OA16446A - Compositions comprising a PI3K inhibitor and a MEK inhibitor and their use for treating cancer. - Google Patents

Compositions comprising a PI3K inhibitor and a MEK inhibitor and their use for treating cancer. Download PDF

Info

Publication number
OA16446A
OA16446A OA1201300235 OA16446A OA 16446 A OA16446 A OA 16446A OA 1201300235 OA1201300235 OA 1201300235 OA 16446 A OA16446 A OA 16446A
Authority
OA
OAPI
Prior art keywords
compound
cancer
combination
tumor
formula
Prior art date
Application number
OA1201300235
Inventor
Laurent Debussche
Carlos GARCIAESCHEVERRIA
Jianguo Ma
Stuart Mcmillan
Janet Anne Meurer Ogden
Loic Vincent
Original Assignee
Sanof
Merck Patent Gmbh
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 Sanof, Merck Patent Gmbh filed Critical Sanof
Publication of OA16446A publication Critical patent/OA16446A/en

Links

Abstract

Methods of treating patients with cancer are provided, wherein the methods comprise administering to the patient an effective amount of a MEK inhibitor and an effective amount of a PI3K inhibitor. Compositions in which the MEK and PI3K inhibitors are combined also are described.

Description

COMPOSITIONS AND METHODS FOR TREATING CANCER USING PI3K INHIBITOR AND MEK INHIBITOR
CROSS-REFERENCE TO RELATED APPLICATIONS
This application daims the benefit of priority of U.S Provisional Application No. 61/421,465 filed December9, 2010, U.S Provisional Application No. 61/436,258 fîled January 26, 2011, and U.S Provisional Application No. 61/467,485 filed March 25, 2011, ail of which are incorporated herein by reference.
BACKGROUND
There is an ongoing need in the art for more efficacious methods and compositions in the treatment of cancer. The instant application is directed, generally, to compositions and methods for the treatment of cancer, and more particularly, to compositions and methods comprising inhibitors of the mitogen activated protein kinase (MEK) and/or phosphoinositide 3-kinase (PI3K) pathways.
Tumor cells treated with inhibitors of MEK kinases typically respond via inhibition of phosphorylation of ERK, down-regulation of Cyclin D, induction of G1 arrest, and finally undergoing apoptosis. Pharmacologically, MEK inhibition completely abrogates tumor growth in BRaf xenograft tumors whereas Ras mutant tumors exhibit only partial inhibition in most cases (D. B. Solit et al., Nature 2006; 439: 358-362). Thus, MEKs hâve been targets of great interest for the development of cancer therapeutics.
A/-((S)-2,3-dihydroxypropyl)-3-(2-fluoro-4-iodo-phenylamino)isonicotinamide (also referred to as MSC1936369 or AS703026) is a novel, allosteric înhibitor of MEK. It possesses relatively high potency and selectivity, having no activity against 217 kinases or 90 non-kinase targets when tested at 10 pM. The in vivo PK profile of AS703026 is acceptable in mice and rats, with relatively high oral bioavailability (52 ~ 57%), medium or high clearance (0.9 - 2.6 L/h/kg) and medium or long half-life (2.2 - 4.7 h). The compound is relatively well-tolerated in mice, with a two-week maximum tolerated dose of 60 mg/kg BID.
N-(3-{[(3-{[2-chloro-5-(methoxy)phenyl]amÎno)quinoxalin-2-y1)amino]sulfonyl}phenyl)-2methylalaninamide (also known as XL147 or SAR245408) and 2-amino-8-ethyl-4-methyl-6(1H-pyrazol-5-yl)pyrido[2,3-d]pyrimidin-7(8/-/)-one (also known as XL765 or SAR245409) are sélective inhibitors of class I PI3K lipid kinases. XL147 inhibits the phosphorylation of downstream effectors Akt and S6 ribosomal protein (S6RP) and targets only PI3K isoforms (Înhibitor concentration, i.e., IC50 values in nanomolar (nM): PI3Ka 39, ΡΙ3Κβ 383, ΡΙ3Κδ 36,
ΡΙ3Κγ 23). XL765 targets both PI3K isoforms (IC50 values in nM: PI3Ka 39, ΡΙ3Κβ 113,
-1 16446
PI3K5 43, ΡΙ3Κγ 9) and mTOR (157 nM).
Oral administration of XL147 orXL765 alone inhibits tumor growth in mice bearing xenografts in which PI3K signaling is activated, such as the PTEN-deficient PC-3 prostate adenocarcinoma, U87-MG gliobastoma, A2058 melanoma and WM-266-4 melanoma, or the PIK3CA mutated MCF7 mammary carcinoma. XL147 is currently undergoing several Phase I trials for patients with solid tumors and/or lymphoma and Phase II trials for patients with endométrial or hormone receptor-positive breast cancer. XL765 is currently undergoing testing in Phase I clinical trials for patients with solid tumor, lymphoma or glioblastoma and in a Phase l/ll trial for patients with hormone receptor-positive breast cancer.
There remains a need, however, for a cancer therapy that is more effective in inhibiting cell prolifération and tumor growth while minimizing patient toxicity. There is a particular need for an MEK or PI3K inhibitor therapy is made more efficacious without substantially increasing, or even maintaining or decreasing, the dosages of MEK or PI3K inhibitor traditionally employed in the art.
SUMMARY
In one aspect, there is provided compositions and uses thereof in the treatment of a variety of cancers.
In particular embodiments, there is provided a composition that includes a compound having the following structural formula:
OH
N
OH (1) (MSC1936369 or AS703026 or MSC6369) and a compound selected from the group consisting of
-216446 and
(XL147orSAR245408)
(2b).
(XL765, SAR245409 or MSC0765)
In another aspect, methods of treating a patient with cancer are provided that comprise administering to the patient a therapeutically effective amount of a compound of Formula (1 ), or a pharmaceutically acceptable sait thereof, in combination with the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof.
In one embodiment, a method of treating a patient with cancer comprises administering to the patient a first dosage of a MEK inhibitor and a second dosage of a PI3K inhibitor, wherein said MEK inhibitor has the following structural formula:
and said PI3K inhibitor is selected from the group consisting of (1)
-316446 and
(2b).
In some embodiments, the methods involve treating cancer selected from the group consisting of non-small cell lung cancer, breast cancer, pancreatic cancer, liver cancer, prostate cancer, bladder cancer, cervical cancer, thyroid cancer, colorectal cancer, liver cancer, muscle cancer, hematological malignancies, melanoma, endométrial cancer and pancreatic cancer. In others, the cancer is selected from the group consisting of colorectal cancer, endométrial cancer, hematological malignancies, thryoid cancer, breast cancer, melanoma, pancreatic cancer and prostate cancer.
In some embodiments, the compositions and methods of use described herein are in amounts (i.e., either in the composition are in an administered dosage) that synergistically reduce tumor volume in a patient In further embodiments, the synergistic combination achieves tumor stasis or tumor régression.
In another aspect, a combination for use in treating cancer is provided, the combination comprising a therapeutïcally effective amount of (A) the compound of Formula (1), or a pharmaceutically acceptable sait thereof, and (B) the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof.
In one embodiment, uses of a combination comprising a therapeutically effective amount of
-416446 (A) the compound of Formula (1), or a pharmaceutically acceptable sait thereof, and (B) the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof, are provided for the préparation of a médicament for use in treatment of cancer.
In another aspect, kits are provided comprisîng: (A) the compound of Formula (1 ), or a pharmaceutically acceptable sait thereof; (B) the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof; and (C) instructions for use.
Other objects, features and advantages will become apparent from the foliowing detailed description. The detailed description and spécifie examples are given for illustration only since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description. Further, the examples demonstrate the principle of the invention and cannot be expected to specifically illustrate the application of this invention to ail the examples where it will be obviously useful to those skilled in the prior art.
BRIEF DESCRIPTION OFTHE DRAWINGS
Figure 1 provides a plot showing body weight change during the évaluation of the antitumor activity of Compound (1) (5 mg/kg) in combination with Compound (2b) (30 mg/kg) and Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice.
Figure 2 provides a plot showing antitumor activity of Compound (1) (5 mg/kg) in combination with Compound (2b) (30 mg/kg) against human HCT 116 bearing SCID female mice.
Figure 3 provides a plot showing antitumor activity of Compound (1) (5 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice. The box indicates combinations achieving therapeutic synergy.
Figure 4 provides a plot showing body weight change during the évaluation of the antitumor activity of Compound (1 ) (10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) and Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice.
Figure 5 provides a plot showing antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) against human HCT 116 bearing SCID female mice.
Figure 6 provides a plot showing antitumor activity of Compound (1) (10 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice.
-516446
Figure 7 provides a plot showing body weight change during the évaluation of the antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice.
Figures 8 provides a plot showing antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice. The box indicates combinations achieving therapeutic synergy.
Figure 9 provides a plot showing body weight change during the évaluation of the antitumor activity of Compound (1)(10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) against human HCT 116 bearing SCID female mice.
Figure 10 provides a plot showing antitumor activity of Compound (1 ) (10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) against human HCT 116 bearing SCID female mice.
Figure 11 provides a plot showing percent body weight of MiaPaCa-2 tumor-bearing mice treated with Compound (1) (5 mg/kg) and Compound (2a) (50 mg/kg) alone or in combination.
Figure 12 provides a plot showing percent body weight of MiaPaCa-2 tumor-bearing mice treated with Compound (1) (5 mg/kg) and Compound (2b) (30 mg/kg) alone or in combination.
Figure 13 provides a plot showing mean tumor volumes of MiaPaCa-2 tumor-bearing mice treated with Compound (1) (5 mg/kg) and Compound (2a) (50 mg/kg) alone or in combination.
Figure 14 provides a plot showing mean tumor volumes of MiaPaCa-2 tumor-bearing mice treated with Compound (1) (5 mg/kg) and Compound (2b) (30 mg/kg) alone or in combination.
Figures 15A and 15B provide charts showing Z-score values of Compound (1 ) for various tumor cell lines identifying spécifie therapeutic applications. Sélection of spécifie therapeutic applications for Compound (1). Individual z-score values for each cell line are plotted within one group corresponding to the tumor origin. An average value for ail values within one group is shown as a green triangle, and can serve as an indîcator for Compound (1 ) activity within one group. As for individual z-scores, z-scores below mean strong efficacy, whereas z-scores >0 approximate résistance.
Figures 16A and 16B provide charts showing Z-score values of Compound (2b) for various
-616446 tumor cell fines identifying spécifie therapeutic applications. Sélection of spécifie therapeutic applications for Compound (2b). Individual z-score values for each cell line are plotted within one group corresponding to the tumor origin. An average value for ail values within one group is shown as a green triangle and can serve as an indicator for Compound (2b) activity within one group. As for individual z-scores, z-scores below zéro mean strong efficacy, whereas a z-score >0 approximate résistance.
