Multi-target nucleoside derivatives

文档序号:1471273 发布日期:2020-02-21 浏览:18次 中文

阅读说明:本技术 多靶标的核苷衍生物 (Multi-target nucleoside derivatives ) 是由 托马斯·I.·卡尔曼 于 2018-04-26 设计创作,主要内容包括:公开了多靶标的嘧啶核苷氨基甲酸酯,其在碱基部分和糖部分均取代有氟原子(参见式I),其中R是直链或支链的烷基(C<Sub>1-7</Sub>);R’是H、羟基保护基;R”是H、磷酸酯、氨基酸烷基(C<Sub>1-7</Sub>)酯氨基磷酸或二氨基磷酸。所公开的化合物是以提供细胞内代谢物的结构组分的新组合为特征的氟嘧啶前药,该细胞内代谢物能够1)抑制DNA合成和适当功能所需的几种细胞酶,和2)通过错误掺入至DNA而引起DNA损伤。通过以不同作用机理作用于多个靶标,式I化合物降低了产生耐药性的可能性,耐药性是使用基于核苷的抗癌药和抗病毒药的主要缺点。<Image he="240" wi="700" file="DDA0002337872580000011.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>(Disclosed are multi-target pyrimidine nucleoside carbamates substituted with a fluorine atom in both the base moiety and the sugar moiety (see formula I), wherein R is a linear or branched alkyl group (C) 1‑7 ) (ii) a R' is H, a hydroxy protecting group; r' is H, phosphate, amino acid alkyl (C) 1‑7 ) Ester phosphoramidates or phosphorodiamidites. The disclosed compounds are fluoropyrimidine prodrugs characterized by providing a novel combination of structural components of intracellular metabolites capable of 1) inhibiting DNA synthesis and several cellular enzymes required for proper function, and 2) causing DNA damage by misincorporation into DNA. By making a differenceThe mechanism of action acts on multiple targets and the compounds of formula I reduce the possibility of developing drug resistance, which is a major drawback of the use of nucleoside-based anticancer and antiviral drugs.)

1. A compound, or a pharmaceutically acceptable salt thereof, having the structure:

Figure FDA0002337872550000011

wherein R is selected from the group consisting of C1To C7Straight chain alkyl radical and C1To C7Branched alkyl groups; r' is selected from the group consisting of H and a hydroxy protecting group; r' is selected from H, phosphate and amino acid C1To C7Alkyl phosphoramidates and phosphorodiamidites.

2. The compound of claim 1, wherein the hydroxyl protecting group is selected from the group consisting of acetyl and benzoyl.

3. The compound of claim 1, wherein R "is a monophosphate, diphosphate, or triphosphate.

4. The compound of claim 1, wherein the compound has the structure:

Figure FDA0002337872550000012

5. a composition comprising a compound of claim 1 and a pharmaceutically acceptable carrier.

6. The composition of claim 5, wherein the compound is:

Figure FDA0002337872550000013

7. a method for treating an individual having cancer comprising administering to the individual the composition of claim 5, wherein administration of the composition results in inhibition of cancer growth in the individual.

8. The method of claim 7, wherein the subject is a human or non-human mammal.

9. The method of claim 7, wherein the cancer is selected from the group consisting of renal adenocarcinoma, myeloid leukemia, colon cancer, non-small cell lung cancer, metastatic malignant melanoma, breast adenocarcinoma, pancreatic adenocarcinoma, and ovarian cancer.

10. The method of claim 7, wherein DNA synthesis in cancer cells is inhibited.

11. The method of claim 7, wherein ribonucleotide reductase, DNA polymerase alpha, thymidylate synthase, DNA (cytosine-5) -methyltransferase, or a combination thereof is inhibited.

12. The method of claim 7, wherein the method induces misincorporation of the analog into the DNA, resulting in a change in the structure and function of the DNA, resulting in death of the cancer cell.

13. The method of claim 7, wherein the composition comprises at least one compound having the structure:

Figure FDA0002337872550000021

Technical Field

The present disclosure relates generally to nucleoside derivatives. More specifically, the disclosure relates to fluoropyrimidines useful for treating cancer and viral infections.

Background

As of 2013, 36 nucleoside or nucleotide analogs approved by the FDA are available for medical use; 11 of these are anti-Cancer drugs, and 25 are anti-Viral drugs (Jordheim, L.P., et al, "Advances in the Development of Nucleotide and Nucleotide analogs for Cancer and Viral Diseases," Naturereviews: Drug Discovery, 2013, No. 12, p. 447) 464). The therapeutic uses of these analogs stem from their ability to interfere with the replication and function of cellular or viral nucleic acids. Their mechanism of action includes inhibition of enzymes required for synthesis and replication of cellular or viral nucleic acids, and/or their ability to incorporate into nucleic acids.

Cancers and viruses are characterized by their ability to develop resistance to the drugs against which they are used in conventional therapies. This is particularly prevalent with nucleic acid component analogs (e.g., nucleobase, nucleoside, and nucleotide analogs) that act as antimetabolites. The practice of using different drug combinations to combat drug resistance is evolving, however, even combination chemotherapy does not eliminate the problem of drug resistance, as cross-resistance may also develop for multiple drugs. The increasing prevalence of these drug resistant cancers and viruses has necessitated the continued discovery and development of more effective new therapeutic agents.

5-Fluorouracil (FU), the prototype anticancer fluoropyrimidine, was an antimetabolite discovered in 1957 and is currently still used alone or in combination with other anticancer drugs or biological response modifiers, particularly for the treatment of colorectal and breast cancer. Major disadvantages of FU include insufficient oral bioavailability, severe toxic side effects and possibly fatal pharmacogenetic liability for patients lacking FU degrading enzymes. During the past sixty years, many prodrug derivatives of FU have been developed, as well as several nucleoside analogs and drug combinations to overcome toxic side effects, improve therapeutic effectiveness and therapeutic index. However, there remains an unmet need to develop novel fluoropyrimidine anticancer drugs that are orally available, which do not have major side effects such as gastrointestinal and bone marrow toxicity, hand-foot syndrome, and pharmacogenetic syndrome (dihydropyrimidine dehydrogenase deficiency (DPD deficiency)), and the like.

