CA2436536C - Methods for inducing apoptosis and inhibiting proliferation in cancer cells - Google Patents

Methods for inducing apoptosis and inhibiting proliferation in cancer cells Download PDF

Info

Publication number
CA2436536C
CA2436536C CA002436536A CA2436536A CA2436536C CA 2436536 C CA2436536 C CA 2436536C CA 002436536 A CA002436536 A CA 002436536A CA 2436536 A CA2436536 A CA 2436536A CA 2436536 C CA2436536 C CA 2436536C
Authority
CA
Canada
Prior art keywords
compound
pharmaceutically acceptable
use according
cells
cancer cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
CA002436536A
Other languages
French (fr)
Other versions
CA2436536A1 (en
Inventor
Thomas E. Adrian
Peter Collin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Creighton University
Original Assignee
Creighton University
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 Creighton University filed Critical Creighton University
Publication of CA2436536A1 publication Critical patent/CA2436536A1/en
Application granted granted Critical
Publication of CA2436536C publication Critical patent/CA2436536C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/201Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids having one or two double bonds, e.g. oleic, linoleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

Disclosed are methods of decreasing proliferation of adenocarcinoma cells, or of inducing apoptosis of adenocarcinoma cells, or of inducing differentiation of adenocarcinoma cells into non-cancerous cells. The methods include contacting a sample comprising adenocarcinoma cells with a compound having the formula ("Formula I"): CH3-(CH2)n-CH=CH-(CH2)m-COOH) wherein n is an integer from 0 to 15, m is an integer from about 1 to 16, and the sum of m and n is an integer from 6 to 16, or a pharmaceutically acceptable ester, salt, amide, solvate, or metabolite thereof. Also disclosed are methods for treating adenocarcinoma in a subject. The methods include administering to the subject an effective amount of a compound having Formula I or a pharmaceutically acceptable ester, salt, amide, solvate, or metabolite thereof.

Description

WO 02I051406 PCTIUSU i149689 METHODS FOR INDUCING APOPTOSIS AND
INHIBITING PROLIFERATION IN CANCER CELLS
FIELD OF THE INVENTION

The subject invention is directed generally to methods for decreasing proliferation of cancer cells, or of inducing apoptosis of cancer cells, or of inducing differentiation of cancer cells into non-cancerous cells and to methods for treating adenocarcinoma in a subject.

BACKGROUND OF THE INVENTION

Pancreatic cancer is one of the most enigmat_c and aggressive malignant diseases facing oncologists (Parker et al., "Cancer Statistics. 1996," CA Cancer J.
Clin.., 46:5-27 (1996) ("Parker") ). it is now the fourth leading cause of cancer death in both men and women in the United States, and the incidence of this disease has significantly increased over the past 20 vears (Parker;
=rede et al., "Survival Afte~.~ Pancreaticoduodenectomy:
118 Consecutive Resections Without ar_ Operative Mortality," Ann. Sura., 211:4S7-458 (1990); Cameron et al., "One Hundred and Forty-five Consecutive Pancreatico-duodenectomies Without Mortality," Ann. Surg., 217:430-438 (1993) ; Horward, "Pancreatic Adenocarcinoma, Curr.
Prob. in Cancer, 20:286-293 (1996) ("Horward"); Poston et al., Gut. Biology of Pancreatic Cancer, 32:800-812 (1991) ("Poston"); and Black et al., "Treatment of Pancreatic Cancer: Current Limitations, Future Possibilities," Oncolociy, 10:301-307 (1996) ("Black")).
Pancreatic cancer is responsible for 27,000 deaths per year in the United States. Because of lack of early diagnosis and poor therapeutic responsiveness of pancreatic cancer, less than 2% of patients survive beyond five years, and the median expectation of life after diagnosis of pancreatic cancer is less than 6 months (Horward; Poston; and Black).
Colonic cancer is the second most common form of cancer in the United States (Doll et al., "Mortality in Relation to Smoking: 20 Years' Observations on Male British Doctors," BMJ, 2:1525-1536 (1976); Hruban et al., "Molecular Diagnosis of Cancer and Micrometastases,"
Adv. Anat. Pathol., 5:175-178 (1998) ("Hruban");
Figueredo et al., "Adjuvant Therapy for Stage II Colon Cancer After Complete Resection. Provincial Gastrointestinal Disease Site Group," Cancer Prev.
Control, 1:379-92 (1997) ("Figueredo"); Ness et al., "Outcome States of Colorectal Cancer: Identification and Description Using Patient Focus Groups," Am. J.
Gastroenterol., 93:1491-7 (1998) ("Ness"); Trehu et al., "Cost of Screening for Colorectal Cancer: Results of a Community Mass Screening Program and Review of Literature," South Med. J., 85:248-253 (1992); and Wingo et al., "Cancer Statistics," CA Cancer J. Clin., 45:8-30 (1995) ("Wi.ngo")). Colonic cancer occurs in more than 138,000 patients and is responsible for more than 55,000 deaths in the United States each year (Wingo). Up to 70 % of patients with colonic cancer develop hepatic metastasises by the time of death, indicating that non-detectable micro-metastases are present at the time of surgery (Hruban; Figueredo; and Ness). Furthermore, metastatic cancer is often not responsive to standard chemotherapeutic regimens, resulting in treatment failure (Figueredo and Ness). The overall response of advanced or non-resectable colorectal cancer patients to chemotherapeutic agents varies from 26 to 44 percent.
For example, less than one third of colorectal cancer patients with liver metastases respond to treatment with agents such as 5-FU and leucovorin (Id.).
Breast cancer has the highest incidence of any cancer in women with the diagnosis being made in more than 275,000 per year in the USA (Richards et al., "Influence of Delay on Survival in Patients with Breast Cancer: A Systematic Review," Lancet, 353:1119-26 (1999);
Norton, "Adjuvant Breast Cancer Therapy: Current Status and Future Strategies -- Growth Kinetics and the Improved Drug Therapy of Breast Cancer," Semin. Oncol., 26:1-4 (1999); Morrow et al., "Current Controversies in Breast Cancer Management," Curr. Probl. Surg., 36:163-216 (1999); and Ruppert et al., "Gene Therapy Strategies for Carcinoma of the Breast," Breast Cancer Res. Treatment, 44:93-114 (1997)). Even though five year survival has increased to more than 80%, more than 77,000 women still die from this disease each year (Id.).
Thus, another dimension in chemotherapeutic agents for pancreatic, colonic, and breast cancer would be extremely beneficial, especially to control metastatic and unresectable disease.
SUNIMARY OF THE INVENTION

