Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
10-SUBSTITUTED COLCHICINOIDS AS POTENT ANTICANCER AGENTS
Document Type and Number:
WIPO Patent Application WO/2016/059650
Kind Code:
A1
Abstract:
The present invention relates to the compounds of formula I wherein R is as herein described. The present invention particularly relates to synthesis and antiproliferative activity of 10-substituted colchicinoids. Compounds of the invention can be used for prevention or in the treatment of cancer disease.

Inventors:
VISHWAKARMA RAM (IN)
BHARATE SANDIP BIBISHAN (IN)
KUMAR AJAY (IN)
SINGH BALJINDER (IN)
KUMAR ASHOK (IN)
BHUSHAN SHASHI (IN)
HAMID ABID (IN)
JOSHI PRASHANT (IN)
GURU SANTOSH KUMAR (IN)
KUMAR SURESH (IN)
HUSSAIN AASHIQ (IN)
QAZI ASIF KHURSHID (IN)
BHARATE SONALI SANDIP (IN)
SHARMA PARDUMAN (IN)
SAXENA AJIT KUMAR (IN)
MONDHE DILIP MANIKRAO (IN)
MAHAJAN GIRISH (IN)
WANI ZAHOOR (IN)
Application Number:
PCT/IN2015/050135
Publication Date:
April 21, 2016
Filing Date:
October 14, 2015
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
COUNCIL SCIENT IND RES (IN)
International Classes:
C07C233/41
Domestic Patent References:
WO2011091114A22011-07-28
WO2011091114A22011-07-28
WO1996011184A11996-04-18
WO2002056872A22002-07-25
WO2011022805A12011-03-03
WO1989012444A11989-12-28
WO2008102397A22008-08-28
Foreign References:
US8232254B22012-07-31
US20110184061A12011-07-28
US20120225912A12012-09-06
US20130011417A12013-01-10
US7935731B22011-05-03
Other References:
SUNG-KUK KIM ET AL: "The colchicine derivative CT20126 shows a novel microtubule-modulating activity with apoptosis", EXPERIMENTAL & MOLECULAR MEDICINE, vol. 45, no. 4, 1 April 2013 (2013-04-01), pages e19, XP055245449, DOI: 10.1038/emm.2013.38
BALJINDER SINGH ET AL: "Colchicine derivatives with potent anticancer activity and reduced P-glycoprotein induction liability", ORGANIC & BIOMOLECULAR CHEMISTRY, vol. 13, no. 20, 1 January 2015 (2015-01-01), GB, pages 5674 - 5689, XP055245352, ISSN: 1477-0520, DOI: 10.1039/C5OB00406C
COCCO G. ET AL., EUR.J. INTERN. MED., vol. 21, 2010, pages 503 - 508
NIEL E. ET AL., JOINT BONE SPINE, vol. 73, 2006, pages 672 - 678
TERKELTAUB, R. A., SEMIN. ARTHRITIS RHEUM., vol. 38, 2009, pages 411 - 419
DRULEY T.E. ET AL., BIOCHEMISTRY, vol. 40, 2001, pages 4323 - 4331
TATEISHI T. ET AL., BIOCHEM. PHARMACOL., vol. 53, 1997, pages 111 - 116
LING V., CANCER CHEMOTHER. PHARMACOL., vol. 40, 1997, pages S3 - S8
AL ER . ET AL., SCIENCE, vol. 323, 2009, pages 1718 - 1722
LAGE H., CELL. MOL. LIFE SCI., vol. 65, 2008, pages 3145 - 3167
COSENTINO L., J. MED. CHEM., vol. 55, 2012, pages 11062 - 11066
JIN M.S., NATURE, vol. 490, 2012, pages 566 - 569
ANDREU J., BIOCHEMISTRY, vol. 21, 1982, pages 6465 - 6476
AS L., BIOCHEMISTRY, vol. 44, 2005, pages 3249 - 3258
CIFUENTES M., BIOORG. MED. CHEM. LETT., vol. 16, 2006, pages 2761 - 2764
ZEFIROVA O.N., BIOORG. MED. CHEM., vol. 19, 2011, pages 5529 - 5538
NAKAGAWA G.K., BIOORG. MED. CHEM. LETT., vol. 15, 2005, pages 235 - 238
BOYER F.D., BIOORG. CHEM., vol. 38, 2010, pages 149 - 158
CHEN J., MINI REV. MED. CHEM., vol. 9, 2009, pages T174 - 1190
Attorney, Agent or Firm:
KOUL, SUNAINA et al. (C-235, Defence Colony, New ., IN)
Download PDF:
Claims:
We claim:

1. 10-substituted colchicinoid represented by the formula lg,

lg

2. A process for the preparation of compound of formula lg as claimed in claim 1, wherein the compound is prepared by reacting colchicine (1) in polar organic solvent with 4-chloro-3-(trifluoromethyl)phenylamine in the ratio in the range of 1 :4 under stirring at room temperature for 24-72 hours and purification by conventional method to obtain compound of formula lg.

2. A process as claimed in claim 2, wherein the polar organic solvent is selected from the group consisting of methanol, acetone, ethanol, dimethyl sulfoxide, ethyl acetate, dimethylformamide.

3. A process as claimed in claim 2, wherein the temperature is preferably in the range of 25-40 °C.

4. A process as claimed in claim 2, wherein the time period is preferably 60-70 hours.

5. A method of treating P-gp mediated multi-drug resistant cancer comprising administering a therapeutically effective amount of 10-substituted colchicinoid as claimed in claim 1.

6. The compound as claimed in claim 1 for use in treatment of P-gp mediated multidrug resistant cancer.

Description:
10-SUBSTITUTED COLCHICINOIDS AS POTENT ANTICANCER AGENTS

FIELD OF THE INVENTION

The present invention relates to 10-substituted colchicinoids for treatment of cancer diseases. The present invention particularly relates to synthesis and anticancer activity of 10-substituted colchicinoids. Compounds of the invention are devoid of P-gp induction liability which is associated with colchicine.