Fluoropyrimidine capecitabine

Figure BDA0002337872560000023

Is the only nucleoside carbamate approved by the FDA and is the most advanced fluoropyrimidine developed to date. It is a prodrug of 5-Fluorouracil (FU). Despite some of the disadvantages of FU, capecitabine can be administered orally, rendering the GI of fluoropyrimidine less toxic by activating mainly in the liver rather than during intestinal absorption. These advances are made by the N of the 5-fluorocytosine moiety of the nucleoside analog4Side chain formation of carbamate at the-position. However, hand-foot syndrome is often a dose-limiting toxicity of capecitabine, leading to significant morbidity. Thymidine phosphorylase is an enzyme used to convert capecitabine exclusively into cytotoxic FU, has significant activity in certain normal tissues (e.g., the palm of the hand) and may play a role in the etiology of hand-foot syndrome. In addition, genetic DPD deficiency (which may put the patient at risk of serious or fatal toxicity) is also associated with the obligate intermediate FU produced by thymidine phosphorylase in the activation of capecitabine metabolism.

There is a continuing and unmet need for nucleosides with fewer deleterious side effects.

Disclosure of Invention

The present disclosure provides novel multi-target pyrimidine nucleoside carbamates substituted with a fluorine atom at both the base moiety and the sugar moiety (see formula I):

Figure BDA0002337872560000021

wherein R is a linear or branched alkyl group (C)1-7) (ii) a R' is H or a hydroxy protecting group (e.g., aminoacyl or acyl)Such as, for example, acetyl or benzoyl); r' is H, phosphate, amino acid alkyl (C)1-7) Ester phosphoramidates or phosphorodiamidites.

The disclosed compounds are prodrugs characterized by a novel combination of structural components that provide intracellular metabolites of several cellular enzymes required for the inhibition of DNA synthesis and proper function.

In addition, the disclosed compounds can cause DNA damage through the misincorporation of analogs into DNA. This DNA damage can lead to the death of cancer cells by apoptosis. By acting on multiple targets with different mechanisms of action, the compounds of formula I reduce the possibility of developing resistance, which is a major drawback of the use of nucleoside-based anticancer and antiviral drugs. Thus, each of the disclosed compounds can achieve the effect of combination chemotherapy via administration of a single compound with a single pharmacokinetic profile. The desired feature of the disclosed compounds is that, unlike all other fluoropyrimidines, they cannot be metabolically converted to extremely toxic FU and therefore they do not have all the adverse effects of FU.

The present disclosure provides methods of preparing compounds of formula I. The process is based on a specific content of TMSOTf used as Lewis acid in the glycosylation reaction.

In one example, a method of making a compound of the present disclosure comprises: provides a composition comprising 5-fluoro-N- (trimethylsilyl) -2- ((trimethylsilyl) oxy) pyrimidin-4-amine

Figure BDA0002337872560000022

(2R,3R,5R) -3- (benzoyloxy) -4, 4-difluoro-5- ((methylsulfonyl) oxy) tetrahydrofuran-2-yl) methyl benzoateAnd a solvent (e.g., dichloroethane), adding 3 equivalents of TMSOTf to the reaction mixture to yield an α/β anomeric mixture of (2R,3R) -5- (4-amino-5-fluoro-2-oxopyrimidin-1 (2H) -yl) -2- ((benzoyloxy) methyl) -4, 4-difluorotetrahydrofuran-3-yl-benzoate in 100% yield using the methods disclosed herein,this product can be used to produce the compounds of the present disclosure.

The present disclosure provides compositions comprising one or more compounds of formula I and pharmaceutically acceptable salts thereof. The composition may comprise one or more pharmaceutically acceptable carriers.

The design principles for producing the compounds of the present disclosure can be summarized as follows. It is generally believed that Thymidine Phosphorylase (TP) activation of capecitabine, an advanced pre-fluoropyrimidine, is beneficial because certain cancers overexpress this Thymidine Phosphorylase (TP). Contrary to the prevailing opinion, it is hypothesized that it might be more advantageous to design a reduction in substrate activity of TP, since this would prevent the formation of FU that causes most toxic side effects, with a beneficial effect not only in those patients with TP-highly expressed cancers but also in all patients. It is believed that the placement of a fluorine atom (which is the most electronegative atom) at the 2 '-position of the analog disrupts the transition state of the TP reaction, which has a positive charge on the adjacent 1' -carbon (see Schwartz, P.A. et al, Journal of the American Chemical Society, 2010, Vol. 132, p. 14425). It is further believed that the addition of another fluorine atom produces a stronger effect. However, the involvement of eliminating TP would prevent metabolic activation of the analog, thus determining the addition of an-OH group at the 5' -position, allowing another pathway of kinase-mediated phosphorylation activation (see figure 2).

The present disclosure provides methods of using one or more compounds of the present disclosure. For example, the compounds may be used to treat cancer and/or viral infections.

Drawings

For a fuller understanding of the nature and objects of the present disclosure, reference should be made to the following detailed description taken together with the accompanying figures.

Figure 1 shows the primary targets of clinically used fluoropyrimidines (unique targets of compounds of formula I) compared to those of formula IItalic bodyShown).

The numbers in the figure correspond to (1) Thymidine Kinase (TK), EC 2.7.1.21, (2) thymidylate kinase (dTMPK), EC 2.7.4.9, (3) nucleoside diphosphate kinase (NDPK), EC2.7.4.6, (4) DNA polymerase α (Pol α), EC 2.7.7.7, (5) nucleoside triphosphatase (NTPase), EC 3.6.1.15, (6)5 '-nucleotidase (5' -NT), EC 3.1.3.5, (7) Cytidine Deaminase (CDA) EC 3.5.4.5, (8) deoxycytidylic acid (dCMP) deaminase (DCTD), EC 3.5.4.12, EC 2.7.4.14, (9) deoxycytidine kinase (dCK), EC 2.7.1.74, (10) UMP/CMP kinase (YMPK), (11) dUTP diphosphatase (dUTPase), EC 3.6.1.23, (12) dCMP kinase (DCMPK), EC 2.7.4.25; NDP/CMP kinase (YMPK), (11) dUTP diphosphatase (dUTP 3.6.1.6), and nucleoside diphosphate (NDMPK 3.6.1.6).

