(19)
(11)EP 3 153 165 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
06.05.2020 Bulletin 2020/19

(21)Application number: 15803906.5

(22)Date of filing:  03.06.2015
(51)International Patent Classification (IPC): 
A61K 31/395(2006.01)
C07D 405/00(2006.01)
C07D 405/14(2006.01)
C07D 491/18(2006.01)
(86)International application number:
PCT/CN2015/080632
(87)International publication number:
WO 2015/184983 (10.12.2015 Gazette  2015/49)

(54)

RAPAMYCIN DERIVATIVE, PREPARATION METHOD THEREFOR, AND PHARMACEUTICAL COMPOSITION AND USES THEREOF

RAPAMYCINDERIVAT, HERSTELLUNGSVERFAHREN DAFÜR UND PHARMAZEUTISCHE ZUSAMMENSETZUNG UND VERWENDUNG DAVON

DÉRIVÉS DE LA RAPAMYCINE, LEUR PROCÉDÉ DE PRÉPARATION, LA COMPOSITION PHARMACEUTIQUE ASSOCIÉE ET SES APPLICATIONS


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30)Priority: 03.06.2014 CN 201410242439

(43)Date of publication of application:
12.04.2017 Bulletin 2017/15

(73)Proprietor: Institute of Pharmacology and Toxicology Academy of Military Medical Sciences P.L.A. China
Beijing 100850 (CN)

(72)Inventors:
  • ZHONG, Wu
    Beijing 100850 (CN)
  • LI, Song
    Beijing 100850 (CN)
  • CAO, Shuang
    Beijing 100850 (CN)
  • CAO, Ruiyuan
    Beijing 100850 (CN)
  • XIAO, Junhai
    Beijing 100850 (CN)
  • ZHOU, Xinbo
    Beijing 100850 (CN)
  • LI, Xingzhou
    Beijing 100850 (CN)

(74)Representative: Weickmann & Weickmann PartmbB 
Postfach 860 820
81635 München
81635 München (DE)


(56)References cited: : 
EP-A2- 1 181 938
US-A- 4 650 803
US-A- 5 162 333
WO-A1-94/25468
US-A- 5 118 677
US-A- 5 349 060
  
      
    Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


    Description

    Technical Field



    [0001] The present invention pertains to the field of pharmaceutical chemicals, and relates to a rapamycin derivative, and a preparation method, pharmaceutical composition and use thereof.

    Background Art



    [0002] Rapamycin is also called as Sirolimus, and was separated and obtained from Streptomyces hygroscopicus by Veniza et al in 1975. In 1989, Rapamycin entered clinical phase as a new immunosuppressor, and it was marketed in 1999. Later, the inhibition effect of rapamycin on T lymphocyte proliferation teaches using it to inhibit cancer cells, and it is found that it shows good antineoplastic activity, and this compound as an anticancer drug has been developed by Wyeth Company of the USA and is about to enter clinic phase.

    [0003] The acting target of rapamycin in human body is mTOR (mammal rapamycin target protein). mTOR is an important component of PI3K-Akt-mTOR signal pathway. PI3K-Akt-mTOR signal pathway regulates proliferation- and apoptosis-related signal transduction pathways of tumor cells, so that on the one hand, proliferative activity of tumor cells is promoted to further enhance infiltration and metastasis abilities of tumor cells; and on the other hand, the acts on tumor apoptosis-related proteins promote activation of endogenous apoptosis inhibitors and/or expression and activation of apoptosis-inhibition-related protein kinases, thereby inhibiting apoptosis of tumor cells. Hence, PI3K-Akt-mTOR signal pathway may be the regulation core for tumor occurrence and development, so that mTOR is a key target for gene therapy of tumors.

    [0004] In animal body, mTOR exists mainly in two complex forms, i.e., mTORC1 and mTORC2, in which mTORC1 (mTOR complex 1) consists of 4 parts: mTOR, raptor (regulatory associated protein of mTOR), mLST8, PRAS40 (Proline-rich AKT Substrate 40 kDa); while mTORC2 (mTOR complex2) consists of 5 parts: mTOR, rictor (raptor independent), mLST8, mSIN1 (mammalian stress-activated protein kinase interacting protein 1), Protor-1 (Proteinobserved with rictor-1). mTORC1 and mTORC2 involves almost all different upstream activators and downstream effectors, and they mutually harmonize and regulate together cell cycle progression. The downstream effectors of mTORC1 mainly include ribosome p70S6 kinase protein (S6 kinase 1, S6K1) and eukaryotic initiation factor 4E binding protein 1(4EBP1). By regulating 4EBP1 and S6K1 phosphorylation, mTORC1 influences translation initiation of specific mRNA, thereby regulating protein synthesis and regulating cell growth and proliferation, in which its major biological function is to regulate protein synthesis. When mTORC1 is stimulated by a factor such as a growth factor, it can promote cell growth and proliferation via activating downstream S6K1 (Thr389 site). Once mTORC1 in vivo is over-activated, downstream 4EBP1 and S6K1 would overexpressed, and then cell proliferation is out of control, and then excessive immune or cancers would occur. mTORC2 mainly takes part in construction of cytoskeletal proteins, and mTORC2 can upregulate Akt level via acting on Ser473 site of Akt. (TANG Yan, Research summary of mTOR inhibitors [J]. Organic Chemistry, 2011, 31(7): 1144-1154.) The immunosuppression and antitumor mechanisms of rapamycin are mainly to inhibit mTORC1 via binding to cell receptor FKBP12, but rapamycin has very slight inhibition effects on mTORC2.



    [0005] Although rapamycin has potent anticancer activity, it has two severe defects: poor stability and poor water solubility. Rapamycin has a solubility of only 2.6 µg/ml in water, i.e., it is almost insoluble in water. After many years of research, it is found that modification of hydroxy at position 42 of rapamycin is a reliable way to improve physical and chemical properties of rapamycin, and some drugs in markets are based on this scheme.

    [0006] Some rapamycin derivatives with position 42 substituted in markets are listed as follows:
    Temsirolimus, of which the trade name is Torisel, is developed by Wyeth-Ayerst, and it is a rapamycin derivative with 42-acrylate, and is a prodrug of rapamycin. It is the earliest anticancer drug among rapamycin derivatives approved by FDA (2007). Temsirolimus is a first-line drug for treatment of advanced renal cell carcinoma (RCC) in patients, and it could extend median survival time by 3-6 months in early RCC patients. Temsirolimus can significantly inhibit T cell proliferation, its median inhibitory concentration (IC50) is 0.8 nmol/L, and it can be used in combination with kinase inhibitors such as Sunitinib or Sorafenib.

    [0007] Everolimus, of which the trade name is Zortress, is developed by Novartis Company, and is the first oral mTOR inhibitor, its chemical structure is 42-O-(2-hydroxyethyl)-rapamycin, the water solubility is superior to rapamycin, and it is rapidly hydrolyzed after entering human body. Experiments show that Everolimus has oral bioavailability of merely 15%- 30%, and half-life of 16 -19 h in human body. It firstly came into the market in Sweden in 2003, initially acted as only an immunosuppressor, and now it is approved to treat RCC, pancreatic neuroendocrine tumor (PNET) and subependymal giant cell astrocytoma (SEGA). FDA has approved that Everolimus oral tablets can be used for preventing adult kidney transplantation patients with lower or moderate immune risk from organ rejection response. Everolimus can be used simultaneously in combination with amount-reduced cyclosporine A and basiliximab and corticosteroids. The approved phase III test showed that Everolimus can prevent acute organ rejection and hold kidney function.

    [0008] Ridaforolimus (Ariad/Merck Company) has modification at C43-position of rapamycin, which improves its solubility and PK value. As for its research work, its phase-III clinic test in treatment of metastatic soft-tissue or bone sarcomas had been completed. In comparison with placebo group, medication group had a mortality risk which was reduced by 28% (NCT00538239). In 2012, Ariad and Merck Company filed with FDA a new drug application of ridaforolimus, but FDA expert panel had doubt in its therapeutic effects, and thus there were some obstruction in application.

    [0009] Zotarolimus (Medtroni Company) was approved by FDA in 2006 in Zotarolimus-coated coronary stent system (Endeavor), and in comparison with the bare-metal coronary stent group, the drug-coated group could effectively reduce death rate of arterial embolism, heart diseases and myocardial infarction in stent users.

    [0010] Umiroliums (Biolimus, Biosensors Company) was approved in Europe in 2007 in Biolimus-coated coronary stent system, biolimus A9. It is more potent than rapamycin in terms of immunosuppression and anti-inflammatory activity, and it hinders cell migration and proliferation by cell cycle arrest at G1, thereby preventing occurrence of vascular restenosis.

    [0011] At present, it is still in need to develop new rapamycin derivatives.

    Contents of Invention



    [0012] With deep research and inventive work, the inventors of the present invention obtain a series of novel rapamycin carboxylic ester derivatives (compounds of Formula I) with quaternary ammonium salt structures, which tremendously overcome defects of rapamycin in terms of water solubility and metabolism (in which, the compound of Example 14 has a water solubility improved by 40,000 times in comparison with rapamycin), and some compounds are superior to rapamycin in vitro antitumor activity, have smaller toxicity on norm cells than rapamycin, and have a very good druggability. Thus, the following invention is provided.

    [0013] One aspect of the present invention relates to a compound of Formula I, a pharmaceutically acceptable salt or hydrate thereof:

    wherein,
    R1 and R2 are independently selected from H, A and B, and R1, R2 cannot be H simultaneously;

    wherein, in Formula A or Formula B,

    arrows refer to sites where A or B links to mother ring of Formula I;

    n independently is 1, 2, 3, 4, 5, 6 or 7;

    R4 is independently selected from fluorine, chlorine, bromine, iodine, nitro, and cyano;

    X1, X2, X3, X4, Y1, Y2, Y3, Y4, Y5 are independently selected from C, S, O, N and Se atoms;

    X1∼X2, X2∼X3, X3∼X4, Y1∼Y2, Y2∼Y3, Y3∼Y4, Y4∼Y5 are independently single bond or double bond; (X1∼X2 and Y1∼Y2 do not from double bond, which is well-known in the art.)

    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 are independently selected from hydrogen atom, hydroxy, aldehyde group, carboxyl, amino, cyano, halogen, C1-C6 alkyl, C3-C10 cycloalkyl, C1-C6 alkoxy, C1-C6alkylthiol, C3-C10 cycloalkoxy, C1-C6 alkenyl, eneynylheterocyclic ring, heterocycloalkyl, substituted heterocycloalkyl, aromatic ring, aromatic heterocyclic ring, benzo-aromatic heterocyclic ring, wherein the C1-C6 alkyl, aromatic ring, aromatic heterocyclic ring, benzo-aromatic heterocyclic ring are not substituted or substituted with 1, 2, 3, 4 or 5 substituents independently selected from: -F, -Cl, -Br, -I, nitro, hydroxy, amino, cyano, C1-C6 alkylthiol, C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl and C1-C6 alkoxy.



