WO1991016309A1 - Synthetic piperidinediones with cytostatic activity - Google Patents

Synthetic piperidinediones with cytostatic activity Download PDF

Info

Publication number
WO1991016309A1
WO1991016309A1 PCT/US1991/002483 US9102483W WO9116309A1 WO 1991016309 A1 WO1991016309 A1 WO 1991016309A1 US 9102483 W US9102483 W US 9102483W WO 9116309 A1 WO9116309 A1 WO 9116309A1
Authority
WO
WIPO (PCT)
Prior art keywords
dione
cells
compound
cell
cytostatic
Prior art date
Application number
PCT/US1991/002483
Other languages
French (fr)
Inventor
Lawrence B. Hendry
Chung K. Chu
Virendra B. Mahesh
Original Assignee
University Of Georgia Research Foundation, Inc.
Stereochemical Genetics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Georgia Research Foundation, Inc., Stereochemical Genetics, Inc. filed Critical University Of Georgia Research Foundation, Inc.
Publication of WO1991016309A1 publication Critical patent/WO1991016309A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/80Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D211/84Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen directly attached to ring carbon atoms
    • C07D211/86Oxygen atoms
    • C07D211/88Oxygen atoms attached in positions 2 and 6, e.g. glutarimide

Definitions

  • This invention is in the area of synthetic organic chemistry, and specifically relates to new
  • a tumor is an unregulated, disorganized
  • a tumor is malignant, or cancerous, if it has the properties of invasiveness and metastasis.
  • Invasiveness refers to the tendency of a tumor to enter surrounding tissue, breaking through the basal laminas that define the boundaries of the tissues, thereby often entering the body's circulatory system.
  • Metastasis refers to the tendency of a tumor to migrate to other areas of the body and establish areas of proliferation away from the site of initial appearance.
  • Cancer is now the second leading cause of death in the United States, Europe, and Japan, resulting in approximately 1,000,000 deaths annually in these countries. In the United States alone, each year over one million people are diagnosed with cancer, and over 500,000 people die from the disease. The number of newly diagnosed cancerous growths in patients in the United States is growing at a rate of 3% a year.
  • Oncogenes are initially normal genes (called prooncogenes) that by mutation or altered context of expression become transforming genes. The products of transforming genes cause inappropriate cell growth. More than twenty different normal cellular genes can become oncogenes by genetic
  • Transformed cells differ from normal cells in many ways, including cell morphology, cell- to-cell interactions, membrane content, cytoskeletal structure, protein secretion, gene expression and mortality (transformed cells can grow indefinitely).
  • the most freguent tumor site is lung, followed by colorectal, breast, prostate, bladder, pancreas, and then ovary.
  • Cancer is now treated with one or a combination of three types of therapies: surgery, radiation, and chemotherapy.
  • Surgery involves the bulk removal of diseased tissue. While surgery is sometimes effective in removing tumors located at certain sites, for example, in the breast, colon, and skin, it cannot be used in the treatment of tumors located in other areas, such as the backbone, nor in the treatment of disseminated neoplastic conditions such as leukemia.
  • Chemotherapy now represents less than 4% of the total expenditures on the treatment of cancer.
  • Chemotherapy involves the disruption of cell
  • replication or cell metabolism is used most often in the treatment of leukemia and breast, lung, and testicular cancer.
  • chemotherapeutic agents there are four major classes of chemotherapeutic agents currently in use for the treatment of cancer; anthracyclines, alkylating agents, antiproliferatives, and hormonal agents. A variety of methods exist to attempt to identify new antineoplastic materials.
  • chemotherapeutic agents including random screening of compounds, preparation of analogs of active compounds, computer or physical modeling, and combinations of these technigues. None of these methods, however, have yet identified the optimal chemotherapeutic agent for neoplastic diseases.
  • U.S. Patent No. 4,461,619 to Hendry et al. discloses a method to determine the relationship of chemical structure to biological activity based on the topology and physicochemical properties of "cavities" or "artificial constructs" in double stranded DNA, double stranded RNA, or double stranded DNA-RNA. While modeling can be very helpful in chemotherapy research, it cannot predict whether a target compound will pass through the cell wall, whether it is stable in vivo generally or in the cytoplasm specifically, or whether the therapeutic index is appropriate for clinical use of the drug.
  • Burzynski has proposed that the human organism is equipped with a corrective system that can reprogram the growth of newly developed neoplastic cells to transform them back into normal cells. He has
  • antineoplastons isolated a number of medium sized peptides, referred to as antineoplastons, that are produced by the body to protect it against the development of neoplastic growth by a nonimmunological process that does not significantly inhibit the growth of normal tissue.
  • the initial hydrolysis product and biological degradation product of A10 is phenylacetylglutamine, which is produced in vivo from phenylacetic acid and glutamine.
  • A10 may be cyclized from
  • Hendry has shown that A10 fits in a stereochemical manner between base pairs of double stranded DNA.
  • A10 is capable of forming a stereospecific hydrogen bond between the imino proton of the piperidinedione ring and the phosphate oxygen on the DNA backbone. A10 does not bind covalently to DNA, which may explain why the compound is cytostatic and not cytotoxic.
  • the acute toxicity of A10 in mice is between 1.35 and 10.33 g/kg. A10 has been administered without serious side effects at a dosage of up to 10 grams per day to humans suffering from cancer. While A10 is a useful drug in the treatment of neoplastic diseases, there is a need for new
  • cytostatic agents that may be more effective in stimulating tumor remission, and that may be effective when administered in lower dosages. The tragic number of deaths that occur each year from this disease accentuates the urgency of this need.
  • invention to provide a compound that has a cytostatic effect on cancer cells.
  • R is OH, NH 2 , OW, or H;
  • X is H, F, Cl, Br, I, OH, OW, NO 2 or NH 2 ;
  • Y is H, F, Cl, I, or Br;
  • W is C(O)Z or a C 1 to C 12 alkyl group;
  • Z is an aliphatic or
  • R is OH or NH 2 .
  • the most preferred compound is (4-hydroxy-3-N- phenylacetylamino-2,6-piperidinedione), in which R is OH, X is H and Y is H.
  • the preferred compound 4-hydroxy-3-N-phenylacetylamino-2, 6- piperidinedione, has significant cytostatic activity against prolactin stimulated Nb2 lymphoma cells (a T cell derived lymphoma), MCF-7 cells (estrogen
  • mice lymphoma (YAK) cells mouse lymphoma
  • Cytostatic activity refers to the ability of a compound to inhibit cell growth or replication.
  • cytotoxic activity refers to the ability of a compound to kill cells.
  • Cytostatic agents typically cause reversible chemical changes in cells.
  • An example of a reversible change is the formation of ionic bonds or hydrogen bonding between the cytostatic agent and molecules in the cells.
  • Cytotoxic compounds typically cause
  • irreversible changes in cells An example of an irreversible change is the formation of a covalent bond.
  • Figure 1 is a schematic illustrating a method of preparation of the cytostatic piperidinediones of the present invention.
  • piperidinedione compounds are abbreviated as follows: a, 4-HO-C 6 H 4 ; b, 2-Cl,6-F-C 6 H 3 ; C, 2-F-C 6 H 4 ; d, 3-F-C 6 H 4 ; e, 4-F-C 6 H 4 ; f, 2,6-di-F-C 6 H 3 ; g, 4-CH 3 O-C 6 H 4 ; h, 4-Cl- C 6 H 4 ; i, 4-Br-C 6 H 4 ; j, 4-CF 3 -C 6 H 4 .
  • the following 4-HO-C 6 H 4 ; b, 2-Cl,6-F-C 6 H 3 ; C, 2-F-C 6 H 4 ; d, 3-F-C 6 H 4 ; e, 4-F-C 6 H 4 ; f, 2,6-di-F-C 6 H 3 ; g, 4-CH 3 O-C 6 H 4 ; h, 4-Cl- C
  • step (i) N,N- dicyclohexylcarbodiimide (DCC) in acetonitrile
  • step (ii) NaHCO 3 in acetonitrile/H 2 O (2/1 ratio)
  • step (iii) DCC in dimethylformamide (DMF)
  • step (iv) DMF.
  • Figure 2 is a bar chart graph indicating the inhibition of prolactin stimulated Nb2 lymphoma cell growth by cytostatic piperidinediones. Equivalent amounts (1 mg/ml) of A10 or A10 analogs were
  • Figure 3 is a graph indicating the inhibitory effect of 3-[N-4-hydroxyphenylacetylaminopiperidine]-
  • FIG. 4 is a bar chart graph indicating the inhibition of mouse lymphoma (YAK) cell proliferation by the cytostatic piperidinediones of the present invention
  • YAK mouse lymphoma
  • PAG phenylacetylglutamine
  • the present invention is a compound with
  • R is OH, NH 2 , OW, or H;
  • X is H, F, Cl, Br, I,
  • Active compounds include 3-[N-4- hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-4- hydroxy-3-fluorophenylacetylaminopiperidine]-2,6- dione, 3-[N-4-hydroxy-3- chlorophenylacetylaminopiperidine]-2,6-dione, 3-[N-
  • the active compound has a proton donating group (such as hydroxyl or amino) in the 4-position of the aromatic ring and an electron withdrawing or proton donating group in the 3-position of the aromatic ring.
  • piperidinedione that is capable of hydrogen bonding to only one strand of the helix, such as A10.
  • these compounds are expected to have greater therapeutic efficacy and lower toxicity than A10.
  • Cytostatic agents are useful to moderate the growth of proliferative cells such as neoplastic cells in cell culture or animal models for purposes of research in the area of proliferative diseases.
  • Cytostatic agents are also useful in the study of the bonding patterns and consequent activity of
  • Cytostatic agents may also have a pharmaceutical use as antineoplastic agents in vivo.
  • cytostatic piperidinedione derivatives can be prepared by condensation of the appropriate
  • a reagent such as N- hydroxysuccinimide in the presence of DCC (N,N- dicyclohexylcarbodiimide), 2-mercaptothiazoline in the presence of DCC, or DCC alone.
  • phenylacetylglutamine derivative is also preferably activated before cyclization by reaction with N- hydroxysuccinimide in the presence of DCC or by reaction with 1,1'-carbonyldiimidazole.
  • Desired derivatives of phenylacetic acid can be purchased commercially or prepared synthetically by methods known to those skilled in the art according to well established rules of electrophilic and
  • 4- hydroxyphenylacetic acid which is commercially available from Aldrich Chemical Company, Inc., can be nitrated with dilute HNO 3 to produce 4-hydroxy-3- nitrophenylacetic acid, that is used as is in the next step of reaction.
  • the nitro group in 4-hydroxy-3-nitrophenylacetic acid can be reduced to the corresponding amine and then reacted with sodium nitrite in acid to form the diazonium salt, that can be converted into a wide range of functional groups, including chloro (CuCl), fluoro (HBF 4 ), bromo (CuBr) and hydroxyl (H 2 SO 4 ).
  • Phenylacetic acid can be nitrated with dilute HNO 3 to produce 4-hydroxy-3- nitrophenylacetic acid, that is used as is in the next step of reaction.