Detailed Description

While the claimed subject matter will be described in terms of certain embodiments, other embodiments, including embodiments that do not provide all of the benefits and features set forth herein, are also within the scope of the present disclosure. Various structural, logical, and process step changes may be made without departing from the scope of the disclosure.

The present disclosure provides novel multi-target pyrimidine nucleoside carbamates, substituted with a fluorine atom on both the base moiety and the sugar moiety (see formula I):

Figure BDA0002337872560000041

wherein R is a linear or branched alkyl group (C)1-7) (ii) a R' is H, a hydroxy protecting group; r' is H, phosphate, amino acid alkyl (C)1-7) Ester phosphoramidates or phosphorodiamidites. In one example, R "can be a monophosphate, diphosphate, or triphosphate.

Examples of hydroxy protecting groups are known in the art. For example, hydroxyl protecting groups may include aminoacyl and acyl groups. Non-limiting examples of acyl groups include acetyl and benzoyl.

The disclosed compounds are prodrugs characterized by providing novel combinations of structural components of intracellular metabolites that are capable of inhibiting several cellular enzymes required for DNA synthesis and proper function, and are capable of causing DNA damage by misincorporation into DNA. By acting on multiple targets with different mechanisms of action, the compounds of formula I reduce the possibility of developing resistance, which is a major drawback of the use of nucleoside-based anticancer and antiviral drugs. Thus, each of the disclosed compounds can achieve the effect of combination chemotherapy via administration of a single compound with a single pharmacokinetic profile. In drug combinations, it is difficult to optimize the differences between the individual ADME properties to achieve maximum benefit due to the different structures of the combined drugs. The desired feature of the disclosed compounds is that, unlike all other fluoropyrimidines, they cannot be metabolically converted to extremely toxic FU and therefore they do not have all the adverse effects of FU.

The compounds disclosed herein undergo prodrug activation pathways that differ from those in the prior art, which do not involve thymidine phosphorylase and do not produce FU (see fig. 2). Thus, in contrast to other fluoropyrimidines used clinically, the compounds of formula I are not expected to have toxic side effects in humans caused by FU.

One of the metabolites of the compound of formula I, F3dUrd (see FIG. 2), can be theoretically cleaved by thymidine phosphorylase to produce FU. However, at the time of the test, F was found3dUrd could not be used as a substrate for thymidine phosphorylase, confirming the initial hypothesis underlying the design of the compounds of formula I. As expected, floxuridine (FdUrd), which does not have two fluorine atoms at the 2' -position, was found to be rapidly cleaved by thymidine phosphorylase to produce FU.

The difference in the basic structure between the only nucleoside carbamate drug present, capecitabine, and representative member 6a of the compound of formula I having the same carbamate side chain that is most similar to capecitabine, is highlighted as follows:

there is a methyl group at the 5' -position of capecitabine. In contrast, 6a has a hydroxymethyl group at the 5' -position, which, unlike the methyl group in capecitabine, allows free nucleoside F after hydrolysis of the carbamate side chain and cellular uptake3Intracellular phosphorylation of dCyd to its monophosphate form (seeFig. 2).

At the 2' -position, there is a 2-point difference. Both the 2 '-hydroxyl and the 2' -hydrogen of capecitabine are absent in formula I. Instead, there are two fluorine atoms at the 2' -position of 6 a. These fluorine atoms stabilize the glycosyl bonds from cleavage and provide a chemical role for the inactivation of key enzymes (ribonucleotide reductase). In contrast, in the metabolic activation of capecitabine, cleavage of the glycosyl bond is an essential step, thus FU is formed, while the activity of ribonucleotide reductase is not affected.

The design principles for producing the compounds of the present disclosure can be summarized as follows. It is generally believed that Thymidine Phosphorylase (TP) activation of capecitabine, an advanced pre-fluoropyrimidine, is beneficial because certain cancers overexpress this Thymidine Phosphorylase (TP). Contrary to the prevailing view, it is hypothesized that it may be more advantageous to design a decrease in the substrate activity of TP, since this would prevent the formation of FU that causes most toxic side effects, with a beneficial effect not only in those patients with TP-highly expressed cancers but also in all patients. It is believed that the placement of a fluorine atom (which is the most electronegative atom) at the 2 '-position of the analog disrupts the transition state of the TP reaction, which has a positive charge on the adjacent 1' -carbon (see Schwartz, P.A. et al, Journal of American Chemical Society, 2010, volume 132, page 14425). It is further believed that the addition of another fluorine atom produces a stronger effect. However, the involvement of eliminating TP would prevent metabolic activation of the analog, thus determining the addition of an-OH group at the 5' -position, allowing another pathway of kinase-mediated phosphorylation activation (see figure 2).

The following scheme outlines the important steps in the metabolic activation pathway of capecitabine to produce FU:

Figure BDA0002337872560000052

carboxyesterases CES1 and CES2 remove the carbamate side chain by hydrolysis to form 5-fluoro-5 '-deoxycytidine, which is then converted to 5-fluoro-5' -deoxyuridine by Cytidine Deaminase (CDA). Thymidine phosphorylase (TPase) cleaves glycosyl bonds to form FU. Additional metabolic steps convert FU into the nucleotide analog FdUMP, which is responsible for the inhibition of thymidylate synthase by capecitabine.

Although the carbamate side chains were removed from both capecitabine and formula I by the same esterase (CES1 and CES2), the remaining prodrug activation of formula I was substantially different from capecitabine and was performed by the enzyme of nucleotide metabolism as shown in figure 2.