Figure 17 provides a chart showing Z-score values of Compound (1) in combination with Compound (2b) for various tumor cell lines.
Figures 18A, 18B, 18C, 18D, 18E and 18F provide plots and graphs showing combination résulte of Compound (1 ) with Compound (2b) in CRC tumor cell lines (synergy plot & mutation analysis).
Figures 19A and 19B provide plots and graphs showing combination results of Compound (1) with Compound (2b) in pancreatic tumor cell lines (synergy plot & mutation analysis).
Figures 20A and 20B provide plots and graphs showing combination results of Compound (1) with Compound (2b) in NSCLC tumor cell lines (synergy plot & mutation analysis).
Figure 21 provides a plot showing body weight change during the évaluation of the antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) and Compound (2a) (75 mg/kg) against human prîmary colon tumors CR-LRB-009C bearing SCID female mice.
Figure 22 provides a plot showing antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) and Compound (2a) (75 mg/kg) against human primary colon tumors CR-LRB-009C bearing SCID female mice.
Figure 23 provides a plot showing body weight change during the évaluation of the antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) and Compound (2a) (75 mg/kg) against human primary colon tumors CR-LRB-013P bearing SCID female mice.
Figure 24 provides a plot showing antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) and Compound (2a) (75 mg/kg) against human primary colon tumors CR-LRB-013P bearing SCID female mice.
Figure 25 graphically depicts the results of leyte ex vivo imaging of Evans Blue tumor extravasation performed after treatment with either Compound (2a) or Compound (2b) as single agents or in combination with Compound (1) in HCT116 xenografts.
Figures 26A and 26B graphically depict results of FMT imagïng after three days of therapy, three hours after AnnexinV-750 administration, four hours post-treatment with Compound (1), Compound (2a) or Compound (2b) as single agents or combinations in HCT116 xenografts. Tumor fluorescence was quantified in pmol of fluorophore and standardized to the tumor volume. Statistics: Newman-Keuls after 2way Anova on Ranked data, NS: P<0.05).
Figures 27A and 27B graphically show protein levels of cleaved-PARP and caspase-3 in tumor extracts following treatment with Compound (1), Compound (2a) or Compound (2b) alone or in selected combination. Statistics: Dunnett's test for one factor after one way
Anova, NS: P<0.05.
Figure 28 provides a plot showing tumor volumes of HCT116 tumor-bearing mice treated with Compound (1) (10 mg/kg), Compound (2a) (50 mg/kg) or Compound (2b)(20 mg/kg) alone or in combination. To quantify apoptosis, fluorescent Annexin-Vivo-750 was injected iv on day 3 and day 7 after start of treatment, 1 hour post daily treatment Animais were imaged by FMT 3 hours post probe injection.
DETAILED DESCRIPTION
In one aspect, methods for treating patients with cancer are provided. In one embodiment, the methods comprise administering to the patient a therapeutically effective amount of a MEK inhibitor and a therapeutically effective amount of a PI3K inhibitor, as further described 20 below.
In one embodiment, the inventive methods and compositions comprise a MEK inhibitor having the following structural formula:
OH
The MEK inhibitor according to formula (1), is referred to herein as Compound (1)” and is known also as MSC1936369, AS703026 or MSC6369. The préparation, properties, and
MEK-inhibiting abilities of Compound (1) are provided in, for example, International Patent
Publication No. WO 06/045514, particularly Example 115 and Table 1 therein. The entire • 816446 contents of WO 06/045514 are incorporated herein by reference. Neutral and sait forms of the compound of Formula (1) are ail considered herein.
In other embodiments, the inventive methods and compositions comprise a PI3K inhibitor having one of the following structures:
or
(2b).
The PI3K inhibitor according to formula (2a), is referred to herein as “Compound (2a) and is known also as XL147 or SAR245408. The PI3K inhibitor according to formula (2b), is referred to herein as Compound (2b)” and is known also as XL765, SAR245409 or MSC0765. The préparation and properties of Compound (2a) are provided in, for example, International Patent Publication No. WO 07/044729, particularly Example 357 therein, The entire contents of WO 07/044729 are incorporated herein by reference. The préparation and properties of Compound (2b) are provided in, for example, International Patent
Publication No. WO 07/044813, particularly Example 56 therein. The entire contents of WO 07/044813 are incorporated herein by reference.
In some embodiments, the compounds described above are unsolvated. In other embodiments, one or both of the compounds used in the method are in solvated form. As known in the art, the solvaté can be any of pharmaceutically acceptable solvent, such as
-916446 water, éthanol, and the like. In general, the presence of a solvaté or lack thereof does not hâve a substantiel effect on the efficacy of the MEK or PI3K inhibitor described above.
Although the compounds ïn Formula (1), Formula (2a) and Formula (2b) are depicted in their neutral forms, in some embodiments, these compounds are used in a pharmaceutically acceptable sait form. The sait can be obtained by any of the methods well known in the art, such as any of the methods and sait forms elaborated upon in WO 07/044729, as incorporated by reference herein. A pharmaceutically acceptable sait of the compound refers to a sait that is pharmaceutically acceptable and that retains pharmacological activity. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington ’s Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, or S. M. Berge, et al., Pharmaceutical Salts, J. Pharm. Sci., 1977;66:1-19, both of which are incorporated herein by reference.
Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, as well as those salts formed with organic acids, such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartane acid, citric acid, benzoic acid, cinnamîc acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonicacid, 2hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, p-toluenesulfonic acid, and salicylic acid.
In a first set of embodiments, the MEK inhibitor of formula (1) is administered simultaneously with the PI3K inhibitor of either formula (2a) or (2b). Simultaneous administration typically means that both compounds enter the patient at precisely the same time. However, simultaneous administration also includes the possibility that the MEK inhibitor and PI3K inhibitor enter the patient at different times, but the différence in time is sufficiently miniscule that the first administered compound is not provided the time to take effect on the patient before entry of the second administered compound. Such delayed times typically correspond to less than 1 minute, and more typically, less than 30 seconds.
In one example, wherein the compounds are in solution, simultaneous administration can be achieved by administering a solution containing the combination of compounds. In another -10-
example, simultaneous administration of separate solutions, one of which contains the MEK inhibitor and the other of which contains the PI3K inhibitor, can be employed. In one example wherein the compounds are in solid form, simultaneous administration can be achieved by administering a composition containing the combination of compounds.
In other embodiments, the MEK and PI3K inhibitors are not simultaneously adminîstered. In this regard, the first adminîstered compound is provided time to take effect on the patient before the second adminîstered compound is adminîstered. Generally, the différence in time does not extend beyond the time for the first adminîstered compound to complété its effect in the patient, or beyond the time the first adminîstered compound is completely or substantially eliminated or deactivated in the patient. In one set of embodiments, the MEK inhibitor is adminîstered before the PI3K inhibitor. In another set of embodiments, the PI3K inhibitor is adminîstered before the MEK inhibitor. The time différence in non-simultaneous administrations is typîcally greater than 1 minute, and can be, for example, precisely, at least, up to, or less than 5 minutes, 10 minutes, 15 minutes, 30 minutes, 45 minutes, 60 minutes, two hours, three hours, six hours, nine hours, 12 hours, 24 hours, 36 hours, or 48 hours.
In one set of embodiments, one or both of the MEK and PI3K inhibitors are adminîstered in a therapeutically effective (î.e., therapeutic) amount or dosage. A “therapeutically effective amount is an amount of the MEK or PI3K inhibitor that, when adminîstered to a patient by itself, effectively treats the cancer (for example, inhibits tumor growth, stops tumor growth, or causes tumor régression). An amount that proves therapeutically effective amount in a given instance, for a particular subject, may not be effective for 100% of subjects simllarly treated for the disease or condition under considération, even though such dosage is deemed a therapeutically effective amount by skilled practitioners. The amount of the compound that corresponds to a therapeutically effective amount is strongly dépendent on the type of cancer, stage of the cancer, the âge of the patient being treated, and other facts. In general, therapeutically effective amounts of these compounds are well-known in the art, such as provided in the supporting references cited above.
In another set of embodiments, one or both of the MEK and PI3K inhibitors are adminîstered in a sub-therapeutically effective amount or dosage. A sub-therapeutically effective amount is an amount of the MEK or PI3K inhibitor that, when adminîstered to a patient by itself, does not completely inhibit over time the biological activity of the intended target.
Whether adminîstered in therapeutic or sub-therapeutic amounts, the combination of MEK inhibitor and PI3K inhibitor should be effective in treating the cancer. A sub-therapeutic amount of MEK inhibitor can be an effective amount if, when combined with the PI3K inhibitor, the combination is effective in the treatment of a cancer.
-11 16446
In some embodiments, the combination of compounds exhibits a synergistic effect (i.e., greater than additive effect) in treating the cancer, particularly in reducing a tumor volume in the patient. In different embodiments, depending on the combination and the effective amounts used, the combination of compounds can either inhibit tumor growth, achieve tumor stasis, or even achieve substantial or complété tumor régression.
In some embodiments, Compound (1) is administered at a dosage of about 7-120 mg po qd. Compound (2a), meanwhile, can be administered at a dosage of about 12-600 mg po qd. Compound (2b) can be administered at a dosage of about 15-90 mg po qd.
As used herein, the term about generally indicates a possible variation of no more than 10%, 5%, or 1% of a value. For example, about 25 mg/kg” will generally indicate, in its broadest sense, a value of 22.5-27.5 mg/kg, i.e., 25 ± 10 mg/kg.
While the amounts of MEK and PI3K inhibitors should resuit in the effective treatment of a cancer, the amounts, when combined, are preferably not excessively toxic to the patient (i.e., the amounts are preferably within toxicity timits as established by medical guidelines). In some embodiments, either to prevent excessive toxicity and/or provide a more efficacious treatment of the cancer, a limitation on the total administered dosage is provided. Typically, the amounts considered herein are per day; however, half-day and two-day or three-day cycles also are considered herein.
Different dosage regîmens may be used to treat the cancer. In some embodiments, a daily dosage, such as any of the exemplary dosages described above, is administered once, twice, three times, or four times a day for three, four, five, six, seven, eight, nine, or ten days. Depending on the stage and severity of the cancer, a shorter treatment time (e.g., up to five days) may be employed along with a high dosage, or a longer treatment time (e.g., ten or more days, or weeks, or a rnonth, or longer) may be employed along with a low dosage. In some embodiments, a once- or twice-daily dosage is administered every other day. In some embodiments, each dosage contains both the MEK and PI3K inhibitors, while in other embodiments, each dosage contains either the MEK or PI3K inhibitors. In yet other embodiments, some of the dosages contain both the MEK and PI3K inhibitors, while other dosages contain only the MEK or the PI3K inhibitor.
