolved in a mixture of THF (18ml) and methanol (2ml), then 10% Pd/C (0.09g) was added and the reaction mixture was stirred under hydrogen atmosphere for 8 h. The reaction mass was passed through celite and the filtrate was evaporated. The residue was treated with ether, the precipitate was filtered off and dried in vacuo to give 0.6g (81%) of: (b)3S, 11aR) -6-hydroxy-N- [ (2, 6-difluoro-pyridin-3-yl) methyl]-3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexa-position Hydrogen [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.19):LC-MS(ESI)07(M+H)+1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.36(t,J=5.8Hz,1H),8.45(s,1H),8.06(dd,J1=17.2Hz,J2=8.4Hz,1H),7.14(dd,J1=8.0Hz,J2=2.0Hz,1H),5.38(dd,J1=10.0Hz,J2=3.6Hz,1H),4.88(dd,J1=12.0Hz,J2=3.6Hz,1H),4.55(d,J=5.6Hz,2H),4.39(dd,J1=8.0Hz,J2=6.8Hz,1H),4.29(m,1H),4.00(t,J=11.0Hz,1H),3.67(dd,J1=8.5Hz,J26.8Hz,1H),1.34(d, J ═ 6.4Hz, 3H); XRD: a simulation of the diffraction pattern of the inhibitor 1.1.19 sample using Pauli technique is shown in fig. 1. Obtained by recrystallization from methanolThe sample was a single phase. Unit cell parameters:α is 65.469(6) °, β is 106.211(6) °, and γ is 131.825(6) °. Unit cell volume ofThe system extinction and the unit cell volume are consistent with space group P-1.

(3S,11aR) -8- ({ [ (2, 6-difluoropyridin-3-yl) methyl]Amino } carbonyl) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a ] s]Pyrido [1,2-d]Pyrazine-6-ol sodium (1.1.20) NaOH (0.05g,1.2mmol) was added to (3S,11aR) -6-hydroxy-N- [ (2, 4-difluoro-3-pyridinyl) methyl at 75 deg.C]-3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazole [3,2-a ]]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.19) (0.05g,1.2mmol) was suspended in 80ml ethanol and 20ml water. The reaction mixture was stirred at rt for 14 h. The resulting precipitate was filtered off, washed with anhydrous ethanol and dried in vacuo to give 0.42g (80%) of (3S,11aR) -8- ({ [ (2, 6-difluoropyridin-3-yl) methyl]Amino } carbonyl) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a ] s]Pyrido [1,2-d]Pyrazine-6-sodium alkoxide (1.1.20):1H NMR(DMSO-d6,400MHz)δ10.76(t,J=5.6Hz,1H),8.00(dd,J1=17.2Hz,J2=8.0Hz,1H),7.90(s,1H),7.11(dd,J1=8.0Hz,J2=1.6Hz,1H),5.21(dd,J1=10.0Hz,J2=4.0Hz,1H),4.59(dd,J1=12.4Hz,J2=3.6Hz,1H),4.52(t,J=5.6Hz,2H),4.25(m,2H),3.74(t,J=10.8Hz,1H),3.60(m,1H),1.27(d,J=5.2Hz,3H)。

the following compounds were obtained in a similar manner:

(3S,11aR) -6-hydroxy-3-methyl-5, 7-dioxo-N- (2-thienylmethyl) -2,3,5,7,11,11 a-hexa-kis Hydrogen [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.1):LC-MS(ESI)376(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.47(brs,1H),10.33(t,J=4.8Hz,1H),8.49(s,1H),7.40(dd,J1=8.8Hz,J2=4.8Hz,1H),7.03(d,J=2.8Hz,1H),6.96(dd,J1=4.8Hz,J2=3.6Hz,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.90(dd,J1=12.0Hz,J2=4.0Hz,1H),4.70(d,J=5.6Hz,2H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.01(t,J=11.2Hz,1H),3.67(dd,J1=8.4Hz,J2=7.2Hz,1H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (4-methyl-4 NEN-1, 3-oxazol-2-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.14): LC-MS(ESI)375(M+H)+,1H NMR(DMSO-d6,300MHz,80℃)δ10.49(brs,1H),7.93(s,1H),5.21(dd,J1=9.6Hz,J2=3.6Hz,1H),4.58-4.85(m,3H),4.29(m,2H),3.64(m,2H),2.30(s,3H),1.30(d,J=5.4Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (1,4, 5-trimethyl-1N-imidazol-2-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.10): LC-MS(ESI)402(M+H)+

(3S,11aR) -6-hydroxy-3-methyl-N- [ (4-methyl-1, 2, 3-thiadiazol-5-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.11): LC-MS(ESI)392(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.52(brs,1H),10.45(t,J=5.6Hz,1H),8.49(s,1H),5.39(dd,J1=9.2Hz,J2=3.2Hz,1H),4.89(m,1H),4.81(d,J=5.6Hz,2H),4.40(dd,J1=7.6Hz,J2=0.4Hz,1H),4.29(m,1H),4.00(t,J=11.2Hz,1H),3.66(t,J=7.6Hz,1H),2.67(s,3H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (3-methyl-1, 2, 4-oxadiazol-5-yl) methyl]-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.13): LC-MS(ESI)376(M+H)+

