CN108976236B - Deuterated PARP inhibitor, salt thereof, preparation method and application thereof - Google Patents
Deuterated PARP inhibitor, salt thereof, preparation method and application thereof Download PDFInfo
- Publication number
- CN108976236B CN108976236B CN201810933066.4A CN201810933066A CN108976236B CN 108976236 B CN108976236 B CN 108976236B CN 201810933066 A CN201810933066 A CN 201810933066A CN 108976236 B CN108976236 B CN 108976236B
- Authority
- CN
- China
- Prior art keywords
- compound
- cancer
- deuterated
- drug
- deuterium
- 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.)
- Active
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D487/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
- C07D487/02—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
- C07D487/06—Peri-condensed systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/02—Antineoplastic agents specific for leukemia
Landscapes
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Hematology (AREA)
- Oncology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
The invention provides a deuterated compound, a pharmaceutically acceptable salt thereof, a preparation method and application. The compound is a novel deuterated PARP inhibitor, the invention also provides a composition containing the deuterated PARP inhibitor, and the application of the compound or the composition as an anti-cancer drug. Pharmacokinetic experiments show that the deuterated compound provided by the invention obviously improves the blood concentration, prolongs the half life of the medicament, and prolongs the time for which the medicament stays in vivo, thereby achieving better curative effect.
Description
Technical Field
The invention belongs to the field of medicinal chemistry, and particularly relates to a novel deuterated PARP inhibitor, a salt thereof, a preparation method thereof, a composition containing the deuterated PARP inhibitor, and application of the compound or the composition as an anti-cancer drug.
Background
Currently, many drugs are limited in their use due to problems with poor properties of absorption, distribution, metabolism or excretion (ADME). Meanwhile, it is also a main reason for the failure of clinical development of drugs. Although the ADME properties of drugs can be improved to some extent by using formulation techniques and prodrug techniques, these approaches do not fundamentally alter the ADME properties of drugs. For example, the problem of fast metabolism is that the drug is not effective when entering the body due to fast metabolism, and is metabolized by the body, and even if the activity is higher, the drug cannot achieve the treatment effect. If the treatment effect is to be achieved, the dosage is increased to increase the blood concentration, so that the treatment cost is increased and more side effects are brought. Therefore, how to improve the metabolic stability of the drug by modifying or adjusting the structure, especially without affecting the activity, is an urgent problem to be solved.
Another limitation of ADME is that many drugs produce toxic metabolites in the body, which exposes the patient to toxic metabolites that are harmful to the body when administered.
Sometimes to modify this condition, a metabolic inhibitor may be introduced which is rapidly metabolized by the body, such as protease inhibitors useful in the treatment of HIV infection. The U.S. FDA recommends that ritonavir be used in combination with such drugs. Ritonavir is an inhibitor of the cytochrome P450 enzyme 3A4(CYP3A4), which is the major cause of metabolism (see Kempf, D.J.et. al., antibodies and chemotherapy,1997,41(3): 654-60). However, ritonavir not only causes side effects, but also increases the economic burden on HIV patients who would otherwise use "cocktail therapy", and increases in the amount of drug administered also decrease patient compliance. Similarly, the CYP2D6 inhibitor, quinidine, is used in combination with dextromethorphan to reduce the problem of rapid metabolism of dextromethorphan, but the side effects produced by quinidine significantly limit its therapeutic potential for use in combination with other drugs (see Wang, L et al, Clinical Pharmacology and Therapeutics,1994,56(6Pt 1): 659-67; or FDA's instructions for quinidine at its website www.accessdata.fda.gov).
One effective way to improve drug metabolism is to modify the drug with deuterium, the isotope of hydrogen.
