CN116917294A - Rapamycin analogues and uses thereof - Google Patents

Rapamycin analogues and uses thereof Download PDF

Info

Publication number
CN116917294A
CN116917294A CN202280011222.6A CN202280011222A CN116917294A CN 116917294 A CN116917294 A CN 116917294A CN 202280011222 A CN202280011222 A CN 202280011222A CN 116917294 A CN116917294 A CN 116917294A
Authority
CN
China
Prior art keywords
compounds
nitrogen
sulfur
oxygen
heteroatoms independently
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280011222.6A
Other languages
Chinese (zh)
Inventor
D·J·奥尼尔
E·赛亚
S·W·A·康
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceutica NV
Original Assignee
Janssen Pharmaceutica NV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Pharmaceutica NV filed Critical Janssen Pharmaceutica NV
Priority claimed from PCT/US2022/070308 external-priority patent/WO2022159976A1/en
Publication of CN116917294A publication Critical patent/CN116917294A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application provides compounds, compositions thereof, and methods of use thereof.

Description

Rapamycin analogues and uses thereof
Cross Reference to Related Applications
The present application is based on the benefits of U.S. C. ≡119 (e) claiming U.S. provisional application 63/140,523 filed on 22 nd 1 year 2021 and U.S. provisional application 63/202,524 filed on 15 th 6 year 2021, the disclosures of each of which are incorporated herein by reference in their entirety.
Technical Field
The present application relates to compounds and methods useful for modulating mTORC1 activity. The application also provides pharmaceutically acceptable compositions comprising the provided compounds of the application, and methods of using such compositions in the treatment of various disorders.
Background
mTOR complex 1 (mTORC 1) positively regulates cell growth and proliferation by promoting biosynthesis of many anabolic processes, including proteins, lipids, and organelles, as well as by limiting catabolic processes such as autophagy. Much of the knowledge about mTORC1 function comes from the use of the bacterial macrolide rapamycin. Upon entry into the Cell, rapamycin binds to the 12kDa FK506 binding protein (FKBP 12) and interacts with the FKBP 12-rapamycin binding domain (FRB) of mTOR, thereby inhibiting mTORC1 function (Guertin, d.a. and Sabatini, d.m., cancer Cell, volume 12, phase 1: pages 9-22 (2007)). In contrast to the effects on mTORC1, FKBP 12-rapamycin is unable to physically interact with or acutely inhibit mTOR complex 2 (mTORC 2) (Janinto, E. Et al, nat.cell Bio., vol.6, 11:1122-1128 (2004); sarbasov, D.D. et al, curr. Biol., vol.14:1296-1302 (2004)). Based on these observations, mTORC1 and mTORC2 have been characterized as rapamycin-sensitive and rapamycin-insensitive complexes, respectively. However, this paradigm may not be entirely accurate, as long-term rapamycin treatment may, in some cases, inhibit the activity of mTORC2 by blocking its assembly (sarbasov, d.d. et al, mol.cell, volume 22, phase 2: pages 159-168 (2006)). Furthermore, recent reports indicate that important mTORC1 functions are resistant to inhibition by rapamycin (Choo, a.y. et al, proc.Natl.Acad.Sci., volume 105, 45, pages 17414-17419 (2008), feldman, M.E., et al, PLoS biol., volume 7, 2, e38 (2009), garcia-Martinez, J.M., et al, biochem J., volume 421, pages 29-42 (2009), thooren, C.C., et al, J.biol.chem., volume 284, 12, pages 8023-8032 (2009)). Thus, selective inhibition of mTORC1 would be able to treat diseases involving dysregulation of protein synthesis and cellular metabolism. Furthermore, this detailed knowledge of the modulation of mTORC1 activation pathway will help to discover new strategies for modulating abnormal disease processes by modulating mTORC1 activity within its functional range.
Many diseases are associated with abnormal cellular responses triggered by the events described above. These diseases include, but are not limited to, autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, alzheimer's disease and hormone-related diseases.
Rapamycin mechanism target protein complex 1 (mTORC 1) is a major growth regulator that senses various environmental cues such as growth factors, cellular stress, and nutrients and energy levels. mTORC1 phosphorylation, when activated, enhances substrates for anabolic processes (such as mRNA translation and lipid synthesis) and limits catabolic processes (such as autophagy). mTORC1 dysfunction occurs in a variety of diseases including diabetes, epilepsy, neurodegeneration, immune responses, skeletal muscle growth inhibition, and cancer, among others (Howell, j.j. Et al biochem. Soc. Trans., volume 41, pages 906-912 (2013); kim, s.g. et al, molecular and cells, volume 35, pages 6: 463-473 (2013); laplante, m. And Sabatini, d.m., cell, volume 149, page 2: 274-293 (2012)).
Rapamycin was originally discovered as an antifungal metabolite produced by streptomyces hygroscopicus in Easter island soil samples. Subsequently, rapamycin was found to have immunosuppressive and antiproliferative properties in mammalian cells, which has stimulated interest in determining the mode of action of rapamycin. Rapamycin has proven to be a potent inhibitor of S6K1 phosphorylation. Meanwhile, rapamycin target proteins (TORs) were identified in yeast and animal cells. Rapamycin forms a functionally acquired complex with the 12kDa FK506 binding protein (FKBP 12) and this complex binds and acts specifically as an allosteric inhibitor of mammalian TOR (mTOR, also known as mechanism TOR) complex 1 (mTORC 1).
Biochemical and genetic analysis of mTOR demonstrated its presence in two functionally distinct complexes. The core component of mTORC1 consists of mTOR, mammalian lethal sec-13 protein 8 (mLST 8), and a regulator-associated protein of TOR (Raptor). Additional components include an mTOR interacting protein (depto) containing a DEP domain and a proline-rich 40kDa Akt substrate (PRAS 40).
The mTOR complex 2 (mTORC 2) core consists of mTOR, rapamycin-insensitive mTOR binding protein (vector), stress-activated protein kinase interacting protein 1 (mSIN 1), and mLST 8. Proteins observed with vector 1/2 (pro 1/2) and DEPTOR are additional regulatory components. S6 kinase 1 (S6K 1) and eukaryotic inhibitor eIF4E binding protein 1 (4E-BP 1) are two well-characterized substrates for mTorrC 1, while AKT is a well-characterized substrate for mTorrC 2 (Li, J. Et al, cell Met, vol.19, 3: pp.373-379 (2014)).
Because FKBP 12-rapamycin does not bind to mTorrC 2, rapamycin was originally thought to inhibit only mTorrC 1 (Sarbasov, D.D. et al, curr. Biol., vol. 14, 14: pages 1296-1302 (2004)). However, in 2006, there were studies showing that rapamycin inhibits the assembly and function of mTORC2 and inhibits pAkt (sarbasov, d.d. et al, molecular Cell, volume 22, phase 2: pages 159-168 (2006)). The effect of rapamycin on phosphorylation of S473 (mTORC 2 substrate) and T389 (mTORC 1 substrate) of Akt, S6K1, was compared in a number of cell lines. Treatment with rapamycin for 1 or 24 hours inhibited S6K1 phosphorylation in PC3, HEK-293t, heLa and H460 cells, consistent with the conclusion of inhibition of mTORC 1. Selective inhibition of S6K1 by rapamycin should result in increased phosphorylation of Akt, and this is indeed the case in HeLa cells. However, in PC3 cells, the drug significantly reduced Akt phosphorylation, indicating that rapamycin is not selective in this cell line. Partial inhibition of pAKT was observed in HEK-293T cells. In about one third of the cell lines, rapamycin causes a strong or partial inhibition of Akt phosphorylation, while in other cell lines, the drug does not affect or increase Akt phosphorylation. Inhibition of pAKT was also observed after 24 hours in both primary and non-transformed cell lines (including endothelial cells and muscle cells). Rapamycin has also been shown to inhibit pAkt in vivo because mice treated with this drug daily for 1 week have reduced Akt phosphorylation in thymus, adipose tissue, heart and lung. These findings indicate that inhibition of Akt phosphorylation by rapamycin is common and occurs in normal cell lines, cancer cell lines, and in vivo.
Sarbassov et al concluded that rapamycin and its analogs (CCI 779, RAD001 (also known as everolimus), AP 23573) can inhibit mTORC2 function in certain cell lines and tissues. Rapamycin mediated inhibition of Akt may help explain the side effects of the drug. For example, rapamycin strongly inhibits Akt phosphorylation in adipose tissue, in which insulin-stimulated Akt activity plays an important role in inhibiting lipolysis. Inhibition of Akt by rapamycin in adipocytes allows lipolysis to remain high even in the presence of insulin, resulting in the accumulation of free fatty acids in the plasma that can be used by the liver to produce triglycerides, providing a molecular mechanism for the treatment of hyperlipidemia common in patients with rapamycin.
Pereira et al (Mol Cell endocrinol.; vol.355, phase 1: pages 96-105 (2012)) explored the effects of rapamycin on glucose uptake and insulin signaling proteins in adipocytes obtained by human donor fat biopsies. At therapeutic concentrations (0.01 μm), rapamycin reduced AKT (PKB) Ser473 phosphorylation and reduced glucose uptake in human adipocytes through impaired insulin signaling.
Lamming et al (science, volume 335, 6076: pages 1638-1643 (2012)) demonstrate that rapamycin destroys mTORC2 in vivo and that mTORC2 is essential for insulin-mediated liver gluconeogenesis inhibition.
Similar results are also shown in humans. Di Paolo et al published a similar discovery in humans (JASN, vol.17, 8: pages 2236-2244 (2006)). Given the critical role of rapamycin in regulating cell growth and survival, and in cellular response to nutrients and growth factors, their primary objective of research was to determine the effect of prolonged exposure to rapamycin on AKT activation. They found that mTOR inhibition was associated with a significant down-regulation of basal and insulin-induced AKT phosphorylation levels. AKT is primarily responsible for many metabolic roles of insulin, so they conclude that inhibition of AKT activation is significantly associated with an increase in insulin resistance in renal transplant recipients.
Kennedy et al have recently reviewed the role of mTORC1 and mTORC2 in metabolism and aging (Cell Metab., 23, volume 6: pages 990-1003 (2016)).
Thus, there remains a need to provide potent and mTORC1 specific inhibitors with improved safety and tolerability due to the lack of direct mTORC2 inhibition.
Disclosure of Invention
The compounds of the present invention and pharmaceutically acceptable compositions thereof have now been found to be effective as mTORC1 inhibitors. Such compounds have the general formula I:
or a pharmaceutically acceptable salt thereof, wherein R 1 、R 2 、R 3 、R 3' 、R 4 、R 5 、R 5' 、R 6 、L 1 、L 2 、X、X 1 、X 2 And X 3 Each as defined and described herein.
The compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders, or conditions associated with mTORC 1. Such diseases, disorders, or conditions include the diseases, disorders, or conditions described herein.
Drawings
FIG. 1 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-59, I-57 and I-55.
FIG. 2 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-69, I-66, I-64 and I-62.
FIG. 3 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-85, I-97 and I-83.
FIG. 4 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-34, I-49 and I-31.
FIG. 5 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-37, I-43 and I-40.
FIG. 6 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin and I-14.
FIG. 7 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-4, I-27 and I-47.
FIG. 8 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-9 and I-21.
FIG. 9 shows a comparison of Western blots performed 24 hours after treatment of PC3 cells with rapamycin, I-18 and I-45.
Detailed Description
1. General description of certain embodiments of the invention
It was surprisingly found that the provided compounds inhibited mTORC1 for extended periods of time (e.g., 8 hours, 24 hours, 30 hours, and 48 hours), but had no effect on mTORC2 (as measured by its effect on pAKT). This novel activity is based on the presence of a sufficiently large group at the C-7 position of rapamycin and its analogues. Small substitutions at this position, such as OMe (as shown in rapamycin), OEt, OBn, do not confer selectivity to mTORC2 for 24 hours. Groups of medium length, e.g. OCH 2 CH 2 OH or OCH 2 CH 2 CH 2 OH showed partial selectivity to mTORC2 over 24 hours, but still showed some degree of inhibition. In contrast, larger groups, such as those of the present invention, provided significant selectivity for mTORC2, as measured by the effect on pAKT.
The position of this substitution is also critical to the selectivity observed. For example, the introduction of a larger substitution at the C-43 position does not result in this unique selectivity feature claimed in the present application.
For clarity, the structure of rapamycin is replicated as follows, and the C-7 and C-43 positions are indicated.
In some embodiments, the present application provides novel rapamycin analogues that are potent mTORC1 inhibitors, as measured by pS 6K. Unlike rapamycin and everolimus, these compounds do not inhibit pAKT at longer time points (e.g., 24 hours and 48 hours). These compounds also show improved solubility and improved pharmacokinetics compared to rapamycin.
The activity of the compounds used as mTORC1 inhibitors in the present application can be measured in vitro, in vivo or in cell lines. In vitro assays include assays that determine inhibition of mTORC 1. The detailed conditions for determining the compounds used as mTORC1 inhibitors in the present application are well known to those of ordinary skill in the art. Such methods are described in detail in Liu et al, cancer Research, volume 73, phase 8: pages 2574-2586 (2013) and Liu et al J.biological Chemistry, volume 287, phase 13: pages 9742-9752 (2012).
In certain embodiments, the invention provides compounds of formula I:
or a pharmaceutically acceptable salt thereof, wherein:
x and X 3 Independently a covalent bond, -CR 2 -、-NR-、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -, -O-, -S-or-SO 2 NR-;
L 1 Is covalent bond or C 1-30 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of said chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-S(O) 2 -、-C(O)-、-C(S)-、-C(R) 2 -、-CH(R)-、-CF 2 -、-P(O)(R)-、-Si(R) 2 -Si (OR) (R) -OR-NR-substitution;
each-Cy 1 -independently is an optionally substituted divalent ring selected from phenylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R is independently hydrogen or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, or
Two R groups on the same atom are joined together with the atoms between them to form a 4-7 membered saturated, partially unsaturated or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur in addition to the same atom to which they are attached;
L 2 Is optionally substituted C 1-6 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1-2 methylene units of said chain are independently and optionally protected by-Cy 1 -substitution;
R 1 and R is 2 Is independently hydrogen, halogen, -OR, -CN, -NO 2 、-NR 2 、-NR(C 1-6 Haloalkyl), -NRCOR, -NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 8-10 membered bicyclic aromatic carbocycle, 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur;
R 3 is hydrogen, halogen, -OR OR-OSiR 3
R 3' Is hydrogen, halogen, -OR OR-OSiR 3
Or R is 3 And R is 3' Taken together to form =o or =s;
R 4 and R is 6 Is independently hydrogen, -OR, -NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 An aliphatic;
R 5 and R is 5' Each hydrogen or taken together to form =o or =nor;
X 1 and X 2 Each independently is-CR 2 -, -S-or-S (O) -,
Wherein X is 1 And X 2 At least one of them is-CR 2 -。
2. Compounds and definitions
The compounds of the present invention include those generally described herein and are further illustrated by the classes, subclasses, and species disclosed herein. Unless otherwise indicated, the following definitions will apply. For the purposes of the present invention, the chemical elements are defined according to the periodic Table of the elements, CAS version Handbook of Chemistry and Physics, 75 th edition. In addition, general principles of organic chemistry are described in "Organic Chemistry", thomas Sorrell, university Science Books, sausalato 1999 and "March's Advanced Organic Chemistry", 5 th edition, smith, m.b. and March, j. Editions, john Wiley & Sons, new york:2001, the entire contents of which are incorporated herein by reference.
As used herein, the term "aliphatic" or "aliphatic group" means a straight (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is fully saturated or contains one or more units of unsaturation, or a mono-or bicyclic hydrocarbon (also referred to herein as "carbocycle", "alicyclic" or "cycloalkyl") that is fully saturated or contains one or more units of unsaturation but is not aromatic, having a single point of attachment to the rest of the molecule. Unless otherwise indicated, aliphatic groups contain 1 to 6 aliphatic carbon atoms. In some embodiments of the present invention, in some embodiments, The aliphatic group contains 1 to 5 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1 to 4 aliphatic carbon atoms. In still other embodiments, the aliphatic group contains 1-3 aliphatic carbon atoms, and in still other embodiments, the aliphatic group contains 1-2 aliphatic carbon atoms. In some embodiments, "cycloaliphatic" (or "carbocycle" or "cycloalkyl") refers to a monocyclic C that is fully saturated or contains one or more units of unsaturation, but which is not aromatic 3 -C 6 Hydrocarbons, which have a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, straight or branched chain, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl) alkyl, (cycloalkenyl) alkyl or (cycloalkyl) alkenyl.
The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus or silicon (including any oxidized form of nitrogen, sulfur, phosphorus or silicon; quaternized forms of any basic nitrogen; heterocyclic substitutable nitrogen, such as N (as in 3, 4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
As used herein, the term "unsaturated" means that a moiety has one or more unsaturated units.
The term "divalent C", as used herein 1-8 (or C) 1-6 ) Saturated or unsaturated straight or branched hydrocarbon chain "means divalent alkylene, alkenylene, and alkynylene chains as defined herein that are straight or branched.
The term "alkylene" refers to a divalent alkyl group. "alkylene chain" is a polymethylene group, i.e., - (CH) 2 ) n -wherein n is a positive integer, preferably a positive integer from 1 to 6, 1 to 4, 1 to 3, 1 to 2 or 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced by a substituent. Suitable substituents include those described below for substituted aliphatic groups.
The term "alkenylene" refers to a divalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced by a substituent. Suitable substituents include those described below for substituted aliphatic groups.
The term "halogen" means F, cl, br or I.
The term "aryl" used alone or as part of a larger moiety such as in "aralkyl", "aralkoxy" or "aryloxyalkyl" refers to a monocyclic or bicyclic ring system having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, and wherein each ring in the system contains 3 to 7 ring members. The term "aryl" may be used interchangeably with the term "aryl ring". In certain embodiments of the present invention, "aryl" refers to an aromatic ring system that may carry one or more substituents, including but not limited to phenyl, biphenyl, naphthyl, anthracenyl, and the like. Also included within the scope of the term "aryl" as used herein are groups in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthalimidyl, phenanthridinyl, tetrahydronaphthyl, and the like.
The terms "heteroaryl" and "heteroaryl-" used alone or as part of a larger moiety, such as "heteroarylalkyl" or "heteroarylalkoxy", refer to a moiety having 5 to 10 ring atoms, preferably 5, 6 or 9 ring atoms; having 6, 10 or 14 pi electrons shared in a ring array; and has one to five hetero atoms in addition to carbon atoms. The term "heteroatom" refers to nitrogen, oxygen or sulfur, and includes any oxidized form of nitrogen or sulfur and any quaternized form of basic nitrogen. Heteroaryl groups include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolyl, purinyl, naphthyridinyl, and piperidinyl. As used herein, the terms "heteroaryl" and "heteroaryl-" also include groups in which a heteroaromatic ring is fused to one or more aryl, alicyclic, or heterocyclic rings, wherein the group or point of attachment is on the heteroaromatic ring. Non-limiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido [2,3-b ] -1, 4-oxazin-3 (4H) -one. Heteroaryl groups may be monocyclic or bicyclic. The term "heteroaryl" may be used interchangeably with the terms "heteroaryl ring", "heteroaryl group" or "heteroaromatic", any of which include optionally substituted rings. The term "heteroarylalkyl" refers to an alkyl group substituted with a heteroaryl, wherein the alkyl and heteroaryl moieties are independently optionally substituted.
As used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic group" and "heterocyclic ring" are used interchangeably and refer to a stable 5-7 membered monocyclic or 7-10 membered bicyclic heterocyclic moiety that is saturated or partially unsaturated and has one or more, preferably one to four, heteroatoms in addition to carbon atoms, as defined above. The term "nitrogen" when used in reference to a ring atom of a heterocycle includes substituted nitrogen. For example, in a saturated or partially unsaturated ring having 0 to 3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3, 4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or + NR (as in N-substituted pyrrolidinyl).
The heterocycle may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure, and any of the ring atoms may be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazanylRadical, oxazal- >Basic, thiazideGroup, morpholinyl, and quinuclidinyl. The terms "heterocycle", "heterocyclyl ring", "heterocyclyl group", "heterocyclic moiety" and "heterocyclic group" are used interchangeably herein and also include groups in which the heterocyclyl ring is fused to one or more aryl, heteroaryl or cycloaliphatic rings, such as indolyl, 3H-indolyl, chromanyl, phenanthridinyl or tetrahydroquinolinyl. The heterocyclyl group may be monocyclic or bicyclic (e.g., bridged bicyclic or spiro). The term "heterocyclylalkyl" refers to an alkyl group substituted with a heterocyclyl group, wherein the alkyl and heterocyclyl moieties are independently optionally substituted.
As used herein, the term "partially unsaturated" refers to a cyclic moiety that includes at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties as defined herein.
The compounds of the invention may contain "optionally substituted" moieties, as described herein. Generally, the term "substituted" means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have suitable substituents at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from the specified group, the substituents may be the same or different at each position. The combinations of substituents contemplated by the present invention are preferably substituents that result in the formation of stable or chemically feasible compounds. As used herein, the term "stable" refers to a compound that does not substantially change when limited by conditions that allow for the production, detection, and in certain embodiments, recovery, purification, and use of the compound for one or more of the purposes disclosed herein.
Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halogen; - (CH) 2 ) 0-4 R o ;-(CH 2 ) 0-4 OR o ;-O(CH 2 ) 0-4 R o 、-O-(CH 2 ) 0-4 C(O)OR o ;-(CH 2 ) 0-4 CH(OR o ) 2 ;-(CH 2 ) 0- 4 SR o ;-(CH 2 ) 0-4 Ph, which may be R o Substitution; - (CH) 2 ) 0-4 O(CH 2 ) 0-1 Ph, which may be R o Substitution; -ch=chph, which may be R o Substitution; - (CH) 2 ) 0-4 O(CH 2 ) 0-1 -pyridinyl, which may be substituted by R o Substitution; -NO 2 ;-CN;-N 3 ;-(CH 2 ) 0-4 N(R o ) 2 ;-(CH 2 ) 0-4 N(R o )C(O)R o ;-N(R o )C(S)R o ;-(CH 2 ) 0-4 N(R o )C(O)NR o 2 ;-N(R o )C(S)NR o 2 ;-(CH 2 ) 0-4 N(R o )C(O)OR o ;-N(R o )N(R o )C(O)R o ;-N(R o )N(R o )C(O)NR o 2 ;-N(R o )N(R o )C(O)OR o ;-(CH 2 ) 0-4 C(O)R o ;-C(S)R o ;-(CH 2 ) 0-4 C(O)OR o ;-(CH 2 ) 0-4 C(O)SR o ;-(CH 2 ) 0-4 C(O)OSiR o 3 ;-(CH 2 ) 0 - 4 OC(O)R o ;-OC(O)(CH 2 ) 0-4 SR-、SC(S)SR o ;-(CH 2 ) 0-4 SC(O)R o ;-(CH 2 ) 0 - 4 C(O)NR o 2 ;-C(S)NR o 2 ;-C(S)SR o ;-SC(S)SR o 、-(CH 2 ) 0-4 OC(O)NR o 2 ;-C(O)N(OR o )R o ;-C(O)C(O)R o ;-C(O)CH 2 C(O)R o ;-C(NOR o )R o ;-(CH 2 ) 0 - 4 SSR o ;-(CH 2 ) 0-4 S(O) 2 R o ;-(CH 2 ) 0-4 S(O) 2 OR o ;-(CH 2 ) 0-4 OS(O) 2 R o ;-S(O) 2 NR o 2 ;-(CH 2 ) 0-4 S(O)R o ;-N(R o )S(O) 2 NR o 2 ;-N(R o )S(O) 2 R o ;-N(OR o )R o ;-C(NH)NR o 2 ;-P(O) 2 R o ;-P(O)R o 2 ;-OP(O)R o 2 ;-OP(O)(OR o ) 2 ;SiR o 3 ;-(C 1-4 Linear or branched alkylene) O-N (R) o ) 2 The method comprises the steps of carrying out a first treatment on the surface of the Or- (C) 1-4 Straight or branched chain alkylene) C (O) O-N (R) o ) 2 Wherein each R is o May be substituted as defined below and independently hydrogen, C 1-6 Aliphatic, -CH 2 Ph、-O(CH 2 ) 0-1 Ph、-CH 2 - (5-6 membered heteroaryl ring) or a 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or, in spite of the above definition, two independently occurring R o Taken together with the atoms between them to form a 3-12 membered saturated, partially unsaturated or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, which may be substituted as defined below.
R o Suitable monovalent substituents on (or by two independently occurring R o Ring formed by bonding together atoms between them) is independently halogen, - (CH) 2 ) 0-2 R · - (halo R) · )、-(CH 2 ) 0 - 2 OH、-(CH 2 ) 0-2 OR · 、-(CH 2 ) 0-2 CH(OR · ) 2 The method comprises the steps of carrying out a first treatment on the surface of the -O (halo R) · )、-CN、-N 3 、-(CH 2 ) 0 - 2 C(O)R · 、-(CH 2 ) 0-2 C(O)OH、-(CH 2 ) 0-2 C(O)OR · 、-(CH 2 ) 0-2 SR · 、-(CH 2 ) 0-2 SH、-(CH 2 ) 0-2 NH 2 、-(CH 2 ) 0-2 NHR · 、-(CH 2 ) 0-2 NR · 2 、-NO 2 、-SiR · 3 、-OSiR · 3 、-C(O)SR · 、-(C 1-4 Straight-chain OR branched alkylene) C (O) OR · or-SSR · Wherein each R is · Unsubstituted or substituted with one or more halogens only when preceded by a "halo" and independently selected from C 1-4 Aliphatic, -CH 2 Ph、-O(CH 2 ) 0-1 Ph or a 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. R is R o Suitable divalent substituents on saturated carbon atoms of (c) include =o and =s.
Suitable divalent substituents on the saturated carbon atoms of an "optionally substituted" group include the following: =o, =s, =nnr * 2 、=NNHC(O)R * 、=NNHC(O)OR * 、=NNHS(O) 2 R * 、=NR * 、=NOR * 、-O(C(R * 2 )) 2-3 O-or-S (C (R) * 2 )) 2-3 S-, wherein each independently occurs R * Selected from hydrogen, C which may be substituted as defined below 1-6 An aliphatic or unsubstituted 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur. Suitable divalent substituents bonded to the ortho-substitutable carbon of an "optionally substituted" group include: -O (CR) * 2 ) 2-3 O-, wherein each independently occurs R * Selected from hydrogen, C which may be substituted as defined below 1-6 An aliphatic or unsubstituted 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
R * Suitable substituents on the aliphatic radical of (2) include halogen, -R · - (halo R) · )、-OH、-OR · (halo) R · )、-CN、-C(O)OH、-C(O)OR · 、-NH 2 、-NHR · 、-NR · 2 or-NO 2 Wherein each R is · Unsubstituted or substituted by one or more halogens only when preceded by a "halo" andindependently C 1-4 Aliphatic, -CH 2 Ph、-O(CH 2 ) 0-1 Ph or a 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
Suitable substituents on the substitutable nitrogen of an "optionally substituted" group include Or->Each of which is->C independently hydrogen, which may be substituted as defined below 1-6 Aliphatic, unsubstituted-OPh or unsubstituted 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or two independently occurring +.>Taken together with the atoms between them to form an unsubstituted 3-12 membered saturated, partially unsaturated or aryl monocyclic or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
R * Suitable substituents on the aliphatic radical of (2) are independently halogen, -R · - (halo R) · )、-OH、-OR · (halo) R · )、-CN、-C(O)OH、-C(O)OR · 、-NH 2 、-NHR · 、-NR · 2 or-NO 2 Wherein each R is · Unsubstituted or substituted with one or more halogens only when preceded by a "halo" and is independently C 1-4 Aliphatic, -CH 2 Ph、-O(CH 2 ) 0-1 Ph or a 5-6 membered saturated, partially unsaturated or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
As used herein, the term "pharmaceutically acceptable salt" refers to a salt that is suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and within the scope of sound medical judgment. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in detail in J.pharmaceutical Sciences,1977, volume 66, pages 1-19, which are incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of the invention include salts derived from suitable inorganic and organic acids and inorganic and organic bases. Examples of pharmaceutically acceptable non-toxic acid addition salts are salts formed from amino groups with inorganic acids such as hydrochloric, hydrobromic, phosphoric, sulfuric and perchloric acids, or with organic acids such as acetic, oxalic, maleic, tartaric, citric, succinic or malonic acid, or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipic acid salts, alginates, ascorbates, aspartic acid, benzenesulfonates, benzoates, bisulphates, borates, butyrates, camphorates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfate, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, caprates, hydroiodinates, 2-hydroxy-ethanesulfonates, lactoaldehyde, lactates, laurates, lauryl sulfates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmates, pamonates, pectinates, persulfates, 3-phenylpropionates, phosphates, trimethylacetates, propionates, stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonates, undecanoates, valerates, and the like.
Salts derived from suitable bases include basesMetal salts, alkaline earth metal salts, ammonium salts and N + (C 1-4 Alkyl group 4 And (3) salt. Representative alkali metal or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Additional pharmaceutically acceptable salts include nontoxic ammonium, quaternary ammonium and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate where appropriate.
Unless otherwise indicated, compounds having the structures shown herein are also meant to include any pharmaceutically acceptable salts, stereoisomers, tautomers, solvates, hydrates, and polymorphs thereof.
Unless otherwise indicated, compounds having the structures shown herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms of these structures); for example, the R and S configuration, Z and E double bond isomers, and Z and E conformational isomers for each asymmetric center. Thus, single stereochemical isomers, as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the compounds of the invention are within the scope of the invention. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention. In addition, unless otherwise indicated, structures shown herein are also intended to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, having a composition comprising replacement of hydrogen with deuterium or tritium or with 13 C-or 14 C-enriched carbon-substituted carbon compounds of the structure of the invention are within the scope of the invention. Such compounds may be used, for example, as analytical tools, probes in biological assays, or as therapeutic agents according to the invention.
As used herein, the terms "measurable affinity" and "measurable inhibition" refer to a measurable change in mTORC1 activity between a sample comprising a compound of the invention or a composition thereof and mTORC1 and an equivalent sample comprising mTORC1 in the absence of the compound or composition thereof.
3. Exemplary embodimentsDescription of the embodiments
As described above, in certain embodiments, the present invention provides compounds of formula I:
or a pharmaceutically acceptable salt thereof, wherein:
x and X 3 Independently a covalent bond, -CR 2 -、-NR-、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -, -O-, -S-or-SO 2 NR-;
L 1 Is covalent bond or C 1-30 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of said chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-SO 2 -、-C(O)-、-C(S)-、-CR 2 -、-CF 2 -、-P(O)(R)-、-SiR 2 -Si (OR) (R) -OR-NR-substitution;
each-Cy 1 -independently is an optionally substituted divalent ring selected from phenylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
Each R is independently hydrogen or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, or
Two R groups on the same atom are joined together with the atoms between them to form a 4-7 membered saturated, partially unsaturated or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur in addition to the same atom to which they are attached;
L 2 is optionally substituted C 1-6 Divalent straight or branched chain saturated or unsaturatedHydrocarbon chain, wherein 1-2 methylene units of said chain are independently and optionally protected by-Cy 1 -substitution;
R 1 and R is 2 Is independently hydrogen, halogen, -OR, -CN, -NO 2 、-NR 2 、-NR(C 1-6 Haloalkyl), -NRCOR, -NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 8-10 membered bicyclic aromatic carbocycle, 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur;
R 3 Is hydrogen, halogen, -OR OR-OSiR 3
R 3' Is hydrogen, halogen, -OR OR-OSiR 3
Or R is 3 And R is 3' Taken together to form =o or =s;
R 4 and R is 6 Is independently hydrogen, -OR, -NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 An aliphatic;
R 5 and R is 5' Each hydrogen or taken together to form =o or =nor;
X 1 and X 2 Each independently is-CR 2 -, -S-or-S (O) -,
wherein X is 1 And X 2 At least one of them is-CR 2 -。
In certain embodiments, the invention provides compounds of formula I':
or a pharmaceutically acceptable salt thereof, wherein:
x and X 3 Independently a covalent bond, -CR 2 -、-NR-、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -, -O-, -S-or-SO 2 NR-;
L 1 Is covalent bond or C 1-30 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of said chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-SO 2 -、-C(O)-、-C(S)-、-CR 2 -、-CF 2 -、-P(O)(R)-、-SiR 2 -Si (OR) (R) -OR-NR-substitution;
each-Cy 1 -independently is an optionally substituted divalent ring selected from phenylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R is independently hydrogen or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, or
Two R groups on the same atom are joined together with the atoms between them to form a 4-7 membered saturated, partially unsaturated or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur in addition to the same atom to which they are attached;
L 2 is optionally substituted C 1-6 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1-2 methylene units of said chain are independently and optionally protected by-Cy 1 -substitution;
R 1 and R is 2 Independently hydrogen, halogen, -OR, -CN, - (CR) 2 ) 1-4 NR 2 、-COR、-CONR 2 、-CONR(CR 2 ) 1-4 NR 2 、-NO 2 、-NR 2 、-NR(C 1-6 Haloalkyl), -NRCOR, -NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 、-P(O)R 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 8-10 membered bicyclic aromatic carbocycle, 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur;
R 3 is hydrogen, halogen, -OR OR-OSiR 3
R 3' Is hydrogen, halogen, -OR OR-OSiR 3
Or R is 3 And R is 3' Taken together to form =o or =s;
R 4 and R is 6 Is independently hydrogen, -OR, -NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 An aliphatic;
R 5 and R is 5' Each hydrogen or taken together to form =o or =nor;
X 1 and X 2 Each independently is-CR 2 -, -S-or-S (O) -,
wherein X is 1 And X 2 At least one of them is-CR 2 -。
It should be understood that the term "rapamycin" and its structures described throughout this specification are intended to encompass rapamycin and its analogues.
The rapamycin analogues (i.e., rapalog) described herein are for illustration and are not intended to limit the invention.
As defined aboveSense and as described herein, X and X 3 Independently a covalent bond, -CR 2 -、-NR-、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -, -O-, -S-or-SO 2 NR-。
In some embodiments, X is a covalent bond. In some embodiments, X is-CR 2 -. In some embodiments, X is-NR-. In some embodiments, X is-NRCO-. In some embodiments, X is-NRCO 2 -. In some embodiments, X is-NRCONR-. In some embodiments, X is-NRSO 2 -. In some embodiments, X is-O-. In some embodiments, X is-S-. In some embodiments, X is-SO 2 NR-。
In some embodiments, X 3 Is a covalent bond. In some embodiments, X 3 is-CR 2 -. In some embodiments, X 3 is-NR-. In some embodiments, X 3 is-NRCO-. In some embodiments, X 3 is-NRCO 2 -. In some embodiments, X 3 is-NRCONR-. In some embodiments, X 3 is-NRSO 2 -. In some embodiments, X 3 is-O-. In some embodiments, X 3 is-S-. In some embodiments, X 3 is-SO 2 NR-。
In some embodiments, wherein X is an asymmetric group, such as-NRCO-, -NRCO 2 -、-NRSO 2 -or-SO 2 NR-, X and L 1 Is combined into-NRCOL 1 -、-NRCO 2 L 1 -、-NRSO 2 L 1 -and-SO 2 NRL 1 -。
In some embodiments, wherein X 3 Is an asymmetric group such as-NRCO-, -NRCO 2 -,-NRSO 2 -or-SO 2 NR-,X 3 And R is R 2 Combine to be-NRCOR 2 、-NRCO 2 R 2 、-NRSO 2 R 2 and-SO 2 NRR 2
In some embodiments, X and X 3 Selected from those shown in the compounds of table 1.
L as defined above and described herein 1 Is covalent bond or C 1-30 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of the chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-SO 2 -、-C(O)-、-C(S)-、-CR 2 -、-CF 2 -、-P(O)(R)-、-SiR 2 -, -Si (OR) (R) -OR-NR-substitutions.
In some embodiments, L 1 Is a covalent bond. In some embodiments, L 1 Is C 1-30 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of the chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-SO 2 -、-C(O)-、-C(S)-、-CR 2 -、-CF 2 -、-P(O)(R)-、-SiR 2 -, -Si (OR) (R) -OR-NR-substitutions.
In some embodiments, L 1 is-CH 2 -. In some embodiments, L 1 is-CH 2 CH 2 -. In some embodiments, L 1 Is- (CH) 2 ) 3 -. In some embodiments, L 1 Is- (CH) 2 ) 4 -. In some embodiments, L 1 Is- (CH) 2 ) 5 -. In some embodiments, L 1 is-CH 2 CH 2 O-. In some embodiments, L 1 Is- (CH) 2 CH 2 O) 2 -. In some embodiments, L 1 Is- (CH) 2 CH 2 O) 3 -. In some embodiments, L 1 is-CH 2 CH 2 OCH 2 CH 2 -. In some embodiments, L 1 is-CH 2 CH 2 SO 2 CH 2 CH 2 O-. In some embodiments, L 1 is-CH 2 CH 2 OCH 2 CH 2 OCH 2 CH 2 -. In some embodiments, L 1 is-CH 2 CH 2 OCH 2 CH 2 -。
In some embodiments, L 1 Selected from those shown in the compounds of table 1.
each-Cy as defined above and described herein 1 -independently is an optionally substituted divalent ring selected from phenylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, -Cy 1 -an optionally substituted divalent ring selected from phenylene. In some embodiments, -Cy 1 -an optionally substituted 4-7 membered saturated or partially unsaturated heterocyclyl having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur. In some embodiments, -Cy 1 -is an optionally substituted 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, -Cy 1 -is
In some embodiments, -Cy 1 Selected from those shown in the compounds of table 1.
As defined above and described herein, each R is independently hydrogen or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, or two R groups on the same atom are joined together with the atoms between them to form a 4-7 membered saturated, partially unsaturated or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur in addition to the same atom to which they are attached.
In some embodiments, R is hydrogen. In some embodiments, R is optionally substituted C 1-6 Aliphatic. In some embodiments, R is C 1-6 A haloalkyl group. In some embodiments, R is an optionally substituted 3-8 membered saturated or partially unsaturated monocyclic carbocycle. At the position of In some embodiments, R is optionally substituted phenyl. In some embodiments, R is an optionally substituted 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, two R groups on the same atom are joined together with the atoms between them to form an optionally substituted 4-7 membered saturated, partially unsaturated, or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur in addition to the same atom to which they are attached.
In some embodiments, R is selected from those shown in the compounds of table 1.
L as defined above and described herein 2 Is optionally substituted C 1-6 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1-2 methylene units of the chain are independently and optionally protected by-Cy 1 -substitution.
In some embodiments, L 2 Is optionally substituted C 1-6 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1-2 methylene units of the chain are independently and optionally protected by-Cy 1 -substitution.
In some embodiments, L 2 Is that
In some embodiments, L 2 Selected from those shown in the compounds of table 1.
R is as defined above and described herein 1 And R is 2 Independently hydrogen, halogen, -OR, -CN, - (CR) 2 ) 1-4 NR 2 、-COR、-CONR 2 、-CONR(CR 2 ) 1-4 NR 2 、-NO 2 、-NR 2 、-NR(C 1-6 Haloalkyl), -NRCOR, -NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 、-P(O)R 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 8-10 membered bicyclic aromatic carbocycle, 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R 1 Is hydrogen. In some embodiments, R 1 Is halogen. In some embodiments, R 1 is-OR. In some embodiments, R 1 is-CN. In some embodiments, R 1 is-NO 2 . In some embodiments, R 1 is-NR 2 . In some embodiments, R 1 is-NR (C) 1-6 Haloalkyl). In some embodiments, R 1 is-NRCOR. In some embodiments, R 1 is-NRCO 2 R is defined as the formula. In some embodiments, R 1 is-NRCONR 2 . In some embodiments, R 1 is-NRSO 2 R is defined as the formula. In some embodiments, R 1 is-SR. In some embodiments, R 1 is-SO 2 NR 2 . In some embodiments, R 1 Is optionally substituted C 1-6 Aliphatic. In some embodiments, R 1 Is an optionally substituted 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, R 1 Is optionally substituted phenyl. In some embodiments, R 1 Is an optionally substituted 8-10 membered bicyclic aromatic carbocyclic ring. In some embodiments, R 1 Is an optionally substituted 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur. In some embodiments, R 1 Is an optionally substituted 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur. In some embodiments, R 1 Is an optionally substituted 5-6 membered single having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfurA cyclo-heteroaromatic ring. In some embodiments, R 1 Is an optionally substituted 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R 1 Is methyl. In some embodiments, R 1 is-NH 2 . In some embodiments, R 1 is-NHMe. In some embodiments, R 1 is-NMe 2 . In some embodiments, R 1 is-CH 2 CF 3 . In some embodiments, R 1 is-SO 2 -NH 2 . In some embodiments, R 1 is-CONH 2 . In some embodiments, R 1 is-CONMe 2 . In some embodiments, R 1 is-OCONHMe. In some embodiments, R 1 is-CO 2 H. In some embodiments, R 1 Is thatIn some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->Some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments of the present invention, in some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is thatIn some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->In some embodiments, R 1 Is->/>
In some embodiments, R 2 Is hydrogen. In some embodiments, R 2 Is halogen. In some embodiments, R 12 is-OR. In some embodiments, R 2 is-CN. In some embodiments, R 2 Is- (CR) 2 ) 1-4 NR 2 . In some embodiments, R 2 is-COR. In some embodiments, R 2 is-CONR 2 . In some embodiments, R 2 is-CONR (CR) 2 ) 1-4 NR 2 . In some embodiments, R 2 is-NO 2 . In some embodiments, R 2 is-NR 2 . In some embodiments, R 2 is-NR (C) 1-6 Haloalkyl). In some embodiments, R 2 is-NRCOR. In some embodiments, R 2 is-NRCO 2 R is defined as the formula. In some embodiments, R 2 is-NRCONR 2 . In some embodiments, R 2 is-NRSO 2 R is defined as the formula. In some embodiments, R 2 is-SR. In some embodiments, R 2 is-SO 2 NR 2 . In some embodiments, R 2 is-P (O) R 2 . In some embodiments, R 2 Is optionally substituted C 1-6 Aliphatic. In some embodiments, R 2 Is an optionally substituted 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, R 2 Is optionally substituted phenyl. In some embodiments, R 2 Is an optionally substituted 8-10 membered bicyclic aromatic carbocyclic ring. In some embodiments, R 2 Is an optionally substituted 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur. In some embodiments, R 2 Is an optionally substituted 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur. In some embodiments, R 2 Is an optionally substituted 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur. In some embodiments, R 2 Is an optionally substituted 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur.
In some embodiments, R 2 Is methyl. In some embodiments, R 2 is-CHF 2 . In some embodiments, R 2 is-CF 3 . In some embodiments, R 2 Is thatIn some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is thatIn some embodiments, R 2 Is->In some embodiments, R 2 Is thatIn some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is->
In some embodiments, R 2 Is thatIn some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is->In some embodiments, R 2 Is->
In some embodiments, R 2 Is thatIn some embodiments, R 2 Is->In some embodiments, R 2 Is->
In some embodiments, R 1 And R is 2 Selected from those shown in the compounds of table 1.
R is as defined above and described herein 3 Is hydrogenHalogen, -OR OR-OSiR 3
In some embodiments, R 3 Is hydrogen. In some embodiments, R 3 Is halogen. In some embodiments, R 3 is-OR. In some embodiments, R 3 is-OSiR 3
In some embodiments, R 3 is-OMe.
R is as defined above and described herein 3' Is hydrogen, halogen, -OR OR-OSiR 3
In some embodiments, R 3 Is hydrogen. In some embodiments, R 3 Is halogen. In some embodiments, R 3 is-OR. In some embodiments, R 3 is-OSiR 3
As defined above and described herein, in some embodiments, R 3 And R is 3' Taken together to form =o or =s;
in some embodiments, R 3 And R is 3' Bonded together to form =o. In some embodiments, R 3 And R is 3' Are combined together to form = S.
In some embodiments, R 3 And R is 3' Selected from those shown in the compounds of table 1.
R is as defined above and described herein 4 And R is 6 Is independently hydrogen, -OR, -NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 Aliphatic.
In some embodiments, R 4 Is hydrogen. In some embodiments, R 4 is-OR. In some embodiments, R 4 is-NR 2 . In some embodiments, R 4 is-NRCOR. In some embodiments, R 4 is-NRCO 2 R is defined as the formula. In some embodiments, R 4 is-NRCONR 2 . In some embodiments, R 4 is-NRSO 2 R is defined as the formula. In some embodiments, R 4 is-SR.In some embodiments, R 4 is-SO 2 NR 2 . In some embodiments, R 4 Is optionally substituted C 1-6 Aliphatic.
In some embodiments, R 4 is-OH. In some embodiments, R 4 is-OMe.
In some embodiments, R 6 Is hydrogen. In some embodiments, R 6 is-OR. In some embodiments, R 6 is-NR 2 . In some embodiments, R 6 is-NRCOR. In some embodiments, R 6 is-NRCO 2 R is defined as the formula. In some embodiments, R 6 is-NRCONR 2 . In some embodiments, R 6 is-NRSO 2 R is defined as the formula. In some embodiments, R 6 is-SR. In some embodiments, R 6 is-SO 2 NR 2 . In some embodiments, R 6 Is optionally substituted C 1-6 Aliphatic.
In some embodiments, R 6 is-OMe.
In some embodiments, R 4 And R is 6 Selected from those shown in the compounds of table 1.
R is as defined above and described herein 5 And R is 5' Each hydrogen or combined together to form =o or =nor.
In some embodiments, R 5 Is hydrogen. In some embodiments, R 5' Is hydrogen. In some embodiments, R 5 And R is 5' Bonded together to form =o. In some embodiments, R 5 And R is 5' Combine together to form = NOR.
In some embodiments, R 5 And R is 5' Selected from those shown in the compounds of table 1.
X is as defined above and described herein 1 And X 2 Each independently is-CR 2 -, -S-or-S (O) -, wherein X 1 And X 2 At least one of them is-CR 2 -。
In some embodimentsWherein X is 1 is-CR 2 -. In some embodiments, X 1 is-S-. In some embodiments, X 1 is-S (O) -. In some embodiments, X 2 is-CR 2 -. In some embodiments, X 2 is-S-. In some embodiments, X 2 is-S (O) -.
In some embodiments, X 1 And X 2 Selected from those shown in the compounds of table 1.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -, and L 2 Is as followsThus providing a compound of formula I-a-1:
or a pharmaceutically acceptable salt thereof, wherein:
x is-CR 2 -、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -or-SO 2 NR-; and is also provided with
X 3 、R 1 、R 2 、R 3 、R 3' 、R 4 、R 5 、R 5' 、R 6 And L 1 As defined and described herein, individually and in combination.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -, and L 2 Is as followsThus providing a compound of formula I-a-2:
or a pharmaceutically acceptable salt thereof, wherein:
X 3 is-CR 2 -、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -or-SO 2 NR-; and is also provided with
X、R 1 、R 2 、R 3 、R 3' 、R 4 、R 5 、R 5' 、R 6 And L 1 As defined and described herein, individually and in combination.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -, and L 2 Is as followsThus providing a compound of formula I-a-3: />
Or a pharmaceutically acceptable salt thereof, wherein:
R 4 is-NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 An aliphatic; and is also provided with
X、X 3 、R 1 、R 2 、R 3 、R 3' 、R 5 、R 5' 、R 6 And L 1 As defined and described herein, individually and in combination.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -, and L 2 Is as followsThus providing a compound of formula I-a-4:
or a pharmaceutically acceptable salt thereof, wherein:
R 6 is-NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 An aliphatic; and is also provided with
X、X 3 、R 1 、R 2 、R 3 、R 3' 、R 4 、R 5 、R 5' And L 1 As defined and described herein, individually and in combination.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -,L 2 Is thatX 3 Is a covalent bond, and R 2 Is hydrogen, as shown below, thereby providing a compound of formula I-b-1: />
Or a pharmaceutically acceptable salt thereof, wherein:
X、R 1 、R 3 、R 3' 、R 4 、R 5 、R 5' 、R 6 and L 1 As defined and described herein, individually and in combination.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -,L 2 Is thatR 3 is-OMe, and R 3' Is hydrogen, as shown below, thereby providing a compound of formula I-b-2:
Or a pharmaceutically acceptable salt thereof, wherein:
X、X 3 、R 1 、R 2 、R 4 、R 5 、R 5' 、R 6 and L 1 As defined and described herein, individually and in combination.
In certain embodiments, the present invention provides compounds of formula I or I', wherein X 1 is-CH 2 -,X 2 is-CH 2 -,L 2 Is thatX 3 is-O-, and R 2 Is->As shown below, thereby providing a compound of formula I-b-3:
or a pharmaceutically acceptable salt thereof, wherein:
X、R 1 、R 3 、R 3' 、R 4 、R 5 、R 5' 、R 6 and L 1 As defined and described herein, individually and in combination.
Rapamycin is under the trade name(common name, sirolimus) is marketed and is known for its antiproliferative and immunosuppressive activity. Rapamycin is FDA approved for use in preventing graft rejection and for coating stents to prevent restenosis. In addition to the noted benefits of rapamycinRapamycin is also known to be associated with a number of serious side effects. Such side effects include diabetes-like symptoms of reduced glucose tolerance and reduced insulin sensitivity. In addition, rapamycin has been reported to activate Akt signaling pathways (including activation of Akt and ERK), thereby increasing the risk of cancer in patients.
As used herein, the phrase "rapamycin alone" is intended to compare the compounds of the present invention to rapamycin or an analog thereof (such as everolimus) as an alternative.
In some embodiments, compounds of formula I or I' are provided that are more potent than rapamycin alone.
In some embodiments, compounds of formulas I-a-1 through I-a-5 are provided that are more potent than rapamycin alone.
In some embodiments, compounds of formulas I-a-1 through I-b-3 are provided that are more potent than rapamycin alone.
In some embodiments, compounds of formula I or I' are provided that, when administered to a patient, produce fewer and/or less severe side effects than when rapamycin is administered.
In some embodiments, compounds of formulas I-a-1 through I-a-5 are provided that produce fewer and/or less severe side effects when administered to a patient than when rapamycin is administered.
In some embodiments, compounds of formulas I-a-1 through I-b-3 are provided that produce fewer and/or less severe side effects when administered to a patient than when rapamycin is administered.
Exemplary compounds of the present invention are listed in table 1 below.
TABLE 1 exemplary Compounds
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
In some embodiments, the invention provides a compound listed in table 1 above, or a pharmaceutically acceptable salt thereof. In some embodiments, the present invention provides the compounds listed in table 1 above, or pharmaceutically acceptable salts, stereoisomers, tautomers, and polymorphs thereof. In some embodiments, the invention provides compounds listed in table 1 above, including substitution of one or more hydrogens with deuterium. It is to be understood that the present invention also provides the compounds listed in table 1 above, or pharmaceutically acceptable salts thereof, in the form of a racemic mixture at the C7 position. Furthermore, it is understood that the compounds listed in table 1 above in the form of a racemic mixture at the C7 hydroxyl position may be separated into diastereomers by various methods (e.g., chiral chromatography).
In some embodiments, the invention provides a compound of formula I or I', or a pharmaceutically acceptable salt thereof, wherein when:
L 2 is that
R 3 And R is 3' Is hydrogen;
R 5 and R is 5' Taken together to form =o;
and X is 1 And X 2 Are all-CH 2 -; then:
-X-L 1 -R 1 、-X 3 -R 2 、R 4 and R is 6 In combinations other than those given in each row of table 1A below.
Table 1A.
/>
/>
/>
/>
/>
/>
/>
/>
In some embodiments, the invention provides a compound of formula I or I', wherein the compound is not one or more of the compounds in table 1A. In some embodiments, the present invention provides compounds of formula I or I' that do not include a compound of table 1A, or pharmaceutically acceptable salts, stereoisomers, tautomers, and polymorphs thereof, wherein one or more hydrogens are replaced with deuterium.
In some embodiments, the invention provides compounds of formula I or I' or pharmaceutically acceptable salts, stereoisomers, tautomers, and polymorphs thereof, wherein when:
L 2 is that
R 3 And R is 3' Is hydrogen;
R 5 and R is 5' Taken together to form =o;
and X is 1 And X 2 Are all-CH 2 -; then:
-X-L 1 -R 1 、-X 3 -R 2 、R 4 and R is 6 In combinations other than those given in each row of table 1A below.
4. Use, formulation and administration
Pharmaceutically acceptable compositions
According to another embodiment, the present invention provides a composition comprising a compound of the present invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant or vehicle. The amount of compound in the compositions of the present invention refers to an amount effective to inhibit mTORC1 in a measurable manner in a biological sample or in a patient. In certain embodiments, the amount of a compound in a composition of the invention refers to an amount effective to inhibit mTORC1 in a biological sample or in a patient in a measurable manner. In certain embodiments, the compositions of the present invention are formulated for administration to a patient in need of such compositions. In some embodiments, the compositions of the present invention are formulated for oral administration to a patient.
As used herein, the term "patient" means an animal, preferably a mammal, and most preferably a human.
The term "pharmaceutically acceptable carrier, adjuvant or vehicle" refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that can be used in the compositions of the invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, carboxymethyl cellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and lanolin.
The compositions of the invention may be administered by oral, parenteral, inhalation spray, topical, rectal, nasal, buccal, vaginal route or via an implanted reservoir. As used herein, the term "parenteral" includes subcutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the composition is administered by oral, intraperitoneal or intravenous route. The sterile injectable form of the compositions of the invention may be an aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids such as oleic acid and its glyceride derivatives may be used in the preparation of injectables, as may natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents commonly used in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants, such as tweens, span and other emulsifying agents or bioavailability promoters, commonly used in the preparation of pharmaceutically acceptable solid, liquid or other dosage forms may also be used for formulation purposes.
The pharmaceutically acceptable compositions of the present invention may be administered orally in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. For tablets for oral use, common carriers include lactose and corn starch. A lubricant, such as magnesium stearate, is also typically added. For oral administration in capsule form, useful fillers include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweeteners, flavoring agents or coloring agents may also be added.
Alternatively, the pharmaceutically acceptable compositions of the present invention may be administered in the form of suppositories for rectal administration. These compositions may be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
The pharmaceutically acceptable compositions of the present invention may also be administered topically, especially when the therapeutic target includes an area or organ that is readily accessible by topical administration, including diseases of the eye, skin or lower intestinal tract. Topical formulations suitable for each of these areas or organs are easy to prepare.
Topical application to the lower intestinal tract may be effected in rectal suppository formulations (see above) or in suitable enema formulations. Topical transdermal patches may also be used.
For topical application, the provided pharmaceutically acceptable compositions may be formulated as a suitable ointment containing the active ingredient suspended or dissolved in one or more carriers. Carriers for topical application of the compounds of the invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compounds, emulsifying wax and water. Alternatively, the provided pharmaceutically acceptable compositions may be formulated as suitable lotions or creams containing the active ingredient suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetostearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
For ophthalmic use, the provided pharmaceutically acceptable compositions may be formulated as micronized suspensions of isotonic, pH adjusted sterile saline, or preferably as solutions of isotonic, pH adjusted sterile saline, with or without a preservative such as benzyl dichloromethane. Alternatively, for ophthalmic use, the pharmaceutically acceptable composition may be formulated as an ointment, such as petrolatum.
The pharmaceutically acceptable compositions of the present invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well known in the art of pharmaceutical formulation and may be prepared as saline solutions using benzyl alcohol or other suitable preservatives, absorption promoters for enhanced bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
Most preferably, the pharmaceutically acceptable compositions of the present invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, the pharmaceutically acceptable compositions of the invention are not administered with food. In other embodiments, the pharmaceutically acceptable compositions of the invention are administered with food.
The amount of a compound of the present invention that can be combined with a carrier material to produce a single dosage form of the composition will vary depending upon the host treated, the particular mode of administration. Preferably, the compositions provided should be formulated so that a dosage of 0.01-100mg/kg body weight/day of inhibitor can be administered to a patient receiving these compositions.
It will also be appreciated that the particular dosage and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the particular compound employed, the age, weight, general health, sex, diet, time of administration, rate of excretion, drug combination and the judgment of the treating physician and the severity of the particular disease undergoing therapy. The amount of the compound of the invention in the composition will also depend on the particular compound in the composition.
Use of compounds and pharmaceutically acceptable compositions
As used herein, the term "treating" refers to reversing, alleviating, delaying the onset of, or inhibiting the progression of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, the treatment may be administered after one or more symptoms have occurred. In other embodiments, the treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to onset of symptoms (e.g., based on a history of symptoms and/or based on genetic or other susceptibility factors). Treatment may also be continued after the symptoms have subsided, for example, to prevent or delay recurrence thereof.
The compounds provided are inhibitors of mTORC1 and are therefore useful in the treatment of one or more conditions associated with mTORC1 activity. Thus, in certain embodiments, the present invention provides a method for treating a mTORC1 mediated disorder, the method comprising the step of administering to a patient in need thereof a compound of the present invention, or a pharmaceutically acceptable composition thereof.
As used herein, the term "mTORC 1-mediated" disorder, disease, and/or condition refers to any disease or other detrimental condition in which mTORC1 is known to function. Thus, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which mTORC1 is known to function. In certain embodiments, the mTORC1 mediated disorder, disease, and/or condition is selected from those described by Matt Kaeberlin scientific, vol.2013, arc ID 849186.
The methods described herein include methods for treating cancer in a subject. As used herein, "treating" means alleviating or ameliorating at least one symptom or clinical parameter of cancer. For example, treatment may result in a decrease in tumor size or a decrease in growth rate. In all subjects, treatment did not require 100% cure of cancer or caused remission.
As used herein, the term "cancer" refers to a cell that has the ability to grow autonomously, i.e., an abnormal state or condition characterized by the growth of rapidly proliferating cells. The term is intended to include all types of cancerous growth or oncogenic processes, metastatic tissue, or malignantly transformed cells, tissues, or organs, regardless of the type of histopathology or stage of invasion. As used herein, the term "tumor" refers to cancerous cells, e.g., a mass of cancerous cells.
Cancers that may be treated or diagnosed using the methods described herein include malignancies of various organ systems, such as those affecting the lung, breast, thyroid, lymph, gastrointestinal and genitourinary tracts, and adenocarcinomas including malignancies, such as most colon, renal cell carcinoma, prostate and/or testicular tumors, non-small cell lung cancer, small intestine cancer and esophageal cancer.
In some embodiments, the methods described herein are used to treat or diagnose cancer in a subject. The term "cancer" is art-recognized and refers to malignant tumors of epithelial or endocrine tissues, including cancers of the respiratory system, gastrointestinal system, genitourinary system, testis, breast, prostate, endocrine system and melanoma. In some embodiments, the cancer is renal cancer or melanoma. Exemplary cancers include cancers that form in the tissues of the cervix, lung, prostate, breast, head and neck, colon, and ovary. The term also includes carcinomatous sarcomas, for example, which include malignant tumors composed of cancerous and sarcomatous tissue. "adenocarcinoma" refers to a carcinoma derived from glandular tissue or in which tumor cells form recognizable glandular structures.
The term "sarcoma" is art-recognized and refers to a malignant tumor of mesenchymal origin.
In some embodiments, the cancer treated by the methods described herein is a cancer having increased levels of mTORC1 or expression or activity of mTORC1 relative to normal tissue or other cancers of the same tissue; methods known in the art and described herein can be used to identify these cancers. In some embodiments, the methods comprise obtaining a sample comprising cancer cells, determining mTORC1 activity in the sample, and administering a treatment (e.g., a provided mTORC1 inhibitor) as described herein. In some embodiments, the cancer is a cancer that has an increased level of mTORC1 activity as shown herein.
In some embodiments, the invention provides methods for treating one or more disorders, diseases, and/or conditions, wherein the disorders, diseases, or conditions include, but are not limited to, cell proliferative disorders.
Cell proliferative disorders
The invention features methods and compositions for the diagnosis and prognosis of cell proliferative disorders (e.g., cancer) and the treatment of such disorders by inhibiting mTORC1 activity. Cell proliferative disorders described herein include, for example, cancer, obesity, and proliferation-dependent diseases. Such conditions may be diagnosed using methods known in the art.
Cancer of the human body
Cancers include, but are not limited to, leukemia (e.g., acute leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphomas (e.g., hodgkin's or non-hodgkin's disease), fahrenheit, multiple myeloma, heavy chain diseases, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary adenocarcinomas, cystic adenocarcinoma, medullary carcinoma, bronchi carcinoma, renal cell carcinoma, liver cancer, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, wilms' tumor, cervical cancer, uterine cancer, testicular cancer, lung cancer, small lung cell carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, neuroblastoma, craniopharyngema, ependymoma, pineal tumor, hemangioblastoma, auditory glioma, oligodendroglioma, neuroblastoma, meningioma, melanoma, neuroblastoma, and retinoblastoma. In some embodiments, the cancer is melanoma or breast cancer.
Fibrotic diseases
The Idiopathic Pulmonary Fibrosis (IPF) PI3K pathway is activated in fibrotic lesions (fundamental lesions in IPF). mTOR kinase inhibitor GSK2126458 reduces PI3K pathway signaling and functional response in IPF-derived lung fibroblasts, and mTOR inhibition reduces collagen expression in IPF patient models. In the bleomycin model of pulmonary fibrosis, rapamycin treatment is anti-fibrotic and rapamycin also reduces the expression of α -smooth muscle actin and fibronectin by fibroblasts in vitro.
In some embodiments, idiopathic Pulmonary Fibrosis (IPF) is treated using methods that inhibit mTorrC 1 activity (see Mercer, P.F. et al, thorax., volume 71, 8:701-711 (2016), patel, A.S. et al, PLoS One, volume 7, 7:e 41394 (2012)). Thus, in some embodiments, the present invention provides a method of treating Idiopathic Pulmonary Fibrosis (IPF) in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
Renal fibrosis mTORC1 is activated in myofibroblasts (the primary pathogenic cell type in renal fibrosis). Inhibition of mTOR with rapamycin in a kidney fibrosis mouse model (UUO) attenuated the expression of markers of fibrosis and tubular interstitial injury.
In some embodiments, renal fibrosis is treated using a method of inhibiting mTORC1 activity (see, e.g., jiang, l. Et al, J Am Soc Nephrol, volume 24, 7: pages 1114-1126 (2013), wu, m.j. Et al, kidney International, volume 69, 11: pages 2029-2036 (2006), chen, g. Et al, PLoS One: volume 7, 3: page E33626 (2012)), liu, c.f. et al, clin Invest Med, volume 37, 34: pages E142-E153 (2014)). Thus, in some embodiments, the present invention provides a method of treating renal fibrosis in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
In some embodiments, scleroderma is treated using a method of inhibiting mTORC1 activity (see Mitra, a. Et al, J Invest Dermatol., volume 135, 11: pages 2873-2876 (2015)). Thus, in some embodiments, the present invention provides a method of treating scleroderma in a patient in need thereof, comprising the step of administering to said patient the provided compound or pharmaceutically acceptable salt thereof.
In some embodiments, hypertrophic scarring and keloid disease are treated using methods of inhibiting mTORC1 activity (see Syed, f. Et al, am J pathol., volume 181, 5: pages 1642-1658 (2012)). Thus, in some embodiments, the present invention provides a method of treating hypertrophic scarring and keloid disease in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
In some embodiments, cardiac fibrosis is treated using a method of inhibiting mTORC1 activity (see, e.g., yano, t. Et al, J Mol Cell cardiol., volume 91: pages 6-9 (2016)). Accordingly, in some embodiments, the present invention provides a method of treating cardiac fibrosis in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
Other proliferative diseases
Other proliferative disorders include, for example, obesity, benign prostatic hyperplasia, psoriasis, dysplasia, lymphoproliferative disorders (e.g., disorders of abnormal proliferation of lymphocytes), chronic rheumatoid arthritis, atherosclerosis, restenosis, and diabetic retinopathy. Proliferative diseases incorporated herein by reference include those described in U.S. patent nos. 5,639,600 and 7,087,648.
Other disorders
Other conditions include lysosomal storage diseases including, but not limited to, pompe disease, gaucher disease, mucopolysaccharidosis, and multiple sulfatase deficiency; neurodegenerative diseases such as Parkinson's disease, alzheimer's disease, huntington's disease, alpha 1-antitrypsin deficiency and spinal bulbar muscular atrophy.
In some embodiments, asthma is treated using methods of inhibiting mTORC1 activity (see Hua, w. et al, virology, volume 20, 7: pages 1055-1065 (2015)). Thus, in some embodiments, the present invention provides a method of treating asthma in a patient in need thereof, the method comprising the step of administering to said patient a provided compound or a pharmaceutically acceptable salt thereof.
In some embodiments, the lysosomal storage disease is treated using a method of inhibiting mTORC1 activity (see Sardiello, m., annals of the New York Academy of Sciences, volume 1371, pages 3-14 (2016), awad, o, et al, volume 24, page 20, pages 5775-5788 (2015), spampanato, c, et al, EMBO Mol med, volume 5, pages 691-706 (2013), medina, d.l., dev cell, volume 21, page 3, pages 421-430 (2011)). Thus, in some embodiments, the present invention provides a method of treating a lysosomal storage disease in a patient in need thereof, the method comprising the step of administering to said patient the provided compound or pharmaceutically acceptable salt thereof.
In some embodiments, parkinson's disease is treated using methods that inhibit mTorrC 1 activity (see Decressac, M. Et al Proc Natl Acad Sci U S A., vol. 110, 19: pages E1817-E1826 (2013)). Thus, in some embodiments, the present invention provides a method of treating parkinson's disease in a patient in need thereof, the method comprising the step of administering to said patient the provided compound or pharmaceutically acceptable salt thereof.
In some embodiments, alzheimer's disease is treated using methods of inhibiting mORC 1 activity (see Polio, V.A. et al, EMBO Mol Med., vol. 6, 9: pages 1142-1160 (2014)). Thus, in some embodiments, the present invention provides a method of treating alzheimer's disease in a patient in need thereof, the method comprising the step of administering to said patient a provided compound or a pharmaceutically acceptable salt thereof.
In some embodiments, huntington's disease is treated using a method of inhibiting mTORC1 activity (see Tsunemi, t. Et al, sci trans l med., volume 4, 142 th, page 142ra97 (2012)). Thus, in some embodiments, the present invention provides a method of treating huntington's disease in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or pharmaceutically acceptable salt thereof.
In some embodiments, alpha 1-antitrypsin deficiency is treated using methods of inhibiting mTORC1 activity (see Pastore, n. Et al, EMBO Mol med., volume 5, 3: pages 397-412 (2013)). Accordingly, in some embodiments, the present invention provides a method of treating alpha 1-antitrypsin deficiency in a patient in need thereof, the method comprising the step of administering to the patient the provided compound or a pharmaceutically acceptable salt thereof.
In some embodiments, spinal and bulbar muscular atrophy is treated using methods that inhibit mTORC1 activity (see cores, c.j. Et al, nat neurosci., volume 17, 9: pages 1180-1189 (2014)). Accordingly, in some embodiments, the present invention provides a method of treating spinal bulbar muscular atrophy in a patient in need thereof, the method comprising the step of administering the provided compound or a pharmaceutically acceptable salt thereof to the patient.
In some embodiments, a method of inhibiting mTORC1 activity is used to treat Fragile X Syndrome (FXS), amyotrophic Lateral Sclerosis (ALS), epilepsy, focal Cortical Dysplasia (FCD), hemimegabrain deformity (HME), familial focal epilepsy with variable origins (FFEV), temporal Lobe Epilepsy (TLE), spasticity, neurodegenerative disorders, down's syndrome, rett syndrome (RTS), or a disease associated with activation or overactivation of mTOR signaling in the brain.
In some embodiments, the invention provides a method of treating Fragile X Syndrome (FXS) in a patient in need thereof, the method comprising administering a compound of the invention or a pharmaceutically salt thereof.
In some embodiments, the invention provides methods of treating Amyotrophic Lateral Sclerosis (ALS) in a patient in need thereof, the methods comprising administering a compound of the invention, or a pharmaceutically salt thereof.
In some embodiments, the present invention provides methods of treating epilepsy in a patient in need thereof, comprising administering a compound of the present invention, or a pharmaceutically salt thereof.
In some embodiments, the invention provides a method of treating Focal Cortical Dysplasia (FCD) in a patient in need thereof, the method comprising administering a compound of the invention or a pharmaceutically salt thereof.
In some embodiments, the invention provides a method of treating hemilateral megabrain deformity (HME) in a patient in need thereof, the method comprising administering a compound of the invention or a pharmaceutically salt thereof.
In some embodiments, the invention provides a method of treating Familial Focal Epilepsy (FFEV) with variable foci in a patient in need thereof, the method comprising administering a compound of the invention, or a pharmaceutically salt thereof.
In some embodiments, the invention provides a method of treating Temporal Lobe Epilepsy (TLE) in a patient in need thereof, comprising administering a compound of the invention, or a pharmaceutically salt thereof.
In some embodiments, the present invention provides methods of treating spasticity in a patient in need thereof, the methods comprising administering a compound of the invention or a pharmaceutically salt thereof.
In some embodiments, the present invention provides a method of treating a neurodegenerative disease in a patient in need thereof, the method comprising administering a compound of the present invention or a pharmaceutically salt thereof.
In some embodiments, the present invention provides a method of treating down syndrome in a patient in need thereof, the method comprising administering a compound of the present invention or a pharmaceutically salt thereof.
In some embodiments, the invention provides a method of treating rett syndrome (RTS) in a patient in need thereof, the method comprising administering a compound of the invention, or a pharmaceutically salt thereof.
In some embodiments, the invention provides a method of treating a disease associated with activation or overactivation of mTOR signaling in the brain in a patient in need thereof, the method comprising administering a compound of the invention or a pharmaceutically salt thereof.
In some embodiments, the compounds of the invention bind to FKBP12 to form a complex. In some embodiments, the complex between the compound of the invention and FKBP12 interacts with the FK 506-rapamycin binding domain of mTOR.
In some embodiments, the compounds of the invention bind to FKBP12 and interfere with protein-protein interactions between FRAP and FKBP 12. In some embodiments, R of the compounds of the invention 1 The group interacts with both FRAP and FKBP 12.
The present invention provides compounds that are inhibitors of mTORC1 activity and that demonstrate a higher selectivity for inhibiting mTORC1 than mTORC2, as measured by pS6K inhibition (a measure of mTORC1 activity) and pAKT activation (a measure of mTORC2 activity). In some embodiments, the provided compounds inhibit mTORC1 with higher selectivity than mTORC2. In some embodiments, the provided compounds do not inhibit mTORC2 in a measurable manner. In some embodiments, compounds are provided having>pAKT activation IC of 10. Mu.M 50 . In some embodiments, the compounds provided are as follows>A 10-fold selectivity for mTORC2 inhibits mTORC1. In some embodiments, the compounds provided are as follows>A 20-fold selectivity for mTORC2 inhibits mTORC1. In some embodiments, the compounds provided are as follows>Selective inhibition of mTORC2 by 50-foldmTORC1. In some embodiments, the compounds provided are as follows>The 100-fold selectivity of mTORC2 inhibits mTORC1. In some embodiments, the compounds provided are as follows>The 150-fold selectivity of mTORC2 inhibits mTORC1. In some embodiments, the compounds provided are as follows>A 200-fold selectivity for mTORC2 inhibits mTORC1. In some embodiments, the compounds provided are as follows >A 500-fold selectivity for mTORC2 inhibits mTORC1. In some embodiments, the compounds provided are as follows>A 1,000-fold selectivity for mTORC2 inhibits mTORC1.
In some embodiments, the provided compounds inhibit mTORC1 with higher selectivity than mTORC2 after prolonged treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 24 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 36 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 48 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 72 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 96 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 120 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about 144 hours of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after about one week of treatment or exposure. In some embodiments, the provided compounds inhibit mTORC1 with a higher selectivity than mTORC2 after treatment or exposure for more than about one week.
In some embodiments, the compounds provided are less immunosuppressive than existing rapalog. In some embodiments, the compounds provided are less immunosuppressive than rapamycin. In some embodiments, the compounds provided are less immunosuppressive than everolimus. In some embodiments, the compounds provided are less immunosuppressive than temsirolimus (temsirolimus). In some embodiments, the compounds provided are less immunosuppressive than lidaforolimus (ridaforolimus). In some embodiments, the compounds provided are less immunosuppressive than that of umimony (umirolimus).
In some embodiments, compounds are provided that are less inhibitory to interferon gamma (IFN-gamma) production than rapalog. In some embodiments, compounds are provided that are less inhibitory to IFN-gamma production than rapamycin. In some embodiments, the compounds provided are less inhibitory to IFN- γ production than everolimus. In some embodiments, the provided compounds are less inhibitory to IFN- γ production than temsirolimus. In some embodiments, the provided compounds are less inhibitory to IFN- γ production than lidaformmus. In some embodiments, the provided compounds have less inhibition of IFN-gamma production than does Ulmoseltamium.
In some embodiments, the provided compounds reduce the expression of a fibrosis biomarker in damaged tissue. In some embodiments, the provided compounds reduce the expression of type I collagen (COL 1 A2) in damaged tissue. In some embodiments, the provided compounds reduce the expression of collagen type III (COL 3 A1) in damaged tissue. In some embodiments, the provided compounds reduce the expression of fibronectin (FN 1) in damaged tissue.
In some embodiments, the provided compounds reduce the propensity of immune cells to infiltrate damaged tissue. In some embodiments, the provided compounds reduce the propensity of macrophages to infiltrate damaged tissue.
In some embodiments, compounds are provided that induce less glucose tolerance than rapalog. In some embodiments, compounds are provided that induce glucose tolerance less than rapamycin. In some embodiments, the provided compounds induce a glucose tolerance that is lower than everolimus. In some embodiments, the provided compounds induce a glucose tolerance that is lower than that of temsirolimus. In some embodiments, the provided compounds induce a glucose tolerance that is lower than that of lidaafolimus. In some embodiments, compounds are provided that induce glucose tolerance less than that of lamimus. In some embodiments, the provided compounds do not induce significantly more glucose tolerance than placebo or vehicle alone.
Thus, in some embodiments, the invention provides a method of treating a disorder associated with mTORC1, the method comprising administering a compound that inhibits mTORC1 to a patient, wherein the compound does not inhibit mTORC2. Such compounds are useful for indications where rapamycin and rapalog show benefit in animal models or in human disease settings. Such indications include:
treatment of metabolic diseases (obesity and insulin resistance in type 2 diabetes) inhibits the mTORC1 pathway resulting in extended life of yeasts, flies and mice, and limiting calories increases life and increases insulin sensitivity. Basic mechanisms have been proposed to function by modulating mTORC1 activation. Rapamycin-induced insulin resistance has been demonstrated to be mediated by inhibition of mTORC2, and selective mTORC1 inhibitors are expected to improve insulin sensitivity and glucose homeostasis.
In some embodiments, metabolic diseases (obesity and insulin resistance in type 2 diabetes) are treated using methods of inhibiting mTORC1 activity (see Yu, z., et al, J Gerontol A Biol Sci Med Sci, volume 70, 4: pages 410-420 (2015), fok, w.c., et al, aging Cell, volume 13, 2: pages 311-319 (2014), shum, m., et al, diabetes, volume 59, volume 3: pages 592-603 (2016), lamming, d.w., et al, science, volume 335, volume 6076: pages 1638-1643 (2012)). Thus, in some embodiments, the present invention provides a method of treating metabolic disorders (obesity and insulin resistance in type 2 diabetes) in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or pharmaceutically acceptable salt thereof.
Neurofibromatosis disease. Type 1 neurofibromatosis (NF 1) is caused by mutations in the NF1 gene. The protein product neurofibromatosis protein thereof is used as a tumor inhibitor and eventually produces constitutive upregulation of mTOR.mTOR inhibitors have been shown to reduce tumor size and induce antiproliferative effects in NF 1-related plexiform neurofibromas.
In some embodiments, neurofibromatosis is treated using methods that inhibit mTORC1 activity (see Franz, d.n. et al Curr Neurol Neurosci rep., volume 12, 3: pages 294-301 (2012)), varin, j. Et al, oncotarget, volume 7, pages 35753-35767 (2016)). Thus, in some embodiments, the present invention provides a method of treating neurofibromatosis in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
-/- Cardiomyopathy and skeletal muscular dystrophy, and an angstrom-deluxe muscular dystrophy model (LMNA). Mutations in LMNA lead to several human diseases including limb banding muscular dystrophy (LGMD 1B), adelomeful muscular dystrophy (EDMD 2/3), dilated Cardiomyopathy (DCM) and conduction system disease (CMD 1A), lipodystrophy, fibular muscular atrophy and hakinsen-Ji Erfu de early senescence syndrome (HGPS). Lmna -/- Mice have increased mTORC1 activity and are treated with rapamycin for Lmna -/- Short term treatment of mice resulted in reduced mTORC1 signaling, improved cardiac and skeletal muscle function, and an increase in survival of about 50%.
In some embodiments, cardiomyopathy and skeletal muscular dystrophy are treated using methods that inhibit mTORC1 activity (see Ramos, f. Et al, sci trans l med., volume 4, 144 th, page 144ra103 (2012), bonne, g. And Quijano-Roy, s., handb Clin neurol., volume 113, pages 1367-1376 (2013)). Thus, in some embodiments, the present invention provides a method of treating cardiomyopathy and skeletal muscular dystrophy in a patient in need thereof comprising the step of administering to the patient the provided compound or pharmaceutically acceptable salt thereof.
Lishi syndrome. Ndefs 4 Knockout (KO) mice were used as a model for Litsea syndrome and exhibited excessive activation and metabolic defects of mTORC 1. Treatment of Ndefs 4 KO mice with rapamycin extends the life cycle and improves metabolism and neurological deficit associated with the diseaseAnd (5) sinking.
In some embodiments, methods of inhibiting mTorrC 1 activity are used to treat Living syndrome (see Johnson, S.C. et al, science, volume 342, 6165:1524-1528 (2013)). Thus, in some embodiments, the present invention provides a method of treating li's syndrome in a patient in need thereof, the method comprising the step of administering to the patient the provided compound or a pharmaceutically acceptable salt thereof.
Oncology. Inhibition of mTOR with rapalog has been demonstrated to have anti-tumor activity in murine models of cancer and cancer patients. Examples of sensitive cancer types include, but are not limited to, hepatocellular carcinoma, breast cancer, mantle cell lymphoma, lung cancer, tuberous sclerosis, and lymphangioleiomyomatosis.
In some embodiments, methods of inhibiting mTORC1 activity are used to treat cancers and neoplastic disorders (see Ilagan, e. And manning, b.d., trends Cancer, volume 2, 5: pages 241-251 (2016)). Thus, in some embodiments, the present invention provides methods of treating cancer and neoplastic disorders in a patient in need thereof, the method comprising the step of administering to the patient the provided compounds or pharmaceutically acceptable salts thereof.
Nonalcoholic steatohepatitis (NASH). The present invention provides inhibitors that induce autophagy to clear degraded cytoplasmic proteins, and NASH disease is characterized by lipid deposition, inflammation and fibrosis in the liver. Inhibition of the mTORC1 pathway induces autophagy and down-regulates SREBP-1 to reduce lipid biosynthesis, thereby reducing lipid storage.
In some embodiments, non-alcoholic steatohepatitis (NASH) is treated using methods that inhibit mTORC1 activity (see pure, p. And Chandra, a., J Clin Exp Hepatol, volume 4, phase 1: pages 51-59 (2014)). Thus, in some embodiments, the present invention provides a method of treating non-alcoholic steatohepatitis (NASH) in a patient in need thereof, the method comprising the step of administering to said patient the provided compound or pharmaceutically acceptable salt thereof.
Tuberous Sclerosis (TSC) and Lymphangioleiomyomatosis (LAM). Failure of mTOR regulation is critical to the pathogenesis of the genetic disorder Tuberous Sclerosis (TSC) and associated lung disease, lymphangioleiomyomatosis (LAM). Both diseases are caused by mutations in either TSC1 or TSC2, which result in inappropriate activity of signaling downstream of mTORC 1. Non-malignant tumors appear in many organs (including the brain) of TSC patients, while certain organs or tissues of LAM patients (mainly females), especially the lung, lymph nodes and kidneys accumulate abnormal muscle-like cells. rapalog, everolimus and sirolimus are currently approved by the U.S. FDA for treatment of both TSC and LAM, respectively.
In some embodiments, methods of inhibiting mORC 1 activity are used to treat tuberous sclerosis and lymphangioleiomyomatosis (see Wander, S.A. et al, J.Clin. Invest., volume 121, p.4:1231-1241 (2011); taveira-DaSilva, A.M. and Moss, J., J.Clin epidemic, volume 7:249-257 (2015)). Thus, in some embodiments, the present invention provides a method of treating tuberous sclerosis and lymphangioleiomyomatosis in a patient in need thereof, the method comprising the step of administering to said patient a provided compound or a pharmaceutically acceptable salt thereof.
Aging and aging diseases. Rapamycin inhibits mammalian TORC1 complexes that regulate translation and extend the life cycle of various species, including mice. Rapamycin has been shown to inhibit the pro-inflammatory phenotype of senescent cells. With age, senescent cells accumulate, and the senescence-associated secretory phenotype (SASP) can destroy tissues and lead to age-related conditions, including cancer. Inhibition of mTOR inhibits senescent cells from secreting inflammatory cytokines. Rapamycin reduces cytokine levels, including IL6, and inhibits translation of the membrane-bound cytokine IL 1A. The reduction of IL1A impairs NF- κB transcriptional activity, thereby controlling SASP. Thus, mTORC1 inhibitors can ameliorate age-related conditions, including advanced cancers, by inhibiting aging-related inflammation.
In some embodiments, methods of inhibiting mORC 1 activity are used to treat aging and aging disorders (see Laberge, R.M. et al, nature Cell Biology, vol.17, 8: pages 1049-1061 (2015); nacarelli, T. Et al, free radio Biol Med., vol.95: pages 133-154 (2016)). Thus, in some embodiments, the present invention provides methods of treating aging and aging disorders in a patient in need thereof, comprising the step of administering to said patient a provided compound or a pharmaceutically acceptable salt thereof.
Diabetic nephropathy and kidney-related complications of type 1 diabetes and type 2 diabetes. Diabetic nephropathy is a renal complication of type 1 and type 2 diabetes mellitus, affected by which up to approximately 40% of diabetics. High levels of glucose force the kidneys to work excessively to filter the blood, resulting in kidney damage. Studies have shown that the mTOR pathway is highly active in diabetic neuropathy patients and can play a role in pathological changes and renal dysfunction caused by chronic high glucose. In addition, mTOR inhibition may alleviate hyperinsulinemia.
In some embodiments, a method of inhibiting mTORC1 activity is used to treat diabetic nephropathy or kidney-related complications of type 1 and type 2 diabetes (see Mori, h. Et al, biochem. Res. Commun. Volume 384, 4: pages 471-475 (2009)). Thus, in some embodiments, the present invention provides a method of treating diabetic nephropathy or kidney-related complications in a patient in need thereof, the method comprising the step of administering to the patient the provided compound or pharmaceutically acceptable salt thereof.
Polycystic kidney disease. Polycystic Kidney Disease (PKD) is characterized by the appearance and accumulation of destructive kidney cysts, ultimately leading to renal failure. PKD may be autosomal dominant (ADPKD) or Autosomal Recessive (ARPKD). Dysfunctional mTOR signaling pathways were observed in ADPKD and ARPKD. Thus, normalization of the mTORC1 pathway may improve cyst development and disease progression.
In some embodiments, PKD is treated using a method of inhibiting mORC 1 activity (see Torres, V.E. et al, clin.J.am. Soc. Nephrol. Vol. 5, 7: pages 1312-1329 (2010)). Thus, in some embodiments, the present invention provides a method of treating PKD in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof. In some embodiments, PKD is autosomal dominant. In some embodiments, the PKD is autosomal recessive.
Focal Segmental Glomerulosclerosis (FSGS) and other diseases associated with renal sclerosis. FSGS is the most common primary glomerular disorder that causes End Stage Renal Disease (ESRD) in the united states. As the disease progresses, podocytes in the bowden cell do not match the surface area of the glomerular basement membrane they cover. Studies have shown that control of podocyte size is regulated by mTOR and that mTOR activation promotes disease progression. Furthermore, in mouse knockdown experiments, constitutive mTORC1 activation has been shown to cause FSGS-like lesions. Thus, mTORC1 inhibition can improve (FSGS) or other diseases associated with kidney cirrhosis by normalizing or increasing autophagy activity.
In some embodiments, methods of inhibiting mTORC1 activity are used to treat FSGS or other diseases associated with kidney cirrhosis (see zschidrich, s. Et al, j.am. Soc. Nephrol. Volume 28, 7: pages 2144-2157 (2017)). Thus, in some embodiments, the present invention provides a method of treating FSGS or other diseases associated with kidney cirrhosis in a patient in need thereof, the method comprising the step of administering to said patient the provided compound or pharmaceutically acceptable salt thereof.
Age-related macular degeneration. Age-related macular degeneration (AMD) is a major cause of blindness characterized by photoreceptor death in the macula. Possible mechanisms of AMD progression include oxidative stress leading to protein deposition and organelle dysfunction, leading to retinal pigment epithelial cell hypertrophy, dedifferentiation, and finally atrophy. mTOR is involved in the dedifferentiation of retinal pigment epithelial cells. Thus, mTORC1 inhibition can improve AMD by blocking hypertrophy and dedifferentiation.
In some embodiments, age-related macular degeneration is treated using methods that inhibit mTORC1 activity (see Kolosova, n.g., et al, am.j.path. 181, volume 2: pages 472-477 (2012) and Zhen, c. And Vollrath, d., aging, volume 3, volume 4: 346-347 (2011)). Accordingly, in some embodiments, the present invention provides a method of treating age-related macular degeneration in a patient in need thereof, the method comprising the step of administering to the patient the provided compound or a pharmaceutically acceptable salt thereof.
Diabetic macular edema. Diabetic Macular Edema (DME) is a major cause of blindness in diabetics, with about 35% of diabetics affected by it. Studies have shown that the pathogenesis of DME is an inflammatory disease involving various cytokines and chemokines. Chronic inflammation and oxidative stress can promote the progression of DME. Thus, inhibition of mTORC1 can improve DME symptoms and progression by reducing inflammatory responses.
In some embodiments, DME is treated using methods that inhibit mORC 1 activity (see Okamoto, T. Et al, PLOS ONE, vol.11, vol.1: page e0146517,https://doi.org/10.1371/ journal.pone.0146517(2016)). Thus, in some embodiments, the present invention provides a method of treating DME in a patient in need thereof, the method comprising the step of administering to said patient a provided compound or a pharmaceutically acceptable salt thereof.
Diabetic retinopathy. Diabetic Retinopathy (DR) is a common ocular disease that accounts for about 5% of adult blindness and is associated with chronic hyperglycemia and defects in the insulin signaling pathway. Retinal blood vessels and neurons of DR patients are continually damaged by inflammation, reactive oxygen species and endoplasmic reticulum stress caused by chronic hyperglycemia. Notably, rapamycin has been shown to block the effects of insulin-induced hypoxia-inducible factor-1 (HIF-1) and retinal cell senescence and to induce autophagy, and may be beneficial in promoting apoptosis of new blood vessels and preventing angiogenesis. Thus, inhibition of mTORC1 can improve DR symptoms and progression by reducing inflammation and inhibiting pathogenic signaling pathways.
In some embodiments, DR is treated using a method that inhibits mTorrC 1 activity (see Di Rosa, M. Et al, curr. Neurobaracol. Vol. 14, 8: pages 810-825 (2016)). Thus, in some embodiments, the present invention provides a method of treating DR in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or pharmaceutically acceptable salt thereof.
Glaucoma. Glaucoma is a common optic neuropathy associated with aging and elevated intraocular pressure and is a major cause of irreversible blindness. Studies have shown that mTOR-dependent autophagy dysfunction may be a factor in the progression of the disease. Thus, inhibiting mTORC1 can slow the progression of glaucoma or improve glaucoma by normalizing or increasing autophagy.
In some embodiments, glaucoma is treated using methods that inhibit mTORC1 activity (see Porter, k. Et al, biochem. Biophys. Acta. Volume 1852, 3: pages 379-385 (2014)). Accordingly, in some embodiments, the present invention provides a method of treating glaucoma in a patient in need thereof, the method comprising the step of administering to the patient the provided compound or a pharmaceutically acceptable salt thereof.
Restoring immune function. mTORC1 inhibition has been shown to reduce CD4 + And CD8 + Expression of the programmed death-1 (PD-1) receptor in T lymphocytes promotes T cell signaling. Thus, mTORC1 inhibition can restore immune function by improving the adaptive immune response.
In some embodiments, the immune function is restored using a method that inhibits mTORC1 activity (see Mannick, j.b. et al, sci. Trans. Med. Volume 6, 268: p ra179 (2014)). Thus, in some embodiments, the present invention provides a method of restoring immune function to a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
Treatment of respiratory tract and/or urinary tract infections. mTORC1 inhibition can reduce infection by up-regulating antiviral gene expression and response. Thus, mTORC1 inhibition may enhance the ability of the patient's immune system to resist respiratory and/or urinary tract infections.
In some embodiments, the respiratory tract and/or urinary tract infections are treated using a method of inhibiting mTORC1 activity. (see Mannich, J.B. et al, sci.Trans. Med. Vol. 10, 449: eaaq1564 (2018)). Thus, in some embodiments, the present invention provides a method of restoring immune function to a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
Heart failure. mTORC1 activity is critical for stress-induced cardiac hypertrophy, and after infarction can lead to cardiac disorders due to cardiac remodeling. Inhibition of mTORC1 reduces pressure overload-induced cardiac remodeling and heart failure. Thus, inhibition of mTORC1 may reduce heart failure in patients with myocardial damage.
In some embodiments, heart failure is treated using methods that inhibit mTORC1 activity (see Sciarretta, S. Et al, circ. Res. Volume 122, 3: pages 489-505 (2018)). Accordingly, in some embodiments, the present invention provides a method of treating heart failure in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
Osteoarthritis. Osteoarthritis (OA) is a chronic degenerative disease that leads to cartilage loss and joint inflammation. mTOR may play an important role in collagen homeostasis as well as in the turnover and remodeling of cartilage. Thus, inhibiting mTORC1 may slow the progression of or ameliorate the symptoms of osteoarthritis by normalizing cartilage turnover.
In some embodiments, osteoarthritis is treated using a method of inhibiting mTORC1 activity (see Pal, b. Et al, drugs R & D, volume 15, phase 1: pages 27-36 (2017)). Accordingly, in some embodiments, the present invention provides a method of treating osteoarthritis in a patient in need thereof, the method comprising the step of administering to the patient a provided compound or a pharmaceutically acceptable salt thereof.
Pulmonary hypertension. Pulmonary Arterial Hypertension (PAH) is a progressive fatal disease associated with increased pulmonary vascular resistance. Pulmonary artery smooth muscle cell proliferation and migration involves the progression of arterial wall thickening, exacerbating vasoconstriction. Thus, inhibition of mTORC1 can be alleviated by reducing vascular remodelingPAH。
In some embodiments, PAH is treated using a method of inhibiting mTORC1 activity (see Ma, x. Et al, inter. Cardioview. Thorac. Surg., volume 25, phase 2: pages 206-211 (2017)). Thus, in some embodiments, the present invention provides a method of treating PAH in a patient in need thereof, the method comprising the step of administering to said patient the provided compound or a pharmaceutically acceptable salt thereof.
Chronic obstructive pulmonary disease. Autophagy is reduced resulting in the accumulation of proteins and other cellular material that accelerate cellular senescence in patients with Chronic Obstructive Pulmonary Disease (COPD). Thus, inhibiting mTORC1 may slow the progression of COPD symptoms or ameliorate COPD symptoms by normalizing or increasing autophagy.
In some embodiments, COPD is treated using methods that inhibit mTORC1 activity (see Fujii, s. Et al, oncoimmunology, volume 1, 5: pages 630-641 (2012)). Thus, in some embodiments, the present invention provides a method of treating COPD in a patient in need thereof, the method comprising the step of administering to said patient a provided compound or a pharmaceutically acceptable salt thereof.
Additional therapeutic indications for which mTORC inhibition may be beneficial are: cardiovascular disease (acute coronary syndrome), coronary occlusion using eluting stents, polycystic kidney disease and kidney disease associated with cyst formation or cyst formation, neurofibromatosis, epilepsy associated with TSC1 and/or TSC2 mutations, polycystic liver disease, congenital creutzfeld-jakob syndrome, fragile X syndrome, friedel ataxia, petzfeldt-jakob syndrome, ocular disease (including neovascular age-related macular degeneration, uveitis, diabetic macular edema), fibroblast growth (including pulmonary fibrosis, renal insufficiency/fibrosis), metabolic syndrome, immune system disease (including immunosenescence, lupus nephritis, chronic immune thrombocytopenia, multiple sclerosis), cancer (including lymphomas, tumors associated with TSC1/2 mutations, vascular fibromas associated with TSC1/2 mutations, breast cancer, hepatocellular carcinoma, leukemia, glioma, adenoid cystic carcinoma), aging, autism, and vascular rheumatoid arthritis.
In some embodiments, cardiovascular disease (acute coronary syndrome), coronary occlusion using eluting stents, polycystic kidney disease, neurofibromatosis, epilepsy associated with TSC1 and/or TSC2 mutations, polycystic liver disease, congenital thick-nail disease, fragile X syndrome, friedel's ataxia, petzfeldt-jakob syndrome, ocular disease (including neovascular age-related macular degeneration, uveitis, diabetic macular edema), fibroblast growth (including pulmonary fibrosis, renal insufficiency/fibrosis), metabolic syndrome, immune system disease (including immune aging, lupus nephritis, chronic immune thrombocytopenia, multiple sclerosis), cancer (including lymphomas, tumors associated with TSC1/2 mutations, vascular myolipomas associated with TSC1/2 mutations, breast cancer, hepatocellular carcinoma, leukemia, glioma, adenoid cystic carcinoma), aging, autoimmune arthritis, and vascular rheumatoid arthritis are treated using methods of inhibiting mTORC1 activity.
Thus, in some embodiments, the invention provides methods of treating cardiovascular disease (acute coronary syndrome), coronary occlusion using eluting stents, polycystic kidney disease, neurofibromatosis, epilepsy associated with TSC1 and/or TSC2 mutations, polycystic liver disease, congenital thick nail disease, fragile X syndrome, friedel ataxia, peter-jetty syndrome, ocular disease (including neovascular age-related macular degeneration, uveitis, diabetic macular edema), fibroblast growth (including pulmonary fibrosis, renal insufficiency/fibrosis), metabolic syndrome, immune system disease (including immunosenescence, lupus nephritis, chronic immune thrombocytopenia, multiple sclerosis), cancer (including lymphoma, tumors associated with TSC1/2 mutations, vascular myolipomas associated with TSC1/2 mutations, breast cancer, hepatocellular carcinoma, leukemia, glioma, adenoid cystic carcinoma), aging, autism, and vascular rheumatoid arthritis, comprising the step of providing a pharmaceutically acceptable salt thereof to a patient.
The pharmaceutically acceptable compositions of the invention may be administered to humans and other animals by oral, rectal, parenteral, intracranial, intravaginal, intraperitoneal, topical (e.g., by powder, ointment or drops), buccal, etc. routes, in the form of an oral or nasal spray, etc., depending on the severity of the infection being treated. In certain embodiments, the compounds of the present invention may be administered orally or parenterally at a dosage level of about 0.01mg/kg to about 50mg/kg, and preferably about 1mg/kg to about 25mg/kg of the subject's body weight, one or more times per day, to achieve the desired therapeutic effect.
Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compound, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Injectable formulations, for example, sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension or emulsion in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that may be employed are water, ringer's solution (U.S. pharmacopoeia) and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
The injectable formulations may be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions which may be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
In order to prolong the effect of the compounds of the invention, it is often desirable to slow down the absorption of the compounds from subcutaneous or intramuscular injection. This can be achieved by using a liquid suspension of crystalline or amorphous material with poor water solubility. In addition, the rate of absorption of a compound depends on its rate of dissolution, which in turn depends on the crystal size and the crystal form. Alternatively, delayed absorption of the compound is achieved by dissolving or suspending the parenterally administered compound form in an oil vehicle. Injectable depot formulations are prepared by forming a matrix of microcapsules of a compound in a biodegradable polymer such as polylactide-polyglycolide. Depending on the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release may be controlled. Examples of other biodegradable polymers include poly (orthoesters) and poly (anhydrides). Long acting injectable formulations can also be prepared by embedding the compounds in liposomes or microemulsions that are compatible with body tissues.
Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycols or suppository waxes which are solid at room temperature but liquid at body temperature, so as to melt in the rectum or vaginal cavity and release the active compound.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is combined with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders (such as starches, lactose, sucrose, glucose, mannitol, and silicic acid), b) binders (such as, for example, carboxy methyl cellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and gum arabic), c) humectants (such as, for example, glycerol), d) disintegrants (such as, for example, agar-agar, calcium carbonate, potato, or tapioca starch, alginic acid, certain silicates, and sodium carbonate), e) solution retarders (such as, for example, paraffin wax), f) absorption accelerators (such as, for example, quaternary ammonium compounds), g) wetting agents (such as, for example, cetyl alcohol and glycerol monostearate), h) absorbents (such as, for example, kaolin and bentonite clay), and i) lubricants (such as, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid compositions of a similar type may also be used as fillers in soft-filled and hard-filled gelatin capsules using lactose or milk sugar as excipients for high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings, and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and may also be a composition which releases the active ingredient(s) only or preferentially in a certain part of the intestinal tract, optionally in a delayed manner. Examples of embedding compositions that may be used include polymers and waxes. Solid compositions of a similar type may also be used as fillers in soft-filled and hard-filled gelatin capsules using lactose or milk sugar as excipients for high molecular weight polyethylene glycols and the like.
The active compound may also be in microencapsulated form with one or more excipients as described above. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings, controlled release coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms, the active compound is admixed with at least one inert filler such as sucrose, lactose or starch. As is normal practice, such dosage forms may also contain additional substances other than inert diluents, such as tabletting lubricants and other tabletting aids (such as magnesium stearate and microcrystalline cellulose). In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and may also be a composition which releases the active ingredient(s) only or preferentially in a certain part of the intestinal tract, optionally in a delayed manner. Examples of embedding compositions that may be used include polymers and waxes.