(3S,11aR) -6-hydroxy-3-methyl-N- [ (3-fluoropyridin-2-yl) methyl]-5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.16):LC-MS(ESI)389(M+H)+,1H NMR(DMSO-d6,400MHz)δ10.61(t,J=5.4Hz,1H),8.57(s,1H),8.42(d,J=4.5Hz,1H),7.77-7.67(m,1H),7.57-7.50(m,2H),7.47-7.28(m,4H),5.40-5.31(m,1H),5.25-5.17(m,1H),5.11-5.03(m,1H),4.84-4.70(m,3H),4.37-4.23(m,2H),4.08-3.97(m,1H),3.68-3.60(m,1H),1.28(d,J=6.1Hz,3H);

(3S,11aR) -6-hydroxy-N- [ (3, 5-difluoropyridin-2-yl) methyl]-3-methyl-5, 7-dioxo-2, 3,5, 7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.17):LC-MS(ESI)407(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.42(brs,1H),10.54(t,J=5.2Hz,1H),8.49(d,J=2.4Hz,1H),8.46(s,1H),7.95(dt,J1=11.4Hz,J2=2.4Hz,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.88(dd,J1=12.0Hz,J2=4.0Hz,1H),4.71(d,J=4.8Hz,2H),4.39(dd,J1=8.0Hz,J2=6.8Hz,1H),4.29(m,1H),4.00(t,J=11.2Hz,1H),3.66(dd,J1=8.0Hz,J2=6.8Hz,1H),1.34(d,J=6.4Hz,3H);

(3S,11aR) -6-hydroxy-N- [ (6-chloropyridin-3-yl) methyl]-3-methyl-5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.18):LC-MS(M+1)=405,1H NMR(DMSO-d6,400MHz)δ10.76(t,J=5.8Hz,1H),8.35(s,1H),7.92(s,1H),7.75(d,J=8.0Hz,1H),7.45(d,J=8.0Hz,1H),5.21(m,1H),4.60(m,1H),4.53(m,2H),4.25(m,2H),3.75(t,J=10.8Hz,1H),3.59(m,1H),1.26(d,J=4.4Hz,3H);

(3S,11aR) -8- ({ [ (2, 6-difluoropyridin-3-yl) methyl]Amino } carbonyl) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a ] s]Pyrido [1,2-d]Pyrazine-6-ol sodium salt(1.1.20):LC-MS(ESI)407(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.36(t,J=5.8Hz,1H),8.45(s,1H),8.06(dd,J1=17.2Hz,J2=8.4Hz,1H),7.14(dd,J1=8.0Hz,J2=2.0Hz,1H),5.38(dd,J1=10.0Hz,J2=3.6Hz,1H),4.88(dd,J1=12.0Hz,J2=3.6Hz,1H),4.55(d,J=5.6Hz,2H),4.39(dd,J1=8.0Hz,J2=6.8Hz,1H),4.29(m,1H),4.00(t,J=11.0Hz,1H),3.67(dd,J1=8.5Hz,J2=6.8Hz,1H),1.34(d,J=6.4Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (2,4, 6-trifluoropyridin-3-yl) methyl]-5, 7-dioxo-2, 3, 5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.22):LC-MS(ESI)425(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.46(s,1H),10.36(t,J=6.0Hz,1H),8.43(s,1H),7.33(d,J=8.4Hz,1H),5.37(dd,J1=10.0Hz,J2=4.0Hz,1H),4.85(dd,J1=12.4Hz,J2=4.0Hz,1H),4.55(d,J=6.0Hz,2H),4.39(dd,J1=8.4Hz,J2=6.8Hz,1H),4.28(sxt,J=6.4Hz,1H),3.99(dd,J1=11.6Hz,J2=10.8Hz,1H),3.66(dd,J1=8.4Hz,J2=6.8Hz,1H),1.33(d,J=6.4Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (3-fluoropyridin-4-yl) methyl]-5, 7-dioxo-2, 3,5,7, 11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.23):LC-MS(ESI)389(M+H)+,1H NMR(DMSO-d6,400MHz)δ10.54-10.44(m,1H),8.57(s,1H),8.52(s,1H),8.38(d,J=4.4Hz,1H),7.55(d,J=7.2Hz,2H),7.42-7.27(m,4H),5.40-5.30(m,1H),5.23(d,J=10.4Hz,1H),5.08(d,J=10.4Hz,1H),4.84-4.74(m,1H),4.65(d,J=5.6Hz,2H),4.38-4.22(m,2H),4.10-3.96(m,1H),3.70-3.59(m,1H),1.28(d,J=5.7Hz,3H);

(3S,11aR) -6-hydroxy-N- [ (3, 5-difluoropyridin-4-yl) methyl]-3-methyl-5, 7-dioxo-2, 3,5, 7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.24):LC-MS(ESI)407(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.46(brs,1H),10.43(t,J=6.0Hz,1H),8.50(s,2H),8.43(s,1H),5.37(dd,J1=10.0Hz,J2=4.0Hz,1H),4.84(dd,J1=12.0Hz,J2=4.0Hz,1H),4.67(d,J=6.0Hz,2H),4.39(dd,J1=8.4Hz,J2=6.8Hz,1H),4.28(m,1H),3.99(dd,J1=12.0Hz,J2=6.4Hz,1H),3.66(dd,J1=8.8Hz,J2=6.8Hz,1H),1.33(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- (pyrazin-2-ylmethyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.25):LC-MS(ESI)372(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.45(brs,1H),10.53(t,J=5.6Hz,1H),8.63(s,1H),8.61(d,J=2.0Hz,1H),8.54(d,J=2.0Hz,1H),8.47(s,1H),5.39(dd,J1=10.0Hz,J2=4.0Hz,1H),4.89(dd,J1=12.0Hz,J2=4.0Hz,1H),4.71(d,J=5.6Hz,2H),4.40(t,J=7.6Hz,1H),4.30(m,1H),4.01(t,J=11.0Hz,1H),3.67(dd,J1=8.0Hz,J2=7.2Hz,1H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (2-chloroimidazo [2, 1-b)][1,3]Thiazol-6-yl) methyl]-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.28):LC-MS(ESI)450(M+H)+,1H NMR(DMSO-d6,400MHz)δ10.31(brs,1H),8.06(s,1H),7.57(s,1H),5.25(brs,1H),4.65(m,1H),4.48(brs,2H),4.28(brs,2H),3.77(brs,1H),3.63(brs,1H),1.29(brs,3H);