The present invention relates to a method of decreasing proliferation of adenocarcinoma cells, or of inducing apoptosis of adenocarcinoma cells, or of inducing differentiation of adenocarcinoma cells into non-cancerous cells. The method includes contacting a sample comprising adenocarcinoma cells with a compound having the formula:
CH3 -( CH2 ) n- CH=CH- ( CH2 ) m- COOH

wherein n is an integer from 0 to 15, m is an integer from about 1 to 16, and the sum of m and n is an integer from 6 to 16, or a pharmaceutically acceptable ester, salt, amide, solvate, or metabolite thereof.
The present invention also relates to a method of treating adenocarcinoma in a subject. The method includes administering to the subject an effective amount of a compound having the formula:

CH3 -( CH2) n- CH=CH- ( CH2 ) R,- COOH

wherein n is an integer from 0 to 15, m is an integer from about 1 to 16, and the sum of m and n is an integer from 6 to 16, or a pharmaceutically acceptable ester, salt, amide, solvate, or metabolite thereof.

BRIEF DESCRIPTION OF THE DRAWINGS
Figures 1A and 1B are bar graphs showing the effects of myristoleic acid.obtained from Sigma Chemicals (St. Louis, Missouri) (Figure lA) and myristoleic acid obtained from Matreya, Inc. (State College, Pennsylvania) (Figure 1B) on the proliferation of AsPC-1 human pancreatic cancer cells.
Figures 2A and 2B are bar graphs showing the effects of impure cetyl myristoleic acid (Figure 2A) and pure palmitoleic acid (Figure 2B) on the proliferation of AsPC-1 human pancreatic cancer cells.
Figures 3A and 3B are bar graphs showing the effects of myristoleic acid on the proliferation of AsPC-1 (Figure 3A) and PANC-1 (Figure 3B) human pancreatic cancer cells.
Figure 4 is a set of four fluorescence micrograph images showing the effect of myristoleic acid on annexin V binding.
Figures 5A and 5B are dot plots showing TUNEL
assay results of AsPC-1 cells pancreatic cancer cells treated with 10 g/ml of myristoleic acid for 24 hours (Figure 5B) as compared to control (Figure 5A).
Figure 6 is an image of human pancreatic cancer xenografts in athymic mice which were treated with 250mg/kg/day of myristoleic acid (labeled "myristoleic acid") or control solution (labeled "control").
Figures 7A and 7B are graphs showing the effects of myristoleic acid on tumor volume of subcutaneous xenografts of AsPC-1 (Figure 7A) and HPAC
(Figure 7B) human pancreatic cancer cells in athymic mice as a function of time.
Figures 8A and 8B are bar graphs showing the effects of myristoleic acid on tumor weight of subcutaneous xenografts of AsPC-1 (Figure 8A) and HPAC
(Figure 8B) human pancreatic cancer cells in athymic mice at the end of the experiment.
Figures 9A-9D are images produced with an in situ TUNEL assay conducted on sections of AsPC-1 pancreatic cancer cell xenografts harvested from athymic mice that were treated with control solution (vehicle only) (Figures 9A and 9B) or myristoleic acid (Figures 9C
and 9D).
DETAILED DESCRIPTION OF THE INVENTION