Examples of types of cancers to be treated with the présent invention include, but are not limited to, lymphomas, sarcomas and carcinomas, e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, ostéogénie sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, synovioma, mesothelioma, lymphangioendotheliosarcoma, Ewing’s tumor, leiomyosarcoma, rhabdomyosarcome, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, gastric
-1216446 cancer, esophageal cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, rénal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testîcular tumor, lung carcinoma, non-small cell lung carcinoma, small cell lung carcinoma, bladder carcinoma, épithélial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, meningîoma, melanoma, neuroblastoma, retinoblastoma; leukemias, e.g., acute lymphocytic leukemia and acute myelocytic leukemia (myeloblastic, promyelocytic, myelomonocytic, monocytic and erythroleukemia); chronic leukemia (chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia); and polycythemia vera, lymphoma (Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom’s macroglobulinemia and heavy chain disease.
In some embodiments, the cancer being treated is selected from the group consisting of non-small cell lung cancer, breast cancer, pancreatic cancer, liver cancer, prostate cancer, bladder cancer, cervical cancer, thyroid cancer, colorectal cancer, liver cancer, and muscle cancer. In other embodiments, the cancer is selected from colorectal cancer, endométrial cancer, hematology cancer, thryoid cancer, triple négative breast cancer or melanoma.
The patient considered herein is typically a human. However, the patient can be any mammal for which cancer treatment is desired. Thus, the methods described herein can be applied to both human and veterinary applications.
The term treating or treatment, as used herein, indicates that the method has, at the least, mitigated abnormal cellular prolifération. For example, the method can reduce the rate of tumor growth in a patient, or prevent the continued growth of a tumor, or even reduce the size of a tumor.
In another aspect, methods for preventing cancer in an animal are provided. In this regard, prévention dénotés causing the clinical symptoms of the disease not to develop in an animal that may be exposed to or predisposed to the disease but does not yet expérience or display symptoms of the disease. The methods comprise administering to the patient a MEK inhibitor and a PI3K inhibitor, as described herein. In one example, a method of preventing cancer in an animal comprises administering to the animal a compound of Formula (1), or a pharmaceutically acceptable sait thereof, in combination with a compound selected from the group consisting of Formula (2a) and Formula (2b), or a pharmaceutically acceptable sait thereof.
The MEK and PI3K inhibiting compounds, or their pharmaceutically acceptable salts or solvaté forms, in pure form or in an appropriate pharmaceutical composition, can be administered via any of the accepted modes of administration or agents known in the art. The compounds can be administered, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intracistemally, or rectally. The dosage form can be, for example, a solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, pills, soft elastic or hard gelatin capsules, powders, solutions, suspensions, suppositories, aérosols, or the like, preferably in unit dosage forms suitable for simple administration of précisé dosages. A particular route of administration is oral, particularly one in which a convenient daily dosage regimen can be adjusted according to the degree of severity of the disease to be treated.
In another aspect, the instant application is directed to a composition that includes the MEK inhibitor shown in Formula (1) and a PI3K inhibitor selected from the compounds shown in Formulas (2a) and (2b). In some embodiments, the composition includes only the MEK and PI3K inhibitors described above. In other embodiments, the composition is in the form of a solid (e.g., a powder or tablet) including the MEK and PI3K inhibitors in solid form, and optionally, one or more auxiliary (e.g., adjuvant) or pharmaceutically active compounds in solid form. In other embodiments, the composition further includes any one or combination of pharmaceutically acceptable carriers (i.e., vehicles or excipients) known in the art, thereby providing a liquid dosage form.
Auxiliary and adjuvant agents may include, for example, preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prévention of the action of microorganisms is generally provided by various antibacterial and antifungal agents, such as, parabens, chlorobutanol, phénol, sorbic acid, and the like. Isotonie agents, such as sugars, sodium chloride, and the like, may also be included. Prolonged absorption of an injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. The auxiliary agents also can include wetting agents, emulsifying agents, pH buffering agents, and antioxidants, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylated hydroxytoluene, and the like.
Dosage forms suitable for parentéral injection may comprise physîologically acceptable stérile aqueous or nonaqueous solutions, dispersions, suspensions or émulsions, and stérile powders for reconstitution into stérile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, éthanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as
-14 16446 ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the requîred particle size in the case of dispersions and by the use of surfactants.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, as for example, cellulose dérivatives, starch, alignâtes, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quatemary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, magnésium stéarate and the like (h) adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stéarate, magnésium stéarate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms also may comprise buffering agents.
Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well-known in the art. They can contain pacifying agents and can be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds also can be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable émulsions, solutions, suspensions, syrups, and élixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., a MEK or PI3K inhibitor compound described herein, or a pharmaceutically acceptable sait thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, éthanol and the like; solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3butyleneglycol, dimethyl formamide; oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
-1516446
Suspensions, in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
Compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds described herein with, for example, suitable non- irritating excipients or carriers such as cocoa butter, polyethyleneglycol or a suppository wax, which are solid at ordinary températures but liquid at body température and therefore, melt while in a suitable body cavity and release the active component therein.
Dosage forms for topicai administration may include, for example, ointments, powders, sprays, and inhalants. The active component is admixed under stérile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as can be required. Ophthalmic formulations, eye ointments, powders, and solutions also can be employed.
Generally, depending on the intended mode of administration, the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of the compounds described herein, or a pharmaceutically acceptable sait thereof, and 99% to 1% by weight of a pharmaceutically acceptable excipient. In one example, the composition will be between about 5% and about 75% by weight of a compounds described herein, or a pharmaceutically acceptable sait thereof, with the rest being suitable pharmaceutical excipients.
Actual methods of preparing such dosage forms are known, or will be apparent, to those skîlled in this art. Reference is made, for example, to Remington’s Pharmaceutical Sciences, 18th Ed., (Mack Publishing Company, Easton, Pa., 1990).
In some embodiments, the composition does not include one or more other anti-cancer compounds. In other embodiments, the composition includes one or more other anti-cancer compounds. For example, administered compositions can comprise standard of care agents for the type of tumors selected for treatment.
In another aspect, kits are provided. Kits according to the invention include package(s) comprising compounds or compositions of the invention. In one embodiment, kits comprise Compound (1 ), or a pharmaceutically acceptable sait thereof, and a compound selected from the group consisting of Compound (2a) and Compound (2b), or a pharmaceutically acceptable sait thereof.
The phrase package means any vessel containing compounds or compositions presented herein. In some embodiments, the package can be a box or wrapping. Packaging materials
-1616446 for use in packaging pharmaceutical products are well-known to those of skill in the art. Examples of pharmaceutical packaging materials include, but are not limited to, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, bottles, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
The kit also can contain items that are not contaîned within the package but are attached to the outside of the package, for example, pipettes.
Kits can contain instructions for administering compounds or compositions of the invention to a patient. Kits also can comprise instructions for approved uses of compounds herein by regulatory agencies, such as the United States Food and Drug Administration. Kits also can contain labeling or product inserts for the inventive compounds. The package(s) and/or any product insert(s) may themselves be approved by regulatory agencies. The kits can include compounds in the solid phase or in a liquid phase (such as buffers provided) in a package. The kits also can include buffers for preparing solutions for conducting the methods, and pipettes for transferring liquids from one container to another.
Examples hâve been set forth below for the purpose of illustration and to describe certain spécifie embodiments of the invention. However, the scope of the claims is not to be in any way limited by the examples set forth herein.
Example 1. In vitro activity of Compound (1 ) in combination with Compound (2b)
This study describes the activity of individual anticancer agents Compound (1) and Compound (2b), as well as their combination, in a panel of 81 cancer cell lines. Cell lines were selected to represent 17 different indications with many different genetic variations and biochemical characteristics. In addition, the study included resting Peripheral Blood Mononuclear Cells, PBMC, as a model for non-proliferating cells. The results of individual activity profiles were further used to perform a combination study of Compound (1) and Compound (2b) using a panel of 81 cell lines. The study also compared the activity profiles of Compound (1) and Compound (2b) with profiles of more than 300 known anticancer agents.
Prior to in vitro combination studîes, the activity of individual agents was investigated using a panel of 82 cell lines. The purpose of testing individual agents was to détermine the independence of their action. In addition, comparison to an activity profile of known anticancer agents may help form a hypothesis regarding potential mechanisms of the compounds’ action.
Materials and methods
-1716446
Cell lines were purchased directly from the ATCC, NCI, CLS, and DSMZ cell line collections. A master bank and working aliquots were prepared. Cells used for the study had undergone less than 20 passages. To ensure the absence of potential contamination and wrong assignment, ail cell lines were tested on the Whole Genome Array (Agitent, USA) and by STR analysis. Absence of mycoplasma and SMRV contamination was confirmed for ail cell lines used in the studies.
The cell lines were grown in the media recommended by the suppliers in the presence of 100 U/ml penicillin G and 100 pg/ml streptomycin supplied with 10% FCS (PAN, Germany). The RPMI 1640, DMEM, and MEM Earle’s medium were from Lonza (Cologne, Germany), suppléments 2mM L-glutamine, 1 mM Na-pyruvate and 1% NEAA were from PAN (Aidenbach, Germany), 2.5% horse sérum and 1 unit/ml insulin from Sigma-Aldrich (Munich, Germany). RPMI medium was used for culturing the foilowing cell lines: 5637, 22RV1, 7860, A2780, A431, A549, ACHN, ASPC1, BT20, BXPC3, CAKI1, CLS439, COLO205, COLO678, DLD1, DU 145, EFO21, EJ28, HCT15, HS578T, IGROV1, JAR, LOVO, MCF7, MDAMB231, MDAMB435, MDAMB436, MDAMB468, MHHES1, MT3, NCIH292, NCIH358M, NCIH460, NCIH82, OVCAR3, OVCAR4, PANC1005 (addition of insulin), PBMC, PC3, RDES, SF268, SF295, SKBR3, SKMEL28, SKMEL5, SKOV3, SW620, U2OS, UMUC3, and UO31.
DMEM was used for A204, A375, A673, C33A, CASKI, HCT116, HEPG2, HS729, HT29, J82, MG63, MIAPACA2 (addition of horse sérum), PANC1, PLCPRF5, RD, SAOS2, SKLMS1, SKNAS, SNB75, T24, and TE671.
MEM Earle’s medium was used for CACO2, CALU6, HEK293, HELA, HT1080, IMR90, JEG3, JIMT1, SKHEP1, SKNSH, and U87MG.
Cells were grown in 5% CO2 atmosphère in a HeraCell 150 incubator (Thermo Scïentific, Germany).
The foilowing is a list of compounds used in the studies:
Container bar code Amount supplied Dissolved in Concentration of a stock solution (max. final concentration) Supplier
Compound (D 10.27 mg 439 pl DMSO 50 mM (50 μΜ) EMD Serono (Rockland MA, USA)
-1816446
Compound (2b) 10.3 mg 762 μΙ DMSO 50 mM (50 μΜ) EMD Serono (Rockland MA, USA)
5-FU NA DMSO 100πιΜ(100μΜ) Lot #22808088 (SigmaAldrich)
Paclitaxel NA DMSO 10 mM (10 μΜ) Lot #ASM- 110 (LC Laboratories)
The stock solutions of Compound (1) and Compound (2b) were prepared in DMSO (SigmaAldrich, Germany) as indicated in table above. Stock solutions were further aliquoted and stored under argon at -20°C.