(3S,11aR) -6-hydroxy-N- (imidazo [2, 1-b)][1,3,4]Thiadiazol-6-ylmethyl) -3-methyl-5, 7- Dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.29):LC-MS(ESI)417(M+H)+,1H NMR(DMSO-d6,400MHz)δ10.64(brs,1H),9.18(s,1H),8.00(s,1H),7.91(s,1H),5.22(m,1H),4.60(m,1H),4.49(s,2H),4.25(s,2H),3.75(m,1H),3.59(m,1H),1.26(brs,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (2-methylimidazo [2, 1-b)][1,3,4]Thiadiazol-6-yl) methyl Base of]-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxylic acid esters Amine (1.1.30):LC-MS(ESI)431(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.43(brs,1H),10.27(t,J=5.2Hz,1H),8.47(s,1H),7.95(s,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.90(dd,J1=12.0Hz,J2=3.6Hz,1H),4.49(d,J=5.2Hz,2H),4.39(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.01(t,J=11.2Hz,1H),3.66(dd,J1=8.4Hz,J2=7.2Hz,1H),2.70(s,3H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (1-methyl-benzimidazol-2-yl) methyl]-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.42):LC-MS(ESI)424(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.49(brs,1H),10.60(t,J=4.8Hz,1H),8.44(s,1H),7.68(d,J=8.0Hz,1H),7.63(d,J=7.6Hz,1H),7.37(t,J=8.0Hz,1H),7.63(t,J=7.6Hz,1H),5.39(dd,J1=10.0Hz,J2=3.6Hz,1H),4.95(d,J=4.8Hz,2H),4.88(m,1H),4.40(dd,J1=8.4Hz,J2=6.8Hz,1H),4.30(m,1H),3.99(t,J=11.2Hz,1H),3.89(s,3H),3.67(dd,J1=8.4Hz,J2=6.8Hz,1H),1.35(d,J=6.4Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (6-chloro-1-isopropyl-1N-benzimidazol-2-yl) methyl]-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.45):LC-MS(ESI)486(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.47(brs,1H),10.57(t,J=5.2Hz,1H),8.51(s,1H),7.80(d,J=2.0Hz,1H),7.61(d,J=8.8Hz,1H),7.20(dd,J1=8.8Hz,J2=2.0Hz,1H),5.40(dd,J1=10.0Hz,J2=4.0Hz,1H),4.91(dd,J1=12.0Hz,J2=4.0Hz,1H),4.86(d,J=5.2Hz,2H),4.83(m,1H),4.40(dd,J1=8.4Hz,J2=7.2Hz,1H),4.29(m,1H),4.02(dd,J1=12.0Hz,J2=10.0Hz,1H),3.66(dd,J1=8.4Hz,J2=7.2Hz,1H),1.54(d,J=6.8Hz,6H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -N- (2,1, 3-benzothiadiazol-5-ylmethyl) -6-hydroxy-3-methyl-5, 7-dioxo-2, 3, 5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.47):LC-MS(ESI)428(M+H)+

(3S,11aR) -6-hydroxy-3-methyl-N- (quinolin-6-yl) methyl) -5, 7-dioxo-2, 3,5,7,11,11a- Hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.53):LC-MS(ESI)421(M+H)+

(3S,11aR) -6-hydroxy-N- (isoquinolin-1-ylmethyl) -3-methyl-5, 7-dioxo-2, 3,5,7,11, 11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.55):LC-MS(ESI)421(M+H)+,1H NMR(DMSO-d6,300MHz,80℃)δ11.25(brs,1H),10.64(t,J=5.2Hz,1H),8.47(m,2H),8.32(d,J=8.0Hz,1H),7.99(d,J=8.0Hz,1H),7.75(m,3H),5.42(dd,J1=10.0Hz,J2=3.6Hz,1H),5.20(d,J=5.2Hz,2H),4.85(dd,J1=12.0Hz,J2=3.6Hz,1H),4.38(m,2H),4.01(t,J=10.8Hz,1H),3.70(m,1H),1.38(d,J=5.6Hz,3H);

(3S,11aR) -6-hydroxy-N- (imidazo [1, 2-a)]Pyridin-3-yl) methyl-3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.56):LC-MS(M+1)=410,1H NMR(DMSO-d6,400MHz)δ10.67(t,J=5.6Hz,1H),8.48(d,J=6.4Hz,1H),7.96(s,1H),7.57(m,2H),7.23(t,J=8.0Hz,1H),6.92(t,J=7.2Hz,1H),5.20(dd,J1=9.6Hz,J2=3.6Hz,1H),4.89(m,2H),4.61(m,1H),4.23(m,2H),3.75(t,J=10.8Hz,1H),3.58(m,1H),1.24(d,J=5.6Hz,3H);

(3S,11aR) -6-hydroxy-3-methylradical-N- (1,2, 4-triazolo [4, 3-a)]Pyridin-3-ylmethyl-5, 7-dioxo Substituted-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.57): LC-MS(ESI)411(M+H)+