BACKGROUND OF THE INVENTION

Colchicine (1), a natural product isolated from Colchicum autumnale (Meadow Saffron) has been used for the treatment of rheumatic condition 'gout' and also for the treatment of familial Mediterranean fever, pericarditis, and Behcet's disease (Cocco G. et al., Eur. J. Intern. Med. 2010, 21, 503-508; Niel E. et al., Joint Bone Spine 2006, 73, 672-678; Terkeltaub, R. A., Semin. Arthritis Rheum. 2009, 38, 411-419). Colchicine exhibits potent anticancer activity by binding to tubulin, inhibiting its self-assembly and microtubule polymerization and finally arresting cell division at metaphase. Colchicine interacts mainly with three proteins viz. tubulin, cytochrome P4503A4 (CYP3A4), and P-glycoprotein (P-gp). CYP3A4 is the major protein found mainly in liver and intestine which metabolizes colchicine by demethylating the -OMe groups at C-2 and C-3 positions. The development of tumor resistance to colchicine mainly occurs via its P-gp substrate and induction activity, and increase in the expression of βΙΙΙ tubulin isotype (Niel E. et al., Joint Bone Spine 2006, 73, 672-678; Druley T.E. et al., Biochemistry 2001, 40, 4323-4331; Tateishi T. et al, Biochem. Pharmacol. 1997, 53, 111-116).

The P-gp induction is responsible for the multidrug resistance particularly in the case of cancer cells and makes the currently available chemotherapy ineffective (Ling V., Cancer Chemother. Pharmacol. 1997, 40, S3-S8; Aller S. et al., Science 2009, 323, 1718-1722). Therefore, the search for cytotoxic compounds which are devoid of multidrug resistance is of great importance (Lage H., Cell. Mol. Life Sci. 2008, 65, 3145-3167). Colchicine (1) is a substrate of P-gp efflux pump and it induces its activity by inducing conformational change (Druley T.E. et al., Biochemistry 2001, 40, 4323- 4331). To overcome the P-gp mediated resistance of colchicine, Cosentino et al. demonstrated that its N-benzylated derivative (2) showed no loss in cytotoxic activity in the resistant A2780AD ovarian carcinoma cells which are known to overexpress the ABCB1 drug transporter (Cosentino L., . Med. Chem. 2012, 55, 11062-11066).

2

Several patents have been published related to colchicine and their compounds for the treatment of inflammatory diseases, antiproliferative, gout, anti-tumor, etc. Thorson et al. have synthesized series of neoglycosides of colchicine with cytotoxic activity (US 8232254). Nielsen et al. (US20110184061), Nielsen et al. (WO2011091114) and Bombardelli et al. (WO1996011184) have designed thiocolchicine and colchicine derivatives suitable for use as a muscle relaxant, an anti-inflammatory agent, as an anti- gout agent, as an anti-proliferative agent, or as an anti-cancer agent. Tuszynski et al. (US20120225912), Brown et al. (WO2002056872) and Alberta health services (WO2011022805) have synthesized and patented colchicine derivatives for targeting cancer diseases. Han et al. have patented several colchicine derivatives for immunomodulatory action (US20130011417). Colchicine is also patented for control of retroviruses by The Rockefeller University USA (WO1989012444). Fringuelli et al. have synthesized and patented colchicine derivatives for the antifibrotic treatment of chronic hepatic diseases (WO2008102397). Mutual Pharmaceutical Company, Philadelphia, USA have used colchicine and macrolide antibiotics to inhibit P- glycoprotein in order to enhance the effect of colchicine. To the best of our knowledge, there is no patent where the P-glycoprotein induction liability of colchicine scaffold has been reduced by the structural modification of colchicine (US7935731). Recent report on the discovery of 3D-structure of P-gp from Caenorhabditis elegans, encouraged us to use 3D-crystal structure of P-gp to design colchicine derivatives for addressing its issue of P-gp induction liability (Jin M.S., Nature 2012, 490, 566-569). The structure-activity relationship (SAR) of colchicine as anti-tubulin agent (Cosentino L., . Med. Chem. 2012, 55, 11062-11066; Andreu J., Biochemistry 1982, 21, 6465-6476; Das L., Biochemistry 2005, 44, 3249-3258; Cifuentes M., Bioorg. Med. Chem. Lett. 2006, 16, 2761-2764; Zefirova O.N., Bioorg. Med. Chem. 2011, 19, 5529-5538; Nakagawa G.K., Bioorg. Med. Chem. Lett. 2005, 15, 235-238; Boyer F.D., Bioorg. Chem. 2010, 38, 149-158; Chen J., Mini Rev. Med. Chem. 2009, 9, 1174- 1190) is available in the literature, however its SAR for P-gp induction activity has not been examined. The limitation of colchicine in the treatment of cancer is due to its P-gp liability. The present invention reports discovery of 10-substituted colchicinoids with better in vitro and in vivo anticancer activity than colchicine, and with reduced P-gp induction liability.

OBJECTIVES OF THE INVENTION

The main object of the invention is to provide better anticancer compounds devoid of P- glycoprotein induction liability.

Another objective of the present invention is to provide 10-substituted colchicinoids. Yet another objective is to provide less toxic compounds than colchicine.

SUMMARY OF THE INVENTION

Accordingly, the present invention provides a compound represented by the formula I thereof,

wherein, R groups is selected from the group consisting of Piperazine, Piperazine carboxylate, 4- Amino piperidine, 4-Aryl piperidine, Prolinol, β-Carboline, or 4-chloro- 3-(Trifluoromethyl)phenylamine.

In a second aspect of the invention, a method is presented for treating or preventing P- gp-mediated multi-drug resistant cancer disease by identifying a patient suffering from or at a risk of developing a cancer disease and administering to the patient a therapeutically-effective amount of a compound represented by the formula I.

I wherein, R group is selected from Piperazine, Piperazine carboxylate, 4-Amino piperidine, 4-Aryl piperidine, Prolinol, β-Carboline, or 4-chloro-3- (Trifluoromethyl)phenylamine.

Another aspects of the invention is a process for the preparation of compound of formula I, wherein the compound is prepared by reacting colchicine (1) in polar organic solvent with a corresponding amine in the ratio in the range of 1 :4 under stirring at room temperature for 24-72 hours and purification by conventional method to obtain compound of formula I.

Another embodiment of the invention is the polar organic solvent selected from the group consisting of methanol, acetone, ethanol, dimethyl sulfoxide, ethyl acetate, dimethylformamide .

In yet another embodiment of the invention the temperature is preferably in the range of 25-40 °C.

In further embodiment the time period is preferably 60-70 hours.

Yet another embodiment of the invention is to provide 10-substituted colchicinoid for use in treatment of P-gp mediated multi-drug resistant cancer.

In one embodiment, the patient is human.

In another embodiment of the invention the said compounds are useful for the treatment of P-gp mediated multidrug resistant cancer.

BRIEF DESCRIPTION OF THE ACCOMPANYING DRAWINGS

Fig. 1 is a scheme illustrating the chemical synthesis of compounds claimed in the invention. Reagents and conditions: (a) amine (1.0 mmol), MeOH (3 mL), rt, 24-72 h, 60-90%.