The present invention relates to a method of decreasing proliferation of adenocarcinoma cells, or of inducing apoptosis of adenocarcinoma cells, or of inducing differentiation of adenocarcinoma cells into non-cancerous cells. The method includes contacting a sample comprising adenocarcinoma cells with a compound having the formula ("Formula I") CH3 -( CH2 ) n- CH= CH -( CH2 ) R, - COOH

wherein n is an integer from 0 to 15, m is an integer from about 1 to 16, and the sum of m and n is an integer from 6 to 16, or a pharmaceutically acceptable ester, salt, amide, solvate, or metabolite thereof.
Examples suitable compounds having Formula I
include those in which m is 1 and n is 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15; in which m is 2 and n is 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14; in which m is 3 and n is 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13; in which in which m is 4 and n is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12; in which m is 5 and n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11; m is 6 and n is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; m is 7 and n is 0, 1, 2, 3, 4, 5, 6, 7, 8, or 9; m is 8 and n is 0, 1, 2, 3, 4, 5, 6, 7, or 8; m is 9 and n is 0, 1, 2, 3, 4, 5, 6, or 7; m is 10 and n is 0, 1, 2, 3, 4, 5, or 6; m is 11 and n is 0, 1, 2, 3, 4, or 5; m is 12 and n is 0, 1, 2, 3, or 4; m is 13 and n is 0, 1, 2, or 3; m is 14 and n is 0, 1, or 2; m is 15 and n is 0 or 1; and m is 16 and n is 0. Preferably, when m is 7, n is 0, 1, 2, 3, 4, 5, 6, 8, or 9.
As indicated above, pharmaceutically acceptable base addition salts of the compounds of Formula I can also be used. Such salts include those derived from inorganic bases, such as ammonium and alkali and alkaline earth metal hydroxides, carbonates, bicarbonates, and the like, as well as salts derived from basic organic amines, such as aliphatic and aromatic amines, aliphatic diamines, hydroxy alkylamines, and the like. Such bases useful in preparing the salts useful in the practice of the present invention thus include ammonium hydroxide, potassium carbonate, sodium bicarbonate, calcium hydroxide, methyl amine, diethyl amine, ethylene diamine, cyclohexylamine, ethanolamine, and the like.
Pharmaceutically acceptable esters and amides of the compound of Formula I can also be employed in the method of the present invention. Examples of suitable esters include alkyl, aryl, and aralkyl esters, such as methyl esters, ethyl esters, propyl esters, dodecyl esters, benzyl esters, and the like. Examples of suitable amides include, unsubstituted amides, monosubstituted amides, and disubstituted amides, such as methyl amide, dimethyl aminde, methyl ethyl amide, and the like.
In addition, the method of the present invention can be practiced using solvate forms of the compounds of Formula I or salts, esters, amides, and/or metabolies thereof, such as ethanol solvates, hydrates, and the like.
It is recognized that the compounds of Formula I can be in the cis or trans configuration. The method of the present invention car: be practiced with pure cis isomer, pure trans isomer, a racemic mixture of cis and trar_s isomers, or any other mixture of cis and trans isomers.
Illustrative compounds which can be used in the practice of the method of the present invention include mvristoleic acid er a pharmaceutically acceptable salt, ester (e.g., a cetyl ester), amide, metabolite, and/or solvate thereof.
Compounds of Formula I can be synthesized bv established methods, such as those set forth in Beilstein 2(2) 423.
As explained above, the above-identified compounds can be used to decrease proliferation of adenocarcinoma cells, and/or induce apoptosis of adenocarcinoma cells, and/or induce differentiation of adenocarcinoma cells into non-cancerous cells. The meaning of the terms "prolzferation", "apoptosis", and "differentiation are readily understood in the art.
Illustrative methods for assa_ving for proliferation, apoptosis, or differentiation are nrovided in the examples which follow.