10% w/v of trichloracetic acid, TCA (Sigma-Aldrich, Germany), was prepared in distilled water. 0.08% wt/v sulforhodamine B, SRB (Sigma-Aldrich, Germany) solution was prepared in 1% acetic acid (Sigma-Aldrich). Tris base was purchased from Karl Roth (Germany).
Cell growth and treatment were performed in 96-well microtitre plates CELLSTAR®(Greiner Bio-One, Germany). Cells harvested from exponential phase cultures by trypsinization were plated in 150 pl of media at optimal seeding densifies. The optimal seeding densifies for each cell line were determined to ensure exponential growth for the duration of the experiment. Ail cells growing without anticancer agents were sub-confluent by the end of the treatment as determined by Visual inspection.
Compound dilutions in DMSO were performed in 96-well rigid PCR plates. Compounds were then diluted 1:250 in RPMI medium.
150 μΙ of cells, after a 24-hour pre-growth period, were treated by mixing with 50 μΙ of the compound containing media (resulting in a final DMSO concentration of 0.1%). The cells were allowed to grow at 37°C for 72 hours. In addition, ail experiments contained a few plates with cells that were processed for measurement immediately after the 24 hours recovery period. These plates contained information about the cell number that existed before treatment, at time zéro, and served to calculate the cytotoxicity.
-1916446
After treatment, cells were precipitated by addition of 10% TCA. Prior to fixation, the media was aspirated as described. After an hour of incubation at 4°C, the plates were washed two times with 400 pl of deionized water. Cells were then stained with 100 pl of a 0.08% wt/v SRB. The plates were allowed to sit for at least 30 min. and washed six times with 1% acetic acid to remove unbound stain. The plates were left to dry at room température and bound SRB was solubilized with 100 μΙ of 10 mM Tris base. Measurement of optical density was performed at 560 nm on a Victor 2 plate reader (Perkin Elmer, Germany). The SRB values for A375 and H460 cell lines were near to saturation (2.5 OD units) due to the high protein content of these cells, but not cell confluence. The measurements for these cells were performed at 520 nm instead of 560 nm.
Prior to in vitro combination studies, the activity of indivîdual agents was investigated using a panel of 80 cell lines. The purpose of testing indivîdual agents was to détermine the independence of their action. In addition, comparison to an activity profile of known anticancer agents may help form a hypothesis regarding potential mechanisms of the compounds’ action.
The calculations used nomenclature introduced by DTP NCI. Unprocessed optical density data from each microtitre plate were stored in MS Excel or as a text file in a databank. The first step of data processing was calculating an average background value for each plate, derived from wells containing medium without cells. The average background optical 20 density was then subtracted from the appropriate control values (containing cells without addition of a drug), from values representing the cells treated with an anticancer agent, and from values of wells containing cells at time zéro. Thus the following values were obtained for each experiment: control cell growth, C; cells in the presence of an anticancer agent T( and cells prior to compound treatment at time zéro, Tz (or To, in some publications).
The Z -factor is a parameter commonly used to assess quality of the assay performance and was calculated according to the following équation:
= 1 _ + ) — Mewhere pc+ and pc_ are denoted for the means of positive and négative control signais and oc+ and Oc- are their standard déviation. In a way, the Z '-factor reflects the significance of the dynamic range of the measurements recorded and should be >0.5. In this study, Z '-factor was applied to détermine the significance of signais over background for Tz and C values.
The results of the screening were accepted only if the Z-factor was above 0.5 for each case.
-2016446
The non-linear curve fitting calculations were performed using in-house developed algorithms and visualization tools. The algorithms are similar to those previously described and were complemented with the mean square error or MSE model. This can be compared to commercial applications, e.g. XLfit (ID Business Solutions Ltd., Guild-ford, UK) algorithm “205. The calculations included the dose response curves with the best approximation line, a 95% confidence interval for the 50% effect (see below).
A common way to express the effect of an anticancer agent is to measure cell viability and survival in the presence of the test agent as % T/C * 100. The relationship between viability and dose is called a dose response curve. Two major values are used to describe this relationship without needing to show the curve: the concentration of test agents giving a % T/C value of 50%, or 50% growth inhibition (IC50), and a % T/C value of 10%, or 90% growth inhibition (IC90).
Using these measurements, cellular responses can be calculated for incomplète inhibition of cell growth (Gl), complété inhibition of cell growth (T Gl) and net loss of cells (LC) due to compound activity. Growth inhibition of 50% (GI50) is calculated as 100 χ [(Τι - TZ)/(C - Tz)] = 50. This is the drug concentration causing a 50% réduction compared to the net protein increase in control cells during the drug incubation period. In other words, GI50 is IC50 corrected for time zéro. Similar to ICgo, calculated GI0O values are also reported for ail compounds tested. TGI was calculated from Tt = Tz. LCM is the concentration of drug causing a 50% réduction in the measured protein at the end of the drug incubation period compared to that at the beginning. It was calculated as 100 *[(T| -TZ)/TJ = -50. However, due to 72 hours treatment, low cell seeding density was required and LC50 could rarely be achieved.
The ICso, IC90, GIsq, Glgo and T Gl values were computed automatically. Visual analysis of ail dose response curves was performed to check the quality of the fitting algorithm. In cases where the effect was not reached or exceeded, the values were either approximated or expressed as In this study ail values were greater than the maximum drug concentration tested. In these cases, the values were either excluded from the analysis, or approximation of IC10 and GI10 were used for analysis.
AU values were Iog10-transformed for analysis. This transformation ensures better data fitting to the normal distribution, a prerequisite to apply any statistical tool. Statistical analyzes were performed using proprietary software developed at Oncolead integrated as a database analysis tool. However, except for database comparison, the analysis can be reproduced using either MS Excel or STATISTICA®(StatSoft, Hamburg). Using MS Excel:
identification of mean, e.g. mean GI50 (function: “Average”); calculation of ,δ, delta (GI50 mean GI50 ); and z-score (function Standardize). Comparison of the activity profile of
-21 16446
Compound (1) and Compound (2b) cross-correlation could be performed using Pearson and Spearman corrélations (for example by using STATISITCA(R)). In addition, Pearson pairwise and Spearman pairwise comparisons were used to increase the confidence of the results. Pairwise comparison was calculated based on pairwise similarity of the agents to ail tested agents in the database.
Z-score is a way to report standard déviations rather than absolute deltas and mean values.
It indicates how far the value deviated from its mean in units of standard déviation:
„ x-μ à σχ σχ where X is a single measured value, e.g. GI50, and μ is a mean of ail measured values (mean Glso) and σχ is a standard déviation of X.
The concept of the mean graph introduced by NCI permits visualization of a cell activity parameter for a given anticancer drug in ail cells. This graph yields a characteristic pattern that provides rich information for Visual comparison. The values are plotted as horizontal bars from the mean values. Each bar, therefore, represents the relative activity of the compound in the given cell lines deviating from the mean in ail cell lines. In contrast to NCI, z-score values were plotted rather than absolute delta. In statistical terms, z-values represent a standard déviation that provides a kind of normalization and simplifies comparison between compounds with different activity distributions. In addition, an averaged combined z-score was calculated for cell lines of the same origin.
Z-score values as well as the range of tested concentrations were included in ail visualizations. The applicability of z-score graphs should be considered with précaution if the agent’s activity does not follow the normal distribution.
The most sensitive and non-sensitive cell lines were visualized by using either a box-plot graph or by selecting the eight most and least sensitive cell lines using the z-score for each agent. This also applied to the cell lines where activity of an agent could not be determined. Box plots were constructed from five values: the smallest value (the lowest whisker), the first quartile (the lowest border of the box), the médian (square in the middle), the third quartile (the upper border of the box), and the largest value (the highest whisker).
The screening was designed to détermine potential synergistic combinations. Ail and/or part of the 5x5 or 7x7 matrix were used to design the study. Bliss independence was used as a basis for calculations, unless otherwise stated. The following parameters were calculated:
0, = Measured valuet — Theorettcal values
-2216446 where i = [1 ,.n] is one of the values of the matrix used and theoretical value, calculated as described for the Bliss Independence method. Vector sum was determined as:
n
Kectorsum = Sign(Ef fectj)Ef feci?
i = l in this term the Vector Sum rather represents scalar:
1
Vector sum average = —57 Effectj = Mean(Ef fecfj) ”i=l
The average values below -0.5 indicate a strong synergy effect: (-0.5, -.02) - Synergy effect, (-0.2, .02) -Zéro effect (additivism), (0.02, 0.5) - potential antagonism, and above 0.5 -strong antagonism. However, it is possible that the effect of the combination is not synergistic (or even antagonistic) but still better than each of the agents alone. Moreover, in vivo, any effect better than a single agent is considered clinically positive (or synergistic). In this case, one considère a potential interaction of two agents that can be determined by the highest single agent, HSA, model. This model détermines the différence between the larger effects produced by one of the single agents at the same concentrations as in the mixture.
Single Beati = Beat o/[A<?ent 1<;
and delta HSA, for two agents can be determined as:
delta H S Ai = ôHSA, = Measured value, — Single Beat, and ν' .4 verage HSA Ef fect = —,-> ----20 Summary of in vitro results
Efficacy of Compound (1) varies broadly from 4-5 nM in sensitive cell lines to minimal activity at 50 pM in the most non-sensitive cell lines. Under the conditions tested, minimal activity could be determined for cancer cell lines: A673, HEK293, J82, JAR, JEG3, MDAMB436, MDAMB468, MHHES1, NCIH82, PANC1, PLCPRF5, and SF268. For cell
Unes CLS439, EFO2,1 PC3, SAOS2, SF295, and SKOV3, activity was estîmated above the highest tested concentration of 50 pM. At the same time, 50% of the cell lines tested exhibited a sensitivity below 500 nM (the médian is 490 nM), and 27 of 82 cell lines were found to be sensitive below 100 nM of Compound (1 ). Action of Compound (1) and Compound (2b) was synergistic in a larger number of human cancer cell lines, which suggests that the mechanisms of compound action are complementary. A673 cells are nonsensitive to the action of Compound (1) or Compound (2b) alone, but can show strong -2316446 synergy in combination. A549 and MCF7 cells show some sensitivity to both agents, which can be further potentiated with their combination. SKBR3 cell line is very sensitive to Compound (2b). However, the effect can be further increased by the combination of both agents. These findings may be related to the ail breast cancer cell Unes with overexpression of the HER2 gene.