Fig. 2 is a scheme illustrating the P-gp activity of colchicine (1) and compounds (la- lg) at 100 nM, measured in terms of the % intracellular accumulation of Rhodamine 123/total protein inside LSI 80 cells. The decrease in the % intracellular accumulation (compared to control) of Rhl23 indicates induction of P-gp. Rifampicin (10 μΜ) was used as a reference P-gp inducer. Statistical comparisons were made between control vs 1 and 1 vs other compounds. The p value <0.5 was considered to be significant. P value *< 0.5, **<0.01, ***<0.001. Comparison between control and 1 are represented by @.

Fig. 3 is a diagram illustrating effect of colchicine (1) and compounds Id and lg on microtubules in HCT-116 cells (National Cancer Institute; U.S). Cells were cultured on coverslips. After 24 h, the cells were treated with 100 nM of compound. Immunocytochemical staining was conducted using anti-a-tubulin antibody and Alexa Flour-488-labeled secondary antibody. Nuclei were stained with DAPI (left panel). The data are representative of three separate sets of experiments.

DETAILED DESCRIPTION OF THE INVENTION

Colchicine (1) is a substrate and inducer of P-glycoprotein. The present invention reports compounds, represented by general structure I, possessing better in vitro and in vivo anticancer activity and with reduced P-gp induction liability.

I

A series of 10-substituted compounds were prepared using scheme depicted in Fig. 1. Treatment of colchicine (1) with different amines in methanol produced corresponding 10-amino-linked compounds la-g in 60-81% yield. Colchicine (1) along with all synthesized compounds were screened for their ability to induce P-gp in P-gp overexpressing LS-180 cells (ECACC type, purchased from Sigma, Cat. no. 87021202, cell passage no. 52) using rhodaminel23 (Rhl23) cell exclusion method. The rifampicin (69% intracellular accumulation of Rhl23) was used as positive control in this study. Results are depicted in Fig. 2. Colchicine showed potent P-gp induction activity as indicated by the decrease in % intracellular accumulation of Rhl23 in LSI 80 cells (62%) compared to the control (100%). However, synthesized compounds displayed significantly reduced P-gp induction activity; which comprises of compounds substituted with Piperazine la, 4-Amino piperidine lb, 4-Phenyl piperidine Id, Tryptoline If, 4-Chloro-3-(trifluoromethyl)aniline lg all showing > 90% intracellular accumulation of Rhl23 in LSI 80 cells compared to the control (100%) (Fig. 2). Further, the EC 50 values of selected compounds Id and lg along with colchicine (1) was calculated for P-gp induction activity. The derivative Id and lg showed higher EC 50 value compared to colchicine (1). These results suggest that synthesized compounds are less liable to P-gp induction compared to colchicine (1) (Table 1).

The in vitro antiproliferative activity of compounds la-lg along with colchicine (1) was determined by MTT assay in two different cancer cell lines viz. HCT-116 (NCI, passage no. 5) and Colo-205 (National Cancer Institute; U.S) 116 (NCI, passage no. 3). The results are summarized in Table 2. The 10-amino substituted compound lg displayed promising antiproliferative activity in both cell lines (HCT-116, Colo-205) with IC 50 values of 0.040 and 0.030 μΜ, respectively. The compound lg showed better antiproliferative activity compared to natural product colchicine. The compounds exhibiting promising cytotoxicity in HCT-116 cells (IC 50 < 1 μΜ) along with complete loss of P-gp induction activity include lg, and Id.

The promising candidates identified through P-gp screening and antiproliferative screening were further studied for tubulin polymerization inhibition activity. Colchicine (1), 10-amino linked derivative Id and lg were tested for their ability to show disruption of microtubule assembly using confocal microscopy. As shown in Fig. 3, the cells treated with colchicine (1), derivative Id and lg showed a remarkable disruption and loss of microtubules compared to the control. The solubility of colchicine (1) and best compounds Id and lg were determined in biological media's such as phosphate buffer saline (PBS), simulated gastric fluid (SGF), and simulated intestinal fluid (SIF). Results are shown in Table 3.

Further, the compound lg was evaluated for pharmacokinetic studies. The pharmacokinetics of lg was evaluated in BALB/c mice following a single 10 mg/Kg dose administration by oral route and 1.0 mg/Kg dose administration by IV route. Following oral administration, elimination half life (t j i) was found to be 5.43 h and AUCo-t was found to be 16899 ng-h/mL. Following IV administration, elimination half life (ti/2 ,f i) was found to be 2.78 h and clearance was ~6 mL/min/Kg. The absolute oral bioavailability was 64%. The pharmacokinetic parameters are summarized in Table 4 and 5. The PK results indicated excellent plasma exposure of the compound lg and thus we decided to investigate the effect of this compound in in vivo anticancer model.

The compound lg along with colchicine (1) was evaluated for their ability to reduce tumor in Ehrlich solid tumor model in mice. The results are summarized in Table 6. The compound lg has shown promising activity at 1 mg/kg dose with 47.35% inhibition in tumor size compared to control. There is no mortality observed in the group treated with lg. On the other hand, colchicine (1) has shown promising activity with 52.29% inhibition at 0.75 mg/kg dose but the mortality was observed in 2 animals in the group of 7 animals. However, colchicine (1) has shown toxicity at 1 mg/kg with mortality of 5 animals in the group of 7 animals. 5-Fluorouracil was used as positive control with 54.78% inhibition at dose 22 mg/kg. These results clearly indicate that our compound lg has better safety profile compared to colchicine.

A class of 10-substituted colchicinoids is presented and defined by structural formula I:

I wherein, R group is selected from Piperazine, Piperazine carboxylate, 4-Amino piperidine, 4-Aryl piperidine, Prolinol, β-Carboline, or 4-chloro-3- (Trifluoromethyl)phenylamine.