"Adenocarcinoma cells", as used herein, are meant to include cancerous epith.eliai cells, such as nrostate cancer cells, lung cancer cells, stomach cancer cells, breast cancer cells, pancreatic cancer cells, and colon cancer cells. The methods of the present invention can be practiced in viLro or in vivo.
More particularly, the method of the present invention can be used in vivo to treat adenocarcinomas, such as prostate cancer, 1-ung cancer, stomach cancer, pancreatic cancer, breast cancer, and colon cancer. In the case where the method of the present invention is carried out in vivo, for example, where the adenocarcinoma cells are present in a human subject, contacting can be carried out by administering a therapeutically effective amount of the compound to the human subject, for example, by directly injecting the compound into a tumor. Details with regard to administering compounds in accordance with the method of the present invention are described below.
The present invention, in another aspect thereof, relates to a method of treating adenocarcinomas, such as prostate cancer, lung cancer, stomach cancer, breast, pancreatic cancer, colon cancer, esophageal cancer, uterine cancer, ovarian cancer, or other cancers involving epithelial cells. The method includes administering, to the subject, a compound of Formula I or a pharmaceutically acceptable ester, salt, amide, solvate, or metabolite thereof.
Suitable subjects include, for example mammals, such as rats, mice, cats, dogs, monkeys, and humans.
Suitable human subjects include, for example, those which have previously been determined to be at risk of having prostate cancer, lung cancer, stomach cancer, pancreatic cancer, colon cancer, and/or breast cancer and those who have been diagnosed as having prostate cancer, lung cancer, stomach cancer, pancreatic cancer, colon cancer, and/or breast cancer. Preferably, the subject suffers from only one of these types of cancers, for example, from only pancreatic cancer.
In subjects who are determined to be at risk of having adenocarcinoma, the above-identified c.ompounds of Formula I or salts, esters, amides, solvates, and metabolites thereof are administered to the subject, preferably under conditions effective to decrease proliferation and/or induce apoptosis and/or induce differentiation of the adenocarcinoma cells in the event that they develop. Such preventive (which is not used in the absolute 100% sense) therapy can be useful in high risk individuals as long as the adverse side effects of the administration of these compounds are outweighed by the potential benefit of prevention.
Any of the compounds described above can be used in the treatment method of the present invention.
For example, compounds may be administered alone or in combination with compatible carriers as a composition.
Compatible carriers include suitable pharmaceutical carriers or diluents. The diluent or carrier ingredients should be selected so that they do not diminish the therapeutic effects of the compounds used in the present invention.
The compositions herein may be made up in any suitable form appropriate for the desired use. Examples of suitable dosage forms include oral, parenteral, or topical dosage forms.
Suitable dosage forms for oral use include tablets, dispersible powders, granules, capsules, suspensions, syrups, and elixirs. Inert diluents and carriers for tablets include, for example, calcium carbonate, sodium carbonate, lactose, and talc. Tablets may also contain granulating and disintegrating agents, such as starch and alginic acid; binding agents, such as starch, gelatin, and acacia; and lubricating agents, such as magnesium stearate, stearic acid, and talc. Tablets may be uncoated or may be coated by known techniques to delay disintegration and absorption. Inert diluents and carriers which may be used in capsules include, for example, calcium carbonate, calcium phosphate, and kaolin. Suspensions, syrups, and elixirs may contain conventional excipients, for example, methyl cellulose, tragacanth, sodium alginate; wetting agents, such as lecithin and polyoxyethylene stearate; and preservatives, such as ethyl-p-hydroxybenzoate.
Dosage forms suitable for parenteral administration include solutions, suspensions, dispersions, emulsions, and the like. They may also be manufactured in the form of sterile solid compositions which can be dissolved or suspended in sterile injectable medium immediately before use. They may contain suspending or dispersing agents known in the art.
Examples of parenteral administration are intraventricular, intracerebral, intramuscular, intravenous, intraperitoneal, rectal, and subcutaneous administration.
In addition to the above, generally non-active components of the above-described formulations, these formulations can include other active materials, particularly, actives which have been identified as useful in the treatment of prostate, lung, stomach, breast, colon, pancreatic cancers and/or other adenocarcinomas. These actives can be broad-based anti-cancer agents, such that they also are useful in treating other types of cancers (i.e., in addition to adenocarcinomas) or they may be more specific, for example, in the case where the other active is useful for treating adenocarcinomas or particular types of adenocarcinomas. The other actives can also have non-anti-cancer pharmacological properties in addition to their anti-adenocarcinoma properties. For example, the other actives can have anti-inflammatory properties, or, alternatively, they can have no such anti-inflammatory properties.
It is understood that some of the compounds described above (i.e., some of the compounds that are useful in the methods of the present invention) are naturally occurring. The compositions used in the treatment method of the present invention can be substantially free of one or more of the components with which the compound is typically found when it is naturally occurring. Alternatively or additionally, the compositions used in the treatment method of the present invention can be substantially free of all but one of the components with which the compound is typically found when it is naturally occurring. Still alternatively or additionally, the compositions used in the treatment method of the present invention can be substantially free of all of the components with which the compound is typically found when it is naturally occurring. For the purposes of the present application, a composition is considered to be substantially free of component X when the amount of component X is less than 10% by weight (such as less than 5% by weight, less than 2% by weight, and/or less than 1% by weight) relative to the weight of the composition.
It will be appreciated that the actual preferred amount of compound to be administered according to the present invention will vary according to the particular compound, the particular composition formulated, and the mode of administration. Many factors that may modify the action of the compound (e.g., body weight, sex, diet, time of administration, route of administration, rate of excretion, condition of the subject, drug combinations, and reaction sensitivities and severities) can be taken into account by those skilled in the art. Administration can be carried out continuously or periodically within the maximum tolerated dose. Optimal administration rates for a given set of conditions can be ascertained by those skilled in the art using conventional dosage administration tests.
The present invention is further illustrated with the following examples.