The present disclosure provides methods of preparing compounds of formula I. Scheme 1 outlines a general synthetic strategy for preparing the disclosed nucleoside derivatives, an example of which is the amino acid ester phosphorodiamidite member of the compound family of formula I (R ═ C)1-C7Alkyl, R ═ amino acid side chain). The reagents used are commercially available and the reaction types and purification steps are well known in the art. This process is based on the use of a specific molar equivalent of the lewis acid TMSOTf required for 100% yield of the glycosylation reaction, which was found during the development of the synthetic process.

Scheme 1. general description of the Synthesis

Figure BDA0002337872560000061

The yield of the anomeric mixture of 2 and 3 coupled to form 4 was found to be highly dependent on the molar excess of trimethylsilyl triflate (TMSOTf). It was found that, despite using 1.0 equivalent of TMSOTf, the yield was only 8%; but increasing TMSOTf to 2.55 equivalents, 2.75 equivalents, and 3.0 equivalents increased the yield to 58%, 85%, and 100%, respectively.

The synthesis of intermediates 4 and 5 was previously disclosed, but their preparation involved only a 2-fold excess of TMSOTf for the condensation reaction and the resolution of anomeric mixture 4 generated during glycosylation required chiral chromatography in addition, the isolation of pure β -anomer 5 required additional chromatographic purification this method was previously reported as an improvement of the literature procedure (Kotra, LP, et al, J.Med.Chem.1997, 40 th, 3635. page 3644) for the preparation of the L-enantiomer of 4, which involved a 2-fold excess of TMSOTf for the condensation reaction to give a 57% anomeric mixture.

In contrast, the processes disclosed herein (see examples 1 and 2) use a 3:1 molar excess of TMSOTf, yielding 100% yield of anomeric mixture in the condensation reaction, and furthermore, isolation of pure β -anomer was performed without resolution by any conventional chromatographic step used in the art, greatly simplifying the procedure.

In one example, a method of making the disclosed compounds comprises: provides a composition comprising 5-fluoro-N- (trimethylsilyl) -2- ((trimethylsilyl) oxy) pyrimidin-4-amine(2R,3R,5R) -3- (benzoyloxy) -4, 4-difluoro-5- ((methylsulfonyl) oxy) tetrahydrofuran-2-yl) methyl benzoate

Figure BDA0002337872560000072

And a solvent (e.g., dichloroethane), adding 3 equivalents of TMSOTf to the reaction mixture to give the product (2R,3R) -5- (4-amino-5-fluoro-2-oxopyrimidin-1 (2H) -yl) -2- ((benzoyloxy) methyl) -4, 4-difluorotetrahydrofuran-3-yl-benzoate

Figure BDA0002337872560000073

A mixture of α anomer and β anomer at 1.2:1.0 pure β anomer of free nucleoside 5 was obtained as described in example 2 without the need for conventional chromatographic separation of the anomer mixture compound 5 was useful to produce the compounds of the present disclosure using the methods disclosed herein.

Based on chemical composition and biological activity, the compounds of formula I are fluoropyrimidines other than other fluoropyrimidines (such as FU, tegafur, doxifluridine, floxuridine, and capecitabine). The primary targets for previous fluoropyrimidines have been identified, including:

(i) the enzyme thymidylate synthase (all fluoropyrimidines),

(ii) nucleic acid DNA that is misincorporated with FU and uracil (floxuridine and capecitabine),

(iii) nucleic acid RNA that was misincorporated into FU (tegafur, doxifluridine and capecitabine, less profound floxuridine).

Misincorporation into RNA is an undesirable effect because it adversely affects RNA synthesis and function in non-dividing normal as well as cancer cells, thereby reducing the selectivity index for all existing fluoropyrimidines. In contrast, misincorporation into DNA only affects dividing cells, contributing to the cytotoxicity of fluoropyrimidine.

It was found that the compound of formula I also targets thymidylate synthase and DNA, but not RNA in addition, various intracellular nucleotide analogs produced by the compound of formula I via its metabolic activation were found to target additional enzymes ribonucleotide reductase, DNA polymerase α and DNA (cytosine-5) methyltransferase the primary targets of known cytidine analogs compared to formula I are shown in FIG. 1.

It was also found that the compounds of formula I, in addition to FU and uracil, also mis-incorporate 5-fluorocytosine into DNA. Furthermore, the incorporation of FU linked to 2, 2-difluoro-2-deoxyribose, rather than to the native 2' -deoxyribose as in the case of other fluoropyrimidines, may have additional effects on the structure and function of DNA.

Various misincorporated nucleotides interfere with DNA replication and/or function and induce specific DNA repair processes, with different results depending on the nature and extent of DNA damage they cause. Without intending to be bound by any particular theory, DNA damage may result in apoptosis of cancer cells exposed to the compound of formula I, resulting in death.

The above comparison shows that the compounds of formula I inhibit more key enzymes and cause DNA damage by misincorporation of more abnormal nucleotides into DNA compared to the previous anticancer fluoropyrimidines used clinically. Thus, the disclosed compounds may reduce the likelihood of developing resistance to themselves and may be less susceptible to cross-resistance with other fluoropyrimidine analogs in their therapeutic use.

A unique aspect of the present disclosure is that the compounds of formula I cause the incorporation of 5-fluorocytosine into DNA, resulting in irreversible inactivation of DNA (cytosine-5) -methyltransferase. None of the FDA approved anti-cancer cytidine analogs are capable of incorporating 5-fluorocytosine into DNA. Inactivation of DNA (cytosine-5) -methyltransferases leads to elimination ("demethylation") of 5-methylcytosine residues in specific CpG sequences in DNA, thereby interfering with epigenetic regulation of cellular metabolism.

The capecitabine molecule also contains a 5-fluorocytosine base, which is converted to the corresponding 5-fluorouracil by deamination during metabolic activation of the drug. Thus, in contrast to the present disclosure, capecitabine cannot cause the incorporation of 5-fluorocytosine into DNA and has no direct effect on DNA (cytosine-5) -methyltransferase.