Compounds of the invention derived from Formula I include, but are not limited to, the following chemical structures:

N-[(7S)-l,2,3-Trimethoxy-9-oxo-10- N-[(7S)- 1 ,2,3-Trimethoxy-9-oxo- 10-(4- (piperazin-l-yl)-5, 6,7,9- amino-piperidin-l-yl)-5, 6,7,9- tetrahydrobenzo[a]heptalen-7-yl]acetamide tetrahydrobenzo[a]heptalen-7-yl]acetamide (la); (lb);

N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-(N- N-[(7S)- 1 ,2,3-Trimethoxy-9-oxo- 10-(4- boc-piperazin-l-yl)-5, 6,7,9- phenyl-piperidin -l-yl)-5, 6,7,9- tetrahydrobenzo[a]heptalen-7-yl]acetamide tetrahydrobenzo[a]heptalen-7-yl]acetamide ( lc); < W);

N-[(7S)-l,2

prolinol- 1 -yl)-5, 6,7,9-

N-[(7S)- 1 ,2,3-Trimethoxy-9-oxo- 10-(2,3 ,4,9- tetrahydrobenzo[a]heptalen-7-yl]acetamide

tetrahydro- 1 H-pyrido [3 ,4-b] indole-2-yl)- (le);

5,6,7,9-tetrahydrobenzo[a]heptalen-7- yl]acetamide ( If);

N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-[3-(trifluoromethyl)-4-chl oro-phenylamino]-5,6,7,9- tetrahydrobenzo[a]heptalen-7-yl]acetamide ( lg);

The compounds of the invention can be used to treat a patient (e.g. a human) that suffers from or is at a risk of suffering from a disease, disorder, condition, or symptom described herein. The compounds of the invention can be used alone or in combination with suitable excipients in methods of treating or preventing P-gp mediated multidrug resistance cancer disease. Each such treatment described above includes the step of administering to a patient in need thereof a therapeutically effective amount of the compound of the invention described herein to delay, reduce or prevent such a disease, disorder, condition, or symptom. The compounds of the invention presented herein may be also useful in efflux of toxins out of the body.

It is understood that the foregoing examples are merely illustrative of the present invention. Certain modifications of the articles and/or methods employed may be made and still achieve the objectives of the invention. Such modifications are contemplated as within the scope of the claimed invention.

EXAMPLES

Following examples are given by way of illustration and should not construed the scope of present invention. Example 1: Synthesis of N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-(piperazin-l-yl)- 5,6,7,9-tetrahydrobenzo[a]heptalen-7-yl]acetamide (la). To the solution of colchicine (1, 0.25 mmol) in MeOH (3 mL) was added corresponding amine (Piperazine, 1.0 mmol). The reaction mixture was stirred at room temperature for 24-72 h, and then the mixture was diluted with cold water and extracted with ethyl acetate followed by purification with silica gel column chromatography (MeOH: CHCI 3 ) to yield compounds la (67 % yield); yellow solid; mp 158-161 °C. 1H NMR (CDCI 3 , 400 MHz): δ 7.44 (1H, s, br, NH), 7.28 (1H, s, C8-H), 7.25 (1H, d, J = 10.8 Hz, C12-H), 6.79 (1H, d, J = 10.8 Hz, Cl l-H), 6.52 (1H, s, C4-H), 4.64-4.61 (1H, m, C7-H), 3.93 (3H, s, Ar-OCH 3 ), 3.89 (3H, s, Ar-OCH 3 ), 3.64 (3H, s, Ar-OCH 3 ), 3.56-3.53 (2H, m, C2'-Ha, C6'-Ha), 3.35-3.32 (2H, m, C2'-Hb, C6'-Hb), 3.14-3.04 (4H, m, C3'-H, C5'-H), 2.50-2.39 (2H, m, C5-Ha, C6-Ha), 2.25-2.18 (1H, m, C5-Hb), 2.01 (3H, s, C14-CH 3 ), 1.87-1.85 (1H, m, C6-Hb); 13 C NMR (CDC1 3 , 125 MHz): δ 181.35 (C9), 169.78 (C13), 158.35 (CIO), 153.15 (C3), 151.29 (CI), 149.63 (C7a), 141.57 (C2), 136.11 (C12), 134.36 (C12a), 134.27 (C4a), 129.22 (C8), 125.96 (C12b), 118.26 (Cl l), 107.29 (C4), 61.46 (Cl-OCH 3 ), 61.41 (C2-OCH 3 ), 56.09 (C3-OCH 3 ), 51.87 (C7), 49.74 (C2'), 49.74 (C6'), 45.85 (C3'), 45.85 (C5'), 36.99 (C6), 30.05 (C5), 23.03 (C14); HR-MS: m/z calcd 454.2317 for C 25 H 3 iN 3 0 5 +H + (454.2336); IR (CHC1 3 ): v max 3283, 2926, 2852, 1660, 1544, 1462, 1349, 1234, 1195, 1118, 1033 cm "1 .

Example 2: Synthesis of N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-(4-amino-piperidin-l- yl)-5,6,7,9-tetrahydrobenzo[a]heptalen-7-yl]acetamide (lb). Procedure for synthesis of lb is similar to example 1 except corresponding amine (4-Amino-piperidine). Yield, 73%; yellow solid; mp 154-155 °C. 1H NMR (CDC1 3 , 400 MHz): δ 7.40 (1H, d, J = 11.6 Hz, C12-H), 7.39 (1H, s, C8-H), 7.22 (1H, d, J = 8 Hz, NH), 6.63 (1H, d, J = 11.6 Hz, Cl l-H), 6.51 (1H, s, C4-H), 4.64-4.61 (1H, m, C7-H), 3.92 (3H, s, Ar-OCH 3 ), 3.88 (3H, s, Ar-OCH 3 ), 3.60 (3H, s, Ar-OCH 3 ), 3.08-3.03 (4H, m, C2'-H, C6'-H), 2.81-2.70 (1H, m, C2'-H), 2.68-2.55 (1H, m, C4'-H), 2.48-2.35 (2H, m, C6-Ha, C5-Ha), 2.26-2.18 (1H, m, C6-Hb), 2.01 (3H, s, C14-CH 3 ), 1.89-1.84 (1H, m, C5-Hb), 1.74-1.70 (4H, m, C3'-H, C5'-H); 1J C NMR (CDCI 3 , 125 MHz): δ 175.08 (C9), 169.90 (C13), 153.01 (CIO), 152.84 (C3), 151.06 (CI), 150.08 (C7a), 141.48 (C2), 139.07 (C12), 134.53 (C4a), 130.17 (C12a), 126.85 (C12b), 122.83 (C8), 108.53 (Cl l), 107.15 (C4), 61.43 (CI-OCH3), 61.35 (C2-OCH3), 56.09 (C3-OCH 3 ), 52.52 (C7), 49.03 (C4'), 45.48 (C2'), 45.13 (C6'), 37.28 (C3'), 37.20 (C6), 32.67 (C5'), 30.07 (C5), 22.94 (C14); HR-MS: m/z calcd 468.2448 for C 26 H 3 3N 3 0 5 +H + (468.2493); IR (CHC1 3 ): v max 3305, 2927, 2853, 2802, 1654, 1579, 1488, 1463, 1427, 1283, 1194, 1095, 1045 cm "1 .