Dosage forms for topical or transdermal administration of the compounds of the invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier and any required preservatives or buffers which may be required. Ophthalmic formulations, ear drops and eye drops are also considered to be within the scope of the present invention. In addition, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of the compound to the body. Such dosage forms may be prepared by dissolving or partitioning the compound in a suitable medium. Absorption enhancers may also be used to increase the flux of a compound across the skin. The rate may be controlled by providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
As used herein, the term "biological sample" includes, but is not limited to, a cell culture or extract thereof; biopsy material obtained from a mammal or an extract thereof; and blood, saliva, urine, stool, semen, tears, or other bodily fluids or extracts thereof.
In other embodiments, the present invention provides a method for treating a mTORC1 mediated disorder in a patient in need thereof, the method comprising the step of administering to the patient a compound according to the present invention, or a pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.
Depending on the particular condition or disease to be treated, additional therapeutic agents that are typically administered to treat the condition may also be present in the compositions of the present invention. As used herein, an additional therapeutic agent that is typically administered to treat a particular disease or condition is referred to as "suitable for the disease or condition being treated.
The compounds of the present invention may also be used to advantage in combination with other antiproliferative compounds. Such antiproliferative compounds include, but are not limited to, aryl enzyme inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; an alkylating compound; histone deacetylase inhibitors; compounds that induce cellular differentiation processes; cyclooxygenase inhibitors; an MMP inhibitor; an mTOR inhibitor; antitumor drug resistanceA metabolite; a platinum compound; compounds that target/reduce protein or lipid kinase activity and additional anti-angiogenic compounds; a compound that targets, reduces or inhibits protein or lipid phosphatase activity; a somatostatin agonist; an antiandrogen; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; a biological response modifier; an anti-proliferative antibody; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; a proteasome inhibitor; a compound for use in the treatment of hematological malignancies; a compound that targets, reduces or inhibits Flt-3 activity; hsp90 inhibitors such as 17-AAG (17-allylamino geldanamycin, NSC 330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC 707545), IPI-504, CNF1010, CNF2024, CNF1010 (from Kang Fuma pharmaceutical company (Conforma Therapeutics)); temozolomide Spindle kinesin inhibitors such as SB715992 or SB743921 (from GlaxoSmithKline), or pentamidine/chlorpromazine (from combinatoprx); MEK inhibitors such as ARRY142886 (from AArray BioPharma), AZD6244 (from AstraZeneca), PD181461 (from Pfizer), and leucovorin. As used herein, the term "aromatase inhibitor" relates to a compound that inhibits estrogen production (e.g., conversion of androstenedione and testosterone to estrogen ketone and estradiol, respectively). The term includes, but is not limited to, steroids, especially atamestane, exemestane and formestane, and in particular, non-steroids, especially aminoglutethimide, lotamitraz, pirimite, trilostane, testosterone lactone, ketoconazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane is available under the trade name aromas TM And (5) selling. Futaman is available under the trade name Lentaron TM And (5) selling. Fatrozole is under the trade name Afema TM And (5) selling. Anastrozole is sold under the trade name Arimidex TM And (5) selling. Letrozole is under the trade name Femara TM Or Femar TM And (5) selling. Ammonia glutethimide is sold under the trade name Orimeten TM And (5) selling. Including aromatase inhibitionThe combination of chemotherapeutic agents of the present invention is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
As used herein, the term "antiestrogen" refers to a compound that antagonizes estrogenic effects at the estrogen receptor level. The term includes, but is not limited to tamoxifen, fulvestrant, raloxifene, and raloxifene hydrochloride. Tamoxifen is available under the trade name Nolvadex TM And (5) selling. Raloxifene hydrochloride is sold under the trade name Evista TM And (5) selling. Fulvestrant is available under the trade name Faslodex TM And (3) application. The combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors.
As used herein, the term "anti-androgen" refers to any substance capable of inhibiting the biological effects of androgens and includes, but is not limited to, bicalutamide (Casodex TM ). As used herein, the term "gonadotropin-releasing hormone agonist" includes, but is not limited to, abarelix, goserelin, and goserelin acetate. Goserelin is available under the trade name Zoladex TM And (3) application.
As used herein, the term "topoisomerase I inhibitor" includes, but is not limited to, topotecan, gematecon, irinotecan, camptothecin and analogs thereof, 9-nitrocamptothecin, and the macromolecular camptothecin conjugate PNU-166148. Irinotecan can be administered, for example, in the form it is marketed, e.g. under the trade name Camptosar TM And (3) application. Topotecan is under the trade name Hycamptin TM And (5) selling.
As used herein, the term "topoisomerase II inhibitor" includes, but is not limited to, anthracyclines (such as doxorubicin (including liposomal formulations, such as Caelyx TM ) Daunorubicin, epirubicin, idarubicin and nemorubicin), anthraquinones (mitoxantrone and chlorpheniramine), and podophyllotoxins (etoposide and teniposide). Etoposide under the trade name etoposics TM And (5) selling. Teniposide is sold under the trade name VM 26-Bristol. Doxorubicin is available under the trade name Acriblastin TM Or Adriamycin TM And (5) selling. Epirubicin is available under the trade name Farmorubicin TM And (5) selling. IdarubiStar trade name Zavedos TM And (5) selling. Mitoxantrone is sold under the trade name Novantron.
The term "microtubule active agent" relates to microtubule stabilizing, microtubule destabilizing compounds and microtubule polymerization inhibitors, including but not limited to taxanes such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; a discoderm sponge lactone medicament; colchicine and epothilones and derivatives thereof. Paclitaxel under the trade name Taxol TM And (5) selling. Docetaxel is under the trade name of Taxote TM And (5) selling. Vinblastine sulfate is sold under the trade name Vinblastin R.P TM And (5) selling. Vincristine sulfate is known by the trade name Farmsin TM And (5) selling.
As used herein, the term "alkylating agent" includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan, or nitrosourea (BCNU or Gliadel). Cyclophosphamide is known by the trade name cyclophosphamide TM And (5) selling. Ifosfamide under the trade name Holoxan TM And (5) selling.
The term "histone deacetylase inhibitor" or "HDAC inhibitor" relates to a compound that inhibits histone deacetylase and has antiproliferative activity. The term includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
The term "anti-tumor antimetabolites" includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylates (such as 5-azacytidine and decitabine), methotrexate and edatrazine, and folic acid antagonists (such as pemetrexed). Capecitabine has the trade name Xeloda TM And (5) selling. Gemcitabine is available under the trade name Gemzar TM And (5) selling.
As used herein, the term "platinum compound" includes, but is not limited to, carboplatin, cisplatin, and oxaliplatin. Carboplatin can be applied, for example, in the form of its sale, for example under the trade name carboput TM And (3) application. Oxaliplatin can be applied, for example, in the form as it is marketed, e.g. under the trade name Eloxatin TM And (3) application.
As used herein, the term "targeting/reducing protein or lipid excitationA compound having enzymatic activity, or protein or lipid phosphatase activity; or additional anti-angiogenic compounds "including, but not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds that target, reduce or inhibit Platelet Derived Growth Factor Receptor (PDGFR) activity, such as compounds that target, reduce or inhibit PDGFR activity, in particular PDGF receptor-inhibiting compounds, such as N-phenyl-2-pyrimidine-amine derivatives, such as imatinib, SU101, SU6668 and GFB-111; b) A compound that targets, reduces or inhibits Fibroblast Growth Factor Receptor (FGFR) activity; c) Compounds that target, reduce or inhibit insulin-like growth factor receptor I (IGF-IR) activity, such as compounds that target, reduce or inhibit IGF-IR activity, in particular compounds that inhibit kinase activity of IGF-I receptor or antibodies that target the extracellular domain of IGF-I receptor or a growth factor thereof; d) A compound or ephrin B4 inhibitor that targets, reduces or inhibits Trk receptor tyrosine kinase family activity; e) A compound that targets, reduces or inhibits AxI receptor tyrosine kinase family activity; f) A compound that targets, reduces or inhibits Ret receptor tyrosine kinase activity; g) Compounds that target, reduce or inhibit Kit/SCFR receptor tyrosine kinase activity, such as imatinib; h) Compounds that target, reduce or inhibit C-Kit receptor tyrosine kinase activity that is part of the PDGFR family, such as compounds that target, reduce or inhibit C-Kit receptor tyrosine kinase family activity, in particular compounds that inhibit C-Kit receptor, such as imatinib; i) Compounds that target, reduce or inhibit the activity of c-Abl family members, their gene fusion products (e.g., BCR-Abl kinase), and mutants, such as compounds that target, reduce or inhibit the activity of c-Abl family members and their gene fusion products, such as N-phenyl-2-pyrimidine-amine derivatives, such as imatinib or nilotinib (AMN 107); PD180970; AG957; NSC 680410; PD173955 (from ParkeDavis); or dasatinib (BMS-354825); j) Protein Kinase C (PKC) and Raf family members that target, reduce or inhibit serine/threonine kinases, MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, ras/MAPK, PI3K, SYK, TYK2, BTK and TEC family members, and/or cell cycle eggs Compounds that are members of the white dependent kinase family (CDK) active include staurosporine derivatives such as midostaurin; examples of additional compounds include UCN-01, sha Fenge, BAY 43-9006, bryostatin 1, pirifloxacin; tamofosin; RO 318220 and RO 320432; GO 6976; lsis 3521; LY333531/LY379196; isoquinoline compounds; FTI; PD184352 or QAN697 (P13K inhibitor) or AT7519 (CDK inhibitor); k) Compounds that target, reduce or inhibit the activity of a protein-tyrosine kinase inhibitor, such as compounds that target, reduce or inhibit the activity of a protein-tyrosine kinase inhibitor, including imatinib mesylate (Gleevec TM ) Or Tyrphostin, such as Tyrphostin A23/RG-50810; g99; tyrphostin AG 213; tyrphostin AG 1748; tyrphostin AG 490; tyrphostin B44; the Tyrphostin B44 (+) enantiomer; tyrphostin AG 555; AG 494; tyrphostin AG 556, AG957 and adafostine (4- { [ (2, 5-dihydroxyphenyl) methyl group]Amino } -benzoic acid adamantyl ester; NSC 680410, adafostine); l) epidermal growth factor family (EGFR) targeting, reducing or inhibiting receptor tyrosine kinases 1 ErbB2, erbB3, erbB4, homo-or heterodimeric forms) and mutants thereof, such as compounds that target, reduce or inhibit epidermal growth factor receptor family activity, in particular compounds, proteins or antibodies that inhibit EGF receptor tyrosine kinase family members (such as EGF receptor, erbB2, erbB3 and ErbB 4) or bind to EGF or EGF-related ligands, such as CP 358774, ZD 1839, ZM 105180; trastuzumab (Herceptin) TM ) Cetuximab (Erbitux) TM ) Iressa, tarceva, OSI-774, cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or E7.6.3, and 7H-pyrrolo- [2,3-d]Pyrimidine derivatives; m) compounds targeting, reducing or inhibiting c-Met receptor activity, such as compounds targeting, reducing or inhibiting c-Met activity, in particular compounds inhibiting kinase activity of c-Met receptor, or antibodies targeting the extracellular domain of c-Met or binding to HGF, n) compounds targeting, reducing or inhibiting kinase activity of one or more members of the JAK family (JAK 1/JAK2/JAK3/TYK2 and/or pan-JAK), including but not limited to PRT-062070, SB-1578, baritinib, parcritinib, moxionLottinib, VX-509, AZD-1480, TG-101348, tofacitinib, and ruxotinib; o) compounds that target, reduce or inhibit the kinase activity of PI3 kinase (PI 3K), including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, bunazox, pitelist, PF-4691502, BYL-719, dacril, XL-147, XL-765 and Aidoris; a kind of electronic device with high-pressure air-conditioning system; q) targeting, reducing or inhibiting signaling effects of hedgehog (Hh) or smooth receptor (SMO) pathways, including but not limited to cyclopamine, viss Mo Jibu, itraconazole, elis Mo Jibu and IPI-926 (sarideji).
As used herein, the term "PI3K inhibitor" includes, but is not limited to, compounds having inhibitory activity against one or more enzymes of the phosphatidylinositol-3-kinase family, including, but not limited to, PI3kα, PI3kγ, PI3kδ, PI3kβ, PI3K-c2α, PI3K-c2β, PI3K-c2γ, vps34, p110- α, p110- β, p110- γ, p110- δ, p85- α, p85- β, p55- γ, p150, p101, and p87. Examples of PI3K inhibitors useful in the present invention include, but are not limited to, ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, bunazomethide, pitelist, PF-4691502, BYL-719, dacril, XL-147, XL-765, and Aitoriris.
As used herein, the term "Bcl-2 inhibitor" includes, but is not limited to, compounds having inhibitory activity against B cell lymphoma 2 protein (Bcl-2), including, but not limited to ABT-199, ABT-731, ABT-737, apogossypol, pan-Bcl-2 inhibitor of Ai Senda, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitor (inflight pharmaceutical (Infinity Pharmaceuticals)/nova pharmaceutical (Novartis Pharmaceuticals)), genasense (G3139), HA14-1 (and analogs thereof; see WO 2008/118802), navitocrax (and analogs thereof), see U.S. patent 7,390,799), NH-1 (university of sunk), obatocrax (and analogs thereof; see WO 2004/106328), S-001 (the trade of reputation), TW series compounds (michigan university), and nettock. In some embodiments, the Bcl-2 inhibitor is a small molecule therapeutic agent. In some embodiments, the Bcl-2 inhibitor is a mimetic peptide.
As used herein, the term "BTK inhibitor" includes, but is not limited to, compounds having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.
As used herein, the term "SYK inhibitor" includes, but is not limited to, compounds having inhibitory activity against spleen tyrosine kinase (SYK), including, but not limited to, PRT-062070, R-343, R-333, excellair, PRT-062607, and fortinib.
Further examples of BTK inhibiting compounds and conditions treatable by the combination of such compounds with the compounds of the present invention can be found in WO 2008/039218 and WO 2011/090760, which are incorporated herein by reference in their entirety.
Further examples of SYK inhibiting compounds and conditions treatable by combinations of such compounds with the compounds of the invention can be found in WO 2003/063794, WO 2005/007423 and WO 2006/078846, which are incorporated herein by reference in their entirety.
Further examples of PI3K inhibitory compounds and conditions treatable by the combination of such compounds with the compounds of the invention can be found in WO 2004/019973, WO 2004/089925, WO 2007/016176, U.S. patent 8,138,347, WO 2002/088112, WO 2007/084786, WO 2007/129161, WO 2006/122806, WO 2005/113554 and WO 2007/044729, the entire disclosures of which are incorporated herein by reference.
Further examples of JAK inhibitory compounds and conditions treatable by the combination of such compounds with the compounds of the invention can be found in WO 2009/114512, WO 2008/109943, WO 2007/053452, WO 2000/142246 and WO 2007/070514, which are incorporated herein by reference in their entirety.
Additional anti-angiogenic compounds include compounds having other mechanisms of activity unrelated to protein or lipid kinase inhibition, such as thalidomide (thamate TM ) And TNP-470.
Examples of proteasome inhibitors that may be used in combination with the compounds of the present invention include, but are not limited to, bortezomib, alcohol-stopped sulfur, epigallocatechin-3-gallate (EGCG), halosporide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.
Compounds that target, reduce or inhibit protein or lipid phosphatase activity are, for example, inhibitors of phosphatase 1, phosphatase 2A or CDC25, such as okadaic acid or a derivative thereof.
Compounds that induce the cell differentiation process include, but are not limited to, retinoic acid, alpha-, gamma-, or delta-tocopherol or alpha-, gamma-, or delta-tocotrienol.
As used herein, the term "cyclooxygenase inhibitor" includes, but is not limited to, cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acids and derivatives, such as celecoxib (Celebrex) TM ) Rofecoxib (Vioxx) TM ) Etoricoxib, valdecoxib or 5-alkyl-2-arylaminophenylacetic acid (such as 5-methyl-2- (2 '-chloro-6' -fluoroanilino) phenylacetic acid), lomecoxib.
As used herein, the term "bisphosphonate" includes, but is not limited to, etidronic acid, clodronic acid, tiludronic acid, pamidronic acid, alendronic acid, ibandronic acid, risedronic acid, and zoledronic acid. Etidronic acid is available under the trade name Didronel TM And (5) selling. Chlorophosphonic acid is known under the trade name Bonefos TM And (5) selling. Teluphosphonic acid is under the trade name Skelid TM And (5) selling. Pamidronate is under the trade name Aredia TM And (5) selling. Alendronic acid is available under the trade name Fosamax TM And (5) selling. Ibandronic acid is available under the trade name Bondraat TM And (5) selling. Risedronic acid is under the trade name Actonel TM And (5) selling. Zoledronic acid is available under the trade name Zometa TM And (5) selling. The term "mTOR inhibitor" relates to compounds that inhibit mammalian target of rapamycin (mTOR) and have antiproliferative activity, such as sirolimusEverolimus (Certifan) TM ) CCI-779 and ABT578.
As used herein, the term "heparanase inhibitor" refers to a compound that targets, reduces or inhibits the degradation of heparin sulfate. The term includes, but is not limited to, PI-88. As used herein, the term "biological response modifier" refers to a lymphokine or an interferon.
As used herein, the term "inhibitors of Ras oncogenic isoforms," such asSuch as H-Ras, K-Ras or N-Ras refers to compounds that target, reduce or inhibit the oncogenic activity of Ras; for example, "farnesyl transferase inhibitors", such as L-744832, DK8G557 or R115777 (Zarnestra) TM ). As used herein, the term "telomerase inhibitor" refers to a compound that targets, reduces, or inhibits telomerase activity. Compounds that target, reduce or inhibit telomerase activity are especially compounds that inhibit the telomerase receptor, such as telomerase.
As used herein, the term "methionine aminopeptidase inhibitor" refers to a compound that targets, reduces or inhibits methionine aminopeptidase activity. Compounds that target, reduce or inhibit methionine aminopeptidase activity include, but are not limited to, bengamide or derivatives thereof.
As used herein, the term "proteasome inhibitor" refers to a compound that targets, reduces, or inhibits proteasome activity. Compounds that target, reduce or inhibit proteasome activity include, but are not limited to, bortezomib (Velcade) TM ) And MLN 341.
As used herein, the term "matrix metalloproteinase inhibitor" or ("MMP" inhibitor) includes, but is not limited to, collagen mimetic and non-mimetic peptide inhibitors, tetracycline derivatives, for example, the hydroxamic acid mimetic peptide inhibitor, pamphlet and its orally bioavailable analogs, margastat (BB-2516), pramasstat (AG 3340), metastat (NSC 683551), BMS-279251, BAY 12-9566, TAA211, MMI270B, or AAJ996.
As used herein, the term "compound for treating hematological malignancies" includes, but is not limited to, FMS-like tyrosine kinase inhibitors, which are compounds that target, reduce or inhibit FMS-like tyrosine kinase receptor (Flt-3R) activity; interferons, 1-beta-D-arabinofuranosyl cytosine (ara-c) and bisulfan; and ALK inhibitors, which are compounds that target, reduce or inhibit anaplastic lymphoma kinase.
Compounds that target, reduce or inhibit FMS-like tyrosine kinase receptor (Flt-3R) activity are, inter alia, compounds, proteins or antibodies that inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, staurosporine derivatives, SU11248 and MLN518.
As used herein, the term "HSP90 inhibitor" includes, but is not limited to, compounds that target, reduce, or inhibit the intrinsic atpase activity of HSP 90; compounds that degrade, target, reduce or inhibit HSP90 client proteins via the ubiquitin proteasome pathway. Compounds that target, reduce or inhibit the intrinsic atpase activity of HSP90, in particular compounds, proteins or antibodies that inhibit the atpase activity of HSP90, such as 17-allylamino, 17-demethoxygeldanamycin (17 AAG), geldanamycin derivatives; other geldanamycin related compounds; radicicol and HDAC inhibitors.
As used herein, the term "anti-proliferative antibody" includes, but is not limited to, trastuzumab (Herceptin) TM ) Trastuzumab-DM1, erbitux, bevacizumab (Avastin) TM ) RituximabPRO64553 (anti-CD 40) and 2C4 antibodies. Antibodies refer to intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
For the treatment of Acute Myelogenous Leukemia (AML), the compounds of the invention can be used in combination with standard leukemia therapies, especially with therapies for the treatment of AML. In particular, the compounds of the invention may be administered in combination with, for example, a farnesyl transferase inhibitor and/or other drugs useful in the treatment of AML, such as daunorubicin, doxorubicin, ara-C, VP-16, teniposide, mitoxantrone, idarubicin, carboplatin and PKC 412.
Other anti-leukemia compounds include, for example, ara-C, pyrimidine analogs, which are 2' -alpha-hydroxy ribose (arabinoside) derivatives of deoxycytidine. Purine analogs of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate are also included. Compounds that target, reduce, or inhibit the activity of Histone Deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (pro FR 901228), trichostatin a, and compounds disclosed in U.S. patent 6,552,065, including but not limited to N-hydroxy-3- [4- [ [ [2- (2-methyl-1H-indol-3-yl) -ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof and N-hydroxy-3- [4- [ (2-hydroxyethyl) {2- (1H-indol-3-yl) ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof, especially a lactate salt. As used herein, a somatostatin receptor antagonist refers to a compound that targets, treats, or inhibits somatostatin receptors such as octreotide and SOM 230. Tumor cell damage methods refer to methods such as ionizing radiation. The term "ionizing radiation" mentioned above refers to ionizing radiation occurring in the form of electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art (see Hellman, principles of Radiation Therapy, cancer, in Principles and Practice of Oncology, edited by Devita et al, 4 th edition, volume 1, pages 248-275 (1993)).
EDG binding agents and ribonucleotide reductase inhibitors are also included. As used herein, the term "EDG binding agent" refers to a class of immunosuppressants that regulate lymphocyte recirculation, such as FTY720. The term "ribonucleotide reductase inhibitor" refers to pyrimidine or purine nucleoside analogues including, but not limited to, fludarabine and/or cytarabine (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or prastatin. Ribonucleotide reductase inhibitors are in particular hydroxyurea or 2-hydroxy-1H-isoindole-1, 3-dione derivatives.
Also specifically included are those compounds, proteins or monoclonal antibodies to VEGF, such as 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine or a pharmaceutically acceptable salt thereof, 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine succinate; angiostatin TM ;Endostatin TM The method comprises the steps of carrying out a first treatment on the surface of the Anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab, VEGF nucleic acid ligands, such as Macugon; FLT (flash light)-4 inhibitors, FLT-3 inhibitors, VEGFR-2IgGI antibodies, angiozyme (RPI 4610) and bevacizumab (Avastin) TM )。
Photodynamic therapy, as used herein, refers to therapy that treats or prevents cancer using certain chemicals known as photoactive compounds. Examples of photodynamic therapy include treatment with compounds such as Visudyne TM And porphin sodium treatment.
As used herein, angiogenesis inhibiting steroids refer to compounds that block or inhibit angiogenesis, such as anecortave, triamcinolone, hydrocortisone, 11-alpha-epihydrocortisone, 11-deoxycortisol, 17 alpha-hydroxyprogesterone, corticosterone, deoxycorticosterone, testosterone, oestrogen, and dexamethasone.
Implants containing corticosteroids refer to compounds such as fluocinolone acetonide and dexamethasone.
Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds, and antagonists; a biological response modifier, preferably a lymphokine or an interferon; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or heteropoly compounds or compounds with other or unknown mechanisms of action.
The structure of the active compound identified by code number, common or trade name may be taken from the actual version of the standard compilation merck index or from a database, for example Patents International (e.g. IMS World Publications).
The compounds of the invention may also be used in combination with known methods of treatment (e.g., administration of hormones or radiation). In certain embodiments, the provided compounds are useful as radiosensitizers, particularly for treating tumors that exhibit poor sensitivity to radiation therapy.
The compounds of the invention may be administered alone or in combination with one or more other therapeutic compounds, with the possible combination therapies taking the form of a fixed combination or the compounds of the invention and one or more other therapeutic compounds being administered either in alternation or independently of each other, or in combination with a fixed combination and one or more other therapeutic compounds. In addition or alternatively, the compounds of the present invention may be administered for tumor therapy, especially with chemotherapy, radiation therapy, immunotherapy, phototherapy, surgical intervention, or a combination thereof. In other treatment strategies, long-term therapy is equally feasible as the adjuvant therapy described above. Other viable treatments are therapies that maintain the patient's state after tumor regression, or even chemopreventive treatments, such as chemoprevention in at-risk patients.
Those additional agents may be administered separately from the compound-containing compositions of the present invention as part of a multi-dose regimen. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of the present invention in a single composition. If administered as part of a multi-dose regimen, the two active agents may be provided simultaneously, sequentially or within a period of time of each other (typically within five hours of each other).
As used herein, the term "combination" and related terms refer to the simultaneous or sequential administration of therapeutic agents according to the present invention. For example, the compounds of the invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms or together in a single unit dosage form. Accordingly, the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
The amounts of the compounds of the invention and additional therapeutic agents (in those compositions described above that comprise additional therapeutic agents) that can be combined with the carrier material to produce a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, the compositions of the present invention should be formulated so that a dose of 0.01-100mg/kg body weight/day of the compounds of the present invention can be administered.
In those compositions comprising an additional therapeutic agent, the additional therapeutic agent and the compound of the invention may act synergistically. Thus, the amount of additional therapeutic agent in such compositions will be less than that required in monotherapy in which the therapeutic agent is administered alone. In such compositions, additional therapeutic agents may be administered at a dose of 0.01-1,000 μg/kg body weight/day.
The amount of additional therapeutic agent present in the compositions of the present invention will not exceed the amount that would normally be administered in a composition comprising the therapeutic agent as the sole active agent. Preferably, the amount of the additional therapeutic agent in the presently disclosed compositions will be in the range of about 50% to 100% of the amount typically present in compositions comprising the agent as the sole therapeutically active agent.
In some embodiments, the additional therapeutic agent administered in combination with the compounds of the invention is another mTOR inhibitor. In some embodiments, the additional mTOR inhibitor inhibits mTOR by binding to the catalytically active site of mTOR. Examples of such additional mTOR inhibitors include: dacliprist, 8- (6-methoxy-pyridin-3-yl) -3-methyl-1- (4-piperazin-1-yl-3-trifluoromethyl-phenyl) -1, 3-dihydro-imidazo [4,5-c ] quinolin-2-one (WO 2006/122806), valdecolonite (AZD 2014; WO 2009/153597); AZD8055 (WO 2009/153597; XL388 (U.S. patent application publication 2010/0305093)), salazepam tibu (MLN 0128; INK128; WO 2015/051043), DS3078; apitolixib (GDC 0980; WO 2008/070740); O Mi Paxi cloth (GSK-2126458; WO 2008/14446)), NVP-BGT226 (Chang, K.Y. et al, clin. Cal Res. 17 volume 22: 7116-26 page (2011)), wo Daxi cloth (XL 765; SAR245409; WO 2007/044813), PF04691502 (WO 2008/0323162), kdaglixib (PF 05212384; PKI-587; WO 2009/143313), SF1126 (WO 2004/089925), GSK1059615 (WO 2007/13685; OSI) 2007 (WO 2007/061737), VS 84 (WO 2010) and (WO 2010/061737), WO 2010-26 Page (PCV) and CD 7), WO 35, PCV.7/support 7 (WO 7, PCV.7/7), PCV.35 (PCV) and so on-35, WO 35, PCV-7, WO 35 (WO 7/044813), volume 15, phase 12: pages 2705-2718 (2016)); ABTL0812 (WO 2010/106211); WYE-132; EXEL-3885 (Eur J Cancer suppl.6 (12): abst 322 (2008)); EXEL-4431 (Eur J Cancer suppl.6 (12): abst 322 (2008)); AR-mTOR-26 (101 th American society of cancer research (AACR) (17-21 days, washington, USA) 2010, abst 4484); NV-128 (A.B. Alvero et al, mol Cancer Ther volume 10, 8: pages 1385-1393 (2011)); salinomycin (VS-507; gupta, P.B. et al, cell, volume 138, 4: pages 645-659 (2009)); BN-107; BN-108; WAY-600; WYE-687; WYE-354 (Yu, k. Et al, cancer res. Volume 69, 15 th edition: pages 6232-6240 (2009)); ku-063794 (Garcia-Martinez, j.m. et al, biochem.j., volume 421, stage 1: pages 29-42 (2009)); tuote (PP 242; apsel, B. Et al, nat. Chem. Biol. Volume 4, 11: pages 691-699 (2008)); PP30; CZ415 (REF); INK1069; EXEL-2044; EXEL-7518; SB2158; SB2280; AR-mTOR-1 (Wallace, E.M et al mol. Canc. Thermo. Volume 8, journal 12: abst. B267 (2009)).
Reference herein to any particular additional mTOR inhibitor also includes any pharmaceutically acceptable salts, stereoisomers, tautomers, solvates, hydrates and polymorphs thereof.
The compounds of the present invention or pharmaceutical compositions thereof may also be incorporated into compositions for coating implantable medical devices such as dentures, prosthetic valves, vascular grafts, stents and catheters. For example, vascular stents have been used to overcome restenosis (restenosis after damage to the vessel wall). However, patients using stents or other implantable devices are at risk of clot formation and platelet activation. These unwanted effects can be prevented or alleviated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Implantable devices coated with the compounds of the present invention are another embodiment of the present invention.
Examples
As shown in the examples below, in certain exemplary embodiments, the compounds are prepared according to the following general procedure. It should be appreciated that while the general procedure shows the synthesis of certain compounds of the present invention, the following general procedure and other procedures known to those of ordinary skill in the art may be applied to all compounds and each subclass and class of these compounds described herein (see also Luengo, j.i. et al, chem. Biol., volume 2, 7: pages 471-481 (1995); grinfeld, a.a. et al, tet. Lett., volume 35, 37: pages 6835-6838 (1994); PCT/US2019/037507; and PCT/US 2020/0632351, which are incorporated herein by reference in their entirety).
The examples hereinafter list only analytical measurements such as LC/MS, 1 H NMR、 19 When F NMR and the like (rather than the reaction step details), it is understood that the appropriate reagents and reactants are selected and substituted according to the procedures as described in the synthetic schemes and examples herein to prepare the title compound, as will be readily appreciated by those skilled in the art.
All temperatures are expressed in degrees celsius (°c) unless otherwise indicated in the examples. Unless otherwise indicated, all reactions were carried out at ambient temperature and under an inert atmosphere. The reaction solution was stirred at room temperature under N unless otherwise specified 2 (g) Or Ar (g) atmosphere. Reagents used without synthetic details are commercially available or prepared according to known methods, for example according to literature procedures. When the solutions are "concentrated to dryness", they are concentrated under reduced pressure using a rotary evaporator, and when the solutions are dried, they are typically subjected to a desiccant such as MgSO 4 Or Na (or) 2 SO 4 And (5) drying. In the case where the listed synthetic products have been separated into residues, those of ordinary skill in the art will understand that the term "residues" does not limit the physical state of the product as it is separated, and may include, for example, solids, oils, foams, gels, slurries, and the like.
In obtaining the compounds described in the examples below and the corresponding analytical data, the following experimental and analytical protocols were followed, unless otherwise indicated.
LC-MS: unless otherwise indicated, the analytical LC-MS system used consisted of Shimadzu LCMS-2020 with positive ion detection mode electrospray ionization (ESI) with 20ADXR pump, SIL-20ACXR autosampler, CTO-20AC column incubator, M20APDA detector and LCMS 2020MS detector. The column is HALO, C18.times.30 mm. Times.5.0 mm, 2.7. Mu.m. Mobile phase a was water containing 0.05% tfa and mobile phase B was acetonitrile containing 0.05% tfa. The gradient was set as follows: at the position ofFrom 5% mobile phase B to 100% (95%) in 2.0 minutes, hold for 0.7 minutes, then recover to 5% mobile phase B in 0.05 minutes and hold for 0.25 minutes. The column oven (CTO-20 AC) was run at 40.0deg.C. The flow rate was 1.5 mL/min and the injection volume was 1. Mu.L. The detection range of the PDA (SPD-M20A) is 190nm to 400nm. An MS detector configured with electrospray ionization as an ionizable source; the acquisition mode is as follows: scanning; atomizing gas flow rate: 1.5L/min; dry gas flow rate: 15L/min; detector voltage: tuning voltage ± 0.2kv; DL temperature: 250 ℃; heating block temperature: 250 ℃; scanning range: 90.00m/z to 900.00m/z. ELSD (Alltech 3300) detector parameters: drift tube temperature: 60+/-5 ℃; n2 flow rate: 1.8 + -0.2L/min. Mobile phase gradients were optimized for each compound. The calculated mass corresponds to the exact mass.
Preparative HPLC: unless otherwise stated, preparative HPLC purification was performed using a Waters Auto purification system (2545-2767) with 2489UV detector. The column is selected from one of the following: waters C18, 19 mm. Times.150 mm,5 μm; XBridge Prep OBD C18 column, 30 mm. Times.150 mm,5 μm; XSelect CSH Prep C18 OBD column, 5 μm,19 mm. Times.150 mm; XBridge Shield RP18 OBD column, 30mm×150mm,5 μm; xselect CSH fluorophenyl, 30 mm. Times.150 mm,5 μm; or YMC-Actus Triart C18, 30 mm. Times.150 mm,5 μm. The mobile phase is composed of acetonitrile (5% -95%) and contains 0.1% FA or 10mmol/L NH 4 HCO 3 Is composed of a mixture in an aqueous solution. The flow rate was kept at 25 mL/min, the injection volume was 1200. Mu.L, and the UV detector used two channels, 254nm and 220nm. Mobile phase gradients were optimized for each compound.
Normal phase flash chromatography: unless otherwise indicated, normal phase Flash Column Chromatography (FCC) uses a pre-packed silica gel column (such as) The execution was performed using ethyl acetate (EtOAc)/hexane, ethyl acetate (EtOAc)/petroleum ether (b.p.60 ℃ C. To 90 ℃ C.), CH 2 Cl 2 MeOH or CH 2 Cl 2 /10%2N NH 3 As eluent.
1 H NMR: unless otherwise indicated 1 H NMR Spectroscopy was performed using a 400MHz spectrometer (or 500MHz spectrometer) in DMSO-d 6 Or CDCl 3 Obtained in solution. Nuclear Magnetic Resonance (NMR) spectroscopic characteristics refer to chemical shifts (δ) in parts per million (ppm). In DMSO-d 6 Tetramethylsilane (TMS) was used as an internal standard in the solution. Coupling constants (J) are reported in hertz (Hz). The properties of shift with respect to multiplicity are reported as s (singlet), d (doublet), t (triplet), q (quartet), dd (doublet), dt (doublet), m (multiplet), br (broad).
Abbreviation form used in the experimental section
CH 3 CN: acetonitrile
DCM: dichloromethane (dichloromethane)
DMAP: dimethylaminopyridine
DMF: n, N-dimethylformamide
DMSO: dimethyl sulfoxide
EDCI: 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide ESI: electrospray ionization
EtOAc: acetic acid ethyl ester
Et 2 O: diethyl ether
EtOH: ethanol
h: hours of
HCl: hydrogen chloride
HF: hydrogen fluoride
HND-8: acidic ion exchange resins (e.g., amberlyst) H 2 O: water and its preparation method
HPLC: high performance liquid chromatography
MeOH: methanol
min: minute (min)
MgSO 4 : magnesium sulfate
mL: milliliters of (milliliters)
mM: millimoles (milli)
mmol: millimoles (milli)
MS: mass spectrometry
N 2 : nitrogen gas
NaHCO 3 : sodium bicarbonate
NaOH: sodium hydroxide
Na 2 SO 4 : sodium sulfate
NH 3 : ammonia
NH 4 Cl: ammonium chloride
And (3) NMR: nuclear magnetic resonance
C: degree centigrade
prep-HPLC: preparation type high performance liquid chromatography PE: petroleum ether
p-TsOH: para-toluene sulfonic acid
And rt: room temperature
TASF: tris (dimethylamino) sulfonium trimethylsilyl difluoride
TEA: triethylamine
TFA: trifluoroacetic acid
THF: tetrahydrofuran (THF)
Synthetic examples: intermediate products
Synthesis of intermediate I
To a solution of rapamycin (0.2 g,0.22 mmol) in toluene (5 mL) was added proton sponge (0.94 g,4.38 mmol) at room temperature followed by methyl triflate (0.54 g,3.28 mmol). The mixture was stirred at 50 ℃ for 6 hours, then cooled and purified by silica gel chromatography and reverse phase chromatography (85% ch 3 Aqueous solutions of CN) to give (24 e,26e,28e,29e,31R,32s,33R,34R,36s,38s,40s,41s,42R,43R, 52R) -41- [ (1R) -2- [ (1 s,3R, 4R) -3, 4-dimethoxycyclohexyl) as a white solid]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone (50 mg,24% yield). ESI-MS (EI) + ,m/z):964.2[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.50-5.80(m,4H),5.62(ddd,J=22.9,14.5,7.9Hz,1H),5.32(dt,J=11.6,7.7Hz,2H),5.18-5.03(m,1H),4.68(s,1H),3.95-3.54(m,5H),3.50-3.33(m,7H),3.32-3.21(m,3H),3.18-2.92(m,8H),2.83-2.48(m,3H),2.25(dd,J=30.1,10.7Hz,2H),2.02(ddd,J=34.0,26.3,9.6Hz,4H),1.88-1.56(m,14H),1.51-1.16(m,9H),1.15-0.82(m,18H),0.79-0.68(m,1H)。
Synthesis of intermediate II
Step 1: 3-iodopropyl triflate. A mixture of 3-iodopropan-1-ol (4 g,21.51 mmol) and 2, 6-lutidine (4.61 g,43 mmol) in DCM (40 mL) was taken in N 2 Cooled down to 0℃and trifluoromethylsulfonyl trifluoromethanesulfonate (6.67 g,23.66 mmol) was added dropwise. The resulting solution was stirred at 0 ℃ for 2 hours, then quenched with 10% etoac in PE and filtered through a short silica gel column. The filtrate was concentrated in vacuo to give 3-iodopropyl triflate (6.72 g,98 yield) as a pale yellow liquid.
Step 2: (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-iodopropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone (intermediate II). A mixture of rapamycin (2 g,2.19 mmol) and N-ethyl-N-isopropyl-propan-2-amine (5.72 mL,32.82 mmol) in toluene (40 mL) was stirred at 50℃for 16 h. The mixture was poured into ice-cold saturated NaHCO 3 In an aqueous solution (50 mL), the mixture was washed with ice water (60 mL. Times.2) and brine (50 mL) and then dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=3:1) to give intermediate II (1.45 g,60% yield) as a pale yellow solid.
ESI-MS(EI+,m/z):1104.5[M+Na] +
Synthesis of intermediates III and VI
Step 1: 2-methoxyethyl triflate. At 0℃under N 2 To a solution of 2-methoxyethanol (4.5 g,59.14 mmol) and DIEA (11.46 g,88.71 mmol) in DCM (50 mL) was added dropwise trifluoromethyl sulfonyl triflate (18.35 g,65.05 mmol). It was stirred at 0deg.C for 2h, then diluted with DCM (50 mL) and saturated NaHCO 3 (50 mL), water (50 mL), brine (50 mL), and then the organic layer was washed with Na 2 SO 4 Drying, filtration and concentration in vacuo gave 2-methoxyethyltriflate (12.3 g,99% yield) as a brown oil, which was used without further purification. 1 H NMR(400MHz,CDCl 3 ):δ4.62(t,J=4.4Hz,2H),3.71(t,J=4.6Hz,2H),3.42(s,3H)。
Step 2: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -9,27-dihydroxy-10, 21-dimethoxy-3- ((R) -1- ((1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl) propan-2-yl) -6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (intermediate III). Toluene (30 mL) containing rapamycin (3 g,3.28 mmol) and N-ethyl-N-isopropyl-propan-2-amine (8.48 g,65.63 mmol) was stirred at 50℃for 3 hours. Pouring the reaction into cold NaHCO 3 In (50 mL), the mixture was washed with ice water (2X 60 mL), brine (50 mL), and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=1:2) to give intermediate III (1.2 g,38% yield) as a white solid. 1 H NMR(400MHz,CDCl 3 ):δ5.95-6.42(m,4H),5.14-5.58(m,4H),4.41-4.81(m,1H),4.17-4.28(m,1H),3.84-4.00(m,1H),3.63-3.79(m,4H),3.49-3.59(m,2H),3.31-3.46(m,10H),3.07-3.22(m,5H),2.55-2.76(m,2H),2.31-2.35(m,1H),1.91-2.10(m,3H),1.61-1.88(m,19H),1.41-1.55(m,4H),1.15-1.36(m,7H),0.83-1.11(m,16H),0.69-0.76(m,1H)。
Step 3: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-9,10,21- Trimethoxy-3- ((R) -1- ((1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl) propan-2-yl) -6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (intermediate VI). At room temperature, at N 2 Next, the reaction mixture was purified to give (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-41, 44-dimethoxy-42- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl group]-1-methyl-ethyl]To a suspension of 32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclohexa-hexadecane-23, 25,27 (45), 28 (46) -tetralin-47,48,49,50,51-pentanone (0.5 g,0.51 mmol) and 1, 8-bis (dimethylamino) naphthalene (1.65 g,7.71 mmol) in toluene (10 mL) was added methyl triflate (1.01 g,6.17 mmol) dropwise. The reaction was heated to 50deg.C for 3 hours, then filtered and diluted with EA (60 mL), saturated NH 4 Aqueous Cl (60 mL. Times.10), water (60 mL) and brine (60 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography (PE: ea=1:1) to give intermediate VI (92 mg,18% yield) as a white solid. 1 H NMR(400MHz,CDCl 3 ):δ6.03-6.42(m,4H),5.08-5.60(m,4H),4.10-4.74(m,1H),3.73-3.93(m,4H),3.49-3.71(m,5H),3.44-3.46(m,3H),3.34-3.41(m,4H),3.24-3.31(m,3H),3.07-3.18(m,7H),2.48-2.82(m,2H),1.95-2.35(m,5H),1.53-1.83(m,18H),1.42-1.52(m,3H),1.22-1.37(m,6H),1.04-1.15(m,10H),0.86-0.97(m,7H),0.69-0.79(m,1H)。
Synthesis of intermediate IV
Step 1: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c][1]Oxa [4 ]]Aza-compoundsCyclothirty-one pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. To a solution of rapamycin (5 g,5.47 mmol) in DMF (60 mL) was added imidazole (1.49 g,21.88 mmol) and tert-butyl-chloro-dimethyl-silane (2.47 g,16.41mmol,3.05 mL). The reaction was stirred at 50 ℃ for 6 hours and then poured into cold saturated NH 4 Cl solution (40 mL) and Et 2 O: PE (60 mL, 2:1). The organic layer was saturated with NH 4 Cl solution (20 mL), water and brine (20 mL), washed with Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc/PE = 10% to 50%) to give (3 s,6R,7e,9R,10R,12R,14s,15e,17e,19e,21s,23s,26R,27R,34 as) -3- ((R) -1- ((1 s,3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ] ][1]Oxa [4 ]]Azacycle Triundecardecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (4 g,71% yield). ESI-MS (EI) + ,m/z):1050.5[M+Na] +
Step 2: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. At room temperature, at N 2 Next, (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c][1]Oxa [4 ]]To a suspension of aziridinyl tricyclopentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (1 g,0.97 mmol) and 1, 8-bis (dimethylamino) naphthalene (2.5 g,11.67 mmol) in toluene (15 mL) was added dropwise methyl triflate (2.39 g,14.59mmol,1.60 mL). The reaction was then heated to 50℃for protection For 6 hours, cooled and filtered. The filtrate was concentrated and purified by silica gel chromatography to give (28E, 30E,32E,33E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl ] as a white solid]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-56-hydroxy-44, 46, 47-trimethoxy-35,36,37,38,48,49-hexamethyl-65, 66-dioxa-58-azatricyclohexa-hexadecane-28, 30,32 (48), 33 (49) -tetraene-50,51,52,53,54-pentanone (0.45 g,44% yield). ESI-MS (EI) + ,m/z):1064.6[M+Na] +
Step 3: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. To (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ] at 0 ℃ ][1]Oxa [4 ]]To a solution of aziridinyl-pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (0.4 g,0.38 mmol) in THF (10 mL) was added hydrogen fluoride pyridine (3.8 g,38.37 mmol) and the mixture was stirred at 45℃for 5 hours. The mixture was washed with DCM and saturated NaHCO 3 Dilute, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give (3 s,6R,7e,9R,10R,12R,14s,15e,17e,19e,21s,23s,26R,27R,34 as) -27-hydroxy-3- ((R) -1- ((1 s,3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadeca-3H-23, 27-epoxypyrido [2,1-c ] as a white solid][1]Oxa [4 ]]Azacycle Triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (0.16 g,45% yield). ESI-MS (EI) + ,m/z):949.9[M+Na] +
Step 4: (1R, 2R, 4S) -4- ((R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-twenty-four-hydrogen-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclotricyclopentadec-3-yl) propyl) -2-methoxycyclohexyl dimethylphosphinate (intermediate IV). To (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadeca-3H-23, 27-epoxypyrido [2,1-c ] ][1]Oxa [4 ]]To a solution of aziridinyl-pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (0.26 g,0.28 mmol) in DCM (10 mL) was added DCM (1 mL) containing 2, 6-di-tert-butyl-4-methylpyridine (0.17 g,0.84 mmol) and dimethylphosphonyl chloride (0.315 g,2.80 mmol). The reaction was stirred at 0deg.C for 5 hours, then diluted with EtOAc and saturated NaHCO 3 The solution was washed with ice-cold 0.5N HCl, water, brine, and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (DCM: meoh=40:1) to give intermediate IV as a white solid (100 mg,36% yield). ESI-MS (EI) + ,m/z):1025.8[M+Na] +
Synthesis of intermediate V/IX
Step 1:2- ((tert-butyldiphenylsilyl) oxy) ethan-1-ol. Tert-butylchlorodiphenylsilane (26.61 g,96.83 mmol) was added to a solution of ethylene glycol (49.28 g,793.97 mmol) in pyridine (44 mL) at 0deg.C. The resulting solution was stirred at room temperature for 1 hour, then poured into water (500 mL) and extracted with EtOAc (200 ml×3). The organic layer was purified by Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EA: pe=1:8) to give 2- (. About.a. as a colourless solidTert-butyldiphenylsilyl) oxy) ethan-1-ol (25 g,86% yield). ESI-MS (EI) + ,m/z):323.1[M+Na] +
Step 2:2- ((tert-butyldiphenylsilyl) oxy) ethyltriflate. At 0℃under N 2 To a solution of 2- ((tert-butyldiphenylsilyl) oxy) ethan-1-ol (17.13 g,57 mmol) and DIEA (11.05 g,85.52 mmol) in DCM (120 mL) was added trifluoromethylsulfonyl triflate (17.69 g,62.71 mmol). The reaction was stirred at 0deg.C for 2 hours, then diluted with DCM (200 mL) and saturated NaHCO 3 (100 mL. Times.3), water (100 mL. Times.2) and brine (100 mL). The organic layer was purified by Na 2 SO 4 Dried, filtered and concentrated in vacuo to give 2- ((tert-butyldiphenylsilyl) oxy) ethyl triflate (24.5 g,99% yield) as a brown oil. This material was used without further purification.
Step 3: (35E, 37E,39E,40E,46R,47S,48R,49R,51S,53S,55S,56S,57R,58R, 67R) -56- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57, 67-dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclo-hexa-hexadecane-35, 37,39 (59), 40 (60) -tetraene-61, 62,63,64, 65-pentanone. To a solution of rapamycin (5 g,5.47 mmol) and 2- ((tert-butyldiphenylsilyl) oxy) ethyl triflate (23.66 g,54.69 mmol) in toluene (100 mL) was added DIEA (8.48 g,65.63 mmol). The reaction was stirred at 58 ℃ for 16 hours and then poured into cold saturated NaHCO 3 The solution (200 mL) was extracted with EtOAc (100 mL. Times.3). The organic layer was washed with water (100 mL. Times.3) and brine (100 mL), dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=3:1) to give (35 e,37e,39e,40e,46R,47s,48R,49R,51s,53s,55s,56s,57R,58R, 67R) -56- [ (1R) -2- [ (1 s,3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl ] as a yellow solid]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57, 67-dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclo-hexa-hexadecane-35, 37,39 (59), 40 (60) -tetraene-6162,63,64,65-pentanone (4.7 g,72% yield). ESI-MS (EI) + ,m/z):1219.5[M+Na] +
Step 4: (36E, 38E,40E,41E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclo-hexa-hexadecane-36, 38,40 (60), 41 (61) -tetraen-62,63,64,65,66-pentanone. At room temperature, at N 2 Next, the reaction mixture was purified to give (35E, 37E,39E,40E,46R,47S,48R,49R,51S,53S,55S,56S,57R,58R, 67R) -56- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl group ]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]To a solution of (57, 67) -dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclo-hexa-hexadecane-35, 37,39 (59), 40 (60) -tetralin-61, 62,63,64, 65-pentanone (2 g,1.67 mmol) and 1, 8-bis (dimethylamino) naphthalene (3.94 g,18.39 mmol) in toluene (40 mL) was added dropwise methyl triflate (2.19 g,13.37 mmol). The mixture was then heated to 50 ℃ for 5 hours, filtered and diluted with EA (60 mL), saturated NH 4 Cl (aqueous) (60 mL. Times.3), water (60 mL) and brine (60 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography (PE: ea=3:1) to give (36 e,38e,40e,41e,47R,48s,49R,50R,52s,54s,56s,57s,58R,59R, 68R) -57- [ (1R) -2- [ (1 s,3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl ] as a yellow solid]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclo-hexa-hexadecane-36, 38,40 (60), 41 (61) -tetraene-62,63,64,65,66-pentanone (700 mg,35% yield). ESI-MS (EI) + ,m/z):1232.7[M+Na] +
Step 5: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]-41,43, 44-trimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-23, 25,27 (45), 28 (46) -tetraen-47,48,49,50,51-pentanone (intermediate)Body V). At 0℃to (36E, 38E,40E,41E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclohexa-hexadecane-36, 38,40 (60), 41 (61) -tetraene-62,63,64,65,66-pentanone (0.6 g, 0.495mmol) to a solution of THF (10 mL) was added pyridine HF (0.39 g,4.96 mmol). The mixture was stirred at 30℃for 3 hours, then saturated NaHCO 3 The solution (20 mL) was quenched and extracted with EA (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (PE: acetone=3:1) to give intermediate V (430 mg,89% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):994.7[M+Na] +
Synthesis of intermediate VII
Step 1: (27E, 29E,31E,32E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 57R) -46, 57-dihydroxy-44, 47-dimethoxy-45- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4-phenoxycarbonylthioxy-cyclohexyl ]-1-methyl-ethyl]-35,36,37,38,48,49-hexamethyl-66, 67-dioxa-58-azatricyclohexa-27, 29,31 (48), 32 (49) -tetraen-50,51,52,53,54-pentanone. At 0deg.C, to (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-39- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-51-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (300 mg,0.328 mmol) to a solution of pyridine (208 mg,2.63 mmol) and phenyl thiocarboxylate chloride (283 mg,1.64 mmol) in DCM (8 mL). The resulting solution was stirred at 0deg.C for 2 hours, then diluted with DCM, and taken up in NH 4 Cl, water and brine, washed with Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatographySpectrometry (CH) 3 CN/water=0% to 100%) to give the title compound as a white solid (150 mg,44% yield). ESI-MS (EI) + ,m/z):1072.3[M+Na] +1 H NMR(500MHz,CDCl 3 )δ7.41(t,J=7.9Hz,2H),7.29(d,J=7.4Hz,1H),7.12(d,J=7.8Hz,2H),6.44-6.09(m,3H),5.96(d,J=10.4Hz,1H),5.61-5.38(m,2H),5.29(d,J=5.2Hz,1H),5.22-5.06(m,2H),4.79(s,1H),4.20(dd,J=16.6,6.0Hz,1H),3.93-3.52(m,4H),3.51-3.28(m,10H),3.14(s,3H),2.91-2.55(m,3H),2.25(dd,J=91.2,12.9Hz,4H),1.97(d,J=4.8Hz,2H),1.90-1.69(m,9H),1.60(t,J=22.2Hz,11H),1.54-1.38(m,7H),1.37-1.19(m,5H),1.11(ddd,J=25.6,13.0,7.6Hz,10H),1.01-0.84(m,10H)。
Step 2: (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-38, 41-dimethoxy-39- [ (1R) -2- [ (1S, 3S) -3-methoxycyclohexyl]-1-methyl-ethyl]-30,31,32,33,42,43-hexamethyl-59, 60-dioxa-51-azatricyclohexa-hexadecane-23, 25,27 (42), 28 (43) -tetraen-44,45,46,47,48-pentanone. To (27E, 29E,31E,32E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 57R) -46, 57-dihydroxy-44, 47-dimethoxy-45- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4-phenoxycarbonylthioxy-cyclohexyl ]-1-methyl-ethyl]-35,36,37,38,48,49-hexamethyl-66, 67-dioxa-58-azatricyclohexa-hexadecane-27, 29,31 (48), 32 (49) -tetraene-50,51,52,53,54-pentanone (1.4 g,1.33 mmol) to a solution of toluene (15 mL) were added triethylborane (157 mg,1.60 mmol) and bis (trimethylsilyl) silyl-trimethyl-silane (994 mg,4mmol,1M in THF). The resulting solution was stirred at 100 ℃ for 1 hour, then concentrated and purified by silica gel chromatography (EtOAc/pe=0% to 50%) to give the title compound (0.6 g,50% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):920.0[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.59-5.85(m,4H),5.68-5.06(m,4H),4.68(dd,J=48.1,31.4Hz,1H),4.49-3.99(m,2H),3.99-3.51(m,4H),3.52-3.27(m,7H),3.29-3(m,5H),2.88-2.53(m,3H),2.20(ddd,J=80.2,58.5,14.9Hz,6H),1.80(dd,J=34.0,5.5Hz,7H),1.63(d,J=16.1Hz,12H),1.52-1.19(m,10H),1.21-0.78(m,19H),0.70(dd,J=16.1,9.9Hz,2H)。