  • the nitro group in 4-hydroxy-3-nitrophenylacetic acid can be reduced to the corresponding amine and then reacted with sodium nitrite in acid to form the diazonium salt, that can
  • Prodrugs of the hydroxyl or amino derivatives of 3-N-phenylacetylamino-2,6-piperidinedione can be prepared by alkylation or acylation of the hydroxyl or amino moieties according to established methods.
  • These protecting groups can be cleaved in vivo or in vitro by the appropriate enzyme, generating the active compound.
  • 2a, 2c-2j were prepared as described below from the appropriate substituted phenylacetic acids (1a, 1c- 1j), respectively.
  • Example 2 General Procedure for the Preparation of Substituted Phenylacetyl-L-Glutamines.
  • N-hydroxysuccimide ester of 2-chloro-6-fluoro-phenyl acetic acid (2b, 12.5 g, 42 mmol) in 120 mL of acetonitrile was added a solution of L-glutamine (6.14 g, 42 mmol) in a mixture of 170 mL of water and 340 mL of acetonitrile containing sodium bicarbonate (7 g, 84 mmol). The mixture was stirred at ambient temperature for 24 hours.
  • N-hydroxysuccinimide (5 g, 42 mmol) in 50 mL of anhydrous dimethylformamide (dried over 4 ⁇ molecular sieve) was added to the solution of 2- chloro-6-fluoro-phenylacetyl-L-glutamine (3b, 15.3 g, 42 mmol) in 300 mL of anhydrous DMF.
  • N,N- Dicyclohexylcarbodiimide (8.66 g, 42 mmol) was added to the mixture with stirring. The mixture was stirred at 80°C for 6 hours and then stirred at ambient temperature for 18 hours. The colorless precipitate obtained was filtered off and the filtrate then used directly in the procedure of Example 4 without
  • the compounds 4a, 4c-4j were prepared at room temperature as described above from 3a, 3c-3j,
  • piperidinediones were tested in three cell lines:
  • the rat Nb2 lymphoma cell line a T-cell derived lymphoma
  • FCS fetal calf serum
  • HS horse serum
  • interleukin-2 IL-2
  • phorbol esters have mitogenic effects in this cell line.
  • Antineoplaston A10 (3-phenylacetylamino-2,6- piperidinedione) was a generous gift from the
  • the mass spectral fragmentation pattern was consistent with 3-phenylacetylamino-2,6- piperidinedione (A10) (m/e 246, M+; 155, M+ -C 7 H 7 ; 127; 118; 110; 99; 91, C 7 H 7 +; 84; 65; 56). While the mass spectrum of A10 was not present in the computer library, a search revealed an expected close match for the hydrolysis product of A10, 3-N- phenylacetylglutamine. No discernable impurities were detected using reconstructed ion chromatograms and the ion chromatograms of the major diagnostic ions (m/e 246, 155, and 91) possessed single superimposable chromatographic peaks. The retention times and mass spectra obtained in all cases were consistent with those previously reported for A10 as well as those obtained from authentic samples.
  • Penicillin/streptomycin, FCS, and Fischer's media were obtained from Gibco Laboratories (Grand Island, NY). HS was purchased from MA Bioproducts
  • Rat prolactin (rPRL-RP3) was supplied by NIAMDD of the National Institute of Rat prolactin (rPRL-RP3) was supplied by NIAMDD of the National Institute of Rat prolactin (rPRL-RP3) was supplied by NIAMDD of the National Institute of Rat prolactin (rPRL-RP3) was supplied by NIAMDD of the National Institute of Rat prolactin (rPRL-RP3) was supplied by NIAMDD of the National Institute of
  • NB2 lymphoma cells The proliferation of NB2 lymphoma cells was determined by the following procedure. The cells were centrifuged (300 x g, 4 minutes), washed three times in media containing only HS, and then resuspended in the same media. Cells were then cultured for 24 hours prior to use, at which time prolactin and test
  • test compound in the culture system was 1 mg/ml. Approximately 5 mg of each of the compounds were weighed and dissolved in heated DMSO (40°C) to give an equivalent concentration of 200 mg/ml. An equal volume of heated 95% ethanol was added to each compound to give a 100 mg/ml
  • FIG. 2 A comparison of the inhibitory effects of the piperidinediones on PRL stimulation of proliferation in the NB2 lymphoma cell line (0.4 ng/ml) is provided in Figure 2. As illustrated, p-OH-A10 provided the greatest inhibition of lymphoma cell growth.
  • HQH-1-45-28 (3-[N-2-chloro,6- fluorophenylacetylaminopiperidine]-2,6-dione), HQH-1- 51-29 (3-[N-4-fluorophenylacetylaminopiperidine]-2,6- dione), and HQH-2-48-30, (3-[N-3- fluorophenylacetylaminopiperidine]-2,6-dione) also significantly inhibited proliferation of these cells.
  • the 2-fluorophenyl-A10 derivative and the 2,2- difluorophenyl-A10 derivative provided some inhibition of cell growth.
  • Example 6 Inhibition of MCF-7 Cells MCF-7 cells were grown in 5% fetal calf serum and 10 ⁇ g insulin/ml in MEM supplemented media. The cells were trypsinized during log phase growth and plated in T25 flasks at a cell number of 20,000 cells in 2 ml media. The cells were allowed to attach during a period of 48-72 hours. The media was then changed.
  • the cell numbers were counted in control flasks and 1, 2.5, 5.0 and 10 nM of A-10 or p-OH A-10 or control medium was added to each flask. Each experiment was done in triplicate. The media was changed at the end of 3 days and new aliquots of A-10 or p-OH A-10 or control media were added. After 3 more days of cell growth, the cells were detached from the culture flasks by adding 2 ml of a solution of 0.05% trypsin and 0.02% EDTA. The cells were removed, and 20 ml isoton was added and counted in a Coulter Counter.
  • Figure 3 is a graph indicating the inhibitory effect of 3-[N-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione (p-OH A10) and 3-[N- phenylacetylaminopiperidine]-2,6-dione (A10) on MCF-7 cells growing in log phase.
  • p-OH A10 3-[N-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione
  • A10 3-[N- phenylacetylaminopiperidine]-2,6-dione
  • Cells were grown in Dalbecco's modified Eagle's media in log phase. These cell grow in suspension. Cells (22,263) were plated and treated with 4 mM quantities of various compounds. Cell numbers were counted after 72 hours of treatment by removing 100 ⁇ l aliquots and diluting the aliquot with 20 ml isoton. The DMSO:ETOH concentration in all samples was 0.53%.
  • Figure 4 is a bar chart graph indicating the inhibition of mouse lymphoma (YAK) cell proliferation by cytostatic piperidinediones. As illustrated, p-OH- A10 provides dramatic inhibition of the growth of these hormone insensitive cells. Other 3-[N- phenylacetylaminopiperidine]-2,6-dione derivatives, including the 4-fluorophenyl, the 2-fluorophenyl, the 3-fluorophenyl, and the 2-chloro,6-fluorophenyl derivatives also provided some inhibition of cell growth. IV. Preparation of Pharmaceutical Compositions and
  • the cytostatic piperidinediones of the present invention are useful in the study of proliferative diseases in animal models and in vitro cell cultures.
  • the active compounds may also have a use in the treatment of neoplastic diseases in vivo.
  • Pharmaceutical compositions including these active compounds can be prepared as described below.
  • the active compound or its pharmaceutically acceptable salt is included in the pharmaceutically acceptable carrier or diluent in an amount sufficient to exert an inhibitory effect on the growth of the target neoplastic or proliferative cell line in vitro or in vivo.
  • the active materials can be administered by any appropriate route, for example, orally, parenterally, intravenously, intradermally,
  • the active compound is included in the
  • inhibitory effect on neoplastic or proliferative cells without serious toxic effect to healthy cells.
  • inhibitory amount is meant an amount of active ingredient sufficient to exert a cytostatic effect as measured by, for example, an assay such as that described in Examples 5 through 7, or measured by blood analysis or radiation analysis of the state of tumorigenesis in vivo.
  • the concentration of active compound in the drug composition will depend on absorption, inactivation, and excretion rates of the active compound as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions.
  • the active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
  • the compound may be provided in a
  • composition that protects it from the acidic
  • the compound can be orally administered in combination with an antacid formulation.
  • the composition can also be administered in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine.
  • Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules.
  • compositions can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a
  • disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • a sweetening agent such as sucrose or
  • saccharin or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • dosage unit form When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil.
  • dosage unit forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or other enteric agents.
  • the active compound or its pharmaceutically acceptable salt can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like.
  • a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes, colorings and flavors.
  • the active compounds can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, including other cytostatic or anticancer compounds.
  • Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens;
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants such as ascorbic acid or sodium
  • ethylenediaminetetraacetic acid ethylenediaminetetraacetic acid
  • buffers such as acetates, citrates or phosphates
  • agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • preferred carriers are physiological saline or phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations are known to those skilled in the art. The materials can also be obtained commercially from Nova Pharmaceutical Corporation.
  • Liposomal suspensions are also preferred as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811 (which is incorporated herein by
  • liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound is then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
  • appropriate lipid(s) such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol
  • R is OH, NH 2 , or OW
  • X is H, F, Cl, Br, OH, OW, NO 2 or NH 2
  • Y is H, F, Cl, or Br
  • W is C(O)Z or a C 1 to C 12 alkyl group
  • Z is an aliphatic or aromatic group of from C 1 to C 12
  • X and Y can both vary within the molecule; or its pharmaceutically acceptable salt.
  • the compound of claim 1 further comprising a pharmaceutical carrier.
  • pharmaceutical carrier is for topical application.
  • pharmaceutical carrier is for systemic administration.
  • controlled delivery device is selected from the group consisting of microcapsules, microspheres, and
  • the compound of claim 6 further comprising a pharmaceutical carrier.
  • pharmaceutical carrier is for topical application.
  • pharmaceutical carrier is for systemic administration.
  • controlled delivery device is selected from the group consisting of microcapsules, microspheres, and
  • a method of inhibiting cell growth comprising providing an effective cytostatic amount of a compound of the formula
  • R is OH, NH 2 , or OW
  • X is H, F, Cl, Br, OH, OW, NO 2 or NH 2
  • Y is H, F, Cl, or Br
  • W is C(O)Z or a C 1 to C 12 alkyl group
  • Z is an aliphatic or aromatic group of from C 1 to C 12
  • X and Y can both vary within the molecule; or its pharmaceutically acceptable salt.