The most sensitive cell fines were HT29, COLO205, TE671, A375, SKMEL5, COLO678, SKNAS, and NCIH292, where Compound (1) showed activity between 4.8 and 8 nM. The différence between the most and least sensitive cell lines was as large as 10,000-fold. Due to such a large window of activity, the activity distribution is broad and does not follow a normal distribution. In such a case, z-score has little statistical meaning; however, it can still be applicable, for example, to group activities according to therapeutic indications.
The rank of Compound (1 ) activity (or rank of z-score values) is another tool that can be applied. These properties of Compound (1 ) stress the necessity of using diverse analysis tools and covering a broad concentration range to test anticancer agents. One possibility is that Compound (1) has a spécifie mechanism of action and acts only on a sub-population of tumor cells.
The 81 human cancer cell lines represented 17 different tumor origins. Figures 15A and 15B show individual z-scores within one tumor origin group, as well as combined z-scores for each therapeutic indication as an average value (green triangle). As in the case of individual z-scores, direction to the left points towards sensitivity to the compound action. A zeroline corresponds to average activity. The data suggest that lung, pancréas, colon, and melanoma cell lines are generally more sensitive to Compound (1), since the average value of z-scores are on the left. Ail but one pancréas (PANC1 ) cell line are very sensitive to Compound (1) action. HT1080 is also a very sensitive cell line. .
Activity, Glgo values, of Compound (2b) in cell lines ranged between < 500 nM in A204, IMR90, MDAMB468, SKBR3, CAKI1, and IGROV1 (most sensitive, as determined by zscore < -1.5) and > 4 pM in SW620, COLO678, and HCT116 (non-sensitive cell lines, z score > 1.5). These results may indicate that cell lines showing the strongest négative déviation of z-scores from the mean will also show activity in other biological Systems, e.g., mouse xenograft models. The average GI50 value in ail 81 cell lines was 1.3-1.4 pM, calculated based on Iog10-transformed data. No activity was shown in arrested PBMC suggesting that Compound (2b) may act preferably on proliferating cells. Figures 16A and 16B show that the activity distribution is narrow, but sensitive cell lines can be welldiscriminated.
-2416446
Comparison of the Compound (2b) activity profile with an internai databank containing more than 300 different anticancer agents identified a number of agents. The most similar agent (average similarity above 0.8) is MSC2208382A. Weaker similarity (above 0.7) is detected with GDC-0941 bismesylate and ZSTK474, and some degree of similarity to MSC2313080A. GDC-0941 bismesylate is an analog of PI-103, a dual PI3K/mTOR inhibitor and considered to be a relatively spécifie inhibitor of class I PI3K enzymes as well as ZSTK474. It could be suggested that Compound (2b) belongs to the class of PI3K inhibitors.
As in the case of individual z-scores, the direction to the left points towards sensitivity to the compound action. A zero-line corresponds to average activity. Ovarian and prostate tumors could be spécifie therapeutic areas. At least for ail cell lines tested, the z-score is below zéro. Applications for breast, lung, and rénal tumors also could be considered. However, each of the indications contains cell lines either very sensitive or non-sensitive to Compound (2b) action.
Although most of the cell lines showed potential synergy for in vitro combination of Compound (1) and Compound (2b), the results with a vector sum of below -1 can be considered significant. Table 1 and Figure 17 summarize the results. Cell line A673 is nonsensitive to the action of Compound (1 ) or Compound (2b) alone, but shows strong synergy in combination. However, from in vivo or clinical perspectives, cell line groups four and five are probably more relevant. Activity (GIM) of Compound (1) is 300 nM and 150 nM in A549 and MCF7 cells, respectively, which is comparable with 100 nM activity in the most sensitive cell lines. Activity (GIM) of Compound (2b) is 1.15 μΜ and 1.6 pM in A549 and MCF7 cells, respectively, below or close to the average activity of 1.3-1.4 pM for this agent. The combination index for these cell lines is close to -1, which is indicative of synergy. Another example is SKBR3. This cell line is very sensitive to Compound (2b) and non-sensitive to Compound (1 ). However, the effect can be further increased by the combination of both agents.
Compound (1) and Compound (2b) act on proliferating cells and showed no activity in resting PBMC. However, these agents differ in their activity. The différence between the most and least sensitive cell lines for Compound (1 ) was as large as 10,000-fold. For the most insensitive cell lines, résistance extends beyond the tested concentration range > 50pM.
Thus, it appears that Compound (1 ) may hâve a spécifie mechanism of action and acts only on a sub-population of tumor cells. Sélection of therapeutic indications in the clinic can be complemented by the mutational analysis. In contrast, Compound (2b) shows narrow activity in cell lines. The séparation between sensitive and insensitive cell lines is statistically significant but the différences in activity are in the range of 10-20-fold. The
-2516446 activity profile of Compound (2b) has similarities to the PI3K inhibitors, e.g. PI-103 or its pharmalog GDC-0941. No prédiction could be made about the agent's activity and the mutational status of genes involved in activation of the PI3K pathway, e.g. EGFR, PTEN, and PI3K. Some markers may be prédictive for induction of apoptosis upon action of this
PI3K inhibitor: EGFR (mutation), HER2 (amplification), MET (mutation/amplification).
Indirectly, this fact can be supported by the observation that SKBR3 cells (HER2 amplification) were among the most sensitive cell lines.
Compound (1 ) and Compound (2b) were further tested in combination in ail cell lines using a 7x7 matrix, with variation around GI50 averaged in ail cell lines for each of the agents. The 10 rationale for selecting this concentration was as follows. First, this concentration is a reference concentration that describes efficacy of the anticancer agents in cellular models, i.e. only cell lines that show significant effects below mean GI50. Second, it is known that efficacy of anticancer agents is limited, based on citations reporting 10-30%. Therefore, sélection of mean Gl® would correspond to the expected efficacy of approximately 50%.
Third, the variation spanned by the 7 x 7 matrix (almost ten-fold in both directions from the mean GI50) allows enough coverage to address the question of whether there are any potential interactions between the two agents.
In almost ail cases, Compound (1) and Compound (2b) in combination showed potential to be synergîstic (Figure 17), as determined by the Bliss Independence model (see, for example, Yan et al., BMC Systems Biology, 4:50 (2010)). See also Figures 18A, 18B, 18C, 18D, 18E, 18F, 19A, 19B, 20A, 20B.
However, the strongest synergîstic effect was detected when the activity of either agent was weak. This may be attributed, at least in part, to experimental set-up, i.e., any effect of combination is considered significant if the agents alone médiate little, if any effect on the 25 cells. Alternatively, the effect of a single agent can be too strong to detect increasing effects. In the later case, the HSA model provides a better view of the potential interaction between two agents.
Example 2. In vivo activity of Compound (1) in combination with Compound (2b) or
Compound (2a) against subcutaneous human colon carcinoma HCT 116 bearinq SCIP mice
To evaluate the antitumor activity of the MEK inhibitor Compound (1 ) in combination with the pan-PI3K inhibitor Compound (2a) or the dual pan-PI3K / mTOR inhibitor Compound (2b), experiments were conducted using female SCID mice bearing human colon carcinoma HCT
116 (KRAS and PIK3CA mutant) xenografts. Four studies were performed:
-2616446
In a first study, a low dose of Compound (1) at 5 mg/kg was tested in combination with
Compound (2b) at 30 mg/kg and Compound (2a) at 50 and 75 mg/kg.
In a second study, the dose of Compound (1 ) was increased to 10 and 20 mg/kg in combination with Compound (2b) at 20 mg/kg, and Compound (1) at 10 mg/kg was combined with Compound (2a) at 50 and 75 mg/kg.
In a third study, used as a confirmation study, the dose of Compound (1) was used at 10 and 20 mg/kg in combination with Compound (2a) at 50 and 75 mg/kg.
In a fourth study, used as a confirmation study, the dose of Compound (1) was used at 10 and 20 mg/kg in combination with Compound (2b) at 20 mg/kg.
Materials and methods
CB17/ICR-Prkdc severe combined immunodeficiency (SCID) /Cri mice, at 8-10 weeks old, were bred at Charles River France (Domaine des Oncins, 69210 L'ArbresIe, France) from strains obtained from Charles River, USA. Mice were over 18 g at start of treatment after an acclimatization time of at least 5 days. The mice had free access to food (UAR reference 113, Villemoisson, 91160 Epinay sur Orge, France) and stérile water. The mice were housed on a 12 hours light/dark cycle. Environmental conditions including animal maintenance, room température (22°C ± 2°C), relative humidity (55% ± 15%) and lighting times were recorded by the supervisor of laboratory animal sciences and welfare (LASW) and archived.
Human colon carcinoma HCT 116 cells were purchased at American Type Culture Collection [(ATCC), Rockville, MD, USA). The HCT 116 cells were cultured in Dulbecco's modified Eagle's medium (DMEM) (Invitrogen). The tumor model was established by împlanting (SC) 3x106 cells mixed with 50% matrigel (Reference 356234, Becton Dickinson Biosciences) per SCID female mice.
Compound (1) formulation was prepared by incorporating the MEK inhibitor into 0.5% CMC 0.25% Tween 20. The préparation was stored at 4°C and resuspended by vortexing before use. The oral form of the compound was prepared every 3 days. The volume of administration per mouse was 10 ml_/kg.
Compound (2a) formulation was prepared in water for injection. The stock solution was chemically stable 7 days in the dark at 4°C. The volume of administration per mouse was
10mL/kg.
-2716446
Compound (2b) formulation was prepared in 1N HCl and water for injection followed by five cycles of vortexing and sonicating. The pH of the final solution was 3. The stock solution was chemically stable 7 days in the dark at 4°C. The volume of PO administration per mouse was 10 mL/kg.
For subcutaneous implantation of tumor cells, skin in the flank of the mice was disinfected using alcohol or Betadine® solution (Alcyon) and a suspension of tumor cells was inoculated SC unilaterally under a volume of 0.2 mL using a 23 G needle.
The activity on tumor growth of Compound (1), Compound (2a) and Compound (2b) used as single agent or in combination was evaluated in four different studîes. The dosages and schedule of administration for each study are described in the results section and detailed in the tables that follow.
The animais required to begin a given experiment were pooled and implanted monolaterally on day 0. Treatments were administered on measurable tumors. The solid tumors were allowed to grow to the desired volume range (animais with tumors not in the desired range were excluded). The mice were then pooled and unselectively distributed to the various treatment and control groups. Treatment started 11 days post HCT 116 tumor cell implantation as indicated in the results section and in each table. The dosages are expressed in mg/kg, based on the body weight at start of therapy. Mice were checked daily, and adverse clinical reactions noted. Each group of mice was weighed as a whole daily until the weight nadir was reached. Then, groups were weighed once to thrice weekly until the end of the experiment. Tumors were measured with a caliper 2 to 3 times weekly until final sacrifice for sampling time, tumor reached 2000 mm3 or until the animal died (whichever cornes first). Solid tumor volumes were estimated from two-dimensional tumor measurements and calculated according to the following équation:
Tumor weight (mg) = Length (mm) x Width2 (mm2)/2
The day of death was recorded. Survïving animais were sacrificed and macroscopie examination of the thoracic and abdominal cavities was performed.