Step 3: (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,39S,40S,41R,42R, 51R) -51-dihydroxy-39, 41, 42-trimethoxy-40- [ (1R) -2- [ (1S, 3S) -3-methoxycyclohexyl]-1-methyl-ethyl]-31,32,33,34,43,44-hexamethyl-59, 60-dioxa-52-azatricyclohexa-24, 26,28 (43), 29 (44) -tetraen-45,46,47,48,49-pentanone (intermediate VI). To (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-38, 41-dimethoxy-39- [ (1R) -2- [ (1S, 3S) -3-methoxycyclohexyl]-1-methyl-ethyl]-30,31,32,33,42,43-hexamethyl-59, 60-dioxa-51-azatricyclohexa-hexadecane-23, 25,27 (42), 28 (43) -tetraene-44,45,46,47,48-pentanone (200 mg,0.222 mmol) to a solution of toluene (8 mL) were added N1, N1, N8, N8-tetramethylnaphthalene-1, 8-diamine (668 mg,3.12 mmol) and methyl triflate (365 mg,2.23 mmol). The resulting solution was stirred at 50 ℃ for 1 hour, then cooled, filtered and concentrated. The residue was purified by silica gel chromatography (45% etoac/PE) to give intermediate VI (50 mg,12% yield) as a white solid. ESI-MS (EI) + ,m/z):934.2[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.54-5.81(m,4H),5.78-5.02(m,5H),4.52(dd,J=105.2,28.6Hz,1H),4.38-3.94(m,1H),3.93-3.53(m,4H),3.54-3.01(m,12H),3.03-2.46(m,3H),2.45-1.88(m,6H),1.90-1.54(m,16H),1.54-1.19(m,9H),1.19-0.76(m,16H),0.70(d,J=11.0Hz,2H)。
Compound synthesis examples
Example 1: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44S,45R,46R, 55R) -55- Hydroxy-45, 46-dimethoxy-44- [ (1R) -2- [ (1S, 3S) -3-methoxycyclohexyl]-1-methyl-ethyl]-43-[2- (2-methoxyethoxy) ethoxy]-35,36,37,38,47,48-hexamethyl-63, 64-dioxa-56-azatricyclo-thirty Synthesis of Hexane-24, 26,28 (47), 29 (48) -tetraene-49,50,51,52,53-pentanone (I-1)
/>
At 50℃under Ar, to the intermediateVII (150 mg,0.164 mmol) and 2- (2-methoxyethoxy) ethanol (399mg, 3.29 mmol) to a solution of THF (5 mL) was added HND-8 (25 mg). The resulting solution was stirred at 50 ℃ for 2 hours, then filtered and concentrated. The residue was purified by reverse phase chromatography (85% CH 3 CN/water) to give the title compound (I-1) as a white solid: 105mg,64% yield). ESI-MS (EI) + ,m/z):1022.0[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.51-5.83(m,4H),5.72-5.08(m,4H),4.41(ddd,J=101.7,68.7,23.6Hz,2H),4.01-3.03(m,22H),2.93-2.50(m,5H),2.42-1.70(m,17H),1.52-1.21(m,16H),1.20-0.78(m,18H),0.77-0.65(m,1H)。
Example 2: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) a- 27-hydroxy-9, 10-dimethoxy-3- ((R) -1- ((1S, 3S) -3-methoxycyclohexyl) propan-2-yl) -21- (2- ((2-methyl) Oxyethyl) sulfonyl) ethoxy) -6,8,12,14,20,26-hexamethyl-9,10,12,13,14,21,22,23,24,25, 26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ] ][1]Oxa [4 ]]Azacyclo-triundec carbon Synthesis of pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (I-2):
step 1:2- [2- [ tert-butyl (diphenyl) silyl group]Oxyethylsulfonyl group]Ethanol. To a solution of 2- (2-hydroxyethylsulfonyl) ethanol (5.01 g,32.47 mmol) in pyridine (20 mL) was added tert-butyl-chloro-diphenyl-silane (2.22 g,8.08 mmol) at 0deg.C. The reaction was stirred at 15℃for 3 hours, then diluted with water (200 mL) and extracted with EtOAc (100 mL. Times.3). The combined organic layers were concentrated and purified by silica gel chromatography (EtOAc: pe=1:2) to give 2- [2- [ tert-butyl (diphenyl) silyl ] as a white solid]Oxyethylsulfonyl group]Ethanol (2.25 g,71% yield). 1 HNMR(400MHz,CDCl 3 ):δ7.65-7.67(m,4H),7.42-7.47(m,6H),4.09-4.14(m,4H),3.44-3.46(m,2H),3.25-3.27(m,2H),2.57-2.60(m,1H),1.06(s,9H)。
Step 2: tert-butyl- [2- (2-methoxyethylsulfonyl) ethoxy]-diphenyl-silane. At 0℃to 2- [2- [ tert-butyl (diphenyl) silyl ]]Oxyethylsulfonyl group]To a solution of ethanol (8.6 g,21.91 mmol) and N1, N1, N8, N8-tetramethylnaphthalene-1, 8-diamine (14.08 g,65.72 mmol) in toluene (20 mL) was added methyl triflate (10.78 g,65.72 mmol). The mixture was stirred at 50 ℃ for 18 hours, then concentrated, treated with water (200 mL) and extracted with EtOAc (150 ml×2). The combined organic layers were concentrated and purified by silica gel column chromatography (PE: etoac=3:1) to give tert-butyl- [2- (2-methoxyethylsulfonyl) ethoxy as a white solid ]Diphenyl-silane (7.9 g,89% yield). ESI-MS (EI) + ,m/z):429.0[M+Na] +1 HNMR(400MHz,CDCl 3 ):δ7.67-7.69(m,4H),7.39-7.45(m,6H),4.07-4.10(m,2H),3.82-3.84(m,2H),3.40-3.43(m,2H),3.37(s,3H),3.29-3.31(m,2H),1.06(s,9H)。
Step 3:2- (2-methoxyethylsulfonyl) ethanol. To tert-butyl- [2- (2-methoxyethylsulfonyl) ethoxy]To a solution of diphenyl-silane (8.6 g,21.15 mmol) in THF (10 mL) was added Py-HF (31.44 g,317.26 mmol). The mixture was stirred at 20 ℃ for 18 hours, then concentrated and treated with EtOAc (100 mL). Addition of NaHCO 3 (150 mL of aqueous solution) the mixture was stirred at room temperature for 1 hour, then filtered and washed with EtOAc (20 mL). The combined organic layers were concentrated and purified by reverse phase chromatography (water) to give 2- (2-methoxyethylsulfonyl) ethanol (3.55 g,99% yield) as a yellow oil. ESI-MS (EI) + ,m/z):169.0[M+H] +1 H NMR(400MHz,CDCl 3 ):δ4.09-4.13(m,2H),3.83-3.86(m,2H),3.40(s,3H),3.31-3.37(m,4H),2.68-2.71(m,1H)。
Step 4: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44S,45R,46R, 55R) -55-hydroxy-45, 46-dimethoxy-44- [ (1R) -2- [ (1S, 3S) -3-methoxycyclohexyl]-1-methyl-ethyl]-43- [2- (2-methoxyethylsulfonyl) ethoxy]-35,36,37,38,47,48-hexamethyl-65, 66-dioxa-56-azatricyclohexa-hexadecane-24, 26,28 (47), 29 (48) -tetraen-49,50,51,52,53-pentanone (I-2). At 50℃to intermediate VII (200 mg,0.22 mmol) and 2- (2-methoxyethyl)HND-8 (80 mg) was added to a solution of alkylsulfonyl) ethanol (369 mg,2.19 mmol) in THF (3 mL). The mixture was stirred at 50℃for 6 hours, then saturated NaHCO 3 (aqueous) (20 mL) quenched and extracted with EtOAc (30 mL). The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (0-100% CH 3 CN/water) to give the title compound (I-2, 25mg,11% yield). ESI-MS (EI) + ,m/z):1069.7[M+Na] +1 H NMR(400MHz,CDCl 3 ):δ6.05-6.43(m,4H),5.10-5.59(m,4H),4.41-4.44(m,1H),3.53-3.90(m,7H),3.24-3.46(m,15H),3.03-3.17(m,4H),2.89-2.95(m,1H),2.70-2.78(m,1H),2.51-2.69(m,2H),2.17-2.34(m,4H),1.94-2.15(m,4H),1.54-1.89(m,25H),1.22-1.53(m,12H),1.01-1.20(m,12H),0.84-0.96(m,8H),0.69-0.82(m,1H)。
Example 3: (22E, 24E,26E,27E,33R,34S,35R,36R,38S,40S,43S,44R,45R, 55R) -43- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]44,55-dihydroxyl Radical-45-methoxy-42- [2- (2-methoxyethoxy) ethoxy]-33,34,35,36,46,47-hexamethyl-64, 65-di Oxa-56-azatricyclohexadecane-22, 24,26 (46), 27 (47) -tetraene-48,49,50,51,52-pentanone (I-3), (22E,24E,26E,27E,33R,34S,35R,36R,38S,40S,42S,43S,44R,45R,55R)-43-[(1R)-2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-44,55-dihydroxy-45-methoxy Radical-42- [2- (2-methoxyethoxy) ethoxy]-33,34,35,36,46,47-hexamethyl-64, 65-dioxa-56-nitrogen Heterotricyclotricetyl-22, 24,26 (46), 27 (47) -tetraene-48,49,50,51,52-pentanone (I-4) and (22E, 24E, 26E,27E,33R,34S,35R,36R,38S,40S,42R,43S,44R,45R,55R)-43-[(1R)-2-[(1S,3R,4R)- 4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-44,55-dihydroxy-45-methoxy-42- [2- (2-) Methoxyethoxy) ethoxy ]-33,34,35,36,46,47-hexamethyl-64, 65-dioxa-56-azatricyclo thirty-six Synthesis of alkane-22, 24,26 (46), 27 (47) -tetraene-48,49,50,51,52-pentanone (I-5)
Step 1: (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclohexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (intermediate VIII). At room temperature, to (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-39- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]To a solution of-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-51-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (1.0 g,1.09 mmol) in DCM (15 mL) was added an aqueous solution (15 mL) of potassium fluorohydride (1.28 g,16.41 mmol) and bromodifluoro (trimethylsilyl) methane (2.22 g,10.94 mmol). The reaction was stirred at 25 ℃ for 18 hours, then diluted with DCM, saturated NH 4 Aqueous Cl solution, water, brine, na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc: pe=1:1.2) to give the title compound (110 mg,10% yield) as a white solid. ESI-MS (EI) + ,m/z):985.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.72-5.83(m,5H),5.62(ddd,J=22.9,14.6,7.9Hz,1H),5.49-5.01(m,3H),4.67(s,1H),3.98-3.54(m,6H),3.52-3.05(m,15H),2.88-2.52(m,3H),2.41-1.68(m,16H),1.56-1.19(m,10H),1.17-0.86(m,17H),0.76(dd,J=24.3,12.0Hz,2H)。
Step 2: (22E, 24E,26E,27E,33R,34S,35R,36R,38S,40S,43S,44R,45R, 55R) -43- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-44,55-dihydroxy-45-methoxy-42- [2- (2-methoxyethoxy) ethoxy ]]33,34,35,36,46,47-hexamethyl-64, 65-dioxa-56-azatricyclohexa-hexa-ne-22, 24,26 (46),27 (47) -tetraene-48,49,50,51,52-pentanone (CP-NAV-067-1410). At 0℃under N 2 Next, the reaction mixture was purified to give (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]To a solution of 40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclohexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (200 mg,0.21 mmol) and 2- (2-methoxyethoxy) ethanol (498 mg,4.15 mmol) in THF (5 mL) was added 4-methylbenzenesulfonic acid hydrate (197mg, 1.04 mmol). The reaction was stirred at this temperature for 2 hours, then cooled with ice-cold NaHCO 3 Dilute with aqueous solution, extract with EtOAc, wash with brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography (76% CH 3 CN/water) to give the title compound (I-3) as a white solid: 40mg,18% yield).
Step 3: (22E, 24E,26E,27E,33R,34S,35R,36R,38S,40S,42S,43S,44R,45R, 55R) -43- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -44,55-dihydroxy-45-methoxy-42- [2- (2-methoxyethoxy) ethoxy ] -33,34,35,36,46,47-hexamethyl-64, 65-dioxa-56-azatricyclo-hexadecane-22, 24,26 (46), 27 (47) -tetraene-48,49,50,51,52-pentanone (CP-NAV-067-1429-P1) and (22E, 24E,26E,27E,33R,34S,35R,36R,38S,40S,42R,43S,44R,45R, 55R) -43- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -44,55-dihydroxy-45-methoxy-42- [2- (2-methoxyethoxy) ethoxy ] -33,34,35,36,46,47-hexamethyl-64, 65-dioxa-56-azatricyclo-hexadecane-22, 24,26 (46), 27 (47) -tetraene-48,49,50,51,52-pentanone (CP-NAV-067-1429-P2). 100mg of the mixture was separated by chiral HPLC and then purified by silica gel chromatography (PE: DCM: etOAc: meOH=3:3:1:0.3) to give the title compound (I-4:28 mg,28% yield) and (I-5:15 mg,15% yield) as a white solid.
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 1mg/mL mobile phase solution
And (3) sample injection: 5mL of
Mobile phase: hexane/etoh=70/30 (V/V)
Flow rate: 30mL/min
Wavelength: UV 254nm
Temperature: 38 DEG C
I-4:ESI-MS(EI + ,m/z):1073.7[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.60-6.05(m,4H),5.91(dd,J=41.4,11.1Hz,1H),5.58-5.07(m,4H),4.74(s,1H),4.19(dd,J=14.0,6.0Hz,1H),3.95-3.26(m,24H),3.12(dd,J=16.8,7.9Hz,1H),2.92-2.51(m,3H),2.40-1.86(m,8H),1.84-1.64(m,11H),1.54-1.16(m,10H),1.16-0.83(m,18H),0.78-0.65(m,1H)。
I-5:ESI-MS(EI + ,m/z):1073.7[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.64-5.88(m,5H),5.75-5.08(m,5H),4.28(s,1H),4.03-3.02(m,26H),2.98-1.90(m,9H),1.86-1.63(m,16H),1.50-1.17(m,6H),1.16-0.81(m,18H),0.78-0.61(m,1H)。
Example 4: (24E, 26E,28E,29E,32R,33S,34R,35R,37S,39S,42S,44R,45R, 55R) -42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41- (1, 4-dioxane) Alk-2-ylmethoxy) -44,55-dihydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-66, 67-dioxa- 56-azatricyclohexa-24, 26,28 (46), 29 (47) -tetraene-48,49,50,51,52-pentanone (I-6), (24E, 26E,28E,29E,32R,33S,34R,35R,37S,39S,41S,42S,44R,45R,55R)-42-[(1R)-2-[(1S,3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41- (1, 4-dioxan-2-ylmethoxy) room-for-one 44,55-dihydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-66, 67-dioxa-56-azatricyclo-thirty Hexane-24, 26,28 (46), 29 (47) -tetraene-48,49,50,51,52-pentanone (I-9) and (24E, 26E,28E,29E,32R, 33S,34R,35R,37S,39S,41R,42S,44R,45R, 55R) -42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) Radical) -3-methylOxy-cyclohexyl]-1-methyl-ethyl]-41- (1, 4-dioxan-2-ylmethoxy) -44,55-dihydroxy-45- Methoxy-32,33,34,35,46,47-hexamethyl-66, 67-dioxa-56-azatricyclo-tricetyl-24, 26,28 (46) 29 (47) -tetraene-48,49,50,51,52-pentanone (I-10)
Step 1: (24E, 26E,28E,29E,32R,33S,34R,35R,37S,39S,42S,44R,45R, 55R) -42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41- (1, 4-dioxan-2-ylmethoxy) -44,55-dihydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-66, 67-dioxa-56-azatricyclohexa-hexa-ne-24, 26,28 (46), 29 (47) -tetraen-48,49,50,51,52-pentanone (I-6). At 50℃under N 2 Next, the reaction mixture was purified to give (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]To a solution of 40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraen-44,45,46,47,48-pentanone (0.1 g,0.1mmol, from example 3) and 2- (oxetan-3-yloxy) ethanol (245 mg,2.07 mmol) in THF (10 mL) was added HND-8 (50 mg). The reaction mixture was stirred at 50 ℃ for 20 hours, cooled, filtered and the filtrate was poured into saturated NaHCO at 0 ℃ 3 Aqueous (2 mL) and extracted with EtOAc (20 mL). The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc: pe=4:1) and reverse phase chromatography (with 60% ch) 3 CN/water elution) to give the title compound as a white solid (30 mg,28% yield). ESI-MS (EI) + ,m/z):1072.5[M+Na] + 。1H NMR(400MHz,CDCl 3 )δ6.58-5.92(m,5H),5.53-4.75(m,5H),4.27-4.09(m,2H),3.84-3.67(m,9H),3.63-3.54(m,2H),3.45-3.28(m,10H),3.25-3.07(m,3H),2.84-2.55(m,3H),2.35-2.20(m,2H),2.13-1.86(m,6H),1.46-1.77(m,37H),1.43-1.17(m,14H),1.11-0.82(m,22H),0.79-0.69(m,1H)。
Step 2: (24E, 26E,28E,29E,32R,33S,34R,35R,37S,39S,41S,42S,44R,45R, 55R) -42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -41- (1, 4-dioxan-2-ylmethoxy) -44,55-dihydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-66, 67-dioxa-56-azatricyclohexadecane-24, 26,28 (46), 29 (47) -tetraene-48,49,50,51,52-pentanone (I-9) and (24E, 26E,28E,29E,32R,33S,34R,35R,37S,39S,41R,42S, R,45R, 55R) -42- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ] -1-methyl-56-azatricyclohexadecane-24, 26,28 (46), 29 (47) -tetraene-48,49,50,51,52-pentanone (I-9) and (24E, 26E,28E, 29R, 33S,34R, 55R) -42- [ (1S, 3R) -4- (difluoromethoxy-cyclohexyl ] -1-methyl-ethyl ] -1-oxa-methyl-56-oxa-6-oxa-methyl-trione (37S, 67). 115mg of the mixture was separated by chiral HPLC and then purified by silica gel chromatography (PE: DCM: etOAc: meOH=3:3:1:0.3) to give the title compound I-9 (35 mg,30% yield) and I-10 (12 mg,10% yield) as white solids.
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 0.2mg/mL mobile phase solution
And (3) sample injection: 5mL of
Mobile phase: hexane/etoh=60/40 (V/V)
Flow rate: 30mL/min
Wavelength: UV 266nm
Temperature: 35 DEG C
I-9:ESI-MS(EI + ,m/z):1072.5[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.59-6.10(m,4H),5.91(dd,J=28.2,10.4Hz,1H),5.58-5.06(m,4H),4.75(dd,J=16.6,9.8Hz,1H),4.69-4.53(m,1H),4.17(d,J=5.7Hz,1H),3.91-3.54(m,12H),3.48-3.01(m,13H),2.91-2.53(m,3H),2.38-1.81(m,7H),1.83-1.64(m,9H),1.52-1.19(m,10H),1.16-0.81(m,18H),0.74(dd,J=24.3,12.0Hz,1H)。
I-10:ESI-MS(EI + ,m/z):1072.5[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.67-5.84(m,5H),5.70-5.07(m,4H),4.37-4.07(m,3H),3.98(t,J=4.3Hz,1H),3.87-3.57(m,8H),3.56-3.05(m,13H),2.93-1.97(m,10H),1.94-1.64(m,15H),1.54-1.20(m,7H),1.18-0.83(m,18H),0.77-0.61(m,1H)。
Example 5: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -47, 57-dihydroxy-48-methoxy-44- [2- (2-methoxyethoxy) ethoxy ]]-45- [ (1R) -2- [ (1S, 3R) -3-methoxy Phenyl-4- (oxetan-3-yloxy) cyclohexyl]-1-methyl-ethyl]-35,36,37,38,49,50-hexamethyl-67 of the formula, 68-dioxa-58-azatricyclohexa-24, 26,28 (49), 29 (50) -tetraene-51,52,53,54,55-pentanone (I) 7) Is synthesized by (a)
Step 1: [ (37S, 39R, 41R) -4- [ (2R) - (22E, 24E,26E,27E,33R,34S,35R,36R,38S,40S,43S,44R,45R, 54R) -44, 54-dihydroxy-45-methoxy-42- [ (2- (2 methoxyethoxy) ethoxy)]-33,34,35,36,46,47-hexamethyl-48,49,50,51,52-pentoxy-66, 67-dioxa-56-azatricyclo-hexa-hexadecane-22, 24,26 (46), 27 (47) -tetra-43-yl]Propyl group]-41-methoxy-39-cyclohexyl]Triflate esters. At 0℃under N 2 Next, the reaction mixture was purified to give (22E, 24E,26E,27E,33R,34S,35R,36R,38S,40S,43S,44R,45R, 54R) -44, 54-dihydroxy-43- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ]-1-methyl-ethyl]-45-methoxy-42- [2- (2-methoxyethoxy) ethoxy]To a solution of-33,34,35,36,46,47-hexamethyl-64, 65-dioxa-55-azatricyclohexa-hexadecane-22, 24,26 (46), 27 (47) -tetraene-48,49,50,51,52-pentanone (compound A prepared according to U.S.10,980,784, 1g,1 mmol) and 2, 6-dimethylpyridine (1.07 g,10 mmol) in DCM (10 mL) was added dropwise trifluoromethanesulfonic anhydride (1.41 g,4.99 mmol) (dissolved in 1mL DCM). The reaction was stirred at 0 ℃ for 0.5 hours. The reaction mixture was used directly in the next step without further purification.
Step 2: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -47, 57-dihydroxylBase-48-methoxy-44- [2- (2-methoxyethoxy) ethoxy]-45- [ (1R) -2- [ (1S, 3R) -3-methoxy-4- (oxetan-3-yloxy) cyclohexyl]-1-methyl-ethyl]-35,36,37,38,49,50-hexamethyl-67, 68-dioxa-58-azatricyclohexa-24, 26,28 (49), 29 (50) -tetraen-51,52,53,54,55-pentanone (I-7). The reaction solution from step 1 was taken up in N 2 Cool down to 0 ℃ and add DIPEA (1.29 g,9.96 mmol) and oxetan-3-ol (0.74 g,9.96 mmol). The reaction was warmed to room temperature and stirred for 20 hours, then concentrated and purified by silica gel chromatography (80% etoac/PE) and reverse phase chromatography (with 60% ch) 3 CN/water elution) to give the title compound as a white solid (0.055 g,5% yield). ESI-MS (EI) + ,m/z):1079.9[M+Na] + 。1H NMR(400MHz,CDCl 3 )δ6.39-5.94(m,4H),5.54-5.12(m,4H),4.79-4.49(m,4H),4.27-3.98(m,2H),3.91-3.74(m,3H),3.63-3.52(m,9H),3.50-3.12(m,13H),2.81-2.49(m,3H),2.26-1.97(m,4H),1.91-1.49(m,29H),1.53-1.12(m,12H),1.14-0.84(m,15H)。
Example 6: (25E, 27E,29E,30E,33R,34S,35R,36R,38S,40S,43S,45R,46R, 56R) -43- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-42- (1, 4-dioxane) Alk-2-ylmethoxy) -56-hydroxy-45, 46-dimethoxy-33,34,35,36,47,48-hexamethyl-66, 67-dioxa- Synthesis of 57-azatricyclohexa-hexa-25, 27,29 (47), 30 (48) -tetraene-49,50,51,52,53-pentanone (I-8)
Step 1:3- (2-benzyloxy-ethoxy) oxetane. To a solution of oxetan-3-ol (8 g,108 mmol) and 2-bromoethoxymethylbenzene (34.84 g,162 mmol) in DMF (20 mL) was added sodium hydride (5.18 g,216 mmol) in several portions. The resulting solution was stirred at 0 ℃ for 2h and at room temperature for 16 h. The reaction was then treated with 50mL NH 4 Cl (saturated aqueous) was quenched, then extracted with EtOAc (50 mL. Times.2), and the organic layers were combined and concentrated. Will beThe residue was purified by silica gel chromatography (eluting with PE: etOAc (8:1)) to give 3- (2-benzyloxyethoxy) oxetane (12.4 g,55% yield). 1 H NMR(400MHz,CDCl 3 )δ7.32(s,4H),7.31-7.26(m,1H),4.72(dd,J=6.3,5.7Hz,2H),4.64-4.60(m,2H),4.58(dd,J=8.6,3.0Hz,1H),4.54(s,2H),3.57(dt,J=5.6,2.7Hz,5H)。
Step 2:2- (oxetan-3-yloxy) ethanol. Pd/C (4.09 g,38.41mmol, 10%) was added to a solution of 3- (2-benzyloxyethoxy) oxetane (8 g,38.41 mmol) in MeOH (20 mL) in several portions. The resulting solution was stirred at 60 ℃ for 16 hours, then filtered and concentrated. The residue was purified by silica gel chromatography (PE: etoac=1:5) to give 2- (oxetan-3-yloxy) ethanol (2.96 g,65% yield). 1 H NMR(400MHz,CDCl 3 )δ4.79(dd,J=8.3,4.3Hz,2H),4.62(dt,J=10.1,4.9Hz,3H),3.75(d,J=3.9Hz,2H),3.54-3.45(m,2H),2.44(d,J=5.9Hz,1H)。
Step 3: (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclohexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone: to a solution of rapamycin (1 g,1.09 mmol) in DCM (15 mL) was added bromodifluoro (trimethylsilyl) methane (2.22 g,10.94 mmol) dissolved in 30mL of water at room temperature. The reaction was stirred at room temperature for 16 hours, then poured into ice-cold saturated NaHCO 3 In aqueous solution (10 mL). The organic layer was washed with water (10 ml×3) and brine (10 ml×3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc: pe=1:1) to give the title compound as a white solid (200 mg,19% yield). ESI-MS (EI) + ,m/z):986.5[M+Na] + ,T=2.428min。
Step 4: (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,39S,40S,41R,42R, 52R) -40- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-52-hydroxy-39, 41, 42-trimethoxy-30,31,32,33,43,44-hexamethyl-60, 61-dioxa-53-aza Tricyclohexa-23, 25,27 (43), 28 (44) -tetraene-45,46,47,48,49-pentanone: at room temperature, to (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetralin-44,45,46,47,48-pentanone (300 mg,0.31 mmol) to a solution of toluene (6 mL) were added N1, N1, N8, N8-tetramethylnaphthalene-1, 8-diamine (867 mg,4.04 mmol) and methyl triflate (0.51 g,3.11 mmol). The reaction was stirred at 50 ℃ for 2 hours, then filtered, concentrated and purified by silica gel chromatography (EtOAc: pe=1:1.5) and reverse phase chromatography (85% ch) 3 CN/water) to give the title compound as a white solid (100 mg,33% yield). ESI-MS (EI) + ,m/z):1000.5[M+Na] +
Step 5: (25E, 27E,29E,30E,33R,34S,35R,36R,38S,40S,43S,45R,46R, 56R) -43- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-42- (1, 4-dioxan-2-ylmethoxy) -56-hydroxy-45, 46-dimethoxy-33,34,35,36,47,48-hexamethyl-66, 67-dioxa-57-azatricyclohexa-hexa-ne-25, 27,29 (47), 30 (48) -tetraen-49,50,51,52,53-pentanone (I-8). At 50℃under N 2 Next, the reaction mixture was purified to give (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,39S,40S,41R,42R, 52R) -40- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-52-hydroxy-39, 41, 42-trimethoxy-30,31,32,33,43,44-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-23, 25,27 (43), 28 (44) -tetraen-45,46,47,48,49-pentanone (50 mg,0.05 mmol) and 2- (oxetan-3-yloxy) ethanol (121 mg,1.02 mmol) to a solution of THF (5 mL) was added HND-8 (25 mg). The reaction mixture was stirred at 50 ℃ for 20 hours, then cooled, concentrated and purified by silica gel chromatography (EtOAc: pe=1:1) to give the title compound (7 mg,13% yield) as a white solid. ESI-MS (EI) + ,m/z):1086.6[M+Na] + ,T=2.479min。 1 H NMR(400MHz,CDCl 3 )δ6.51-5.88(m,4H),5.34(d,J=3.5Hz,4H),4.86-4.10(m,2H),3.94-3.51(m,8H),3.51-2.99(m,13H),2.85-2.45(m,3H),2.46-1.97(m,6H),1.97-1.54(m,20H),1.55-1.21(m,11H),1.21-0.81(m,17H),0.83-0.65(m,2H)。
Example 7: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -27-hydroxy 1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9, 10-dimethoxy-6, 8,12, 14,20,26-hexamethyl-21- (2- (oxetan-3-yloxy) ethoxy) -9,10,12,13,14,21,22,23,24, 25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-thirty-one Synthesis of carbon pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (I-11)
Step 1: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. To a solution of rapamycin (5 g,5.47 mmol) in DMF (60 mL) was added imidazole (1.49 g,21.88 mmol) at room temperature followed immediately by tert-butyl-chloro-dimethyl-silane (2.47 g,16.41 mmol). The mixture was stirred at 50℃for 6 hours, then poured into ice-cold saturated NH 4 Aqueous Cl (40 mL) and Et 2 O in a mixture of petroleum ether (60 mL, 2:1). The organic layer was then treated with saturated NH 4 Aqueous Cl (20 mL) was washed, water and brine (20 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc/petroleum ether=10% to 50%) to give the title compound (4 g,71% yield) as a white solid. ESI-MS (EI) + ,m/z):1050.5[M+Na] +
Step 2: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((t-butyldi-)Methylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadeca-hydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. At room temperature, at N 2 Next, (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c][1]Oxa [4 ]]To a suspension of aziridinyl tricyclopentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (1 g,0.97 mmol) and 1, 8-bis (dimethylamino) naphthalene (2.5 g,11.67 mmol) in toluene (15 mL) was added dropwise methyl triflate (1.6 mL,14.59 mmol). The mixture was then heated to 50 ℃ for 6 hours, then cooled, filtered and concentrated. The residue was purified by silica gel chromatography to give the title compound (0.45 g,0.43 mmol) as a white solid. ESI-MS (EI) + ,m/z):1064.6[M+Na] +
Step 3: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. To (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ] at 0 ℃ ][1]Oxa [4 ]]To a solution of aziridinyl-pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (0.4 g,0.38 mmol) in THF (10 mL) was added hydrogen fluoride pyridine (3.34 mL,38.37 mmol). The reaction was stirred at 45 ℃ for 5 hours, then poured into DCM and NaHCO 3 In a mixture of aqueous solutions, usingWashing with water and brine, passing through Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography to give the title compound (160 mg,45% yield) as a white solid. ESI-MS (EI) + ,m/z):949.9[M+Na] +
Step 4: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-21- (2- (oxetan-3-yloxy) ethoxy) -9,10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (I-11). To (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ] ][1]Oxa [4 ]]To a solution of aziridinyl-pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (500 mg, 538.68. Mu. Mol) in DCM (15 mL) was added TFA (1.66 mL,21.55 mmol). The mixture was stirred at the same temperature for 10 min, then 2- (oxetan-3-yloxy) ethanol (1.91 g,16.16 mmol) dissolved in DCM (0.2 mL) was added and the mixture was stirred at-10 ℃ for 5 h. The reaction was diluted with a mixture of DCM and aqueous NaHCO3, washed with water and brine, and concentrated over Na 2 SO 4 Dried, filtered and concentrated. The residue was then purified by reverse phase chromatography (70% CH 3 CN/water) to give I-11.ESI-MS (EI) + ,m/z):1036.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.51-5.80(m,4H),5.75-5.03(m,4H),4.83-4.09(m,4H),3.99-3.53(m,7H),3.52-3.02(m,15H),3.01-2.44(m,5H),2.11(ddd,J=99.8,49.8,39.7Hz,7H),1.83-1.61(m,13H),1.52-1.20(m,10H),1.18-0.80(m,17H),0.69(dd,J=23.8,11.9Hz,1H)。
Example 8: (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 61R) -48- (1, 4-dioxan-2-ylmethoxy) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [(1S, 3R, 4R) -3-methoxy Phenyl-4- (3-morpholinopropoxy) cyclohexyl]-1-methyl-ethyl]-39,40,41,42,53,54-hexamethyl-74, 75-dioxo Synthesis of hetero-63-azatricyclohexa-28, 30,32 (53), 33 (54) -tetraene-55,56,57,58,59-pentanone (I-12) Finished products
Step 1:1, 4-dioxan-2-yl methanol. A mixture of 2- (oxetan-3-yloxy) ethanol (7.77 g,65.77 mmol) and HND-8 (2.33 g,65.77 mmol) in THF (120 mL) was stirred at 50℃for 3 hours. The mixture was filtered and concentrated to give 1, 4-dioxan-2-yl methanol (6.97 g,90% yield) as a colorless oil. 1 H NMR(400MHz,CDCl 3 )δ3.87-3.39(m,9H),2.37-2.11(m,1H)。
Step 2: 3-iodopropyl triflate. A mixture of 3-iodopropan-1-ol (4 g,21.51 mmol) and 2, 6-lutidine (4.61 g,43.01 mmol) in DCM (40 mL) was taken in N 2 Cooled down to 0℃and trifluoromethylsulfonyl trifluoromethanesulfonate (6.67 g,23.66 mmol) was added dropwise. The resulting solution was stirred at 0 ℃ for 2 hours, then quenched with 10% etoac in petroleum ether and passed through a short silica gel column, filtered and concentrated to give 3-iodopropyl triflate (6.72 g,98% yield).
Step 3: (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-iodopropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraen-47,48,49,50,51-pentanone. A mixture of rapamycin (2 g,2.19 mmol) and N-ethyl-N-isopropyl-propan-2-amine (4.24 g,32.82 mmol) in toluene (40 mL) was stirred at 50℃for 16 h. The mixture was poured into ice-cold saturated NaHCO 3 In (50 mL), the mixture was washed with ice water (60 mL) and brine (50 mL) twice, and then dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (stone Oil ether ea=3:1) to give the title compound as a pale yellow solid (1.45 g,61% yield). ESI-MS (EI) + ,m/z):1104.5[M+Na] +
Step 4: (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 57R) -47, 57-dihydroxy-45, 48-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl]-1-methyl-ethyl]-36,37,38,39,49,50-hexamethyl-68, 69-dioxa-59-azatricyclohexa-hexadecane-26, 28,30 (49), 31 (50) -tetraen-51,52,53,54,55-pentanone. (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-iodopropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]A mixture of-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone (1.35 g,1.25 mmol) and N-ethyl-N-isopropyl-propan-2-amine (0.48 g,3.74 mmol) in DCM (7.2 mL) was stirred at room temperature for 16 h. The reaction mixture was diluted with DCM and acidified to pH 5 with 1N HCl. The organic phase is treated with H 2 O is washed, passed through a phase separator and then over anhydrous Na 2 SO 4 Drying and concentrating. The residue was purified by silica gel chromatography (EA: 5% NH) 3 Purification with MeOH) afforded the title compound as a pale yellow solid (0.5 g,37% yield). ESI-MS (EI+, m/z): 1042.0[ M+Na ]] +
Step 5: (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 61R) -48- (1, 4-dioxan-2-ylmethoxy) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl]-1-methyl-ethyl]-39,40,41,42,53,54-hexamethyl-74, 75-dioxa-63-azatricyclohexa-28, 30,32 (53), 33 (54) -tetraen-55,56,57,58,59-pentanone (I-12). At 0℃under N 2 Next, the reaction mixture was purified to give (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 57R) -47, 57-dihydroxy-45, 48-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl]-1-methyl-ethyl]-36,37,38,39,49,50-hexamethyl-68, 69-dioxa-59-azatricyclotricetyl-26, 28,30 (49), 31 (50) -tetraen-51,52,53,54,55-pentanone (0.4 g,0.38 mmol) and 1, 4-dioxan-2-ylTo a solution of methanol (1.36 g,11.52 mmol) in DCM (16 mL) was added 2, 2-trifluoroacetic acid (1.75 g,15.36 mmol). The reaction mixture was stirred at-10 ℃ for 20 hours, then saturated with ice NaHCO 3 The aqueous solution (10 mL), water (10 mL. Times.3) and brine (10 mL. Times.3) were washed, dried over anhydrous sodium sulfate, filtered and concentrated. The reaction mixture was purified by reverse phase chromatography (with 50% ch 3 CN/water elution) to give I-12 (156 mg,34% yield). ESI-MS (EI) + ,m/z):1150.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.51-6.01(m,4H),5.47(d,J=43.7Hz,3H),5.33-5.10(m,2H),4.22(d,J=31.5Hz,2H),3.73(dd,J=48.2,40.9Hz,12H),3.39(dd,J=28.6,10.3Hz,10H),3.02(d,J=10.8Hz,3H),2.71(d,J=16.9Hz,9H),2.32(s,2H),2.12-1.37(m,31H),1.35-0.75(m,20H)。
Example 9: (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,48S,49S,51R,52R, 61R) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy Radical) cyclohexyl]-1-methyl-ethyl]-39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) Ethoxy group]-73, 74-dioxa-63-azatricyclohexa-27, 29,31 (53), 32 (54) -tetraene-55, 56,57, 58, 59-pentanone (I-14) and (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,48R,49S,51R,52R, 61R) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy Radical) cyclohexyl]-1-methyl-ethyl]-39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) Ethoxy group]-73, 74-dioxa-63-azatricyclohexa-27, 29,31 (53), 32 (54) -tetraene-55, 56,57, synthesis of 58, 59-pentanone (I-13)
Step 1:3- (2-benzyloxy-ethoxy) oxetane. To a solution of oxetan-3-ol (10 g,135 mmol) in DMF (160 mL) at 0deg.CSodium hydride (3.24 g,135 mmol) was added. The resulting solution was stirred at this temperature for 30 minutes, then 2-bromoethoxymethyl benzene (43.55 g,202.49 mmol) was added. The resulting solution was stirred at 0 ℃ for 2 hours, then at room temperature for 16 hours. The reaction was purified by adding 800mL NH 4 Cl (saturated aqueous solution) was quenched, then extracted with 2X 120mL ethyl acetate and the organic layers were combined and concentrated. The residue was purified by silica gel chromatography (eluting with petroleum ether/EA (8:1)) to give the title compound (16.4 g,78.75 mmol) as a colorless liquid. ESI-MS (EI) + ,m/z):231[M+Na] +1 H NMR(400MHz,CDCl 3 )δ7.41-7.23(m,6H),4.79-4.70(m,2H),4.68-4.52(m,6H),3.62-3.53(m,4H)。
Step 2:2- (oxetan-3-yloxy) ethanol. At N 2 Pd/C (2.04 g,19.21 mmol) was added to a solution of 3- (2-benzyloxyethoxy) oxetane (4 g,19.21 mmol) in MeOH (20 mL) followed by H 2 Stirred overnight at 40 ℃, then filtered and concentrated. The residue was purified by silica gel chromatography (eluting with petroleum ether: ea=1:5) to give the title compound (2.1 g,93% yield) as a colorless liquid. 1 H NMR(400MHz,CDCl 3 )δ4.79(td,J=5.8,2.1Hz,2H),4.62(dt,J=10.2,4.9Hz,3H),3.80-3.69(m,2H),3.52-3.44(m,2H),2.36(s,1H)。
Step 3: 3-iodopropyl triflate. At 0℃under N 2 To a mixture of 3-iodopropan-1-ol (4 g,21.5 mmol) and 2, 6-lutidine (4.61 g,43 mmol) in DCM (40 mL) was added dropwise trifluoromethyl sulfonyl triflate (6.67 g,23.66 mmol). The resulting solution was stirred at 0 ℃ for 2 hours, then quenched with 10% etoac in petroleum ether and filtered through a short silica gel column; the filtrate was concentrated in vacuo to give the title compound as a pale yellow liquid (6.72 g,98% yield).
Step 4: (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-iodopropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-ne-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone. A mixture of rapamycin (2 g,2.19 mmol) and N-ethyl-N-isopropyl-propan-2-amine (5.72 mL,32.82 mmol) in toluene (40 mL) was stirred at 50deg.C for 16 hours and then poured into ice-cold saturated NaHCO 3 (50 mL) of the mixture was washed with ice water (60 mL. Times.2) and brine (50 mL) and then with anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether: ea=3:1) to give the title compound (1.45 g,61% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1104.5[M+Na] +
Step 5: (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 57R) -47, 57-dihydroxy-45, 48-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl]-1-methyl-ethyl]-36,37,38,39,49,50-hexamethyl-68, 69-dioxa-59-azatricyclohexa-hexadecane-26, 28,30 (49), 31 (50) -tetraen-51,52,53,54,55-pentanone. (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-iodopropoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]A mixture of-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone (intermediate II,1.35g,1.25 mmol) and N-ethyl-N-isopropyl-propan-2-amine (0.65 mL,3.74 mmol) in DCM (7.2 mL) was stirred at room temperature for 16 h. The reaction mixture was diluted with DCM and acidified with 1N aqueous HCl to ph=5. The organic phase is treated with H 2 O was washed, filtered through a phase separator, washed with anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EA: 5%7M NH) 3 Meoh=4:1) to give the title compound as a pale yellow solid (498 mg,37% yield). ESI-MS (EI) + ,m/z):1042.0[M+Na] +
Step 6: (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 61R) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl]-1-methyl-ethyl]-39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy ]]-73, 74-dioxa-63-azatricyclohexa-27, 2931 (53), 32 (54) -tetraene-55,56,57,58,59-pentanone. at-50deg.C, at N 2 Next, the mixture was purified to give (26E, 28E,30E,31E,37R,38S,39R,40R,42S,44S,46S,47S,48R,49S, 58R) -48-ethyl-49, 58-dihydroxy-46-methoxy-47- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl ]-1-methyl-ethyl]-37,38,39,40,50,51-hexamethyl-69, 70-dioxa-60-azatricyclohexa-hexadecane-26, 28,30 (50), 31 (51) -tetraene-52,53,54,55,56-pentanone (200 mg,0.19 mmol) 2, 2-trifluoroacetic acid (0.59 mL,7.70 mmol) was added dropwise to a solution of DCM (30 mL). After the addition, the reaction mixture was stirred at-50 ℃ for 10 min, then 2- (oxetan-3-yloxy) ethanol (682 mg,5.77mmol, dissolved in DCM) was added to the reaction mixture at the same temperature. The reaction mixture was stirred at-10℃for 2 hours, then poured into saturated NaHCO at 0 ℃ 3 Aqueous solution (15 mL) and extracted with DCM (20 mL). The organic layer was washed with water (50 mL) and brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (with 50% ch 3 CN/water elution) to give the title compound as a white solid (40 mg,18% yield). ESI-MS (EI) + ,m/z):1126.69[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.39-5.98(m,4H),5.55-5.03(m,5H),4.78-4.43(m,4H),4.15(d,J=40.7Hz,2H),3.71(t,J=21.3Hz,6H),3.60-3.45(m,3H),3.46-3.14(m,10H),2.96(d,J=11.0Hz,3H),2.56(d,J=54.3Hz,8H),2.26(s,2H),2.17-2.03(m,2H),1.94(s,4H),1.80-1.32(m,15H),1.28-1.10(m,11H),1.06-0.56(m,19H)。
Step 7: (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,48S,49S,51R,52R, 61R) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl ] -1-methyl-ethyl ] -39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy ] -73, 74-dioxa-63-azatricyclohexadecan-27, 29,31 (53), 32 (54) -tetraene-55,56,57,58,59-pentanone (I-14) and (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,48R,49S,51R,52R, 61R) -51, 61-dihydroxy-52-methoxy-49- [ (1R) -2S, 3-morpholinopropoxy) ethoxy ] -73, 74-dioxa-63-azatricyclohexadecan-27, 29,31 (53), 32 (54) -tetraene-55,56,57,58,59-pentanone (I-14) and (27E, 29E, 31R, 39R, 40R, 42R, 41R. 120mg (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 61-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (3-morpholinopropoxy) cyclohexyl ] -1-methyl-ethyl ] -39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy ] -73, 74-dioxa-63-azatricyclohexa-hexadecane-27, 29,31 (53), 32 (54) -tetraen-55,56,57,58,59-pentanone are isolated by chiral preparative HPLC and purified by silica gel chromatography (17% MeOH/petroleum ether: EA: 3:3:1) to give I-14 as a white solid (7.2 mg,6% yield) and I-13 as a white solid (14.8 mg,12% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 2.5cm inside diameter x 25cm length, 10 μm
Sample solution: 1.3mg/mL mobile phase solution
And (3) sample injection: 8mL of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 20mL/min
Wavelength: UV 254nm
Temperature: 38 DEG C
I-14:ESI-MS(EI + ,m/z):1126.8[M+H] + ;1148.9[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.41-5.74(m,4H),5.51-5.01(m,4H),4.80-4.41(m,5H),4.10(d,J=5.4Hz,1H),3.88-2.88(m,26H),2.72-2.13(m,12H),2.00-1.27(m,26H),1.10-0.59(m,20H)。
I-13:ESI-MS(EI + ,m/z):1126.8[M+H] + ;1148.9[M+Na] +
1 H NMR(500MHz,CDCl 3 )δ6.41-5.80(m,4H),5.63-5.01(m,4H),4.76-4.45(m,5H),4.25-3.92(m,2H),3.87-2.88(m,25H),2.71-2.04(m,12H),1.91(d,J=28.3Hz,5H),1.62(ddt,J=39.3,32.9,10.5Hz,14H),1.47-1.29(m,7H),1.08-0.55(m,20H)。
Example 10: (28E, 30E,32E,33E,40R,41S,42R,43R,45S,47S,50S,52R,53R, 62R) the reaction products 52, 62-dihydroxy-53-methoxy-50- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (4-methylpiperazin-1-yl) Propoxy group]Cyclohexyl group]-1-methyl-ethyl]-40,41,42,43,54,55-hexamethyl-49- [2- (oxetan-3-yloxy) Radical) ethoxy]-74, 75-dioxa-65-azatricyclohexa-28, 30,32 (54), 33 (55) -tetraene-56, 57, synthesis of 58,59,60-pentanone (I-15)
Step 1: (27E, 29E,31E,32E,37R,38S,39R,40R,42S,44S,46S,47S,48R,49R, 58R) -48, 58-dihydroxy-46, 49-dimethoxy-47- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (4-methylpiperazin-1-yl) propoxy]Cyclohexyl group]-1-methyl-ethyl]-37,38,39,40,50,51-hexamethyl-69, 70-dioxa-61-azatricyclohexa-27, 29,31 (50), 32 (51) -tetraen-52,53,54,55,56-pentanone. (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-iodopropoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]A mixture of-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone (intermediate II,1.45g,1.34 mmol), 1-methylpiperazine (0.16 g,1.61 mmol) and N-ethyl-N-isopropyl-propan-2-amine (0.52 g,4.02 mmol) in DCM (30 mL) was stirred at room temperature for 16 hours, then diluted with DCM and acidified with 1N HCl to pH 5. The organic phase is treated with H 2 O is washed, passed through a phase separator and then over anhydrous Na 2 SO 4 Drying and concentrating. The residue was purified by silica gel chromatography (EA: 5% NH) 3 Purification with MeOH) afforded the title compound (0.96 g,68% yield) as a pale yellow solid. 1 H NMR(400MHz,CDCl 3 ):δ5.95-6.39(m,4H),5.16-5.55(m,4H),4.10-4.22(m,2H),3.55-3.87(m,6H),3.30-3.43(m,8H),2.98-3.17(m,6H),2.67-2.86(m,9H),2.50-2.64(m,3H),2.46(S,2H),2.27-2.35(m,2H),1.95-2.05(m,7H),1.79-1.86(m,3H),1.74-1.76(m,3H),1.58-1.71(m,8H),1.46-1.54(m,3H),1.31-1.35(m,2H),0.86-1.35(m,23H)。
Step 2: (28E, 30E,32E,33E,40R,41S,42R,43R,45S,47S,50S,52R,53R, 62R) -52, 62-dihydroxy-53-methoxy-50- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (4-methylpiperazin-1-yl) propoxy)]Cyclohexyl group]-1-methyl-ethyl]-40,41,42,43,54,55-hexamethyl-49- [2- (oxetan-3-yloxy) ethoxy ]]-74, 75-dioxa-65-azatricyclohexa-28, 30,32 (54), 33 (55) -tetraene-56,57,58,59,60-pentanone (I-15). At-40 ℃ under N 2 Next, (27E, 29E,31E,32E,37R,38S,39R,40R,42S,44S,46S,47S,48R,49R, 58R) -48, 58-dihydroxy-46, 49-dimethoxy-47- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (4-methylpiperazin-1-yl) propoxy ]Cyclohexyl group]-1-methyl-ethyl]-37,38,39,40,50,51-hexamethyl-69, 70-dioxa-61-azatricyclohexa-hexadecane-27, 29,31 (50), 32 (51) -tetraene-52,53,54,55,56-pentanone (0.78 g,0.74 mmol) 2, 2-trifluoroacetic acid (3.38 g,29.65 mmol) was added dropwise to a solution of DCM (20 mL). After the addition, the reaction mixture was stirred at-40 ℃ for 10 min, then 2- (oxetan-3-yloxy) ethanol (2.63 g,22.23mmol in DCM) was added to the reaction mixture at the same temperature. The reaction mixture was stirred at-20℃for 2 hours, then poured into saturated NaHCO at 0 ℃ 3 Aqueous solution (25 mL) and extracted with DCM (25 mL). The organic layer was washed with water (25 mL) and brine (25 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated in vacuo. The residue was purified by reverse phase chromatography (with 50% ch 3 CN/0.01% HCOOH/water elution) to give I-15 (120 mg,14% yield) as a white solid. ESI-MS (EI) + ,m/z):1140.8[M+Na] +1 H NMR(400MHz,CDCl 3 ):δ5.97-6.36(m,4H),5.15-5.49(m,4H),4.57-4.79(m,5H),4.04-4.27(m,2H),3.73-3.86(m,2H),3.54-3.64(m,3H),3.33-3.52(m,11H),2.64-3.09(m,15H),2.48-2.63(m,4H),2.28-2.35(m,2H),1.86-2.11(m,8H),1.61-1.79(m,11H),1.14-1.56(m,12H),0.82-1.09(m,18H)。
Example 11: [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R, 41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexa Methyl-51,52,53,54,55-pentaoxo-71, 72-dioxoHetero-60-aza-tricyclo-hexa-27, 29,31 (49), 32 (50) -tetraen-46-yl]Propyl group]-44-methoxy-42-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamic acid ester (I-16)、[(43S,45R,47R)-4-[(2R)-2-[(28E,30E,32E,33E,39R,40S,41R,42R,44S,46S, 48S,49S,51R,52R, 62R) -51,62-dihydroxy-52-methoxy-39,40,41,42,53,54-hexamethyl-48- [2 ] (oxetan-3-yloxy) ethoxy]-55,56,57,58,59-pentoxy-76, 77-dioxa-64-azatricyclotri Hexadecan-28, 30,32 (53), 33 (54) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]N-methyl-N- (2-) Morpholinoethyl) carbamate (I-18) and [ (43S, 45R, 47R) -4- [ (2R) -2- [ (28E, 30E,32E,33E, 39R), 40S,41R,42R,44S,46S,48R,49S,51R,52R, 62R) -51,62-dihydroxy-52-methoxy-39,40,41,42, 53, 54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy]55,56,57,58,59-pentoxy-76, 77- Dioxa-64-aza-tricyclotricetyl-28, 30,32 (53), 33 (54) -tetraen-49-yl]Propyl group]-47-methoxy-45- Cyclohexyl radical]Synthesis of N-methyl-N- (2-morpholinoethyl) carbamate (I-17)
Step 1: tert-butyl N- (2-morpholinoethyl) carbamate. To a solution of 2-morpholinoethylamine (10 g,76.81 mmol) in DCM (5 mL) at 0deg.C was added triethylamine (5.35 mL,38.41 mmol) and tert-butyl tert-butoxycarbonyl carbonate (18.44 g,84.5 mmol), and the resulting solution was stirred overnight at 25deg.C. The reaction mixture was diluted with 200mL of dichloromethane and then washed with 30mL of 10% sodium bicarbonate and 30mL of brine. The organic layer was dried over sodium sulfate, filtered and concentrated to give the title compound as an off-white solid (17 g,96% yield). ESI-MS (EI) + ,m/z):231.3[M+H] +1 H NMR(400MHz,CDCl 3 )δ3.78-3.62(m,4H),3.24(d,J=5.5Hz,2H),2.45(dd,J=8.0,3.9Hz,6H),1.49-1.42(m,9H)。
Step 2: tert-butyl N-methyl-N- (2-morpholinoethyl) carbamate. Tert-butyl N- (2-morpholinoethyl) carbamate (18 g,78.16 mmol) was dissolved in DMF (240 mL) cooled to 0deg.C and NaH (9.38 g,234.47mmol,60% purity) was added. The reaction was stirred at room temperature for 20 minutes, then cooled to 0 ℃, methyl iodide (12.2 g,85.97 mmol) was added, and the mixture was stirred for an additional 3 hours. The reaction was then diluted with ethyl acetate (500 mL) and washed sequentially with saturated aqueous ammonium chloride (300 mL) and brine (300 ml×5). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure to give the crude title compound (14 g,73% yield) as a white solid. ESI-MS (EI) + ,m/z):245.3[M+H] +1 H NMR(400MHz,CDCl 3 )δ3.74-3.64(m,4H),3.34(s,2H),2.93-2.81(m,3H),2.48(d,J=4.8Hz,6H),1.46(s,10H)。
Step 3: n-methyl-2-morpholino-ethylamine. To hydrochloric acid (4M, 143.25 mL) at 0deg.C was added tert-butyl N-methyl-N- (2-morpholinoethyl) carbamate (14 g,57.3 mmol), and the mixture was stirred at room temperature for 50 min. The reaction mixture was concentrated and the residue was taken up with NH 3 (7M, 81.86 mL) was treated and stirred for 1 hour, then concentrated. The residue was purified by silica gel chromatography (DCM: meOH: tea=90:10:0.1) to give the title compound (7.4 g,90% yield) as a yellow solid. ESI-MS (EI) + ,m/z):145.1[M+H] +1 H NMR(400MHz,DMSO-d 6 )δ9.03(s,2H),3.80(s,4H),3.26(dd,J=44.9,20.4Hz,8H),2.63(s,3H)。
Step 4: [ (43S, 45R, 47R) -4- [ (2R) -2- [ (30E, 32E,34E,35E,39R,40S,41R,42R,44S,46S,48S,49S,50R,51R, 61R) -61-hydroxy-48, 51-dimethoxy-39,40,41,42,52,53-hexamethyl-54, 55,56,57, 58-pentaoxo-50-trimethylsilyloxy-73, 74-dioxa-63-azatricyclohexadecane-30, 32,34 (52), 35 (53) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate. At 0deg.C under argon, the mixture was injected into (25E, 27E,29E,30E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-43-trimethylsilyloxy-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-25, 27,29 (45), 30 (46) -tetraene-47,48,49,50,51-pentanone (0.5 g,0.507 mmol) and pyridine (160.4 mg,2.03mmol,164 μl) were added dropwise triphosgene (150.43 mg,0507 mmol) in THF (20 mL) in DCM (5 mL). The reaction mixture was stirred at 0deg.C for 1 hour, then triethylamine (0.41 g,4.06 mmol) and N-methyl-2-morpholino-ethylamine (1.46 g,10.14 mmol) were added, and the resulting solution was stirred at 0deg.C for another 1 hour, diluted with DCM, and concentrated with NH 4 Aqueous Cl solution, water, brine, na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (8% meoh/DCM) to give the title compound (3836 mg,66% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1156.4[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.57-5.93(m,4H),5.73-5.47(m,1H),5.27-4.98(m,2H),4.72(s,1H),4.56(s,1H),4.36-3.54(m,12H),3.54-3.05(m,12H),2.93(s,4H),2.40(dt,J=34.4,23.8Hz,11H),2.04(s,5H),1.88-1.52(m,12H),1.52-1.17(m,10H),1.20-0.73(m,17H),0.10--0.14(m,9H)。
Step 5: [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-51,52,53,54,55-pentaoxo-71, 72-dioxa-60-azatricyclohexa-hexadecane-27, 29,31 (49), 32 (50) -tetraen-46-yl]Propyl group]-44-methoxy-42-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate. To [ (43S, 45R, 47R) -4- [ (2R) -2- [ (30E, 32E,34E,35E,39R,40S,41R,42R,44S,46S,48S,49S,50R,51R, 61R) -61-hydroxy-48, 51-dimethoxy-39,40,41,42,52,53-hexamethyl-54, 55,56,57, 58-pentoxy-50-trimethylsilyloxy-73, 74-dioxa-63-azatricyclo-hexadecan-30, 32,34 (52), 35 (53) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]To a solution of N-methyl-N- (2-morpholinoethyl) carbamate (1.8 g,1.56 mmol) in acetone (5 mL) and water (5 mL) was added 0.5N sulfuric acid (4.67 mL). The resulting solution was stirred at 0deg.C for 2 hours, then poured into 100mL EtOAc and 100mL saturated NaHCO 3 In a mixture of aqueous solutions. Using organic layersWashing with water and brine, passing through Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (5% meoh/DCM) to give the title compound (1.4 g,83% yield) as a pale yellow solid. 1 H NMR(400MHz,CDCl 3 )δ6.47-5.84(m,4H),5.60-5.05(m,4H),4.77(s,1H),4.55(s,1H),4.34-4.10(m,1H),3.92-3.52(m,7H),3.52-3.23(m,10H),3.13(d,J=2.7Hz,4H),2.92(s,3H),2.78-2.39(m,8H),2.40-2.00(m,5H),2.03-1.53(m,18H),1.53-1.11(m,12H),1.11-0.87(m,13H),0.83(d,J=6.5Hz,2H)。
Step 6: [ (43S, 45R, 47R) -4- [ (2R) -2- [ (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 62R) -51,62-dihydroxy-52-methoxy-39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy ]]-55,56,57,58,59-pentoxy-76, 77-dioxa-64-azatricyclohexa-28, 30,32 (53), 33 (54) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate (I-16). To [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-51,52,53,54,55-pentaoxo-71, 72-dioxa-60-azatricyclohexa-hexadecane-27, 29,31 (49), 32 (50) -tetraen-46-yl under nitrogen]Propyl group]-44-methoxy-42-cyclohexyl]To a solution of N-methyl-N- (2-morpholinoethyl) carbamate (0.2 g,0.18 mmol) in DCM (5 mL) was added TFA at-10℃ (426. Mu.L, 5.53 mmol), followed by 2- (oxetan-3-yloxy) ethanol (0.236 g,3.69 mmol), and the mixture was stirred at-10℃for 2 hours, then with NaHCO 3 The aqueous solution, water and brine were washed, filtered and concentrated. The residue was purified by reverse phase chromatography (65% CH 3 CN/water) to give I-16 (63 mg,29% yield) as a white solid. ESI-MS (EI) + ,m/z):1170.8[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.46-6.01(m,4H),5.56-5.15(m,4H),4.75(s,2H),4.60(s,3H),4.18(s,2H),3.72(s,6H),3.64-3.03(m,13H),2.94(s,3H),2.80-2.28(m,9H),2.13-1.87(m,4H),1.84-1.38(m,24H),1.85-0.75(m,25H)。
Step 7: [ (43S, 45R, 47R) -4- [ (2R) -2- [ (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,48S,49S,51R,52R, 62R) -51,62-dihydroxy-52-methoxy-39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy ] -55,56,57,58,59-pentaoxo-76, 77-dioxa-64-azatricyclohexadecane-28, 30,32 (53), 33 (54) -tetraen-49-yl ] propyl ] -47-methoxy-45-cyclohexyl ] N-methyl-N- (2-morpholinoethyl) carbamate (I-18) and [ (43S, 45R, 47R) -4- [ (2R) -2- [ (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,48R,49S,51R,52R, 62R) -51,62-dihydroxy-52-methoxy-39,40,41,42,53,54-hexamethyl-48- [2- (oxetan-3-yloxy) ethoxy ] -55,56,57,58,59-pentoxy-76, 77-dioxa-64-azatricyclohexadecane-28, 30,32 (53), 33 (54) -tetraen-49-yl ] propyl ] -47-methoxy-45-cyclohexyl ] N-methyl-N- (2-morpholinoethyl) carbamate (I-17). 120mg of I-16 were isolated by chiral preparative HPLC to give I-18 as a white solid (27.5 mg,23% yield) and I-17 as a white solid (16.1 mg,13% yield).
Chiral analysis method
Chromatographic column: CHIRALPAK IC (IC 00CE-UF 123)
Column dimensions: 0.46cm inside diameter x 25cm length
And (3) sample injection: 20 μl of
Mobile phase: etoh=100%
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-18:ESI-MS(EI + ,m/z):1170.8[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.40-5.78(m,4H),5.62-5.03(m,4H),4.76-4.39(m,6H),4.12(d,J=5.9Hz,1H),3.88-3.59(m,6H),3.56-3.00(m,17H),2.86(s,3H),2.79-2.18(m,11H),2.15-1.81(m,5H),1.59(t,J=15.2Hz,13H),1.49-1.15(m,11H),1.10-0.66(m,18H)。
I-17:ESI-MS(EI + ,m/z):1170.7[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.49-5.81(m,4H),5.64-5.06(m,4H),4.66(d,J=65.7Hz,3H),4.23(d,J=26.2Hz,2H),3.94-3.03(m,28H),2.98-2.22(m,15H),2.21-1.69(m,11H),1.54-1.18(m,13H),1.15-0.69(m,19H)。
Example 12: [ (42S, 44R, 46R) -4- [ (2R))-2-[(27E,29E,31E,32E,38R,39S,40R,41R, 43S,45S,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy ]]-54,55,56,57, 58-pentoxy-75, 76-dioxa-64-aza-tri Cyclotriacontan-27, 29,31 (52), 32 (53) -tetraen-48-yl]Propyl group]-46-methoxy-44-cyclohexyl]N- (2-morpholin) Ethyl) carbamate (I-19), [ (42S, 44R, 46R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,38R,39S, 40R,41R,43S,45S,47S,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52, 53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy]-54,55,56,57, 58-pentaoxo-75, 76-dioxo Hetero-64-aza-tricyclohexa-27, 29,31 (52), 32 (53) -tetraen-48-yl]Propyl group]-46-methoxy-44-cyclohexane Base group]N- (2-morpholinoethyl) carbamic acid (I-21) and [ (42S, 44R, 46R) -4- [ (2R) -2- [ (27E, 29E, 31E), 32E,38R,39S,40R,41R,43S,45S,47R,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38, 39,40,41,52,53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy]-54,55,56,57, 58-pentaoxy Substituted-75, 76-dioxa-64-azatricyclohexa-27, 29,31 (52), 32 (53) -tetraen-48-yl]Propyl group]-46- Methoxy-44-cyclohexyl]N- (2-morpholinoethyl) carbamate (I-20)
Step 1: (25E, 27E,29E,30E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-43-trimethylsilyloxy-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-25, 27,29 (45), 30 (46) -tetraene-47,48,49,50,51-pentanone. To a solution of rapamycin (5.5 g,6.02 mmol) and imidazole (3.2 g,48 mmol) in EtOAc (35 mL) was added dropwise TMSCl (5.2 g,48 mmol). After the addition, the resulting solution was stirred at room temperature for 1 hour, and then 0.5. 0.5N H was added at 0 ℃ 2 SO 4 Solution (24 mL). After addition, the resulting solution was stirred at 0deg.C for 1.5 hours, then diluted with EtOAc (100 mL) and brine (50 mL) and the organic layer was taken up in anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography to give the title compound (4.33 g, 73% in 2 steps) as a white solid. ESI-MS (EI) + ,m/z):1008.2[M+Na] +1 H-NMR(400MHz,CDCl 3 )δ6.50-5.80(m,4H),5.61(ddd,J=23.0,14.1,7.6Hz,1H),5.37-5.19(m,2H),5.07(ddd,J=11.3,9.1,5.2Hz,1H),4.71(d,J=1.4Hz,1H),3.89-3.56(m,4H),3.50-3.30(m,6H),3.29-3.18(m,3H),3.18-3.04(m,3H),2.97-2.86(m,1H),2.84-2.45(m,3H),2.43-2.05(m,4H),1.97(dd,J=10.0,5.4Hz,2H),1.86-1.50(m,19H),1.49-0.81(m,23H),0.68(dd,J=23.6,11.9Hz,1H),0.05-0.07(m,9H)。
Step 2: [ (42S, 44R, 46R) -4- [ (2R) -2- [ (29E, 31E,33E,34E,38R,39S,40R,41R,43S,45S,47S,48S,49R,50R, 60R) -60-hydroxy-47, 50-dimethoxy-38,39,40,41,51,52-hexamethyl-53,54,55,56,57-pentoxy-49-trimethylsilyloxy-72, 73-dioxa-63-azatricyclohexadecane-29, 31,33 (51), 34 (52) -tetraen-48-yl]Propyl group]-46-methoxy-44-cyclohexyl]N- (2-morpholinoethyl) carbamate. At 0deg.C under argon, the mixture was injected into (25E, 27E,29E,30E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-43-trimethylsilyloxy-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-25, 27,29 (45), 30 (46) -tetraene-47,48,49,50,51-pentanone (2 g,2.03 mmol) and pyridine (0.64 g,8.11 mmol) were added dropwise triphosgene (0.6 g,2.03 mmol) in DCM (40 mL). The reaction mixture was stirred at 0deg.C for 1 hour, then TEA (0.62 g,6.08 mmol) and 2-morpholinoethylamine (0.53 g,4.06 mmol) were added to the mixture, and the resulting solution was stirred at 0deg.C for another 1 hour, then diluted with DCM, and taken up in NH 4 Aqueous Cl solution, water, brine, na 2 SO 4 Dried, filtered and concentrated. Will remain behindThe material was purified by silica gel chromatography (8% meoh/DCM) to give the title compound (2 g,86% yield) as a pale yellow solid.
Step 3: [ (39S, 41R, 43R) -4- [ (2R) -2- [ (26E, 28E,30E,31E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 57R) -46, 57-dihydroxy-44, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-50,51,52,53,54-pentaoxo-70, 71-dioxa-60-azatricyclohexadecane-26, 28,30 (48), 31 (49) -tetraen-45-yl]Propyl group]-43-methoxy-41-cyclohexyl]N- (2-morpholinoethyl) carbamate. To [ (42S, 44R, 46R) -4- [ (2R) -2- [ (29E, 31E,33E,34E,38R,39S,40R,41R,43S,45S,47S,48S,49R,50R, 60R) -60-hydroxy-47, 50-dimethoxy-38,39,40,41,51,52-hexamethyl-53,54,55,56,57-pentoxy-49-trimethylsilyloxy-72, 73-dioxa-63-azatricyclo-hexadecane-29, 31,33 (51), 34 (52) -tetraen-48-yl]Propyl group]-46-methoxy-44-cyclohexyl]N- (2-morpholinoethyl) carbamate (2 g,1.75 mmol) in acetone (40 mL) and H 2 0.5. 0.5N H was added to a solution of O (10 mL) 2 SO 4 (2.63 mmol,5.3 mL). The resulting solution was stirred at 0deg.C for 8 hours, then poured into 100mL EtOAc and 100mL saturated NaHCO 3 In a mixture of aqueous solutions. The organic layer was washed with water and brine, dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (5% meoh/DCM) to give the title compound (1.5 g,80% yield) as a pale yellow solid.