EXAMPLES
Example 1 -- Effect of Myristoleic Acid on Proliferation of Human Pancreatic Cancer Cells Myristoleic acid, obtained from two separate commercial sources, caused a concentration-dependent inhibition of proliferation of AsPC-1 human pancreatic cancer cells as measured by thymidine incorporatiori at 24 hours. This can be seen in Figure lA (results using myristoleic acid obtained from Sigma Chemicals (St.
Louis, Missouri)) and Figure 1B (results using myristoleic acid obtained from Matreya, Inc. (State College, Pennsylvania)).

Example 2 -- Effect of Cetyl Myristoleic Acids and Palmitoleic Acid on Proliferation of Human Pancreatic Cancer Cells Impure cetyl myristoleic acid (approximately 5%
pure) and pure palmitoleic acid also caused a concentration-dependent inhibition of proliferation of AsPC-1 human pancreatic cancer cells as measured by thymidine incorporation. The results are presented in Figure 2A (cetyl myristoleic acid) and Figure 2B
(palmitoleic acid). As Figures 2A and 2B show, the effects of impure cetyl myristoleic acid and palmitoleic acid on AsPC-1 human pancreatic cancer cells proliferation are similar to, though less potent than, the effects of myristoleic acid (shown in Figures 1A and 1B). Irrespective of whether myristoleic acid, cetyl myristoleic acid, or palmitoleic acid was used, the inhibition of proliferation was accompanied by morphological changes associated with apoptosis, including membrane blebbing, cellular rounding, and detachment from the culture plates (data not shown).
Example 3 -- Effect of Myristoleic Acid on Proliferation of AsPC-1 and PANC-1 Human Pancreatic Cancer Cells The thymidine incorporation experiments described in Example 1 were repeated in triplicate on each of two malignant human pancreatic cancer cell lines, AsPC-1 and PANC-l. The results are set forth in Figure 3A (AsPC-1) and Figure 3B (PANC-1). In each case, concentration-dependent effects of myristoleic acid on pancreatic cancer cell proliferation were observed. From these experiments it was determined that myristoleic acid has potent anti-proliferative activity in AsPC-1 and PANC-1 pancreatic cancer cells with effects in the low micromolar range.

Example 4 -- Effect of Myristoleic Acid on Annexin V
Binding in AsPC-1 Human Pancreatic Cancer Cells The effect of myristoleic acid (10 g/ml) ("MA") on annexin V binding in AsPC-1 cells at 3 or 5 hours after beginning treatment is shown in Figure 4.
Marked membrane fluorescence indicates early apoptosis in these cells. This specific test utilizes the translocation of phophatidyl serine from the inner to outer plasma membrane, which is a feature of early apoptosis. The results set forth in Figure 4 are representative of three separate experiments.
Example 5 -- Effect of Myristoleic Acid on DNA
Fragmentation by TUNEL Assay The effect of myristoleic acid (10 g/ml) ("MA") on terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling ("TUNEL") assay at 24 hours in AsPC-1 cells was investigated. The results are presented in Figures 5A (control) and 5B (10 g/ml myristoleic acid). As Figures 5A and 5B demonstrate, 10 g/ml myristoleic acid caused a marked increase in the number of apoptotic cells, shown in the upper right panel compared with control in the upper left. The results set forth in Figures 5A and 5B are representative of three separate experiments.