Hydrogen has three isotopes: protium (a)1H, Hydrogen, titanium), deuterium (2H, Deuterium) and tritium (3H, Tritium). Wherein deuterium (2H or D) is one of the most widely used isotopes, which is hydrogen (C) occurring in nature1H protium), non-radioactive, was first discovered by urea in 1932 in water. The nucleus of deuterium consists of one seed and a proton, whereas hydrogen (protium) has only one proton. Deuterium is present in nature in an amount of about 0.015%, and a large amount of deuterium is currently separated from water in the form of deuterated water, and the content of deuterium can reach 99.9%. Deuterium-substituted water, also called deuterium oxide, is currently the most economical and readily available source of deuterium.
The deuterium-substituted drug is prepared by replacing hydrogen (H) in drug molecules with deuterium (D), wherein the substitution has little influence on the activity due to the slight difference between H and D, but because deuterium is heavier than hydrogen, the formed chemical bond is difficult to break, so that the deuterium-substituted drug has great influence on the drug metabolism, and particularly when the deuterium-substituted drug is positioned at a metabolic site, the drug substitution effect can be well improved, the side effect can be remarkably reduced, and the like. Research on deuterated drugs has been greatly developed since the reports on impurities from Science 1961 (Science 1961,133,102-104), among which companies such as Auspex, Concert, Deuteria/DeuterRx have achieved many good results on deuterated drugs. Dancing medications by Auspex: deuterated tetrabenazine (SD-809) changes metabolism through deuteration of an active site, so that the safety and the effectiveness of the medicament are improved, the probability of depression, drowsiness, insomnia and difficulty in sitting in clinical observation is very low, and due to the excellent clinical effect, Teva spends 32 billion of dollars in burdening the Teva. This event also makes the study of deuterated drugs a very interesting area (nat. rev. drug discov.2016,15, 219-) -221).
Deuterium isotopes and deuterated compounds thereof are widely applied in a plurality of research fields, the deuterated compounds can be used as internal standards for clinical drug analysis and can be used for researching pharmacokinetics, drug metabolic pathways and drug toxicology, and in recent years, the deuterated compounds can be developed as better drugs.
The method of deuteration is used for trying to reduce the metabolic rate of the drug, increase the half-life, or reduce the formation of harmful metabolites through deuteration. The increase of bond energy after deuteration can improve ADME property of the medicine, thereby improving the efficacy, safety and compliance of the medicine. Meanwhile, as the size and the shape of the deuterium atom are basically consistent with those of the hydrogen atom, the selectivity and the biochemical activity of the medicine cannot be changed after the hydrogen is replaced by the deuterium.
Poly (ADP-ribose) transferase (PARP), which functions to catalyze the transfer of ADP-ribose from nicotinamide adenine dinucleotide to various receptor proteins and to repair single stranded DNA by means of base excision repair, is a novel target for cancer treatment today. In tumor cells, when PARP activity is inhibited, DNA damage repair is prone to error, and as DNA damage increases, tumor cells die, thereby achieving the goal of treating tumors. The current research shows that (Hanwe et al, research progress of PARP inhibitor for tumor treatment, China New medicine journal, 2011, 20(12), 1086-.
Ruipab (Invitrogen name: Rucaparib; chemical name: 8-fluoro-1, 3,4, 5-tetrahydro-2- [4- [ (methylamino) methyl ] phenyl ] -6H-pyrrolo [4,3,2-EF ] [2] benzazepin-6-one) is a poly (ADP-ribose) transferase inhibitor used for single drug treatment of patients with advanced ovarian cancer who have BRCA gene mutations and have been treated by two or more chemotherapy regimens.
Currently, research on Riclpa is mainly focused on itself, such as intermediate preparation process research (e.g., Gunn, et al, synthetic research of poly (adenosine diphosphate ribose) polymerase inhibitor Riclpa, fine chemical intermediates, 2012, 42(5), 48-52) and pharmacological research (e.g., PARP and CHK inhibitors intercation to cause DNA damageand cell death in mammary cells, Cancer Biology & Therapy (2013),14(5), 458-465).
However, research on the analogs of Riclpabu is still poorly explored. Therefore, it is very important to develop a new drug for effectively treating cancer based on the structure of the Ruipab.
Disclosure of Invention
The invention aims to provide a compound for treating tumors and application thereof.
The present invention provides a compound as shown below or a pharmaceutically acceptable salt thereof:
wherein, X1、X2、X3、X4、X5Each independently selected from H or D, and at least one selected from D.
Further preferably, X1=X2。
Further preferably, X1、X2When all are hydrogen, X3、X4、X5All of the components are deuterium, and the total amount of deuterium,
further preferably, X1、X2When all are hydrogen, X3、X4、X5Any two are deuterium and the other is hydrogen.
Further preferably, X1、X2When all are hydrogen, X3、X4、X5One is deuterium and the other two are hydrogen.
Further preferably, X1、X2When all are deuterium, X3、X4、X5All are hydrogen, or two are hydrogen, one is deuterium, or one is hydrogen, two are deuterium, or all are deuterium.
Further preferably the compound is one of the following compounds:
or a pharmaceutically acceptable salt thereof.
The document Organic Process Research & Development,2012,16(12): 1897-,
the compound of the present invention is prepared by the process shown in FIG. 2,
wherein, the compound 1 can be prepared according to the document Organic Process Research & Development,2012,16(12): 1897-1904.
among them, 2a can be prepared according to the Journal of the American Chemical Society (2000),122(14),3358-3366, and 2b can be commercially available.
among them, 4a can be prepared according to WO2011113369, 4b can be prepared according to the literature Synthesis (1971), (12), 654-.
The invention also provides the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a PARP inhibitor.
The invention also provides a pharmaceutical composition.
A pharmaceutical composition is a preparation prepared by taking the compound or the pharmaceutically acceptable salt thereof as an active ingredient and adding pharmaceutically common auxiliary materials or auxiliary ingredients.
On the basis of maintaining good antitumor proliferation of the original compound, the compound shown in the formula (I) is modified by deuteration, so that the pharmacokinetic property of the compound in blood plasma is better, the peak concentration of the blood drug is high, the effective blood drug concentration is maintained for a long time, the administration dosage can be reduced, the problem of poor metabolism of the drug can be further solved, and the drug toxicity and other side effects can be reduced.
Drawings
FIG. 1 is a scheme for the preparation of Rucaparib.
FIG. 2 is a scheme for the synthesis of compounds.
Figure 3 is a scheme for the synthesis of compound 101.
Figure 4 is a scheme for the synthesis of compound 102.
Figure 5 is a scheme for the synthesis of compound 103.
Figure 6 is a scheme for the synthesis of compound 104.
Figure 7 is a scheme for the synthesis of compound 201.
Figure 8 is a scheme for the synthesis of compound 202.
Figure 9 is a scheme for the synthesis of compound 202.
Detailed Description
In the present invention, the abbreviations or English-denoted Chinese names are as follows:
Pd(dppf)2Cl2[1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride
DMAC N, N-Dimethylacetamide
MeOH methanol
Na2CO3Sodium carbonate
THF tetrahydrofuran
DCM dichloromethane
HCl hydrogen chloride
NaOH sodium hydroxide
DMSO-d6 hexa-deuterated dimethyl sulfoxide
NaBH4Sodium borohydride
1H NMR hydrogen nuclear magnetic resonance spectrum
ESI/MS electrospray ionization liquid chromatography mass spectrometry
Example 1
The synthesis of 101 is shown in figure 3,
step 1: synthesis of Compound 3
Compound 1(2.8g, 100mmol) was added to DMAC (30ml), followed by addition of Pd (dppf)2Cl2Dichloromethane complex (0.002g), stirred at room temperature for 30min, heated to 95 deg.C and incubated for 1h, then cooled to room temperature, and Compound 2b (1.5g,100mmol) and Na were added2CO3(2.1g,200mmol) and water (10ml) were heated to 90 ℃ and stirred for 4h, the reaction was monitored by dot plate for completion, cooled to room temperature, water (150ml) was added, the solid precipitated, filtered, the filter cake washed with water, then slurried with hot MeOH, cooled to room temperature and filtered to give compound 3 as a pale green solid (2.77g, 90% yield).
H(400MHz,DMSO-d6)3.11(s,br,2H),3.42(s,br,2H),7.38(d,1H)7.47(d,1H),7.87(d,2H),8.06(d,2H),8.29(s,br,1H),10.06(s,br,1H)11.89(s,br,1H);
ESI/MS:m/z=309(M+H)+。
Step 2: synthesis of Compound 5
Adding compound 3(2.5g,8.1mmol) into a mixture of MeOH (20ml) and THF (10ml), adding 4c, stirring at room temperature for 2h, monitoring the completion of the reaction of compound 3 by a dot plate, cooling to 0-5 ℃, and then adding NaBH slowly in batches4(0.65g,17mmol), stirring reaction at below 10 ℃ for 2h, then raising the temperature to room temperature and stirring reaction for 2h, monitoring the reaction completion by a dot plate, slowly dropwise adding 1N diluted hydrochloric acid (15ml), adding activated carbon (5g), stirring for 6h, filtering, concentrating the filtrate, adding water and ethyl acetate for extraction and liquid separation, drying and concentrating the organic phase, recrystallizing the residue with ethanol, dissolving in DCM, introducing dry HCl gas, and filtering to obtain an off-white solid compound 5(2.4g, yield 83%).
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,2H),3.35-3.37(m,2H),4.13(d,br,2H),7.33(dd,1H),7.40(dd,1H),7.65(s,4H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=325(M+H)+。
And step 3: 101 Synthesis
Compound 5(2.2g,6.1mmol) was added to NaOH-H2O-MeOH (0.48g NaOH +15ml H)2O +5ml), stirred at room temperature for 2h, filtered, the filter cake washed with water and dried under vacuum to give compound 101 as an off-white solid (1.78g, yield 90%).
Example 2
The synthesis of 102 is shown in figure 4,
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,2H),3.35-3.37(m,1H),4.13(d,br,2H),7.33(dd,1H),7.40(dd,1H),7.65(s,4H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=326(M+H)+。
Example 3
The synthesis of 103 is shown in figure 5,
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,2H),4.13(d,br,2H),7.33(dd,1H),7.40(dd,1H),7.65(s,4H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=327(M+H)+。
Example 4
The composition of 104 is shown in figure 6,
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,3H),4.13(d,br,1H),7.65(s,3H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=329(M+H)+。
Example 5
The synthesis of 201 is shown in figure 7,
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,2H),4.13(d,br,1H),7.65(s,3H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=330(M+H)+。
Example 6
The synthesis of 202 is shown in figure 8,
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,2H),4.13(d,br,1H),7.65(s,3H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=331(M+H)+。
Example 7
The synthesis of 203 is shown in figure 9,
H(400MHz,DMSO-d6)2.53(t,br,3H),3.01-3.02(m,2H),7.65(s,3H),8.23(t,br,1H),9.38(s,br,1H),11.84(s,1H)。
ESI/MS:m/z=332(M+H)+。
Example 8
Pharmacokinetic testing
Pharmacokinetic experiments were performed on 102, 202 and Ricapab.
The tested animals were: male CD1 mice, weighing 18-22 g. The test animals should be adaptively raised at the test site 3-7 days before the test day, and male CD1 mice are randomly divided into 3 groups, and are respectively administered with gastric lavage, fasted for 12h before the test and freely drunk. The administration was followed by a uniform meal. The samples to be tested are administrated by a single gavage with a dose of 100mg/kg, and the samples to be tested of the experimental group and the positive control group are dissolved by Ethanol, PEG400 and water (5:45:50, v/v/v). Samples were taken at 0.5, 1.0, 2.0, 3.0, 5.0, 8.0, 10 and 24h post-dose, respectively; at each time point, 0.3mL of venous blood was collected from retrobulbar venous plexus of mice after animal anesthesia at the above set time points, placed in heparinized tubes, centrifuged at 11000g for 5min, plasma was separated, and frozen in a refrigerator at-20 ℃. During sample detection, after protein is precipitated from a plasma sample through methanol, the concentrations of the compounds 102 and 202 and the Ruipab in the plasma are determined by an LC-MS/MS method, and the linear range is 30.0-30000 ng/mL.
The WinNonlin6.3 software was used to calculate the major pharmacokinetic parameters (T) after gastric gavage in micemax,CmaxAUC, MRT and t 1/2). Wherein the peak concentration C is reachedmaxAnd time to peak TmaxIs an actual measurement value.
Area under plasma concentration-time curve AUC0-tThe value: and calculating by adopting a trapezoidal method.
AUC0-∞=AUC0-t+Ct/ke,
Ct is the blood concentration at the last measurable time point, ke is the elimination rate constant;
elimination of half-life t1/2=0.693/ke;
The mean residence time MRT is AUMC/AUC.
102. 202 and licarpb are shown in table 1.
Pharmacokinetic parameters of tables 1102, 202 and Ricapabu
From the experimental data in table 1, it can be seen that T of deuterated compounds 102 and 202 prepared by the inventionmaxT with RuicapabmaxSimilarly, deuterated compounds 102, 202 are shown to be absorbed in vivo similarly to Ricapab. However, C of 102, 202max1.31 times and 1.47 times of the benznidazole respectively; 102. 202 AUC0-tRespectively 1.34 times and 1.42 times of Ruipab; 102. 202 elimination half-life t1/2Is 1.28 times and 1.34 times of the benznidazole, which shows that the deuterated compounds 102 and 202 provided by the invention obviously improve the blood concentration, prolong the half-life period of the drug and prolong the time of the drug staying in the body compared with the Ruicapab, thereby achieving better curative effect. Meanwhile, compared with Ruicapa, the deuterated compound provided by the invention has smaller dosage, so that the problem of poor metabolism of the medicament can be further eliminated, and the toxicity and other side effects of the medicament can be reduced.
Example 9
Pharmaceutical composition
The phosphate salt of compound 202 was prepared according to conventional methods in the art.
Phosphate salt of Compound 202 10g
Starch 50g
Microcrystalline cellulose 45g
The materials are mixed evenly and put into common gelatin capsules according to the conventional method to prepare 1000 capsules.
Claims (5)
2. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to claim 1, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or diluent.
3. Use of a compound of claim 1 or a pharmaceutically acceptable salt thereof for the preparation of a PARP inhibitor.
4. The use of claim 3, wherein said PARP inhibitor is an anti-tumor drug.
5. Use according to claim 3 or 4, wherein the tumour is selected from: breast cancer, colon cancer, uterine cancer, pancreatic cancer, lung cancer, stomach cancer, blood cancer, lymph cancer, prostate cancer, liver cancer, cervical cancer, neuroblastoma, melanoma, solid tumor or intracranial tumor.
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201810933066.4A CN108976236B (en) | 2018-08-16 | 2018-08-16 | Deuterated PARP inhibitor, salt thereof, preparation method and application thereof |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201810933066.4A CN108976236B (en) | 2018-08-16 | 2018-08-16 | Deuterated PARP inhibitor, salt thereof, preparation method and application thereof |
Publications (2)
Publication Number | Publication Date |
---|---|
CN108976236A CN108976236A (en) | 2018-12-11 |
CN108976236B true CN108976236B (en) | 2020-11-10 |
Family
ID=64553732
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
CN201810933066.4A Active CN108976236B (en) | 2018-08-16 | 2018-08-16 | Deuterated PARP inhibitor, salt thereof, preparation method and application thereof |
Country Status (1)
Country | Link |
---|---|
CN (1) | CN108976236B (en) |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN109651377B (en) * | 2017-10-12 | 2020-10-20 | 成都海创药业有限公司 | Compound for treating cancer and application thereof |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN1342161A (en) * | 1999-01-11 | 2002-03-27 | 阿古龙制药公司 | Tricyclic inhibitors of poly(ADP-ribose) polymerases |
CN109651377A (en) * | 2017-10-12 | 2019-04-19 | 成都海创药业有限公司 | A kind of compound for the treatment of cancer and application thereof |
-
2018
- 2018-08-16 CN CN201810933066.4A patent/CN108976236B/en active Active
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN1342161A (en) * | 1999-01-11 | 2002-03-27 | 阿古龙制药公司 | Tricyclic inhibitors of poly(ADP-ribose) polymerases |
CN109651377A (en) * | 2017-10-12 | 2019-04-19 | 成都海创药业有限公司 | A kind of compound for the treatment of cancer and application thereof |
Non-Patent Citations (1)
Title |
---|
氘代作用在药物研究中的应用;江文峰 等;《齐鲁药事》;20101231;第29卷(第11期);第682-684页 * |
Also Published As
Publication number | Publication date |
---|---|
CN108976236A (en) | 2018-12-11 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2021129820A1 (en) | Spiro ring-containing quinazoline compound | |
CN115335379B (en) | Spirocyclic quinazoline compounds | |
TWI772291B (en) | Substituted pyrrolopyrimidine CDK inhibitors, pharmaceutical compositions containing the same and uses thereof | |
CN103788075B (en) | A kind of crystal form of thymidine phosphorylase inhibitor and preparation method thereof | |
CN116390728B (en) | Quinazoline derivative, preparation method and application thereof | |
CN111732575B (en) | N- (3- (pyrimidine-2-yl) phenyl) benzene sulfonamide derivative, pharmaceutical composition, preparation method and application | |
CN108976236B (en) | Deuterated PARP inhibitor, salt thereof, preparation method and application thereof | |
CN116283997A (en) | Deuterated HGFR inhibitor drug and application thereof | |
CN102617676B (en) | Zidovudine quinoline conjugated compound, preparation method thereof and application for liver cancer resistance | |
CN109651377B (en) | Compound for treating cancer and application thereof | |
TW201815793A (en) | Crystalline form of free alkali of imidazo isoindole derivative and a preparation method thereof | |
CN102688234A (en) | Synthesis of indolone derivative as RSK2 inhibitor and application | |
CN110054627A (en) | A kind of novel IDO inhibitor, preparation method, medical composition and its use | |
CN111303163B (en) | Compound with JAK kinase inhibitory activity, preparation method, composition and application | |
WO2022105542A1 (en) | Deuterated 1,4-benzodiazepine-2,5-dione compound and use thereof | |
CN105541810B (en) | Coumarin NEDD8 activating enzyme inhibitor and preparation method and application thereof | |
TW202104216A (en) | Crystal form of plk4 inhibitor | |
CN102688250A (en) | Synthesis and application of azo derivatives as inhibitor of RSK2 | |
KR100715358B1 (en) | 2-o-carboran-1-ylmethylpiperidine derivative, a process for the preparation thereof and a pharmaceutical composition comprising the same | |
CN104788372B (en) | A kind of deuterated card is rich to replace Buddhist nun's derivative, its preparation method, application and its intermediate | |
WO2022083733A1 (en) | Bruton tyrosine kinase inhibitor compound in solid form and use thereof | |
CN115433169B (en) | Preparation method of oxitinib mesylate dimer | |
CN108727404B (en) | Thieno [3,2-d ] pyrimidine compound, preparation method and application thereof | |
CN116514828A (en) | Water-soluble antitumor prodrug, and pharmaceutical composition and application thereof | |
WO2020057605A2 (en) | Glucose compound, pharmaceutical composition, 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 | ||
GR01 | Patent grant | ||
GR01 | Patent grant |