Abstract

The present invention are novel compounds of formula (I), wherein R is OH, NH2, OW, or H; X is H, F, Cl, Br, I, OH, OW, NO2 or NH2; Y is H, F, Cl, I, or Br; W is C(O)Z or a C1 to C12 alkyl group; Z is an aliphatic or aromatic group of from C1 to C12; X and Y can both vary within the molecule; and if R is H, at least one of X or Y is not H. In a preferred embodiment, R is OH or NH2. The most preferred compound is (4-hydroxy-3-N-phenylacetylamino-2,6-piperidinedione), in which R is OH, X is H and Y is H. These compounds have cytostatic activity and insert stereochemically into DNA.

Description

SYNTHETIC PIPERIDINEDIONES WITH CYTOSTATIC ACTIVITY
BACKGROUND OF THE INVENTION
This invention is in the area of synthetic organic chemistry, and specifically relates to new
piperidinedione derivatives with cytostatic
properties.
A tumor is an unregulated, disorganized
proliferation of cell growth. A tumor is malignant, or cancerous, if it has the properties of invasiveness and metastasis. Invasiveness refers to the tendency of a tumor to enter surrounding tissue, breaking through the basal laminas that define the boundaries of the tissues, thereby often entering the body's circulatory system. Metastasis refers to the tendency of a tumor to migrate to other areas of the body and establish areas of proliferation away from the site of initial appearance.
Cancer is now the second leading cause of death in the United States, Europe, and Japan, resulting in approximately 1,000,000 deaths annually in these countries. In the United States alone, each year over one million people are diagnosed with cancer, and over 500,000 people die from the disease. The number of newly diagnosed cancerous growths in patients in the United States is growing at a rate of 3% a year.
Cancer is not fully understood on the molecular level. It is known that exposure of a cell to a carcinogen such as certain viruses, certain chemicals, or radiation, leads to DNA alteration that activates an "oncogene." Oncogenes are initially normal genes (called prooncogenes) that by mutation or altered context of expression become transforming genes. The products of transforming genes cause inappropriate cell growth. More than twenty different normal cellular genes can become oncogenes by genetic
alteration. Transformed cells differ from normal cells in many ways, including cell morphology, cell- to-cell interactions, membrane content, cytoskeletal structure, protein secretion, gene expression and mortality (transformed cells can grow indefinitely).
All of the various cell types of the body can be transformed into malignant cells. The most freguent tumor site is lung, followed by colorectal, breast, prostate, bladder, pancreas, and then ovary.
Cancer is now treated with one or a combination of three types of therapies: surgery, radiation, and chemotherapy. Surgery involves the bulk removal of diseased tissue. While surgery is sometimes effective in removing tumors located at certain sites, for example, in the breast, colon, and skin, it cannot be used in the treatment of tumors located in other areas, such as the backbone, nor in the treatment of disseminated neoplastic conditions such as leukemia.
Chemotherapy now represents less than 4% of the total expenditures on the treatment of cancer.
Chemotherapy involves the disruption of cell
replication or cell metabolism. It is used most often in the treatment of leukemia and breast, lung, and testicular cancer.
There are four major classes of chemotherapeutic agents currently in use for the treatment of cancer; anthracyclines, alkylating agents, antiproliferatives, and hormonal agents. A variety of methods exist to attempt to identify new antineoplastic
chemotherapeutic agents, including random screening of compounds, preparation of analogs of active compounds, computer or physical modeling, and combinations of these technigues. None of these methods, however, have yet identified the optimal chemotherapeutic agent for neoplastic diseases. U.S. Patent No. 4,461,619 to Hendry et al., discloses a method to determine the relationship of chemical structure to biological activity based on the topology and physicochemical properties of "cavities" or "artificial constructs" in double stranded DNA, double stranded RNA, or double stranded DNA-RNA. While modeling can be very helpful in chemotherapy research, it cannot predict whether a target compound will pass through the cell wall, whether it is stable in vivo generally or in the cytoplasm specifically, or whether the therapeutic index is appropriate for clinical use of the drug.
Burzynski has proposed that the human organism is equipped with a corrective system that can reprogram the growth of newly developed neoplastic cells to transform them back into normal cells. He has
isolated a number of medium sized peptides, referred to as antineoplastons, that are produced by the body to protect it against the development of neoplastic growth by a nonimmunological process that does not significantly inhibit the growth of normal tissue.
The most potent antineoplaston isolated by Burzynski is 3-[N-phenylacetylaminopiperidine]-2,6-dione
(referred to below as A10). Antineoplastons are described in U.S. Patent No. 4,444,890, entitled
"Testing Procedure to Aid Diagnosis of Cancer and Evaluate the Progress of Cancer Therapy"; U.S. Patent No. 4,593,038, entitled "Topical Use of 3- Phenylacetylamino-2,6-Piperidinedione for Treatment of Skin Wrinkles and Hyperpigmentation"; and U.S. Patent Nos. 4,558,057, 4,559,325 and 4,470,970, entitled "Purified Antineoplaston Fractions and Methods of Treating Neoplastic Disease." According to these patents, administration of antineoplastons to cancer patients has resulted in symptomatic improvement in 93% of the patients treated. A remission of the tumor was noted in about 45% of the patients.
The initial hydrolysis product and biological degradation product of A10 is phenylacetylglutamine, which is produced in vivo from phenylacetic acid and glutamine. In fact, A10 may be cyclized from
phenylacetylglutamine in vivo. Markaverich, et al., report that a compound structurally related to
phenylacetic acid, methyl p-hydroxyphenylacetate, inhibits MCF-7 human breast cancer cells in vitro. Markaverich, et al., J. of Biol. Chem. 263(15), 7203 (1988).
Hendry has shown that A10 fits in a stereochemical manner between base pairs of double stranded DNA.
Hendry, L.B., et al., "Modeling Studies Suggest the Modified Dipeptide Analog Phenylacetylamino-2,6- piperidinedione may interact with DNA, " Advances in Experimental and Clinical Chemotherapy. 15th
International Congress of Chemotherapy, Istanbul, Turkey, 1987. Specifically, A10 is capable of forming a stereospecific hydrogen bond between the imino proton of the piperidinedione ring and the phosphate oxygen on the DNA backbone. A10 does not bind covalently to DNA, which may explain why the compound is cytostatic and not cytotoxic. The acute toxicity of A10 in mice is between 1.35 and 10.33 g/kg. A10 has been administered without serious side effects at a dosage of up to 10 grams per day to humans suffering from cancer. While A10 is a useful drug in the treatment of neoplastic diseases, there is a need for new
cytostatic agents that may be more effective in stimulating tumor remission, and that may be effective when administered in lower dosages. The tragic number of deaths that occur each year from this disease accentuates the urgency of this need.
In light of the above, it is clear that there is a strong need for new cytostatic agents that can
effectively insert stereochemically into DNA.
Therefore, it is an object of the present
invention to provide a compound that has a cytostatic effect on cancer cells.
It is a further object of the present invention to provide a compound that can insert stereochemically into DNA.
SUMMARY OF THE INVENTION The present invention is a compound with
cytostatic activity of the formula:
Figure imgf000007_0001
wherein R is OH, NH2, OW, or H; X is H, F, Cl, Br, I, OH, OW, NO2 or NH2; Y is H, F, Cl, I, or Br; W is C(O)Z or a C1 to C12 alkyl group; Z is an aliphatic or