A dosage producing a 15% body weight loss (BWL) during three consecutive days (mean of group), 20% BWL during 1 day or 10% or more drug deaths was considered an excessively toxic dosage. Animal body weights included the tumor weight.
The primary efficacy end points are ΔΤ/ΔΟ, percent médian régression, partial and complété régressions (PR and CR).
-2816446
Changes in tumor volume for each treated (T) and control (C) group were calculated for each tumor by subtracting the tumor volume on the day of first treatment (staging day) from the tumor volume on the specified observation day. The médian ΔΤ is calculated for the treated group, and the médian AC is calculated for the control group. Then the ratio ΔΤ/AC is calculated and expressed as a percentage. The dose is considered as therapeutically active when ΔΤ/AC is lower than 40% and very active when ΔΤ/AC is lower than 10%. If ΔΤ/AC is equal to or lower than 0, the dose is considered as highly active and the percentage of régression is dated.
The percent of tumor régression is defined as the % of tumor volume decrease in the treated group at a specified observation day compared to its volume on the first day of treatment. At a spécifie time point and for each animal, % régression is calculated. The médian % régression is then calculated for the group using the following équation:
% régression (at t) = (volume at to - volume at t)/volume at to) x 100
Partial régression: Régressions are defined as partial if the tumor volume decreases to 50 % of the tumor volume at the start of treatment.
Complété régression: The CR is achieved when tumor volume - 0 mm3 (CR is considered when tumor volume cannot be recorded).
The term “therapeutic synergy is used when the combination of two products at given doses is more efficacîous than the best of the two products alone considerîng the same doses. In order to study therapeutic synergy, each combination was compared to the best single agent using estimâtes obtained from a two-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume.
Statistical analyses were performed on SAS system release 8.2 for SUN4 via Everstat V5 software and SAS 9.2 software. A probability less than 5% (p<0.05) was considered as signîficant.
Results of in vivo studies
First study: antitumor activity of Compound (1)(5 mg/kg) in combination with Compound (2b) (30 mg/kg) or Compound (2a) (50 and 75 mg/kg) against HCT 116 bearinq SCID mice
The médian tumor burden at start of therapy was 198 to 221 mm3. As single agents,
Compound (1) (5 mg/kg/administration (Adm)), Compound (2b) (30 mg/kg/adm) and
Compound (2a) (50 and 75 mg/kg/adm) were administered PO daily from days 11 to 18 post
-2916446 tumor implantation. In the combination groups, the dose of Compound (1) was combined with each dose of Compound (2a) and Compound (2b), as shown in Table 2.
As single agents or used in combination, Compound (1 ) and Compound (2a) were welltolerated, inducing minimal BWL (Figure 1 and Table 2). As single agents, Compound (1), Compound (2a) and Compound (2b) achieved a AT/AC>40 %) under these test conditions.
In combination, treatment with Compound (1) at 5 mg/kg/adm and Compound (2b) at 30 mg/kg/adm achieved a ΔΤ/AC of 27 % (Figure 2 and Table 1 ), but as shown by Table 3, therapeutic synergy was not reached (p = 0.0606 for global analysis). Treatment with Compound (1) at 5 mg/kg/adm and Compound (2a) at 50 and 75 mg/kg/adm achieved a ΔΤ/AC of 22 % and 21 %, respectively (Figure 3 and Table 2). As shown by Table 2, therapeutic synergy was achieved for both combinations (p=0.0091 and p<0.0001 globally, respectively). See also Tables 11A and 11 B.
Second study: antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2b) (20 mq/kq) and Compound (1) (10 mg/kg) in combination with Compound (2a) (50 and 75 mq/kq) against HCT 116 bearinq SCIP mice
The médian tumor burden at start of therapy was 180 to 198 mm3. As single agents, Compound (1)(10 and 20 mg/kg/adm), Compound (2b) (20 mg/kg/adm) and Compound (2a) (50 and 75 mg/kg/adm) were administered PO daily from days 11 to 18 post tumor implantation. In the combination groups, the dose of Compound (1) was combined with each dose of Compound (2a) and Compound (2b), as shown in Table 3.
As single agents, Compound (1), Compound (2a) and Compound (2b) were well-tolerated, inducing minimal BWL (Figure 4 and Table 4).
As single agents, Compound (1) (10 and 20 mg/kg/adm) achieved a ΔΤ/AC of 20 % and 22 %, respectively, while Compound (2b) at 20 mg/kg/adm achieved a AT/AC>40 %. As shown in Table 4, Compound (2a) at both doses tested achieved a AT/AC>40%.
In combination, treatment with Compound (1) at 10 or 20 mg/kg/adm and Compound (2b) at 20 mg/kg/adm achieved a ΔΤ/AC of 0, and therapeutic synergy was reached with Compound (1 ) at 10 mg/kg/adm (p=0.0004 globally). As shown by Table 5, therapeutic synergy was not reached with Compound (1) at 20 mg/kg/adm (p=0.2169 globally). Partial régression (PR) was observed in 2/7 mice for the combination treatment of Compound (1) at 10 mg/kg/adm and Compound (2b) at 20 mg/kg/adm (Figure 5 and Table 4). When Compound (1) was used at 10 mg/kg/adm, the combinations with Compound (2a) at 75 and 50 mg/kg/adm achieved, respectively a ΔΤ/AC of 5 % and ΔΤ/Δΰ<0, with 1/7 PR occurring for both combination treatments (Figure 6 and Table 4). As shown by Table 5, both
-3016446 combinations (p=0.0063 and p=0.0019 globally, respectively) achieved therapeutic synergy.
In ail combination groups, tumor stasis was achieved (Figure 5 and Figure 6). See also
Tables 12A and 12B below.
Third study: antitumor activity of Compound (11 (10 and 20 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against HCT 116 bearing SCIP mice
The médian tumor burden at start of therapy was 187 to 189 mm3. As single agents, Compound (1)(10 and 20 mg/kg/adm) and Compound (2a) (50 and 75 mg/kg/adm) were administered PO daily from days 11 to 20 post tumor implantation. In the combination groups, the dose of Compound (1) was combined with each dose of Compound (2a), as shown in Table 6.
As single agents, Compound (1) and Compound (2a) were well-tolerated, inducing minimal BWL (Figure 7 and Table 6).
As a single agent, Compound (1) achieved a AT/AC of 34 % at a dose of 20 mg/kg/adm and AT/AC>40 % at a dose of 10 mg/kg/adm (Figure 7). As shown in Table 6, Compound (2a) at both doses tested achieved a AT/AC>40 %.
In the combination, treatment with Compound (1 ) at 10 or 20 mg/kg/adm and Compound (2a) at 75 mg/kg/adm achieved AT/AC of 18 % and 9 %, respectively) (Figure 10 and Table 6), and therapeutic synergy was reached (p=0.0109 and p=0.0003 globally, respectively) (Table 6). The treatment with Compound (1) at 10 or 20 mg/kg/adm and Compound (2a) at 50 mg/kg/adm achieved AT/AC of 19 % and 22 %, respectively) (Figure 10 and Table 6). Therapeutic synergy was reached only for the combination with Compound (1) at 10 mg/kg (p=0.0088 globally) (Table 7). As shown by Table 7, therapeutic synergy was not reached with Compound (1 ) at 20 mg/kg/adm (p=0.0764 globally). In ail combination groups, tumor stasis was achieved (Figure 8). See also Table 13 below.
Fourth study: antitumor activity of Compound (1) (10 and 20 mq/kq) in combination with Compound (2b) (20 mg/kg) against HCT 116 bearing SCIP mice
The médian tumor burden at start of therapy was 189 to 196 mm3. As single agents, Compound (1) (10 and 20 mg/kg/adm) and Compound (2b) (20 mg/kg/adm) were administered PO daily from days 11 to 20 post tumor implantation. In the combination groups, the dose of Compound (2b) was combined with each dose of Compound (1), as shown in Table 8.
As single agents, Compound (1) and Compound (2b) were well-tolerated, inducing minimal BWL (Figure 9 and Table 8).
-31 16446
As single agents, Compound (1) (10 and 20 mg/kg/adm) and Compound (2b) at 20 mg/kg achieved a AT/AC>40 % (Figure 10 and Table 8).
In the combination, the treatment with Compound (1) at 10 or 20 mg/kg/adm and Compound (2b) at 20 mg/kg/adm achived a ΔΤ/AC of 30 % and 15 %, respectively (Figure 10 and Table 8), and therapeutic synergy was reached (p=0.0002 and p=0.0008 globally, respectively) (Table 9). See also Table 14 below.
Example 3. In vivo activity of Compound (1) in combination with Compound (2a) or Compound (2b) against subcutaneous human pancreatic MiaPaCa-2 bearing nude mice
To evaluate the antitumor activity of the MEK inhibitor Compound (1) (5 mg/kg) in combination with the pan-PI3K inhibitor Compound (2a) (50 mg/kg) or the dual pan-PI3K / mTOR inhibitor Compound (2b) (30 mg/kg), experiments were conducted using female nude mice bearing human pancreatic MiaPaCa-2 (KRAS mutant) xenografts.
A low dose of Compound (1 ) at 5 mg/kg was tested in combination with Compound (2b) at 30 mg/kg and Compound (2a) at 50 mg/kg.
Materiais and methods
The human pancreatic cancer cell line MiaPaCa-2 (American Type Culture Collection, Manassas VA), was cultured in MEM medium containing 10% fêtai bovine sérum, 1% essential amino acid, 1% sodium pyruvate (Life Technologies, Carlsbad, CA). Cells were trypsonized during the log phase of growth at 60-85% confluence, collected and washed once with PBS. Cells were re-suspended in PBS (Life Technologies, Carlsbad, CA) and then mixed 1:1 with Matrigel (BD Biosciences, San José, CA). Cells were stored at 4°C until implantation.
MiaPaCa-2 cells (10x10e in a 200μΙ PBS:Matrigel (1:1) suspension) were subcutaneously injected into the right flank area of female nude (Crl:NU-Foxn1nu) mice (6-8 weeks old, Charles River Laboratories, Wilmington, MA). Ail mice in this study were used according to the guidelines approved by the EMD-Serono Institutional Care and Animal Use Committee (IACUC), #07-003.
A solution of 0.5% CMC (carboxymethylcellulose; Sïgma-Aldrich, St Louis, MO) and 0.25%
Tween 20 (Acros Organics, Morris Plains, NJ) in water was used as the vehicle for this study. Compound (1) (Lot #27) was prepared by suspending 10 mg of compound in 20 mL of 0.5% CMC 0.25% Tween 20 in water to make a 0.5 mg/mL (5.0 mg/kg) dosing solution.