Example 3: Synthesis of N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-(N-boc-piperazin-l- yl)-5,6,7,9-tetrahydrobenzo[a]heptalen-7-yl]acetamide (lc). Procedure for synthesis of lc is similar to example 1 except corresponding amine (N-boc-Piperazine). Yield, 60%; yellow solid; mp 158-160 °C. 1H NMR (MeOD, 500 MHz): δ 7.32 (1H, d, J = 1 1.0 Hz, C12-H), 7.18 ( 1H, s, C8-H), 7.05 (1H, d, J = 11.0 Hz, Cl l-H), 6.72 ( 1H, s, C4- H), 4.48-4.46 ( 1H, m, C7-H), 3.91-3.81 (2H, m, C2'-Ha, C6'-Ha), 3.89 (3H, s, Ar- OCH 3 ), 3.58 (3H, s, Ar-OCH 3 ), 3.45-3.42 (2H, m, C2'-Hb, C6'-Hb), 3.30 (3H, s, Ar- OCH 3 ), 2.65-2.57 (2H, m, C3'-Ha, C5'-Ha). 2.39-2.31 (2H, m, C3'-Hb, C5'-Hb), 2.20- 2.12 (2H, m, C6-Ha, C5-Ha), 1.98 (3H, s, C14-CH 3 ), 1.95-1.94 (2H, m, C5-Hb, C6- Hb), 1.48 (9H, -CH 3 x3); 13 C NMR (CDC1 3 , 125 MHz): δ 181.28 (C9), 169.43 (C13), 157.98 (CIO), 154.73 (C7'), 153.24 (C3), 151.37 (CI), 149.15 (C7a), 141.74 (C2), 135.93 (C12), 134.34 (C4a), 134.24 (C12a), 129.41 (C8), 125.96 (C12b), 118.03 (Cl l), 107.39 (C4), 80.07 (C8'), 61.40 (CI-OCH3), 61.40 (C2-OCH 3 ), 56.13 (C3-OCH 3 ), 51.88 (C7), 48.53 (C2'), 48.53 (C6'), 37.24 (C6), 30.02 (C5), 29.70 (C3'), 29.70 (C5'), 28.45 (C9'), 28.45 (CIO'), 28.45 (Cl l'), 22.69 (C14); HR-MS: m/z calcd 554.2885 for C 3 oH 39 N 3 0 7 +H + (554.2861); IR (CHC1 3 ): v max 3390, 2924, 2854, 1653, 1422, 1322, 1232, 1160, 1044 cm "1 .

Example 4: Synthesis of N-[(7S)-l,2,3-Timethoxy-9-oxo-10-(4-phenyl-piperidin-l- yl)-5,6,7,9-tetrahydrobenzo[a]heptalen-7-yl]acetamide (Id). Procedure for synthesis of Id is similar to example 1 except corresponding amine (4-Phenyl-piperidine). Yield, 78%; yellow solid; mp 169- 172 °C. 1H NMR (CDCI 3 , 400 MHz): δ 7.35-7.31 (m, 3H, C9'-H, C10'-H, Cl l'-H) 7.28-7.20 (2H, C8'-H, C12'-H), 7.26 (IH, s, C8-H), 7.23 (IH, d, J = 11.2 Hz, C12-H), 6.84 (IH, d, J = 11.2 Hz, Cl l-H), 6.51 (IH, s, C4-H), 4.65-4.62 (IH, m, C7-H), 4.27-4.24 (IH, m, C2'-Ha), 4.11-4.08 (IH, m, C6'-Ha), 3.94 (3H, s, Ar- OCH 3 ), 3.89 (3H, s, Ar-OCHs), 3.66 (3H, s, Ar-OCH 3 ), 3.06-3.05 (IH, m, C4'-H), 2.86-2.80 (2H, m, C2'-Hb, C6'-Hb), 2.48-2.42 (2H, m, C6-Ha, C5-Ha), 2.22-2.17 (IH, m, C6-Hb), 2.04 (3H, s, C14-CH 3 ), 2.03-1.92 (3H, m, C5-Hb, C3'-Ha, C5'-Ha). 1.85- 1.84 (2H, m, C3'-Hb, C5'-Hb); 13 C NMR (CDC1 3 , 125 MHz): 181.51 (C9), 169.79 (C13), 158.62 (CIO), 153.06 (C3), 151.31 (CI), 149.10 (C7a), 145.60 (C7'), 141.57 (C2), 136.19 (C12), 134.41 (C4a), 134.30 (C12a), 128.80 (C8), 128.86 (C9'), 128.86 (Cl l'), 126.86 (C8'), 126.86 (C12'), 128.41 (CIO'), 126.11 (C12b), 118.03 (Cl l), 107.27 (C4), 61.47 (Cl-OCH 3 ), 61.42 (C2-OCH 3 ), 56.09 (C3-OCH 3 ), 51.87 (C7), 49.76 (C2'), 49.68 (C6'), 42.84 (C3'), 36.99 (C6), 33.55 (C3'), 31.14 (C5'), 30.09 (C5), 23.05 (C14); HR-MS: m/z calcd 529.2696 for C 3 2H 36 N 2 0 5 +H + (529.2697); IR (CHC1 3 ): v max 3291, 2932, 2850, 1654, 1601, 1543, 1486, 1399, 1321, 1216, 1095, 1007 cm "1 .

Example 5: Synthesis of N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-(L-prolinol-l-yl)- 5,6,7,9-tetrahydrobenzo[a]heptalen-7-yl]acetamide (le). Procedure for synthesis of le is similar to example 1 except corresponding amine (L-Prolinol). Yield, 81%; yellow solid; mp 164-166 °C. 1H NMR (CDC1 3 , 500 MHz): δ 7.91 (IH, d, J = 6.2 Hz, NH), 7.33 (IH, d, J = 11.8 Hz, C12-H), 7.25 (IH, s, C8-H), 6.53 (IH, s, C4-H), 6.48 (IH, d, J = 11.8 Hz, Cl l-H), 6.2 (IH, s, br, C5'-OH), 5.35-5.33 (IH, m, C3'-H), 4.55-4.53 (IH, m, C7-H), 3.93 (3H, s, Ar-OCH 3 ), 3.88 (3H, s, Ar-OCH 3 ), 3.73-3.69 (2H, m, C6'-H), 3.62 (3H, s, Ar-OCH 3 ), 3.55-3.52 (IH, m, C2'-Ha), 3.47-3.41 (IH, m, C5'-Ha), 2.44- 2.42 (IH, m, C6-Ha), 2.34-2.25 (2H, m, C6-Hb, C5-Ha), 2.12-2.11 (2H, m, C2'-Hb, C5'-Hb), 2.01 (3H, s, C14-CH 3 ), 2.01-1.97 (2H, m, C4'-H), 1.97-1.87 (IH, m, C6-Hb); 13 C NMR (CDC1 3 , 125 MHz): δ 176.77 (C9), 169.85 (C13), 156.25 (CIO), 152.92 (C3), 151.34 (CI), 149.89 (C7a), 141.49 (C2), 138.27 (C12), 134.69 (C4a), 129.65 (C12a), 126.20 (C12b), 123.54 (C8), 113.10 (Cl l), 107.35 (C4), 64.15 (C6'), 62.20 (C2'), 61.43 (Cl-OCH 3 ), 61.27 (C2-OCH 3 ), 56.08 (C3-OCH 3 ), 52.04 (C7), 50.58 (C5'), 37.47 (C6), 30.15 (C5), 28.51 (C5'), 23.01 (C4'), 22.45 (C14); HR-MS: m/z calcd 469.2336 for C 26 H 3 2N 2 06+H + (469.2333); IR (CHC1 3 ): v max 3350, 2928, 2853, 1653, 1598, 1527, 1486, 1455, 1401 , 1382, 1349, 1237, 1 145, 1096, 1019 cm "1 .