Step 4: [ (42S, 44R, 46R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,38R,39S,40R,41R,43S,45S,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy ]]-54,55,56,57, 58-pentaoxo-75, 76-dioxa-64-azatricyclohexa-hexa-dec-27, 29,31 (52), 32 (53) -tetraen-48-yl]Propyl group]-46-methoxy-44-cyclohexyl]N- (2-morpholinoethyl) carbamate. To [ (39S, 41R, 43R) -4- [ (2R) -2- [ (26E, 28E,30E,31E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 57R) -46, 57-dihydroxy-44, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-50,51,52,53,54-pentaoxo-70, 71-dioxa-60-azatricyclo-hexadecan-26, 28,30 (48), 31 (49) -tetraen-45-yl under nitrogen at-40 ℃]Propyl group]-43-methoxy-41-cyclohexyl]N- (2-morpholin)To a solution of ethyl carbamate (02 g,0.18 mmol) in DCM (4 mL) was added TFA (0.85 g,7.47 mmol). 2- (oxetan-3-yloxy) ethanol (0.22 g,1.87 mmol) was added and the mixture was stirred at-30℃for 2 hours. Pouring the mixture into cold saturated NaHCO 3 In aqueous solution (30 mL), extracted with DCM (30 mL), washed with water (30 mL) and brine (30 mL), and dried over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was passed through a reverse phase column (with 80% CH 3 CN/water elution) to give I-19 (35 mg,16% yield) as a white solid. ESI-MS (EI) + ,m/z):1179.6[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.41-5.90(m,4H),5.58-5.39(m,2H),5.30-5.15(m,2H),4.80-4.51(m,5H),4.32-3.95(m,2H),3.92-3.66(m,7H),3.60-3.40(m,4H),3.39-3.20(m,11H),3.19-3.05(m,2H),2.79-2.62(m,2H),2.61-2.40(m,7H),2.37-2.20(m,2H),2.15-1.90(m,5H),1.84-1.58(m,17H),1.54-1.16(m,7H),1.15-0.83(m,17H),0.82-0.75(m,1H)。
Step 5: [ (42S, 44R, 46R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,38R,39S,40R,41R,43S,45S,47S,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy ] -54,55,56,57, 58-pentaoxo-75, 76-dioxa-64-azatricyclotricyclohexadecane-27, 29,31 (52), 32 (53) -tetraen-48-yl ] propyl ] -46-methoxy-44-cyclohexyl ] N- (2-morpholinoethyl) carbamate (I-21) and [ (42S, 44R, 46R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,38R,39S,40R,41R,43S,45S,47R,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy ] -54,55,56,57, 58-pentoxy-75, 76-dioxa-64-azatricyclohexadecane-27, 29,31 (52), 32 (53) -tetraen-48-yl ] propyl ] -46-methoxy-44-cyclohexyl ] N- (2-morpholinoethyl) carbamate (I-20)
115mg [ (42S, 44R, 46R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,38R,39S,40R,41R,43S,45S,48S,50R,51R, 61R) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-47- [2- (oxetan-3-yloxy) ethoxy ] -54,55,56,57, 58-pent-oxa-64-azatricyclo-hexadecane-27, 29,31 (52), 32 (53) -tetraen-48-yl ] propyl ] -46-methoxy-44-cyclohexyl ] N- (2-morpholinoethyl) carbamate are isolated by chiral preparative HPLC and then purified by silica gel chromatography (12% MeOH/petroleum ether: DCM: EA=3:3:1) to give I-21 (23 mg,20% yield) as a white solid and I-20 (10 mg,8.7% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 2.5cm inside diameter x 25cm length, 10 μm
Sample solution: 3mg/mL mobile phase solution
And (3) sample injection: 10mL
Mobile phase: etoh=100%
Flow rate: 10mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-21:ESI-MS(EI + ,m/z):1156.9[M+H] + ,1178.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.51-5.78(m,4H),5.74-5.01(m,5H),4.68(ddd,J=37.1,11.1,6.1Hz,6H),4.32-4.13(m,1H),3.96-3.06(m,23H),2.79-2.24(m,10H),2.17-1.18(m,29H),1.17-0.76(m,19H)。
I-20:ESI-MS(EI + ,m/z):1156.9[M+H] + ,1178.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.58-5.81(m,4H),5.67-5.02(m,5H),4.44(dd,J=176.8,44.7Hz,6H),4.03-3.05(m,26H),2.83-2.29(m,10H),2.17-1.19(m,27H),1.16-0.69(m,19H)。
Example 13: (22E, 24E,26E,27E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) a- 44- [2- (2-Aminoethoxy) ethoxy]-56-hydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) p-henate 3-methoxy-cyclohexyl]-1-methyl-ethyl]-46, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-66, 67- Dioxa-58-aza-tricyclohexa-22, 24,26 (48), 27 (49) -tetraene-50,51,52,53,54-pentanone (I-23)
Step 1: (36E, 38E,40E,41E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclo-hexa-hexadecane-36, 38,40 (60), 41 (61) -tetraen-62,63,64,65,66-pentanone. At room temperature, at N 2 Next, the reaction mixture was purified to give (35E, 37E,39E,40E,46R,47S,48R,49R,51S,53S,55S,56S,57R,58R, 67R) -56- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl group ]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57, 67-dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclo-hexa-hexadecane-35, 37,39 (59), 40 (60) -tetralin-61, 62,63,64, 65-pentanone (intermediate IX prepared according to example 22, 2g,1.67 mmol) and 1, 8-bis (dimethylamino) naphthalene (3.94 g,18.39 mmol) were added dropwise to a solution of methyl triflate (2.19 g,13.37 mmol) in toluene (40 mL). After addition, the mixture was heated to 50 ℃ for 5 hours, then cooled, filtered and diluted with EA (60 mL), with saturated NH 4 Cl (aqueous) (60 mL. Times.3), water (60 mL) and brine (60 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography (petroleum ether: ea=3:1) to give the title compound (700 mg,35% yield) as a yellow solid. ESI-MS (EI) + ,m/z):1232.7[M+Na] +
Step 2: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41,43, 44-trimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-23, 25,27 (45), 28 (46) -tetraen-47,48,49,50,51-pentanone. At 0℃to (36E, 38E,40E,41E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl ]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclo-hexa-hexadecane-36, 38,40 (60), 41 (61) -tetraene-62, 63To a solution of 64,65, 66-pentanone (600 mg,0.496 mmol) in THF (10 mL) was added pyridine HF (399mg, 4.96 mmol). The mixture was stirred at 30 ℃ for 3 hours, then by addition of saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with EA (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether: acetone=3:1) to give the title compound (430 mg,89% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):994.7[M+Na] +
Step 3: (22E, 24E,26E,27E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -44- [2- (2-azidoethoxy) ethoxy]-56-hydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-46, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-68, 69-dioxa-60-azatricyclo-hexa-hexadecane-22, 24,26 (48), 27 (49) -tetraen-50,51,52,53,54-pentanone. At 50℃under N 2 Next, the reaction mixture was purified to give (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]To a solution of-41, 43, 44-trimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-23, 25,27 (45), 28 (46) -tetraene-47,48,49,50,51-pentanone (450 mg, 462.85. Mu. Mol) and 2- (2-azidoethoxy) ethanol (1.21 g,9.26 mmol) in THF (10 mL) was added HND-8 (100 mg). The reaction mixture was stirred at 50 ℃ for 22 hours, then cooled and filtered. Pouring the filtrate into saturated NaHCO at 0 DEG C 3 Aqueous (10 mL) and extracted with EA (30 mL), the organic layer was washed with water (10 mL) and brine (10 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by reverse phase chromatography (70% CH 3 CN/water) to give the title compound as a pale yellow solid (250 mg,50% yield). ESI-MS (EI) + ,m/z):1093.4[M+Na] +
Step 4: (22E, 24E,26E,27E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -44- [2- (2-aminoethoxy) ethoxy]-56-hydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-46, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-66, 67-dioxa-58-azatricyclohexa-hexa-ne-22, 24,26 (48), 27 (49) -tetraene-50,51,52,53,54-pentanone (I-23). To (22E, 24E,26E,27E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -44- [2- (2-azidoethoxy) ethoxy ]-56-hydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-46, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-68, 69-dioxa-60-azatricyclo-hexa-hexadecane-22, 24,26 (48), 27 (49) -tetraene-50,51,52,53,54-pentanone (0.6 g,0.56 mmol) to a solution of THF (10 mL) was slowly added triphenylphosphine (0.44 g,1.68 mmol). The resulting solution was stirred at 60℃for 2 hours, 0.05ml of water was added, the reaction was stirred at room temperature for 6 hours, and then concentrated. The residue was purified by reverse phase chromatography (CH 3 CN/H 2 O was purified with 0.025% TFA) to give I-23 (40 mg,7% yield) as a white solid. ESI-MS (EI) + ,m/z):1045.7[M+H] +1 H NMR(400MHz,CDCl 3 )δ4.61-5.96(m,4H),5.69-5.07(m,4H),4.51-4.01(m,3H),3.82-3.52(m,7H),3.47-3.37(m,5H),3.31-3.04(m,13H),2.88-2.52(m,2H),2.38-1.97(m,7H),1.85-1.52(m,17H),1.38-1.13(m,7H),1.12-0.98(m,5H),0.98-0.77(m,17H),0.75-0.68(m,1H)。
Example 14: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E, 19E), 23S,26R,27R,34 aS) -21- ((1, 4-dioxan-2-yl) methoxy) -27-hydroxy-9, 10-dimethoxy-6, 8,12, 14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1,4,5,6,9,10,11,12,13,14,21,22,23,24,25, 26,27,28,29,31,32,33,34 a-icosatetrahydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Aza-compounds Cyclothirty-one-pentadecen-3-yl) propyl) -2-methoxycyclohexyldimethylphosphinate (I-24) and (27E, 29E,31E, 32E,35R,36S,37R,38R,40S,42S,44S,45S,47R,48R, 57R) -45- [ (1R) -2- [ (1S, 3R, 4R) -4-di Methyl phosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-44- (1, 4-dioxan-2-ylmethoxy) -57-hydroxy Phenyl-47, 48-dimethoxy-35,36,37,38,49,50-hexamethyl-68, 69-dioxa-58-azatricyclo-hexa-hexadecane 27,2Synthesis of 9,31 (49), 32 (50) -tetraene-51,52,53,54,55-pentanone (I-25)
Step 1: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. Imidazole (1.49 g,21.88 mmol) and tert-butyl-chloro-dimethyl-silane (2.47 g,16.41 mmol) were added to a solution of rapamycin (5 g,5.47 mmol) in DMF (60 mL) at room temperature. The reaction was stirred at 50℃for 6 hours and then poured into ice-cold saturated NH 4 Aqueous Cl (40 mL) and Et 2 O in a mixture of petroleum ether (60 mL, 2:1). The organic layer was saturated with NH 4 Aqueous Cl (20 mL) was washed, water and brine (20 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc/petroleum ether=10% to 50%) to give the title compound (4 g,71% yield) as a white solid. ESI-MS (EI) + ,m/z):1050.5[M+Na] +
Step 2: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. At room temperature, at N 2 Next, (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-9,10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]To a suspension of aziridinyl tricyclopentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (1 g,0.97 mmol) and 1, 8-bis (dimethylamino) naphthalene (2.5 g,11.67 mmol) in toluene (15 mL) was added dropwise methyl triflate (1.6 mL,14.59 mmol). After the addition, the mixture was heated to 50 ℃ for 6 hours. Then cooled, filtered and the filtrate was purified by silica gel chromatography to give the title compound (0.45 g,0.43 mmol) as a white solid. ESI-MS (EI+, m/z): 1064.6[ M+Na ] ] +
Step 3: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone. To (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -3- ((R) -1- ((1S, 3R, 4R) -4- ((tert-butyldimethylsilyl) oxy) -3-methoxycyclohexyl) propan-2-yl) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ] at 0 ℃][1]Oxa [4 ]]To a solution of aziridinyl-pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (400 mg,038 mmol) in THF (10 mL) was added hydrogen fluoride pyridine (3.34 mL,38.37 mmol). The reaction was warmed to 45 ℃, stirred for 5 hours, then taken up with DCM and NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography to give the title compound (0.16 g,45% yield) as a white solid. ESI-MS (EI) + ,m/z):949.9[M+Na] +
Step 4: (1R, 2R, 4S) -4- ((R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-twenty-four-hydrogen-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-thirty-one pentadecene3-yl) propyl) -2-methoxycyclohexyl dimethyl phosphinate. To (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4-hydroxy-3-methoxycyclohexyl) propan-2-yl) -9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-9, 10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadeca-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]To a solution of aziridinyl-pentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (0.26 g,0.28 mmol) in DCM (10 mL) was added DCM (1 mL) containing 2, 6-di-tert-butyl-4-methylpyridine (0.173 g,0.84 mmol) and dimethylphosphonyl chloride (0.315 g,2.8 mmol). The resulting solution was stirred at 0deg.C for 5 hours, then diluted with EtOAc, washed with aqueous NaHCO3, ice-cold 0.5N HCl solution, water, brine, and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (DCM: meoh=40:1) to give the title compound (0.1 g,36% yield) as a white solid. ESI-MS (EI) + ,m/z):1025.8[M+Na] +
Step 5: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -21- ((1, 4-dioxan-2-yl) methoxy) -27-hydroxy-9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-icosano-3H-23, 27-epoxypyrido [2, 1-c)][1]Oxa [4 ]]Azacyclo-triundec-pentadec-3-yl) propyl) -2-methoxycyclohexyl dimethyl phosphinate (I-24). at-50deg.C, to (1R, 2R, 4S) -4- ((R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -27-hydroxy-9,10,21-trimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-twenty-four-hydro-3H-23, 27-epoxypyrido [2, 1-c)][1]Oxa [4 ]]To a solution of aziridinylpentadec-3-yl) propyl-2-methoxycyclohexyldimethylphosphinate (129 mg,0.129 mmol) in DCM (5 mL) was added TFA (0.49 mL,6.42 mmol). The mixture was stirred at the same temperature for 10 min, then 2- (oxetan-3-yloxy) ethanol (0.758 g,6.42 mmol) dissolved in DCM (0.5 mL) was added and the mixture was stirred at 0 ℃ for 6 h. Then the reaction is carried out The material was treated with DCM and NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was then purified by reverse phase chromatography (70% CH 3 CN/water) to give I-24 (30 mg,21% yield) as a white solid. ESI-MS (EI) + ,m/z):1112.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.49(br,4H),5.63-5.06(m,4H),4.78-4.05(m,3H),3.66(ddd,J=23.6,18.4,8.5Hz,9H),3.49-2.97(m,17H),2.82-2.48(m,2H),2.37-1.86(m,7H),1.56-1.23(m,22H),1.18-0.68(m,24H)。
Step 6: (27E, 29E,31E,32E,35R,36S,37R,38R,40S,42S,44S,45S,47R,48R, 57R) -45- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoxy-3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -44- (1, 4-dioxan-2-ylmethoxy) -57-hydroxy-47, 48-dimethoxy-35,36,37,38,49,50-hexamethyl-68, 69-dioxa-58-azatricyclohexadecane-27, 29,31 (49), 32 (50) -tetraen-51,52,53,54,55-pentanone (I-25). 100mg of I-24 were isolated by chiral preparative HPLC and then purified by silica gel chromatography (8% MeOH/petroleum ether: DCM: EA: 3:3:1) to give I-25 as a white solid (14 mg,14% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 2.5cm inside diameter x 25cm length, 10 μm
Sample solution: 9mg/mL mobile phase solution
And (3) sample injection: 15mL
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 30mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
ESI-MS(EI + ,m/z):1112.6[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.49-5.80(m,4H),5.67-5.15(m,4H),4.20-4.02(m,2H),3.98-3.55(m,12H),3.52-3.00(m,17H),2.60(ddd,J=39.8,34.1,28.4Hz,6H),2.37-1.83(m,7H),1.66(dt,J=39.0,20.7Hz,12H),1.42-1.19(m,8H),1.18-0.66(m,20H)。
Example 15: (25E, 27E,29E,30E,33R,34S,35R,36R,38S,40S,43S,45R,46R,55R)- 42- (1, 4-dioxan-2-ylmethoxy) -55-hydroxy-43- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclo Hexyl group]-1-methyl-ethyl]-45, 46-dimethoxy-33,34,35,36,47,48-hexamethyl-66, 67-dioxa-56-nitrogen Heterotricyclotricetyl-25, 27,29 (47), 30 (48) -tetraene-49,50,51,52,53-pentanone (I-26) and (25E, 27E, 29E,30E,33R,34S,35R,36R,38S,40S,42S,43S,45R,46R, 55R) -42- (1, 4-dioxan-2-ylmethoxy Phenyl) -55-hydroxy-43- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 46-dimethoxy-33,34,35,36,47,48-hexamethyl-66, 67-dioxa-56-azatricyclotricyclo-hexadecane-25, 27, 29 (47), synthesis of 30 (48) -tetraene-49,50,51,52,53-pentanone (I-27)
Step 1: (25E, 27E,29E,30E,33R,34S,35R,36R,38S,40S,43S,45R,46R, 55R) -42- (1, 4-dioxan-2-ylmethoxy) -55-hydroxy-43- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 46-dimethoxy-33,34,35,36,47,48-hexamethyl-66, 67-dioxa-56-azatricyclo-hexa-hexadecane-25, 27,29 (47), 30 (48) -tetraene-49,50,51,52,53-pentanone (I-26). At 0deg.C under nitrogen, to (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,39S,40S,41R,42R, 51R) -51-hydroxy-40- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-39,41, 42-trimethoxy-30,31,32,33,43,44-hexamethyl-60, 61-dioxa-52-azatricyclo-hexa-hexadecane-23, 25,27 (43), 28 (44) -tetraen-45,46,47,48,49-pentanone (intermediate X prepared according to example 16, 0.312g,0.336 mmol) in THF (15 mL) was added 2- (oxetan-3-yloxy) ethanol (0.397 g,3.36 mmol) and HND-8 (624 mg). The mixture was stirred at 50 ℃ for 5 hours, then purified by reverse phase chromatography (with 80% ch 3 CN/water elution) and purification by TLC (petroleum ether: etoac=1:2) afforded I-26 (30 mg,9% yield) as a white solid. ESI-MS(EI + ,m/z):1035.8[M+Na] +1 H NMR(500MHz,CDCl 3 ): 1 H NMR(500MHz,CDCl 3 )δ6.57-5.90(m,3H),5.71-5.00(m,3H),4.72-4.10(m,1H),3.91-3.52(m,7H),3.38(dd,J=22.8,12.9Hz,5H),3.30-3.15(m,3H),3.16-3.02(m,3H),3.00-2.46(m,4H),2.15(dd,J=97.2,37.0Hz,5H),1.85-1.53(m,23H),1.52-1.21(m,9H),1.19-0.82(m,14H),0.69(d,J=11.9Hz,1H)。
Step 2: (25E, 27E,29E,30E,33R,34S,35R,36R,38S,40S,42S,43S,45R,46R, 55R) -42- (1, 4-dioxan-2-ylmethoxy) -55-hydroxy-43- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -45, 46-dimethoxy-33,34,35,36,47,48-hexamethyl-66, 67-dioxa-56-azatricyclohexadecane-25, 27,29 (47), 30 (48) -tetraen-49,50,51,52,53-pentanone (I-27). 85mg of the epimeric mixture was purified by preparative chiral HPLC followed by silica gel chromatography (hexane: DCM: etOAc: meOH=3:3:1:0.3) to give I-27 as a white solid (25 mg,29% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 0.3mg/mL mobile phase solution
And (3) sample injection: 3mL of
Mobile phase: hexane/etoh=70/30 (V/V)
Flow rate: 25mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
ESI-MS(EI + ,m/z):1036.4[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.44-5.80(m,4H),5.65-5.01(m,4H),4.64(d,J=15.9Hz,1H),3.99-3.52(m,11H),3.47-3.02(m,16H),3.02-2.46(m,5H),2.43-1.85(m,8H),1.83-1.64(m,9H),1.46-1.19(m,10H),1.16-0.83(m,18H),0.79-0.59(m,1H)。
Example 16: (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) a- 57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-47, 48-bis Methoxy-45- [2- [2- (2-methoxyethoxy) ethoxy ] ]Ethoxy group]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-23, 25,27 (49), 28 (50) -tetraen-51,52,53,54,55-pentanone (I) 28 ) synthesis of
Step 1: (27E, 29E,31E,32E,34R,35S,36R,37R,39S,41S,43S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 55-dihydroxy-43, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-65, 66-dioxa-57-azatricyclohexa-hexadecane-27, 29,31 (47), 32 (48) -tetraene-49,50,51,52,53-pentanone. At room temperature, to (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-39- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-51-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (2 g,2.19 mmol) to a solution of DMF (30 mL) was added imidazole (0.596 g,8.75 mmol) and t-butylchlorodimethylsilane (0.989 g,6.56 mmol). The mixture was stirred at 20℃for 5 hours, then poured into ice-cold saturated NH 4 Aqueous Cl (40 mL) and Et 2 O in Petroleum ether (60 mL, 2:1). The organic layer was saturated with NH 4 Aqueous Cl (20 mL) was washed, water and brine (20 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc/petroleum ether=10% to 50%) to give the title compound (1.5 g,67% yield) as a white solid. ESI-MS (EI) + ,m/z):1049.8[M+Na] +
Step 2: (28E, 30E,32E,33E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-56-hydroxy-44, 46, 47-trimethoxy-35,36,37,38,48,49-hexamethyl-65, 66-dioxa-58-azatricyclo-thirtyHexane-28, 30,32 (48), 33 (49) -tetraene-50,51,52,53,54-pentanone. At room temperature, at N 2 Next, the reaction mixture was purified to give (27E, 29E,31E,32E,34R,35S,36R,37R,39S,41S,43S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl group]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]To a suspension of-45, 55-dihydroxy-43, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-65, 66-dioxa-57-azatricyclo-hexa-hexadecane-27, 29,31 (47), 32 (48) -tetralin-49,50,51,52,53-pentanone (0.6 g,0.58 mmol) and 1, 8-bis (dimethylamino) naphthalene (1.5 g,7 mmol) in toluene (20 mL) was added methyl triflate (0.957 g,5.83 mmol) dropwise. After addition, the mixture was heated to 50 ℃ for 6 hours, then cooled, filtered and the filtrate purified by silica gel chromatography (EtOAc: petroleum ether=4:1) to give the title compound (0.24 g,39% yield) as a white solid. ESI-MS (EI) + ,m/z):1063.8[M+Na] +
Step 3: (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,39S,40S,41R,42R, 51R) -51-hydroxy-40- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-39,41, 42-trimethoxy-30,31,32,33,43,44-hexamethyl-60, 61-dioxa-52-azatricyclo-hexa-hexadecane-23, 25,27 (43), 28 (44) -tetraen-45,46,47,48,49-pentanone. At 0℃to (28E, 30E,32E,33E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-56-hydroxy-44, 46, 47-trimethoxy-35,36,37,38,48,49-hexamethyl-65, 66-dioxa-58-azatricyclohexa-hexadecane-28, 30,32 (48), 33 (49) -tetraene-50,51,52,53,54-pentanone (0.24 g,0.23 mmol) to a solution of THF (10 mL) was added hydrogen fluoride pyridine (2.28 g,23.02mmol,2 mL). The reaction was stirred at 45℃for 5 hours, then with DCM and saturated NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography (78% CH 3 CN/water) to give the title compound as a white solid (0.105 g,49% yield). ESI-MS (EI) + ,m/z):949.7[M+Na] +
Step 4: (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,46S,47R,48R,57R ) -57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-47, 48-dimethoxy-45- [2- [2- (2-methoxyethoxy) ethoxy ]]Ethoxy group]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-hexadecane-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-28). To (23E, 25E,27E,28E,30R,31S,32R,33R,35S,37S,39S,40S,41R,42R, 51R) -51-hydroxy-40- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl under nitrogen at 0 ℃]-1-methyl-ethyl]-39,41, 42-trimethoxy-30,31,32,33,43,44-hexamethyl-60, 61-dioxa-52-azatricyclohexa-hexadecane-23, 25,27 (43), 28 (44) -tetraene-45,46,47,48,49-pentanone (0.15 g,0.16 mmol) to a solution of THF (15 mL) was added 2- (2-methoxyethoxy) ethoxy ethanol (0.265 g,1.62 mmol) and HND-8 (0.3 g), and the mixture was stirred at 50 ℃ for 8 hours. The reaction mixture was purified by reverse phase chromatography (with 80% ch 3 CN/water elution) and then purification by preparative TLC (petroleum ether: ethyl acetate=1:2) gave (23 e,25e,27e,28e,36R,37s,38R,39R,41s,43s,46s,47R,48R, 57R) -57-hydroxy-46- [ (1R) -2- [ (1 s,3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl as a white solid ]-1-methyl-ethyl]-47, 48-dimethoxy-45- [2- [2- (2-methoxyethoxy) ethoxy ]]Ethoxy group]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-hexadecane-23, 25,27 (49), 28 (50) -tetraen-51,52,53,54,55-pentanone (36.5 mg,21% yield). ESI-MS (EI) + ,m/z):1035.8[M+Na] +1 HNMR(500MHz,CDCl 3 ): 1 H NMR(400MHz,CDCl 3 )δ6.59-5.88(m,3H),5.85-4.93(m,4H),4.72-4.18(m,1H),4.15-3.76(m,2H),3.74-3.52(m,8H),3.50-3.30(m,8H),3.29-3.03(m,5H),3.03-2.47(m,5H),2.45-1.89(m,6H),1.90-1.52(m,21H),1.32(ddd,J=28.1,22.9,5.8Hz,9H),1.19-0.78(m,14H),0.69(d,J=12.0Hz,1H)。
Example 17: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) a- 45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy Radical-44- [2- (2-methoxyethoxy) ethoxy]-35,36,37,38,48,49-hexamethyl-64, 65-dioxa-57-nitrogen Heterotricyclotricetyl-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-29), (24E, 26E, 28E,29E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R,56R)-45-[(1R)-2-[(1S,3R,4R)- 3, 4-Dimethoxycyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethoxy) Radical) ethoxy]-35,36,37,38,48,49-hexamethyl-64, 65-dioxa-57-azatricyclohexa-24, 26,28 (48) 29 (49) -tetraene-50,51,52,53,54-pentanone (I-31) and (24E, 26E,28E,29E,35R,36S,37R,38R, 40S,42S,44R,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl Radical-ethyl radical]-56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethoxy) ethoxy ] ]-35,36,37,38,48, 49-hexamethyl-64, 65-dioxa-57-azatricyclohexa-24, 26,28 (48), 29 (49) -tetraene-50, 51,52, synthesis of 53, 54-pentanone (I-30)
Step 1: (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone. At room temperature, to (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-39- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-51-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (0.2 g,0.219 mmol) to a solution of toluene (5 mL) was added proton sponge (0.938 g,4.38 mmol) followed by methyl triflate (0.539 g,3.28 mmol). The mixture was stirred at 50 ℃ for 6 hours, cooled and chromatographed on silica gel followed by reverse phase chromatography (85% ch 3 CN/water) to give a white colorThe title compound (50 mg,24% yield) was a coloured solid. ESI-MS (EI) + ,m/z):964.2[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.50-5.80(m,4H),5.62(ddd,J=22.9,14.5,7.9Hz,1H),5.32(dt,J=11.6,7.7Hz,2H),5.18-5.03(m,1H),4.68(s,1H),3.95-3.54(m,5H),3.50-3.33(m,7H),3.32-3.21(m,3H),3.18-2.92(m,8H),2.83-2.48(m,3H),2.25(dd,J=30.1,10.7Hz,2H),2.02(ddd,J=34.0,26.3,9.6Hz,4H),1.88-1.56(m,14H),1.51-1.16(m,9H),1.15-0.82(m,18H),0.79-0.68(m,1H)。
Step 2: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethoxy) ethoxy ]]-35,36,37,38,48,49-hexamethyl-64, 65-dioxa-57-azatricyclohexa-24, 26,28 (48), 29 (49) -tetraen-50,51,52,53,54-pentanone (I-29). At 50℃under N 2 Next, the reaction mixture was purified to (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]To a solution of 52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone (0.17 g,0.18 mmol) and 2- (2-methoxyethoxy) ethanol (0.43 g,3.61 mmol) in sulfolane (5 mL) was added HND-8 (35 mg). The resulting solution was stirred at 50℃for 3 hours, filtered and the filtrate was passed through a C18 column with 85% CH 3 CN/water elution gave I-29 as a white solid (65 mg,35% yield). ESI-MS (EI) + ,m/z):1052.5[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.46
-5.81(m,4H),5.74-5.03(m,4H),4.68-4.15(m,2H),3.99-3.52(m,11H),3.50
-3.22(m,16H),3.21-2.98(m,6H),2.94-2.44(m,3H),2.37-1.89(m,7H),1.86
-1.69(m,7H),1.52-1.24(m,9H),1.22-0.84(m,21H),0.74(dd,J=22.3,10.9Hz,1H)。
Step 3: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethoxy) ethoxy ] -35,36,37,38,48,49-hexamethyl-64, 65-dioxa-57-azatricyclotricyclo-hexadecane-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-31) and (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44R,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R) -4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -56-hydroxy-46, 47-dimethoxy-44- [2- (62-methoxyethoxy) -29-oxa-48, 29-oxa-48 (48, 48) tetraene-29-oxa-2- (29, 29-oxa-trione (I-31). 130mg of the mixture was separated by chiral preparative HPLC and then purified by silica gel chromatography (hexane: DCM: etOAc: meOH=3:3:1:0 to 3:3:1:0.4) to give I-31 as a white solid (45 mg,35% yield) and I-30 as a white solid (40 mg,31% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 1.4mg/mL mobile phase solution
And (3) sample injection: 15mL
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 60mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-31:ESI-MS(EI + ,m/z):1052.1[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.51-5.82(m,4H),5.76-5.03(m,4H),4.51(dd,J=56.0,27.4Hz,1H),4.35-4.06(m,1H),4.00-3.20(m,26H),3.19-2.98(m,5H),2.88-2.48(m,3H),2.40-1.85(m,7H),1.82-1.65(m,11H),1.38(ddd,J=37.8,31.6,21.3Hz,10H),1.21-0.83(m,18H),0.79-0.68(m,1H)。
I-30:ESI-MS(EI + ,m/z):1052.2[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.52-5.81(m,4H),5.77-5.04(m,5H),4.70-4.14(m,2H),4.01-2.97(m,31H),2.64(dd,J=50.7,36.3Hz,3H),2.42-1.68(m,16H),1.50-0.61(m,30H)。
Example 18: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) a- 46- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphinyloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57-hydroxy Radical-47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I) 32)、(25E,27E,29E,30E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R,57R)-46-[(1R)-2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl ]]-1-methyl-ethyl]-57-hydroxy-47, 48-di Methoxy-45- [2- (2-methoxyethoxy) ethoxy]36,37,38,39,49,50-hexamethyl-66, 67-dioxa- 58-azatricyclohexa-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-34) and (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,45R,46S,47R,48R,57R)-46-[(1R)-2-[(1S,3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]57-hydroxy-47, 48-dimethoxy- 45- [2- (2-methoxyethoxy) ethoxy]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatris Synthesis of Cyclotriacontane-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-33)
Step 1: (27E, 29E,31E,32E,34R,35S,36R,37R,39S,41S,43S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 55-dihydroxy-43, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-65, 66-dioxa-57-azatricyclohexa-hexadecane-27, 29,31 (47), 32 (48) -tetraene-49,50,51,52,53-pentanone. To (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-39- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-51-azatricyclo-triTo a solution of hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (2 g,2.19 mmol) in DMF (15 mL) was added imidazole (0.298 g,4.38 mmol) and tert-butyl-chloro-dimethyl-silane (0.495 g,3.28 mmol). The mixture was stirred at 20℃for 5 hours, then poured into ice-cold saturated NH 4 Aqueous Cl (10 mL) and Et 2 O in Petroleum ether (300 mL, 2:1), the organic layer was saturated with NH 4 Aqueous Cl (100 mL) was washed, water and brine (100 mL), and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (EtOAc/petroleum ether=10% to 50%) to give the title compound (1.85 g,82% yield) as a white solid. ESI-MS (EI) + ,m/z):1050.2[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.44-5.83(m,4H),5.60-5.07(m,4H),4.32-4.04(m,2H),3.79(d,J=13.4Hz,1H),3.70(d,J=6.1Hz,1H),3.65(dd,J=9.8,5.5Hz,1H),3.62-3.53(m,1H),3.43-3.28(m,8H),3.13(s,3H),2.94-2.81(m,1H),2.73(dd,J=16.8,5.9Hz,2H),2.63-2.47(m,1H),2.33(d,J=12.7Hz,2H),2.07-1.89(m,4H),1.89-1.40(m,19H),1.38-1.02(m,15H),1.02-0.76(m,18H),0.69(s,1H),0.05(dd,J=8.2,5.1Hz,6H)。
Step 2: (27E, 29E,31E,32E,38R,39S,40R,41R,43S,45S,48S,49R,50R, 59R) -48- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-49,59-dihydroxy-50-methoxy-47- [2- (2-methoxyethoxy) ethoxy ]]-38,39,40,41,51,52-hexamethyl-69, 70-dioxa-61-azatricyclohexa-27, 29,31 (51), 32 (52) -tetraen-53,54,55,56,57-pentanone. At 50℃under N 2 Next, the reaction mixture was purified to give (27E, 29E,31E,32E,34R,35S,36R,37R,39S,41S,43S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl group]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 55-dihydroxy-43, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-65, 66-dioxa-57-azatricyclo-hexa-hexadecane-27, 29,31 (47), 32 (48) -tetraene-49,50,51,52,53-pentanone (1.7 g,1.65 mmol) and 2- (2-methoxyethoxy) ethanol (3.97 g,33.06 mmol) to a solution of sulfolane (20 mL) was added HND-8 (255 mg), and the resulting solution was stirred at 50℃for 2 hours. The reaction mixture was poured into water, extracted with EtOAc, and water was used And brine, washed with Na 2 SO 4 Dried, filtered and concentrated. The residue was chromatographed on silica gel (50% EtOAc/petroleum ether) followed by reverse phase chromatography (85% CH) 3 CN/water) to give the title compound (950 mg,51% yield) as a white solid. ESI-MS (EI) + ,m/z):1138.2[M+Na] +
Step 3: (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,49S,50R,51R, 60R) -49- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-60-hydroxy-50, 51-dimethoxy-48- [2- (2-methoxyethoxy) ethoxy ]]-39,40,41,42,52,53-hexamethyl-69, 70-dioxa-62-azatricyclohexa-28, 30,32 (52), 33 (53) -tetraen-54, 55,56,57, 58-pentanone. At room temperature, to (27E, 29E,31E,32E,38R,39S,40R,41R,43S,45S,48S,49R,50R, 59R) -48- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl) silyl]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-49,59-dihydroxy-50-methoxy-47- [2- (2-methoxyethoxy) ethoxy ]]-38,39,40,41,51,52-hexamethyl-69, 70-dioxa-61-azatricyclohexa-hexadecane-27, 29,31 (51), 32 (52) -tetraene-53,54,55,56,57-pentanone (0.5 g,0.448 mmol) to a solution of toluene (15 mL) were added N1, N1, N8, N8-tetramethylnaphthalene-1, 8-diamine (1.92 g,8.96 mmol) and methyl triflate (1.10 g,6.72 mmol). The resulting solution was stirred at 50 ℃ for 3 hours, then filtered and concentrated. The residue was purified by silica gel chromatography (eluting with 0% to 50% EtOAc/petroleum ether) followed by reverse phase chromatography (CH 3 CN/water=0% to 100%) to give the title compound as a pale yellow solid (160 mg,32% yield). ESI-MS (EI) + ,m/z):1152.2[M+Na] +
Step 4: (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,44S,45R,46R, 55R) -55-hydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 46-dimethoxy-43- [2- (2-methoxyethoxy) ethoxy ]]-34,35,36,37,47,48-hexamethyl-64, 65-dioxa-56-azatricyclohexa-23, 25,27 (47), 28 (48) -tetraen-49,50,51,52,53-pentanone. At 0℃to (28E, 30E,32E,33E,39R,40S,41R,42R,44S,46S,49S,50R,51R, 60R) -49- [ (1R) -2- [ (1S, 3R, 4R) -4- [ tert-butyl (dimethyl)Radical) silyl groups]Oxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-60-hydroxy-50, 51-dimethoxy-48- [2- (2-methoxyethoxy) ethoxy ]]-39,40,41,42,52,53-hexamethyl-69, 70-dioxa-62-azatricyclohexa-hexadecane-28, 30,32 (52), 33 (53) -tetraene-54, 55,56,57, 58-pentanone (0.58 g,0.513 mmol) to a solution of THF (20 mL) was added Py HF (2.54 g,25.65 mmol). The reaction was stirred at room temperature for 3 hours, then with DCM and saturated NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography (75% CH 3 CN/water) to give the title compound as a white solid (200 mg,38% yield). ESI-MS (EI) + ,m/z):1038.1[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.49-5.79(m,4H),5.69-5.03(m,4H),4.62(d,J=13.2Hz,1H),4.00-3.07(m,28H),3.02-2.47(m,6H),2.41-1.68(m,16H),1.54-1.21(m,11H),1.17-0.82(m,18H),0.79-0.55(m,1H)。
Step 5: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) -46- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57-hydroxy-47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ]]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-hexadecane-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-32). At 0℃to (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,44S,45R,46R, 55R) -55-hydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45, 46-dimethoxy-43- [2- (2-methoxyethoxy) ethoxy ]]To a solution of-34,35,36,37,47,48-hexamethyl-64, 65-dioxa-56-azatricyclohexa-hexadecane-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone (0.18 g,0.177 mmol) in DCM (3 mL) was added 2, 6-di-tert-butyl-4-methylpyridine (0.273 g,1.33 mmol) and dimethylphosphonyl chloride (0.1 g,0.89mmol, dissolved in 0.5mL of DCM. The resulting solution was stirred at 0deg.C for 3.5 hours, then diluted with EtOAc and saturated NaHCO 3 Aqueous wash, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography (CH 3 CN/water) to give the purified product in the form ofI-32 as a white solid (90 mg,47% yield). ESI-MS (EI) + ,m/z):1114.1[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.49-5.81(m,4H),5.74-4.96(m,4H),4.67-4.03(m,2H),4.00-3.01(m,29H),2.99-2.46(m,4H),2.44-1.73(m,17H),1.59-1.22(m,15H),1.19-0.83(m,18H),0.82-0.59(m,1H)。
Step 6: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 57R) -46- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -57-hydroxy-47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ] -36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexadecane-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-34) and (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,45R,46S,47R, 48R) -46- [ (1R) -2- [ (1S, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl ] -1-methyl-58-aza-hexadecane-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-34) and (25E, 27E,29E, 30R, 37S, 39R,41S,43S, 45R) -46- [ (1R, 3-methoxy-cyclohexyl ] -1-methyl-hydroxy-47, 4-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ] -1-methyl-cyclohexyl ] -58-oxa-methyl-oxa-58-aza-methyl-75-oxa-75-one (29). 125mg of the epimeric mixture was separated by chiral preparative HPLC and then purified by silica gel chromatography (hexane: DCM: etOAc: meOH=3:3:1:0 to 3:3:1:0.3) to give I-34 as a white solid (25 mg,20% yield) and I-33 as a white solid (15 mg,12% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 1.2mg/mL mobile phase solution
And (3) sample injection: 10mL
Mobile phase: hexane/etoh=40/60 (V/V)
Flow rate: 25mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-34:ESI-MS(EI + ,m/z):1114.1[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.41-6.01(m,3H),5.87(dd,J=83.6,10.7Hz,1H),5.57-5.40(m,1H),5.38-4.97(m,3H),4.57(s,1H),4.02(d,J=20.9Hz,1H),3.92-3.62(m,3H),3.61-2.94(m,26H),2.78-2.40(m,3H),2.29-1.79(m,9H),1.60-1.38(m,15H),1.36-1.11(m,9H),1.08-0.76(m,18H),0.75-0.64(m,1H)。
I-33:ESI-MS(EI + ,m/z):1114.1[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.48-5.79(m,4H),5.63-5.02(m,4H),4.56(d,J=62.6Hz,1H),3.99-3.09(m,28H),3.01-2.49(m,5H),2.40-1.72(m,18H),1.54-1.19(m,14H),1.18-0.81(m,19H),0.78-0.59(m,1H)。
Example 19: (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) a- 57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl Base group]-47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ]]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-23, 25,27 (49), 28 (50) -tetraen-51,52,53,54,55-pentanone (I) 35 (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 57R) -57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ]]-1-methyl-ethyl]-47, 48-dimethyl Oxy-45- [2- (2-methoxyethoxy) ethoxy]36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58- Azatricyclohexa-hexa-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-37) and (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,45R,46S,47R,48R, 57R) -57-hydroxy-46- [ (1R) -2 ] [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl radical ]-1-methyl-ethyl]-47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ]]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclic Synthesis of tricetyl-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-36)
Step 1: (36E, 38E,40E,41E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclo-hexa-hexadecane-36, 38,40 (60), 41 (61) -tetraen-62,63,64,65,66-pentanone. At room temperature, at N 2 Next, the reaction mixture was purified to give (35E, 37E,39E,40E,46R,47S,48R,49R,51S,53S,55S,56S,57R,58R, 67R) -56- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl group]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]To a suspension of-57, 67-dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclohexa-hexadecane-35, 37,39 (59), 40 (60) -tetralin-61, 62,63,64, 65-pentanone (intermediate IX prepared according to example 22, 1.8g,1.5 mmol) and 1, 8-bis (dimethylamino) naphthalene (6.45 g,30.08 mmol) in toluene (40 mL) was added methyl triflate (3.70 g,22.56 mmol) dropwise. After addition, the mixture was heated to 50 ℃ for 5 hours, then the mixture was quenched by addition of water (50 mL) and extracted with EtOAc (50 mL) at 0 ℃. The organic layer was washed with water (50 ml×3) and brine (50 ml×3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether: etoac=3:1) to give the title compound (700 mg,38% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1232.2[M+Na] +1 HNMR(400MHz,CDCl 3 )δ7.70-7.68(m,4H),7.43-7.26(m,6H),6.40-5.87(m,4H),5.68-5.07(m,4H),4.67(s,1H),4.48-4.13(m,1H),3.81-3.57(m,7H),3.47-3.33(m,5H),3.20-3.08(m,7H),3.07-2.97(m,1H),2.71-2.50(m,2H),2.35-2.20(m,2H),2.09-1.97(m,3H),1.70-1.66(m,6H),1.61-1.58(m,11H),1.38-1.20(m,10H),1.15-1.10(m,5H),1.09-1.05(m,10H),0.98-0.73(m,13H),0.71-0.66(m,1H)。
Step 2: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41,43, 44-trimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-23, 25,27 (45), 28 (46) -tetraen-47,48,49,50,51-pentanone. At the position ofAt 0℃to (36E, 38E,40E,41E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-68-hydroxy-56,58,59-trimethoxy-47,48,49,50,60,61-hexamethyl-77, 78-dioxa-70-azatricyclohexa-hexadecane-36, 38,40 (60), 41 (61) -tetraene-62,63,64,65,66-pentanone (0.7 g,0.578 mmol) to a solution of THF (7 mL) was added Py-HF (0.457 g,5.78 mmol). The mixture was stirred at 30 ℃ for 3 hours, then by addition of saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with EtOAc (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether: acetone=3:1) to give the title compound (250 mg,44% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):995.0[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.48-6.01(m,4H),5.71-5.08(m,4H),4.68(s,1H),4.50-4.08(m,1H),3.83-3.55(m,7H),3.45-3.08(m,17H),3.00-2.51(m,2H),2.40-2.32(m,2H),2.16-1.97(m,3H),1.75-1.58(m,15H),1.30-1.24(m,6H),1.15-1.10(m,5H),0.98-0.82(m,17H),0.78-0.68(m,1H)。
Step 3: (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) -57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ]]-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexa-hexadecane-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-35). At 0℃to (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -53-hydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl)]-1-methyl-ethyl]To a solution of-41, 43, 44-trimethoxy-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclo-hexa-hexadecane-23, 25,27 (45), 28 (46) -tetraene-47,48,49,50,51-pentanone (0.25 g,0.257 mmol) and 2- (2-methoxyethoxy) ethanol (0.618 g,5.14 mmol) in THF (4 mL) was added HND-8 (80 mg). The mixture was stirred at 50 ℃ for 4 hours, then by addition of saturated NaHCO 3 Aqueous (20 mL) was quenched and extracted with EtOAc (30 mL) at 0deg.CTaking. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase (85% CH 3 CN/water) to give I-35 (0.12 g,44% yield) as a white solid. ESI-MS (EI) + ,m/z):1082.8[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.42-5.98(m,4H),5.85-5.08(m,4H),4.72-4.65(m,1H),4.51-4.10(m,1H),3.83-3.75(m,2H),3.65-3.55(m,7H),3.40-3.06(m,17H),2.71-2.46(m,2H),2.40-2.20(m,2H),2.15-1.88(m,3H),1.75-1.58(m,21H),1.42-1.30(m,5H),1.19-1.00(m,13H),0.97-0.82(m,10H),0.78-0.68(m,1H)。
Step 4: (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 57R) -57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -47, 48-dimethoxy-45- [2- (2-methoxyethoxy) ethoxy ] -36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclohexadecane-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-37) and (23E, 25E,27E,28E,36R,37S,38R,39R,41S,43S,45R,46S,47R, 48R) -57-hydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ] -1-methyl-58-azatricyclohexadecane-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-37) and (23E, 25E,27E,28E,36R,37S, 39R, 45R, 41S, 45R,46S, 47R) -57-hydroxy-46- [ (1R) -2-methoxy-ethyl ] -47. 140mg of the epimeric mixture was purified by preparative chiral HPLC to give I-37 as a white solid (30 mg,30% yield) and I-36 as a white solid (30 mg,30% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 4mg/mL mobile phase solution
And (3) sample injection: 5mL of
Mobile phase: hexane/etoh=70/30 (V/V)
Flow rate: 30mL/min
Wavelength: UV 254nm
Temperature: 38 DEG C
I-37:ESI-MS(EI + ,m/z):1081.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.52-6.10(m,3H),5.96(dd,J=62.3,11.6Hz,1H),5.62(ddd,J=40.8,14.6,7.8Hz,1H),5.24(ddd,J=66.7,18.2,10.9Hz,3H),4.68(s,1H),3.93-3.52(m,9H),3.51-3.03(m,17H),3.01-2.49(m,3H),2.40-1.63(m,24H),1.53-1.18(m,12H),1.18-0.81(m,18H),0.78-0.62(m,1H)。
I-36:ESI-MS(EI + ,m/z):1081.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.56-5.81(m,4H),5.75-5.15(m,4H),4.01-3.51(m,16H),3.51-3.06(m,20H),2.85-2.49(m,2H),2.45-1.64(m,18H),1.47-1.19(m,10H),1.17-0.61(m,19H)。
Example 20: (26E, 28E,30E,31E,34R,35S,36R,37R,39S,41S,44S,46R,47R, 56R) a- 44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-43- (1, 4-dioxane-2-ylmethyl) Oxy) -56-hydroxy-46, 47-dimethoxy-34,35,36,37,48,49-hexamethyl-66, 67-dioxa-57-azatris Cyclotriacontane-26, 28,30 (48), 31 (49) -tetraene-50,51,52,53,54-pentanone (I-38), (26E, 28E,30E, 31E,34R,35S,36R,37R,39S,41S,43S,44S,46R,47R,56R)-44-[(1R)-2-[(1S,3R,4R)-3,4- dimethoxy cyclohexyl]-1-methyl-ethyl]-43- (1, 4-dioxan-2-ylmethoxy) -56-hydroxy-46, 47-dimethoxy Base-34,35,36,37,48,49-hexamethyl-66, 67-dioxa-57-azatricyclo-tricetyl-26, 28,30 (48), 31 (49) Tetraene-50,51,52,53,54-pentanone (I-40) and (26E, 28E,30E,31E,34R,35S,36R,37R,39S, 41S,43R,44S,46R,47R, 56R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl Base group]-43- (1, 4-dioxan-2-ylmethoxy) -56-hydroxy-46, 47-dimethoxy-34,35,36,37,48,49-hexamethyl Radical-66, 67-dioxa-57-azatricyclohexa-hexa-26, 28,30 (48), 31 (49) -tetraene-50,51,52,53,54- Synthesis of pentanone (I-39)
Step 1: (26E, 28E,30E,31E,34R,35S,36R,37R,39S,41S,44S,46R,47R, 56R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ]-1-methyl-ethyl]-43- (1, 4-dioxan-2-ylmethoxy) -56-hydroxy-46, 47-dimethoxy-34,35,36,37,48,49-hexamethyl-66, 67-dioxa-57-azatricyclohexa-hexa-ne-26, 28,30 (48), 31 (49) -tetraen-50,51,52,53,54-pentanone (I-38). At 50℃under N 2 Next, the reaction mixture was purified to (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]To a solution of 52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraen-46,47,48,49,50-pentanone (0.05 g,0.053 mmol) and 2- (oxetan-3-yloxy) ethanol (0.125 g,1.06 mmol) in THF (5 mL) was added HND-8 (0.02 g). The reaction mixture was stirred at 50 ℃ for 16 hours, then cooled, filtered and concentrated. The residue was purified by reverse phase chromatography (with 80% ch 3 CN/water elution) to give I-38 (0.019 g,35% yield) as a white solid. ESI-MS (EI) + ,m/z):1050.1[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.44-5.98(m,4H),5.69-5.01(m,4H),4.66-4.27(m,2H),3.89-3.56(m,9H),3.44-3.31(m,10H),3.28-3.21(m,3H),3.07-2.96(m,7H),2.95-2.51(m,4H),2.34-1.82(m,7H),1.77-1.48(m,27H),1.44-1.22(m,8H),1.20-1.01(m,13H),1.01-0.88(m,8H),0.85-0.65(m,2H)。
Step 2: (26E, 28E,30E,31E,34R,35S,36R,37R,39S,41S,43S,44S,46R,47R, 56R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -43- (1, 4-dioxan-2-ylmethoxy) -56-hydroxy-46, 47-dimethoxy-34,35,36,37,48,49-hexamethyl-66, 67-dioxa-57-azatricyclohexadecane-26, 28,30 (48), 31 (49) -tetraene-50,51,52,53,54-pentanone (I-40) and (26E, 28E,30E,31E,34R,35S,36R,37R,39S,41S,43R,44S,46R,47R, 56R) -44- [ (1R) -2- [ (1S, 3R, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -43- (1, 4-dioxan-2-ylmethoxy) -66, 67-dioxan-57-azatricyclohexadecane-26, 28,30 (48), 31 (49) -tetraene-50,51,52,53,54-pentanone (I-40) and (26E, 28E,30E,31E, 35R, 37R,39S, 43R. 140mg of the epimeric mixture was purified by preparative chiral HPLC to give I-40 as a white solid (36.6 mg,26% yield) and I-39 as a white solid (17.2 mg,12% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 2mg/mL mobile phase solution
And (3) sample injection: 5mL of
Mobile phase: hexane/etoh=70/30 (V/V)
Flow rate: 30mL/min
Wavelength: UV 254nm
Temperature: 38 DEG C
I-40:ESI-MS(EI + ,m/z):1049.8[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.47-5.80(m,4H),5.75-5.50(m,1H),5.49-5.04(m,3H),4.69-4.41(m,1H),4.36-4.11(m,1H),3.91-3.50(m,10H),3.48-2.99(m,19H),2.79-2.51(m,2H),2.38-1.85(m,7H),1.83-1.58(m,12H),1.53-1.17(m,10H),1.14-0.84(m,18H),0.75(d,J=10.9Hz,1H)。
I-39:ESI-MS(EI + ,m/z):1049.8[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.60-5.70(m,4H),5.66-5.01(m,4H),4.72-4.14(m,2H),4.10-3.50(m,9H),3.49-2.98(m,18H),2.59(dd,J=79.6,49.4Hz,3H),2.40-1.64(m,19H),1.52-1.20(m,10H),1.19-0.65(m,20H)。
Example 21: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) a- 45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy Radical-44- [2- (2-methoxyethylsulfonyl) ethoxy]35,36,37,38,48,49-hexamethyl-66, 67-dioxa- 57-azatricyclohexa-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-41), (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R,56R)-45-[(1R)-2-[(1S,3R, 4R) -3, 4-Dimethoxycyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethyl) Alkylsulfonyl) ethoxy]35,36,37,38,48,49-hexamethyl-66, 67-dioxa-57-azatricyclo-hexa-hexadecane- 24,26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-43) and (24E, 26E,28E,29E,35R,36S, 37R,38R,40S,42S,44R,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl Base group]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethylsulfonyl) ethoxy ] ]-35, 36,37,38,48,49-hexamethyl-66, 67-dioxa-57-azatricyclo-tricetyl-24, 26,28 (48), 29 (49) -tetralin Synthesis of alkene-50,51,52,53,54-pentanone (I-42)
Step 1: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethylsulfonyl) ethoxy ]]-35,36,37,38,48,49-hexamethyl-66, 67-dioxa-57-azatricyclohexa-hexadecane-24, 26,28 (48), 29 (49) -tetraen-50,51,52,53,54-pentanone (I-41). At 0℃to (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]To a solution of 52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone (intermediate I,0.15g,0.16 mmol) and 2- (2-methoxyethylsulfonyl) ethanol (0.268 g,1.59 mmol) in THF (5 mL) was added HND-8 (50 mg). The mixture was stirred at 50 ℃ for 10 hours, then by addition of saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with EtOAc (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (85% CH 3 CN/water) to give I-41 (44 mg,26% yield) as a white solid. ESI-MS(EI + ,m/z):1100.0[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.50-5.96(m,4H),5.80-5.02(m,4H),4.83-4.75(m,1H),4.76-4.39(m,1H),3.85-3.80(m,2H),3.75-3.53(m,4H),3.45-3.10(m,17H),3.09-2.85(m,3H),2.81-2.48(m,3H),2.35-1.85(m,7H),1.76-1.57(m,21H),1.39-1.22(m,5H),1.17-0.83(m,18H),0.79-0.66(m,1H)。
Step 2: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -56-hydroxy-46, 47-dimethoxy-44- [2- (2-methoxyethylsulfonyl) ethoxy ] -35,36,37,38,48,49-hexamethyl-66, 67-dioxa-57-azatricyclo-hexadecane-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-43) and (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44R,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -56-hydroxy-46, 47-aza-tricyclo-hexadecane-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-43) and (24E, 26E, 29E,35R,36S,37R, 40S, 44R,45S, 45R, 46R) -45- [ (1R) -2- [ (1S, 3R, 4-dimethoxycyclohexyl ] -1-methyl-hydroxy-46-oxa-2-oxa-sulfonyl) -52-oxa-2-trione (52-oxa-75-oxa-trione (52). 140mg of the epimeric mixture was purified by preparative chiral HPLC to give I-43 as a white solid (18 mg,20% yield) and I-42 as a white solid (26 mg,29% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 2.5mg/mL mobile phase solution
And (3) sample injection: 8mL of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 40mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-43:ESI-MS(EI + ,m/z):1099.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.48-5.83(m,4H),5.56(dd,J=14.8,8.1Hz,1H),5.49-5.02(m,3H),4.75(s,1H),3.91-3.51(m,9H),3.46-3.18(m,18H),3.16-2.98(m,6H),2.96-2.45(m,3H),2.38-1.66(m,17H),1.54-1.16(m,13H),1.25-0.65(m,19H)。
I-42:ESI-MS(EI + ,m/z):1100.0[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.65-5.86(m,4H),5.75-5.02(m,5H),4.81-4.31(m,2H),4.08-2.99(m,34H),2.97-2.49(m,4H),2.45-1.65(m,17H),1.51-0.53(m,25H)。
Example 22: (23E, 25E,27E,28E,35R,36S,37R,38R,40S,42S,44S,45S,47R,48R, 57R) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1- Methyl-ethyl group]-48-methoxy-35,36,37,38,49,50-hexamethyl-44- [2- (oxetan-3-yloxy) ethoxy Base group]-68, 69-dioxa-58-azatricyclohexa-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55- Pentanone (I-45) and (23E, 25E,27E,28E,35R,36S,37R,38R,40S,42S,44R,45S,47R,48R, 57R) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl Base group]-48-methoxy-35,36,37,38,49,50-hexamethyl-44- [2- (oxetan-3-yloxy) ethoxy ]]-68, 69-dioxa-58-azatricyclohexa-23, 25,27 (49), 28 (50) -tetraen-51,52,53,54,55-pentanone (I) 44 ) synthesis of
Step 1:3- [ tert-butyl (diphenyl) silyl group]Oxypropyl triflate. At 0℃under N 2 Down to 3- [ tert-butyl (diphenyl) silyl group]To a mixture of oxypropan-1-ol (7 g,22.26 mmol) and DIPEA (5.82 mL,33.39 mmol) in DCM (80 mL) was added trifluoromethylsulfonyl triflate (6.91 g,24.48 mmol) and the reaction stirred at 0 ℃ for 2 hours. The mixture was diluted with DCM (150 mL) and saturated NaHCO 3 (50 mL), water (50 mL), and brine (50 mL). The organic layer was purified by Na 2 SO 4 Drying, filtering and concentrating to obtain 3- [ tert-butyl (diphenyl) as brown oilRadical) silyl groups]Oxypropyl triflate (9.9 g,99.6% yield). The crude material was used in the next step without further purification. 1 H NMR(400MHz,CDCl 3 )δ7.67-7.63(m,4H),7.47-7.37(m,6H),4.77-4.73(t,J=6Hz,2H),3.79-3.75(t,J=6Hz,2H),2.04-1.98(m,2H),1.06(s,1H)。
Step 2: (35E, 37E,39E,40E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ tert-butyl (diphenyl) silyl]Oxypropoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-58, 68-dihydroxy-56,59-dimethoxy-47,48,49,50,60,61-hexamethyl-78, 79-dioxa-70-azatricyclohexa-hexa-hexadecane-35, 37,39 (60), 40 (61) -tetraene-62,63,64,65,66-pentanone. (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 50R) -40, 50-dihydroxy-39- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-51-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraen-44,45,46,47,48-pentanone (2 g,2.19 mmol), 3- [ tert-butyl (diphenyl) silyl]A mixture of oxypropyl triflate (9.77 g,21.88 mmol) and N-ethyl-N-isopropyl-propan-2-amine (4.57 mL,26.25 mmol) in toluene (40 mL) was stirred at 58℃for 18 hours. The mixture was poured into ice-cold saturated NaHCO 3 In (150 mL), extracted with EtOAc (200 mL) and the organic layer was washed with water (150 mL. Times.3) and brine (150 mL), over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether: ea=3:1) to give the title compound (1.8 g,68% yield) as a yellow solid. ESI-MS (EI) + ,m/z):1232.7[M+Na] +
Step 3: (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-63, 64-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraen-47,48,49,50,51-pentanone. To (35E, 37E,39E,40E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ tert-butyl (diphenyl) silyl]Oxypropoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-58, 68-dihydroxy-56,59-dimethoxy-47,48,49,50,60,61-hexamethyl-78, 79-dioxa-70-azatricyclohexa-hexadecane-35, 37,39 (60), 40 (61) -tetraene-62,63,64,65,66-pentanone (1.8 g,1.49 mmol) to a solution of THF (15 mL) was added pyridine HF (1.2 mL,14.87 mmol), and the reaction was stirred at 30 ℃ for 3 hours. The mixture was purified by addition of saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with EA (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (petroleum ether: acetone=3:1) to give the title compound (1.1 g,76% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):994.7[M+Na] +
Step 4: (23E, 25E,27E,28E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-48-methoxy-35,36,37,38,49,50-hexamethyl-44- [2- (oxetan-3-yloxy) ethoxy ]]-68, 69-dioxa-58-azatricyclohexa-23, 25,27 (49), 28 (50) -tetraen-51,52,53,54,55-pentanone. At-40℃under nitrogen, to (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-63, 64-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone (0.2 g,0.206 mmol) to a solution of DCM (4 mL) was added TFA (0.32 mL,4.11 mmol) followed by 2- (oxetan-3-yloxy) ethanol (0.49 g,4.11 mmol). The reaction was stirred at-40 ℃ for 3 hours and then poured into ice-cold saturated NaHCO 3 In aqueous solution and extracted with DCM, dried, filtered and concentrated. The residue was then purified by reverse phase chromatography (with 80% CH 3 CN/water elution) to give the title compound as a white solid (30 mg,14% yield). ESI-MS (EI) + ,m/z):1080.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.41-5.92(m,4H),5.57-5.08(m,4H),4.70-4.55(m,5H),4.35-4.0(m,3H),3.92-3.69(m,5H),3.68-3.54(m,3H),3.53-3.30(m,7H),3.29-2.98(m,4H),2.88-2.40(m,4H),2.38-2.25(m,2H),2.22-1.90(m,5H),1.87-1.57(m,17H),1.54-1.16(m,10H),1.15-0.83(m,17H),0.76-0.62(m,1H)。
Step 5: (23E, 25E,27E,28E,35R,36S,37R,38R,40S,42S,44S,45S,47R,48R, 57R) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -48-methoxy-35,36,37,38,49,50-hexamethyl-44- [2- (oxetan-3-yloxy) ethoxy ] -68, 69-dioxa-58-azatricyclohexadecane-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-45) and (23E, 25E,27E,28E,35R,36S,37R,38R,40S,42S, R,45S,47R, 57R) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl ] -68, 69-dioxa-58-azatricyclohexadecane-23, 25,27 (49), 28 (50) -tetraene-51,52,53,54,55-pentanone (I-45) and (23E, 25E,27E, 35R,36S,37R,38R,40S, 42R, 47R. 139mg (23E, 25E,27E,28E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -48-methoxy-35,36,37,38,49,50-hexamethyl-44- [2- (oxetan-3-yloxy) ethoxy ] -68, 69-dioxa-58-azatricyclohexa-hexadecane-23, 25,27 (49), 28 (50) -tetraen-51,52,53,54,55-pentanone were isolated by chiral preparative HPLC and purified by silica gel chromatography (13% MeOH/petroleum ether: DCM: EA=3:3:1) to give I-45 as a white solid (30 mg,22% yield) and I-44 as a white solid (17 mg,12% yield).
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 2.5cm inside diameter x 25cm length, 10 μm
Sample solution: 2mg/mL mobile phase solution
And (3) sample injection: 8mL of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 23mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-45:ESI-MS(EI + ,m/z):1079.9[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.46-5.83(m,4H),5.62-5.02(m,4H),4.87-4.51(m,6H),4.17(d,J=5.0Hz,1H),3.94-2.96(m,24H),2.90-2.52(m,3H),2.41-1.71(m,15H),1.62-1.40(m,8H),1.39-1.18(m,7H),1.15-0.79(m,18H),0.76-0.65(m,1H)。
I-44:ESI-MS(EI + ,m/z):1079.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.48-5.81(m,4H),5.75-5.08(m,4H),4.87-4.53(m,5H),4.40-4.11(m,2H),4.06-3.71(m,5H),3.70-2.89(m,24H),2.87-1.74(m,17H),1.55-1.17(m,11H),1.16-0.82(m,18H),0.73-0.65(m,1H)。
Example 23: (24E, 26E,28E,29E,37R,38S,39R,40R,42S,44S,47S,48R,49R, 58R) a- 58-hydroxy-48, 49-dimethoxy-46- [2- (2-methoxyethoxy) ethoxy ]]-47-[(1R)-2-[(1S,3R,4R)- 3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-37,38,39,40,50,51-hexamethyl-66, 67-dioxa-59-azatricyclohexa-24, 26,28 (50), 29 (51) -tetraen-52,53,54,55,56-pentanone (I) 46 (24E, 26E,28E,29E,37R,38S,39R,40R,42S,44S,46S,47S,48R,49R, 58R) -58-hydroxy- 48, 49-dimethoxy-46- [2- (2-methoxyethoxy) ethoxy ]]-47- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy- ] 4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]37,38,39,40,50,51-hexamethyl-66, 67-dioxa- Synthesis of 59-azatricyclohexa-24, 26,28 (50), 29 (51) -tetraene-52,53,54,55,56-pentanone (I-47)
Step 1: (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone. At room temperatureUnder N 2 Next, the reaction mixture was purified to give (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-41, 44-dimethoxy-42- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl group]-1-methyl-ethyl]To a solution of (32,33,34,35,45,46) -hexamethyl-62, 63-dioxa-54-azatricyclohexa-hexadecane-23, 25,27 (45), 28 (46) -tetralin-47,48,49,50,51-pentanone (intermediate III,1.4g,1.44 mmol) and 1, 8-bis (dimethylamino) naphthalene (4.63 g,21.6 mmol) in toluene (24 mL) was added dropwise methyl triflate (2.36 g,14.4mmol,1.58 mL). After addition, the mixture was heated to 50deg.C for 3 hours, then filtered, diluted with EtOAc (60 mL) and saturated NH 4 Aqueous Cl (60 mL. Times.2), water (60 mL) and brine (60 mL). The organic layer was dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by silica gel chromatography (petroleum ether: etoac=7:3) and reverse phase chromatography (with 80% ch 3 CN/water elution) to give the title compound as a white solid (0.22 g,15% yield). ESI-MS (EI+, m/z): 1009.5[ M+Na ] ] +
Step 2: (24E, 26E,28E,29E,37R,38S,39R,40R,42S,44S,47S,48R,49R, 58R) -58-hydroxy-48, 49-dimethoxy-46- [2- (2-methoxyethoxy) ethoxy]-47- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-37,38,39,40,50,51-hexamethyl-66, 67-dioxa-59-azatricyclohexa-hexadecane-24, 26,28 (50), 29 (51) -tetraene-52,53,54,55,56-pentanone (I-46). At 50℃under N 2 Next, (24E, 26E,28E,29E,33R,34S,35R,36R,38S,40S,42S,43S,44R,45R, 54R) -54-hydroxy-42, 44, 45-trimethoxy-43- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]To a solution of-33,34,35,36,46,47-hexamethyl-62, 63-dioxa-55-azatricyclohexa-hexadecane-24, 26,28 (46), 29 (47) -tetraene-48,49,50,51,52-pentanone (0.1 g,0.101 mmol) and 2- (2-methoxyethoxy) ethanol (0.244 g,2.03 mmol) in THF (10 mL) was added HND-8 (0.04 g). The reaction mixture was stirred at 50 ℃ for 20 hours, then cooled, filtered and the filtrate was poured into saturated NaHCO at 0 ℃ 3 Aqueous (20 mL) and extracted with EtOAc (15 mL). The organic layer was washed with water (15 mL) and brine (15 mL), dried over anhydrous sodium sulfate, filtered and concentrated . The residue was purified by silica gel chromatography (EtOAc: petroleum ether=4:1) to give I-46 (0.065 g,60% yield) as a white solid. ESI-MS (EI) + ,m/z):1095.8[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.43-5.84(m,4H),5.72-5.06(m,4H),4.84-4.17(m,2H),3.96-3.73(m,4H),3.70-3.52(m,10H),3.50-3.43(m,4H),3.41-3.30(m,8H),3.29-3.20(m,3H),3.18-2.99(m,5H),2.96-2.50(m,4H),2.35-2.14(m,3H),2.05-1.84(m,5H),1.80-1.56(m,21H),1.55-1.23(m,10H),1.16-1.00(m,11H),0.97-0.84(m,9H),0.81-0.69(m,1H)。
Step 3: (24E, 26E,28E,29E,37R,38S,39R,40R,42S,44S,46S,47S,48R,49R, 58R) -58-hydroxy-48, 49-dimethoxy-46- [2- (2-methoxyethoxy) ethoxy ] -47- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl ] -1-methyl-ethyl ] -37,38,39,40,50,51-hexamethyl-66, 67-dioxa-59-azatricyclo-hexadecane-24, 26,28 (50), 29 (51) -tetraen-52,53,54,55,56-pentanone (I-47). 50mg of the epimeric mixture was purified by preparative chiral HPLC followed by silica gel chromatography (petroleum ether: DCM: etOAc: meOH=3:1:0.2) to give I-47 (13 mg,26% yield) as a white solid.
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 0.55mg/mL mobile phase solution
And (3) sample injection: 15mL
Mobile phase: hexane/etoh=70/30 (V/V)
Flow rate: 30mL/min
Wavelength: UV 254nm
Temperature: 38 DEG C
ESI-MS(EI+,m/z):1095.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.44-5.72(m,4H),5.72-4.98(m,4H),3.96-3.14(m,32H),3.05(d,J=7.9Hz,5H),2.76-2.42(m,3H),2.37-1.57(m,22H),1.46-1.17(m,16H),1.14-0.77(m,18H),0.73-0.61(m,1H)。
Example 24: (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,43S,44S,45R,46R, 55R) -55-hydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ]-1-methyl-ethyl]-45, 46-dimethoxy-43- [2- (2-methoxyethoxy) ethoxy]-34,35,36,37,47,48-hexamethyl-64, 65-dioxo Hetero-56-azatricyclohexa-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone (I-49) and (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,43R,44S,45R,46R, 55R) -55-hydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ]]-1-methyl-ethyl]45, 46-dimethoxy-43- [2- (2-methoxyethoxy) ethoxy ]]-34,35,36,37,47,48-hexamethyl-64, 65-dioxa-56-azatricyclic Synthesis of hexacosane-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone (I-48)
Step 1: (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,43S,44S,45R,46R, 55R) -55-hydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -45, 46-dimethoxy-43- [2- (2-methoxyethoxy) ethoxy ] -34,35,36,37,47,48-hexamethyl-64, 65-dioxa-56-azatricyclo-hexadecane-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone (I-49) and (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,43R,44S,45R,46R, 55R) -55-hydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ] -1-methyl-45-dioxa-45, 46-dimethoxy-45- (48-oxa-2-ethoxy) -62-pentanone (52, 27-oxa-48). 116mg of the epimeric mixture was separated by chiral preparative HPLC and then purified by silica gel chromatography (hexane: DCM: etOAc: meOH=3:3:1:0.4) to give I-49 (40 mg,34% yield) as a white solid and I-48 (35 mg,30% yield) as a white solid.
Chiral separation method
Chromatographic column: CHIRALPAK IC
Column dimensions: 5.0cm inside diameter by 25cm length, 10 μm
Sample solution: 0.7mg/mL mobile phase solution
And (3) sample injection: 18mL
Mobile phase: hexane/etoh=60/40 (V/V)
Flow rate: 60mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
I-49:ESI-MS(EI + ,m/z):1038.1[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.46-5.81(m,4H),5.75-5.02(m,4H),4.61(d,J=16.7Hz,1H),3.99-3.21(m,25H),3.21-3.06(m,3H),3.01-2.50(m,5H),2.41-1.68(m,14H),1.63-1.19(m,14H),1.17-0.82(m,18H),0.77-0.64(m,1H)。
I-48:ESI-MS(EI + ,m/z):1038.1[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.62-5.87(m,4H),5.77-5.02(m,4H),4.72-4.27(m,1H),3.99-3.06(m,28H),3.00-2.47(m,6H),2.43-1.70(m,15H),1.52-1.20(m,12H),1.18-0.79(m,18H),0.69(d,J=11.7Hz,1H)。
Example 25: (25E, 27E,29E,30E,34R,35S,36R,37R,39S,41S,44S,46R,47R, 56R) a- 44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy Phenyl-34,35,36,37,48,49-hexamethyl-43- [2- (oxetan-3-yloxy) ethoxy]65, 66-dioxa- 57-azatricyclohexa-25, 27,29 (48), 30 (49) -tetraene-50,51,52,53,54-pentanone (I-50)
Step 1: (25E, 27E,29E,30E,34R,35S,36R,37R,39S,41S,44S,46R,47R, 56R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-34,35,36,37,48,49-hexamethyl-43- [2- (oxetan-3-yloxy) ethoxy ]]-65, 66-dioxa-57-azatricyclohexa-25, 27,29 (48), 30 (49) -tetraen-50,51,52,53,54-pentanone (I)-50). at-55deg.C, at N 2 Next, the reaction mixture was purified to (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone (intermediate I,0.72g,0.76 mmol) 2, 2-trifluoroacetic acid (1.18 mL,15.28 mmol) was added dropwise to a solution of DCM (40 mL). After the addition, the reaction mixture was stirred at-45 ℃ for 10 min, then 2- (oxetan-3-yloxy) ethanol (1.81 g,15.28mmol, dissolved in DCM) was added to the reaction mixture at the same temperature. The reaction mixture was stirred at-45℃for 1 hour, then poured into saturated NaHCO at 0 ℃ 3 Aqueous solution (60 mL) and extracted with DCM (60 mL). The organic layer was washed with water (60 mL) and brine (60 mL), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated in vacuo. The residue was chromatographed on silica gel (100% EA) and then on reversed phase (67% CH) 3 CN/water elution) to give I-50 (0.07 g,9% yield) as a white solid. ESI-MS (EI) + ,m/z):1049.9[M+Na] + 。1H NMR(400MHz,CDCl 3 )δ6.41-6.01(m,4H),5.35-4.94(m,4H),4.78-4.57(m,5H),4.50-4.13(m,1H),3.89-3.58(m,4H),3.55-3.31(m,11H),3.28-3.201(m,4H),3.21-3.10(m,3H),3.07-2.97(m,2H),2.78-2.54(m,3H),2.30-2.27(m,2H),2.10-1.95(m,5H),1.79-1.48(m,13H),1.45-1.04(m,19H),0.97-0.84(m,8H)0.78-0.73(m,1H)。
Example 26: (24E, 26E,28E,29E,37R,38S,39R,40R,42S,44S,47S,48R,49R, 58R) a- 47- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-58-hydroxy-48, 49-dimethoxy Base-46- [2- [2- (2-methoxyethoxy) ethoxy ]]Ethoxy group]-37,38,39,40,50,51-hexamethyl-66, 67-di Oxa-59-azatricyclohexa-24, 26,28 (50), 29 (51) -tetraene-52,53,54,55,56-pentanone (I-51) Synthesis
Step 1: (24E, 26E,28E,29E,37R,38S,39R,40R,42S,44S,47S,48R,49R, 58R) -47- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-58-hydroxy-48, 49-dimethoxy-46- [2- [2- (2-methoxyethoxy) ethoxy ]]Ethoxy group]-37,38,39,40,50,51-hexamethyl-66, 67-dioxa-59-azatricyclohexa-hexadecane-24, 26,28 (50), 29 (51) -tetraene-52,53,54,55,56-pentanone (I-51). At 50℃under N 2 Next, the reaction mixture was purified to (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclo-hexa-hexadecane-24, 26,28 (44), 29 (45) -tetraen-46,47,48,49,50-pentanone (intermediate I,0.2g,0.212 mmol) and 2- [2- (2-methoxyethoxy) ethoxy]HND-8 (50 mg) was added to a solution of ethanol (0.349 g,2.12 mmol) in THF (5 mL). The resulting solution was stirred for 15 hours, then diluted with EtOAc, filtered, then washed with water, brine, over Na 2 SO 4 Dried, filtered again and concentrated. The residue was purified by silica gel chromatography (EtOAc: petroleum ether e=1:0.8) and reverse phase chromatography (85% ch) 3 CN/water) to give I-51 (40 mg,18% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1095.8[M+Na] +1 HNMR(400MHz,CDCl 3 )δ6.60-5.79(m,4H),5.76-5.06(m,4H),3.93-2.97(m,33H),2.92-2.49(m,3H),2.47-1.75(m,22H),1.51-0.63(m,29H)。
Example 27: (25E, 27E,29E,30E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) a- 45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy Phenyl-35,36,37,38,48,49-hexamethyl-44- [3- (1, 2, 4-triazol-4-yl) propoxy]-67, 68-dioxa-60-nitrogen Synthesis of Heterotricyclotricetyl-25, 27,29 (48), 30 (49) -tetraene-50,51,52,53,54-pentanone (I-52)
Step 1:3- (1, 2, 4-triazol-4-yl) propan-1-ol. A mixture of formylhydrazine (10 g,166.51 mmol) and diethoxymethoethane (29.61 g,199.82 mmol) in methanol (200 mL) was heated at reflux for 2 hours, followed by dropwise addition of 3-aminopropan-1-ol (12.51 g,166.51 mmol). The reaction was refluxed for 4 hours, then concentrated and purified by reverse phase chromatography (10% ch) 3 CN/water) then purified by silica gel chromatography (DCM: CH 3 Oh=12:1) to give the title compound as an off-white solid (20.6 g,97% yield). ESI-MS (EI+, m/z): 128.1[ M+H ]] + ,T=0.189min。 1 H NMR(400MHz,MeOD-d 4 )δ8.49(s,2H),4.18(t,J=7.0Hz,2H),3.48(t,J=5.9Hz,2H),2.00-1.90(m,2H)。
Step 2 (25E, 27E,29E,30E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-44- [3- (1, 2, 4-triazol-4-yl) propoxy]-67, 68-dioxa-60-azatricyclohexa-25, 27,29 (48), 30 (49) -tetraene-50,51,52,53,54-pentanone (I-52). To 3- (1, 2, 4-triazol-4-yl) propan-1-ol (0.22 g,1.75 mmol), (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone (intermediate I,0.33g,0.35 mmol) and TFA (0.48 g,4.20 mmol) were added to a mixture of DCM (20 mL) 3- (1, 2, 4-triazol-4-yl) propan-1-ol (0.22 g,1.75 mmol) and stirred at-30 ℃ for 3 hours. The mixture was poured into saturated NaHCO 3 In aqueous solution, and the organic layer was washed twice with water, then with brine. After concentration, the residue was purified by reverse phase chromatography (MeOH: dcm=1:15) to give I-52 (60 mg,17% yield). ESI-MS (EI+, m/z): 1038.3[ M+H ]] +1 H NMR(400MHz,CDCl 3 )δ8.36-7.82(m,1H),6.49-5.92(m,4H),5.75-4.96(m,5H),4.51-3.92(m,2H),3.64(ddd,J=34.7,33.2,24.8Hz,4H),3.48-3.20(m,11H),3.08(dd,J=38.8,18.3Hz,7H),2.92-2.42(m,5H),2.25(dd,J=76.9,68.3Hz,8H),1.94-1.46(m,19H),1.44-0.96(m,20H),0.96-0.62(m,9H)。
Example 28: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,42S,44R,45R, 55R) a- 42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-44,55-two Hydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy]- 65, 66-dioxa-56-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraen-48,49,50,51,52-pentanone (I-108)、(23E,25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,44R,45R,55R)-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]44,55-dihydroxyl Phenyl-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy]-65, 66-dioxa-56-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraen-48,49,50,51,52-pentanone (I) 105 (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S,44R,45R, 55R) -42- [ (1R)/(2R) 2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-44,55-dihydroxy-45-methyl ester Oxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy]-65, 66-dioxy Hetero-56-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraen-48,49,50,51,52-pentanone (I-104) Synthesis
Step 1: (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]-40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclohexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone. Rapamycin (2 g,2.19 mmol) was dissolved in DCM (30 mL) at room temperatureKHF is added into the liquid 2 (2.56 g,32.82mmol in 2mL of water) and bromodifluoro (trimethylsilyl) methane (4.44 g,21.88 mmol). The reaction mixture was stirred for 16 hours, then with DCM and NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=3:1) to give (22 e,24e,26e,27e,29R,30s,31R,32R,34s,36s,38s,39s,40R,41R, 51R) -39- [ (1R) -2- [ (1 s,3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl as a white solid]-1-methyl-ethyl]-40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (0.4 g,19% yield). ESI-MS (EI) + ,m/z):986.5[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.43-5.86(m,4H),5.58-5.07(m,4H),4.49(s,1H),4.18-4.09(m,2H),3.89-3.56(m,4H),3.47-3.28(m,7H),3.19-3.02(m,4H),2.90-2.55(m,3H),2.41-2.21(m,2H),2.20-1.91(m,6H),1.90-1.41(m,20H),1.40-1.13(m,7H),1.12-0.81(m,14H),0.80-0.67(m,1H)。
Step 2: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,42S,44R,45R, 55R) -42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]-44,55-dihydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ]]-65, 66-dioxa-56-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraene-48,49,50,51,52-pentanone (I-108). At-40℃under nitrogen, to (22E, 24E,26E,27E,29R,30S,31R,32R,34S,36S,38S,39S,40R,41R, 51R) -39- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-40, 51-dihydroxy-38, 41-dimethoxy-29,30,31,32,42,43-hexamethyl-60, 61-dioxa-52-azatricyclo-hexa-hexadecane-22, 24,26 (42), 27 (43) -tetraene-44,45,46,47,48-pentanone (0.3 g,0.31 mmol) to a solution of DCM (6 mL) was added TFA (0.71 g,6.22mmol,0.48 mL). The mixture was stirred for 10 minutes, then 2- (oxetan-3-yloxy) ethanol (0.74 g,6.22 mmol) was added and the mixture was stirred at-20 ℃ for 2 hours. The mixture was purified by addition of saturated NaHCO 3 Aqueous solution (20 mL) was quenched and quenched with DCM (30 mL) at 0deg.C) And (5) extracting. The organic layer was washed with water (20 mL), brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by reverse phase column chromatography (with 80% CH 3 CN/water elution) to give I-108 (50 mg,15% yield) as a white solid. ESI-MS (EI) + ,m/z):1072.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.41-5.93(m,4H),5.57-5.07(m,4H),5.82-4.53(m,5H),4.31-3.99(m,2H),3.93-3.65(m,3H),3.63-3.04(m,13H),2.90-2.27(m,5H),2.26-1.86(m,5H),1.85-1.55(m,17H),1.53-1.17(m,9H),1.16-0.77(m,17H),0.76-0.65(m,1H)。
Step 3: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,44R,45R, 55R) -42- [ (1R) -2- [ (1S, 3R, 4R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -44,55-dihydroxy-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ] -65, 66-dioxa-56-azatricyclohexadecane-23, 25,27 (46), 28 (47) -tetraene-48,49,50,51,52-pentanone (I-105), (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S, 45R, 55R) -42- [ (1S, 3R) -4- (difluoromethoxy) -3-methoxy-cyclohexyl ] -1-methyl-56-oxa-dioxa-56-azatricyclohexadecane-23, 25,27 (46), 28 (47) -tetraene-48,49,50,51,52-pentanone (I-105), (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R, 42R- [ (1S, 45R). 0.17g I-108 was isolated by chiral preparative HPLC and then purified by silica gel chromatography (8% meoh in PE mixture: DCM: ea=3:3:1) to give I-105 as a white solid (18 mg,11% yield) and I-104 as a white solid (15 mg,9% yield).
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-NA 012)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 10 μl of
Mobile phase: hexane/etoh=60/40 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-105:ESI-MS(EI + ,m/z):1072.6[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.67-5.84(m,5H),5.56-5.02(m,4H),4.81-4.53(m,5H),4.17(d,J=5.6Hz,1H),3.93-3.63(m,4H),3.60-3.04(m,14H),2.94-2.52(m,3H),2.40-1.82(m,7H),1.81-1.40(m,19H),1.25(ddd,J=23.7,20.7,10.9Hz,5H),1.14-0.65(m,18H)。
I-104:ESI-MS(EI + ,m/z):1072.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.63-5.90(m,5H),5.73-5.03(m,4H),4.80-4.54(m,5H),4.31-3.66(m,5H),3.59-3.04(m,14H),2.93-1.96(m,10H),1.94-1.59(m,12H),1.54-1.19(m,11H),1.15-0.63(m,19H)。
Example 29: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E, 19E), 23S,26R,27R,34 aS) -21- ((1, 4-dioxan-2-yl) methoxy) -9,27-dihydroxy-10-methoxy-6, 8,12, 14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1,4,5,6,9,10,11,12,13,14,21,22,23,24,25, 26,27,28,29,31,32,33,34 a-icosatetrahydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Aza-compounds Cyclothirty-one-pentadecen-3-yl) propyl) -2-methoxycyclohexyl (2-morpholinoethyl) carbamate (I-107), [(42S,44R,46R)-4-[(2R)-2-[(28E,30E,32E,33E,38R,39S,40R,41R,43S,45S,47S,48S, 50R,51R, 61R) -47- (1, 4-dioxan-2-ylmethoxy) -50, 61-dihydroxy-51-methoxy-38, 39,40,41, 52, 53-hexamethyl-54, 55,56,57, 58-pentaoxo-76, 77-dioxa-64-azatricyclohexa-hexadecane-28, 30,32 (52) 33 (53) -tetraen-48-yl]Propyl group]-46-methoxy-44-cyclohexyl]N- (2-morpholinoethyl) carbamic acid ester (I-103) and [ (42S, 44R, 46R) -4- [ (2R) -2- [ (28E, 30E,32E,33E,38R,39S,40R,41R,43S,45S, 47R,48S,50R,51R, 61R) -47- (1, 4-dioxan-2-ylmethoxy) -50, 61-dihydroxy-51-methoxy-38, 39, 40,41,52,53-hexamethyl-54, 55,56,57, 58-pentoxy-76, 77-dioxa-64-azatricyclotricyclo-hexadecane-28, 30,32 (52), 33 (53) -fourAlkene-48-yl]Propyl group]-46-methoxy-44-cyclohexyl]N- (2-morpholinoethyl) carbamide Synthesis of acid ester (I-102)
Step 1: (1R, 2R, 4S) -4- ((R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,21S,23S,26R,27R,34 aS) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-icosamhydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclotridecyl-pentadec-3-yl) propyl) -2-methoxycyclohexyl (2-morpholinoethyl) carbamate. To a solution of rapamycin (1 g,1.09 mmol) and pyridine (0.35 mL,4.38 mmol) in DCM (15 mL) was added dropwise by syringe under argon at 0deg.C DCM (0.325 g,1.09 mmol). The reaction mixture was stirred at 0deg.C for 1 hour, then TEA (1.22 mL,8.75 mmol) and 2-morpholinoethylamine (2.85 mL,21.88 mmol) were added to the mixture, and the resulting solution was stirred at 0deg.C for 1 hour, then diluted with DCM, and taken up in NH 4 Aqueous Cl solution, water, brine, and Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (8% meoh/DCM) to give (1R, 2R,4 s) -4- ((R) -2- ((3 s,6R,7e,9R,10R,12R,14s,15e,17e,19e,21s,23s,26R,27R,34 as) -9,27-dihydroxy-10, 21-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-eicoshydro-3H-23, 27-epoxypyrido [2,1-c ] as a pale yellow solid ][1]Oxa [4 ]]Azacyclotridecyl pentadec-3-yl) propyl) -2-methoxycyclohexyl (2-morpholinoethyl) carbamate (0.25 g,21% yield). ESI-MS (EI) + ,m/z):1070.4[M+H] +
Step 2: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -21- ((1, 4-dioxan-2-yl) methoxy) -9,27-dihydroxy-10-methoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-1, 4,5, 6)9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-icosatetrahydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclotridecyl pentadec-3-yl) propyl) -2-methoxycyclohexyl (2-morpholinoethyl) carbamate (I-107). at-50deg.C, to [ (39S, 41R, 43R) -4- [ (2R) -2- [ (26E, 28E,30E,31E,35R,36S,37R,38R,40S,42S,44S,45S,46R,47R, 57R) -46, 57-dihydroxy-44, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-50,51,52,53,54-pentaoxo-70, 71-dioxa-60-azatricyclohexadecane-26, 28,30 (48), 31 (49) -tetraen-45-yl]Propyl group]-43-methoxy-41-cyclohexyl]To a solution of N- (2-morpholinoethyl) carbamate (0.4 g,0.37 mmol) in DCM (6 mL) was added TFA (1.15 mL,14.95 mmol). The mixture was stirred for 10 min, then 1, 4-dioxan-2-yl methanol (1.32 g,11.21 mmol) dissolved in DCM (10 mL) was added and the mixture was stirred at-10 ℃ for 5 h. The reaction was taken up with DCM and NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-107 (63 mg,15% yield) as a white solid. ESI-MS (EI) + ,m/z):1179.6[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.41-5.86(m,4H),5.61-4.99(m,4H),4.43(dt,J=105.0,47.9Hz,3H),3.88-3.52(m,11H),3.46-3.01(m,14H),2.82-2.18(m,10H),2.15-1.59(m,22H),1.53-0.65(m,29H)。
Step 3: [ (42S, 44R, 46R) -4- [ (2R) -2- [ (28E, 30E,32E,33E,38R,39S,40R,41R,43S,45S,47S,48S,50R,51R, 61R) -47- (1, 4-dioxan-2-ylmethoxy) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-54, 55,56,57, 58-pentaoxo-76, 77-dioxa-64-azatricyclohexa-hexadecane-28, 30,32 (52), 33 (53) -tetraen-48-yl ] propyl ] -46-methoxy-44-cyclohexyl ] N- (2-morpholinoethyl) carbamate (I-103) and [ (42S, 44R, 46R) -4- [ (2R) -2- [ (28E, 30E,32E,33E,38R,39S,40R,41R,43S,45S,47R,48S,50R,51R, 61R) -47- (1, 4-dioxan-2-ylmethoxy) -50, 61-dihydroxy-51-methoxy-38,39,40,41,52,53-hexamethyl-54, 55,56,57, 58-pentoxy-76, 77-dioxa-64-azatricyclohexadecane-28, 30,32 (52), 33 (53) -tetraen-48-yl ] propyl ] -46-methoxy-44-cyclohexyl ] N- (2-morpholinoethyl) carbamate (I-102). 124mg of I-107 was isolated by chiral preparative HPLC to give I-103 as a white solid (23.7 mg,19% yield) and I-102 as a white solid (21.3 mg,17% yield).
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-NA 012)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 20 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 0.8mL/min
Wavelength: UV 254nm
Temperature: room temperature
HPLC apparatus: shimadzu LC-20AD
I-103:ESI-MS(EI + ,m/z):1156.8[M+H] + ,1178.8[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.35-5.75(m,4H),5.52-4.93(m,4H),4.50(s,1H),4.14(dd,J=25.0,13.4Hz,1H),3.85-3.44(m,14H),3.41-2.93(m,15H),2.83-1.77(m,17H),1.76-1.11(m,17H),1.08-0.62(m,24H)。
I-102:ESI-MS(EI + ,m/z):1156.8[M+H] + ,1178.8[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.58-5.85(m,4H),5.57-5.08(m,4H),4.63-3.92(m,3H),3.88-3.04(m,24H),2.80-1.94(m,14H),1.87-1.44(m,28H),1.13-0.60(m,20H)。
Example 30: (27E, 29E,31E,32E,39R,40S,41R,44R,46S,48S,51S,53R,54R, 63R) a- 51- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl]Propoxy group]-3-methoxy-ring Hexyl group]-1-methyl-ethyl]-53, 63-dihydroxy-54-methoxy-39,40,41,44,55,56-hexamethyl-50- [2- (oxy) Azetidin-3-yloxy) ethoxy]-75, 76-dioxa-64-azatricyclohexa-27, 29,31 (55), 32 (56) Tetraene-57, 58,59,60, 61-pentanone (I-109), (27E, 29E,31E,32E,39R,40S,41R,44R,46S, 48S,50S,51S,53R,54R, 63R) -51- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholino ] 4-yl group]Propoxy group]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-53, 63-dihydroxy-54-methoxy-39, 40,41, 44,55,56-hexamethyl-50- [2- (oxetan-3-yloxy) ethoxy]-75, 76-dioxa-64-azatricyclic tricyclic derivatives Hexadecane-27, 29,31 (55), 32 (56) -tetraene-57, 58,59,60, 61-pentanone (I-101) and (27E, 29E,31E,32E, 39R,40S,41R,44R,46S,48S,50R,51S,53R,54R,63R)-51-[(1R)-2-[(1S,3R,4R)-4-[3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl ]Propoxy group]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-53, 63-dihydroxyl Group-54-methoxy-39,40,41,44,55,56-hexamethyl-50- [2- (oxetan-3-yloxy) ethoxy]-75, 76-dioxa-64-azatricyclohexa-27, 29,31 (55), 32 (56) -tetraene-57, 58,59,60, 61-pentanone (I) 100 ) synthesis of
Step 1: (26E, 28E,30E,31E,36R,37S,38R,41R,43S,45S,47S,48S,49R,50R, 59R) -48- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl]Propoxy group]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-49,59-dihydroxy-47, 50-dimethoxy-36,37,38,41,51,52-hexamethyl-70, 71-dioxa-60-azatricyclohexa-hexadecane-26, 28,30 (51), 31 (52) -tetraene-53,54,55,56,57-pentanone. A solution of intermediate II (0.5 g,0.46 mmol) and N-ethyl-N-isopropyl-propan-2-amine (179.14 mg,1.39mmol,0.24 mL) in DCM (10 mL) was stirred at 25℃for 20 h. The reaction mixture was diluted with DCM (30 mL) and saturated NH 4 Cl (30 mL. Times.3), water (30 mL. Times.3) and brine (30 mL. Times.3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (with 50% ch 3 CN/water elution) to give (26 e,28e,30e,31e,36R,37s,38R,41R,43s,45s,47s,48s,49R,50R, 59R) -48- [ (1R) -2- [ (1 s,3R, 4R) -4- [3- [ (2 s, 6R) -2, 6-dimethylmorpholin-4-yl as a white solid ]Propoxy group]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-49,59-dihydroxy-47, 50-dimethoxy-36,37,38,41,51,52-hexamethyl-70, 71-dioxa-60-azatricyclotricetyl-26,28,30 (51), 31 (52) -tetraene-53,54,55,56,57-pentanone (0.2 g,40.5% yield). ESI-MS (EI) + ,m/z):1069.1[M+H] + T=1.918 min,98% purity, 254nm.
Step 2: (27E, 29E,31E,32E,39R,40S,41R,44R,46S,48S,51S,53R,54R, 63R) -51- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl]Propoxy group]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-53, 63-dihydroxy-54-methoxy-39,40,41,44,55,56-hexamethyl-50- [2- (oxetan-3-yloxy) ethoxy ]]-75, 76-dioxa-64-azatricyclohexa-27, 29,31 (55), 32 (56) -tetraene-57, 58,59,60, 61-pentanone (I-109). At-45 ℃ under N 2 Next, the mixture was purified to (26E, 28E,30E,31E,36R,37S,38R,41R,43S,45S,47S,48S,49R,50R, 59R) -48- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl]Propoxy group]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-49,59-dihydroxy-47, 50-dimethoxy-36,37,38,41,51,52-hexamethyl-70, 71-dioxa-60-azatricyclo-hexa-hexadecane-26, 28,30 (51), 31 (52) -tetraene-53,54,55,56,57-pentanone (1.12 g,1.05 mmol) in DCM (50 mL) 2, 2-trifluoroacetic acid (3.58 g,31.42mmol,2.42 mL) was added dropwise and the reaction stirred for 10 min. 2- (oxetan-3-yloxy) ethanol (2.47 g,20.95mmol in DCM) was added and the mixture stirred at-45℃for 2 h. Pouring the reactant into saturated NaHCO at 0 DEG C 3 (5 mL) and extracted with DCM (10 mL). The organic layer was washed with water (10 mL) and brine (10 mL), dried over anhydrous sodium sulfate, filtered and the filtrate concentrated. The residue was passed through a reverse phase column (with 40% CH 3 CN/water elution) (0.001% HCOOH) to afford I-109 as a white solid (0.074 g,6% yield). ESI-MS (EI) + ,m/z):1155.8[M+H] + ,T=1.849min,254nm。 1 H NMR(400MHz,CDCl 3 )δ6.40-5.94(m,4H),5.55-5.13(m,5H),4.79-4.54(m,5H),4.45-4.03(m,4H),3.89-3.58(m,4H),3.15-3.54(m,15H),3.14-2.91(m,5H),2.86-2.39(m,3H),2.35-1.85(m,11H),1.85-1.40(m,30H),1.40-1.12(m,19H),1.09-0.85(m,21H),0.76-0.52(m,2H)。
Step 3: (27E, 29E,31E,32E,39R,40S,41R,44R,46S,48S,50S,51S,53R,54R, 63R) -51- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl ] propoxy ] -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -53, 63-dihydroxy-54-methoxy-39,40,41,44,55,56-hexamethyl-50- [2- (oxetan-3-yloxy) ethoxy ] -75, 76-dioxa-64-azatricyclo-hexadecane-27, 29,31 (55), 32 (56) -tetraen-57, 58,59,60, 61-pentanone (I-101) and (27E, 29E,31E,32E,39R,40S,41R,44R,46S,48S,50R,51S,53R,54R, 63R) -51- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ (2S, 6R) -2, 6-dimethylmorpholin-4-yl ] propoxy ] -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -53, 63-dihydroxy-54-methoxy-39,40,41,44,55,56-hexamethyl-50- [2- (oxetan-3-yloxy) ethoxy ] -75, 76-dioxa-64-azatricyclohexadecane-27, 29,31 (55), 32 (56) -tetraen-57, 58,59,60, 61-pentanone (I-100). 94mg of I-109 were isolated by chiral preparative HPLC to give (27E, 29E,31E,32E,39R,40S,41R,44R,46S,48S,50S,51S,53R,54R, 63R) -51- [ (1R) -2- [ (1S, 3R, 4R) -4- [3I-101 (14 mg,15% yield) and I-100 (5 mg,5% yield) as white solids.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-NA 012)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 20 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 0.8mL/min
Wavelength: UV 254nm
Temperature: room temperature
HPLC apparatus: shimadzu LC-20AD
I-101:ESI-MS(EI + ,m/z):1155.8[M+H] +1 H NMR(500MHz,CDCl 3 )δ6.36-5.74(m,4H),5.51-4.97(m,4H),4.71-4.42(m,5H),4.10(d,J=5.7Hz,1H),3.87-3.06(m,19H),3.03-2.87(m,2H),2.84-2.46(m,4H),2.39-1.62(m,18H),1.56-1.32(m,12H),1.11-0.89(m,13H),0.88-0.58(m,20H)。
I-100:ESI-MS(EI + ,m/z):1155.8[M+H] +1 H NMR(500MHz,CDCl 3 )δ6.55-5.80(m,4H),5.55-5.03(m,4H),4.85-4.45(m,5H),4.11(dd,J=100.9,30.7Hz,3H),3.88-3.15(m,19H),3.10-2.09(m,14H),2.01-1.74(m,18H),1.54-1.14(m,15H),1.09-0.63(m,20H)。
Example 31: [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R, 41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexa Methyl-51,52,53,54,55-pentaoxo-71, 72-dioxa-60-azatricyclo-tricetyl-27, 29,31 (49), 32 (50) -tetraen-46-yl]Propyl group]-44-methoxy-42-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamic acid ester Synthesis of (I-99)
Step 1: tert-butyl N- (2-morpholinoethyl) carbamate. To a solution of 2-morpholinoethylamine (10 g,76.81 mmol) in DCM (5 mL) at 0deg.C was added triethylamine (5.35 mL,38.41 mmol) and tert-butyl tert-butoxycarbonyl carbonate (18.44 g,84.49 mmol), and the resulting solution was stirred overnight at 25deg.C. The reaction was diluted with 200mL of dichloromethane and then washed with 30mL of 10% sodium bicarbonate and 30mL of brine. The organic layer was dried over sodium sulfate, filtered and concentrated to give tert-butyl N- (2-morpholinoethyl) carbamate (17 g,96% yield) as an off-white solid. ESI-MS (EI) + ,m/z):231.3[M+H] +1 H NMR(400MHz,CDCl 3 )δ3.78-3.62(m,4H),3.24(d,J=5.5Hz,2H),2.45(dd,J=8.0,3.9Hz,6H),1.49-1.42(m,9H)。
Step 2: tert-butyl N-methyl-N- (2-morpholinoethyl) carbamate. Tert-butyl N- (2-morpholinoethyl) carbamate (18 g,78.16 mmol) was dissolved in DMF (240 mL) cooled to 0deg.C and NaH (9.38 g,234.47mmol,60% purity) was added. The reaction was stirred at room temperature for 20 minutes, then cooled to 0 ℃ and methyl iodide (12.2 g,85.97 mmol) was added. The reaction mixture was stirred for 3 hours, then diluted with ethyl acetate (500 mL) and washed sequentially with saturated aqueous ammonium chloride (300 mL) and brine (300 ml×5). The organic layer was dried over sodium sulfate and then concentrated to give tert-butyl N-methyl-N- (2-morpholinoethyl) carbamate (14 g, 7)3% yield). ESI-MS (EI+, m/z): 245.3[ M+H ]] +1 H NMR(400MHz,CDCl 3 )δ3.74-3.64(m,4H),3.34(s,2H),2.93-2.81(m,3H),2.48(d,J=4.8Hz,6H),1.46(s,10H)。
Step 3: n-methyl-2-morpholino-ethylamine. To tert-butyl N-methyl-N- (2-morpholinoethyl) carbamate (14 g,57.30 mmol) was added hydrochloric acid (4M, 143.25 mL) at 0deg.C. The reaction was stirred at room temperature for 50 min, concentrated and NH was added 3 (7M, 81.86 mL). After stirring for 1 hour, the reaction was concentrated and purified by silica gel chromatography (DCM: meOH: tea=90:10:0.1) to give N-methyl-2-morpholino-ethylamine (7.4 g,90% yield) as a yellow solid. ESI-MS (EI+, m/z): 145.1[ M+H ] ] +1 H NMR(400MHz,DMSO-d 6 )δ9.03(s,2H),3.80(s,4H),3.26(dd,J=44.9,20.4Hz,8H),2.63(s,3H)。
Step 4: [ (43S, 45R, 47R) -4- [ (2R) -2- [ (30E, 32E,34E,35E,39R,40S,41R,42R,44S,46S,48S,49S,50R,51R, 61R) -61-hydroxy-48, 51-dimethoxy-39,40,41,42,52,53-hexamethyl-54, 55,56,57, 58-pentaoxo-50-trimethylsilyloxy-73, 74-dioxa-63-azatricyclohexadecane-30, 32,34 (52), 35 (53) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate. Triphosgene (150.43 mg,0.507mmol in 20mL THF) was added dropwise to a solution of rapamycin (0.5 g,0.507 mmol) and pyridine (2.03 mmol,0.164 mL) in DCM (5 mL) under argon at 0deg.C. The reaction mixture was stirred at 0deg.C for 1 hour, then TEA (410 mg,4.06 mmol) and N-methyl-2-morpholino-ethylamine (1.46 g,10.14 mmol) were added, and the resulting solution was stirred at 0deg.C for 1 hour more. The reaction was diluted with DCM and NH 4 Aqueous Cl, water, brine, then Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (8% meoh/DCM) to give [ (43 s,45R, 47R) -4- [ (2R) -2- [ (30 e,32e,34e,35e,39R,40s,41R,42R,44s,46s,48s,49s,50R,51R, 61R) -61-hydroxy-48, 51-dimethoxy-39,40,41,42,52,53-hexamethyl-54, 55,56,57, 58-pentoxy-50-trimethylsilyloxy-73, 74-dioxa-63-azatricyclo-tricyclo-hexadeca-30, 32,34 (52), 35 (53) -tetraen-49-yl as a pale yellow solid ]Propyl group]-47-methoxy-45-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate (3836 mg,66% yield). ESI-MS (EI) + ,m/z):1156.4[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.57-5.93(m,4H),5.73-5.47(m,1H),5.27-4.98(m,2H),4.72(s,1H),4.56(s,1H),4.36-3.54(m,12H),3.54-3.05(m,12H),2.93(s,4H),2.40(dt,J=34.4,23.8Hz,11H),2.04(s,5H),1.88-1.52(m,12H),1.52-1.17(m,10H),1.20-0.73(m,17H),0.10--0.14(m,9H)。
Step 5: [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-51,52,53,54,55-pentaoxo-71, 72-dioxa-60-azatricyclohexa-hexadecane-27, 29,31 (49), 32 (50) -tetraen-46-yl]Propyl group]-44-methoxy-42-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate. To [ (43S, 45R, 47R) -4- [ (2R) -2- [ (30E, 32E,34E,35E,39R,40S,41R,42R,44S,46S,48S,49S,50R,51R, 61R) -61-hydroxy-48, 51-dimethoxy-39,40,41,42,52,53-hexamethyl-54, 55,56,57, 58-pentoxy-50-trimethylsilyloxy-73, 74-dioxa-63-azatricyclo-hexadecan-30, 32,34 (52), 35 (53) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]To a solution of N-methyl-N- (2-morpholinoethyl) carbamate (1.8 g,1.56 mmol) in acetone (5 mL) and water (5 mL) was added 0.5N sulfuric acid (0.5M, 4.67 mL). The reaction was stirred at 0deg.C for 2 hours, then poured into 100mL EtOAc and 100mL saturated NaHCO 3 In a mixture of solutions. The organic layer was washed with water and brine, dried over Na 2 SO 4 Dried, filtered and concentrated. The crude material was purified by silica gel chromatography (5% MeOH/DCM) to give [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-51,52,53,54,55-pentaoxo-71, 72-dioxa-60-azatricyclotricetyl-27, 29,31 (49), 32 (50) -tetraen-46-yl as a pale yellow solid]Propyl group]-44-methoxy-42-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate (1.4 g,83% yield). 1 HNMR(400MHz,CDCl 3 )δ6.47-5.84(m,4H),5.60-5.05(m,4H),4.77(s,1H),4.55(s,1H),4.34-4.10(m,1H),3.92-3.52(m,7H),3.52-3.23(m,10H),3.13(d,J=2.7Hz,4H),2.92(s,3H),2.78-2.39(m,8H),2.40-2.00(m,5H),2.03-1.53(m,18H),1.53-1.11(m,12H),1.11-0.87(m,13H),0.83(d,J=6.5Hz,2H)。
Step 6: [ (43S, 45R, 47R) -4- [ (2R) -2- [ (29E, 31E,33E,34E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 62R) -48- (1, 4-dioxan-2-ylmethoxy) -51,62-dihydroxy-52-methoxy-39,40,41,42,53,54-hexamethyl-55,56,57,58,59-pentoxy-77, 78-dioxa-64-azatricyclohexadecane-29, 31,33 (53), 34 (54) -tetraen-49-yl]Propyl group]-47-methoxy-45-cyclohexyl]N-methyl-N- (2-morpholinoethyl) carbamate (I-99). At-40 ℃ under N 2 Next, the reaction mixture was purified to [ (40S, 42R, 44R) -4- [ (2R) -2- [ (27E, 29E,31E,32E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-51,52,53,54,55-pentaoxo-71, 72-dioxa-60-azatricyclotricyclohexadecane-27, 29,31 (49), 32 (50) -tetraen-46-yl ]Propyl group]-44-methoxy-42-cyclohexyl]To a solution of N-methyl-N- (2-morpholinoethyl) carbamate (0.4 g,0.37 mmol) in DCM (15 mL) was added trifluoroacetic acid (1.14 mL,14.76 mmol). 1, 4-dioxan-2-yl methanol (0.87 g,7.38 mmol) was then added and the mixture was stirred at-40℃for 2 hours. The reaction mixture was then poured into DCM and ice-cold NaHCO 3 In a mixture of aqueous solutions, and the organic layer was subjected to Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography (68% CH 3 CN/water) to give I-99 (70 mg,16% yield) as a white solid. ESI-MS (EI) + ,m/z):1170.8[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.52-5.80(m,4H),5.61-5.04(m,4H),4.33(dt,J=75.6,73.9Hz,5H),3.93-3.03(m,26H),3.00-1.89(m,18H),1.88-1.58(m,6H),1.52-1.18(m,11H),1.14-0.69(m,19H)。
Example 32: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) 1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-18, 19-dimethyl Oxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy]-11, 36-dioxygen 4,9 Hetero-4-azatricyclo [30 ].3.1.0]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-97) Synthesis
220mg of I-11 were isolated by chiral preparative HPLC and then purified by silica gel chromatography to give I-97 (71.5 mg,33% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 80 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-97:ESI-MS(EI + ,m/z):1036.6[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.48-5.82(m,4H),5.72-5.04(m,4H),4.81-4.50(m,5H),3.97-3.09(m,20H),3.00-2.48(m,5H),2.36-1.86(m,7H),1.83-1.55(m,14H),1.52-1.20(m,9H),1.17-0.61(m,19H)。
Example 33: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) a- 46- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-45- [2- [2- (dimethylamino) Ethoxy group]Ethoxy group]-57-hydroxy-47, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-66, 67-dioxa- Synthesis of 58-azatricyclohexa-hexa-25, 27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-95)
/>
Step 1: (23E, 2)5E,27E,28E,34R,35S,36R,37R,39S,41S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-55-hydroxy-43- [2- (2-iodoethoxy) ethoxy ]]-45, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-63, 64-dioxa-56-azatricyclo-hexa-hexadecane-23, 25,27 (47), 28 (48) -tetraen-49,50,51,52,53-pentanone. At-40℃under nitrogen, to (24E, 26E,28E,29E,31R,32S,33R,34R,36S,38S,40S,41S,42R,43R, 52R) -41- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-52-hydroxy-40, 42, 43-trimethoxy-31,32,33,34,44,45-hexamethyl-60, 61-dioxa-53-azatricyclohexa-hexadecane-24, 26,28 (44), 29 (45) -tetraene-46,47,48,49,50-pentanone (0.5 g,0.53 mmol) to a solution of TFA (1.82 g,15.92mmol,1.23 mL) in DCM (10 mL). 2- (2-iodoethoxy) ethanol (2.29 g,10.61 mmol) was then added and the mixture stirred at-20℃for 3 hours. The reaction was purified by addition of saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with DCM (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=1:1) to give (23 e,25e,27e,28e,34R,35s,36R,37R,39s,41s,44s,45R,46R, 55R) -44- [ (1R) -2- [ (1 s,3R, 4R) -3, 4-dimethoxycyclohexyl as a white solid]-1-methyl-ethyl]-55-hydroxy-43- [2- (2-iodoethoxy) ethoxy ]]-45, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-63, 64-dioxa-56-azatricyclo-hexa-hexadecane-23, 25,27 (47), 28 (48) -tetraen-49,50,51,52,53-pentanone (0.2 g,33% yield). ESI-MS (EI) + ,m/z):1148.4[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.46-5.97(m,4H),5.71-5.03(m,4H),4.19-4.04(m,1H),3.93-3.53(m,7H),3.50-3.38(m,8H),3.37-3.21(m,7H),3.20-2.97(m,6H),2.96-2.50(m,4H),2.40-2.19(m,4H),2.18-1.85(m,6H),1.82-1.55(m,13H),1.53-1.21(m,10H),1.20-0.81(m,13H),0.79-0.69(m,1H)。
Step 2: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,46S,47R,48R, 57R) -46- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-45- [2- [2- (dimethylamino) ethoxy ]]Ethoxy group]-57-hydroxy-47, 48-dimethoxy-36,37,38,39,49,50-hexamethyl-66, 67-dioxa-58-azatricyclotricyclo-hexadecane-25,27,29 (49), 30 (50) -tetraene-51,52,53,54,55-pentanone (I-95). (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl ]-55-hydroxy-43- [2- (2-iodoethoxy) ethoxy ]]A solution of-45, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-63, 64-dioxa-56-azatricyclo-hexa-hexadecane-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone (0.3 g,0.27 mmol), N-methyl methylamine (120 mg,2.66 mmol) and N-ethyl-N-isopropyl-propan-2-amine (344 mg,2.66 mmol) in DCM (5 mL) was stirred at 30℃for 18 h. The reaction mixture was diluted with DCM (10 mL) and saturated NH 4 Cl (10 mL), water (10 mL) and brine (10 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (with 50% ch 3 CN/water elution) to give I-95 (70 mg,25% yield) as a white solid. ESI-MS (EI) + ,m/z):1044.7[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.81-6.70(m,1H),6.39-5.87(m,4H),5.58-5.39(m,2H),4.50-3.91(m,4H),3.87-3.49(m,7H),3.48-3.35(m,7H),3.34-3.20(m,5H),3.19-2.97(m,6H),2.90-2.76(m,6H),2.69-1.97(m,17H),1.88-1.40(m,11H),1.37-0.92(m,21H),0.91-0.77(m,3H)。
Example 34: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,46S,48R,49R, 58R) A- 58-hydroxy-48, 49-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl Base group]-1-methyl-ethyl]-36,37,38,39,50,51-hexamethyl-45- [2- (oxetan-3-yloxy) ethoxy]- 67, 68-dioxa-59-azatricyclohexa-25, 27,29 (50), 30 (51) -tetraen-52,53,54,55,56-pentanone (I-94), (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1-hydroxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]1-methyl-) Ethyl group]-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxygen 4,9 Hetero-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-83) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1-hydroxy-18, 19-dimethyl Oxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]- 15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy]-11, 36-dioxa-4-nitrogen 4,9 Heterotricyclo [30.3.1.0]Synthesis of tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-82)
Step 1: (25E, 27E,29E,30E,36R,37S,38R,39R,41S,43S,46S,48R,49R, 58R) -58-hydroxy-48, 49-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-36,37,38,39,50,51-hexamethyl-45- [2- (oxetan-3-yloxy) ethoxy]-67, 68-dioxa-59-azatricyclohexa-25, 27,29 (50), 30 (51) -tetraene-52,53,54,55,56-pentanone (I-94). at-55deg.C, at N 2 Next, (24E, 26E,28E,29E,33R,34S,35R,36R,38S,40S,42S,43S,44R,45R, 54R) -54-hydroxy-42, 44, 45-trimethoxy-43- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl ]-1-methyl-ethyl]-33,34,35,36,46,47-hexamethyl-62, 63-dioxa-55-azatricyclohexa-hexadecane-24, 26,28 (46), 29 (47) -tetraen-48,49,50,51,52-pentanone (intermediate VI,0.62g,0.63 mmol) 2, 2-trifluoroacetic acid (1.43 g,12.57mmol,0.97 mL) was added dropwise to a solution of DCM (30 mL). The reaction was stirred at-45 ℃ for 10 min, 2- (oxetan-3-yloxy) ethanol (1.49 g,12.57mmol in DCM) was added and the mixture was stirred at-45 ℃ for 2 h. The reaction mixture was poured into saturated NaHCO at 0deg.C 3 Aqueous solution (40 mL) and extracted with DCM (40 mL). The organic layer was washed with water (40 mL) and brine (40 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by chromatography on silica gel (EA: pe=9:1) and then by reverse phase chromatography (with 40% ch 3 CN/water elution) to give I-94 (0.074 g,11% yield) as a white solid. ESI-MS (EI) + ,m/z):1094.8[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.45-5.98(m,4H),5.66-4.97(m,4H),4.43-4.78(m,5H),4.31-4.18(m,1H),3.91-3.69(m,4H),3.67-3.24(m,17H),3.21-2.99(m,5H),2.86-2.50(m,3H),2.30-1.84(m,6H),1.78-1.59(m,20H),1.51-1.23(m,10H),1.20-1.03(m,11H),0.97-0.84(m,8H),0.78-0.69(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1-hydroxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-83) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1-hydroxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-82). 130mg of I-94 were isolated by chiral preparative HPLC and then purified by silica gel chromatography to give I-83 (50.8 mg,39% yield) as a white solid and I-82 (6.2 mg,5% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 10 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu-LC-20AD
I-83:ESI-MS(EI + ,m/z):1094.7[M+H] +1 H NMR(500MHz,CDCl 3 )δ6.46-5.82(m,4H),5.71-5.02(m,4H),4.81-4.49(m,5H),4.00-3.20(m,24H),3.19-2.98(m,5H),2.95-2.43(m,3H),2.36-1.84(m,7H),1.80-1.56(m,13H),1.52-1.22(m,9H),1.19-0.68(m,19H)。
I-82:ESI-MS(EI + ,m/z):1094.6[M+H] +1 H NMR(500MHz,CDCl 3 )δ6.46-5.84(m,4H),5.78-5.02(m,4H),4.74-4.42(m,5H),3.91-2.79(m,29H),2.72-1.56(m,24H),1.46-0.59(m,27H)。
Example 35: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) a- 44- (1, 4-dioxan-2-ylmethoxy) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy Phenyl) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-48-methoxy-35,36,37,38,49,50-hexamethyl-69, 70-di Oxa-58-azatricyclohexadecane-24, 26,28 (49), 29 (50) -tetraene-51,52,53,54,55-pentanone (I-106), (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44S,45S,47R,48R, 57R) -44- (1, 4-dioxane- 2-ylmethoxy) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclo Hexyl group]-1-methyl-ethyl]-48-methoxy-35,36,37,38,49,50-hexamethyl-69, 70-dioxa-58-azatris Cyclotriacontane-24, 26,28 (49), 29 (50) -tetraene-51,52,53,54,55-pentanone (I-93) and (24E, 26E,28E, 29E,35R,36S,37R,38R,40S,42S,44R,45S,47R,48R, 57R) -44- (1, 4-dioxan-2-ylmethoxy) propan-one 47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]1-methyl-) Ethyl group]-48-methoxy-35,36,37,38,49,50-hexamethyl-69, 70-dioxa-58-azatricyclotricyclohexadecane-24, 26,28 (49), 29 (50) -tetraene-51,52,53,54,55-pentanone (I-92)
Step 1:3- (2-benzyloxy-ethoxy) oxetane. To a solution of oxetan-3-ol (10 g,135 mmol) in DMF (160 mL) was added sodium hydride (3.24 g,135 mmol) at 0deg.C, the resulting solution was stirred at that temperature for 30 min, and then 2-bromoethoxymethylbenzene (43.55 g,202.49 mmol) was added. The reaction was stirred in an ice water bath at 0 ℃ for 2 hours, then warmed to room temperature and stirred for 16 hours. The reaction was treated with 1200mL NH 4 Cl (saturated aqueous) was quenched, extracted with ethyl acetate (2X 120 mL) and the organic layers were combined and concentrated. The residue was purified by silica gel chromatography with PE/EA (8:1) to give 3- (2-benzyloxy-ethoxy) oxetane (16.4 g,58% yield) as a colourless liquid. ESI-MS (EI) + ,m/z):231[M+Na] +1 H NMR(400MHz,CDCl 3 )δ7.41-7.23(m,6H),4.79-4.70(m,2H),4.68-4.52(m,6H),3.62-3.53(m,4H)。
Step 2:2- (oxetan-3-yloxy) ethanol. At N 2 To a solution of 3- (2-benzyloxyethoxy) oxetane (4 g,19.21 mmol) in MeOH (20 mL) was added palladium (10% on carbon) (2.04 g,19.21 mmol). The solution was taken up in H 2 Stirred overnight at 40 ℃, then filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=1:5) to give 2- (oxetan-3-yloxy) ethanol (2.1 g,93% yield) as a colorless liquid. 1 HNMR(400MHz,CDCl 3 )δ4.79(td,J=5.8,2.1Hz,2H),4.62(dt,J=10.2,4.9Hz,3H),3.80-3.69(m,2H),3.52-3.44(m,2H),2.36(s,1H)。
Step 3:1, 4-dioxan-2-yl methanol. A mixture of 2- (oxetan-3-yloxy) ethanol (4 g,33.86 mmol) and HND-8 (1.2 g) in THF (60 mL) was stirred at 50℃for 3 hours. The mixture was filtered and concentrated to give 1, 4-dioxan-2-yl methanol (3.66 g,92% yield) as a colorless oil. 1 HNMR(400MHz,CDCl 3 )δ3.86-3.42(m,9H),1.98(s,1H)。
Step 4:2- [ tert-butyl (diphenyl) silyl group]Oxyethyl triflate. At 0℃under N 2 Next, a 2- [ tert-butyl (diphenyl) silyl group is introduced ]Oxyethanol (7 g,23.3 mmol) and DIEA (4.52 g,34.95 mmol) inTo a solution of DCM (20 mL) was added trifluoromethylsulfonyl trifluoromethanesulfonate (7.23 g,25.63 mmol). The mixture was stirred at 0deg.C for 2 hours, then diluted with DCM (150 mL) and saturated NaHCO 3 (50 mL), water (50 mL), brine (50 mL), washed with Na 2 SO 4 Drying, filtering and concentrating to obtain 2- [ tert-butyl (diphenyl) silyl ] as brown oil]Oxyethyl triflate. This material was used without further purification. 1 H NMR(400MHz,CDCl 3 )δ7.68-7.65(m,4H),7.47-7.38(m,6H),4.59-4.53(m,2H),3.94-3.86(m,2H),1.07(d,J=5.4Hz,9H)。
Step 5: (35E, 37E,39E,40E,46R,47S,48R,49R,51S,53S,55S,56S,57R,58R, 67R) -56- [ (1R) -2- [ (1S, 3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57, 67-dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclo-hexa-hexadecane-35, 37,39 (59), 40 (60) -tetraene-61, 62,63,64, 65-pentanone. Rapamycin (2 g,2.19 mmol), 2- [ tert-butyl (diphenyl) silyl]A mixture of oxyethyltriflate (9.46 g,21.88 mmol) and N-ethyl-N-isopropyl-propan-2-amine (3.39 g,26.25 mmol) in toluene (20 mL) was stirred at 58℃for 18 hours and then poured into cold saturated NaHCO 3 (150 mL). It was extracted with EtOAc (200 mL) and the organic layer was washed with water (150 mL. Times.3) and brine (150 mL) over anhydrous Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=3:1) to give (35 e,37e,39e,40e,46R,47s,48R,49R,51s,53s,55s,56s,57R,58R, 67R) -56- [ (1R) -2- [ (1 s,3R, 4R) -4- [2- [ tert-butyl (diphenyl) silyl ] as a yellow solid]Oxyethoxy radical]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-57, 67-dihydroxy-55, 58-dimethoxy-46,47,48,49,59,60-hexamethyl-77, 78-dioxa-69-azatricyclo-hexa-hexadecane-35, 37,39 (59), 40 (60) -tetraene-61, 62,63,64, 65-pentanone. ESI-MS (EI) + ,m/z):1218.6[M+Na] +
Step 6: (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41,44-dimethoxy-32,33,34,35,45,46-hexamethyl-63, 64-dioxa-54-azatricyclohexa-hexadecane-22, 24,26 (45), 27 (46) -tetraene-47,48,49,50,51-pentanone. At 0℃to (35E, 37E,39E,40E,47R,48S,49R,50R,52S,54S,56S,57S,58R,59R, 68R) -57- [ (1R) -2- [ (1S, 3R, 4R) -4- [3- [ tert-butyl (diphenyl) silyl]Oxypropoxy radical ]-3-methoxy-cyclohexyl]-1-methyl-ethyl]-58, 68-dihydroxy-56,59-dimethoxy-47,48,49,50,60,61-hexamethyl-78, 79-dioxa-70-azatricyclohexa-hexadecane-35, 37,39 (60), 40 (61) -tetraene-62,63,64,65,66-pentanone (3.46 g,2.86 mmol) to a solution of THF (70 mL) was added pyridine HF (2.26 g,28.58 mmol). The mixture was stirred at 30 ℃ for 3 hours, then cooled to 0 ℃ and saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with EA (30 mL). The organic layer was washed with water (100 mL) and brine (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (PE: ea=2:3) to give (22 e,24e,26e,27e,32R,33s,34R,35R,37s,39s,41s,42s,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1 s,3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl as a pale yellow solid]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-63, 64-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraen-47,48,49,50,51-pentanone. ESI-MS (EI) + ,m/z):994.5[M+Na] +
Step 7: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -44- (1, 4-dioxan-2-ylmethoxy) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]-48-methoxy-35,36,37,38,49,50-hexamethyl-69, 70-dioxa-58-azatricyclohexa-hexadecane-24, 26,28 (49), 29 (50) -tetraene-51,52,53,54,55-pentanone (I-106). At-40 ℃ under N 2 Next, the reaction mixture was purified to give (22E, 24E,26E,27E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-41, 44-dimethoxy-32,33,34,35,45,46-hexamethyl-63, 64-dioxa-54-azatricyclo-hexa-hexadecane-22, 24,26 (45), 27 (46) -tetraen-47,48,49,50,51-pentanone (0.6 g,0.62 mmol) and 1, 4-dioxan-2-yl methanol (2.19 g,18.51 mmol) in DCM (40 mL)2, 2-trifluoroacetic acid (2.81 g,24.69 mmol) was added. The reaction mixture was stirred at-10℃for 2 hours, then ice-cooled with saturated NaHCO 3 (100 mL), water (100 mL. Times.3) and brine (100 mL. Times.3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase column chromatography (with 65% CH 3 CN/water elution) to give I-106.ESI-MS (EI) + ,m/z):1080.4[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.44-5.89(m,4H),5.61-5.37(m,2H),5.31-5.12(m,2H),4.79(d,J=18.7Hz,1H),4.29(d,J=12.8Hz,1H),3.95-3.53(m,11H),3.53-3.27(m,9H),3.27-2.96(m,5H),2.71(s,1H),2.58(d,J=13.9Hz,1H),2.34(d,J=11.8Hz,2H),2.08(s,3H),1.87-1.57(m,20H),1.47(dd,J=22.8,10.6Hz,3H),1.26-0.77(m,19H),0.70(dd,J=23.6,12.0Hz,1H)。
Step 8: (24E, 26E, 29E,35R,36S,37R,38R,40S,42S,44S,45S,47R,48R, 57R) -44- (1, 4-dioxan-2-ylmethoxy) -47, 57-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4- (3-hydroxypropoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -48-methoxy-35,36,37,38,49,50-hexamethyl-69, 70-dioxa-58-azatricyclohexadecane-24, 26,28 (49), 29 (50) -tetraen-51,52,53,54,55-pentanone (I-93) and (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,44R,45S,47R, 48R) -44- (1, 4-dioxan-2-ylmethoxy) -47, 57-dihydroxy-45R) -2- [ (1R) -2-oxan-5-yloxy ] -48-oxa-one (29R), 29 (50) -tetraen-51,52,53,54,55-pentanone (I-93) and (24E, 26E, 29E,35R,36S,37R,38R,40S,42S, 47R. 240mg of I-106 was isolated by chiral preparative HPLC and then purified by silica gel chromatography (9% MeOH in DCM mixture: PE: EA=3:3:1) to give I-93 (45 mg,19% yield) as a white solid and I-92 (25 mg,10% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 100 μl of
Mobile phase: hexane/etoh=60/40 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-93:ESI-MS(EI + ,m/z):1080.8[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.37-5.74(m,4H),5.51-5.00(m,4H),4.10(d,J=6.0Hz,1H),3.86-3.44(m,14H),3.40-2.90(m,15H),2.82-2.45(m,3H),2.34-1.34(m,24H),1.30-1.10(m,7H),1.05-0.53(m,19H)。
I-92:ESI-MS(EI + ,m/z):1080.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.46-5.88(m,4H),5.70-5.06(m,4H),4.23(t,J=32.2Hz,2H),4.01-3.52(m,12H),3.52-2.88(m,16H),2.78-1.98(m,9H),1.92-1.72(m,8H),1.51-1.20(m,17H),1.14-0.60(m,19H)。
Example 36: (27E, 29E,31E,32E,39R,40S,41R,42R,44S,46S,49S,51R,52R, 62R) the 51,62-dihydroxy-52-methoxy-49- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (2-oxa-6-azaspiro) [3.3]Heptane-6-yl) propoxy]Cyclohexyl group]-1-methyl-ethyl]-39,40,41,42,53,54-hexamethyl-48- [2- (oxy) Azetidin-3-yloxy) ethoxy]-74, 75-dioxa-63-azatricyclohexa-27, 29,31 (53), 32 (54) Synthesis of-tetraene-55,56,57,58,59-pentanone (I-91)
Step 1: (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -45, 48-dihydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (2-oxa-6-azaspiro [ 3.3)]Heptane-6-yl) propoxy]Cyclohexyl group]-1-methyl-ethyl]-36,37,38,39,49,50-hexamethyl-69, 70-dioxa-59-azatricyclohexa-hexadecane-26, 28,30 (49), 31 (50) -tetraen-51,52,53,54,55-pentanone. Intermediate II (2.9 g,2.68 mmol), 2-oxa-6-azaspiro [3.3 ] ]Heptane (0.797 g,8.04mmol,0.123 ml) and N-ethyl-N-isopropyl-propan-2-amine (1.04 g,8.04mmol, 1)40 mL) in DCM (50 mL) was stirred at 22℃for 20 h. The reaction mixture was diluted with DCM (10 mL) and saturated NH 4 Cl (10 mL. Times.3), water (10 mL. Times.3) and brine (10 mL. Times.3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (with 50% ch 3 CN/water elution) to give (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (2-oxa-6-azaspiro [3.3 ] as a white solid]Heptane-6-yl) propoxy]Cyclohexyl group]-1-methyl-ethyl]-36,37,38,39,49,50-hexamethyl-69, 70-dioxa-59-azatricyclohexa-hexadecane-26, 28,30 (49), 31 (50) -tetraen-51,52,53,54,55-pentanone (1.5 g,53% yield). ESI-MS (EI+, m/z): 1053.8[ M+H ]]T=1.882min, 100% purity, 254nm.
Step 2: (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -45, 48-dihydroxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (2-oxa-6-azaspiro [ 3.3)]Heptane-6-yl) propoxy]Cyclohexyl group]-1-methyl-ethyl ]-36,37,38,39,49,50-hexamethyl-69, 70-dioxa-59-azatricyclohexa-hexadecane-26, 28,30 (49), 31 (50) -tetraene-51,52,53,54,55-pentanone (I-91). at-55deg.C, at N 2 Next, the reaction mixture was purified to give (26E, 28E,30E,31E,36R,37S,38R,39R,41S,43S,45S,46S,47R,48R, 58R) -47, 58-dihydroxy-45, 48-dimethoxy-46- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- [3- (2-oxa-6-azaspiro [ 3.3)]Heptane-6-yl) propoxy]Cyclohexyl group]-1-methyl-ethyl]-36,37,38,39,49,50-hexamethyl-69, 70-dioxa-59-azatricyclohexa-hexadecane-26, 28,30 (49), 31 (50) -tetraene-51,52,53,54,55-pentanone (0.5 g,0.47 mmol) 2, 2-trifluoroacetic acid (1.89 g,16.61mmol,1.28 mL) was added dropwise to a solution of DCM (25 mL). The reaction was stirred at-45 ℃ for 10 min, then 2- (oxetan-3-yloxy) ethanol (1.12 g,9.49mmol in DCM) was added and the reaction stirred at-20 ℃ for 2 h. The reaction mixture was poured into saturated NaHCO at 0deg.C 3 (40 mL) and extracted with DCM (40 mL). The organic layer was washed with water (40 mL) and brine (40 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (with 40% ch 3 CN/water elution) (0.01% hcooh) purification,i-91 (0.06 g,11% yield) was obtained as a white solid. ESI-MS (EI) + ,m/z):1139.8[M+H] + T=1.814 min,98% purity, 254nm. 1 H NMR(400MHz,CDCl 3 )δ6.40-5.95(m,4H),5.54-5.12(m,4H),4.85(br,4H),4.79-4.54(m,5H),4.45-4.03(m,6H),3.94-3.64(m,4H),3.57-3.19(m,15H),3.13-2.95(m,6H),2.77-2.13(m,6H),2.03-1.56(m,27H),1.53-1.37(m,5H),1.38-081(m,33H),0.69-0.61(m,3H)。
Example 37: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) a- 45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxy cyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy Base-35,36,37,38,48,49-hexamethyl-44- [2- [2- (methylamino) ethoxy ]]Ethoxy group]65, 66-dioxa- Synthesis of 58-azatricyclohexa-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-90)
Step 1: (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -55-hydroxy-43- [2- (2-iodoethoxy) ethoxy ] -45, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-63, 64-dioxa-56-azatricyclo-tricetyl-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone. To a solution of intermediate I (2.9 g,2.68 mmol) in DCM (10 mL) was added TFA (1.82 g,15.92mmol,1.23 mL) under nitrogen at-40 ℃. 2- (2-iodoethoxy) ethanol (2.29 g,10.61 mmol) was then added and the mixture stirred at-20℃for 3 hours. The reaction mixture was purified by silica gel chromatography (PE: ea=1:1) to give (23 e,25e,27e,28e,34R,35s,36R,37R,39s,41s,44s,45R,46R, 55R) -44- [ (1R) -2- [ (1 s,3R, 4R) -3, 4-dimethoxycyclohexyl ] -1-methyl-ethyl ] -55-hydroxy-43- [2- (2-iodoethoxy) ethoxy ] -45, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-63, 64-dioxa-56-azatricyclohexa-hexadecane-23, 25,27 (47), 28 (48) -tetraen-49,50,51,52,53-pentanone (0.3 g,50% yield) as a white solid.
Step 2: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,46R,47R, 56R) -45- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-56-hydroxy-46, 47-dimethoxy-35,36,37,38,48,49-hexamethyl-44- [2- [2- (methylamino) ethoxy ]]Ethoxy group]-65, 66-dioxa-58-azatricyclohexa-24, 26,28 (48), 29 (49) -tetraene-50,51,52,53,54-pentanone (I-90). (23E, 25E,27E,28E,34R,35S,36R,37R,39S,41S,44S,45R,46R, 55R) -44- [ (1R) -2- [ (1S, 3R, 4R) -3, 4-dimethoxycyclohexyl]-1-methyl-ethyl]-55-hydroxy-43- [2- (2-iodoethoxy) ethoxy ]]A solution of-45, 46-dimethoxy-34,35,36,37,47,48-hexamethyl-63, 64-dioxa-56-azatricyclo-hexa-hexadecane-23, 25,27 (47), 28 (48) -tetraene-49,50,51,52,53-pentanone (0.38 g,0.34 mmol), methylamine (0.105 g,3.37mmol,0.117 mL) and N-ethyl-N-isopropyl-propan-2-amine (0.436 g,3.37mmol,0.588 mL) in DCM (8 mL) was stirred at 22℃for 24 h. The reaction was diluted with DCM (10 mL) and saturated NH 4 Cl (10 mL. Times.3), water (10 mL. Times.3) and brine (10 mL. Times.3), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography (with 50% ch 3 CN/water elution) to give I-90 (55 mg,16% yield) as a white solid.
Example 38: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S,44R,45R, 54R) -44, 54-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl Base group]-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ]]-65, 66-dioxa-55-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraen-48,49,50,51,52-pentanone (I) 88 (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1, 18-dihydroxyl- 12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-a reaction of 19-methoxy-15, 17, 21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy]-11, 36-dioxa-4-azatricyclic 4,9 [30.3.1.0]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-7)2) And (1R, 9S,12S, 15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexanediyl-methyl ester 4,9 Radical-30- [2- (oxetan-3-yloxy) ethoxy]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Thirty-one pieces of Synthesis of hexa-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-71)
Step 1: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S,44R,45R, 54R) -44, 54-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ]-1-methyl-ethyl]-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ]]-65, 66-dioxa-55-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraene-48,49,50,51,52-pentanone (I-88). To a solution of rapamycin (0.5 g,0.547 mmol) in DCM (10 mL) under nitrogen was added TFA (1.87 g,16.41 mmol) followed by 2- (oxetan-3-yloxy) ethanol (1.29 g,10.94 mmol) and the mixture stirred at-20℃for 2 h. The reaction mixture was purified by addition of saturated NaHCO 3 The aqueous solution (20 mL) was quenched and extracted with DCM (30 mL) at 0deg.C. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase column chromatography (with 80% CH 3 CN/water elution) to give I-88 (120 mg,22% yield) as a white solid. ESI-MS (EI) + ,m/z):1022.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.42-5.88(m,4H),5.58-5.08(m,4H),4.83-4.54(m,5H),4.35-3.93(m,2H),3.91-3.68(m,3H),3.53-3.21(m,13H),2.99-2.41(m,5H),2.38-1.87(m,7H),1.85-1.58(m,13H),1.55-1.17(m,11H),1.16-0.82(m,17H),0.80-0.63(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R)4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-72) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-71). 200mg of I-88 were isolated by chiral preparative HPLC and then purified by silica gel chromatography to give I-72 (31 mg,16% yield) as a white solid and I-71 (18 mg,9% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC-3 (IC 30CE-NJ 008)
Column dimensions: 0.46cm inside diameter x 25cm length
And (3) sample injection: 10 mu L
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-72:ESI-MS(EI + ,m/z):1022.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.37-5.77(m,4H),5.51-4.99(m,4H),4.77-4.45(m,5H),4.12(dd,J=13.5,6.1Hz,1H),3.84-3.58(m,3H),3.54-2.99(m,15H),2.93-2.48(m,5H),2.34-1.63(m,14H),1.49-1.10(m,14H),1.08-0.71(m,19H),0.59(dt,J=16.8,8.4Hz,1H)。
I-71:ESI-MS(EI + ,m/z):1022.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.44-5.86(m,4H),5.66-4.99(m,4H),4.77-4.46(m,5H),4.25-3.59(m,5H),3.55-3.02(m,15H),2.96-1.83(m,13H),1.81-1.59(m,10H),1.46-1.10(m,10H),1.08-0.48(m,19H)。
Example 39: (1R, 9S,12S, 1)5R,16E,18R,19R,21R,23S,24E,26E,28E,32S,35R)-12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphinyloxy-3-methoxy-cyclohexyl ]]-1-methyl-ethyl]-1, 18-di Hydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy]- 11, 36-dioxa-4-azatricyclo [30.3.1.04,9 ] ]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentane Synthesis of Ketone (I-89)
At 0deg.C, to (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S,44R,45R, 54R) -44, 54-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ]]To a solution of 65, 66-dioxa-55-azatricyclohexa-hexa-ne-23, 25,27 (46), 28 (47) -tetraene-48,49,50,51,52-pentanone (I-88) (0.290 g,0.290 mmol) in DCM (7 mL) was added 2, 6-di-tert-butyl-4-methylpyridine (0.447 g,2.17 mmol) followed by a solution of dimethylphosphonyl chloride (0.16 g,1.45 mmol) in DCM (2 mL). The resulting solution was stirred at 0deg.C for 7 hours, then diluted with DCM, and saturated NaHCO 3 (30 mL), 0.5N aqueous HCl, water (30 mL), brine (50 mL) and then the organic layer was washed with Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by silica gel chromatography (MeOH/PE: EA: DCM (3:3:10) =0% to 15%) followed by reverse phase chromatography (60% ch) 3 CN/water) to give I-89 (0.13 g,42% yield) as a white solid.
Example 40: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -27-hydroxy 1- ((1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxycyclohexyl) propan-2-yl) -9, 10-dimethoxy Phenyl-6,8,12,14,20,26-hexamethyl-21- (2- (oxetan-3-yloxy) ethoxy) -9,10,12,13,14, 21,22,23,24,25,26,27,32,33,34 a-hexadecyl-3H-2327-epoxy pyrido [2,1-c ]][1]Oxa [4 ]] Synthesis of Azacyclo-Triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (I-87)
Step 1: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -27-hydroxy-3- ((R) -1- ((1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxycyclohexyl) propan-2-yl) -9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-21- (2- (oxetan-3-yloxy) ethoxy) -9,10,12,13,14,21,22,23,24,25,26,27,32,33,34 a-hexadeca-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-triundecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (I-87). To a solution of intermediate V (0.66 g,0.679 mmol) in DCM (10 mL) was added TFA (3.1 g,27.15mmol,2.09 mL) at-50deg.C. After 10 min, a solution of 2- (oxetan-3-yloxy) ethanol (2.41 g,20.37 mmol) in DCM (0.5 mL) was added and the mixture stirred at 0deg.C for 5 h. Addition of DCM and NaHCO 3 Aqueous solution, and the organic layer was washed with water and brine, dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-87 (129.4 mg,18% yield) as a white solid. ESI-MS (EI) + ,m/z):1080.6[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.46-5.93(m,4H),5.65-5.01(m,4H),4.82-4.15(m,4H),3.92-3.54(m,9H),3.51-3.07(m,16H),2.95-2.48(m,3H),2.37-1.83(m,6H),1.82-1.46(m,19H),1.44-1.21(m,7H),1.17-0.81(m,18H),0.74(d,J=11.9Hz,1H)。
Example 41: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphinyloxy-3-methoxy-cyclohexyl ]]-1-methyl-ethyl]-1, 18-di Hydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11,36- 4,9 Dioxa-4-azatricyclo [30.3.1.0 ]]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I) 86 ) synthesis of
Step 1: (25E, 27E,29E,30E,34R,35S,36R,37R,40S,42S,45S,46R,47R, 56R) -46, 56-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-47-methoxy-34,35,36,37,48,49-hexamethyl-44- (2-tetrahydropyran-4-yloxyethoxy) -67, 68-dioxa-57-azatricyclo-hexa-hexadecane-25, 27,29 (48), 30 (49) -tetraen-50,51,52,53,54-pentanone. At 0℃under N 2 To a solution of rapamycin (0.5 g,0.547 mmol) and 2-tetrahydropyran-4-yloxyethanol (2.4 g,16.41 mmol) in DCM (15 mL) was added 2, 2-trifluoroacetic acid (2.49 g,21.88 mmol). The reaction mixture was stirred at 0deg.C for 2 hours, then cooled with saturated NaHCO 3 The solution (10 mL), water (10 mL. Times.3) and brine (10 mL. Times.3) were washed, dried over anhydrous sodium sulfate, filtered and concentrated, and the residue was purified by reverse phase column chromatography (with 70% CH) 3 CN/water elution) to give (25E, 27E,29E,30E,34R,35S,36R,37R,40S,42S,45S,46R,47R, 56R) -46, 56-dihydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl) as a white solid]-1-methyl-ethyl]-47-methoxy-34,35,36,37,48,49-hexamethyl-44- (2-tetrahydropyran-4-yloxyethoxy) -67, 68-dioxa-57-azatricyclo-hexa-hexadecane-25, 27,29 (48), 30 (49) -tetraen-50,51,52,53,54-pentanone (175 mg,31% yield). ESI-MS (EI+, m/z): 1050.6[ M+Na ]] +
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1, 18-dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-86). At 0℃under N 2 Next, (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]To a solution of tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (0.25 g,0.24 mmol) in DCM (9 mL) was added 2, 6-di-tert-butyl-4-methylpyridine (0.37 g,1.82 mmol) and [ chloro (methyl) phosphoryl group]A solution of methane (0.137 g,1.22 mmol) in DCM (3 mL). The mixture was stirred at 0deg.C for 6 hours, then diluted with DCM (30 mL) and saturated NaHCO 3 (30 mL), 0.5N aqueous HCl, water (30 mL), brine (50 mL) and then the organic layer was washed with Na 2 SO 4 Dried, filtered and concentrated in vacuo, and the crude material was purified by silica gel chromatography (MeOH/PE: EA: DCM (3:3:10) =0% to 15%) to give I-86 (130 mg,48% yield) as a white solid. ESI-MS (EI) + ,m/z):1126.4[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.53-5.81(m,4H),5.6-5.12(m,4H),4.33-3.70(m,7H),3.65-2.90(m,16H),2.84-1.97(m,8H),1.95-1.17(m,36H),1.14-0.63(m,19H)。
Example 42: (24E, 26E,28E,29E,34R,35S,36R,37R,39S,41S,44S,46R,47R, 56R) a- 43- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-46, 56-dihydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxy) Hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-47-methoxy-34,35,36,37,48,49-hexamethyl-) 68, 69-dioxa-57-azatricyclohexa-24, 26,28 (48), 29 (49) -tetraen-50,51,52,53,54-pentanone (I-110)、(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S,35R)-30- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethyl) Oxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-11, 36- Dioxa-4-azatricyclo [30.3.1.04,9 ]]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I) 85 (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -30- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethyl)Oxy) -3-) Methoxy-cyclohexyl]-1-methyl-ethyl]19-methoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa- 4-azatricyclo [30.3.1.04,9 ]]Synthesis of hexacosane-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-84) Finished products
Step 1: (24E, 26E,28E,29E,34R,35S,36R,37R,39S,41S,44S,46R,47R, 56R) -43- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-46, 56-dihydroxy-44- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-47-methoxy-34,35,36,37,48,49-hexamethyl-68, 69-dioxa-57-azatricyclohexa-hexadecane-24, 26,28 (48), 29 (49) -tetraen-50,51,52,53,54-pentanone (I-110). at-55deg.C, at N 2 Next, to a solution of everolimus (1 g,1.04 mmol) in DCM (60 mL) was added dropwise 2, 2-trifluoroacetic acid (2.38 g,20.88mmol,1.61 mL). The reaction mixture was stirred at-45 ℃ for 10 minutes, then [ (2R) -1, 4-dioxan-2-yl was added ]A solution of methanol (0.493 g,4.17 mmol) in DCM was added and the reaction stirred at-20deg.C for 1 hr. The mixture was poured into saturated NaHCO at 0deg.C 3 Aqueous solution (80 mL) and extracted with DCM (80 mL). The organic layer was washed with water (80 mL) and brine (80 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography and then repurified by silica gel chromatography (100% ea) to give I-110 (65 mg,6% yield) as a white solid. ESI-MS (EI) + ,m/z):1066.4[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.39-5.92(m,4H),5.56-4.81(m,5H),4.26-3.98(m,2H),3.84-3.68(m,9H),3.62-3.53(m,3H),3.48-3.04(m,15H),2.87-2.55(m,4H),2.35-1.83(m,7H),1.79-1.38(m,27H),1.34-1.22(m,7H),1.18-0.79(m,19H),0.76-0.67(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -30- [ [ (2S) -1, 4-dioxan-2-yl ] methoxy ] -1, 18-dihydroxy-12- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ] -1-methyl-ethyl ] -19-methoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.04,9] triacontan-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-85) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -30- [ (2S) -1, 4-dioxan-2-yl ] methoxy ] -1, 18-dihydroxy-12- [ (2S) -12-dioxan-2-yl ] methoxy ] -1, 18-hydroxy-12- [ (2S) -12-oxa-1, 35R-oxa-1, 36-dioxan-4-azatricyclo [30.3.1.04,9] triacontan-8-e-pentanone (I-85) and (1R, 9R, 28R, 23S, 29S, 30R, 24-dioxan-2-yl ] methoxy ] -30- [ (2-3-2-yl ] methoxy ] -1. 110mg (I-110 was isolated by chiral preparative HPLC and then purified by silica gel chromatography to give I-85 as a white solid (32.2 mg,29% yield) and I-84 as a white solid (12 mg,11% yield).
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 20 mu L
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-85:ESI-MS(EI + ,m/z):1066.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.42-5.82(m,4H),5.59-5.07(m,4H),4.81(s,1H),4.17(d,J=6.1Hz,1H),3.89-3.53(m,15H),3.47-3.01(m,16H),2.90-2.52(m,3H),2.41-1.85(m,8H),1.82-1.42(m,8H),1.39-1.18(m,10H),1.15-0.61(m,19H)。
I-84:ESI-MS(EI + ,m/z):1066.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.42-5.86(m,4H),5.59-5.07(m,4H),4.30-3.95(m,3H),3.87-3.03(m,28H),2.97-1.71(m,21H),1.52-1.17(m,14H),1.13-0.64(m,19H)。
Example 43: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy 1-methoxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]- 1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Second step 4,9 Oxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14, 20-pentanone (I-81), (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1- Hydroxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl Base group]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ] 4,9 Base group]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2, 3, synthesis of 10,14,20-pentanone (I-69)
Step 1: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl ]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-81). at-55deg.C, at N 2 To a solution of intermediate VI (0.5 g,0.507 mmol) in DCM (20 mL) was added dropwise 2, 2-trifluoroacetic acid (1.16 g,10.14mmol,0.78 mL). The reaction was stirred at-45 ℃ for 10 minutes, then 2- [2- (oxetan-3-yloxy) ethoxy ] was added at the same temperature]A solution of ethanol (1.64 g,10.14 mmol) in DCM. The mixture was warmed to 0 ℃ over 2 hours. Pouring the reactant into saturated NaHCO at 0 DEG C 3 Aqueous solution (40 mL) and extracted with DCM (40 mL). The organic layer was washed with water (40 mL) and brine (40 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (100% ea) and by reverse phase chromatography (with 60% ch) 3 CN/water elution) and purifying again to obtain the product in the form ofI-81 as a white solid (0.03 g,5% yield). ESI-MS (EI) + ,m/z):1138.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.45-6.00(m,4H),5.54-5.08(m,4H),4.78-4.57(m,5H),4.45-4.30(m,1H),3.88-3.79(m,4H),3.70-3.50(m,9H),3.47-3.42(m,4H),3.38-3.30(m,5H),3.28-3.23(m,3H),3.22-3.03(m,5H),2.75-2.50(m,2H),2.31-1.84(m,6H),1.76-1.48(m,18H),1.53-1.21(m,10H),1.18-1.04(m,11H),0.98-0.83(m,8H),0.80-0.67(m,2H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1-hydroxy-18, 19-dimethoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl ]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-69). 100mg of I-81 were isolated by chiral preparative HPLC and then by silica gel chromatography to give I-69 (25 mg,25% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CE-BN 011)
Column dimensions: 0.46cm inside diameter x 25cm length
And (3) sample injection: 50 mu L
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-69:ESI-MS(EI + ,m/z):1138.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.42-5.73(m,4H),5.72-4.98(m,4H),4.72-4.47(m,5H),3.94-2.92(m,32H),2.90-2.39(m,3H),2.33-1.49(m,17H),1.47-1.13(m,12H),1.07-0.57(m,20H)。
Example 44: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy 1-2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl-1R]-1-methyl-ethyl]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Second step 4,9 Oxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14, 20-pentanone (I-80) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1- Hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]- 18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]] 4,9 Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2, 3,10, synthesis of 14, 20-pentanone (I-62)
Step 1: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-80). At-40 ℃ under N 2 Next, TFA (2.11 g,18.51mmol,1.43 mL) was added to a solution of intermediate V (0.6 g,0.62 mmol) in DCM (15 mL), followed by the addition of 2- [2- (oxetan-3-yloxy) ethoxy]Ethanol (2 g,12.34 mmol) and the mixture was stirred at-20 ℃ for 2 hours. Pouring the reactant into saturated NaHCO at 0 DEG C 3 (aqueous) solution (20 mL) and extracted with DCM (20 mL). The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-80 (50 mg,7% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1124.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.46-5.97(m,4H),5.73-5.03(m,4H),4.70-4.54(m,5H),4.50-4.12(m,2H),3.93-3.73(m,3H),3.72-3.50(m,8H),3.49-3.03(m,13H),2.98-2.51(m,4H),2.38-1.87(m,7H),1.83-1.55(m,17H),1.54-1.15(m,10H),1.14-0.81(m,17H),0.80-0.68(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-62). 129mg of I-80 were isolated by chiral preparative HPLC and then purified by silica gel chromatography to give I-62 (30.6 mg,24% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 10 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC equipment Shimadzu-LC-20AD
I-62:ESI-MS(EI + ,m/z):1124.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.51-5.80(m,4H),5.73-5.03(m,4H),4.86-4.53(m,5H),3.99-3.03(m,31H),2.99-2.50(m,4H),2.40-1.83(m,10H),1.82-1.44(m,9H),1.43-1.18(m,9H),1.18-0.67(m,19H)。
Example 45: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -30- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R)/(R) 3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 4,9 36-dioxa-4-azatricyclo [30.3.1.0 ]]Trihexadecane-16,24,26,28-tetraene-2,3,10,1 4, 20-pentanone (I-76)、(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S,35R)-30-[[(2S)- 1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy Phenyl-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxo 4,9 Hetero-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-66) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -30- [ [ (2S) -1, 4-bis Oxalan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-) Methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-nitrogen 4,9 Heterotricyclo [30.3.1.0]Synthesis of tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-65)
Step 1: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -30- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-76). at-55deg.C, at N 2 Next, to a solution of intermediate III (0.5 g,0.51 mmol) in DCM (40 mL) was added dropwise 2, 2-trifluoroacetic acid (1.17 g,10.29 mmol). The reaction was stirred for 10 minutes, then [ (2R) -1, 4-dioxan-2-yl was added at the same temperature ]Methanol (1.03 g,8.74mmol in DCM). The reaction was warmed to-10 ℃ over 2 hours and then poured into saturated NaHCO at 0 ℃ 3 Aqueous solution (40 mL) and extracted with DCM (50 mL). The organic layer was washed with water (50 mL) and brine (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatographySpectrometry (EA 100%) followed by reverse phase chromatography (with 65% CH 3 CN/water elution) to give I-76 (0.08 g,15% yield) as a white solid. ESI-MS (EI) + ,m/z):1080.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.39-5.92(m,4H),5.56-4.81(m,5H),4.28-3.98(m,3H),3.90-3.68(m,9H),3.65-3.28(m,16H),3.26-2.97(m,5H),2.88-2.46(m,4H),2.35-1.91(m,6H),1.89-1.60(m,18H),1.55-1.16(m,10H),1.14-0.83(m,18H),1.76-0.65(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -30- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-66) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -30- [ [ (2S) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-65). 155mg of I-76 were isolated by chiral preparative HPLC and purified by silica gel chromatography to give I-66 (33.2 mg,21.42% yield) as a white solid and I-65 (13.8 mg,9% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 10 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu-LC-20AD
I-66:ESI-MS(EI + ,m/z):1080.4[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.43-5.82(m,4H),5.60-5.07(m,4H),4.82(s,1H),4.17(d,J=5.7Hz,1H),3.88-3.00(m,31H),2.88-2.51(m,3H),2.40-1.68(m,13H),1.55-1.30(m,8H),1.29-1.15(m,7H),1.14-0.62(m,19H)。
I-65:ESI-MS(EI + ,m/z):1080.4[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.45-5.87(m,4H),5.71-5.10(m,4H),4.10(dd,J=85.6,30.3Hz,3H),3.86-2.83(m,30H),2.82-1.71(m,17H),1.54-1.14(m,14H),1.12-0.59(m,19H)。
Example 46: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) 12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphinyloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1,18- Dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy]- 4,9 11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentane Ketones (I-78) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphinyloxy-3-methoxy-cyclohexyl ]]-1-methyl-ethyl]-1, 18-di Hydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]- 4,9 11, 36-dioxa-4-azatricyclo [30.3.1.0 ]]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentane Synthesis of Ketone (I-77)
Step 1: (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S,44R,45R, 54R) -44, 54-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ]]-65, 66-dioxa-55-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraen-48,49,50,51,52-pentanone. at-40deg.CTo a solution of rapamycin (0.5 g,0.547 mmol) in DCM (10 mL) under nitrogen was added TFA (1.87 g,16.41mmol,1.26 mL). 2- (oxetan-3-yloxy) ethanol (1.29 g,10.94 mmol) was added and the mixture was stirred at-20℃for 2 hours. The reaction mixture was quenched by addition of saturated aqueous NaHCO3 (20 mL) and extracted with DCM (30 mL) at 0 ℃. The organic layer was washed with water (20 mL) and brine (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by reverse phase column chromatography (with 80% CH 3 CN/water elution) to give (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41R,42S,44R,45R, 54R) -44, 54-dihydroxy-42- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl as a white solid ]-1-methyl-ethyl]-45-methoxy-32,33,34,35,46,47-hexamethyl-41- [2- (oxetan-3-yloxy) ethoxy ]]-65, 66-dioxa-55-azatricyclohexa-23, 25,27 (46), 28 (47) -tetraen-48,49,50,51,52-pentanone (120 mg,22% yield). ESI-MS (EI) + ,m/z):1022.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.42-5.88(m,4H),5.58-5.08(m,4H),4.83-4.54(m,5H),4.35-3.93(m,2H),3.91-3.68(m,3H),3.53-3.21(m,13H),2.99-2.41(m,5H),2.38-1.87(m,7H),1.85-1.58(m,13H),1.55-1.17(m,11H),1.16-0.82(m,17H),0.80-0.63(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1, 18-dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone. At 0℃to (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]To a solution of tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (0.29 g,0.29 mmol) in DCM (7 mL) was added 2, 6-di-tert-butyl-4-methylpyridine (0.45 g,2.17 mmol). Dimethyl phosphonochloride (0.163 g, 1.45 mmol) in DCM (2 mL) and the reaction stirred at 0deg.C for 7h, then diluted with DCM, taken up in saturated NaHCO 3 (30 mL), 0.5N aqueous HCl, water (30 mL), and brine (50 mL). The organic layer was purified by Na 2 SO 4 Dried, filtered, concentrated in vacuo, and the residue was purified by silica gel chromatography (MeOH/PE: EA: DCM (3:3:10) =0% to 15%) and by reverse phase chromatography (60% ch) 3 CN/water) to give (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl as a white solid]-1-methyl-ethyl]-1, 18-dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (130 mg,42% yield). ESI-MS (EI) + ,m/z):1098.7[M+Na] +
Step 3: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1, 18-dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-78) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1, 18-dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-77). 150mg of (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1, 18-dihydroxy-19-methoxy-15,17,21,23,29,35-hexamethyl-30- [2- (oxetan-3-yloxy) ethoxy ]]-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]The tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone is separated by chiral preparative HPLC and then purified by silica gel chromatography to obtainTo I-78 as a white solid (28.5 mg,19% yield) and I-77 as a white solid (12.3 mg,8% yield).
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CE-QE 014)
Column dimensions: 0.46cm inside diameter x 25cm length
And (3) sample injection: 10 mu L
Mobile phase: hexane/etoh=40/60 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AD
I-78:ESI-MS(EI + ,m/z):1098.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.37-5.78(m,4H),5.52-5.02(m,4H),4.79-4.44(m,5H),4.24-3.94(m,2H),3.89-3.57(m,3H),3.55-2.88(m,15H),2.80-2.42(m,3H),2.36-1.78(m,8H),1.75-1.35(m,16H),1.32-1.10(m,11H),1.08-0.57(m,19H)。
I-77:ESI-MS(EI + ,m/z):1098.7[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.39-5.93(m,4H),5.65-5.01(m,4H),4.80-4.46(m,5H),4.26-3.91(m,4H),3.51-3.10(m,13H),3.04-1.91(m,11H),1.86-1.52(m,20H),1.49-1.11(m,10H),1.08-0.57(m,19H)。
Example 47: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -30- [ [ (2R) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R)/(R) 3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 4,9 36-dioxa-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-79)、(1R,9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S,35R)-30-[[(2R)- 1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy Phenyl-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxo 4,9 Hetero-4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-64) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -30- [ [ (2R) -1, 4-bis Oxalan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-) Methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-nitrogen 4,9 Heterotricyclo [30.3.1.0]Synthesis of tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-63)
Step 1: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -30- [ [ (2R) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-79). at-55deg.C, at N 2 Next, to a solution of intermediate III (0.5 g,0.51 mmol) in DCM (35 mL) was added dropwise 2, 2-trifluoroacetic acid (1.17 g,10.29mmol, -0.79 mL). The reaction was stirred at-45 ℃ for 10 minutes and [ (2S) -1, 4-dioxane-2-yl was added]Methanol (0.97 g,8.23mmol in DCM) and then the mixture was warmed to-10℃over 1 hour. Pouring the reactant into saturated NaHCO at 0 DEG C 3 Aqueous solution (40 mL) and extracted with DCM (40 mL). The organic layer was washed with water (40 mL) and brine (40 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (100% ea) and then by reverse phase chromatography (with 60% ch 3 CN/water elution) and purification gave I-79 (0.1 g,18% yield) as a white solid. ESI-MS (EI) + ,m/z):1080.6[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.39-5.92(m,4H),5.57-4.77(m,5H),4.31-3.98(m,3H),3.85-3.67(m,8H),3.65-3.24(m,17H),3.22-2.97(m,3H),2.75-2.26(m,5H),2.17-1.90(m,5H),1.86-1.58(m,17H),1.51-1.16(m,10H),1.15-0.81(m,18H),0.76-0.65(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -30- [ [ (2R) -1, 4-dioxan-2-yl ]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-64) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -30- [ [ (2R) -1, 4-dioxan-2-yl]Methoxy group]-1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.0 ] 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-63). 146mg of I-79 were isolated by chiral preparative HPLC and then purified by silica gel chromatography to give I-64 (31.2 mg,21% yield) as a white solid and I-63 (15.4 mg,11% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CE-BN 011)
Column dimensions: 0.46cm inside diameter x 25cm length
And (3) sample injection: 10 mu L
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu-LC-20AD
I-64:ESI-MS(EI + ,m/z):1080.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.43-5.81(m,4H),5.58-5.08(m,4H),4.77(s,1H),4.17(d,J=5.6Hz,1H),3.89-3.26(m,28H),3.22-2.99(m,4H),2.89-2.46(m,3H),2.38-1.67(m,13H),1.55-1.16(m,13H),1.13-0.59(m,20H)。
I-63:ESI-MS(EI + ,m/z):1080.3[M+Na] +1 H NMR(400MHz,CDCl 3 )δ6.47-5.93(m,4H),5.70-5.14(m,4H),4.34-3.94(m,3H),3.86-2.93(m,30H),2.87-1.87(m,9H),1.72(t,J=14.6Hz,8H),1.51-1.16(m,12H),1.13-0.59(m,21H)。
Example 48: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E, 19E), 23S,26R,27R,34 aS) -27-hydroxy-9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-21- (2- (oxa-cyclic ring) Butan-3-yloxy) ethoxy) -1,5,11,28,29-pentoxy-1,4,5,6,9,10,11,12,13,14,21,22,23, 24,25,26,27,28,29,31,32,33,34 a-icosanhydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa-type [4]Synthesis of Azacyclotridecyl-pentadec-3-yl) propyl) -2-methoxycyclohexyl dimethylphosphinate (I-75)
To a solution of intermediate IV (0.5 g,0.498 mmol) in DCM (15 mL) was added TFA (2.27 g,19.92mmol,1.53 mL) at-50 ℃. After 10 min, a solution of 2- (oxetan-3-yloxy) ethanol (1.76 g,14.94 mmol) in DCM (0.5 mL) was added and the mixture stirred at-10℃for 5 h. The reaction was taken up with DCM and NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-75 (180.7 mg,33% yield) as a white solid. ESI-MS (EI+, m/z): 1112.5[ M+Na ]] +1 H NMR(500MHz,CDCl 3 )δ6.47-5.79(m,4H),5.38(dddd,J=91.9,76.3,49.1,19.1Hz,4H),4.80-4.03(m,7H),3.94-2.94(m,22H),2.93-1.83(m,11H),1.67-1.30(m,22H),1.30-0.82(m,21H),0.77(dd,J=24.4,12.2Hz,1H)。
Example 49: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E, 19E), 23S,26R,27R,34 aS) -27-hydroxy-9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-21- (3-morpholinopropyl) Oxy) -1,5,11,28,29-pentoxy-1,4,5,6,9,10,11,12,13, 14,21,22,23,24,25,26,27,28, 29,31,32,33,34 a-icosanhydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclo-thirty-one Synthesis of C-pentadecen-3-yl) propyl) -2-methoxycyclohexyldimethylphosphinate (I-74)
To a solution of intermediate IV (0.35 g,0.349 mmol) in DCM (15 mL) was added TFA (1.59 g,13.94 mmol) at-50 ℃. The reaction was stirred for 10 min, then 3-morpholinopropan-1-ol (1.52 g,10.46 mmol) dissolved in DCM (0.5 mL) was added and the mixture was stirred at-10℃for 5 h. The mixture was washed with DCM and NaHCO 3 Dilute with water, wash with water and brine, over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-74 (138.8 mg,36% yield) as a white solid. ESI-MS (EI) + ,m/z):1118.7[M+H] +1 H NMR(400MHz,CDCl 3 )δ6.19(dd,J=78.7,69.0Hz,4H),5.32(d,J=60.0Hz,4H),4.11(s,2H),3.93-3.54(m,9H),3.47-2.93(m,18H),2.90-1.93(m,17H),1.32(dd,J=60.9,36.3Hz,17H),1.19-0.62(m,26H)。
Example 49: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl Base group]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa- 4,9 4-azatricyclo [30.3.1.0 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-68), (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1, 18-dihydroxy-19-methoxy 1-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15, 4 17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 ,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-57) and (1R, 9S,12S,15R,16E,18R, 19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-Hexanail 4,9 Phenyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0]Trihexadecane-16,24,26 was used as a starting material, 28-tetraene-2,3,10,14,20-pentanone (I-56)
Step 1: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-68). At 0℃under N 2 Next, the reaction mixture was purified to give (23E, 25E,27E,28E,32R,33S,34R,35R,37S,39S,41S,42S,43R,44R, 53R) -43, 53-dihydroxy-41, 44-dimethoxy-42- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl group ]-1-methyl-ethyl]To a solution of-32,33,34,35,45,46-hexamethyl-62, 63-dioxa-54-azatricyclohexa-hexadecane-23, 25,27 (45), 28 (46) -tetraen-47,48,49,50,51-pentanone (0.5 g,0.51 mmol) and pyrazin-2-ylmethanol (0.96 g,8.74 mmol) in THF (15 mL) was added 4-methylbenzenesulfonic acid hydrate (0.49 g,2.57mmol, 0.399 mL). The reaction was stirred at 40 ℃ for 22 hours and then poured into cold saturated NaHCO 3 Aqueous solution (30 mL) and extracted with DCM (10 mL). The organic layer was washed with water (30 mL) and brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was chromatographed on silica gel (100% ea) and then on reversed phase (70% ch) 3 CN/water elution) to give I-68 (0.08 g,15% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1072.5[M+Na] +1 H NMR(400MHz,CDCl 3 )δ8.73-8.70(m,1H),8.56-8.48(m,2H),6.42-5.98(m,4H),5.60-4.82(m,4H),4.62-4.15(m,4H),4.07-3.86(m,2H),3.75-3.48(m,6H),3.47-3.20(m,12H),3.16-2.95(m,4H),2.98-2.10(m,6H),2.05-1.54(m,23H),1.56-1.16(m,10H),1.15-0.82(m,19H),0.79-0.64(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-57) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1, 18-dihydroxy-19-methoxy-12- [ (1R) -2- [ (1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl ]-1-methyl-ethyl]-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4 ,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-56). 200mg of I-68 were isolated by chiral preparative HPLC and then purified by silica gel chromatography (11% MeOH/PE: DCM: EA 3:3:1) to give I-57 (24.4 mg,12% yield) as a white solid and I-56 (21.5 mg,10% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 10 μl of
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-57:ESI-MS(EI + ,m/z):1072.6[M+Na] +1 H NMR(500MHz,CDCl 3 )δ8.71(s,1H),8.49(d,J=2.4Hz,2H),6.41-5.90(m,4H),5.59-5.08(m,4H),4.91(s,1H),4.58(d,J=13.8Hz,1H),4.44-4.32(m,1H),4.18(t,J=16.3Hz,1H),3.94(dd,J=21.3,14.0Hz,2H),3.71(ddd,J=25.5,13.1,7.6Hz,3H),3.60-3.26(m,15H),3.22-2.95(m,3H),2.86-2.54(m,3H),2.37-2.16(m,2H),2.01(dd,J=31.2,14.8Hz,5H),1.70(dd,J=31.5,12.5Hz,9H),1.51-1.16(m,11H),1.14-0.79(m,18H),0.71(dd,J=23.8,12.1Hz,1H)。
I-56:ESI-MS(EI + ,m/z):1072.7[M+Na] +1 H NMR(500MHz,CDCl 3 )δ8.73(s,1H),8.51(d,J=2.5Hz,2H),6.43-5.78(m,4H),5.71-5.01(m,4H),4.65-4.16(m,4H),4.03-3.61(m,4H),3.56-2.90(m,18H),2.85-1.69(m,16H),1.41(ddd,J=79.6,43.9,14.6Hz,14H),1.14-0.59(m,20H)。
Example 50: (3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -9,27- Dihydroxy-10-methoxy-3- ((R) -1- ((1S, 3R, 4R) -3-methoxy-4- (2-methoxyethoxy) cyclohexyl) propan-2-) Yl) -6,8,12,14,20,26-hexamethyl-21- (2- (pyrazin-2-yl) ethoxy) -9,10,12,13,14,21,22,23, 24,25,26,27,32,33,34 a-hexadechydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxa [4 ]]Azacyclic three Synthesis of undecarpentadecen-1,5,11,28,29 (4H, 6H, 31H) -pentanone (I-60)
To a solution of intermediate III (0.5 g,0.51 mmol) in DCM (15 mL) was added TFA (2.35 g,20.57 mmol) at-50deg.C. The reaction was stirred for 10 min, then 2-pyrazin-2-ylethanol (1.92 g,15.43 mmol) dissolved in DCM (0.5 mL) was added and the mixture was stirred at-20 ℃ for 5 h. The mixture was washed with DCM and NaHCO 3 The aqueous solution was diluted, and then the organic layer was washed with water and brine, and dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-60 (162.8 mg,30% yield) as a white solid. ESI-MS (EI) + ,m/z):1086.6[M+Na] +1 H NMR(500MHz,CDCl 3 )δ8.65-8.29(m,1H),6.53-5.95(m,4H),5.56-5.14(m,4H),4.59-3.65(m,6H),3.62-2.43(m,25H),2.15(dt,J=144.5,40.3Hz,6H),1.56-1.16(m,16H),1.15-0.54(m,26H)。
Example 51: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl Base group]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-nitrogen 4,9 Heterotricyclo [30.3.1.0]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-67), (1R, 9S, 12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2 ] [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl radical]-1-methyl-ethyl]-a reaction of 19-methoxy-15, 17, 4,9 21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0]Three kinds of Hexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-59) and (1R, 9S,12S,15R,16E,18R,19R, 21R,23S,24E,26E,28E,30R,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxy) Ethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl ]19-methoxy-15,17,21,23,29,35-hexamethyl-30- 4,9 (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0]Trihexadecane-16,24,26,28-tetratetrahexadecane Alkene-2,3,10,14,20-pentanone (I-58)
Step 1: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-67). At 0℃under N 2 The lower part of the upper part is provided with a lower part,to a solution of everolimus (0.977 g,8.87 mmol) in THF (15 mL) was added 4-methylbenzenesulfonic acid hydrate (0.496 g,2.61 mmol). The reaction was stirred at 40 ℃ for 10 hours and then poured into cold saturated NaHCO 3 Aqueous (30 mL) and extracted with EA (30 mL). The organic layer was washed with water (30 mL) and brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated. The residue was purified by silica gel chromatography (DCM: meoh=1:5) followed by reverse phase chromatography (with 60% ch 3 CN/water elution) to give I-67 (0.11 g,20% yield) as a pale yellow solid. ESI-MS (EI) + ,m/z):1059.6[M+Na] +1 H NMR(400MHz,CDCl 3 )δ8.743-8.70(m,1H),8.56-8.48(m,2H),6.42-5.93(m,4H),5.60-4.89(m,5H),4.63-4.06(m,4H),4.017-3.53(m,7H),3.46-3.28(m,8H),3.25-2.91(m,4H),3.16-2.95(m,4H),2.88-2.42(m,4H),2.32-1.97(m,8H),1.96-1.61(m,23H),1.56-1.13(m,12H),1.11-0.82(m,17H),0.76-0.63(m,1H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl ]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-59) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -1, 18-dihydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-19-methoxy-15,17,21,23,29,35-hexamethyl-30- (pyrazin-2-ylmethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-58). 220mg of I-67 was isolated by chiral preparative HPLC and then purified by silica gel chromatography (11% MeOH/PE: DCM: EA 3:3:1) to give I-59 (40.2 mg,18% yield) as a white solid and I-58 (32.1 mg,15% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 10 mu L
Mobile phase: hexane/etoh=50/50 (V/V)
Flow rate: 1.0mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-59:ESI-MS(EI + ,m/z):1059.6[M+Na] +1 H NMR(500MHz,CDCl 3 )δ8.71(s,1H),8.49(d,J=2.0Hz,2H),6.45-5.91(m,4H),5.60-5.09(m,4H),4.90(s,1H),4.58(dd,J=13.6,4.1Hz,1H),4.46-4.33(m,1H),4.19(dd,J=20.7,6.5Hz,1H),4.02-3.51(m,8H),3.48-3.02(m,12H),2.88-2.54(m,3H),2.36-1.88(m,7H),1.85-1.63(m,11H),1.52-1.17(m,10H),1.14-0.79(m,18H),0.75-0.64(m,1H)。
I-58:ESI-MS(EI + ,m/z):1059.0[M+Na] +1 H NMR(500MHz,CDCl 3 )δ8.79(s,1H),8.51(d,J=2.3Hz,2H),6.43-5.96(m,4H),5.74-5.08(m,4H),4.90(s,1H),4.65-4.18(m,4H),4.01-3.54(m,6H),3.50-2.82(m,14H),2.76-1.69(m,14H),1.56-1.19(m,16H),1.16-0.60(m,19H)。
Example 52: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E, 19E), 23S,26R,27R,34 aS) -27-hydroxy-9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29- Pentoxy-21- (2- ((tetrahydro-2H-pyran-4-yl) oxy) ethoxy) -1,4,5,6,9,10,11,12,13,14,21,22, 23,24,25,26,27,28,29,31,32,33,34 a-icosatetrahydro-3H-23, 27-epoxypyrido [2,1-c ]][1]Oxygen gas Impurity [4 ]]Azacyclo-triundec-pentadec-3-yl) propyl) -2-methoxycyclohexyl dimethyl phosphinate (I-70), (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S,35R)-12-[(1R)-2-[(1S,3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]1-hydroxy-18, 19-dimethoxy- 15,17,21,23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11, 36-dioxa-4-azatris (aza) can be used as a protective agent 4,9 Ring [30.3.1.0 ]]Trihexadecane-16,24,26,28-tetraene-2,3,10,14,20-pentaneThe reaction of ketone (I-55) and (1R, 9S,12S, 15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-di Methyl phosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1-hydroxy-18, 19-dimethoxy-15,17,21, 23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11, 36-dioxa-4-azatricyclo 4,9 [30.3.1.0]Synthesis of tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-54)
Step 1: (1R, 2R, 4S) -4- ((2R) -2- ((3S, 6R,7E,9R,10R,12R,14S,15E,17E,19E,23S,26R,27R,34 aS) -27-hydroxy-9, 10-dimethoxy-6,8,12,14,20,26-hexamethyl-1,5,11,28,29-pentoxy-21- (2- ((tetrahydro-2H-pyran-4-yl) oxy) ethoxy) -1,4,5,6,9,10,11,12,13,14,21,22,23,24,25,26,27,28,29,31,32,33,34 a-twenty-four-hydro-3H-23, 27-epoxypyrido [2,1-c ] ][1]Oxa [4 ]]Azacyclo-triundec-pentadec-3-yl) propyl) -2-methoxycyclohexyl dimethyl phosphinate (I-70). To a solution of intermediate IV (0.5 g,0.498 mmol) in DCM (15 mL) was added TFA (2.27 g,19.92mmol,1.53 mL) at-50 ℃. The reaction was stirred for 10 min, then 2-tetrahydropyran-4-yloxyethanol (2.18 g,14.94 mmol) dissolved in DCM (0.5 mL) was added. The mixture was stirred at-10℃for 24 hours, then with DCM and NaHCO 3 Diluting the aqueous solution. The organic layer was washed with water and brine, dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give I-70 (89.8 mg,16% yield) as a white solid. ESI-MS (EI) + ,m/z):1140.5[M+Na] +1 HNMR(500MHz,CDCl 3 )δ6.61-5.89(m,4H),5.77-5.06(m,4H),4.66-4.01(m,2H),4.01-3.47(m,6H),3.47-2.93(m,15H),2.92-2.33(m,3H),2.33-1.84(m,7H),1.71-1.34(m,29H),1.33-0.66(m,26H)。
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30S,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphineAcyloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1-hydroxy-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (I-55) and (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,30R,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl ]-1-methyl-ethyl]-1-hydroxy-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- (2-tetrahydropyran-4-yloxyethoxy) -11, 36-dioxa-4-azatricyclo [30.3.1.0 4,9 ]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-54). 100mg of I-70 were isolated by chiral preparative HPLC and then purified by silica gel chromatography (8% MeOH/PE: DCM: EA 3:3:1) to give I-55 (26.5 mg,25% yield) as a white solid and I-54 (11.4 mg,11% yield) as a white solid.
Chiral analysis method
Chromatographic column: CHIRALPAKIC (IC 00CD-TB 016)
Column dimensions: 0.46cm inside diameter by 15cm length
And (3) sample injection: 50 μl
Mobile phase: etoh=100%
Flow rate: 0.5mL/min
Wavelength: UV 254nm
Temperature: 35 DEG C
HPLC apparatus: shimadzu LC-20AT
I-55:ESI-MS(EI + ,m/z):1140.6[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.50-5.76(m,4H),5.58-4.96(m,4H),4.19-3.49(m,9H),3.46-2.86(m,17H),2.81-2.46(m,2H),2.37-1.68(m,18H),1.60-1.17(m,22H),1.13-0.70(m,20H)。
I-54:ESI-MS(EI + ,m/z):1140.4[M+Na] +1 H NMR(500MHz,CDCl 3 )δ6.36-5.75(m,4H),5.46-4.97(m,4H),4.65(s,1H),4.11(d,J=5.7Hz,1H),3.90-3.58(m,5H),3.53-3.17(m,18H),2.89-2.38(m,6H),2.31-1.64(m,20H),1.55-1.20(m,17H),1.07-0.56(m,20H)。
Example 53: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S,35R)-12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphinyloxy-3-methoxy-cyclohexyl ]]-1-methyl-ethyl]1-hydroxy-) 18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]] Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.04,9 ]]Tricetyl-16,24,26,28-tetraene-2, 3,10, synthesis of 14, 20-pentanone (I-73)
Step 1: (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -57-hydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl ]-1-methyl-ethyl]-47, 48-dimethoxy-35,36,37,38,49,50-hexamethyl-44- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-67, 68-dioxa-58-azatricyclohexa-24, 26,28 (49), 29 (50) -tetraen-51,52,53,54,55-pentanone. at-55deg.C, at N 2 To a solution of intermediate I (0.5 g, 0.39 mmol) in DCM (5 mL) was added dropwise 2, 2-trifluoroacetic acid (1.23 g,10.77mmol,0.83 mL). After stirring for 10 minutes at-45℃2- [2- (oxetan-3-yloxy) ethoxy ] was added]Ethanol (1.75 g,10.77mmol in DCM) and heating the mixture to 0deg.C over 1 hr, then pouring saturated NaHCO at 0deg.C 3 Aqueous solution (70 mL) and extracted with DCM (70 mL). The organic layer was washed with water (70 mL) and brine (70 mL), dried over anhydrous sodium sulfate, filtered and the filtrate concentrated. The residue was chromatographed on silica gel (100% ea) and then on reversed phase chromatography (60% ch 3 CN/water elution) to give (24E, 26E,28E,29E,35R,36S,37R,38R,40S,42S,45S,47R,48R, 57R) -57-hydroxy-45- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl as a white solid]-1-methyl-ethyl]-47, 48-dimethoxy-35,36,37,38,49,50-hexamethyl-44- [2- [2- (oxetan-3-yloxy) ethoxy ] ]Ethoxy group]-67, 68-dioxa-58-azatricyclohexa-24, 26,28 (49), 29 (50) -tetraen-51,52,53,54,55-pentanone (120 mg,21% yield).
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -12- [ (1R) -2- [ (1S, 3R, 4R) -4-dimethylphosphoryloxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-1-hydroxy-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.04,9 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-73). At 0℃to (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4-hydroxy-3-methoxy-cyclohexyl]-1-methyl-ethyl]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-30- [2- [2- (oxetan-3-yloxy) ethoxy ]]Ethoxy group]-11, 36-dioxa-4-azatricyclo [30.3.1.04,9 ]]To a solution of tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (0.3 g,0.28 mmol) in DCM (6 mL) was added 2, 6-di-tert-butyl-4-methylpyridine (0.437 g,2.13 mmol) and dimethylphosphonyl chloride (159.45 mg,1.42 mmol). The reaction was stirred at 0deg.C for 3.5 hours, then diluted with EtOAc and NaHCO 3 Washing with aqueous solution, washing with water, brine, washing with Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography (CH 3 CN/water=0% to 75%) to give I-73 (110 mg,34% yield) as a white solid. ESI-MS (EI) + ,m/z):1156.6[M+Na] +
Example 54: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -30- [2- [2- (dimethylamino) ethoxy ]]Ethoxy group]-1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) Phenyl) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.04,9 ]]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone Synthesis of (I-53)
Step 1: (1R, 9S,12S,15R,16E,18R,19R, 21)R,23S,24E,26E,28E,32S, 35R) -1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-30- [2- (2-iodoethoxy) ethoxy ]]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.04,9]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone. To a solution of intermediate V (0.24 g,0.247 mmol) in DCM (10 mL) was added TFA (844.38 mg,7.41mmol,0.57 mL) at-50deg.C. After 10 min, a solution of 2- (2-iodoethoxy) ethanol (1.07 g,4.94 mmol) in DCM (0.05 mL) was added and the mixture was stirred at-20℃for 5 h. The reaction was taken up with DCM and NaHCO 3 The aqueous solution was diluted and the organic layer was washed with water and brine, dried over Na 2 SO 4 Dried, filtered and concentrated. The residue was purified by reverse phase chromatography to give (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl as a white solid]-1-methyl-ethyl]-30- [2- (2-iodoethoxy) ethoxy ]]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.04,9]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (130 mg,46% yield).
Step 2: (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -30- [2- [2- (dimethylamino) ethoxy]Ethoxy group]-1-hydroxy-12- [ (1R) -2- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.04,9]Tricetyl-16,24,26,28-tetraene-2,3,10,14,20-pentanone (I-53). (1R, 9S,12S,15R,16E,18R,19R,21R,23S,24E,26E,28E,32S, 35R) -1-hydroxy-12- [ (1S, 3R, 4R) -4- (2-hydroxyethoxy) -3-methoxy-cyclohexyl]-1-methyl-ethyl]-30- [2- (2-iodoethoxy) ethoxy ] ]-18, 19-dimethoxy-15,17,21,23,29,35-hexamethyl-11, 36-dioxa-4-azatricyclo [30.3.1.04,9]A solution of tricetyl-16,24,26,28-tetraen-2,3,10,14,20-pentanone (0.36 g,0.31 mmol), N-methyl methylamine (0.42 g,9.34mmol,0.54 mL) and N-ethyl-N-isopropyl-propan-2-amine (1.21 g,9.34mmol,1.63 mL) in DCM (3.92 mL) was stirred at 25℃for 17 h. The reaction mixture was diluted with DCM (10 mL) and taken upSaturated NH 4 Cl (10 mL. Times.3), water (10 mL. Times.3) and brine (10 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate concentrated in vacuo. The residue was passed through a reverse phase column (with 50% CH 3 CN/water elution) to give I-53 (40 mg,12% yield) as a yellow solid. ESI-MS (EI) + ,m/z):1095.4[M+Na] +
Example 55: alphaLISA Ultra pS6K1 assay
Assay protocol
1. MCF-7 cells were seeded in Corning 3701 plates and incubated for 20 to 24 hours. From 12,000 to 16,000 cells will be seeded in 36 μl of medium per well.
2. The medium was replaced with fresh medium and incubated for an additional 2 hours.
3. 12. Mu.L (4X) of compound was added to the cell plate by HAMILTON. The final DMSO concentration was 0.5%. Incubate for 90 minutes.
4. 38. Mu.L was aspirated by HAMILTON, leaving 10. Mu.L per well.
5. mu.L of 2 Xlysis buffer was added using HAMILTON; the total volume in the wells was 20. Mu.L. The cells were allowed to oscillate for 30 minutes. The plates were covered with plastic foil and stored at-80 ℃ until analysis.
6. Cell lysates were thawed at room temperature and 10. Mu.L of lysates were transferred to assay plates (Optiplate-384).
7. mu.L of receptor beads were added to the assay plate and incubated for 2 hours
8. Add 5. Mu.L donor beads and incubate for 2 hours
9. Counting plate of multi-mode plate reader through EnSpire
Table 2: critical reagents/consumables
Example 56: alphaLISA Ultra pAKT assay
Assay protocol
1. MCF-7 cells were seeded in Corning 3701 plates and incubated for 20 to 24 hours.
16,000 to 20,000 cells will be seeded in 36 μl of medium per well.
2. The medium was replaced with fresh medium and incubated for an additional 90 minutes.
3. 12. Mu.L (4X) of compound was added to the cell plate by HAMILTON. The final DMSO concentration was 0.5%. Incubate for 2 hours.
4. 38. Mu.L was aspirated by HAMILTON, leaving 10. Mu.L per well.
5. mu.L of 2 Xlysis buffer was added using HAMILTON; the total volume in the wells was 20. Mu.L. The cells were allowed to oscillate for 30 minutes. The plates were covered with plastic foil and stored at-80 ℃ until analysis.
6. Cell lysates were thawed at room temperature and 10. Mu.L of lysates were transferred to assay plates (Optiplate-384).
7. mu.L of receptor beads were added to the assay plate and incubated for 2 hours
8. Add 5. Mu.L donor beads and incubate for 2 hours
9. Counting plate of multi-mode plate reader through EnSpire
Table 3: critical reagents/consumables
Example 57: western blot based pS6K1 and pAKT assays at time points of 24 hours and 48 hours
Assay protocol
1. Six well plates were seeded with 500,000 PC3 cells per well and incubated for 20 to 24 hours.
2. Compounds were added to the cell plates. Incubation was carried out for 24 to 48 hours.
4. Plates were placed on ice and the medium was removed by aspiration. The wells were washed with 1mL of 1 XPBS and then aspirated completely.
5. 110. Mu.L of 1% Triton lysis buffer was added and each well was scraped with force.
6. The cell homogenate was transferred to a 1.5mL Eppendorf tube on ice and spun at 10,000rpm for 10 minutes at 4 ℃.
7. The protein concentration of the resulting cell lysates was quantified using the Bradford assay and samples were analyzed by western blotting with 1×mes buffer on 4% -12% bis/Tris gels.
8. The gel was transferred onto a membrane at 50V for 100 min, blocked with Odyssey blocking buffer for 30 min, then incubated with primary antibodies (pS 6K 1T 389 rabbits or pAktS473 rabbits) overnight at 4 ℃ on a rotator.
9. Membranes were washed 3 times with TBS-T, incubated for 5 minutes between each wash, and then incubated with secondary antibody (LiCor IRDye 800 donkey anti-rabbit) for at least 30 minutes.
10. The membranes were washed 3 times with TBS-T and incubated for 5 minutes between each wash.
11. The gel was then incubated with PBS for 5 minutes at room temperature and then imaged using Li-Cor.
Table 4 shows the inhibitory activity (IC) of selected compounds of the invention in the pS6K1 assay and the pAKT assay 50 ) And their solubility in 100mM phosphate buffer (pH 7.4). The compound numbers correspond to those in table 1.
In the "mORC 1 selectivity @90 min" column of Table 4, pS6K1 and pAKT IC were compared by kinase assay 50 Whereas the compounds of the present invention that inhibit mTORC1 with higher selectivity than mTORC2 are denoted "yes". In the "mORC 1 selectivity @90 min" column of Table 4, pS6K1 and pAKT IC were compared by kinase assay 50 And the non-selective compounds are indicated by "no". In the "mTORC1 selectivity @24 hours" column of table 4, compounds of the invention that inhibit mTORC1 with higher selectivity than mTORC2 and maintain selectivity for at least 24 hours as determined by western blotting are represented by images of "yes" and western blotting assays as shown in fig. 1-9. In the "mTORC1 selectivity @24 hours" column of table 4, there is no selectivity at the 24 hour markThe compounds of (2) are indicated as "no". "N/A" means "not measured" and "N/C" means "not calculated".
The compound denoted "A" exhibited less than 1nM (x<1 nM) IC 50 . The compound denoted as "B" exhibits a concentration of greater than or equal to 1nM and less than 10nM (1 nM. Ltoreq.x)<10 nM) IC 50 . The compound denoted as "C" exhibits a concentration of greater than or equal to 10nM and less than 100nM (10 nM. Ltoreq.x)<100 nM) IC 50 . The compound denoted as "D" exhibits a concentration of greater than or equal to 100nM and less than 1. Mu.M (100 nM. Ltoreq.x)<1 μm) of IC 50 . The compound denoted as "E" exhibits an IC of greater than or equal to 1. Mu.M (1. Mu.M.ltoreq.x) 50
Table 4: assay data for exemplary Compounds
/>
/>
While we have described various embodiments of the invention, it is apparent that our basic examples can be varied to provide other embodiments that utilize the compounds and methods of the invention. It is, therefore, to be understood that the scope of the invention is defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

Claims (20)

1. A compound of formula I':
or a pharmaceutically acceptable salt thereof, wherein:
x and X 3 Independently a covalent bond, -CR 2 -、-NR-、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -, -O-, -S-or-SO 2 NR-;
L 1 Is covalent bond or C 1-30 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of said chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-SO 2 -、-C(O)-、-C(S)-、-CR 2 -、-CF 2 -、-P(O)(R)-、-SiR 2 -Si (OR) (R) -OR-NR-substitution;
each-Cy 1 -independently is an optionally substituted divalent ring selected from phenylene, 4-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur;
each R is independently hydrogen or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, or
Two R groups on the same atom are joined together with the atoms between them to form a 4-7 membered saturated, partially unsaturated or aryl ring having 0-3 heteroatoms independently selected from nitrogen, oxygen and sulfur in addition to the same atom to which they are attached;
L 2 is optionally substituted C 1-6 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1-2 methylene units of said chain are independently and optionally protected by-Cy 1 -substitution;
R 1 and R is 2 Independently hydrogen, halogen, -OR, -CN, - (CR) 2 ) 1-4 NR 2 、-COR、-CONR 2 、-CONR(CR 2 ) 1-4 NR 2 、-NO 2 、-NR 2 、-NR(C 1-6 Haloalkyl group),-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 、-P(O)R 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 8-10 membered bicyclic aromatic carbocycle, 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, 7-10 membered saturated or partially unsaturated bicyclic heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur;
R 3 is hydrogen, halogen, -OR OR-OSiR 3
R 3' Is hydrogen, halogen, -OR OR-OSiR 3
Or R is 3 And R is 3' Taken together to form =o or =s;
R 4 and R is 6 Is independently hydrogen, -OR, -NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 An aliphatic;
R 5 and R is 5' Each hydrogen or taken together to form =o or =nor;
X 1 and X 2 Each independently is-CR 2 -, -S-or-S (O) -,
wherein X is 1 And X 2 At least one of them is-CR 2 -。
2. The compound of claim 1, wherein the compound is selected from any one of the following formulas:
wherein:
x is-CR 2 -、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -or-SO 2 NR-,
Wherein:
X 3 is-CR 2 -、-NRCO-、-NRCO 2 -、-NRCONR-、-NRSO 2 -or-SO 2 NR-,
Wherein:
R 4 is-NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 The aliphatic group of the aliphatic hydrocarbon is a hydrocarbon,
wherein:
R 6 is-NR 2 、-NRCOR、-NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or optionally substituted C 1-6 The aliphatic group of the aliphatic hydrocarbon is a hydrocarbon,
or a pharmaceutically acceptable salt thereof.
3. The compound according to any one of claims 1 to 2, wherein L 1 Is C 1-10 A divalent straight or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 5 methylene units of said chain are independently and optionally protected by-Cy 1 -、-O-、-S-、-SO 2 -、-C(O)-、-C(S)-、-CR 2 -、-CF 2 -P (O) (R) -or-NR-substitution.
4. A compound according to any one of claims 1 to 3, wherein L 1 Selected from-CH 2 -、-CH 2 CH 2 -、-(CH 2 ) 3 -、-(CH 2 ) 4 -、-(CH 2 ) 5 -、-CH 2 CH 2 O-、-(CH 2 CH 2 O) 2 -、-(CH 2 CH 2 O) 3 -、-CH 2 CH 2 OCH 2 CH 2 -、-CH 2 CH 2 SO 2 CH 2 CH 2 O-and-CH 2 CH 2 OCH 2 CH 2 OCH 2 CH 2 -。
5. The compound according to any one of claims 1 to 2, wherein L 1 Is a covalent bond.
6. The compound of any one of claims 1 to 5, wherein each R is independently hydrogen or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 4-7 membered saturated or partially unsaturated heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and 5-6 membered heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
7. The compound according to any one of claims 1 to 6, wherein R 1 Selected from hydrogen, -OR, -CN, -NR 2 、-NR(C 1-6 Haloalkyl), -NRCOR, -NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 memberedA saturated or partially unsaturated monocyclic carbocyclic ring, a phenyl group, an 8-10 membered bicyclic aromatic carbocyclic ring, a 4-8 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, a 7-10 membered saturated or partially unsaturated bicyclic heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfur, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur.
8. The compound according to any one of claims 1 to 7, wherein R 1 Is methyl, -CH 2 CF 3 、-NH 2 、-NHMe、-NMe 2 、-SO 2 NH 2 、-CONH 2 、-CONMe 2 、-OCONHMe、-CO 2 H、
9. The compound according to any one of claims 1 to 8, wherein R 2 Selected from hydrogen, -OR, -CN, -NR 2 、-NR(C 1-6 Haloalkyl), -NRCOR, -NRCO 2 R、-NRCONR 2 、-NRSO 2 R、-SR、-SO 2 NR 2 Or an optionally substituted group selected from C 1-6 Aliphatic, 3-8 membered saturated or partially unsaturated monocyclic carbocycle, phenyl, 8-10 membered bicyclic aromatic carbocycle, 4-8 membered saturated or partially unsaturated monocyclic heterocycle having 1-2 heteroatoms independently selected from nitrogen, oxygen and sulfur, 7-10 membered heterocycle having 1-3 heteroatoms independently selected from nitrogen, oxygen and sulfurA saturated or partially unsaturated bicyclic heterocycle, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen and sulfur, and an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen and sulfur.
10. The compound according to any one of claims 1 to 9, wherein R 2 Is methyl, -CHF 2
11. The compound according to any one of claims 1 to 10, wherein the compound is selected from those shown in table 1.
12. A pharmaceutically acceptable composition comprising a compound according to any one of claims 1 to 11, and a pharmaceutically acceptable carrier, adjuvant or vehicle.
13. A method of treating a mTORC-mediated disease, disorder, or condition in a patient in need thereof, the method comprising administering to the patient a compound according to any one of claims 1 to 11, or a pharmaceutical composition thereof.
14. The method of claim 13, further comprising administering an additional therapeutic agent in combination with the compound.
15. The method of claim 13, wherein the mTORC-mediated disease, disorder, or condition is selected from the group consisting of type 1 and type 2 diabetic nephropathy and kidney-related complications, autosomal Dominant Polycystic Kidney Disease (ADPKD), autosomal Recessive Polycystic Kidney Disease (ARPKD), kidney disease associated with cyst formation or cyst formation, focal Segmental Glomerulosclerosis (FSGS) and other diseases associated with kidney sclerosis, laminopathies, age-related macular degeneration (AMD), diabetic macular edema, diabetic retinopathy, glaucoma, age-related retinal disease, immune system aging, respiratory tract infections, urinary tract infections, heart failure, osteoarthritis, pulmonary Arterial Hypertension (PAH), and Chronic Obstructive Pulmonary Disease (COPD).
16. The method of claim 13, wherein the mTORC-mediated disease, disorder, or condition is selected from Fragile X Syndrome (FXS), amyotrophic Lateral Sclerosis (ALS), epilepsy, focal Cortical Dysplasia (FCD), hemilateral megabrain malformation (HME), familial focal epilepsy with variable origins (FFEV), temporal Lobe Epilepsy (TLE), spasticity, neurodegenerative disease, down syndrome, rett syndrome (RTS), and diseases associated with activation or over-activation of mTOR signaling in the brain.
17. Use of a compound according to claims 1 to 11, or a pharmaceutical composition thereof, in the manufacture of a medicament for the treatment of a mTORC mediated disease, disorder, or condition in a patient in need thereof.
18. The use of claim 17, further comprising an additional therapeutic agent in combination with the compound.
19. The use of claim 17, wherein the mTORC-mediated disease, disorder, or condition is selected from the group consisting of type 1 and type 2 diabetic nephropathy and kidney-related complications, autosomal Dominant Polycystic Kidney Disease (ADPKD), autosomal Recessive Polycystic Kidney Disease (ARPKD), kidney disease associated with cyst formation or cyst formation, focal Segmental Glomerulosclerosis (FSGS) and other diseases associated with kidney sclerosis, laminopathies, age-related macular degeneration (AMD), diabetic macular edema, diabetic retinopathy, glaucoma, age-related retinal disease, immune system aging, respiratory tract infections, urinary tract infections, heart failure, osteoarthritis, pulmonary Arterial Hypertension (PAH), and Chronic Obstructive Pulmonary Disease (COPD).
20. The use of claim 17, wherein the mTORC-mediated disease, disorder, or condition is selected from Fragile X Syndrome (FXS), amyotrophic Lateral Sclerosis (ALS), epilepsy, focal Cortical Dysplasia (FCD), hemilateral megabrain malformation (HME), familial focal epilepsy with variable origins (FFEV), temporal Lobe Epilepsy (TLE), spasticity, neurodegenerative disease, down syndrome, rett syndrome (RTS), and diseases associated with activation or overactivation of mTOR signaling in the brain.
CN202280011222.6A 2021-01-22 2022-01-24 Rapamycin analogues and uses thereof Pending CN116917294A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/140523 2021-01-22
US202163202524P 2021-06-15 2021-06-15
US63/202524 2021-06-15
PCT/US2022/070308 WO2022159976A1 (en) 2021-01-22 2022-01-24 Rapamycin analogs and uses thereof

Publications (1)

Publication Number Publication Date
CN116917294A true CN116917294A (en) 2023-10-20

Family

ID=88351586

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280011222.6A Pending CN116917294A (en) 2021-01-22 2022-01-24 Rapamycin analogues and uses thereof

Country Status (1)

Country Link
CN (1) CN116917294A (en)

Similar Documents

Publication Publication Date Title
US11819476B2 (en) Rapamycin analogs and uses thereof
EP3813946B1 (en) Rapamycin analogs and uses thereof
EP3704120B1 (en) Heterocyclic compounds as prmt5 inhibitors
JP6082397B2 (en) Macrocyclic LRRK2 kinase inhibitor
WO2019127008A1 (en) Compound for targeted degradation of btk and application thereof
WO2020188467A1 (en) Condensed tricyclic compound used as kinase inhibitor
US10414727B2 (en) Phenyl amino piperidine mTORC inhibitors and uses thereof
US20220267352A1 (en) Rapamycin analogs and uses thereof
CN116917294A (en) Rapamycin analogues and uses thereof
EA045993B1 (en) RAPAMYCIN ANALOGUES AND THEIR APPLICATIONS
CN117964643A (en) Pyrrole [2,3-b ] pyridine derivative and preparation method and application thereof

Legal Events

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