Example 6 -- Effect of Myristoleic Acid on Growth of Human Pancreatic Cancer Xenografts in Athymic Mice The effects of daily intraperitoneal injection of myristoleic acid (250mg/kg/day) on the growth of AsPC-1 and HPAC xenografts in athymic mice were investigated.
The results are presented in Figure 6. Animals were injected daily with myristoleic acid (250mg/kg/day) (labeled "myristoleic acid" in Figure 6) or with a control solution (labeled "control" in Figure 6) once visible tumors were established (about five days after implantation). As the sizes of the tumors in Figure 6 demonstrate, myristoleic acid caused a marked reduction in tumor size throughout the experimental period. The experiment was repeated twice with AsPC-1 cells and also with HPAC cells. The results set forth in Figure 6 are representative of these experiments.
Tumor volumes of the above-described athymic mice were measured as a function of time, and the results are presented in Figure 7A (AsPC-1 cancer cells) and Figure 7B (HPAC cancer cells). Figures 7A and 7B
demonstrate that myristoleic acid caused a marked reduction in both AspC-1 and HPAC tumor volume throughout the experimental period.
At the end of the experiment, the AsPC-1 and HPAC xenografts were harvested from the athymic mice and the weights of the tumors were measured. The results, presented in Figure 8A (AspC-1 cancer cells) and Figure 8B (HPAC cancer cells), show that myristoleic acid caused a marked reduction in AspC-1 and in HPAC tumor weight in all three experiments.

Example 7 -- In Situ TUNEL Assay of Human Pancreatic Cancer Xenografts in Athymic Mice To determine whether the effects observed in the athymic mouse experiments described in Example 7 might be due to apoptosis, in situ TUNEL assays were carried out on the AsPC-1 xenografts that were harvested from the athymic mice at the end of the experiment.
Images of xenograft sections from mice treated with the control solution (vehicle only) (Figures 9A and 9B) show minimal staining. In contrast, images of xenograft sections from mice treated with the myristoleic acid (Figures 9C and 9D) show many apoptotic cells (visualized by the dark brown staining). Thus, Figures 9A-9D
demonstrate that cells in the myristoleic acid-treated tumor are undergoing apoptosis (programmed cell death).
Although preferred embodiments have been depicted and described in detail herein, it will be apparent to those skilled in the relevant art that 30' various modifications, additions, substitutions and the like can be made without departing from the spirit of the invention and these are therefore considered to be within the scope of the invention as defined in the claims which follow.

Claims (41)