aromatic group of from C1 to C12; X and Y can both vary within the molecule; and if R is H, at least one of X or Y is not H. In a preferred embodiment, R is OH or NH2. The most preferred compound is (4-hydroxy-3-N- phenylacetylamino-2,6-piperidinedione), in which R is OH, X is H and Y is H.
These compounds exhibit cytostatic activity and inhibit the uptake of radioactive thymidine in a variety of neoplastic cell lines. The preferred compound, 4-hydroxy-3-N-phenylacetylamino-2, 6- piperidinedione, has significant cytostatic activity against prolactin stimulated Nb2 lymphoma cells (a T cell derived lymphoma), MCF-7 cells (estrogen
sensitive cells), and mouse lymphoma (YAK) cells.
"Cytostatic activity" as used herein refers to the ability of a compound to inhibit cell growth or replication. In contrast, "cytotoxic activity" refers to the ability of a compound to kill cells.
Cytostatic agents typically cause reversible chemical changes in cells. An example of a reversible change is the formation of ionic bonds or hydrogen bonding between the cytostatic agent and molecules in the cells. Cytotoxic compounds typically cause
irreversible changes in cells. An example of an irreversible change is the formation of a covalent bond.
Modeling studies suggest that 4-hydroxy-3-N- phenylacetylamino-2,6-piperidinedione (referred to below as p-OH-A10) fits easily between base pairs of DNA to form a complex with significantly lower energy than either structure alone. It has now been
discovered that the compounds described herein in which R is a proton donating group such as hydroxyl or amino have significantly greater cytostatic effect than A10. Further, it has been discovered that the preferred compounds of this invention are capable of binding to both strands of the DNA, whereas A10 is only capable of binding to one of the strands of DNA. Thymidine uptake studies indicate that the compounds inhibit DNA synthesis.
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is a schematic illustrating a method of preparation of the cytostatic piperidinediones of the present invention. The aryl groups in the
piperidinedione compounds are abbreviated as follows: a, 4-HO-C6H4; b, 2-Cl,6-F-C6H3; C, 2-F-C6H4; d, 3-F-C6H4; e, 4-F-C6H4; f, 2,6-di-F-C6H3; g, 4-CH3O-C6H4; h, 4-Cl- C6H4; i, 4-Br-C6H4; j, 4-CF3-C6H4. The following
reagents were used: step (i), N,N- dicyclohexylcarbodiimide (DCC) in acetonitrile; step (ii), NaHCO3 in acetonitrile/H2O (2/1 ratio); step (iii), DCC in dimethylformamide (DMF); and step (iv), DMF.
Figure 2 is a bar chart graph indicating the inhibition of prolactin stimulated Nb2 lymphoma cell growth by cytostatic piperidinediones. Equivalent amounts (1 mg/ml) of A10 or A10 analogs were
introduced simultaneously with prolactin (PRL) (0.4 ng/ml) to quiescent Nb2 lymphoma cells. Cell counts were made 48 hours later. The following abbreviations were used for the compounds tested: HQH-1-45-28, 3-[N- -chloro, 6-fluorophenylacetylaminopiperidine]-2,6- dione; HQH-1-44-11, 3-[N-2- fluorophenylacetylaminopiperidine]-2,6-dione; HQH-1- 41-27, 3-[N-2,6-difluorophenylacetylaminopiperidine]- 2,6-dione; HQH-1-36-36, 3-[N-4- hydroxyphenylacetylaminopiperidine]-2,6-dione; HQH-1- 51-29, 3-[N-4-fluorophenylacetylaminopiperidine]-2,6- dione ; and HQH-2-48-30 , 3- [N-3- fluorophenylacetylaminopiperidine]-2,6-dione. The following is the legend for Figure 2:
Figure imgf000010_0002
= Control;
PRL;
Figure imgf000010_0006
= A10; = HQH-1-45-28;
Figure imgf000010_0003
= HQH-1-44-11; = HQH-1-41-27;
Figure imgf000010_0005
= HQH-1-36-36;
Figure imgf000010_0004
= HQH-1-51-29; and
Figure imgf000010_0007
= HQH-2-48-30.
Figure 3 is a graph indicating the inhibitory effect of 3-[N-4-hydroxyphenylacetylaminopiperidine]-
2,6-dione (A10) and 3-[N-phenylacetylaminopiperidine]- 2,6-dione (p-OH-A10) on MCF-7 cells growing in log phase. The following is the legend for Figure 3:
Figure imgf000010_0008
-control;
Figure imgf000010_0009
= A10; and
Figure imgf000010_0010
= pOH-A10.
Figure 4 is a bar chart graph indicating the inhibition of mouse lymphoma (YAK) cell proliferation by the cytostatic piperidinediones of the present invention (PAG = phenylacetylglutamine; PA =
phenylacetic acid). The compound numbers are as indicated in Figure 2. DETAILED DESCRIPTION OF THE INVENTION
The present invention is a compound with
cytostatic activity of the formula:
Figure imgf000010_0001
wherein R is OH, NH2, OW, or H; X is H, F, Cl, Br, I,
OH, OW, NO2 or NH2; Y is H, F, Cl, I, or Br; W is C(O)Z or a C1 to C12 alkyl group; Z is an aliphatic or aromatic group of from C1 to C12; X and Y can both vary within the molecule; and if R is H, at least one of X or Y is not H. Active compounds include 3-[N-4- hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-4- hydroxy-3-fluorophenylacetylaminopiperidine]-2,6- dione, 3-[N-4-hydroxy-3- chlorophenylacetylaminopiperidine]-2,6-dione, 3-[N-
3,4-dihydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-3-amino-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-aminophenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-methoxy-3- hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-4- amino-3-fluorophenylacetylaminopiperidine]-2,6-dione, 3-[N-2-fluoro-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione, 3-[N-2-fluorophenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-acyloxyphenylacetylaminopiperidine]- 2,6-dione, and 3-[N-4- alkoxyphenylacetylaminopiperidine]-2,6-dione.
In a preferred embodiment, the active compound has a proton donating group (such as hydroxyl or amino) in the 4-position of the aromatic ring and an electron withdrawing or proton donating group in the 3-position of the aromatic ring. The 2-position of the aromatic ring is preferably unsubstituted (Y=H) or substituted with a small group that does not hinder free rotation of the aromatic ring.
These compounds are synthetic derivatives of the naturally occurring antineoplaston, A10. It has been demonstrated through thymidine incorporation studies that A10 significantly inhibits DNA synthesis (16% at 20 hours; 30% at 40 hours, p<0.01). Modeling has indicated that A10 can insert into DNA and form a stabilizing hydrogen bond through its imide group to a phosphate oxygen on one strand of the double stranded DNA.
It has been now been established through thymidine studies that the compounds described herein also significantly inhibit DNA synthesis. Further, it has been discovered that the novel compounds described herein that have a proton donating group, such as hydroxyl or amino, in the 4-position of the aromatic ring have significantly more cytostatic activity than those piperidinedione compounds without proton
donating groups in these positions. It is postulated that the increased activity is due to the ability of the proton donating group at the 4-position of the molecule to form a stabilizing stereospecific hydrogen bond with a phosphate oxygen on the nucleic acid strand opposite to that hydrogen bonded to the imide group of the synthetic piperidinedione. These active compounds thus appear to be capable of securing themselves to both strands of DNA in the double helix, resulting in a complex of significantly lower energy than a complex of double stranded DNA with an
piperidinedione that is capable of hydrogen bonding to only one strand of the helix, such as A10. As a result, these compounds are expected to have greater therapeutic efficacy and lower toxicity than A10.
Cytostatic agents are useful to moderate the growth of proliferative cells such as neoplastic cells in cell culture or animal models for purposes of research in the area of proliferative diseases.
Cytostatic agents are also useful in the study of the bonding patterns and consequent activity of
ribonucleic acids. Cytostatic agents may also have a pharmaceutical use as antineoplastic agents in vivo.
The present invention will be further understood with reference to the following non-limiting examples describing the synthesis, activity, and preparation of pharmaceutical compositions of these compounds. I. Method of Preparation of Cytostatic
Piperidinediones The cytostatic piperidinedione derivatives can be prepared by condensation of the appropriate