Example 6: Synthesis of N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-(2,3,4,9-tetrahydro- lH-pyrido[3,4-b]indole-2-yl)-5,6,7,9-tetrahydrobenzo[a]hepta len-7-yl]acetamide

(If). Procedure for synthesis of If is similar to example 1 except corresponding amine (Tryptoline). Yield, 62%; yellow solid; mp 158-160 °C. 1H NMR (CD 3 OD, 500 MHz): δ 7.44 (IH, d, J = 10.0 Hz, C12-H), 7.35 (IH, d, J = 10.0 Hz, Cl l-H), 7.30 (IH, m, C9'- H), 7.18 (IH, s, C8-H), 7.15-6.98 (2H, m, C10'-H, Cl l'-H,), 6.75-6.73 (IH, m, C8'-H), 6.72 (IH, s, C4-H), 4.75-4.69 ( IH, m, C7-H), 4.51-4.43 (IH, m, C6'-Ha), 4.10-4.01 (IH, m, C6'-Hb), 3.89 (3H, s, Ar-OCH 3 ), 3.87 (3H, s, Ar-OCH 3 ), 3.63-3.61 (IH, m, C2'- Ha), 3.57 (3H, s, Ar-OCH 3 ), 3.10-2.90 (2H, m, C2'-Hb, C3'-Ha), 2.61-2.51 (IH, m, C6- Ha), 2.45-2.25 (2H, m, C3'-Hb, C6-Hb), 2.24-2.10 ( IH, m, C5-Ha), 1.99 (3H, s, C14- CH 3 ), 1.99-1.97 (IH, m, C5-Hb); 13 C NMR (CD 3 OD, 100 MHz): δ 171.65 (C9), 167.16 (C13), 159.69 (CIO), 158.44 (C3), 154.83 (CI), 151.92 (C7a), 142.69 (C2), 138.47 (C12), 138.07 (C12'), 136.23 (C4a), 132.09 (C12a), 130.38 (C8), 128.83 (C7), 128.32 (C12b), 127.26 (C5'), 122.16 (C9'), 120.47 (C4'), 1 19.86 (C10'), 118.60 (C8'), 116.20 (Cl l'), 1 11.92 (Cl l), 108.83 (C4), 61.67 (Cl-OCH 3 ), 61.67 (C2-OCH 3 ), 56.65 (C3- OCH 3 ), 53.39 (C7), 48.66 (C2'), 44.54 (C6'), 37.71 (C6), 30.73 (C5), 22.50 (C3'), 22.45 (C14); HR-MS: m/z calcd 540.2493 for C 32 H 33 N 3 0 5 +H + (540.2493); IR (CHC1 3 ): v max 3392, 2923, 2852, 1649, 1602, 1537, 1487, 1454, 1399, 1349, 1321 , 1219, 1095, 1043, 1019 cm "1 .

Example 7: Synthesis of N-[(7S)-l,2,3-Trimethoxy-9-oxo-10-[3-(trifluoromethyl)-4- chloro-phenylamino]-5,6,7,9-tetrahydrobenzo[a]heptalen-7-yl] acetamide (lg). Procedure for synthesis of lg is similar to example 2 except corresponding amine (3- Trifluoromethyl-4-chloro-phenylamine). Yield, 62%; yellow solid; mp 168-171 °C. 1 H NMR (CDC1 3 , 500 MHz): δ 8.71 (IH, s, CIO-NH), 7.66 (IH, s, C8-H), 7.59 (IH, d, J = 4 Hz, C5'-H), 7.56 (IH, s, C2'-H), 7.50 (IH, d, J = 4 Hz, C6'-H), 7.40 (IH, d, J = 12.0 Hz, C12-H), 7.23 (1H, d, J = 12.0 Hz, Cl l-H), 6.54 (1H, s, C4-H), 4.71-4.69 (1H, m, C7-H), 3.93 (3H, s, Ar-OCH 3 ), 3.90 (3H, s, Ar-OCH 3 ), 3.65 (3H, s, Ar-OCH 3 ), 2.54- 2.51 (1H, m, C5-Ha), 2.44-2.29 (2H, m, C5-Hb, C6-Ha), 2.02 (3H, s, C14-CH 3 ) 1.94- 1.92 (1H, m, C6-Hb); 13 C NMR (CDC1 3 , 100 MHz): δ 175.83 (C9), 169.73 (C13), 153.35 (CIO), 152.14 (C3), 151.12 (CI), 150.79 (C7a), 141.58 (C2), 138.46 (C12), 137.40 (C3'), 134.53 (CI'), 134.26 (C4a), 132.79 (C8), 132.79 (C7'), 128.41 (C12a), 127.21 (C6'), 126.27 (C12b), 125.90 (C5'), 122.58 (C4'), 122.54 (C2'), 110.45 (Cl l), 107.27 (C4), 61.47 (Cl-OCH 3 ), 61.41 (C2-OCH 3 ), 56.13 (C3-OCH 3 ), 52.70 (C7), 37.21 (C6), 29.97 (C5), 22.99 (C14); HR-MS: m/z calcd 563.1556 for C 2 8H 2 6C1F 3 N 2 05+H + (563.1555); IR (CHC1 3 ): v max 3270, 3001, 2929, 2855, 1660, 1601, 1582, 1543, 1502, 1487, 1463, 1402, 1349, 1322, 1236, 1194, 1142, 1096, 1047, 1015 cm "1 .