-3216446
Compound (2a) was weighed (5 mg for 1 mL of solution) and water added for injection (60% of final volume i.e. 0.60 ml). Solution was mixed via five cycles of vortexing and sonicating in a sonicating water bath for 1 min each. Completed with water for dosing. Compound (2b) was weighed (3 mg for 1 mL of solution), 10 pL HCl 1N was added and then water was added for injection (60% of final volume i.e. 0.60 ml). Solution was mixed via five cycles of vortexing and sonicating in a sonicating water bath for 1 min each. 1N NaOH was added to adjust the pH up to 3 and finally completed with water for injection.
Developing tumors located in the right flank area of female nude mice were measured over time with digital calipers. Seven days after cell implantation, the tumors had reached an average volume of 165 mm3 in an ample number of mice to begin the study. Mice bearing a tumor that was significantly different from the average tumor volume were excluded from the study. The remaining tumor-bearing mice were randomized into seven experimental groups (n=9), so that each group had the same mean tumor volume.
In ail combination groups, both agents were administered to the animais at the same time, within approximately 5-10 minutes of each other. The treatments began on the seventh day following implantation of the Miapaca-2 cells, which was designated as Day 0 for data évaluation purposes. Animais underwent 21 days of treatment. Body weights and tumor volumes were assessed twice per week post treatment initiation. On Day 22, ail animais were euthanized via progressive hypoxia with CO2.
Efficacy was determined by analyzing tumor volumes and the percent ΔΤ/Δ0 (%ΔΤ/ΔΟ). Tumor volume was determined by using the tumor length (I) and width (w) measurements and calculating the volume with the équation l*w*/2. The length was measured along the longest axis of the tumor and width was measured perpendicular to that length. The mean percent of actual tumor growth inhibited by the treatments was calculated as follows: [%ΔΤ/ΔΟ= ( (TVf- TV/TVfctri-TVictri)) x 100%], where TV=tumor volume, f=final, rinitial and Ctrl=control group. Tolerability was assessed by regarding percent body weight différence during the treatment period. Percent body weight différence was calculated as follows: [%Body weight différence = (BWC-BW() / BW, x 100%], where BW = body weight, c = current, / = initial.
Tumor volume data and percent body weight différences were analyzed by Repeated Measures Analysis of Variance (RM-ANOVA) followed by Tukey’s post-hoc multiple pairwise comparisons (□ = 0.05).
Results of in vivo studies
-3316446
No groups experienced more than 5% body weight loss during the study. No clinical signs were noted (Figure 11) for the combination with Compound (2a) or (Figure 12) for the combination with Compound (2b).
As single agents, Compound (1) (5 mg/kg/adm), Compound (2a) (50 mg/kg) and Compound 5 (2b) (30 mg/kg) achieved AT/AC>40 % in these assays (Figure 13 and 14 and Table 10).
In combination, treatment with Compound (1) at 5 mg/kg/adm and Compound (2b) at 30 mg/kg/adm achieved ΔΤ/AC = 27.3 % (Figure 14 and Table 10), and therapeutic synergy was reached (p<0.05) (Table 10). In contrast, the treatment with Compound (1) at 5 mg/kg/adm and Compound (2a) at 50 mg/kg/adm achieved AT/AC>40 % (Figure 13 and
Table 10), and therapeutic synergy was not reached (p>0.05) (Table 10).
Summary of in vivo results
The in vivo work presented here reports the in vivo antitumor activity of combining Compound (1), an oral potent and sélective allosteric inhibitor of MEK1/2, with oral, potent, and spécifie inhibitors of class I PI3K lipid kinases Compound (2a), a pan-PI3K inhibitor, and 15 Compound (2b), a dual pan-PI3K and mTOR inhibitor. This work has been performed against human colon carcinoma HCT 116 xenografts harboring a G13D activating mutation of KRAS and an activating mutation of PIKC3A known to reduce the sensitîvity to MEK inhibition and against human pancreatic MiaPaCa-2 xenografts harboring a KRAS mutation.
In the studies described above, combination treatment was highly effective in inducing a sustained tumor stasis during the treatment phase and realizing therapeutic synergy.
In conclusion, a potent antitumor activity with therapeutic synergy has been achieved in PIKC3A and KRAS mutant HCT 116 driven xenograft model when combining the inhibitor of MEK1/2 Compound (1 ) with Compound (2a), a pan-PI3K inhibitor, and in both PIKC3A and KRAS mutant HCT 116 driven xenograft model and KRAS mutant MiaPaCa-2 driven xenograft model, when combining Compound (1) with Compound (2b), a dual pan-PI3K and mTOR inhibitor.
Example 4. Fluorescence molecular tomooraphv study of combination of Compound (11 with Compound (2b) or Compound (2bj against subeutaneous human colon carcinoma HCT 116 bearing SCIP mice
To evaluate the apoptotic activity of the MEK inhibitor Compound (1 ) in combination with the pan-PI3K inhibitor Compound (2a) or the dual pan-PI3K / mTOR inhibitor Compound (2b), experiments were conducted using female SCIP mice bearing human colon carcinoma HCT
-3416446
116 (KRAS and PIK3CA mutant) xenografts in which apoptosis induction was monïtored non-invasively using fluoresence molecular tomography (FMT).
Methods
HCT116 tumor cells were implanted subcutaneously in the intra-scapular région in SCID mice. Implanted animais received 50 mg/kg Compound (2a) or 20 mg/kg Compound (2b) from day 11 to day 17, as single agents or combined with 10mg/kg Compound (1), Each agent was given by oral route on a daily schedule. Tumor growth was monïtored throughout the experiment by callipering the tumors. To quantify apoptosis, fluorescent Annexin-Vivo750 was injected intravenously one hour post daily treatment on days three and seven after start of treatment. Animais were imaged by FMT three hours post probe injection to document fluorescent Annexin uptake in the tumor. Ex vivo apoptosis was assessed on tumor lysâtes using Meso Scale Discovery assays for cleaved caspase-3 and cleavedPARP détection.
Results
Under these regimens, Compound (1 ), Compound (2a) and Compound (2b) used as single agents showed marginal activity on HCT116 tumor growth with ΔΤ/AC = 40% (NS), 36% (p= 0.023) and 80% (NS) respectively at the end of study (Figure 28). Conversely, both Compound (2a) and Compound (2b) in combination with Compound (1) induced strong tumor growth inhibition (AT/AC <0, associated with 23% médian tumor régression (p<0.0001)for Compound (2a)/ Compound (1) and (AT/AC <0 with 5% médian tumor régression (p= 0.0009) for Compound (2b)/ Compound (1 )). Both combination thérapies were associated with a clear enhancement of ex vivo cleaved caspase-3 (3.7 & 5.2 fold) (Figure 27B) and cleaved-PARP (8.4 & 12.8 fold) (Figure 27A) after four days treatment. Compound (2a)/ Compound (1) combination therapy was associated with a significant enhancement of Annexin-V-750 uptake in the tumor, reflecting apoptosis induction after three and seven days of combined therapy (p=0.005 and <0.0001) (Figure 26B). The ratios of Annexin fluorescence in treated animal groups relative to control were respectively 2.1 after 3 days and 3.8 after 7 days of combination therapy (Figure 26A).
Summarv
The combination of the MEK1/2 inhibitor Compound (1) with the Pan-PI3K inhibitor
Compound (2a) or the Pan-PI3K/mTOR Compound (2b) resulted in significantly enhanced anti-tumor activity in a dual KRAS/PIK3CA mutated tumor xenograft model, with synergistic induction of tumor apoptosis as demonstrated ex vivo for both combinations and in vivo using longitudinal FMT imaging for the Compound (2a)/ Compound (1) combination.
-3516446
Example 5. In vivo activity of Compound (1 ) in combination with Compound (2b) or Compound (2a) against subcutaneous human colon tumors CR-LRB-009C bearing SCIP female mice
To evaluate the antitumor activity of the MEK inhibitor Compound (1) in combination with the 5 pan-PI3K inhibitor Compound (2a) or the dual pan-PI3K / mTOR inhibitor Compound (2b), experiments were conducted using female SCID mice bearing human primary colon tumors CR-LRB-009C (KRAS and PIK3CA mutant) xenografts. In this study, Compound (1) at 20 mg/kg was tested in combination with Compound (2b) at 20 mg/kg and Compound (2a) at 75 mg/kg.
Materials and methods
CB17/ICR-Prkdc severe combined immunodeficiency (SCID) /Cri mice, at 8-10 weeks old, were bred at Charles River France (Domaine des Oncins, 69210 L'ArbresIe, France) from strains obtained from Charles River, USA. Mice were over 18 g at start of treatment after an 15 acclimatization time of at least 5 days. The mice had free access to food (UAR reference 113, Villemoisson, 91160 Epinay sur Orge, France) and stérile water. The mice were housed on a 12 hours light/dark cycle. Environmental conditions including animal maintenance, room température (22°C ± 2°C), relative humidity (55% ± 15%) and lighting times were recorded by the supervisor of laboratory animal sciences and welfare (LASW) 20 and archived.
The human primary colon carcinoma CR-LRB-009C tumor model was established by implanting (SC) small tumor fragments and was maintained in SCID female mice using serial passages.
Compound (1) formulation was prepared by incorporating the MEK inhibitor into 0.5% CMC 25 0.25% Tween 20. The préparation was stored at 4°C and resuspended by vortexing before use. The oral form of the compound was prepared every 3 days. The volume of administration per mouse was 10 mL/kg.
Compound (2a) formulation was prepared in water for injection. The stock solution was chemically stable 7 days in the dark at 4°C. The volume of administration per mouse was 10 30 mL/kg.
Compound (2a) and Compound (2b) formulations were prepared in 1N HCl and water for injection, final pH was 3, followed by five cycles of vortexing and sonicating. The stock solution was chemically stable 7 days in the dark at 4°C. The volume of PO administration per mouse was 10 ml_/kg.
-3616446
For subcutaneous implantation of tumor cells, skin in the flank of the mice was disinfected using alcohol or Betadine® solution (Alcyon) and a suspension of tumor cells was inoculated
SC unilaterally under a volume of 0.2 mL using a 23 G needle.
The dosages and schedule of administration of Compound (1), Compound (2a) and Compound (2b) used as single agent or in combination are described in the results section and detailed in Tables 15-17.
The animais required to begin a given experiment were pooled and implanted monolaterally on day 0. Treatments were administered on measurable tumors. The solid tumors were allowed to grow to the desired volume range (animais with tumors not in the desired range were excluded). The mice were then pooled and unselectively distributed to the various treatment and control groups. Treatment started 11 days post CR-LRB-009C tumor fragment implantation as indicated in the results section and in each table. The dosages are expressed in mg/kg, based on the body weight at start of therapy. Mice were checked daily, and adverse clinical reactions noted. Each group of mice was weighed as a whole daily until the weight nadir was reached. Then, groups were weighed once to thrice weekly until the end of the experiment. Tumors were measured with a caliper 2 to 3 limes weekly until final sacrifice for sampling time, tumor reached 2000 mm3 or until the animal died (whichever cornes first). Solid tumor volumes were estimated from two-dimensional tumor measurements and calculated according to the foilowing équation:
Tumor weight (mg) = Length (mm) x Width2 (mm2)/2
The day of death was recorded. Surviving animais were sacrificed and macroscopie examination of the thoracic and abdominal cavities was performed.