CLAIMS:
1. An in vitro method of decreasing proliferation of adenocarcinoma cells, or of inducing apoptosis of adenocarcinoma cells, said method comprising:
contacting a sample comprising adenocarcinoma cells with a compound of the formula:
CH3-(CH2)n-CH=CH-(CH2)m-COOH
wherein m is 7 and n is 3, or a pharmaceutically acceptable ester, salt, amide or solvate thereof.
2. The in vitro method according to claim 1, wherein the compound is in the cis configuration.
3. The in vitro method according to claim 1, wherein the sample comprises prostate cancer cells, lung cancer cells, stomach cancer cells, breast cancer cells, pancreatic cancer cells, colon cancer cells, or combinations thereof.
4. The in vitro method according to claim 1, wherein said contacting is carried out with the compound or a pharmaceutically acceptable salt, amide or solvate thereof.
5. The in vitro method according to claim 1, wherein said contacting is carried out with the compound.
6. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable salt of the compound.
7. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable amide of the compound.
8. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable solvate of the compound.
9. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable aryl or aralkyl ester of the compound.
10. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable alkyl ester of the compound.
11. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable methyl, ethyl, dodecyl, or cetyl ester of the compound.
12. The in vitro method according to claim 1, wherein said contacting is carried out with a pharmaceutically acceptable cetyl ester of the compound.
13. A use of an effective amount of a compound of the formula:
CH3-(CH2)n-CH=CH-(CH2)m-COOH
wherein m is 7 and n is 3, or a pharmaceutically acceptable ester, salt, amide or solvate thereof, for treating adenocarcinoma in a subject.
14. A use of an effective amount of a compound of the formula:
CH3-(CH2)n-CH=CH-(CH2)m-COOH
wherein m is 7 and n is 3, or a pharmaceutically acceptable ester, salt, amide or solvate thereof, for the preparation of a medicament for treating adenocarcinoma in a subject.
15. The use according to claim 13 or 14, wherein the subject is a human subject.
16. The use according to claim 13 or 14, wherein the amount is effective to decrease proliferation of cancer cells in the subject.
17. The use according to claim 13 or 14, wherein the amount is effective to induce apoptosis of cancer cells in the subject.
18. The use according to claim 13 or 14, wherein the compound is in the cis configuration.
19. The use according to claim 13 or 14, wherein the adenocarcinoma is selected from the group consisting of prostate cancer, lung cancer, stomach cancer, breast cancer, colon cancer, pancreatic cancer, and combinations thereof.
20. The use according to claim 13 or 14, wherein the compound or a pharmaceutically acceptable salt, amide or solvate thereof is used.
21. The use according to claim 13 or 14, wherein the compound is used.
22. The use according to claim 13 or 14, wherein a pharmaceutically acceptable salt of the compound is used.
23. The use according to claim 13 or 4, wherein a pharmaceutically acceptable amide of the compound is used.
24. The use according to claim 13 or 14, wherein a pharmaceutically acceptable solvate of the compound is used.
25. The use according to claim 13 or 14, wherein a pharmaceutically acceptable aryl or aralkyl ester of the compound is used.
26. The use according to claim 13 or 14, wherein a pharmaceutically acceptable alkyl ester of the compound is used.
27. The use according to claim 13 or 14, wherein a pharmaceutically acceptable methyl, ethyl, dodecyl or cetyl ester of the compound is used.
28. The use according to claim 13 or 14 wherein a pharmaceutically acceptable cetyl ester of the compound is used.
29. A use of a compound of the formula:
CH3-(CH2)n-CH=CH-(CH2)m-COOH

wherein m is 7 and n is 3, or a pharmaceutically acceptable ester, salt, amide or solvate thereof, for decreasing proliferation of adenocarcinoma cells or inducing apoptosis of adenocarcinoma cells.
30. A use of a compound of the formula:
CH3-(CH2)n-CH=CH-(CH2)m-COOH
wherein m is 7 and n is 3, or a pharmaceutically acceptable ester, salt, amide or solvate thereof, for the preparation of a medicament for decreasing proliferation of adenocarcinoma cells or inducing apoptosis of adenocarcinoma cells.
31. The use according to claim 29 or 30 wherein the compound is in the cis configuration.
32. The use according to claim 29 or 30 wherein the cells comprise prostate cancer cells, lung cancer cells, stomach cancer cells, breast cancer cells, pancreatic cancer cells, colon cancer cells, or combinations thereof.
33. The use according to claim 29 or 30, wherein the compound or a pharmaceutically acceptable salt, amide or solvate thereof is used.
34. The use according to claim 29 or 30, wherein the compound is used.
35. The use according to claim 29 or 30, wherein a pharmaceutically acceptable salt of the compound is used.
36. The use according to claim 29 or 30, wherein a pharmaceutically acceptable amide of the compound is used.
37. The use according to claim 29 or 30, wherein a pharmaceutically acceptable solvate of the compound is used.
38. The use according to claim 29 or 30, wherein a pharmaceutically acceptable aryl or aralkyl ester of the compound is used.
39. The use according to claim 29 or 30, wherein a pharmaceutically acceptable alkyl ester of the compound is used.
40. The use according to claim 29 or 30, wherein a pharmaceutically acceptable methyl, ethyl, dodecyl, or cetyl ester of the compound is used.
41. The use according to claim 29 or 30, wherein a pharmaceutically acceptable cetyl ester of the compound is used.
CA002436536A 2000-12-23 2001-12-24 Methods for inducing apoptosis and inhibiting proliferation in cancer cells Expired - Fee Related CA2436536C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US25780900P 2000-12-23 2000-12-23
US60/257,809 2000-12-23
PCT/US2001/049689 WO2002051406A1 (en) 2000-12-23 2001-12-24 Methods for inducing apoptosis and inhibiting proliferation in cancer cells

Publications (2)

Publication Number Publication Date
CA2436536A1 CA2436536A1 (en) 2002-07-04
CA2436536C true CA2436536C (en) 2008-07-08