Several intracellular targets of the present disclosure were identified as key enzymes required for DNA biosynthesis and function. These enzymes are susceptible to inhibition by various cellular metabolites of the present disclosure, resulting in the observed cytotoxicity. Intracellular metabolites that affect enzyme inhibitory activity are all phosphorylated derivatives and are found as follows: 2',2', 5-Trifluorodeoxycytidine monophosphate, F3dCMP, an inhibitor of deoxycytidine deaminase (DCTD); 2',2', 5-Trifluorodeoxyuridine monophosphate, F3dUMP, an inhibitor of Thymidylate Synthase (TS); 2',2', 5-Trifluorodeoxycytidine diphosphate, F3dCDP, an inhibitor of Ribonucleotide Reductase (RR); 2',2', 5-Trifluorodeoxycytidine triphosphate, F3dCTP, DNA polymerase α (Pol α), 2', 5-Trifluorodeoxyuridine triphosphate, F3dUTP, DNA polymerase α (Pol α), 5-fluorocytosine, FC incorporated into DNA, DNA (cytosine-5) -methyltransferase (MTase) in one example, these metabolites are summarized in table 1.

TABLE 1 growth inhibitory Activity against human cancer cell lines

Target Inhibitory metabolites Reversibility
DCTD (Secondary) F3dCMP Reversible
TS (Main) F3dUMP Irreversible
RR (Main) F3dCDP Irreversible
Pol α (Main) F3dCTP,F3dUTP Irreversible
MTase (Main) 5-FC in DNA Irreversible

Inactivation by formation of covalent bonds at the active site; inhibition by DNA strand termination and/or DNA fragmentation.

A unique aspect of the present disclosure is that inhibitory activity of the nucleotide metabolites of the compounds of formula I is observed at all phosphorylation levels: phosphoric acid (F)3dCMP), diphosphonic acid (F)3dCDP), triphosphate (F)3dCTP,F3dUTP) and polynucleotides (DNA containing 5-FC).

Inhibition of the major enzyme targets (TS, RR and MTase) is irreversible due to mechanism-based inactivation by inhibitory metabolites. The molecular mechanism of inactivation involves the use of the active site to catalyze the formation of covalent bonds from the sulfhydryl group of a cysteine residue (see, examples 6, 7 and 8).

The observed misincorporation into nucleic acids may contribute to the biological activity of the disclosed compounds. Uracil, instead of thymine, is mistakenly incorporated into DNA, resulting in DNA fragmentation due to inhibition of Thymidylate Synthase (TS), causing cell death. Similar misincorporation of 5-fluorouracil into DNA instead of thymine produced similar results. In contrast, misincorporation of 5-fluorocytosine into DNA instead of cytosine leads to inactivation of DNA (cytosine-5) -methyltransferase (MTase), which in turn leads to DNA demethylation, disrupting epigenetic regulation of cellular metabolism. This represents the unexpected effect of the fluoropyrimidines of this disclosure in view of the previous fluoropyrimidines used for sixty years of treatment.

The erroneous incorporation of traditional fluoropyrimidines into RNA, while possibly a cause of its cytotoxicity, can also affect non-dividing cells in normal tissues, thereby reducing the therapeutic index of these drugs.

Testing against human cell lines cultured by the national cancer institute of america found that the compounds of the present disclosure have a broad spectrum of significant growth inhibitory activity. Table 2 shows that the growth of samples of various human cancer cell lines was inhibited by more than 50% by 10 micromolar of the proto-drug 6 a.

TABLE 2 multiple enzyme targets

Cell lines % growth inhibition
ACHN 86.4
HL60 75.8
NCI-H522 61.1
HCT-116 63.5
M14 85.9
MCF-7 51.9
OVCAR-8 71.6

Generated from 10 micromoles of 6a

The description of the human cancer cell lines used is as follows: ACHN, human renal adenocarcinoma; HL-60(TB), human acute myeloid leukemia; HCT-116, human colon cancer; HOP-62, human adenocarcinoma (non-small cell lung carcinoma); m14, human metastatic malignant melanoma; MCF7, human breast adenocarcinoma; OVCAR-8, human ovarian cancer.

These results demonstrate that the present disclosure demonstrates inhibitory activity against cancers derived from different tissues (leukemia as well as solid tumors).

Using cell viability assays of Promega, the following IC was obtained for KG-1 human acute myeloid leukemia cells50Values (table 3). The nucleoside carbamate capecitabine

Figure BDA0002337872560000092

Included for comparison.

TABLE 3 cell viability data

Figure BDA0002337872560000091

Figure BDA0002337872560000101

IC of capecitabine50value/IC of test Compound50Value of

Prototype compound 6a of formula I with a pentoxycarbonyl side chain showed 69-fold more potency than capecitabine with the same side chain. Compound 5 is the major intracellular nucleoside metabolite of the compound of formula I (F)3dCyd, see fig. 2), showed 253-fold efficacy compared to capecitabine.

The present disclosure provides compositions comprising one or more compounds of formula I. The composition may comprise one or more pharmaceutically acceptable carriers.

Compositions comprising one or more compounds of the present disclosure and a pharmaceutical carrier can be prepared at the patient's bedside or by a pharmaceutical manufacturer. In either case, the composition or its ingredients may be provided in any suitable container, such as a sealed sterile vial or ampoule, and may be further packaged to include instructions for use by a pharmacist, physician or other health care provider. The composition may be provided in combination with any suitable delivery form or vehicle, examples of which include liquids, caplets, capsules, tablets, inhalants or aerosols, and the like. The delivery device may include components that facilitate release of the drug over a period of time and/or interval, and may include compositions that enhance drug delivery, such as nanoparticle formulations, microsphere formulations, or liposome formulations, a variety of which are known in the art and commercially available. Additionally, procedures and specific compositions that enable delayed or sustained release of the active pharmaceutical ingredient are known in the art. In addition, each composition described herein may comprise one or more pharmaceutical agents.

The compositions described herein may comprise one or more standard pharmaceutically acceptable carriers. Pharmaceutically acceptable carriers depend, in part, on the particular composition being administered and the particular method used to administer the composition. Thus, there are a variety of suitable formulations of the pharmaceutical compositions of the present disclosure. Some examples of pharmaceutically acceptable carriers can be described in Remington, The Science and Practice of Pharmacy (2005), 21 st edition, Philadelphia, PA, Risperter Williams and Wilkins. Effective formulations include oral and nasal formulations, formulations for parenteral administration and compositions formulated for sustained release.