phenylacetic acid derivative with L-glutamine to produce the corresponding phenylacetylglutamine derivative, that is then intramolecularly cyclized to the desired 3-(N-phenylacetylamino)-2,6- piperidinedione. The condensation reaction is
facilitated by prior activation of the phenylacetic acid derivative with a reagent such as N- hydroxysuccinimide in the presence of DCC (N,N- dicyclohexylcarbodiimide), 2-mercaptothiazoline in the presence of DCC, or DCC alone. The
phenylacetylglutamine derivative is also preferably activated before cyclization by reaction with N- hydroxysuccinimide in the presence of DCC or by reaction with 1,1'-carbonyldiimidazole. These
reactions are described in more detail in Burzynski, Drugs of the Future. 10(2), 1003 (1985).
Desired derivatives of phenylacetic acid can be purchased commercially or prepared synthetically by methods known to those skilled in the art according to well established rules of electrophilic and
nucleophilic aromatic substitution. For example, 4- hydroxyphenylacetic acid, which is commercially available from Aldrich Chemical Company, Inc., can be nitrated with dilute HNO3 to produce 4-hydroxy-3- nitrophenylacetic acid, that is used as is in the next step of reaction. Alternatively, the nitro group in 4-hydroxy-3-nitrophenylacetic acid can be reduced to the corresponding amine and then reacted with sodium nitrite in acid to form the diazonium salt, that can be converted into a wide range of functional groups, including chloro (CuCl), fluoro (HBF4), bromo (CuBr) and hydroxyl (H2SO4). Phenylacetic acid can
alternatively be nitrated in the 4-position to produce 4-nitrophenylacetic acid, that is used as is in the reaction or converted to the diazonium salt and derivatized. The nitro group can be reduced to the corresponding amino group as a final step of reaction by methods known to those skilled in the art,
including catalytic hydrogenation with palladium on carbon.
Prodrugs of the hydroxyl or amino derivatives of 3-N-phenylacetylamino-2,6-piperidinedione can be prepared by alkylation or acylation of the hydroxyl or amino moieties according to established methods.
These protecting groups can be cleaved in vivo or in vitro by the appropriate enzyme, generating the active compound.
In Figure 1, a general reaction scheme is
illustrated for the preparation of the cytostatic piperidinediones. As shown, the substituted
phenylacetic acids 1 were reacted with N- hydroxysuccinimide to produce the corresponding N- hydroxysuccinimide esters 2, that were reacted with L- glutamine to form the phenylacetylglutamine
derivatives 3. Compound 3 was then reacted with N- hydroxysuccinimide and cyclized to form the
substituted 3-N-phenylacetylamino-2,6-piperidinedione derivatives 5. Detailed experimental procedures are provided in Examples 1-4. (The numbering scheme for the compounds described in these Examples is specified in Figure 1.) Physical and NMR data for the N- hydroxysuccinimide esters of substituted phenylacetic acids are provided in Tables 1 and 2, respectively. NMR data for the substituted phenylacetyl-L-glutamines and substituted-3-N-phenylacetylamino-2,6- piperidinediones are provided in Tables 3 and 4, respectively.
In the following working examples, melting points were determined on a Thomas Hoover capillary apparatus and are uncorrected. 1N NMR spectra were recorded on a JEOL FX 90Q fourier transform spectrometer for the 90-MHz 1H NMR spectra, using Me4Si as internal
standard: chemical shifts are reported in parts per million (δ) and signals are quoted as s (singlet), d
(doublet), t (triplet), q (quartet), or m (multiplet). UV spectra were obtained on a Beckman DU-7
spectrophotometer. Thin layer chromatography was performed on Uniplates (silica gel) purchased from Analtech Co. Elemental analyses were performed by Atlantic Microlab Inc., Norcross, GA or Galbrait
Laboratories, Inc., Knoxville, TN.
Figure imgf000017_0001
Figure imgf000018_0001
2
Figure imgf000019_0001
Figure imgf000020_0001
*
Figure imgf000021_0001
Example 1: General Procedure for the Preparation of N-Hydroxysuccimide Esters of substituted Phenylacetic Acids.
The following is a detailed description for the preparation of the N-hydroxysuccinimide ester of 2- chloro-6-fluorophenylacetic acid 2b. The compounds
2a, 2c-2j were prepared as described below from the appropriate substituted phenylacetic acids (1a, 1c- 1j), respectively.
A solution of N-hydroxysuccinimide (5 g, 42 mmol) in 30 mL of anhydrous acetonitrile (dried over 4 Å molecular sieve) was mixed with a solution of 2- chloro-6-fluoro-phenylacetic acid (1b, 7.92 g, 42 mmol, purchased from Aldrich Chemical Company, Inc., in 50 mL of anhydrous acetonitrile. The clear
solution was cooled in an ice bath and N,N- dicyclohexylcarbodiimide (8.66 g, 42 mmol) in 50 mL of anhydrous acetonitrile was added to the mixture with stirring. The reaction mixture was stirred at ambient temperature for 23 hours, until thin layer
chromatography indicated the disappearance of the starting material (ethyl acetate/hexanes, 1/1). The colorless precipitate obtained (dicyclohexylurea) was filtered off. The filter cake was washed with
acetonitrile and the combined filtrate was then evaporated under vacuum to give a colorless solid (2b, 12.57 g) that was used for next reaction without further purification. Analytical samples were
prepared by preparative thin layer chromatography using ethyl acetate/hexanes (1/1). Example 2: General Procedure for the Preparation of Substituted Phenylacetyl-L-Glutamines. To a solution of the N-hydroxysuccimide ester of 2-chloro-6-fluoro-phenyl acetic acid (2b, 12.5 g, 42 mmol) in 120 mL of acetonitrile was added a solution of L-glutamine (6.14 g, 42 mmol) in a mixture of 170 mL of water and 340 mL of acetonitrile containing sodium bicarbonate (7 g, 84 mmol). The mixture was stirred at ambient temperature for 24 hours. The organic layer was separated, and the aqueous layer then extracted with 100 mL acetonitrile. The combined organic layer was evaporated under vacuum to give 3b (15.3 g) as colorless solid. A small amount of the sample was purified by thin layer chromatography
(chloroform/methanol, 10.1) for spectroscopic
characterization.
The compounds of 3a, 3e-3j were prepared as described above from 2a, 2c-2j, respectively.
Example 3: General Procedure for the Preparation of
N-Hydroxysuccinimide Esters of
(Substituted Phenylacetyl)-L-Glutamine.
A solution of N-hydroxysuccinimide (5 g, 42 mmol) in 50 mL of anhydrous dimethylformamide (dried over 4Å molecular sieve) was added to the solution of 2- chloro-6-fluoro-phenylacetyl-L-glutamine (3b, 15.3 g, 42 mmol) in 300 mL of anhydrous DMF. N,N- Dicyclohexylcarbodiimide (8.66 g, 42 mmol) was added to the mixture with stirring. The mixture was stirred at 80°C for 6 hours and then stirred at ambient temperature for 18 hours. The colorless precipitate obtained was filtered off and the filtrate then used directly in the procedure of Example 4 without
isolation of the product. A small amount of sample (4b) was purified by thin layer chromatography
(chloroform.methanol, 1/1) for spectroscopic
characterization.
The compounds 4a, 4c-4j were prepared at room temperature as described above from 3a, 3c-3j,
respectively. Example 4: General Procedure for the Preparation of
Substituted-3-N-Phenylacetylamino-2,6- Piperidinedione. The filtrate obtained in Example 3 (4b) was heated at 95-100°C for 6 hours with stirring, during which time a colorless precipitate formed. The mixture was cooled and the precipitate filtered off. The filtrate was stored in the refrigerator overnight. Needle-like crystals that formed were filtered off. The filtrate was then concentrated under vacuum at 60°C to give a syrup that was recrystallized from hot methanol to provide 5b as a colorless crystal (5.3 g, overall yield 42.5%).
Compounds 5c-5j were prepared as described above from 4c-4j, respectively. However, 5a was purified by a vacuum fresh column chromatography of the syrup using chloroform/methanol (10/1) as the eluent, to get a pale yellow crystal.
II. Activity of Cytostatic Piperidinediones
The cytostatic activities of the active
piperidinediones were tested in three cell lines:
prolactin sensitive cells (Nb2 lymphoma cells), estrogen sensitive cells (MCF-7), and cells that are not hormone sensitive (YAK mouse lymphoma cells). In all three cell lines, the preferred compound, p-OH- A10, dramatically inhibited cell growth in a dose- dependent fashion. The other piperidinedione
derivatives also significantly inhibited cell growth as a function of concentration.
Example 5: Inhibition of Prolactin Stimulated Nb2
Lymphoma Cells
The rat Nb2 lymphoma cell line, a T-cell derived lymphoma, has a specific requirement of lactogenic hormones for growth and provides a useful system to determine the effect of the active compounds on hormone-dependent neoplasia. When Nb2 lymphoma cells are cultured in media containing fetal calf serum (FCS) and horse serum (HS), they demonstrate a
doubling time of approximately 15 hours. If, however, the cells are cultured in media devoid of FCS
(lactogen source), they enter into a quiescent state of growth. Upon addition of exogenous prolactin
(PRL), the cells resume proliferation in a dose- dependent manner. Additionally, it has been
demonstrated that interleukin-2 (IL-2) and phorbol esters have mitogenic effects in this cell line.
Antineoplaston A10 (3-phenylacetylamino-2,6- piperidinedione) was a generous gift from the
Burzynski Research Institute (Stafford, TX). The A10 analogs were obtained by the syixthetic scheme illustrated in Figure 1. Samples of A10 were analyzed for chemical composition, purity and stability.
Analyses were routinely performed on sample solutions dissolved in dimethylformamide using a Finnigan 4023 computerized gas chromatograph-mass spectrometer under the following conditions: injector, 300°C; source, 300°C; oven, 265°C isothermal; DB5 30 M capillary column; electron energy, 70 ev; scan range, 35-450 amu. In each case, the results showed a single chromatographic peak with the same electron impact mass spectrum. The mass spectral fragmentation pattern was consistent with 3-phenylacetylamino-2,6- piperidinedione (A10) (m/e 246, M+; 155, M+ -C7H7; 127; 118; 110; 99; 91, C7H7+; 84; 65; 56). While the mass spectrum of A10 was not present in the computer library, a search revealed an expected close match for the hydrolysis product of A10, 3-N- phenylacetylglutamine. No discernable impurities were detected using reconstructed ion chromatograms and the ion chromatograms of the major diagnostic ions (m/e 246, 155, and 91) possessed single superimposable chromatographic peaks. The retention times and mass spectra obtained in all cases were consistent with those previously reported for A10 as well as those obtained from authentic samples.
Penicillin/streptomycin, FCS, and Fischer's media were obtained from Gibco Laboratories (Grand Island, NY). HS was purchased from MA Bioproducts
(Walkersville, MD). Rat prolactin (rPRL-RP3) was supplied by NIAMDD of the National Institute of
Health. Tissue culture flasks (75 cm2), 24- and 96- well plates were obtained from Fisher (Pittsburgh, PA). Nb2 rat lymphoma cells were maintained in
suspension cultures in 75 cm2 tissue culture flasks in Fischer's media supplemented with 5% horse serum (HS), 5% fetal calf serum (FCS), 10-4M 2-mercaptoethanol, 50 units/ml penicillin, 50 μg/ml streptomycin, in an atmosphere of 5% CO2; 95% air at 37°C.
The proliferation of NB2 lymphoma cells was determined by the following procedure. The cells were centrifuged (300 x g, 4 minutes), washed three times in media containing only HS, and then resuspended in the same media. Cells were then cultured for 24 hours prior to use, at which time prolactin and test
compound were added. For cell counting, 100 μl aliquots of cells were added to 10 ml of isoton
(Coulter Counter Electronics); the cell number was determined in a Coulter counter (model ZM). All experiments were performed in triplicate.
The final concentration of test compound in the culture system was 1 mg/ml. Approximately 5 mg of each of the compounds were weighed and dissolved in heated DMSO (40°C) to give an equivalent concentration of 200 mg/ml. An equal volume of heated 95% ethanol was added to each compound to give a 100 mg/ml
equivalent followed by a 1:50 dilution in Fischer's media containing 5% horse serum, pen/strp, and 2- mercaptoethanol (A10 and analog concentration - 2 mg/ml). The least soluble compound was the parent A10. It was noted that p-OH-A10 had a yellow
appearance. A volume of 400 μl of cells containing solvent were added. Following a 48 hour culture, cell counts were made.
A comparison of the inhibitory effects of the piperidinediones on PRL stimulation of proliferation in the NB2 lymphoma cell line (0.4 ng/ml) is provided in Figure 2. As illustrated, p-OH-A10 provided the greatest inhibition of lymphoma cell growth.
Compounds HQH-1-45-28 (3-[N-2-chloro,6- fluorophenylacetylaminopiperidine]-2,6-dione), HQH-1- 51-29 (3-[N-4-fluorophenylacetylaminopiperidine]-2,6- dione), and HQH-2-48-30, (3-[N-3- fluorophenylacetylaminopiperidine]-2,6-dione) also significantly inhibited proliferation of these cells. The 2-fluorophenyl-A10 derivative and the 2,2- difluorophenyl-A10 derivative provided some inhibition of cell growth.
Example 6: Inhibition of MCF-7 Cells MCF-7 cells were grown in 5% fetal calf serum and 10 μg insulin/ml in MEM supplemented media. The cells were trypsinized during log phase growth and plated in T25 flasks at a cell number of 20,000 cells in 2 ml media. The cells were allowed to attach during a period of 48-72 hours. The media was then changed.
The cell numbers were counted in control flasks and 1, 2.5, 5.0 and 10 nM of A-10 or p-OH A-10 or control medium was added to each flask. Each experiment was done in triplicate. The media was changed at the end of 3 days and new aliquots of A-10 or p-OH A-10 or control media were added. After 3 more days of cell growth, the cells were detached from the culture flasks by adding 2 ml of a solution of 0.05% trypsin and 0.02% EDTA. The cells were removed, and 20 ml isoton was added and counted in a Coulter Counter.
The results are provided in Table 5 and illustrated in Figure 3.
Figure imgf000029_0001
Figure 3 is a graph indicating the inhibitory effect of 3-[N-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione (p-OH A10) and 3-[N- phenylacetylaminopiperidine]-2,6-dione (A10) on MCF-7 cells growing in log phase. As illustrated, at concentrations above 1 millimolar, p-OH-A10 is highly active in this cell line. For example, in the absence of test compound, MCF-7 cells growing in log phase reached a count of approximately 80,000. When grown in log phase under the same conditions but in the presence of 5 millimolar of A10, the cell count was reduced to slightly less than 40,000. When grown under the same conditions in the presence of 5
millimolar of p-OH-A10, the cell count was reduced to less than 10,000. Example 7: Inhibition of Mouse Lymphoma (YAK) cells
Cells were grown in Dalbecco's modified Eagle's media in log phase. These cell grow in suspension. Cells (22,263) were plated and treated with 4 mM quantities of various compounds. Cell numbers were counted after 72 hours of treatment by removing 100 μl aliquots and diluting the aliquot with 20 ml isoton. The DMSO:ETOH concentration in all samples was 0.53%.
The results are provided in Table 6 and Figure 4.
Figure imgf000030_0001
Figure 4 is a bar chart graph indicating the inhibition of mouse lymphoma (YAK) cell proliferation by cytostatic piperidinediones. As illustrated, p-OH- A10 provides dramatic inhibition of the growth of these hormone insensitive cells. Other 3-[N- phenylacetylaminopiperidine]-2,6-dione derivatives, including the 4-fluorophenyl, the 2-fluorophenyl, the 3-fluorophenyl, and the 2-chloro,6-fluorophenyl derivatives also provided some inhibition of cell growth. IV. Preparation of Pharmaceutical Compositions and
Mode of Administration
As stated above, the cytostatic piperidinediones of the present invention are useful in the study of proliferative diseases in animal models and in vitro cell cultures. The active compounds may also have a use in the treatment of neoplastic diseases in vivo. Pharmaceutical compositions including these active compounds can be prepared as described below.
The active compound or its pharmaceutically acceptable salt is included in the pharmaceutically acceptable carrier or diluent in an amount sufficient to exert an inhibitory effect on the growth of the target neoplastic or proliferative cell line in vitro or in vivo. The active materials can be administered by any appropriate route, for example, orally, parenterally, intravenously, intradermally,
subcutaneously, intraperitoneally, or topically, in liquid or solid form.
The active compound is included in the
pharmaceutically acceptable carrier in an amount sufficient to exert a therapeutically useful
inhibitory effect on neoplastic or proliferative cells without serious toxic effect to healthy cells. By "inhibitory amount" is meant an amount of active ingredient sufficient to exert a cytostatic effect as measured by, for example, an assay such as that described in Examples 5 through 7, or measured by blood analysis or radiation analysis of the state of tumorigenesis in vivo.
The concentration of active compound in the drug composition will depend on absorption, inactivation, and excretion rates of the active compound as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. The active ingredient may be administered at once, or may be divided into a number of smaller doses to be administered at varying intervals of time.
If oral administration is desired, although not required, the compound may be provided in a
composition that protects it from the acidic
environment of the stomach. The compound can be orally administered in combination with an antacid formulation. The composition can also be administered in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine.
Oral compositions will generally include an inert diluent or an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules.
Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a
disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or
saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or other enteric agents.
The active compound or its pharmaceutically acceptable salt can be administered as a component of an elixir, suspension, syrup, wafer, chewing gum or the like. A syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes, colorings and flavors.
The active compounds can also be mixed with other active materials that do not impair the desired action, or with materials that supplement the desired action, including other cytostatic or anticancer compounds.
Solutions or suspensions used for parenteral, intradermal, subcutaneous, or topical application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens;
antioxidants such as ascorbic acid or sodium
bisulfite; chelating agents such as
ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. The parental preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
If administered intravenously, preferred carriers are physiological saline or phosphate buffered saline (PBS). In a preferred embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including
implants and microencapsulated delivery systems.
Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations are known to those skilled in the art. The materials can also be obtained commercially from Nova Pharmaceutical Corporation.
Liposomal suspensions are also preferred as pharmaceutically acceptable carriers. These may be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811 (which is incorporated herein by
reference in its entirety). For example, liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container. An aqueous solution of the active compound is then introduced into the container. The container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
Modifications and variations of the present invention, synthetic piperidinediones with cytostatic activity, will be obvious to those skilled in the art from the foregoing description. Such modifications and variations are intended to come within the scope of the appended claims.
We claim.
1. A compound of the formula:
Figure imgf000036_0001
wherein R is OH, NH2, or OW; X is H, F, Cl, Br, OH, OW, NO2 or NH2; Y is H, F, Cl, or Br; W is C(O)Z or a C1 to C12 alkyl group; Z is an aliphatic or aromatic group of from C1 to C12; X and Y can both vary within the molecule; or its pharmaceutically acceptable salt.
2. The compound of claim 1, wherein R is OH or NH2, and Y is H.
3. The compound of claim 1, wherein R is OH or NH2, X is OH or NH2 and Y is H.
4. The compound of claim 1, wherein R is OH or NH2, X is F, Cl, Br or I, and Y is H.
5. The compound of claim 1 selected from the group consisting of 3-[N-
4-hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N- 4-hydroxy-3-fluorophenylacetylaminopiperidine]-2,6- dione, 3-[N-4-hydroxy-3- chlorophenylacetylaminopiperidine]-2,6-dione, 3-[N- 3,4-dihydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-3-amino-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-aminophenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-methoxy-3- hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-4- amino-3-fluorophenylacetylaminopiperidine]-2,6-dione, 3-[N-2-fluoro-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione, 3-[N-2-fluorophenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-acyloxyphenylacetylaminopiperidine]- 2,6-dione and 3-[N-4- alkoxyphenoxyacetylaminopiperidine]2,6-dione.
6. The compound of claim 1, wherein R is OH, and X and Y are H.
7. The compound of claim 1 further comprising a pharmaceutical carrier.
8. The compound of claim 7 wherein the
pharmaceutical carrier is for topical application.
9. The compound of claim 7 wherein the
pharmaceutical carrier is for systemic administration.
10. The compound of claim 1 wherein the active agent is provided in a dosage that inhibits the growth of neoplastic cells.
11. The compound of claim 1 in a controlled delivery device.
12. The compound of claim 11 wherein the
controlled delivery device is selected from the group consisting of microcapsules, microspheres, and
polymeric matrices.
13. The compound of claim 6 further comprising a pharmaceutical carrier.
14. The compound of claim 13 wherein the
pharmaceutical carrier is for topical application.
15. The compound of claim 13 wherein the
pharmaceutical carrier is for systemic administration.
16. The compound of claim 6 wherein the active agent is provided in a dosage that inhibits the growth of neoplastic cells.
17. The compound of claim 6 in a controlled delivery device.
18. The compound of claim 17 wherein the
controlled delivery device is selected from the group consisting of microcapsules, microspheres, and
polymeric matrices. 19. A method of inhibiting cell growth comprising providing an effective cytostatic amount of a compound of the formula
Figure imgf000038_0001
wherein R is OH, NH2, or OW; X is H, F, Cl, Br, OH, OW, NO2 or NH2; Y is H, F, Cl, or Br; W is C(O)Z or a C1 to C12 alkyl group; Z is an aliphatic or aromatic group of from C1 to C12; X and Y can both vary within the molecule; or its pharmaceutically acceptable salt.
20. The method of claim 19 wherein the compound is selected from the group consisting of 3-[N- 4-hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N- 4-hydroxy-3-fluorophenylacetylaminopiperidine]-2,6- dione, 3-[N-4-hydroxy-3- chlorophenylacetylaminopiperidine]-2,6-dione, 3-[N- 3,4-dihydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-3-amino-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-aminophenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-methoxy-3- hydroxyphenylacetylaminopiperidine]-2,6-dione, 3-[N-4- amino-3-fluorophenylacetylaminopiperidine]-2,6-dione, 3-[N-2-fluoro-4-hydroxyphenylacetylaminopiperidine]- 2,6-dione, 3-[N-2-fluorophenylacetylaminopiperidine]- 2,6-dione, 3-[N-4-acyloxyphenylacetylaminopiperidine]- 2,6-dione and 3-[N-4- alkoxyphenoxyacetylaminopiperidine]2,6-dione.
PCT/US1991/002483 1990-04-12 1991-04-11 Synthetic piperidinediones with cytostatic activity WO1991016309A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US50883990A 1990-04-12 1990-04-12
US508,839 1990-04-12

Publications (1)

Publication Number Publication Date
WO1991016309A1 true WO1991016309A1 (en) 1991-10-31

Family

ID=24024294

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1991/002483 WO1991016309A1 (en) 1990-04-12 1991-04-11 Synthetic piperidinediones with cytostatic activity

Country Status (2)

Country Link
AU (1) AU7742491A (en)
WO (1) WO1991016309A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0740708A1 (en) * 1993-11-26 1996-11-06 Lawrence B. Hendry Design of drugs involving receptor-ligand-dna interactions
WO1997042939A1 (en) * 1996-05-14 1997-11-20 Burzynski Research Inst Liposomal antineoplaston therapies with markedly improved antineoplastic activity

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4423047A (en) * 1980-01-10 1983-12-27 Nyegaard & Co. A/S Pyrimidine-2-sulphides and their S-oxides for use in medicine and methods of use therefor, pharmaceutical compositions containing them, processes for their preparation and per se novel sulphides and S-oxides
US4444890A (en) * 1982-02-05 1984-04-24 Burzynski Stanislaw R Testing procedure to aid diagnosis of cancer and evaluate the progress of cancer therapy
US4461619A (en) * 1981-12-29 1984-07-24 Medical College Of Georgia Research Inst. Method of predicting biological activity of compounds by nucleic acid models
US4470970A (en) * 1981-07-02 1984-09-11 Burzynski Stanislaw R Purified antineoplaston fractions and methods of treating neoplastic disease
US4558057A (en) * 1981-12-15 1985-12-10 Burzynski Stanislaw R Purified antineoplaston fractions and methods of treating neoplastic disease
US4559325A (en) * 1981-12-15 1985-12-17 Burzynski Stanislaw R Purified antineoplaston fractions and methods of treating neoplastic disease
US4593038A (en) * 1985-04-03 1986-06-03 Burzynski Stanislaw R Topical use of 3-phenylacetylamino-2,6-piperidinedione for treatment of skin wrinkles and hyperpigmentation

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4423047A (en) * 1980-01-10 1983-12-27 Nyegaard & Co. A/S Pyrimidine-2-sulphides and their S-oxides for use in medicine and methods of use therefor, pharmaceutical compositions containing them, processes for their preparation and per se novel sulphides and S-oxides
US4470970A (en) * 1981-07-02 1984-09-11 Burzynski Stanislaw R Purified antineoplaston fractions and methods of treating neoplastic disease
US4558057A (en) * 1981-12-15 1985-12-10 Burzynski Stanislaw R Purified antineoplaston fractions and methods of treating neoplastic disease
US4559325A (en) * 1981-12-15 1985-12-17 Burzynski Stanislaw R Purified antineoplaston fractions and methods of treating neoplastic disease
US4461619A (en) * 1981-12-29 1984-07-24 Medical College Of Georgia Research Inst. Method of predicting biological activity of compounds by nucleic acid models
US4444890A (en) * 1982-02-05 1984-04-24 Burzynski Stanislaw R Testing procedure to aid diagnosis of cancer and evaluate the progress of cancer therapy
US4593038A (en) * 1985-04-03 1986-06-03 Burzynski Stanislaw R Topical use of 3-phenylacetylamino-2,6-piperidinedione for treatment of skin wrinkles and hyperpigmentation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Volume 105, No. 25, issued 22 December 1986, (Columbus, Ohio, U.S.A.), A.F. LEHNEN et al., "3-Phenylacetylamino-2,6-Piperidinedione, a Naturally Occurring Peptide Analog with Apparatus Antineoplastic Activity May Bind to DNA", see page 26-27, Abstract No. 218408x; & DRUGS EXP. CLIN. RES., 1986, 12 (Suppl. 1), 57-72, (Eng.). *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0740708A1 (en) * 1993-11-26 1996-11-06 Lawrence B. Hendry Design of drugs involving receptor-ligand-dna interactions
EP0740708A4 (en) * 1993-11-26 1999-12-01 Lawrence B Hendry Design of drugs involving receptor-ligand-dna interactions
WO1997042939A1 (en) * 1996-05-14 1997-11-20 Burzynski Research Inst Liposomal antineoplaston therapies with markedly improved antineoplastic activity
US6013278A (en) * 1996-05-14 2000-01-11 Burzynski Research Institute Liposomal antineoplaston therapies with markedly improved antineoplastic activity

Also Published As

Publication number Publication date
AU7742491A (en) 1991-11-11

Similar Documents

Publication Publication Date Title
EP1541554B1 (en) Crystal for oral solid drug and oral solid drug for dysuria treatment containing the same
WO2018075993A1 (en) Compounds and methods to sensitize cancer cells to tyrosine kinase inhibitors
JP2019528318A (en) Substituted quinazoline compounds with ability to cross the blood-brain barrier
KR20220107020A (en) Spiro compounds acting as ERK inhibitors and their applications
CN113336768B (en) Multi-target tyrosine kinase inhibitor
CN110452176A (en) Indazole analog derivative and its preparation method and application
US5238947A (en) Synthetic piperidinediones with cytostatic activity
CN112574191A (en) Isoxazole heterocyclic compound and application thereof
CN113336697A (en) CDK9 inhibiting compound and application thereof
US9399644B2 (en) [1,3] dioxolo [4,5-G] quinoline-6(5H)thione derivatives as inhibitors of the late SV40 factor (LSF) for use in treating cancer
CN108976159A (en) The indoles alcohol derivative of ArAA modification, synthesis, activity and application
HUT63843A (en) Process for producing new kumarin derivatives and their analogs inhibiting mammal cell proliferation and tumour growth, as well as pharmaceutical comkpositions comprising such compounds
WO1991016309A1 (en) Synthetic piperidinediones with cytostatic activity
JPH07502975A (en) Adenosine diphosphoribose polymerase-binding nitrosoaromatic compounds useful as antitumor and anti-retroviral agents
TW201922690A (en) Inhibitors of cyclic-AMP response element-binding protein
US4897423A (en) Dinitrobenzenesulfonamides
EP3279198B1 (en) Crystal form of n-[6-(cis form-2,6-dimethylmorpholine-4-group)pyridine-3- group]-2-methyl-4&#39;-(trifluoromethoxy)[1,1&#39;-biphenyl]-3- formamide monophosphate, and preparation method therefor
CN111138449B (en) Preparation of dual-targeting ERK1 and ERK5 inhibitors and anti-tumor application thereof
CN109438279B (en) Small molecule compound for overcoming EGFR drug-resistant mutation and preparation method and application thereof
CN112574061A (en) TRPV1 agonist and preparation method and application thereof
EP4273143A1 (en) Derivative of 2,5-diketopiperazine compound, and preparation method therefor, pharmaceutical composition thereof and use thereof
CN105745187B (en) For treating the compound of tumor disease
CN113845484B (en) Quinazoline small molecule inhibitor and application thereof in antitumor drugs
US20190062307A1 (en) Deuterium-substituted quinoline derivatives
RU2800042C1 (en) Spiro compounds as erk inhibitors and their use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU NL SE

NENP Non-entry into the national phase

Ref country code: CA