    [0014] The N atom together with X1, X2, X3, X4, or with Y1, Y2, Y3, Y4, Y5 form a heterocyclic structure; namely:

    in Formula A, N atom together with X1, X2, X3, X4 form a thiazole ring, and/or

    in Formula B, N atom together with Y1, Y2, Y3, Y4, Y5 form a pyridine ring.



    [0015] As for the compound of Foumula I according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, preferably:
    Z1, Z2, Z3, Z4, Z5, Z6 Z7, Z8, Z9 are independently selected from hydrogen atom, hydroxy, aldehyde group, carboxyl, amino, cyano, halogen, C1-C3 alkyl, C3-C6 cycloalkyl, C1-C3 alkoxy, C1-C3 alkylthiol, C3-C6 cycloalkoxy, and C1-C3 alkyl-enyl.

    [0016] As for the compound of Formula I according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, preferably:
    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 are independently selected from hydrogen atom, hydroxyl and methyl.

    [0017] As for the compound of Formula I according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, preferably:

    R1 and R2 are independently selected from H, carbonylmethyl-(4-methyl-thiazole R4 salt-3-yl), carbonylmethyl-(4, 5-dimethyl-thiazole R4 salt-3-yl), carbonylmethyl-(pyridine R4 salt-1-yl), carbonylmethyl-(3-hydroxy-pyridine R4 salt-1-yl), carbonylmethyl-(3-methyl-pyridine R4 salt-1-yl) and carbonylmethyl-(4-methyl-pyridine R4 salt-1-yl), wherein R4 is independently selected from fluorine, chlorine, bromine, iodine, nitro and cyano;

    and R1, R2 cannot be H simultaneously.



    [0018] As for the compound of Formula I according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, preferably:

    R1 and R2 are independently selected from H, carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl), carbonylmethyl-(4, 5-dimethyl-thiazole bromide salt-3-yl), carbonylmethyl-(pyridine bromide salt-1-yl), carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl), carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl) and carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl);

    and R1, R2 cannot be H simultaneously.



    [0019] In one embodiment, the compounds, their pharmaceutically acceptable salts or their hydrates of the present invention are selected from Table 1.
    Table 1: Some compounds of the present invention
    Example/ Compound No.NameStructure Formula
    1 31,42-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin

    2 42-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin

    3 31-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin

    4 31,42-O-carbonylmethyl-(4, 5-dimethyl-thiazole bromide salt-3-yl)-rapamycin

    5 42-O-carbonylmethyl-(4, 5-dimethyl-thiazole bromide salt-3-yl)-rapamycin

    6 31-O-carbonylmethyl-(4, 5-dimethyl-thiazole bromide salt-3-yl)-rapamycin

    7 31,42-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin

    8 31,42-O-carbonylmethyl-(3 -hydroxy-pyridine bromide salt-1-yl)-rapamycin

    9 31,42-O-carbonylmethyl-(3 -methyl-pyridine bromide salt-1-yl)-rapamycin

    10 42-O-carbonylmethyl-(py ridine bromide salt-1-yl)-rapamycin

    11 42-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin

    12 42-O-carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl)-rapamycin

    13 42-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin

    14 31,42-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin

    15 31-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin

    16 31-O-carbonylmethyl-(py ridine bromide salt-1-yl)-rapamycin

    17 31-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin



    [0020] Another aspect of the present invention relates to a method for preparing the compound of Formula I according to any one of items of the present invention, which comprises steps of any one of the following processes (1) to (3):

    Process (1): Preparation of compounds of Formula I with position 31,42 bis-substituted

    Process (2): Preparation of compounds of Formula I with position 42 mono-substituted

    Process (3): Preparation of compounds of Formula I with position 31 mono-substituted

    wherein in the above processes (1) to (3),
    R3 is independently selected from F, Cl, Br and I;
    the meanings of other notations are independently the same as stated in any one of the above items.

    [0021] In Process (1), rapamycin is firstly reacted with acyl halide to generate 31- and 42-bis-esterification product (Intermediate 1), then the product is reacted with heterocyclic ring to generate quaternary ammonium salt type rapamycin derivatives with position 31, 42 bis-subsituted.

    [0022] In Process (2), rapamycin firstly reacts with sufficient trimethylcholosilane to generate Intermediate 2, then its ether bond at position 31 is hydrolyzed in the presence of an acid to generate Intermediate 3, then this intermediate is substituted at position 42 with halogenated acryl halide to obtain Intermediate 4, then the trimethylsilyl ether at position 31 is hydrolyzed with an acid to obtain 42-mono-esterification product (Intermediate 5); finally, Intermediate 5 is reacted with azacyclic ring to generate quaternary ammonium salt type rapamycin derivatives with position mono-substituted.

    [0023] In Process (3), Intermediate 3 is substituted at position 42 with tert-butyldimethylchlorosilane to obtain Intermediate 6; subsequently, trimethylsilyl ether at position 31 is hydrolyzed with an acid to obtain Intermediate 7; Intermediate 7 reacts with halogenated acyl halide to obtain Intermediate 8; then tert-butyldimethylsilyl ether is hydrolyzed under acidic condition to obtain Intermediate 9; finally, Intermediate 9 is reacted with azacyclic ring to generate quaternary ammonium salt type rapamycin derivatives with position 31 mono-substituted.

    [0024] In the last step of the Process (1), i.e., in the reaction from Intermediate 1 to the final product in Process (1), a reaction reagent (hereinafter cited as R5) can be added.

    [0025] In the last step of the Process (2), i.e., in the reaction from Intermediate 5 to the final product in Process (2), a reaction reagent (hereinafter cited as R5) can be added.

    [0026] In the last step of the Process (3), i.e., in the reaction from Intermediate 9 to the final product in Process (3), a reaction reagent (hereinafter cited as R5) can be added.

    [0027] The reagent R5 is independently selected from thiazole, pyridine, thiazole substituted with one or more C1-C6 alkyl, pyridine substituted with one or more C1-C6 alkyl, thiazole substituted with one or more halogen atoms, and pyridine substituted with one or more halogen atoms; more preferably, selected from 4-methylthiazole, 4, 5-dimethylthiazole, pyridine, 3-hydroxypyridine, 3-methylpyridine and 4-methylpyridine.

    [0028] Another aspect of the present invention relates to a pharmaceutical composition, which comprises the compound of any one of items of the preset invention, a pharmaceutically acceptable salt or hydrate thereof, and optionally a pharmaceutically acceptable excipient.

    [0029] The compounds of the present invention all have very good water solubility, are convenient to prepare various dosage forms. When used for oral administration, the compound of the present invention can be processed in any preparation dosage forms suitable for oral administration, including but not being limited to tablets, capsules, aqueous solutions or aqueous suspensions, in which carriers of tablets usually include lactose and corn starch, and lubricants such as magnesium stearate may also be added. Diluents of capsules usually include lactose and dry corn starch. In aqueous suspensions, active ingredient is usually used together with suitable emulsifying agents and suspending agents. If required, the above dosage forms for oral administration may further be added with some sweeting agents, fragrances or coloring agents.

    [0030] The compound of the present invention can also be administered in sterile injection dosage form, including sterile injection water or oil suspension or sterile injection solution, wherein the usable carriers and solvents include water, Ringer's solution and isotonic NaCl solution.

    [0031] The compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention, can be used as a medicament.

    [0032] The compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention, can be used for suppressing immune, inhibiting mTOR, inhibiting mTORC1, inhibiting PI3K-Akt-mTOR signal pathway, inhibiting T lymphocyte proliferation, anti-tumor, promoting tumor cell apoptosis, allowing cell cycle arrest at G1, reducing arterial embolism, anti-aging, anti-Alzheimer's disease, preventing organ rejection, anti-inflammatory or anti-microbial; preferably, the anti-tumor refers to treatment and/or prophylaxis and/or adjuvant therapy of kidney cancer, lymphoma, lung cancer, liver cancer, breast cancer, neuroendocrine cancer, uterine sarcoma or gastric cancer.

    [0033] The present invention further relates to a method for treatment and/or prophylaxis and/or adjuvant therapy of kidney cancer, lymphoma, lung cancer, liver cancer, breast cancer, neuroendocrine cancer, uterine sarcoma or gastric cancer, comprising a step of using an effective amount of the compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention.

    [0034] The administration dosage of the compound of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention, depends on many factors, for examples, properties and severities of diseases to which the treatment or adjuvant therapy is applied, gender, age, body weight and individual responses of patients or animals, specific compounds to be used, administration routes and times, etc. The dosage can be administrated in a single dosage form or in 2, 3, 4 dosage forms.

    [0035] Further another aspect of the present invention relates to a use of the compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention, in the manufacture of an immunosuppressant, a mTOR inhibitor, a mTORC1 inhibitor, a drug for inhibiting PI3K-Akt-mTOR signal pathway, a drug for inhibiting T lymphocyte proliferation, an antitumor drug, drug for promoting tumor cell apoptosis, a drug allowing cell cycle arrest at G1, a drug for preventing organ rejection, a drug for lowering arterial embolism, an anti-aging drug, drug against Alzheimer's disease, an anti-inflammatory drug or an antibacterial drug; preferably, the antitumor drug is a medicament for treatment and/or prophylaxis and/or adjuvant therapy of kidney cancer, lymphoma, lung cancer, liver cancer, breast cancer, neuroendocrine cancer, uterine sarcoma or gastric cancer.

    [0036] Further another aspect of the present invention relates to a method for suppressing immune, inhibiting mTOR, inhibiting mTORC1, inhibiting PI3K-Akt-mTOR signal pathway, inhibiting T lymphocyte proliferation, combating tumors, promoting tumor cell apoptosis, allowing cell cycle arrest at G1, lowering arterial embolism, combating aging, combating Alzheimer's disease, preventing organ rejection, combating inflammation or combating microbes in vivo or in vitro, comprising a step of using an effective amount of the compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention.

    [0037] In one embodiment of the present invention, the "in vitro" method is for non-therapeutic purpose.

    [0038] Further another aspect of the present invention relates to a coronary stent, comprising a drug coating comprising the compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention.

    [0039] The present invention further relates to a use of the compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention, in the manufacture of the coronary stent.

    [0040] The present invention further relates to a method for preparing the coronary stent, comprising a step of using an effective amount of the compound according to any one of items of the present invention, a pharmaceutically acceptable salt or hydrate thereof, or the pharmaceutical composition of the present invention.

    In the present invention,



    [0041] The term "C1-C6 alkyl" refers to a straight or branched alkyl having 1-4 carbon atoms, for example, methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, pentyl, 2-pentyl, isopentyl, neopentyl, hexyl, 2-hexyl, 3-hexyl, etc.; C1-C4 alkyl, C1-C3 alkyl or C1-C2 alkyl can be interpreted similarly. Specific alkyl can be C1-C4 alkyl, C1-C3 alkyl or C1-C2 alkyl.

    [0042] The term "C1-C6 alkoxy" refers to a straight or branched alkoxy having 1-6 carbon atoms, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentoxy, isopentoxy, neo-pentoxy, hexyloxy, 2-hexyloxy, 3-hexyloxy, etc.; C1-C4 alkoxy, C1-C3 alkoxy or C1-C2 alkoxy can be interpreted similarly. Specific alkoxy is C1-C4 alkoxy, C1-C3 alkoxy or C1-C2 alkoxy.

    [0043] The term "C1-C6alkylthio"can be interpreted in a way similar to "C1-C6 alkoxy", which difference lies in oxygen atom is replaced with sulfur atom.

    [0044] The term "C3-C10cycloalkyl" refers to saturated carbocyclic group having 3-10 carbon atoms. The cycloalkyl can be monocyclic or polycyclic fused system, and can be fused on aromatic ring. Examples of these groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. In the text, the cycloalkyl is not substituted or substituted with various groups at one or more substitutable positions. For example, these cycloalkyls can be substituted with following groups: C1-C6 alkyl, C1-C6 alkoxy, cyano, halogen, hydroxy, amino, nitro, mono(C1-C6) alkylamino, di(C1-C6) alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 halogenated alkyl, C1-C6 halogenated alkoxy. C3-C6 cycloalkyl can be interpreted similarly.

    [0045] The term "C3-C10cycloalkoxy" refers to saturated cycloalkoxy group havine3-10 carbon atoms. The cycloalkoxy can be monocyclic or polycyclic fused system, and can be fused on an aromatic ring. Examples of these groups include cyclopropoxy, cycloprobutoxy, cyclopentoxy and cyclohexyloxy. In the test, the cycloalkoxy is not substituted or substituted with various groups at one or more substitutable positions. For example, these cycloalkoxys can be optionally substituted with following groups: C1-C6 alkyl, C1-C6 alkoxy, cyano, halogen, hydroxy, amino, nitro, mono(C1-C6) alkylamino, di(C1-C6) alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 halogenated alkyl, C1-C6 halogenated alkoxy. C3-C6cycloalkoxy can be interpreted similarly.

    [0046] The term "C2-C6 alkenyl" refers to alkenyl having 2-6 carbon atoms and at least one double bond, and includes ethenyl, propenyl, 1-buten-3-yl, 1-penten-3-yl, 1-hexen-5-yl; C3-C5 alkenyl can be interpreted similarly. C3-C5 alkenyl is preferred.

    [0047] The term "C2-C6 alkynyl" refers to hydrocarbonyl having 2-6 carbon atoms and at least one triple bond, and includes ethynyl, propynyl, butynyl, pentyn-2-yl; C3-C5 alkynyl can be interpreted similarly. C3-C5 alkynyl is preferred.

    [0048] The term "halogen" or "halogen atom" refers to fluorine, chlorine, bromine, or iodine atom.

    [0049] The term "aromatic ring" or "aryl" refers to aromatic carbocyclic group having monocyclic ring (e.g., phenyl), polycyclic ring (e.g., biphenyl) or polycyclic fused rings with at least one aromatic ring (e.g., 1,2,3,4-tetrahydronaphthyl, naphthyl), which can be optionally mono-, bis- or tri-substituted with halogen, lower alkyl, lower alkoxy, trifluoromethyl, aryl, heteroaryl and hydroxy.

    [0050] The term "arylalkyl" refers to alkyl (as defined above) substituted with one or more aryl groups (as defined above). Preferably, aryl alkyl is aryl-C1-C3 alkyl. Examples include benzyl, phenylethyl.

    [0051] The term "aromatic heterocyclic ring" or "heteroaryl" refers to 5-, 6- or 7-membered monocyclic or polycyclic aromatic ring system, including fused ring system comprising 5-10 atoms (in which at least one ring is aromatic ring), and the ring system contains at least one and at most four heteroatoms selected from nitrogen, oxygen or sulfur. Examples of heteroaryl is pyridinyl, imidazolyl, pyrimidyl, pyrazolyl, triazolyl, pyrazinyl, tetrazyl, furyl, thienyl, isoxazole, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolone ring, isoquinolone ring, indole ring, benzo-imidazole, benzo-furan ring, benzo-thiophene, benzo-thiazole ring, pyridazine ring, etc., which is optionally mono-, bis- or tri-substituted with halogen, lower alkyl, lower alkoxy, trifluoromethyl, aryl, heteroaryl and hydroxy.

    [0052] The term "heterocyclic ring" or "heterocyclic group" refers to 5-, 6- or 7-membered monocyclic or polycyclic ring system, including fused ring system comprising 4-10 atoms, and the ring system contains at least one and at most four heteroatoms selected from nitrogen, oxygen or sulfur, with proviso that the ring group does not contain two adjacent O or S atoms. The fused ring system can be heterocyclic ring fused on aromatic group. Preferred heterocyclic rings include but are not limited to pyrrolylalkyl, tetrahydrofuryl, dihydrofuryl, tetrahydrothiophanyl, piperidyl, morpholine ring, hexamethylene ring, piperazinyl, and they can be substituted with following groups: C1-C6 alkyl, C1-C6 alkoxy, cyano, halogen, hydroxy, amino, nitro, mono(C1-C6) alkylamino, di(C1-C6) alkylamino, C2-C6 alkenyl, C2-C6alkynyl, C1-C6 halogenated alkyl, C1-C6 halogenated alkoxy.

    [0053] When applied to "in vivo", the term "effective amount" refers to a dose that can realize therapy, prophylaxis, alleviation and/or remission of the diseases or disorders of the present invention.

    [0054] The term "subject" can refer to a patient or an animal, especially a mammal, such as human, dog, monkey, cattle, horse, etc., which accepts the composition of the present invention for therapy, prophylaxis, alleviation and/or remission of the diseases or disorders of the present invention.

    [0055] The term "disease and/or disorder" refers to a physical state of the subject, in which the physical state relates to the diseases and/or disorders of the present invention.

    Brief Description of the Drawings



    [0056] Fig.1: Inhibition effects of compounds on phosphorylation of Thr389 of S6K1 and Ser473 of Aktin tumor cell A549; A, compounds of Examples No. 1-8; B, Compounds of Examples No. 9-17.

    Specific Models for Carrying Out the Invention



    [0057] The embodiments of the present invention are illustrated in details in conjunction with examples. However, those skilled in the art would understand the following examples are merely to illustrate the present invention, rather than to limit the scope of the present invention. When specific conditions are not given in the examples, conventional conditions or conditions recommended by manufacturers are applied. The reagents or instruments which manufactures are not given are all conventional products commercially available in markets.

    [0058] Melting points of compounds are measured by SRY-1 type melting point instrument, in which temperatures are not calibrated. 1H-NMR spectra are measured by VARIAN INOVA 600 type NMR spectrometer. Mass spectra are measured byAPI-150EX LC/MS high resolution magnetic mass spectrometer.

    Example 1: Preparation of 31,42-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin (Compound 1)



    [0059] 

    Step 1: 1g (1.09mmol) of rapamycin was dissolved in 10ml of dry dichloromethane, cooled to -10°C, then added with 0.87g (11mmol) of dry pyridine. To the reaction solution, 2.2g (10mmol) of bromoacetyl bromide in 5ml of dichloromethane solution was slowly added dropwise, the addition was finished within 30min, and the reaction was continued for 10min then stopped. The reaction solution was adjusted to neutral pH with 1mol/L hydrochloric acid, washed with distilled water 20ml×3, then purified with column chromatography, to obtain 0.73g of Intermediate 1 in form of white foam (n=1, R3=Br, R4=Br).

    Step 2: 0.73g (0.63mmol) of Intermediate 1 (n=1, R3=Br, R4=Br) was dissolved in 20ml of acetone, added with 0.94g (9.45mmol) of 4-methylthiazole, reacted at 60°C for 5h, the product was purified by column to obtain 0.7g of Compound 1 in form of light yellow granules.



    [0060] M.p.142-145 °C;MS:1193 [M-2Br-H]+,597[(M-2Br)/2]+; 1H-NMR (600MHz,DMSO-d6 δ ppm), 10.18 (d,2H), 8.07 (s,2H), 6.46 (s,1H), 6.38 (m, 1H), 6.24 (m, 1H), 6.22 (m, 1H), 5.71 (s, 1H), 5.59(s, 2H), 5.46 (m, 1H), 5.33 (s, 1H), 4.91 (m, 3H), 4.53 (s, 2H).

    Example 2: Preparation of 42-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin (Compound 2)



    [0061] 

    Step 1: 2g (2.19mmol) of rapamycin was dissolved in 25mlof dry ethyl acetate, cooled to 0-5°C. To the reaction solution,1.50g (22mmol) imidazole was added, 1.2g (11mmol) of trimethylchlorosilane in 5ml of ethyl acetate solution was slowly added dropwise within 30min, until the conversion to Intermediate 2 was completed. To the reaction solution, 10ml of 0.5mol/L H2SO4 solution was slowly added dropwise within 3h, until the conversion of Intermediate 2 was completed, and then the product was purified with column chromatography, to obtain 1.3g of Intermediate 3 in form of white foam.

    Step 2: 1.3g (1.3mmol) of Intermediate 3 was dissolved in 15ml of dichloromethane, cooled to -10°C, then added with 1.03g (13mmol) of dry pyridine. To the reaction solution, 1.31g (6.5mmol) of bromoacetyl bromide in 5ml solution was slowly added dropwise, and the addition was completed within 30min, the reaction was continued for 10min then stopped. The reaction solution was adjusted to neutral pH with 1mol/L hydrochloric acid, washed with distilled water20ml×3, then purified with column chromatography, to obtaining of Intermediate 4 (n=1, R3=Br, R4=Br) in form of white foam.

    Step 3: 1.2g (1.1mmol) of Intermediate 4 (n=1, R3=Br, R4=Br) was dissolved in 15mlof acetone, added dropwise with 0.7ml of 1mol/LH2SO4 aqueous solution, the addition was completed within 1h, the reaction was continued for 30min, and the conversion of the reactant was completed. By column separation and purification, 1.05g of Intermediate 5 (n=1, R3=Br, R4=Br) in form of white foam was obtained.

    Step 4: 1.05g (0.95mmol) of Intermediate 5 (n=1, R3=Br, R4=Br) was dissolved in 20mlof acetone, added with 0.94g (9.5mmol) of 4-methylthiazole, reacted at 60°C for 5h, the product was purified by column to obtain 1.02g of Compound 2 in form of light yellow granules.



    [0062] M.p.130-133°C;MS:1053.8 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 10.16 (d,1H), 8.06 (s, 1H), 6.45 (s,1H), 6.38 (m, 1H), 6.22 (m, 1H), 6.15 (m, 1H), 5.62 (s, 2H), 5.46(M, 1H), 5.24 (s, 1H), 5.08 (d, 1H), 4.97 (s, 1H), 4.93 (s, 1H), 4.68 (m, 1H), 4.00 (s, 2H), 3.97 (s, 1H).

    Example 3: Preparation of 31-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin (Compound 3)



    [0063] 

    Step 1: 2g (2mmol) of Intermediate 3 was dissolved in 20ml of ethyl acetate, cooled to 0∼5°C. To the reaction solution, 1.36g (20mmol) of imidazole was added, 1.5g (10mmol) of tert-butyldimethylchlorosilane in 5ml of ethyl acetate solution was slowly added dropwise with within 30min, the reaction was continued for 30min; by column separation, 1.3g of Intermediate 6 was obtained. To the reaction solution, 0.5ml of 0.5mol/LH2SO4 solution was added within 30min, and the addition was finished within 1h, until the conversion of reactant was completed. By column separation and purification, 1.05g of Intermediate 7 in form of white foam was obtained.

    Step 2: 1.05g (1.02mmol)Intermediate 7 was dissolved in 10ml of dichloromethane, cooled to -10°C, then added with 0.81g (10.2mmol)of dry pyridine. To the reaction solution, 1.03g (5.1mmol) of bromoacetyl bromide in 5ml solution was slowly added dropwise, the addition was completed within 30min. The reaction was continued for 10min and then stopped. The reaction solution was adjusted to neutral pH with 1mol/L hydrochloric acid, washed with distilled water 20ml×3, then purification was carried out by using column chromatography method, to obtain 0.73g of Intermediate 8 (n=1, R3=Br, R4=Br) in form of white foam.

    Step 3: 0.73g (0.64mmol) of Intermediate 8 (n=1, R3=Br, R4=Br) was dissolved in 10ml of acetone, added dropwise with 0.5ml of 2mol/L H2SO4 solution, the addition was completed within 30min, the reaction was continued for 30min, until the conversion of reactant was completed. By column separation and purification, 0.4g of Intermediate 9 (n=1, R3=Br, R4=Br) in form of white foam was obtained.

    Step 4: 0.4g (0.39mmol) of Intermediate 9 (n=1, R3=Br, R4=Br) was dissolved in 20mlof acetone, added with 0.38g (3.90mmol) of 4-methylthiazole, reacted at 60°C for 5h; by column separation, 0.27g (0.24mmol) of Compound 3 in form of light yellow granules was obtained.



    [0064] M.p.130-132°C; MS:1054 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 10.16 (d,1H), 8.05 (s, 1H), 6.45 (s,1H), 6.37 (m, 1H), 6.25 (m, 1H), 6.18 (m, 2H), 5.70 (s, 2H), 5.46(m, 1H), 5.35 (s, 1H), 4.55 (d 2H), 4.04 (s, 1H).

    Example 4: Preparation of 31,42-O-carbonylmethyl-(4,5-dimethyl-thiazole bromide salt-3-yl)-rapamycin (Compound 4)



    [0065] Preparation method was referred to Example 1 (n=1, R3=Br, R4=Br, R5= 4, 5-dimethylthiazole).

    [0066] m.p.135-139°C;MS:1220.9 [M-2Br-H]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 10.03 (d,2H), 6.46 (s,1H), 6.37 (m, 1H), 6.24 (m, 1H), 6.18 (m, 1H), 6.12 (m, 1H), 5.76 (m, 3H), 5.62(s, 2H), 5.46 (m, 1H), 5.32 (s, 1H), 5.32 (s, 1H), 4.93 (s, 1H), 4.83 (m, 2H), 4.61 (s, 1H), 4.55 (s, 1H), 4.52 (s, 1H).

    Example 5: Preparation of 42-O-carbonylmethyl-(4,5-dimethyl-thiazole bromide salt-3-yl)-rapamycin (Compound 5)



    [0067] Preparation method was referred to Example 2 (n=1, R3=Br, R4=Br, R5= 4, 5-dimethylthiazole).
    m.p.135-138°C; MS:1067.9 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 10.03 (d,1H), 6.49 (s,1H), 6.37 (m, 1H), 6.21 (m, 1H), 6.14 (m, 2H), 5.62 (m, 2H), 5.46 (m, 1H), 5.23 (s, 1H), 5.08 (s, 1H), 4.97 (s, 1H), 4.93 (s, 1H), 4.68 (s, 1H), 4.06 (s, 2H), 4.01 (s, 1H).

    Example 6: Preparation of 31-O-carbonylmethyl-(4, 5-dimethyl-thiazole bromide salt-3-yl)-rapamycin (Compound 6)



    [0068] Preparation method was referred to Example 3 (n=1, R3=Br, R4=Br, R5= 4, 5-dimethylthiazole).
    m.p.125-127°C; MS:1067.7 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 10.02 (d,1H), 6.45 (s,1H), 6.37 (m, 1H), 6.25 (m, 1H), 6.20 (m, H), 6.15 (m, H), 5.70 (m, 2H), 5.45 (m, 1H), 5.33 (s, 1H), 5.08 (s, 1H), 4.93 (s, 1H), 4.81 (m, 1H), 4.56 (m, 3H), 4.03 (m, 1H).

    Example 7: Preparation of 31,42-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin (Compound 7)



    [0069] Preparation method was referred to Example 1 (n=1, R3=Br, R4=Br, R5=pyridine).
    m.p.140-142°C; MS:1153 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 8.96 (d, 4H), 8.72 (m, 2H), 8.22 (m, 4H), 6.44 (s, 1H), 6.40 (m, 1H), 6.24 (m, 1H), 6.17 (m, 2H), 5.87 (m, 1H), 5.79 (m, 1H), 5.48 (m, 1H), 5.29 (s, 1H), 4.95 (m, 2H), 4.81 (s, 1H), 4.52 (m, 3H).

    Example 8: Preparation of 31,42-O-carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl)-rapamycin (Compound 8)



    [0070] Synthesis method was referred to Example 1 (n=1, R3=Br, R4=Br, R5= 3-hydroxypyridine).
    m.p.120-124°C; MS:1185.4 [M-2Br-H]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 12.04 (s, 2H), 8.60 (m, 4H), 8.00 (m, 4H), 6.43 (m, 2H), 6.12 (m, 3H), 5.61 (m, 3H), 5.56 (m, 1H).

    Example 9: Preparation of 31,42-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin (Compound 9)



    [0071] Preparation method was referred to Example 1 (n=1, R3=Br, R4=Br, R5= 3-methylpyridine).
    m.p.159-162°C; MS:1181.2 [M-2Br-H]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 9.00 (s, 1H), 8.89 (m, 2H), 8.81 (m, 1H), 8.57 (s, 2H), 8.14 (m, 2H), 6.46 (s, 1H), 6.38 (m, 1H), 6.23 (m, 1H), 6.14 (m, 2H), 5.80 (m, 5H), 5.46 (m, 1H), 5.29 (m, 1H).

    Example 10: Preparation of 42-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin (Compound 10)



    [0072] Preparation method was referred to Example 2 (n=1, R3=Br, R4=Br, R5=pyridine).
    m.p.135-137°C; MS:1034.1 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 9.07 (d, 1H), 9.02 (m, 1H), 8.71 (m, 1H), 8.24 (m, 2H), 8.14 (m, 2H), 6.45 (s, 1H), 6.41 (m, 1H), 6.22 (m, 1H), 6.14 (m, 1H), 5.80 (m, 5H), 5.67 (m, 2H), 5.44 (m, 1H), 5.25 (s, 1H), 5.08 (m, 1H).

    Example 11: Preparation of 42-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin (Compound 11)



    [0073] Preparation method was referred to Example 2 (n=1, R3=Br, R4=Br, R5= 3-methylpyridine).
    m.p.138-140°C; MS:1048 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 9.07 (d, 1H), 9.02 (m, 1H), 8.71 (m, 1H), 8.24 (m, 2H), 8.14 (m, 2H), 6.45 (s, 1H), 6.41 (m, 1H), 6.22 (m, 1H), 6.14 (m, 1H), 5.80 (m, 5H), 5.67 (m, 2H), 5.44 (m, 1H), 5.25 (s, 1H), 5.08 (m, 1H).

    Example 12: Preparation of 42-O-carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl)-rapamycin (Compound 12)



    [0074] Preparation method was referred to Example 2 (n=1, R3=Br, R4=Br, R5= 3-hydroxypyridine).
    m.p.146-148°C; MS:1050 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 12.08 (s, 1H), 8.60 (m, 1H), 8.53 (m, 1H), 8.02 (m, 2H), 6.45 (m, 2H), 6.38 (s, 1H), 6.22 (m, 1H), 6.12 (m, 2H), 5.60 (m, 2H), 5.48 (m, 1H), 5.25 (m, 1H), 5.08 (m, 1H).

    Example 13: Preparation of 42-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin (Compound 13)



    [0075] Preparation method was referred to Example 2 (n=1, R3=Br, R4=Br, R5= 4-methylpyridine).
    m.p.157-160°C; MS:1048.1 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 8.89 (d, 1H), 8.05 (m, 2H), 6.45 (s, 1H), 6.41 (m, 1H), 6.22 (m, 1H), 6.12 (m, 2H), 5.61 (m, 3H), 5.46 (m, 1H), 5.25 (m, 1H), 5.08 (d, 1H), 4.93 (m, 2H).

    Example 14: Preparation of 31,42-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin (Compound 14)



    [0076] Preparation method was referred to Example 1 (n=1, R3=Br, R4=Br, R5= 4-methylpyridine).
    m.p.181-183°C; MS:1181.2 [M-2Br-H]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 8.90 (d, 2H), 8.78 (d, 2H), 8.06 (m, 4H), 6.46 (d, 1H), 6.38 (m, 1H), 6.20 (m, 1H), 6.14 (m, 2H), 5.80 (d, 1H), 5.62 (m, 4H), 5.47 (m, 1H), 5.29 (s, 1H).

    Example 15: Preparation of 31-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin (Compound 15)



    [0077] Preparation method was referred to Example 3 (n=1, R3=Br, R4=Br, R5= 3-methylpyridine).
    m.p.138-140°C; MS:1048.1 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 8.88 (d, 1H), 8.80 (d, 1H), 8.57 (m, 1H), 8.12 (m, 1H), 6.45 (m, 1H), 6.40 (m, 1H), 6.23 (m, 1H), 6.17 (m, 2H), 5.81 (d, 1H), 5.74(m, 2H), 5.47 (m, 1H) 5.28 (s, 1H).

    Example 16: Preparation of 31-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin (Compound 16)



    [0078] Preparation method was referred to Example 3 (n=1, R3=Br, R4=Br, R5=pyridine).
    m.p.140-142°C; MS:1034 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 8.96 (d, 2H), 8.72 (m, 1H), 8.22 (m, 2H), 6.44 (s, 1H), 6.40 (m, 1H), 6.24 (m, 1H), 6.17 (m, 2H), 5.87 (m, 1H), 5.79 (m, 1H), 5.48 (m, 1H), 5.29 (s, 1H), 4.95 (m, 2H), 4.81 (s, 1H), 4.52 (m, 3H).

    Example 17: Preparation of 31-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin (Compound 17)



    [0079] Preparation method was referred to Example 3 (n=1, R3=Br, R4=Br, R5= 4-methylpyridine).
    m.p. 145-148°C; MS:1048.1 [M-Br]+; 1H-NMR (600MHz, DMSO-d6 δ ppm), 8.77 (d, 2H), 8.03 (d, 2H), 6.44 (s, 1H), 6.40 (m, 1H), 6.23 (m, 1H), 6.16 (m, 2H), 5.80 (d, 1H), 5.70 (d, 1H), 5.44 (m, 1H), 5.29 (m, 1H), 5.29 (s, 1H).

    Example 18: Solubility test



    [0080] The Compounds 1-17 of the present invention were taken, and separately tested according to the Solubility Test Method of the Part II of Chinese Pharmacopoeia, 2010 Edition. The results are shown in Table 2:
    Table 2: Water solubility of Compounds 1-17
    Compound No.

    Water solubility (mg/ml)Comparisont o water solubility of rapamycin
     R1R2
    Rapamycin H H 0.0026 1
    1



    34.5 13269
    2

    H 0.028 11
    3 H

    10 3846
    4



    100 38462
    5

    H 4.2 1615
    6 H

    23.3 8962
    7



    85 32692
    8



    0.1 38
    9



    47 18077
    10

    H 0.025 10
    11

    H 6.7 2577
    12

    H 0.01 4
    13

    H 6 2308
    14



    120 46154
    15 H

    26.7 10269
    16 H

    52.6 20231
    17 H

    62.5 24038


    [0081] It can be seen from data of Table 2 that the compounds of the present invention have water solubility significantly higher than that of rapamycin, and even by 3 to 4 orders of magnitudes for some of these compounds.

    Example 19: Experimental evaluation of toxicity on rat primary hepatocytes



    [0082] Experimental method: Liver lobes of SD rat were placed in sterilized plate, added with a suitable amount of liver cell cleaning solution (precooled at 4°C), the liver was shredded and fibrous connective tissues were removed to form a liver cell suspension, screened with 200 mesh screen and placed in 50ml centrifuge tube, centrifuged at 500rpm for 1-2 min; the supernatant was removed, the precipitate was added with 20-30ml of RPMI 1640 (precooled at 4°C) and washed 3 times; isovolumetric liver cell suspension was taken and suspended in Percoll separating solution I, mixed by turning upside down, centrifuged at4°Cand 800rpm for 10min, the supernatant was discarded, the liver cells were washed with PBS once (800rpm, 5min). The liver cell precipitate was added into liver cell culture fluid, resuspended and diluted to 10ml, and a suitable amount thereof was taken for counting and survival rate was determined by using 0.4% trypan blue. The liver cells with survival rate of 90% or more were inoculated in a density of 2x103 on 96-well plate, 100µl of RPMI 1640 culture medium was added to each well, culture was performed at 37°C, 5% CO2 for 24h. The compounds were diluted with DMSO to desired concentrations and added to each well in an amount of 1µl(final compound concentrations were usually initiated from 1000µM, 10x gradient dilution, 6 gradients), and blank control was added with 1µl of DMSO.

    [0083] After the cells were cultured at 37°C, 5% CO2 for 24h, 100µl of ATP detection reagent was added, incubated under shock for 15min. fluorescence values of wells were read by ELIASA. Cell survival rates for each example compounds were calculated, and then GI50 values (concentrations of added compounds at which growth of 50% cells was inhibited) were calculated. The results were shown in Table 4. Obviously, the toxicities of the compounds were significantly less than that of rapamycin.
    Table 4: GI50 values of some example compounds on rat primary hepatocytes
    CompoundGI50 (µM)
    Rapamycin 24.822
    Compound of Example 11 71.753
    Compound of Example 13 44.936
    Compound of Example 17 54.311


    [0084] The data of Table 4 show that the compounds of the present invention have toxicities significantly less than rapamycin.

    Example 20: Experiment of inhibition activities of the compounds to mTORC1 and mTORC2



    [0085] Molecular mechanism of action of the example compounds in tumor cell A549.

    [0086] Experimental materials: DMEM high-glucose cell culture media (Hyclone Company), fetal calf serum (FBS) (Gibco Company), penicillin and streptomycin were purchased from North China Pharmaceutical Co.,Ltd, phosphate buffer saline (PBS) was purchased from Gibco Company, pancreatic enzyme and dimethylsulfoxide (DMSO) were products of Sigma Company. A549 cell line (human lung adenocarcinoma cell line) was purchased from ATCC. Mouse anti-human p-p70S6K(T389) monoclonal antibody was purchased from Cell Signaling Technology Company, mouse anti-human p70S6Kmonoclonal antibody was purchased from Cell Signaling Technology Company, mouse anti-human p-AKT (S473)monoclonal antibody was purchased from Cell Signaling Technology Company, mouse anti-human AKTmonoclonal antibody was purchased from Cell Signaling Technology Company. Horseradish peroxidase-labeled goat anti-mouse, goat anti-mouse monoclonal antibodies were purchased from Cell Signaling Technology Company. Cell lysis solution was purchased from Beijing Solarbio Science and Technology Co., Ltd., loading buffer was purchased from Biyuntian Company, SDS electrophoretic buffer solution was purchased from Biyuntian Company, Towbin transfer buffer was purchased from Biyuntian Company, TBS was purchased from Biyuntian Company, TBST was purchased from Biyuntian Company. ECL chemiluminescence solution was purchased from Beijing Pulilai Genetic Technology Co., Ltd., developing solution and fixing solution were purchased from the Tenth Chemical Plant of Shijiazhuang, NC membrane was purchased from Whatman Company, photographic film was purchased from Carestream (Xiamen) Medical Equipment Co., Ltd., skimmed milk powder was purchased from Beijing Xikai Creative Technology Co., Ltd.

    Experimental method:



    [0087] A549 cells were spread on a 96-well plate, the cells grew to density of 80%-90% and then cultured overnight under serum deprivation condition, co-incubated on the second day with 167nM insulin and the compounds for 2 hours, then the cells were lysed and the phosphorylation levels for the example compounds at Thr389 site of S6K1 (corresponding to mTORC1) and Ser473 site of Akt (corresponding to mTORC2) were detected by Western Blot method, and which were used to semi-quantitatively reflect the inhibition levels of the compounds to mTORC1 and mTORC2 (in the bands of the results, absence of blot represented expression of corresponding protein was inhibited, while presence of blot represented the expression was not inhibited). Positive control compound was Rapamycin. The results of evaluation are shown in Fig.1 (Fig.1A and Fig.1B). Specific steps were as follows:

    1. Treatment of cells:



    [0088] 
    1. 1)the cells were inoculated on 6-well plate, and the cells spread 80%-90% after 24h;
    2. 2)the cells were washed with PBS once, then 2 mL serum-free culture media was used for replacement and the culture was performed overnight under serum deprivation condition;
    3. 3) to the treated cells, 2µL of cell culture fluid containing the compound to be tested (20µM) and insulin (167nM) was added and co-incubation was performed for 2 h.

    2. Lysis of cells:



    [0089] 
    1. 1)the cells on the 6-well plate were washed off with 10ml PBS, centrifuged at 1500rpm for 5min;
    2. 2) the supernatant was discarded then 60µl cell lysis buffer (containing protease inhibitor) was added, the cells were lysed on ice for 20-30min, after 10min, oscillated with oscillator for 2-3s, and put back on ice;
    3. 3) oscillation was performed again for 3s before centrifugation, then centrifuged at 4°C, 13000rpm, for 10 min;
    4. 4)2×loading buffer 60µl was added, subjected to boiling water bath for 5min, and cryopreservated at -20°C or -80°C for standby use;

    3. Western blotting analysis:



    [0090] 
    1. 1)SDS-PAGE, 80 V voltage was firstly used for running to spacer gel, then 120 V voltage was used for running separation gel;
    2. 2) transfer, wet transfer, in ice-bath, 250 mA, 150 min;
    3. 3) 5% milk was used for sealing at room temperature for 1-2h (or overnight at 4°C);
    4. 4) primary antibody was added (diluted with TBS), stood overnight at 4°C;
    5. 5)TBST washing 3×10 min.
    6. 6) secondary antibody was added (1: 2000 diluted with 3% skimmed milk powder in TBS), incubation was performed at room temperature at sealing film for 2 hours;
    7. 7)TBST washing 3×10 min.
    8. 8)ECL chemiluminescence developing solutions A, B, each in 500µl, were mixed in a plate dish, and washed for 5 min;
    9. 9) the film was placed in exposure clamp, exposed in darkroom for a certain time, then placed in developing solution for 2 min, in fixing solution for 5min, the film was washed with tap water, and dried by baking;
    10. 10) pictures were taken and kept.


    [0091] The results showed that the compounds of the present invention could effectively inhibit mTORC1, and their inhibition effects are not inferior to rapamycin (rapamycin could only inhibit mTORC1, so that our compounds had activities not inferior to rapamycin).

    Example 21: Experiment of tumor cell inhibition



    [0092] The activities of example compounds to tumor cell A549 were evaluated.

    [0093] Experimental materials: DMEM high-glucose cell culture media (Hyclone Company), fetal calf serum (FBS) (Gibco Company), penicillin and streptomycin were purchased from North China Pharmaceutical Co.,Ltd, phosphate buffer saline (PBS) was purchased from Gibco Company, Cell Titer-Glo® cell viability detection reagent was purchased from Promega Company, pancreatin and dimethylsulfoxide (DMSO) were products of Sigma Company. A549 cell line (human lung adenocarcinoma cell line) was purchased from ATCC.

    Experimental method:



    [0094] The cells were inoculated on a 96-well plate with white wall and penetrative bottom (Costar) in amount of 5000 cells per well, cultured at condition of 37°C, 5%CO2 for 24h. The compounds to be tested were dissolved with DMSO and diluted to 100mM, to obtain mother liquids of the compounds.

    [0095] DMEM culture solution containing 2% FBS was used to diluted the compounds, in which concentration gradients were 3, and concentration range was 100µM to 3nM. The compounds in various dilution degrees were added to the cultured cells on the 96-well plate, 100µl per well. Culture was performed at 37°C, CO2 for 72h, supernatant was discarded, and cell viability detection test was performed.

    [0096] AfterCell Titer-Glo® reaction buffer solution was mixed with isometric substrate, the mixture was added to 96-well plate, 100µl per well. Cell lysis was induced by horizontally shaking for 4min. Reaction signal was stabilized by balancing at room temperature for 15min. Chemiluminescence units in each well of the 96-well plate were detected by using chemiluminescence detector.

    [0097] Inhibition rates at different dilution degrees of each compound were calculated according to chemiluminescence value of each well, and Origin 8.0 software was used for S-type curve fitting of each compound, and for calculating EC50 value. The results were shown in Table 5.
    Table 5: EC50 values of concentrations of some example compounds for tumor cell A549
    CompoundIC50 (µM)
    rapamycin 49.35
    Compound of Example 1 17.39
    Compound of Example 2 19.31
    Compound of Example 3 3.04
    Compound of Example 4 6.23
    Compound of Example 5 7.40
    Compound of Example 6 52.26
    Compound of Example 7 46.68
    Compound of Example 8 45.98
    Compound of Example 9 25.12
    Compound of Example 10 66.32
    Compound of Example 11 52.80
    Compound of Example 12 51.48
    Compound of Example 13 44.80
    Compound of Example 14 44.0
    Compound of Example 15 52.70
    Compound of Example 16 57.71
    Compound of Example 17 40.80


    [0098] The results showed that the compound of the present invention could effectively inhibit tumor cells, and some compounds had effects even superior to rapamycin, and thus are promising in the manufacture of anti-tumor drugs.

    Example 22: Experiment of inhibition to drug-resistant tumor cell line



    [0099] The activity of example compounds to multi-drug resistant tumor cell line MES-SA was evaluated.

    [0100] Experimental materials: DMEM high-glucose cell culture media (Hyclone Company), fetal calf serum (FBS) (Gibco Company), penicillin and streptomycin were purchased from North China Pharmaceutical Co.,Ltd,phosphate buffer saline (PBS) was purchased from Gibco Company, Cell Titer-Glo® cell viability detection reagent was purchased from Promega Company, pancreatinand dimethylsulfoxide (DMSO) were products of Sigma Company.MES-SA cell line (human uterine sarcoma cell line) was purchased from ATCC. MRC5 cell (human embryonic lung fibroblast) was purchased from ATCC.

    Experimental method:



    [0101] The cells were inoculated on a 96-well plate with white wall and penetrative bottom (Costar) in amount of 5000 cells per well, cultured at condition of 37°C, 5%CO2 for 24h. The compounds to be tested were dissolved with DMSO and diluted to 100mM, to obtain mother liquids of the compounds.

    [0102] DMEM culture solution containing 2% FBS was used to diluted the compounds, in which concentration gradients were 3, and concentration range was 100µM to 0.046µM. The compounds in various dilution degrees were added to the cultured cells on the 96-well plate, 100µl per well. Culture was performed at 37°C, CO2 for 72h, supernatant was discarded, and cell viability detection test was performed.

    [0103] AfterCell Titer-Glo® reaction buffer solution was mixed with isometric substrate, the mixture was added to 96-well plate, 100µl per well. Cell lysis was induced by horizontally shaking for 4min. Reaction signal was stabilized by balancing at room temperature for 15min. Chemiluminescence units in each well of the 96-well plate were detected by using chemiluminescence detector.

    [0104] Inhibition rates at different dilution degrees of each compound were calculated according to chemiluminescence value of each well, and Origin 8.0 software was used for S-type curve fitting of each compound, and for calculating EC50 value. The ratio of EC50 value of compound for MRC5 cell to EC50 value of compound for MES-SA cell was used as therapeutic index for the compound.The experimental results were shown in Table 6.
    Table 6: EC50 values and therapeutic indexes of some example compounds for tumor cell MES-SA
    CompoundIC50 (µM)Therapeutic index
    rapamycin <0.046 >635.09
    Compound of Example 1 <0.046 >259.8
    Compound of Example 2 <0.046 >1811.9
    Compound of Example 3 <0.046 >1501.1
    Compound of Example 4 <0.046 >787.2
    Compound of Example 5 <0.046 >1371.1
    Compound of Example 6 <0.046 >455.1
    Compound of Example 7 <0.046 >442.7
    Compound of Example 8 <0.046 >1441.6
    Compound of Example 9 7.21 12.1
    Compound of Example 10 10.93 3.4
    Compound of Example 11 <0.046 >1888.7
    Compound of Example 12 <0.046 >146.5
    Compound of Example 13 <0.046 >1559.0
    Compound of Example 14 <0.046 >2136.8
    Compound of Example 15 <0.046 >331.5
    Compound of Example 16 <0.046 >364.8
    Compound of Example 17 <0.046 >317.7


    [0105] The results showed that the compounds of the present invention could effectively inhibit proliferation of multi-drug resistant tumor cell line MES-SA, and their inhibition activities to normal cells were generally lower than their inhibition activities to MES-SA, so that they had good therapeutic indexes and good safety. Some compounds had effects superior to rapamycin, and were promising for making or acting as anti-tumor drugs.

    [0106] Although specific models for carrying out the invention were described in details, those skilled in the art would understand that these details can be modified and changed according to the teachings of disclosures, and all these changes fall into the protection scope of the present invention. The whole protection scope of the present invention is given by the appended claims.


    Claims

    1. A compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof:

    wherein,
    R1 and R2 are independently selected from H, A and B, and R1, R2 cannot be H simultaneously;

    wherein, in Formula A or Formula B,

    arrows refer to sites where A or B links to mother ring of Formula I;

    n independently is 1, 2, 3, 4, 5, 6 or 7;

    R4 is independently selected from fluorine, chlorine, bromine, iodine, nitro, and cyano;

    X1, X2, X3, X4, Y1, Y2, Y3, Y4, and Y5 are independently selected from C, S, O, N and Se atoms;

    X1∼X2, X2∼X3, X3∼X4, Y1∼Y2, Y2∼Y3, Y3∼Y4, Y4∼Y5 are independently single bond or double bond;

    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 are independently selected from hydrogen atom, hydroxy, aldehyde group, carboxyl, amino, cyano, halogen,C1-C6 alkyl, C3-C10 cycloalkyl, C1-C6 alkoxy, C1-C6 alkylthiol, C3-C10 cycloalkoxy, C1-C6 alkenyl, eneynylheterocyclic ring, heterocycloalkyl, substituted heterocycloalkyl, aromatic ring, aromatic heterocyclic ring, benzo-aromatic heterocyclic ring, wherein the C1-C6 alkyl, aromatic ring, aromatic heterocyclic ring, benzo-aromatic heterocyclic ring are not substituted or substituted with 1, 2, 3, 4 or 5 substituents independently selected from the following groups: -F, -Cl,-Br, -I, nitro, hydroxy, amino, cyano, C1-C6 alkylthiol, C1-C6 alkyl, C1-C6 alkenyl, C1-C6 alkynyl and C1-C6 alkoxy;

    wherein in Formula A, the N atom together with X1, X2, X3, X4 forms a thiazole ring; and

    wherein in Formula B, the N atom together with Y1, Y2, Y3, Y4, Y5 forms a pyridine ring.


     
    2. The compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof according to claim 1, wherein:
    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 are independently selected from hydrogen atom, hydroxy, aldehyde group, carboxyl, amino, cyano, halogen, C1-C3 alkyl, C3-C6 cycloalkyl, C1-C3 alkoxy, C1-C3 alkylthiol, C3-C6 cycloalkoxy and C1-C3 alkyl-enyl.
     
    3. The compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof according to any one of claims 1 to 2, wherein Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 are independently selected from hydrogen atom, hydroxy, and methyl.
     
    4. The compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof according to claim 1, wherein:
    R1 and R2 are independently selected from H, carbonylmethyl-(4-methyl-thiazole R4 salt-3-yl), carbonylmethyl-(4, 5-dimethyl-thiazole R4 salt-3-yl), carbonylmethyl-(pyridine R4 salt-1-yl), carbonylmethyl-(3-hydroxy-pyridine R4 salt-1-yl), carbonylmethyl-(3-methyl-pyridine R4 salt-1-yl) and carbonylmethyl-(4-methyl-pyridine R4 salt-1-yl), wherein R4 is independently selected from fluorine, chlorine, bromine, iodine, nitro and cyano; and R1, R2 cannot be H simultaneously.
     
    5. The compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof according to claim 1, wherein:
    R1 and R2 are independently selected from H, carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl), carbonylmethyl-(4, 5-dimethyl-thiazole bromide salt-3-yl), carbonylmethyl-(pyridine bromide salt-1-yl), carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl), carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl) and carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl), and R1, R2 are not H simultaneously.
     
    6. The compound of Formula I, or a pharmaceutically acceptable salt or hydrate thereof according to claim 1, wherein the compound is selected from the following compounds:

    31,42-bis-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin,

    42-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin,

    31-O-carbonylmethyl-(4-methyl-thiazole bromide salt-3-yl)-rapamycin,

    31,42-bis-O-carbonylmethyl-(4,5-dimethyl-thiazole bromide salt-3-yl)-rapamycin,

    42-O-carbonylmethyl-(4,5-dimethyl-thiazole bromide salt-3-yl)-rapamycin,

    31-O-carbonylmethyl-(4,5-dimethyl-thiazole bromide salt-3-yl)-rapamycin,

    31,42-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin,

    31,42-O-carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl)-rapamycin,

    31,42-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin,

    42-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin,

    42-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin,

    42-O-carbonylmethyl-(3-hydroxy-pyridine bromide salt-1-yl)-rapamycin,

    42-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin,

    31,42-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin,

    31-O-carbonylmethyl-(3-methyl-pyridine bromide salt-1-yl)-rapamycin,

    31-O-carbonylmethyl-(pyridine bromide salt-1-yl)-rapamycin, and

    31-O-carbonylmethyl-(4-methyl-pyridine bromide salt-1-yl)-rapamycin.


     
    7. A method for preparing the compound of Formula I according to any one of claims 1 to 6, which comprises steps of any one of the following processes (1) to (3):

    Process (1): Preparation of compounds of Formula I with position 31,42 bis-substituted

    Process (2): Preparation of compounds of Formula I with position 42 mono-substituted

    Process (3): Preparation of compounds of Formula I with position 31 mono-substituted

    wherein in the above methods (1) to (3),
    R3 is independently selected from F, Cl, Br and I;
    the other symbols are independently as defined in any one of claims 1 to 6.


     
    8. The method according to claim 7, wherein in the last reaction step of Process (1), Process (2) or Process (3), a reagent is added, and the reagent is independently selected from a thiazole ring and a pyridine ring.
     
    9. The method according to claim 8, wherein the reagent is selected from thiazole, pyridine, thiazole substituted with one or more C1-C6 alkyl, pyridine substituted with one or more C1-C6 alkyl, thiazole substituted with one or more halogen atoms, and pyridine substituted with one or more halogen atoms.
     
    10. The method according to claim 8, wherein the reagent is selected from 4-methylthiazole, 4,5-dimethylthiazole, pyridine, 3-hydroxypyridine, 3-methylpyridine and 4-methylpyridine.
     
    11. A pharmaceutical composition, which comprises the compound or a pharmaceutically acceptable salt or hydrate thereof of any one of claims 1 to 6, and optionally a pharmaceutically acceptable excipient.
     
    12. A coronary stent, comprising a drug coating comprising the compound or a pharmaceutically acceptable salt or hydrate thereof according to any one of claims 1 to 6, or the pharmaceutical composition according to claim 11.
     
    13. The compound or a pharmaceutically acceptable salt or hydrate thereof according to any one of claims 1 to 6, or the pharmaceutical composition of claim 11,for use as a medicament.
     
    14. The compound or a pharmaceutically acceptable salt or hydrate thereof according to any one of claims 1 to 6, or the pharmaceutical composition of claim 11, for use in suppressing immune, inhibiting mTOR, inhibiting mTORC1, inhibiting PI3K-Akt-mTOR signal pathway, inhibiting T lymphocyte proliferation, combating tumors, promoting tumor cell apoptosis, allowing cell cycle arrest at G1, reducing arterial embolism, combating aging, combating Alzheimer's disease, preventing organ rejection, combating inflammation or combating bacteria.
     
    15. The compound or a pharmaceutically acceptable salt or hydrate thereof according to any one of claims 1 to 6, or the pharmaceutical composition of claim 11, for used in treatment and/or prophylaxis and/or adjuvant therapy of kidney cancer, lymphoma, lung cancer, liver cancer, breast cancer, neuroendocrine cancer, uterine sarcoma or gastric cancer.
     


    Ansprüche

    1. Verbindung nach Formel I oder ein pharmazeutisch verträgliches Salz oder Hydrat davon:

    wobei,
    R1 und R2 unabhängig ausgewählt sind aus H, A und B, und R1, R2 nicht gleichzeitig H sein können;

    wobei sich, in Formel A oder Formel B,

    Pfeile auf Stellen beziehen, an denen A oder B an Mutter-Ring von Formel I bindet;

    n unabhängig 1, 2, 3, 4, 5, 6 oder 7 ist;

    R4 unabhängig ausgewählt ist aus Fluor, Chlor, Brom, Iod, Nitro und Cyano;

    X1, X2, X3, X4, Y1, Y2, Y3, Y4, und Y5 unabhängig ausgewählt sind aus C, S, O, N und Se Atomen;

    X1∼X2, X2∼X3, X3∼X4, Y1∼Y2, Y2∼Y3, Y3∼Y4, Y4∼Y5 unabhängig eine Einfachbindung oder eine Doppelbindung sind;

    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 unabhängig ausgewählt sind aus Wasserstoffatom, Hydroxy, Aldehydgruppe, Carboxyl, Amino, Cyano, Halogen, C1-C6 Alkyl, C3-C10 Cycloalkyl, C1-C6 Alkoxy, C1-C6 Akylthiol, C3-C10 Cycloalkoxy, C1-C6 Alkenyl, eneynylheterocyclischem Ring, Heterocycloalkyl, substituiertem Heterocycloalkyl, aromatischem Ring, aromatischem heterocyclischen Ring, benzoaromatischem heterocyclischen Ring, wobei das/der C1-C6 Alkyl, aromatische Ring, aromatische heterocyclische Ring, benzoaromatische heterocyclische Ring nicht substituiert oder substituiert sind mit 1, 2, 3, 4 oder 5 Substituenten, unabhängig ausgewählt aus den folgenden Gruppen: -F, -Cl, -Br, -I, Nitro, Hydroxy, Amino, Cyano, C1-C6 Alkylthiol, C1-C6 Alkyl, C1-C6 Alkenyl, C1-C6 Alkinyl und C1-C6 Alkoxy;

    wobei das N-Atom zusammen mit X1, X2, X3, X4 einen Thiazolring in Formel A bildet; und

    wobei das N-Atom zusammen mit Y1, Y2, Y3, Y4, Y5 einen Pyridin-Ring in Formel B bildet.


     
    2. Verbindung nach Formel I oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach Anspruch 1, wobei:
    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 unabhängig ausgewählt sind aus Wasserstoffatom, Hydroxy, Aldehydgruppe, Carboxyl, Amino, Cyano, Halogen, C1-C3 Alkyl, C3-C6 Cycloalkyl, C1-C3 Alkoxy, C1-C3 Alkylthiol, C3-C6 Cycloalkoxy und C1-C3 Alkylenyl.
     
    3. Verbindung nach Formel I oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach einem der Ansprüche 1 oder 2, wobei Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 unabhängig ausgewählt sind aus Wasserstoffatom, Hydroxy und Methyl.
     
    4. Verbindung nach Formel I oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach Anspruch 1, wobei:
    R1 und R2 unabhängig ausgewählt sind aus H, Carbonylmethyl-(4-methyl-thiazol R4 Salz-3-yl), Carbonylmethyl-(4, 5-dimethyl-thiazol R4 Salz-3-yl), Carbonylmethyl-(Pyridine R4 Salz-1-yl), Carbonylmethyl-(3-hydroxy-pyridin R4 Salz-1-yl), Carbonylmethyl-(3-methyl-pyridin R4 Salz-1-yl) und Carbonylmethyl-(4-methyl-pyridin R4 Salz-1-yl), wobei R4 unabhängig ausgewählt ist aus Fluor, Chlor, Brom, Iod, Nitro und Cyano; und R1, R2 nicht gleichzeitig H sein können.
     
    5. Verbindung nach Formel I oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach Anspruch 1, wobei:
    R1 und R2 unabhängig ausgewählt sind aus H, Carbonylmethyl-(4-methyl-thiazolbromid Salz-3-yl), Carbonylmethyl-(4, 5-dimethyl-thiazolbromid Salz-3-yl), Carbonylmethyl-(pyridinbromid Salz-1-yl), Carbonylmethyl-(3-hydroxy-pyridinbromid Salz-1-yl), Carbonylmethyl-(3-methyl-pyridinbromid Salz-1-yl) und Carbonylmethyl-(4-methyl-pyridinbromid Salz-1-yl), und R1, R2 nicht gleichzeitig H sind.
     
    6. Verbindung nach Formel I oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach Anspruch 1, wobei die Verbindung aus folgenden Verbindungen ausgewählt ist:

    31,42-Bis-O-carbonylmethyl-(4-methyl-thiazolbromid Salz-3-yl)-rapamycin,

    42-O-Carbonylmethyl-(4-methyl-thiazolbromid Salz-3-yl)-rapamycin,

    31-O-Carbonylmethyl-(4-methyl-thiazolbromid Salz-3-yl)-rapamycin,

    31, 42-Bis-O-carbonylmethyl-(4,5-dimethyl-thiazolbromid Salz-3-yl)-rapamycin,

    42-O-Carbonylmethyl-(4,5-dimethyl-thiazolbromid Salz-3-yl)-rapamycin,

    31-O-Carbonylmethyl-(4,5-dimethyl-thiazolbromid Salz-3-yl)-rapamycin,

    31,42-O-Carbonylmethyl-(pyridinbromid Salz-1-yl)-rapamycin,

    31,42-O-Carbonylmethyl-(3-hydroxy-pyridinbromid Salz-1-yl)-rapamycin,

    31,42-O-Carbonylmethyl-(3-methyl-pyridinbromid Salz-1-yl)-rapamycin,

    42-O-Carbonylmethyl-(pyridinbromid Salz-1-yl)-rapamycin,

    42-O-Carbonylmethyl-(3-methyl-pyridinbromid Salz-1-yl)-rapamycin,

    42-O-Carbonylmethyl-(3-hydroxy-pyridinbromid Salz-1-yl)-rapamycin,

    42-O-Carbonylmethyl-(4-methyl-pyridinbromid Salz-1-yl)-rapamycin,

    31,42-O-Carbonylmethyl-(4-methyl-pyridinbromid Salz-1-yl)-rapamycin,

    31-O-Carbonylmethyl-(3-methyl-pyridinbromid Salz-1-yl)-rapamycin,

    31-O-Carbonylmethyl-(pyridinbromid Salz-1-yl)-rapamycin, und

    31-O-Carbonylmethyl-(4-methyl-pyridinbromid Salz-1-yl)-rapamycin.


     
    7. Verfahren zur Herstellung der Verbindung von Formel I nach einem der Ansprüche 1 bis 6, welches die Schritte von einem der folgenden Prozesse (1) bis (3) umfasst:

    Prozess (1): Herstellung von Verbindungen der Formel I mit 31,42 bis-substituierten Stellen

    Prozess (2): Herstellung von Verbindungen der Formel 1 mit 42 mono-substituierter Stelle

    Prozess (3): Herstellung von Verbindungen der Formel I mit 31 mono-substituierter Stelle

    Wobei in den obigen Verfahren (1) bis (3)
    R3 unabhängig ausgewählt ist aus F, Cl, Br und I;
    die anderen Symbole unabhängig, wie in einem der Ansprüche 1 bis 6 definiert, sind.


     
    8. Verfahren nach Anspruch 7, wobei in dem letzten Reaktionsschritt von Prozess (1), Prozess (2) oder Prozess (3) ein Reagenz hinzugefügt wird und das Reagenz unabhängig ausgewählt ist aus einem Thiazolring und einem Pyridinring.
     
    9. Verfahren nach Anspruch 8, wobei das Reagenz ausgewählt ist aus Thiazol, Pyridin, Thiazol, das mit einem oder mehreren C1-C6 Alkyl substituiert ist, Pyridin, das mit einem oder mehreren C1-C6 Alkyl substituiert ist, Thiazol, das mit einem oder mehreren Halogenatomen substituiert ist und Pyridin, das mit einem oder mehreren Halogenatomen substituiert ist.
     
    10. Verfahren nach Anspruch 8, wobei das Reagenz ausgewählt ist aus 4-Methylthiazol, 4,5-Dimethylthiazol, Pyridin, 3-Hydroxypyridin, 3-Methylpyridin und 4-Methylpyridin.
     
    11. Pharmazeutische Zusammensetzung, die die Verbindung oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach einem der Ansprüche 1 bis 6 und gegebenenfalls einen pharmazeutisch verträglichen Hilfsstoff umfasst.
     
    12. Koronarer Stent, umfassend eine Arzneimittelbeschichtung, umfassend die Verbindung oder ein pharmazeutisch verträgliches Salz oder Hydrat davon nach einem der Ansprüche 1 bis 6 oder die pharmazeutische Zusammensetzung nach Anspruch 11.
     
    13. Verbindung oder pharmazeutisch verträgliches Salz oder Hydrat davon nach einem der Ansprüche 1 bis 6 oder die pharmazeutische Zusammensetzung nach Anspruch 11 zur Verwendung als ein Medikament.
     
    14. Verbindung oder pharmazeutisch verträgliches Salz oder Hydrat davon nach einem der Ansprüche 1 bis 6 oder die pharmazeutische Zusammensetzung nach Anspruch 11 zur Verwendung bei Unterdrückung des Immunsystems, Hemmung von mTOR, Hemmung von mTORC1, Hemmung des PI3K-Akt-mTOR-Signalweges, Hemmung der T-Lymphozyten-Proliferation, Bekämpfung von Tumoren, Förderung der Tumorzell-Apoptose, Ermöglichung des Zellzyklus-Stopps bei G1, Verringerung der arteriellen Embolie, Bekämpfung der Alterung, Bekämpfung der Alzheimer-Krankheit, Verhinderung der Organabstoßung, Bekämpfung von Entzündungen oder Bekämpfung von Bakterien.
     
    15. Verbindung oder pharmazeutisch verträgliches Salz oder Hydrat davon nach einem der Ansprüche 1 bis 6 oder die pharmazeutische Zusammensetzung nach Anspruch 11 zur Verwendung bei Behandlung und/oder Vorbeugung und/oder adjuvanter Therapie von Nierenkrebs, Lymphom, Lungenkrebs, Leberkrebs, Brustkrebs, neuroendokrinem Krebs, Uterussarkom oder Magenkrebs.
     


    Revendications

    1. Composé de formule I, ou sel ou hydrate pharmaceutiquement acceptable de celui-ci :

    dans lequel, R1 et R2 sont indépendamment choisis parmi H, A et B, et R1, R2 ne peuvent pas être H simultanément ;

    dans lequel, dans la formule A ou la formule B,

    les flèches se réfèrent à des sites où A ou B se lient au cycle de base de la formule I ; n vaut indépendamment 1, 2, 3, 4, 5, 6 ou 7 ;

    R4 est indépendamment choisi parmi le fluor, le chlore, le brome, l'iode, le nitro et le cyano ;

    X1, X2, X3, X4, Y1, Y2, Y3, Y4 et Y5 sont indépendamment choisis parmi les atomes C, S, O, N et Se ;

    X1∼X2, X2∼X3, X3∼X4, Y1∼Y2, Y2∼Y3, Y3∼Y4, Y4∼Y5 sont indépendamment une simple liaison ou une double liaison ;

    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 sont indépendamment choisis parmi un atome d'hydrogène, hydroxy, un groupe aldéhyde, carboxyle, amino, cyano, halogène, alkyle en C1 à C6, cycloalkyle en C3 à C10, alcoxy en C1 à C6, alkylthiol en C1 à C6, cycloalcoxy en C3 à C10, alcényle en C1 à C6, cycle éneynylhétérocyclique, hétérocycloalkyle, hétérocycloalkyle substitué, cycle aromatique, cycle hétérocyclique aromatique, cycle hétérocyclique benzo-aromatique, dans lequel les alkyles en C1 à C6, cycles aromatiques, cycles hétérocycliques aromatiques, cycles hétérocycliques benzo-aromatiques ne sont pas substitués ou substitués par 1, 2, 3, 4 ou 5 substituants indépendamment choisis parmi les groupes suivants : -F, -Cl, -Br, -I, nitro, hydroxy, amino, cyano, alkylthiol en C1 à C6, alkyle en C1 à C6, alcényle en C1 à C6, alcynyle en C1 à C6 et alcoxy en C1 à C6 ;

    dans lequel dans la formule A, l'atome N conjointement avec X1, X2, X3, X4 forme un cycle thiazole ; et

    dans lequel dans la formule B, l'atome N conjointement avec Y1, Y2, Y3, Y4, Y5 forme un cycle pyridine.


     
    2. Composé de formule I, ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon la revendication 1, dans lequel :
    Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 sont indépendamment choisis parmi un atome d'hydrogène, hydroxy, un groupe aldéhyde, carboxyle, amino, cyano, halogène, alkyle en C1 à C3, cycloalkyle en C3 à C6, alcoxy en C1 à C3, alkylthiol en C1 à C3, cycloalcoxy en C3 à C6 et alkyl-ényle en C1 à C3.
     
    3. Composé de formule I, ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon l'une quelconque des revendications 1 ou 2, dans lequel Z1, Z2, Z3, Z4, Z5, Z6, Z7, Z8, Z9 sont indépendamment choisis parmi un atome d'hydrogène, hydroxy et méthyle.
     
    4. Composé de formule I, ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon la revendication 1, dans lequel :
    R1 et R2 sont indépendamment choisis parmi H, carbonylméthyl-(3-yl-sel de R4 de 4-méthyl-thiazole), carbonylméthyl-(3-yl-sel de R4 de 4,5-diméthyl-thiazole), carbonylméthyl-(1-yl-sel de R4 de pyridine), carbonylméthyl-(1-yl-sel de R4 de 3-hydroxy-pyridine), carbonylméthyl-(1-yl-sel de R4 de 3-méthyl-pyridine) et carbonylméthyl-(1-yl-sel de R4 de 4-méthyl-pyridine), dans lequel R4 est indépendamment choisi parmi le fluor, le chlore, le brome, l'iode, le nitro et le cyano ; et R1, R2 ne peuvent pas être H simultanément.
     
    5. Composé de formule I, ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon la revendication 1, dans lequel :
    R1 et R2 sont indépendamment choisis parmi H, carbonylméthyl-(3-yl-sel de bromure de 4-méthyl-thiazole), carbonylméthyl- (3-yl-sel de bromure de 4,5-diméthyl-thiazole), carbonylméthyl-(1-yl-sel de bromure de pyridine), carbonylméthyl-(1-yl-sel de bromure de 3-hydroxy-pyridine), carbonylméthyl-(1-yl-sel de bromure de 3-méthyl-pyridine) et carbonylméthyl-(1-yl-sel de bromure de 4-méthyl-pyridine), et R1, R2 ne sont pas H simultanément.
     
    6. Composé de formule I, ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon la revendication 1, dans lequel le composé est choisi parmi les composés suivants :

    31,42-bis-O-carbonylméthyl-(3-yl-sel de bromure de 4-méthyl-thiazole)-rapamycine,

    42-O-carbonylméthyl-(3-yl-sel de bromure de 4-méthyl-thiazole)-rapamycine,

    31-O-carbonylméthyl-(3-yl-sel de bromure de 4-méthyl-thiazole)-rapamycine,

    31,42-bis-O-carbonylméthyl-(3-yl-sel de bromure de 4,5-diméthyl-thiazole)-rapamycine,

    42-O-carbonylméthyl-(3-yl-sel de bromure de 4,5-diméthyl-thiazole)-rapamycine,

    31-0 -carbonylméthyl-(3-yl-sel de bromure de 4,5-diméthyl-thiazole)-rapamycine,

    31,42-O-carbonylméthyl-(1-yl-sel de bromure de pyridine)-rapamycine,

    31,42-O-carbonylméthyl-(1-yl-sel de bromure de 3-hydroxy-pyridine)-rapamycine,

    31,42-O-carbonylméthyl-(1-yl-sel de bromure de 3-méthyl-pyridine)-rapamycine,

    42-O-carbonylméthyl-(1-yl-sel de bromure de pyridine)-rapamycine,

    42-O-carbonylméthyl-(1-yl-sel de bromure de 3-méthyl-pyridine)-rapamycine,

    42-O-carbonylméthyl-(1-yl-sel de bromure de 3-hydroxy-pyridine)-rapamycine,

    42-O-carbonylméthyl-(1-yl-sel de bromure de 4-méthyl-pyridine)-rapamycine,

    31,42-O-carbonylméthyl-(1-yl-sel de bromure de 4-méthyl-pyridine)-rapamycine,

    31- O-carbonylméthyl-(1-yl-sel de bromure de 3-méthyl-pyridine)-rapamycine,

    31-O-carbonylméthyl-(1-yl-sel de bromure de pyridine)-rapamycine et

    31-O-carbonylméthyl-(1-yl-sel de bromure de 4-méthyl-pyridine)-rapamycine.


     
    7. Procédé de préparation du composé de formule I selon l'une quelconque des revendications 1 à 6, qui comprend des étapes de l'un quelconque des processus (1) à (3) suivants :

    Processus (1) : préparation de composés de formule I bis-substitués à la position 31,42

    Processus (2) : préparation de composés de formule I monosubstitués à la position 42

    Processus (3) : préparation de composés de formule I monosubstitués à la position 31

    dans lequel dans les procédés (1) à (3) ci-dessus,
    R3 est choisi indépendamment parmi F, Cl, Br et I ;
    les autres symboles sont indépendamment tels que définis dans l'une quelconque des revendications 1 à 6.


     
    8. Procédé selon la revendication 7, dans lequel dans la dernière étape de réaction du processus (1), du processus (2) ou du processus (3), un réactif est ajouté, et le réactif est indépendamment choisi parmi un cycle thiazole et un cycle pyridine.
     
    9. Procédé selon la revendication 8, dans lequel le réactif est choisi parmi le thiazole, la pyridine, le thiazole substitué par un ou plusieurs alkyles en C1 à C6, la pyridine substituée par un ou plusieurs alkyles en C1 à C6, le thiazole substitué par un ou plusieurs atomes d'halogène, et la pyridine substituée par un ou plusieurs atomes d'halogène.
     
    10. Procédé selon la revendication 8, dans lequel le réactif est choisi parmi le 4-méthylthiazole, le 4,5-diméthylthiazole, la pyridine, la 3-hydroxypyridine, la 3-méthylpyridine et la 4-méthylpyridine.
     
    11. Composition pharmaceutique, qui comprend le composé ou un sel ou hydrate pharmaceutiquement acceptable de celui-ci de l'une quelconque des revendications 1 à 6, et facultativement un excipient pharmaceutiquement acceptable.
     
    12. Endoprothèse coronaire, comprenant une élution médicamenteuse comprenant le composé ou un sel ou hydrate pharmaceutiquement acceptable de celui-ci selon l'une quelconque des revendications 1 à 6, ou la composition pharmaceutique selon la revendication 11.
     
    13. Composé ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon l'une quelconque des revendications 1 à 6, ou composition pharmaceutique de la revendication 11, pour son utilisation comme médicament.
     
    14. Composé ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon l'une quelconque des revendications 1 à 6, ou composition pharmaceutique selon la revendication 11, pour son utilisation dans la suppression d'immunité, l'inhibition de mTOR, l'inhibition de mTORC1, l'inhibition de la voie de signalisation PI3K-Akt-mTOR, l'inhibition de la prolifération de lymphocytes T, la lutte contre les tumeurs, la promotion d'apoptose de cellules tumorales, l'autorisation de l'arrêt de cycle cellulaire en G1, la réduction d'une embolie artérielle, la lutte contre le vieillissement, la lutte contre la maladie d'Alzheimer, la prévention du rejet d'organe, la lutte contre une inflammation ou la lutte contre les bactéries.
     
    15. Composé ou sel ou hydrate pharmaceutiquement acceptable de celui-ci selon l'une quelconque des revendications 1 à 6, ou composition pharmaceutique selon la revendication 11, pour son utilisation dans le traitement et/ou prophylaxie et/ou le traitement complémentaire du cancer du rein, d'un lymphome, du cancer du poumon, du cancer du foie, du cancer du sein, d'un cancer neuroendocrinien, du sarcome utérin ou du cancer gastrique.
     




    Drawing








    Cited references

    REFERENCES CITED IN THE DESCRIPTION



    This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

    Non-patent literature cited in the description