(3S,11aR) -6-hydroxy-N- (imidazo [1, 2-a)]Pyrimidin-2-ylmethyl) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.58):LC-MS(ESI)411(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.46(brs,1H),10.41(t,J=5.6Hz,1H),8.94(d,J=5.6Hz,1H),8.50(m,2H),7.79(s,1H),7.03(m,1H),5.40(m,1H),4.91(m,1H),4.67(d,J=5.6Hz,2H),4.39(dd,J1=8.0Hz,J2=7.2Hz,1H),4.30(m,1H),4.02(m,1H),3.67(dd,J1=8.0Hz,J2=7.2Hz,1H),1.34(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-N- (imidazo [1, 2-a)]Pyrazin-3-ylmethyl) -3-methyl-5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamide (1.1.59):LC-MS(ESI)411(M+H)+,1H NMR(DMSO-d6,300MHz,80℃)δ10.39(brs,1H),8.98(s,1H),8.59(d,J=6.0Hz,1H),8.07(brs,1H),7.88(d,J=6.0Hz,1H),7.76(s,1H),5.23(m,1H),4.89(m,2H),4.65(m,1H),4.26(s,2H),3.72(brs,1H),3.60(m,1H),1.28(d,J=6.0Hz,3H);

(3S,11aR) -6-hydroxy-3-methyl-N- [ (6-chloro-1, 2, 4-triazolo [4, 3-b)]Pyridazin-3-yl) methyl]- 5, 7-dioxo-2, 3,5,7,11,11 a-hexahydro [1,3]]Oxazolo [3,2-a]Pyrido [1,2-d]Pyrazine-8-carboxamides (1.1.60):LC-MS(ESI)446(M+H)+,1H NMR(DMSO-d6,400MHz)δ11.50(brs,1H),10.59(brs,1H),8.47(m,2H),7.52(d,J=9.6Hz,1H),5.38(dd,J1=10.0Hz,J2=4.0Hz,1H),5.04(d,J=5.6Hz,2H),4.88(m,1H),4.39(dd,J1=8.2Hz,J2=7.2Hz,1H),4.29(sxt,J=6.4Hz,1H),4.00(t,J=11.0Hz,1H),3.66(dd,J1=8.2Hz,J2=7.2Hz,1H),1.34(d,J=6.4Hz,3H);

Example 4 general synthetic procedure for fused 9-hydroxy-1, 8-dioxo-1, 3,4, 8-tetrahydro-2 n-pyrido [1,2-a ] pyrazine-7-carboxamide of formula 1.2 or 1.3.

Starting from (4R,12aS) -7-benzyloxy-9-bromo-4-methyl-6, 8-dioxo-3, 4,6,8,12,12 a-hexahydro-2H-pyrido [1',2':4,5] pyrazino- [2,1-b ] [1,3] oxazine and (2R,5S,13aR) -8-benzyloxy-10-bromo-7, 9-dioxo-2, 3,4,5,7,9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5] pyrazino [2,1-b ] [1,3] oxazepine, respectively, inhibitors 1.2 and 1.3 are prepared in analogy to the synthesis of the compound of general formula 1.1 (example 3), comprises the following steps:

(4R,12aS) -7-hydroxy-N- [ (2, 6-difluoropyridin-3-yl) methyl]-4-methyl-6, 8-dioxo-3, 4,6, 8,12,12 a-hexahydro-2H-pyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazine-9-carboxamide (1.2.1):LC-MS(ESI)421(M+H)+,1H NMR(DMSO-d6,400MHz)δ12.51(brs,1H),10.40(t,J=5.8Hz,1H),8.48(s,1H),8.05(q,J=8.8Hz,1H),7.14(dd,J1=8.0Hz,J2=2.4Hz,1H),5.44(m,1H),4.79(m,1H),4.55(m,3H),4.34(dd,J1=13.6Hz,J2=5.6Hz,1H),4.03(dt,J1=12.0Hz,J2=1.6Hz,1H),3.89(m,1H),2.01(m,1H),1.55(m,1H),1.33(d,J=7.2Hz,3H);

(4R,12aS) -7-hydroxy-4-methyl-N- [ (2,4, 6-trifluoropyridin-3-yl) methyl]-6, 8-dioxo-3, 4, 6,8,12,12 a-hexahydro-2H-pyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazine-9-carboxamide (1.2.3): LC-MS(ESI)439(M+H)+,1H NMR(DMSO-d6,400MHz)δ12.49(brs,1H),10.39(t,J=6.0Hz,1H),8.46(s,1H),7.33(d,J=8.4Hz,1H),5.43(dd,J1=9.6Hz,J2=4.0Hz,1H),4.78(p,J=6.6Hz,1H),4.53(m,3H),4.32(dd,J1=13.6Hz,J2=5.6Hz,1H),4.02(t,J=12.0Hz1H),3.88(m,1H),2.01(m,1H),1.54(m,1H),1.32(d,J=7.2Hz,3H);

(4R,12aS) -7-hydroxy-N- [ (3, 5-difluoropyridin-4-yl) methyl]-4-methyl-6, 8-dioxo-3, 4,6, 8,12,12 a-hexahydro-2H-pyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazine-9-Formamide (1.2.4):LC-MS(ESI)421(M+H)+,1H NMR(DMSO-d6,400MHz)δ12.49(s,1H),10.47(t,J=5.6Hz,1H),8.50(s,2H),8.45(brs,1H),5.43(m,1H),4.78(m,1H),4.66(d,J=5.6Hz,2H),4.53(m,1H),4.32(m,1H),4.02(t,J1=12.0Hz,1H),3.89(m,1H),2.00(m,1H),1.54(d,J=13.6Hz,1H),1.32(d,J=7.2Hz,3H);