Family

ID=22977837

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002436536A Expired - Fee Related CA2436536C (en) 2000-12-23 2001-12-24 Methods for inducing apoptosis and inhibiting proliferation in cancer cells

Country Status (4)

Country Link
AU (1) AU2002231190B2 (en)
CA (1) CA2436536C (en)
GB (1) GB2389788B (en)
WO (1) WO2002051406A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2186576B1 (en) 2001-10-11 2004-09-16 Universitat De Les Illes Balears 2-HYDROXYOLEIC ACID TO USE AS A MEDICINAL PRODUCT.
CA2950800C (en) * 2014-05-09 2023-01-10 Tangent Reprofiling Limited Modulators of androgen synthesis
CN114948967A (en) * 2021-12-31 2022-08-30 广州医科大学附属第五医院 Application of NU2058 in preparation of antitumor drugs

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS552601A (en) * 1978-06-20 1980-01-10 Yamasa Shoyu Co Ltd Anti-tumor agent for non-injection use
CN1035925C (en) * 1988-11-16 1997-09-24 许正 Cytotropic non-homogeneous molecular lipid and its preparation method
US5457130A (en) * 1989-03-20 1995-10-10 Cancer Research Campaign Technology Limited Eicosapentaenoic acid used to treat cachexia
EP0436682B1 (en) * 1989-07-21 1994-01-19 Marigen S.A. Sterols, esters with fatty acids and glucosides of such sterols; a method of preparation thereof; spontaneously dispersable agents containing these compounds; and the use of such agents to treat tumours
AU3973793A (en) * 1992-04-27 1993-11-29 New England Deaconess Hospital Corporation Method of treating cancer
US6214875B1 (en) * 1998-04-14 2001-04-10 Zhenhua Yang Anticancer effects of specific branched-chain fatty acids and related production process

Also Published As

Publication number Publication date
AU2002231190B2 (en) 2005-11-03
WO2002051406A9 (en) 2003-05-01
GB0317152D0 (en) 2003-08-27
WO2002051406A1 (en) 2002-07-04
CA2436536A1 (en) 2002-07-04
GB2389788A (en) 2003-12-24
GB2389788B (en) 2005-07-20

Similar Documents

Publication Publication Date Title
US10245240B2 (en) Treatment of prostate carcinoma
EP0954299B1 (en) Thyroxine analogues having no significant hormonal activity for treating malignant tumours
US20230114220A1 (en) Compositions and methods for modulating hair growth
US6117908A (en) Use of GABAB receptor agonists as reflux inhibitors
US5736576A (en) Method of treating malignant tumors with thyroxine analogues having no significant hormonal activity
RU2161487C2 (en) Use of derivatives of 2-hydroxy-5-phenylazobenzoic acid as chemoprophylactic and chemotherapeutic agents incolon cancer
EP1293205A1 (en) Valproic acid and derivatives thereof for the combination therapy of human cancers, for the treatment of tumour metastasis and minimal residual disease
WO1997046228A9 (en) Method of treating malignant tumors with thyroxine analogues having no significant hormonal activity
EA011074B1 (en) Hydrazide-containing cftr inhibitor compounds and uses thereof
WO2020142413A1 (en) Compositions and methods for modulating hair growth
CN100427139C (en) Water-soluble shps as novel alkylating agents
CA2436536C (en) Methods for inducing apoptosis and inhibiting proliferation in cancer cells
PT642341E (en) USE OF 3-HYDROXY-3-METHYLGLUTARIL COENZYME REDUCED INHIBITORS IN CANCER TREATMENT
JP2024023269A (en) Antitumor agents and combination drugs
AU2002231190A1 (en) Methods for inducing apoptosis and inhibiting proliferation in cancer cells
US20040072902A1 (en) Methods for inducing apoptosis and inhibiting proliferation in cancer cells
JP6900067B2 (en) Composition for suppressing cancer metastasis and treating
US20110046223A1 (en) Treatment of neurofibromatosis
EP1326605A1 (en) Methods for inhibiting proliferation and inducing apoptosis in cancer cells
EP3854392A1 (en) Use of carbamate compound for prevention, alleviation, or treatment of concurrent seizures
WO2022164901A1 (en) Enhanced anti-proliferative and antitumor immune effects of mitochondria-targeted hydroxyurea
NZ759233B2 (en) Compositions and methods for modulating hair growth
JP2000500500A (en) Methods and compositions for treating neoplasms using optically pure (-)-fotemustine
JP2000500495A (en) Methods and compositions for treating neoplasms using (+)-fotemustine

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed