The present invention relates to a pharmaceutical composition for inhibiting cancer cells, specifically to a pharmaceutical composition for inhibiting cancer cells through enhancing Glycine N-methyltransferase (GNMT) expression.
Hepatocellular carcinoma (HCC) is a global health burden with increasing incidence. Liver cancer is the fifth most common cancer worldwide and the third most common cause of cancer mortality. Because of the late diagnosis, poor treatment options and high recurrence, the incidence of HCC still nearly equals to its mortality rate. Treatment options for HCC are not a lot. Currently, the drugs that have been approved by the Food and Drug Administration (FDA) for HCC treatment are sorafenib and regorefenib. These drugs can only work in some individuals, but they have side effect and patients quickly develop drug resistance. Therefore, there is a high unmet need for HCC. The pace of new drug for HCC is very slow and many compounds failed in clinical trial. The developments of preventive agents, novel therapy or drug combination strategies are urgently needed to improve the patient's survival rate or help the patients to control disease.
GNMT is a tumor suppressor gene (TSG) for HCC. GNMT includes 1-3% of the cytosolic proteins in normal mammalian livers, while it is down regulated in more than 75% HCC patients. GNMT is one of the major enzymes in one-carbon metabolism pathway. It affects genetic stability by regulating the ratio of S-adenosylmethionine to S-adenosylhomocystine, and serves as the major folate-binding protein in hepatocytes. In addition, a previous study suggested that GNMT is an example of a moonlighting protein. GNMT is able to switch its function from acting as a metabolic enzyme to being a 4S polycyclic aromatic hydrocarbons (PAH) binding protein based on its phosphorylation state, which changes the subunit configuration. By changing the cellular localization of GNMT from the cytoplasm to the nucleus, GNMT is able to act as a transcription cofactor and regulate the expression of detoxification (and/or other target) genes.
Previously, it was found in a GNMT−/− mouse model that both male and female GNMT−/− mice developed chronic hepatitis at the age of 4-11 weeks. These GNMT−/− mice were followed up until 24 months old and the results showed that all female and half of male GNMT−/− mice developed HCC. By contrast, re-expression of GNMT inhibits cell proliferation in cancer cells. On top of that, transgenic mice overexpressing GNMT prevent the carcinogenicity and liver cancer progression induced by aflatoxin B1 (AFB1). More importantly, it was demonstrated that GNMT reduced the proliferation of Huh7 cells and sensitized them to rapamycin treatment both in vitro and in vivo. Recently, it was further demonstrated that the overexpression of GNMT sensitized Huh7 cells not only to rapamycin but also to sorafenib treatment both in vitro and in vivo. Therefore, considering the strong relevance between GNMT deficiency and liver disorders as well as the protective role of GNMT against hepatic carcinogenesis, the precise targeting of small molecule compounds that are able to enhance GNMT expression will have important implications for the treatment of HCC.
Preliminary results showed that a small molecule K78 is a potent lead for HCC treatment with the IC50 value of 2.5 μM in Huh7 cells. With the collaboration of the medicinal chemistry team, a comprehensive structure-activity relationship (SAR) study on K78 was performed. In this invention, K-series compounds (K78 derivatives) are synthesized to identify the abilities of K78 and its derivatives to induce a GNMT-promoter reporter activity.
In view of the deficiencies in the prior art, the Applicant have developed the present invention to overcome the deficiencies in the prior art. The summary of the present invention is described below.
The main object of the present invention is to provide novel GNMT enhancers for treating cancers via increasing GNMT expression in cancer cells.
Therefore, the present invention provides a nicotinohydrazide derivative, a stereoisomer thereof or a pharmaceutically acceptable salt thereof having a structure of formula (I):
wherein each of X, Y and Z is one of N and CH, at least one of X, Y and Z is CH, at least one of X, Y and Z is N, and R is one of OH and NH2.
In order to synthesize the preferred embodiment of compound having formula (I), the present invention provides a method for preparing a nicotinohydrazide derivative, including the steps of reacting an ethyl nicotinate with a hydrazine hydrate to form a carbohydrazide, and performing a condensation of the carbohydrazide with an acetylpyridine to form N′-(1-(pyridin-2-yl)ethylidene)-2-carbohydrazide (K117) having a structure below:
The present invention further provides a pharmaceutical composition including an effective amount of the nicotinohydrazide derivative, its stereoisomer or its pharmaceutically acceptable salt, wherein the nicotinohydrazide derivative has a structure of formula (I).
In a further aspect, the present invention provides a use of the above-mentioned nicotinohydrazide derivative, the stereoisomer thereof or the pharmaceutically acceptable salt thereof for preparing a drug in treating a cancer disease. The nicotinohydrazide derivative has a structure of formula (I), wherein the formula (I) is represented as
wherein X is one of N and CH, each of Y and Z is CH, and R is one of OH and NH2.
In accordance with an aspect of the present invention, a nicotinohydrazide derivative, a stereoisomer thereof or a pharmaceutically acceptable salt thereof is provided. In one preferred embodiment, the nicotinohydrazide derivative has a structure of formula (I)
wherein each of X, Y and Z is one of N and CH, at least one of X, Y and Z is CH, at least one of X, Y and Z is N, and R is one of OH and NH2.
The term “nicotinohydrazide derivative” used herein refers to the nicotinohydrazide derivatives, their stereoisomers or their pharmaceutically acceptable salts.
In another aspect of the present invention, a pharmaceutical composition including the nicotinohydrazide derivatives, their stereoisomers or their pharmaceutically acceptable salts is provided. The definitions of the nicotinohydrazide derivatives, their stereoisomers or their pharmaceutically acceptable salts are as described above. Preferably, the pharmaceutical composition further includes at least one of a pharmaceutically acceptable carrier and an excipient.
In a further aspect of the present invention, a method for preparing the preferred embodiment of the nicotinohydrazide derivatives is provided. The method includes the steps of: reacting ethyl nicotinate with hydrazine hydrate to form carbohydrazide, and performing a condensation of the carbohydrazide with acetylpyridine to form N′-(1-(pyridin-2-yl)ethylidene)-2-carbohydrazide (K117) having a structural formula below:
According to the present invention, the ethyl nicotinate is ethyl 2-aminonicotinate, the carbohydrazide is 2-carbohydrazide, and the acetylpyridine is 2-acetylpyridine.
In a further aspect, the present invention provides a pharmaceutical composition including an effective amount of a nicotinohydrazide derivative, its stereoisomer or its pharmaceutically acceptable salt. The nicotinohydrazide derivative has a structure of formula (I) as described above. Preferably, the pharmaceutical composition further includes at least one of a pharmaceutically acceptable carrier and an excipient.
In a further aspect, the present invention provides a use of the above-mentioned nicotinohydrazide derivative, the stereoisomer and the pharmaceutically acceptable salt thereof for preparing a drug in treating a cancer disease. The nicotinohydrazide derivative has a structure of formula (I), wherein the formula (I) is represented as
wherein X is one of N and CH, each of Y and Z is CH, and R is one of OH and NH2.
As used herein, the term “cancer” refers to a cellular disorder characterized by the uncontrolled or disordered cell proliferation, the decreased cellular differentiation, the inappropriate ability to invade surrounding tissue, and/or the ability to establish new growth at ectopic sites. The term “cancer” includes, but is not limited to, solid tumors and bloodborne tumors. The term “cancer” includes diseases of skin, tissues, organs, bone, cartilage, blood and blood vessels. The term “cancer” further includes primary and metastatic cancers. Non-limiting examples of cancer in the present invention include the liver cancer, the prostate cancer and the pancreatic cancer. Preferably, the liver cancer is hepatocellular carcinoma.
The pharmaceutical composition of the present invention is used for treating the cancers via enhancing GNMT expression in cancer cells.
Please refer to
In another aspect, different ethyl nicotinates are used to synthesize other K-series compounds. Ethyl 2-hydroxynicotinate, ethyl 3-aminoisonicotinate and ethyl 3-aminopyrazine-2-carboxylate respectively reacted with hydrazine hydrate to obtain their respective 2-carbohydrazides. Then, the respective 2-carbohydrazides respectively condensed with 2-acetylpyridine to obtain other compounds having the structure of formula (I). The structures of these compounds are represented as follows:
Human liver cancer cell lines (Huh7 and HepG2), human pancreatic cancer cell line (PANC1) and prostate cancer cell line (PC3) were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (HyClone, Logan, UT, USA), L-glutamine (2 mM), streptomycin (100 μg/mL), penicillin (100 U/mL) and nonessential amino acids (0.1 mM). Human pancreatic cancer cell line BxPC3 was cultured in the medium of Roswell Park Memorial Institute (RPMI) 1640 with the same supplements as in DMEM. All cell lines were maintained at 37ºC in a humidified CO2 incubator. H7GPL stable cells established by lentiviral-system was grown in DMEM supplemented with 1 μg/mL puromycin. The sorafenib were purchased from Sigma-Aldrich (St Louis, MO, USA).
A −1812/+14 GNMT promoter-luciferase reporter cell line H7GPL was constructed. For the primary analysis, H7GPL cells seeded in 96-well plates were treated with individual drugs of 10 μM for 24 hours and then were lysed for the luciferase activity assay using Luciferase Assay System (Promega, Madison, WI, USA). Then, the secondary analysis was performed using the compounds with different doses. The same confirmed hits were used for cytotoxicity by using alamarBlue® reagent (AbD serotec, Oxford, UK). The cytotoxicity was measured according to the manufacturer's recommendation. Top hits were used for further studies.
Huh7, HepG2, PANC1, PC3, and BxPC3 cells were seeded in 96-well plates for 24 hours and exposed to the drugs with various concentrations. AlamarBlue® was added in to the cells and further incubated for 4 hours to assess the cytotoxic effect of the tested drugs. Blank wells (without cells) containing the same amount of test substances were used for background subtraction, and the relative number of viable cells were represented as the percentage of the mean of the relative solvent controls. AlamarBlue® assay (AbD Serotec, Oxford, UK) was used to assess cell viability according to the manufacturer's instructions.
Cells or xenograft tumors were harvested for lysate preparation by using RIPA buffer containing protease inhibitor cocktail (Roche Molecular Biochemicals) and phosphatase inhibitors (10 mM Na3VO4, 1 mM NaF, 5 mM NaPPi). Protein concentrations were quantified by Bio-Rad Protein Assay (Bio-Rad) and immunoblotting was carried out as previous description (Yen et al., 2012). Anti-MYC (D84C12, Cell Signaling Technology), anti-GNMT (YMAC-Bioteck) and anti-β-actin (AC-15, Sigma-Aldrich) antibodies were used according to the descriptions in manufacturer's instructions.
6. Quantitative Real-Time PCR (qRT-PCR)
RNA was isolated by using Trizol reagent (Life Technologies) according to the manufacturer's protocol. Cellular RNA was reverse transcribed into complementary DNA using a SuperScript II RNase H-Reverse Transcriptase Kit (Life Technologies). PCR reactions were carried out using a Super Script II Reverse Transcriptase Kit (Invitrogen Inc., Carlsbad, CA, USA) on an Applied Biosystems Step-One-plus real-time PCR system (Applied Biosystems, Foster City, CA, USA). The mRNA level of TATA-box binding protein (TBP) was used as an internal control. The primer pairs used for qRT-PCR were:
Plasmids for lentivirus production (pCMV-ΔR8.91 and pMD.G), shRNAs for MYC (MYC-lentiviral shRNA, SEQ ID NO: 11) and control plasmid for the RNA interference (pLKO.1-shLacZ) were obtained from the National RNAi Core Facility (Academia Sinica, Taipei, Taiwan). The vector control plasmid and pcDNA-MYC plasmid for overexpression experiments were provided by Dr. Shih-Ping Liu (China Medical University, Taichung, Taiwan). The Renilla luciferase control reporter vector (Promega, Madison, WI, USA) was used for standardizing transfection efficiency. Plasmid DNAs were transfected by using TurboFect Reagent (Fermentas, Hanover, MD) and Lipofectamine 3000 (Life Technologies, Mulgrave, Australia) according to the manufacturers' recommendations.
All animal experiments were reviewed and approved by the Institutional Animal Care and Use Committee of Kaohsiung Medical University, and performed in accordance with relevant guidelines. Five to six-week-old male NOD-SCID mice were subcutaneously injected with Huh7 cells (1×106) in the right flank. Upon detection of palpable tumor, mice were randomly grouped and treated with 300 mpk, 100 mpk or 25 mpk of LKY4606D or KYT5397 and drug vehicle orally every day for 28 days. For K117, doses of 10 mpk and 25 mpk were used. For Sorafenib, dose of 25 mpk was used. Tumor growth was monitored (every day or three times per week) by measuring the length (L) and the width (W) of the tumor using Vernier caliper. Tumor volume (TV) was calculated by using the formula: volume=(L×W2)/2. All drugs were dissolved in 0.5% carboxymethyl cellulose sodium salt.
Statistical analyses were performed using MS excel and Graph Pad Prism 5.0 software. To compare various groups, P-values were determined by student's two-sided t-tests and considered to be statistically significant at P<0.05.
Preliminary studies showed that a naphthohydrazide derivative, K78, whose structure is shown in
To determine the therapeutic potential of K78 against HCC, the inhibitory effect of K78 on the growth of Huh7 cells was firstly evaluated. K78 suppressed the proliferation of Huh7 cells in a dose-dependent manner after 72 hours of treatment (
By performing structure-activity relationship (SAR) study, we identified more potent HCC inhibiting drugs with ability to enhance GNMT expression. K117 and K102 (K78 derivatives) were more effective than K78 in inducing GNMT expression. The structure of K102 is shown in
By performing structure-activity relationship (SAR) study, as shown in Table 1, K-series compounds (GNMT inducers) were synthesized by different ethyl nicotinates. Table 1 shows GNMT induction capacity of K-series compounds.
Next, we analyzed the effect of K117 on cell proliferation in different liver cancer cell lines (including Huh7, Hep3B, and HepG2). Results showed that K117 inhibited proliferation of all tested liver cancer cells in a dose-dependent manner (
As shown in
It has been reported that GNMT expression in normal prostatic and benign prostatic hyperplasia tissues was abundant whereas it was reduced in 82.2% of the prostate cancer tissues. This suggested that GNMT is a tumor susceptibility gene for the prostate cancer.
It is found in many cancers including pancreatic cancer that hypermethylation is utilized to silence gene. In such condition, GNMT downregulation may aid the availability of the methyl donor SAM to perform gene silencing. Previously, GNMT was found significantly down-regulated in the pancreatic cancer, and the secondary tumor from the pancreatic cancer often aim at liver. In this regard, it is similar to HCC that GNMT may be a TSG in the pancreatic cancer, and the re-activation of this down-regulated TSG in the pancreatic cancer would be a therapeutic regimen. Table 2 shows IC50 (μM) of the pancreatic and prostate cancer cell-lines. It indicates that the induction of GNMT inhibits the growth of cancer cells.
5. Mechanism of k78-Series GNMT Inducers in HCC
Next, the mechanism by which K78 enhances GNMT expression and inhibits liver cancer cell proliferation was explored in the present invention. It is known that the GNMT inducer drug 1,2,3,4,6-penta-O-galloyl-Beta-D-glucopyranoside (PGG) enhanced GNMT expression through MYC downregulation in the pathogenesis of liver cancer. Therefore, in order to determine whether K78-series compounds also exert their effect through MYC inhibition in HCC, the effects of K78-series compounds (K78, K90 and K102) treating on mRNA expression of MYC in Huh7 cells were examined in the present invention. The structure of K90 is shown in
The present invention may be modified in any way by those skilled in the art without departing from the scope of the appended claims.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/CN2020/139547 | 12/25/2020 | WO |