Example 8: P-gp induction assay. All synthesized compounds were screened for their ability to induce P-gp by using rhodaminel23 (Rhl23) cell exclusion method. In this method, the P-gp function was evaluated in terms of rhodamine 123 (Rhl23) accumulation and efflux. Briefly, the protocol used is as follows: Colorectal LS-180 cells (ECACC type, purchased from Sigma, Cat. no. 87021202, cell passage no. 52) were seeded at a density of 2xl0 4 per well of 96 well plate and were allowed to grow for next 24 h. Cells were further incubated with the test compounds, and were diluted to a final concentration of 100 nM and rifampicin (standard) to a final concentration of 10 μΜ in complete media for 48 h. The final concentration of DMSO was kept at 0.1%. Drugs were removed and cells were incubated with HANKS buffer for 40 minutes before further incubation with HANKS buffer (containing 10 μΜ of Rhl23 as a P-gp substrate) for 90 minutes. At the end of Rhl23 treatment cells were washed four times with cold PBS followed by cell lysis for 1 h by using 200 μΐ of lysis buffer (0.1% Triton X-100 and 0.2 N NaOH). A total of 100 μΐ of lysate was used for reading fluorescence of Rhl23 at 485 nm/ 529 nm. Samples were normalized by dividing fluorescence of each sample with total protein present in the lysate. For EC 50 determination, different concentrations of compound were used to treat LSI 80 cells for 48 h. EC 50 was determined by plotting fluorescence of Rhl23 against concentration of compound. Colchicine showed potent P-gp induction activity as indicated by the decrease in % intracellular accumulation of Rhl23 in LS I 80 cells (62%) compared to the control (100%). However, synthesized compounds displayed significantly reduced P-gp induction activity; which comprises of compounds substituted with Piperazine la, 4- Amino piperidine lb, 4-Phenyl piperidine Id, Tryptoline If, 4-Chloro-3- (trifluoromethyl)aniline lg all showing > 90% intracellular accumulation of Rhl23 in LS I 80 cells compared to the control (100%) (Fig. 2). Further, the EC 50 values of selected compounds Id and lg along with colchicine (1) was calculated for P-gp induction activity. The derivative Id and lg showed higher EC 50 value compared to colchicine (1). These results suggest that synthesized compounds are less liable to P-gp induction compared to colchicine (1) (Table 1).

Table 1. Induction of P-gp in P-gp overexpressing LS-180 cells using rhodamine 123 (Rh 123) cell exclusion method

Entry EC50 (nM)

1 14.4

Id 29.6 lg 69

Example 9. Antiproliferative activity. Human colon cancer cell lines HCT-116 (NCI, passage no. 5) and Colo-205 116 (NCI, passage no. 3) were procured from National Cancer Institute, USA. HCT-116 and Colo-205 cells were grown in McCoy's 5 A and RPMI growth medium respectively containing 10% FCS, 100U penicillin and 100 mg streptomycin per mL medium. Cells were grown in CO 2 incubator (Thermocon Electron Corporation, Houston, TX) at 37 °C with 95% humidity and 5% CO 2 gas environment. Cells treated with tested materials were dissolved in DMSO while the untreated control cultures received only the vehicle (DMSO < 0.2%). Cells were seeded in 96 well plates and exposed to tested compounds at various concentrations for 48 h time interval. MTT dye (2.5 mg/ml in PBS) was added 4 hrs priors to experiment termination. The plates were then centrifuged at 1500 rpm for 15 min and the supernatant was discarded, and MTT formazan crystals were dissolved in 150 μΐ of DMSO. The OD measured at 570 nm with reference wavelength of 620 nm.

The percentages of cell viability and growth inhibition were calculated using formulas:

Absorbance of treated cells - Absorbance of Blank

The % of cell viability = X 100

Absorbance of control cells - Absorbance of Blank

% Growth inhibition = 100 - % of cell viability

The results are summarized in Table 2. The 10-amino substituted compound lg displayed promising antiproliferative activity in both cell lines (HCT-116, Colo-205) with IC 50 values of 0.040 and 0.030 μΜ, respectively. The compound lg showed better antiproliferative activity compared to natural product colchicine. The compounds exhibiting promising cytotoxicity in HCT-116 cells (IC 50 ≤ 1 μΜ) along with complete loss of P-gp induction activity include lg, and Id.

Table 2. Antiproliferative activities of colchicine and synthesized compounds (la-lg)

I

la-lg

Entry R HCT-116 Colo-205

(ICso, μΜ) (ICso, μΜ)

1 (Colchicine as standard-OCH 3 0.050 0.032

control)

la -\- >10 8.0

Id ΟΟ 1 · 0 0.8

If W 3.0 0.8 Example 10. Effect of compounds on microtubules. For detection of tubulins, HCT- 116 cells (1 x 10 3 cells/well) were seeded on to 18 mm square cover slips in six well plates in complete medium. Cells were allowed to adhere for 24 h and were treated with respective concentrations of compounds 1, Id and lg for 24 h. After the treatment period, cells were fixed in 4% paraformaldehyde for 10 min at room temperature and permeabilized using 0.5% Triton-X in PBS for 5 min. The cells were blocked with 10% goat serum for 20 min at room temperature. Microtubules were detected with a monoclonal a-tubulin antibody (Sigma, Catalogue number: T9026) diluted 1: 100 in 0.1% Triton X-100 in PBS for 1 h at room temperature and Alexa Fluor 488 conjugated secondary antibody (Invitrogen) diluted 1:500 in PBS for 1 h at room temperature. Cells were then washed three times in PBS and stained with 4',6-diamidino-2-phenylindole (DAPI 1 μg/ml in PBS). The cover slips were mounted over glass slides and cells were imaged by a laser scanning confocal microscope (Olympus Fluoview FV1000). The promising candidates identified through P-gp screening and antiproliferative screening were further studied for tubulin polymerization inhibition activity. Colchicine (1), 10- amino linked derivative Id and lg were tested for their ability to show disruption of microtubule assembly using confocal microscopy. As shown in Fig. 3, the cells treated with colchicine (1), derivative Id and lg showed a remarkable disruption and loss of microtubules compared to the control.