Examples of compositions that may be suitable for oral administration include, but are not limited to: (a) a liquid solution, such as an effective amount of a compound of the present disclosure suspended in a diluent (e.g., water, saline, or PEG 400); (b) capsules, caplets, depot or tablets, such as liquid, solid, granules or gelatin preparations, each containing a predetermined amount of the active ingredient; (c) suspensions in appropriate liquids; (d) a suitable emulsion; and (e) a patch. The liquid solutions described above may be sterile solutions. The compositions may comprise, for example, one or more of lactose, sucrose, mannitol, sorbitol, calcium phosphate, corn starch, potato starch, microcrystalline cellulose, gelatin, colloidal silicon dioxide, talc, magnesium stearate, stearic acid, and other excipients, colorants, fillers, binders, diluents, buffering aids, wetting aids, preservatives, flavoring agents, dyes, disintegrants, and pharmaceutically compatible carriers.

The compositions may have a unit dosage form. The compositions are subdivided in such dosage forms into multiple unit doses containing appropriate quantities of the active component. The unit dosage form can be a packaged preparation, the package containing discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or ampoules. Likewise, the unit dosage form may itself be a capsule, tablet, caplet, or lozenge, or it may be the appropriate number of any of these in packaged form. The compositions may also contain other compatible therapeutic agents, if desired. The compositions may deliver the compounds of the present disclosure in sustained release formulations.

The present disclosure provides methods of using one or more compounds of the present disclosure. For example, the compounds may be used to treat cancer and/or viral infections.

The steps of the methods described in the various embodiments and examples disclosed herein are sufficient to practice the methods of the present invention. Thus, in one embodiment, the method consists essentially of a combination of the steps of the methods disclosed herein. In another embodiment, the method consists of such steps.

For example, a method of treatment comprises administering to an individual one or more compounds of the disclosure or a composition comprising one or more compounds of the disclosure.

The method may be performed in an individual who has been diagnosed with or is suspected of having cancer or a viral infection (i.e., therapeutic use). The method may also be performed in individuals who relapse or are at high risk of relapse after treatment for cancer or viral infection.

For example, the methods of the disclosure can be performed such that upon exposure of a compound or composition of the disclosure to a cancer cell results in misincorporation of the analog into DNA, resulting in a change in the structure and function of the DNA, potentially resulting in death of the cancer cell.

For example, the methods of the present disclosure can be performed such that a ribonucleotide reductase, DNA polymerase a, thymidylate synthase, DNA (cytosine-5) -methyltransferase, or a combination thereof is inhibited using a compound of the present disclosure.

The methods of the present disclosure can be performed in an individual in need of prevention or treatment of a viral infection/disease. Viral targets include, but are not limited to, HIV, HBV, HCV, HSV1, and HSV 2. For example, a method of treating a viral infection comprises administering a compound or composition of the disclosure to an individual in need of treatment, thereby eliminating the virus or curing the individual with the virus.

Compositions comprising the compounds described herein can be administered to a subject using any known method and route, including oral, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intranasal, and intracranial injection. Parenteral infusion includes intramuscular, intravenous, intraarterial, intraperitoneal and subcutaneous administration. Administration also includes topical and/or transdermal administration.

The dosage of a composition comprising a compound of the present disclosure and a pharmaceutical agent will necessarily depend on the needs of the individual to whom the composition of the present disclosure is administered. These factors include, for example, body weight, age, sex, medical history, and the nature and stage of the disease for which treatment or prevention is desired. The compositions may be used in conjunction with any other conventional treatment modality designed to ameliorate the condition for which a desired therapeutic or prophylactic effect is intended, non-limiting examples of which include surgical intervention and radiation therapy. For example, the composition is used in combination (e.g., co-administered) with one or more known anticancer agents (e.g., DNA-damaging anticancer agents) or known antiviral agents.

The methods of the present disclosure may be used for a wide variety of individuals. In various examples, the individual is a human or non-human mammal. Examples of non-human mammals include, but are not limited to, farm animals (e.g., cattle, pigs, sheep, etc.) and pet or sports animals (e.g., horses, dogs, cats, etc.). Other non-limiting examples of individuals include rabbits, rats, mice, and the like. The compounds or compositions of the present disclosure can be administered to an individual, for example, in a pharmaceutically acceptable carrier that facilitates transport of the compound from one organ or portion of the body to another organ or portion of the body.

The following examples are presented to illustrate the present disclosure. They are not intended to be limiting in any way.

Example 1

This example provides a description of the synthesis of 3',5' -di-O-benzoyl-2 ' -deoxy-2 ',2', 5-trifluorocytidine (4).

Under argon in ammonium sulfate (NH)4SO4679mg) 5-fluorocytosine (1, 23.1g, 178.9mmol) was treated with hexamethyldisilazane (HMDS, 1,000mL) and refluxed at 125 ℃ for 5 hours. Volatiles were removed under vacuum under an argon atmosphere to give 2 as a white solid. This was dissolved in anhydrous dichloroethane (400mL), and a solution of 3, 5-di-O-benzoyl-2-deoxy-2, 2-difluoro-1-O-methanesulfonyl-a-D-furanoside (3, 32g, 70.16mmol) in anhydrous dichloroethane (300mL) was added. The reaction mixture was stirred at room temperature for 10 minutes (min). TMSOTf (38mL, 210.5mmol) was then added and the mixture was stirred under argon at 95 ℃ for 16 hours (hrs). The reaction mixture was cooled to room temperature and then NaHCO was added3The solution was saturated and then stirred for 10 minutes. The resulting phases were separated and the aqueous phase was extracted with dichloromethane. NaHCO for organic phase3And brine treatment with MgSO4Drying and concentration in vacuo gave 4(37g) as a pale yellow solid. Warp beamNMR and LCMS analysis gave compound 4 as a mixture of a and β anomer at 1.2:1.0 compound 4 was used in the next step without further purification.