(2R,5S,13aR) -8-hydroxy-N- [ (2, 6-difluoropyridin-3-yl) methyl]-7, 9-dioxo-2, 3,4,5,7, 9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazepine-10- Formamide (1.3.1):LC-MS(M+1)=433,1H NMR(DMSO-d6,400MHz)δ12.47(s,1H),10.41(m,1H),8.45(s,1H),8.04(m,1H),7.15(d,J=7.2Hz,1H),5.44(m,1H),5.10(brs,1H),4.68(m,1H),4.60(brs,1H),4.55(m,2H),4.03(m,1H),1.94(brs,4H),1.84(m,1H),1.58(m,1H);

(2R,5S,13aR) -8-hydroxy-N- [ (2,4, 6-trifluoropyridin-3-yl) methyl]-7, 9-dioxo-2, 3,4,5, 7,9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazepine- 10-carboxamide (1.3.2):LC-MS(ESI)451(M+H)+,1H NMR(DMSO-d6,400MHz)δ12.46(s,1H),10.40(t,J=5.6Hz,1H),8.42(s,1H),7.33(d,J=8.4Hz,1H),5.43(dd,J1=10.0Hz,J2=4.0Hz,1H),5.09(brs,1H),4.65(dd,J1=12.8Hz,J2=4.0Hz,1H),4.60(brs,1H),4.54(m,2H),4.01(dd,J1=12.4Hz,J2=9.6Hz,1H),1.94(brs,4H),1.84(d,J=12.0Hz,1H),1.57(m,1H);

(2R,5S,13aR) -8-hydroxy-N- [ (3, 5-difluoropyridin-4-yl) methyl]-7, 9-dioxo-2, 3,4,5,7, 9,13,13 a-octahydro-2, 5-methylenepyrido [1',2':4,5]Pyrazino [2,1-b ] s][1,3]Oxazepine-10- Formamide (1.3.3):LC-MS(ESI)433(M+H)+,1H NMR(DMSO-d6,400MHz)δ12.46(s,1H),10.47(t,J=5.8Hz,1H),8.49(s,2H),8.42(s,1H),5.43(dd,J1=9.2Hz,J2=4.0Hz,1H),5.09(brs,1H),4.66(m,3H),4.60(brs,1H),4.01(dd,J1=12.4Hz,J2=9.6Hz,1H),1.94(brs,4H),1.84(d,J=12.0Hz,1H),1.57(m,1H);

example 5. pharmaceutical composition in tablet form.

Starch (1600mg), ground lactose (1600mg), talc (400mg) and 900mg of a compound of formula 1 or 2 were mixed and pressed into bars. The resulting strands were crushed into granules and sieved to collect 14-16 mesh sized granules. The granules thus obtained are formed into tablets of suitable form, each weighing 225 or 450 mg.

Example 6 pharmaceutical composition in the form of a capsule.

The compound of formula 1 or 2 is thoroughly mixed with lactose powder in a 2:1 ratio. The resulting powdered mixture is packaged into suitable gelatin capsules of 30, 60 or 120mg per capsule.

Example 7. lyophilized pharmaceutical nano-compositions.

Zircon sand (150ml) (grinding medium: 0.5 mm)Zirconia milling and dispersion media; tosoh USA, Inc.) was loaded into a container (tank) with a solution of poloxamer P338(10.0g) and sucrose or mannitol (11.6g) in 400ml phosphate buffered saline (PBS, pH 7.4). The resulting solution (150ml) was placed in a jar and zircon sand (150ml) (grinding media: 0.5 mm)Zirconia milling and dispersion media; tosoh USA, Inc.) and adding a compound of formula 1 or 2 (-15 g). The jar was placed on a jar mill (u.s. storage 700Series), the rotational speed was set at 104rpm, and the mixture was milled for 24 hours. The end of the process was controlled by particle size distribution measurements on a Malvern Zetasizer Nano ZS instrument. At the end of the milling process, the rotation is stopped and the mixture is left at 2-8 ℃ for 16-20 hours to precipitate the zircon sand. The suspension was filtered through a glass filter having a porosity of 5-15 μm and analyzed to determine the content (concentration) of the compound of formula 1 or 2. The obtained particles having a particle size of 200-900nm, preferably 200-250nmThe nanosuspension was aseptically poured in 2ml into 5-ml sterile glass vials (vical), frozen and lyophilized to yield the pharmaceutical nanocomposite in the form of a lyophilizate.

Example 8 injectable drug.

The previously prepared sterile PBS solution with pH 6.8 was added to the nano-composition obtained in example 7 in the form of a lyophilizate at a rate of 2.2ml per 5ml vial. The vials were stoppered with pre-sterilized stoppers and then sealed. The obtained drug nanosuspension can be used.

Example 9 injectable drug.

The compound of formula 1 or 2 was sterilized with gamma radiation and subjected to clean grinding of wet granules together with mannitol, polysorbate 20, polyethylene glycol 3350 and water for injection to reach an average cabozet particle size of 200 nm. The nanosuspensions were placed in pre-sterilized glass vials. The vials were stoppered with pre-sterilized stoppers and then sealed. The obtained drug nanosuspension can be used.

Example 10 anti-HIV Activity of Compounds of formula 1 or 2 and prototype (CAB).

A) lymphotropic virus (lymphotropic virus) strain HIV-1(IIIB) in CEM-SS cell line The inhibitory activity of (3).

Evaluation was performed by ImQuest BioSciences, Inc. (Frederick, MD, USA). CEM-SS cells [ Foley G.E. et al, Continuous cultures of Human lysoblasts from Peripheral Blood of a Child with Acute Leucemea.cancer 1965,18,522-529] obtained by the NIH AIDS Research and Reference Reagent Program (NIH AIDS Research and Reference Reagent Program) (Manassas, VA, USA) were passaged in a T-75 flask and then subjected to antiviral analysis using RPMI1640 medium (Lonza; Walsvilk MD, USA) supplemented with 10% heat-inactivated fetal bovine serum (Gibco; Grand Island, NY), 2. mu. M L-glutamine (Lonza), 100U/. mu.l penicillin (Lonza) and 100. mu.g/. mu.l streptomycin (Lonza). CEM-SS cells were maintained in RPMI 140 medium supplemented with 10% heat-inactivated fetal bovine serum, 2mM glutamine, 100U/ml penicillin and 100. mu.g/ml streptomycin.

The lymphotropic (lymphotropic) HIV-1IIIB strain was selected for the virus to be analyzed [ Popovic, M. et al protection, isolation, and continuous production of cytopathic retroviruses (HTLV-III) from tissues with AIDS and pre-AIDS science 1984,224(4648),497-500 ]. Viruses were obtained by NIH AIDS research and reference reagents program and cultured in CEM-SS cells to generate stock virus stocks. A pre-titration aliquot of the virus was removed from the freezer (-80 ℃) and allowed to thaw slowly to room temperature in a biosafety cabinet. The virus was diluted in tissue culture medium so that the amount of virus added in 50. mu.l/well was sufficient to kill 85-95% of the cells 6 days after infection. HIV-1IIIB was cultured and titrated in CEM-SS cells. The inhibitory effect of CEM-SS compounds on HIV-1 compounds was evaluated in a microtiter plate anti-HIV assay in CEM-SS cells to quantitatively assess the ability of the compounds to inhibit HIV-induced cell death as previously described [ Nara, P.L. et al, Simple, rapid, quantitative, synthetic-formed microassay for the detection of human immunological toxicity. Quantification was performed using 2, 3-bis- (2-methoxy-4-nitro-5-sulfonyl) - (2H) -tetrazol-5-carboxyanilide (XTT) tetrazolium, which was metabolized by living cells into a stained formazan product.

The compounds given in Table 1 were dissolved in DMSO at 10. mu.M and stored at-20 ℃. For antiviral assays, test materials were evaluated in triplicate using concentrations of 30 and 10 nM. The compounds shown in Table 1 were dissolved in DMSO at 10. mu.M and stored at-20 ℃. Test materials were evaluated in triplicate at concentrations of 30 and 10nM using an antiviral assay. Each compound was prepared at a concentration of 60 nanomolar (2-fold the experimental concentration in the well) as follows: mu.M DMSO stock was diluted 1:100 in cell culture medium (10. mu.l of compound added to 990. mu.l medium), followed by 1:10 dilution (10. mu.l of compound added to 90. mu.l medium), followed by 6. mu.l to 994. mu.l medium; 100 μ l were added in triplicate to a 96-well microtiter plate to ensure efficacy and one colorimetric measurement well. Each compound was prepared at 20 nanomolar concentration (2-fold the experimental concentration in the wells) as follows: mu.M DMSO stock was diluted 1:100 in cell culture medium (10. mu.l of compound was added to 990. mu.l of medium), followed by 1:100 dilution (10. mu.l of compound was added to 900. mu.l of medium), followed by 20. mu.l to 980. mu.l of medium; 100 μ l were added in triplicate to a 96-well microtiter plate to ensure efficacy and one colorimetric measurement well.

Azidothymidine (AZT) was purchased from Sigma Aldrich (USA) and evaluated as a positive control compound in an antiviral assay using a 6-concentration reaction curve on each assay plate.

Test compounds were initially evaluated on HIV-1IIIB strain in CEM-SS cells at test concentrations of 30 and 10 nM. Antiviral efficacy data are summarized in table 1. Compounds with activity at 10 and 30nM were evaluated at 2nM in an HIV cytoprotective assay. For compounds exhibiting activity at 2nM, the inhibitory activity (EC) was determined50). Data on antiviral efficacy are also shown in table 1.

AZT control compounds were evaluated in parallel with the test compounds presented and gave EC in the range of less than 2nM to 7nM50The value is obtained. AZT exhibited EC in the range of 10-20nM when compound antiviral activity was evaluated at 2nM50The value is obtained.

TABLE 1 inhibitory Activity of the Compounds of general formulae 1 and 2 on HIV-1 (strain IIIB) (EC) in CEM-SS cell line50).

(a)Inhibitory Activity of HIV-1 carrying a GFP reporter gene (strain NL 4.3) (NL4.3-GFP) in the SupT1 cell line.

Inhibitory Activity of HIV-1 carrying GFP reporter Gene (strain NL 4.3) (NL4.3-GFP) in SupT1 cell line And (4) sex.

Evaluation was performed by RetroVirox Inc (San Diego, CA, USA). For use withThe SupT1 cells were infected with HIV NL4.3 strain (NL4.3-GFP) carrying the green fluorescent protein gene. Viral samples were produced by transfecting 293T cells with proviral DNA. Samples were frozen 48 hours after transfection and stored for use. To increase the efficiency of infection, a suspension of SupT1 cells was pelleted from the infectious mixture by centrifugation. The test substance is added to the cells immediately before the addition of the virus. After 2 hours of incubation, the infectious mixture was replaced with fresh medium containing the test sample. The effectiveness of infection was determined after 45 hours by counting the percentage of fluorescent cells relative to uninfected cell culture. The cytotoxicity of test compounds was determined in parallel on the same, but uninfected, SupT1 cell line using XTT reagents. Serial 10-fold dilutions of the samples were used (starting from 10 μ M when determining antiviral activity, or 100 μ M when determining cytotoxicity). 0.1% DMSO was used as a negative control. Calculation of EC50The value of (c). The properties of the test were determined using the following controls: signal to noise ratio, integrase inhibitor latilatavir (1 μ M) and assay reproducibility. The results are presented in table 1.

Example 11 evaluation of the thermodynamic solubility of the compound of formula 1 or 2.

Test compound of formula 1 or 2 (2mg) was weighed into a glass vial. A pION universal buffer (0.5ml) having a pH of 7.4 or deionized water was added to the appropriate vial. The vial was closed with a cap, placed on a rotary shaker, and incubated for 24 hours to equilibrate the solution and solid phase at the saturation point. Each resulting solution (200. mu.l) was removed from a vial (in duplicate), placed in a 96-well MultiScreen solublity Filter Plate (Millipore), and filtered into a polypropylene Plate with a U-shaped bottom using vacuum (10 "Hg). The filtrate is diluted with an appropriate buffer (water) if necessary. Then, 120. mu.l of the filtrate (or diluted filtrate) was transferred to a 96-well plate with a UV-clear bottom, and 60. mu.l/well acetonitrile and 20. mu.l/well DMSO were added and mixed thoroughly with a multichannel pipettor (acetonitrile and DMSO were added to obtain the desired composition of the calibration standard, as described below). The optical density of the resulting solution was measured in 10nm steps on a multifunctional Infinite 200 PRO plate reader over a wavelength range of 240nm to 400 nm. In order to draw correction curveLine-selected wavelength to assess OD of solutionλThe value of (c).

The concentration of the substance in the solution after the solubility test was evaluated using a calibration curve. Stock solutions in DMSO were prepared from individual samples at a concentration of 2 mg/ml. Using serial dilutions (step 2), 6 10-fold solutions in DMSO were prepared and then diluted 10-fold in the appropriate buffer containing 30% acetonitrile to ensure complete dissolution of the test compound in the mixture. The final concentrations of the calibration curves ranged from 0.2, 0.1, 0.05, 0.025, 0.0125, 0.00625, and 0 mg/ml. Calibration solutions were prepared in 96-well UV plates as follows: mu.l of a 10-fold stock solution in DMSO, 60. mu.l of acetonitrile and 120. mu.l of the appropriate buffer were added to the wells and stirred. The optical density of the resulting solution was measured in 10nm steps on a multifunctional Infinite 200 PRO plate reader over a wavelength range of 240nm to 400 nm. Selecting an optimum wavelength that generally corresponds to the absorption maximum and obtaining the OD at a given wavelengthλLinear dependence on compound concentration.

The concentration of the compound in the filtrate after the solubility test was calculated according to the following formula:

shake flask solubility ═ ODλfiltrate-ODλBlank)/(slope X1.67X filtrate dilution)

Wherein the slope refers to the slope of the calibration curve; 1.67 is the dilution ratio of the filtrate to acetonitrile + DMSO; filtrate dilution is additional dilution of the filtrate, if applicable. Data on the thermodynamic solubility of the compounds of formulae 1 and 2 are presented in table 2.

TABLE 2 thermodynamic solubility of the compounds of the general formulae 1 and 2

Example 12 pharmacokinetics of compounds of general formula 1.1.

Male Sprague-Dawley rats from Charles River Nuclear (Germany) were studied. The age of the animals at the time of administration is 8-10 weeks. The animals were pre-catheterized so that all blood samples during the study of each substance could be taken from the same animal. Each substance was tested in 3 animals per route of administration.

A solution of the test substance at a concentration of 0.2mg/ml in 10% DMSO/10% Solutol/80% 0.05M N-methyl reduced glucosamine is administered intravenously to rats in the tail vein at a dose of 1 mg/kg. Blood samples were collected at 5, 15, 30 minutes, 1,2,4, 8 and 24 hours post-administration. Intragastric administration involves gavage with a solution of 0.5mg/ml in 0.5% methylcellulose at a dose of 5 mg/kg. Blood samples were collected at 15, 30 minutes, 1,2,4, 8 and 24 hours post-administration. Blood (0.2ml) was collected in a polypropylene tube containing 20. mu.l of 5% NaEDTA. Plasma was separated by centrifugation at 1500 × g for 10min and stored until tested at a temperature of-80 ℃.

A pure acetonitrile-water (1:1) mixture (5. mu.l) was added to 45. mu.l of the test sample. The sample was vortexed thoroughly for 10 seconds. Tolbutamide (100. mu.l) (200 ng/ml in MeCN) cooled to +4 ℃ was added to the sample and the mixture was vortexed. The samples were pelleted on ice at +4 ℃ for 15 minutes and then centrifuged at 2700g for 10min at +4 ℃. The supernatants (100. mu.l each) were transferred to a blank plate for HPLC/MS/MS analysis.

HPLC/MS/MS analysis was performed using an Agilent 1290 Infinity system coupled to an AB Sciex QTrap 6500 mass spectrometer.

Using PhoenixTMWinThe software (Pharsight Corp.) carried out pharmacokinetic analysis of plasma concentration versus time data by a model-independent method. The following pharmacokinetic parameters were calculated (table 3): maximum concentration in liver (C)max) And the time (T) to reach itmax) Half life (T)1/2) Area under PC Curve (AUC)0-t、AUC0-inf) Clearance (Cl) and elimination constant (kel).

TABLE 3 pharmacokinetics of Compound 1.1.19 and B in rat plasma when administered Intravenously (IV) and orally (oral)

Example 13 evaluation of the stability of compositions comprising compounds of formulae 1 and 2 in biological media.

A) stability in plasma test compound at a final concentration of 1 μ M was incubated in pooled human plasma (Innovative Research). The incubation was performed in a VorTemp56 shaking incubator at 37 ℃ with stirring at 300 rpm. At certain intervals (0, 0.25, 0.5, 1,2,4, 6,8 and 24h), 30 μ l of sample was taken and the reaction was stopped by adding 180 μ l of cold acetonitrile containing an internal standard to the selected sample. The sample was then centrifuged at 3000rpm for 10min and 150. mu.l of the supernatant was taken for analysis. Incubations were performed in duplicate and each sample was measured 2 times. Samples were analyzed by HPLC-MS/MS developed for each prodrug to be tested using a 1290 Infinity II chromatography system (Agilent Technologies) coupled to a QTRAP5500 tandem mass spectrometer (AB Sciex). When developing conditions for mass spectrometric detection, solutions of test compounds at a concentration of 100ng/ml in a 1:1 acetonitrile-water mixture were analyzed by direct injection into the mass spectrometer with a syringe pump using electrospray ionization in the positive ion mode. Molecular ions for each compound were determined while scanning in total ion current mode (MS1), and the main product ions were recorded in MS2 mode. The MS/MS technique is then optimized in MRM mode to achieve maximum sensitivity. In the quantitative processing of chromatograms, the most intense MRM transition was used for the analyte and the internal standard. Chromatography was performed in mobile phase on a YMC Triart C18,50 x 2.mm,1.9 μm column in gradient elution mode with a composition of 0.1% formic acid in water-0.1% formic acid in acetonitrile. Tolbutamide (Fluka) was used as internal standard. Half-life (T) was calculated from the kinetics of experimental prodrug loss in antiviral compositions during incubation in biological media1/2). In the calculation, values of chromatographic peak areas of substances in the experimental samples normalized to the internal standard signal were used. Calculation of the linear dependence of the log normalized area of the chromatographic peaks on timeDivided by the rate constant (k is the slope of the linear portion). Then, the half-life was calculated: t (T)1/20.693/k. (FIG. 2) it was found that the compounds of formula 1.1 are stable in human and rat plasma.

b) Stability in the S9 fraction.The reaction mixture was prepared in a total final volume of 250. mu.l in 0.1M potassium phosphate buffer (pH 7.4BD Gentest) and contained 1mM NADPH tetrasodium salt (AppliChem), 7mM glucose-6-phosphate sodium salt (Sigma), 1.5U/ml glucose-6-phosphate dehydrogenase (Sigma), 3.3mM MgCl2(Sigma), 5mM uridine-5-diphospho-glucuronic acid trisodium salt (UDPGA, Sigma) and 1. mu.M test compound. The metabolic reaction was started by adding a suspension of the S9 human (BD Gentest) or rat liver fractions, the final protein concentration being 1 mg/ml. The reaction mixture was incubated at 37 ℃ with stirring at 400rpm on a shaker (vortex 56). At certain time intervals (0, 0.25, 0.5, 1,2,4, 6,8, 24h), 30- μ l samples were taken and the reaction stopped by adding 180 μ l of cold acetonitrile containing an internal standard to the selected samples. Proteins were pelleted on ice for 15 minutes, samples were centrifuged at 3000rpm for 10min, and 150 μ Ι of supernatant was collected for analysis. Incubations were performed in duplicate and 2 measurements were made for each sample. (FIG. 3) it was established that the compounds of general formula 1.1 are stable in the S9 fraction of human or rat liver.

Example 14 binding of compound 1.1.17 to plasma proteins of 1.1.19.

To assess protein binding, pooled plasma samples diluted up to 50% with phosphate buffer were used. The experiments were performed in teflon-coated 48-well dialysis plates. Each well contains 2 individual chambers separated by a vertical semi-permeable dialysis membrane with 8kDa pores. A plasma sample containing 1 μ M of the test compound was placed in one of the chambers and a buffer solution (pH 7.2) was placed in the other chamber. Over time, passive diffusion of unbound compounds occurred with shaking at 37 ℃, and after 4 hours, an equilibrium state between the chamber and the plasma and buffer solution was reached. The amount of free fraction was evaluated by HPLC-MS/MS followed by precipitation of the protein with acetonitrile. Also, the study involved evaluating the stability of the substance over a 4-hour period and passive permeability across the dialysis membrane in buffer by mass balance. Warfarin was used as a control compound. The results are presented in table 4 from which it can be seen that the compounds of general formula 1.1 have a high binding to human and rat plasma proteins.

TABLE 4 binding of Compounds 1.1.17 and 1.1.19 to human and rat plasma proteins

Type (B) Compound (I) Permeability, of Extracting, according to Binding, according to Binding classes
Human being 1.1.17 84.8 94.6 98.8 Height of
Rat 1.1.17 90.9 108 99.9 Height of
Human being 1.1.19 84.0 95.0 99.0 Height of
Rat 1.1.19 83.8 94.5 98.9 Height of

INDUSTRIAL APPLICABILITY

The invention can be applied in human and veterinary medicine.

76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:新型偶氮苯衍生物、其制备方法及其在与电离辐射联合的治疗性治疗中的用途

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!