Example 11. Determination of thermodynamic equilibrium solubility. The compounds were first dissolved in methanol to prepare stock solutions (100 and 1000 μg/mL). Different concentrations of stock solutions were pipetted into the 96-well plates and the solvent was evaporated to ensure that solid drug was present in the beginning of the experiment. Thereafter, 200 μΐ of the dissolution medium was added to the wells and 96-well plate was shaken horizontally at 300 rpm (Eppendorf Thermoblock Adapter, North America) for 4 h at room temperature (25 ± 1 °C). The plates were kept overnight for equilibration of drug in medium. Later, the plates were centrifuged at 3000 rpm for 15 min (Jouan centrifuge BR4i). Supernatant (50 μΐ) was pipetted into UV 96-well plates (Corning 96 Well Clear Flat Bottom UV-Transparent Microplate) for analyses with plate reader (SpectraMax Plus384) at of 350 nm. The analyses were performed in triplicate for each compound. The solubility curve of concentration ^g/mL) vs absorbance was plotted to find out saturation point and the corresponding concentration was noted. The solubility of colchicine (1) and best compounds Id and lg were determined in biological medias such as phosphate buffer saline (PBS), simulated gastric fluid (SGF), and simulated intestinal fluid (SIF). Results are shown in Table 3.

Table 3. Solubility of compounds 1, Id and lg in water, PBS, SGF and SIF

Compound Solubility in μg/mL

Water PBS SGF SIF

1 >1500 800 200 800

Id 80 80 >1500 20 lg >1500 >1500 >1500 >1500

Example 12. Pharmacokinetic studies. Oral and intravenous (IV) pharmacokinetic studies of compound lg were carried out in BALB/c male mice (Jubilant Biosys Limited,Bangalore) of age 4-6 weeks, by administering lg orally and IV at dose of 10 mg/kg for oral and 1 mg kg for IV. Plasma samples were collected at appropriate time points between the range of 0 hours to 24 hours and analyzed by LC-MS-MS. Mean plasma concentration calculated and data were further analyzed for PK parameters using WinNonlin 5.3 software package. Following oral administration, elimination half life (ti/2 ,f i) was found to be 5.43 h and AUCo-t was found to be 16899 ng-h/mL. Following IV administration, elimination half life (t ji) was found to be 2.78 h and clearance was ~6 mL/min/Kg. The absolute oral bioavailability was 64%. The pharmacokinetic parameters are summarized in Table 4 and 5. Table 4. Pharmacokinetic parameters of compound lg post PO dose at 10 mg/Kg to BALB/c mice.

Parameter Unit Value tl/2,B (h) 5.43

AUCo t (ng-h/mL) 16899

AUC 0-∞ (ng-h/mL) 17334

Bioavailability (% F) 64.4

Time points considered for tin, β calculation: 8 - 24 h

Table 5. Pharmacokinetic parameters of compound lg post IV dose at 1 mg/Kg to BALB/c mice.

Parameter Unit Value

tl/2, β (h) 2.78

c (ng/mL) 698

Co (ng/mL) 819

AUCo t (ng-h/mL) 2343

AUCo- (ng-h/mL) 2691

CL (mL/min/Kg) 6.19

v d (L/Kg) 1.49

Tlast (h) 8.00

Time points considered

2 - 8 h

for ti 2 , p calculation

Co calculated manually using initial 3 time points /2,e>- terminal half life; AUCo- t : the area under the plasma concentration-time curve from 0 to last measurable time point; AUCo- : area under the plasma concentration-time curve from time zero to infinity; C max : maximum observed plasma concentration; Co: extrapolated concentration at zero time point; CL: clearance; Vd: volume of distribution; Vdss: volume of distribution at steady state; Ti ast : time at which last concentration was found; F: bioavailability. Example 13. In vivo activity in Ehrlich solid tumor model. Ehrlich ascites carcinoma (EAC) cells were collected from the peritoneal cavity of the swiss mice (animal house of IIIM registered eith CPCSEA registration no. 67/99/CPCSEA)weighing 18-23 gm, harbouring 8-10 days old ascitic tumor. 1x10 EAC cells were injected intramuscularly in the right thigh of 38 swiss male mice selected for the experiment on day 0. The next day, animals were randomized and divided into five groups. Four treatment groups contained 7 animals each and one control group contained 10 animals. Treatment was given as follows:

Group I: 1 (0.75 mg/kg, i/p) from day 1-9. Group II: lg (lmg/kg, i/p) from day 1-9.

The third treatment group was treated with 5-fluorouracil (22 mg/kg, i/p) from day 1 -9 and it served as positive control. The control group was similarly administered normal saline (0.2 ml, i/p) from day 1-9. On day 9 & 13, tumor bearing thigh of each animal was shaved and longest and shortest diameters of the tumor were measured with the help of vernier caliper. Tumor weight of each animal was calculated using the following formula.

Length (mm) X [width (mm)] 2

Tumor weight (mg) =

The percent tumor growth inhibition was calculated on day 13 by comparing the average values of treated groups with that of control group. Tumor growth in saline treated control animals was taken to be 100 %.

The results are summarized in Table 6. The compound lg has shown promising activity at 1 mg/kg dose with 47.35% inhibition in tumor size compared to control. There is no mortality observed in the group treated with 4o. On the other hand, colchicine (1) has shown promising activity with 52.29 % inhibition at 0.75 mg/kg dose but the mortality was observed in 2 animals in the group of 7 animals. However, colchicine (1) has shown toxicity at 1 mg/kg with mortality of 5 animals in the group of 7 animals. 5-Fluorouracil was used as positive control with 54.78% inhibition at dose 22 mg kg. These results clearly indicate that our compound lg has better safety profile compared to colchicine.

Table 6. In vivo activity of 1 and lg in Ehrlich solid tumor model

Avg. body

weights (g) of Day 13

animals on days % Tumor

Treatment Mortali

Avg. Avg. Growth

Groups ty

body tumor Inhibition

weights weights

(g) (mg)

1 (Colchicine,

22.2 21.4

standard control; 20.2 19.25 705.0 52.29 2/7

8 2

0.75 mg/kg, i/p)

1 (1 mg/kg, i/p)* Nd Nd Nd Nd Nd Nd 5/7

22.2 21.2

lg (l mg/kg, i/p) 22.0 21.66 778.0 47.35 0/7

8 8

5-Fluorouracil

22.4 22.2

(Positive control; 19.4 19.4 668.2 54.78 0/7

2 8

22 mg/kg, i/p)

Normal Control

23.4 24.4 25.4 25.33 1477.94 — 0/10

NS (0.2 ml, i/p)

* Colchicine (1) was toxic at 1 mg/kg with mortality of 5 animals in the group of 7 animals, therefore the tumor growth inhibition could not be determined. ADVANTAGES OF THE INVENTION

The main advantages of the present invention are:

• Compounds of the invention showed better antiproliferative activity, both in vitro as well as in vivo than natural product colchicine.

• Compounds of the invention have significantly reduced P-gp substrate and induction liability compared to natural product colchicine.

• Compounds of the invention have better solubility in biological fluids PBS, SGF and SIF compared to natural product colchicine.

• Compounds of invention are not toxic as compared to natural product colchicine.