Example 2

This example provides a description of the synthesis of 2' -deoxy-2 ',2', 5-trifluorocytidine (5).

With 7N NH4OH in MeOH (190mL) Compound 4 (see example 1, 33.9g, 69.3mmol) was stirred at room temperature overnight. Evaporating the volatiles and reacting in CH2Cl2And H2The residue was partitioned between O. By CH2Cl2(3X) the aqueous phase is extracted and then evaporated to dryness. The residue was slurried in i-PrOH (75mL) and heated to 60 ℃ before concentration. To one portion was added HCl (15 mL). The thick suspension was cooled to room temperature and white crystals formed immediately, which were filtered off and washed with cold i-PrOH, petroleum ether and Et2O washing to obtain compound 5 which is a mixture of alpha/β anomer at a ratio of 1.5:1.0, suspending the compound 5 in H at 60 ℃2O, then 4N NaOH was slowly added to put the mixture in solution, after which the pH was adjusted to-8.3 with 1N NaOH, the solid was precipitated, collected and washed with ice water, dried in vacuo to give amine-free 5(5.85g) as a white solid in a yield of 30% over 3 steps (β -anomer 100% by HPLC) mp 118 ℃; [ a ] a]D ═ 79.64 ° (C0.28, MeOH), LCMS (C-18, 5% isocratic H)2O/MeCN), ELSD (Peak, 1.065min), UV (Peak, 0.96min), Positive mode: M/z 282[ M + H ]]+(ii) a Negative mode, M/z 280[ M-H ═ M/z]-,316[M+Cl]-.C9H10F3N3O4(281.19)。

1H NMR(DMSO-d6)δ=3.79(dd,1H,H-5'),3.80(m,2H,H-4',H-5"),4.18(m,1H,H-3'),5.34(br t,1H,5'-OH),6.05(t,J1',F=7.35Hz,1H,H-1'),6.25(br d,1H,3'-OH),7.75,8.00(2s,2H,NH2),8.04(d,J6,F=7.23Hz,1H,H-6).

H, H-NOESY:4.18(H-3') is related to 8.04 (H-6); 6.05(H-1') correlates with 3.8 (H-4').

1H HNR(D2O):δ=3.88(dd,1H,Jgem=13.23Hz,J5',4'=3.66Hz,H-5'),4.03(dd,Jgem=12.99Hz,H-5"),4.08(m,1H,H-4'),4.35(m,1H,H-3'),6.19(t,J1',F=7.29Hz,1H,H-1'),7.93(d,J6,F=6.36Hz,1H,H-6).

13C NMR(DMSO-d6)δ=58.67(s,1C,C-5'),68.11(t,J3',F=22.23Hz,1C,C-3'),80.44(s,1C,C-4'),83.60(t,J1',F=31.51Hz,1C,C-1'),122.98(t,J2',F=258.06Hz,1C,C-2'),124.82(d,J6,F=32.18Hz,1C,C-6),136.20(d,J5,F=242.39Hz,1C,C-5),152.88(s,1C,C-2),157.62(d,J4,F=13.77Hz,1C,C-4).

Example 3

This example provides a description of the synthesis of 2' -deoxy-2 ',2', 5-trifluorocytidine hydrochloride (5. HCl).

To a suspension of compound 5 (see, example 2, 5.85g, 20.8mmol) was added concentrated HCl (5mL) at 60 ℃. Cooled to room temperature and then crystallized. Filtering the crystals with i-PrOH, petroleum ether and Et2O wash, then vacuum dry to give 5.HCl as a white solid (5.85g, 89% from 5): mp 128-135 deg.C; [ a ] A]D +52.23 ° (C0.28, MeOH), LCMS (C-18; 5% isocratic H)2O/MeCN), ELSD (Peak at 1.08 min), UV (Peak at 0.975 min), Positive mode, M/z 282[ M + H ]]+(ii) a Negative mode, M/z 280[ M-H ═ M/z]-,316[M+Cl]-.C9H11F3N3O4Cl(317.20)。

1H NMR(DMSO-d6)δ=3.63(dd,1H,H-5'),3.79(m,2H,H-4',H-5"),4.18(m,1H,H-3'),5.34(t,JOH,5'=5.16Hz,1H,5'-OH),6.06(t,J1',F=7.05Hz,1H,H-1'),6.25(br,1H,3'-OH),7.75,8.01(2s,2H,NH2),8.04(d,J6,F=7.17Hz,1H,H-6).

1H HNR(D2O):δ=3.88(dd,1H,Jgem=13.17Hz,J5',4'=3.6Hz,H-5'),4.04(dd,Jgem=13.59Hz,H-5"),4.09(m,1H,H-4'),4.35(m,1H,H-3'),6.20(t,J1',F=6.78Hz,1H,H-1'),7.94(d,J6,F=6.24Hz,1H,H-6).

13C NMR(DMSO-d6)δ=58.68(s,1C,C-5'),68.13(t,J3',F=22.57Hz,1C,C-3'),80.51(s,1C,C-4'),83.61(t,J1',F=32.84Hz,1C,C-1'),122.99(t,J2',F=258.1Hz,1C,C-2'),124.80(d,J6,F=32.63Hz,1C,C-6),136.22(d,J5,F=242.42Hz,1C,C-5),152.94(s,1C,C-2),157.67(d,J4,F=13.75Hz,1C,C-4).

Example 4

This example provides synthesis of N4-pentyloxycarbonyl-2 ' -deoxy-2 ',2', 5-trifluorocytidine (6 a).

A solution of trifluoronucleoside 5(1.0g, 3.5mmol) in acetonitrile (30mL) and pyridine (2 equiv.) was cooled to 4 deg.C and HMDS (1.1 equiv.) added thereto. A solution of amyl chloroformate (1.0 eq) in methylene chloride was then added dropwise, maintaining the solution at 4 ℃. The mixture was then warmed to room temperature and stirring was continued for 2 hours. Water (30mL) was added, the organics extracted with dichloromethane (2X 30mL), MgSO4Dried and concentrated in vacuo. The crude product 6a (1.4g) thus obtained is shown as a mixture of 6a and 5' -carbonate. To a solution of the crude product (1.4g) in MeOH (30mL) was added sodium methoxide (0.6mL, 25 wt% in MeOH), and the reaction mixture was stirred at room temperature for 2.5 hours. Then using Amberlite H+The resin adjusted the pH of the reaction mixture to 7. The mixture was then filtered and concentrated in vacuo. Purification by flash chromatography (0-10% MeOH in EtOAc) afforded 6a as a white solid (0.89g, 66% yield): mp 75-86 ℃; 396[ M + H ] M/z]+;HPLC(UV,260nm)99.46%。

1H NMR(DMSO-d6):δ=0.86-0.91(m,3H,CH3),1.31-1.33(m,4H,CH2),1.59-1.63(m,2H,CH2),3.62-3.67(m,1H,H-5'),3.84-3.90(m,2H,H-4',H-5'),4.08-4.20(m,2H,CH2),4.24-4.67(m,1H,H-3'),5.42(m,1H,OH),6.06(m,1H,H-1'),6.34(d,J6,F=6.3Hz,1H,H-6),8.40(br s,1H,OH),10.66(br s,1H,NH).

13C NMR(DMSO-d6)δ=14.9(s,1C,CH3),21.2(s,1C,CH2),28.6(d,1C,CH2),58.6(s,1C,C-5'),65.1(s,1C,CH2),67.4(t,1C,C-3'),80.2(s,1C,C-4'),84.1(t,1C,C-1'),119.7(s,1C,C-2'),122.3(s,1C,C-6),125.5(s,1C,C-5).

HSQC (D2O) 7.85ppm (H-6) correlated with 130ppm (C-6).

19F NMR(DMSO-d6):δ=-159(s,1F,F-5),-117(s,2F,F-2').

Example 5

This example provides a description of the structure-activity relationship.

The structural determinants required for the pharmacological activity of the compounds of formula I have been identified. The molecular characteristics characteristic of formula I are shown in circles:

Figure BDA0002337872560000141

part 1: the fluorine atom at the 5-position of the pyrimidine ring is essential for inhibiting both Thymidylate Synthase (TS) and DNA (cytosine-5) -methyltransferase;

part 2: is a 5-fluorocytosine base in N4-a prodrug modifying substituent at position that directs the compound of formula I primarily to the liver for metabolic activation by liver carboxyesterases CES1 and CES2, thereby protecting against gastrointestinal toxicity, i.e. toxic side effects characteristic of fluoropyrimidines;

part 3: the bis-fluoro disubstituted group at the 2 '-position of the 2' -deoxyribose is critical for the inhibition of ribonucleotide reductase and DNA polymerase a;

part 4: is a prodrug modifying substituent of 2',2' -difluoro-2 ' -deoxyribose at the 5' -OH group that can deliver the intact 5' -monophosphate form of the compound of formula I into a cell; when P is triphosphate, the corresponding metabolite can serve as a substrate for DNA polymerase and can lead to misincorporation into DNA;

and part 5: is a hydrolyzable 3' -OH-protecting group that modulates solubility and permeability; it is removed by enzymatic hydrolysis.

Example 6

This example provides a solution by F3Illustrative of the molecular mechanism by which dUMP metabolites inactivate Thymidylate Synthase (TS).

F3dUMP passage with thymidineThe synthetase and the cofactor 5, 10-methylenetetrahydrofolate form a covalent ternary complex that serves as a suicide substrate for the thymidylate synthetase. Binding of the inhibitor to the enzyme is achieved by forming a covalent bond with the SH-group of the active site cysteine residue involved in catalysis:

Figure BDA0002337872560000151

example 7

This example provides a solution by F3An illustration of the molecular mechanism by which dCDP metabolites inactivate Ribonucleotide Reductase (RR).

Formation of a binary complex between the inhibitor and the enzyme by covalent bond formation with the SH-group of the cysteine residue of the active site, whereby F3dCDP as a suicide substrate for the enzyme:

Figure BDA0002337872560000152

example 8

This example provides a solution by F3The incorporation of dCMP metabolites into DNA exemplifies the molecular mechanism by which DNA (cytosine-5) -methyltransferase (MTase) is inactivated.

F misincorporated into DNA by forming a covalent binary complex with the enzyme and being methylated in the process3The 5-fluorocytosine portion of dCMP acts as a suicide substrate for the enzyme, DNA (cytosine 5) -methyltransferase (MTase). The binding to the enzyme is achieved by forming a covalent bond with the SH-group of the cysteine residue of the active site involved in catalysis. The mechanism of enzyme inactivation is similar to that described for TS in example 6.

Figure BDA0002337872560000161

Example 9

This example provides for N4-description of the metabolic activation mechanism of alkyl carbamate prodrug moieties.

Enzymatic hydrolysis catalyzed by liver carboxyesterases CES1 and CES2 releases the free amino group at the 4-position of the pyrimidine ring:

Figure BDA0002337872560000162

example 10

This example provides a description of the mechanism of metabolic activation of the amino acid ester phosphorodiamidate prodrug (prodrug nucleotide) moiety at the 5 '-position of the 2',2 '-difluoro-2' -deoxyribose moiety.

Intracellular esterases hydrolyze the 5' -substituent of formula I (R ″ ═ phosphorodiamidite) and subsequent cyclization results in the folding of the cyclic phosphate intermediate into a monophosphoramide compound. Further hydrolysis by phosphoamidase activity of HINT1 leads to F3Formation of dCMP, F3dCMP is the first intracellular phosphorylated metabolite of the compound of formula I, which acts as a substrate and competitive inhibitor for the enzyme dCMP deaminase and is a precursor to all other inhibitory nucleotides mentioned above.

Figure BDA0002337872560000163

Although the present disclosure has been described with respect to one or more particular embodiments and/or examples, it is to be understood that other embodiments and/or examples of the disclosure may be made without departing from the scope of the disclosure.

18页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:治疗帕金森氏病(PD)和相关疾病的组合物和方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!