A dosage producing a 15% body weight loss (BWL) during three consecutive days (mean of group), 20% BWL during 1 day or 10% or more drug deaths was considered an excessively toxic dosage. Animal body weights included the tumor weight.
The primary efficacy end points are AT/AC, percent médian régression, partial and complété régressions (PR and CR). Statistical analyses were performed on SAS system release 8.2 for SUN4 via Everstat V5 software and SAS 9.2 software. A probability less than 5% (p<0.05) was considered as significant.
Results of in vivo studies
The médian tumor burden at start of therapy was 126 to 144 mm3. As single agents, Compound (1) (20 mg/kg/administration (Adm)), Compound (2b) (20 mg/kg/adm) and Compound (2a) (75 mg/kg/adm) were administered PO daily from days 11 to 21 post tumor
implantation. In the combination groups, the dose of Compound (1 ) was combined with each dose of Compound (2a) and Compound (2b), as shown in Table 15.
As single agents or used in combination, Compound (1), Compound (2b) and Compound (2a) were tolerated, inducing some BWL but not reaching toxicity (Figure 21 and Table 15). As single agents, Compound (1) and Compound (2b) achieved a ΔΤ/ΔΟ>40 %, while Compound (2a) achieved a ΔΤ/AC of 39 % under these test conditions.
In the combination, the treatment with Compound (1 ) at 20 mg/kg/adm and Compound (2b) at 20 mg/kg/adm achieved a ΔΤ/ΔΟ of 4 % (Figure 22 and Table 15), and as shown by Table 16, therapeutic synergy was reached (p < 0,0001 for global analysis). The treatment with Compound (1) at 20 mg/kg/adm and Compound (2a) at 75 mg/kg/adm achieved a ΔΤ/AC of 21 % (Figure 22 and Table 15), and as shown by Table 16, therapeutic synergy was achieved (p=0.0386 globally). See a/so Table 17.
Summarv of in vivo results
The in vivo work presented here reports the in vivo antitumor activity of combining Compound (1), an oral potent and sélective allosteric inhibitor of MEK1/2, with oral, potent, and spécifie inhibitors of class I PI3K lipid kinases Compound (2a), a pan-PI3K inhibitor, and Compound (2b), a dual pan-PI3K and mTOR inhibitor. This work has been performed against human primary colon carcinoma CR-LRB-009C xenografts harboring a dual KRAS and PIKC3A mutation known to reduce the sensitivity to MEK inhibition.
In the study, combination treatment induced a sustained tumor stasis during the treatment phase and reached therapeutic synergy.
Accordingly, a potent antitumor activity with therapeutic synergy has been achieved in a PIKC3A- and KRAS-mutant CR-LRB-009C driven xenograft model when combining the inhibitor of MEK1/2 Compound (1) with Compound (2a), a pan-PI3K inhibitor or Compound (2b), a dual pan-PI3K and mTOR inhibitor.
Example 6. In vivo activity of Compound (1) in combination with Compound (2a) or Compound (2b) against subeutaneous human colon tumors CR-LRB-013P bearinq SCIP female mice
To evaluate the antitumor activity of the MEK inhibitor Compound (1) in combination with the pan-PI3K inhibitor Compound (2a) or the dual pan-PI3K / mTOR inhibitor Compound (2b), experiments were conducted using female SCID mice bearing human primary colon tumors
CR-LRB-013P (KRAS mutant) xenografts. In this study, Compound (1) at 20 mg/kg was tested in combination with Compound (2b) at 20 mg/kg or Compound (2a) at 75 mg/kg.
-3816446
Materials and methods
CB17/ICR-Prkdc severe combined immunodeficiency (SCID) /Cri mice, at 8-10 weeks old, were bred at Charles River France (Domaine des Oncins, 69210 L’Arbresie, France) from strains obtained from Charles River, USA. Mice were over 18 g at start of treatment after an acclimatization time of at least 5 days. The mice had free access to food (UAR reference 113, Villemoisson, 91160 Epinay sur Orge, France) and stérile water. The mice were housed on a 12 hours light/dark cycle. Environmental conditions including animal maintenance, room température (22°C ± 2°C), relative humidity (55% ± 15%) and lighting times were recorded by the supervisor of laboratory animal sciences and welfare (LASW) and archived.
The human primary colon carcinoma CR-LRB-013P tumor model was established by implanting (SC) small tumor fragments and was maintained in SCID female mice using serial passages.
Compound (1) formulation was prepared by incorporating the MEK inhibitor into 0.5% CMC 0.25% Tween 20. The préparation was stored at 4°C and resuspended by vortexing before use. The oral form of the compound was prepared every 3 days. The volume of administration per mouse was 10 mL/kg.
Compound (2a) formulation was prepared in water for injection. The stock solution was chemically stable 7 days in the dark at 4°C. The volume of administration per mouse was 10 mL/kg.
Compound (2a) and Compound (2b) formulations were prepared in 1N HCl and water for injection, final pH was 3, followed by five cycles of vortexing and sonicating, The stock solution was chemically stable 7 days in the dark at 4°C. The volume of PO administration per mouse was 10 mL/kg.
For subcutaneous implantation of tumor cells, skin in the flank of the mice was disinfected using alcohol or Betadine® solution (Alcyon) and a suspension of tumor cells was inoculated SC unilaterally under a volume of 0.2 mL using a 23 G needle.
The dosages and schedule of administration of Compound (1 ), Compound (2a) and Compound (2b) used as single agent or in combination are described in the results section and detailed in the tables that follow.
The animais required to begin a given experiment were pooled and implanted monolaterally on day 0. Treatments were administered on measurable tumors. The solid tumors were allowed to grow to the desired volume range (animais with tumors not in the desired range
-3916446 were excluded). The mice were then pooled and unselectively distributed to the various treatment and control groups. Treatment started 33 days post CR-LRB-013P tumor fragment implantation as indicated in the results section and in each table. The dosages are expressed in mg/kg, based on the body weight at start of therapy. Mice were checked daily, and adverse clinical reactions noted. Each group of mice was weighed as a whole daily until the weight nadir was reached. Then, groups were weighed once to thrice weekly until the end of the experiment. Tumors were measured with a calliper 2 to 3 times weekly until final sacrifice for sampling time, tumor reached 2000 mm3 or until the animal died (whichever cornes first). Solid tumor volumes were estimated from two-dimensional tumor measurements and calculated according to the following équation:
Tumor weight (mg) = Length (mm) x Width2 (mm2)/2
The day of death was recorded. Surviving animais were sacrificed and macroscopie examination of the thoracic and abdominal cavîties was performed.
A dosage producing a 15% body weight loss (BWL) during three consecutive days (mean of group), 20% BWL during 1 day or 10% or more drug deaths was considered an excessively toxic dosage. Animal body weights included the tumor weight.
The primary efficacy end points are ΔΤ/AC, percent médian régression, partial and complété régressions (PR and CR). Statistical analyses were performed on SAS System release 8.2 for SUN4 via Everstat V5 software and SAS 9.2 software. A probability less than 5% (p<0.05) was considered as signifîcant.
Results of in vivo studies
The médian tumor burden at start of therapy was 144 to 162 mm3. As single agents, Compound (1 ) (20 mg/kg/administration (Adm)), Compound (2b) (20 mg/kg/adm) and Compound (2a) (75 mg/kg/adm) were administered PO daily from days 33 to 50 post tumor implantation. In the combination groups, the dose of Compound (1) was combined with each dose of Compound (2a) and Compound (2b), as shown in Table 18.
As single agents or used in combination, Compound (1), Compound (2b) and Compound (2a) were tolerated, inducing some BWL but not reaching toxicity (Figure 23 and Table 18). As single agents under these test conditions, Compound (2a) and Compound (2b) achieved a ΔΤ/Δΰ>40 %, while Compound (1 ) achieved a ΔΤ/AC of 30 %.
In combination, treatment with Compound (1) at 20 mg/kg/adm and Compound (2b) at 20 mg/kg/adm achieved a ΔΤ/AC of 26 % (Figure 24 and Table 18) with 1/7 partial régression, and as shown by Table 19, therapeutic synergy was reached (p = 0.0302 for global
-4016446 analysis). The treatment with Compound (1) at 20 mg/kg/adm and Compound (2a) at 75 mg/kg/adm achieved a AT/AC of -5 % (Figure 24 and Table 18) with 5/7 partial régression, and as shown by Table 19, therapeutic synergy was achieved (p<0.0001 globally). See also Table 20.
Summary of in vivo results
The in vivo work presented here reports the in vivo antitumor activity of combining Compound (1 ), an oral potent and sélective allosteric inhibitor of MEK1/2, with oral, potent, and spécifie inhibitors of class I PI3K lipid kinases Compound (2a), a pan-PI3K inhibitor, and Compound (2b), a dual pan-PI3K and mTOR inhibitor. This work has been performed against human primary colon carcinoma CR-LRB-013P xenografts harboring a KRAS mutation.
In the study, combination treatment induced a sustained tumor stasis or partial régressions during the treatment phase and reached therapeutic synergy.
Accordingly, a potent antitumor activity with therapeutic synergy has been achieved in KRAS mutant CR-LRB-013P driven xenograft model when combining the inhibitor of MEK1/2 Compound (1 ) with Compound (2a), a pan-PI3K inhibitor or Compound (2b), a dual pan-PI3K and mTOR inhibitor.
Example 7. Evaluation of tumor permeability
The following experiment was conducted to evaluate the impact of Compound (2a) and Compound (2b), alone or in combination with Compound (1 ), on tumor vascular permeability.
Methods
HCT116 tumor cells were implanted subcutaneously in the intra-scapular région in SCID mice. Implanted animais received Compound (2a) 50mg/kg or Compound (2b) 20mg/kg from day 11 to day 13, as single agents or combined with Compound (1) 10mg/kg (five animais per group). Each agent was given by oral route on a daily schedule. Tumor growth was monitored throughout the experiment by callipering the tumors. To quantify tumor vascular permeability, tumors were excised under ketamine/Xylazine (120/6 mg/kg ip) anesthésia at day 13, 4 hours post last treatment, 30 min after 0.5% Evans Blue iv injection, and 2 min post Dextran-Fitc 100mg/kg iv injection. Tumors were then snap frozen, and 25pm sections obtained for fluorescence quantification. Tumors sections were imaged with leyte at 488 nm for vascular Dextran-Fitc détermination and at 633 nm for Evans-Blue extravasation détermination. Respective fluorescence were quantified as the sum of intégral
-41 16446 phantoms of fluorescence intensity and expressed as the mean ratio of Evans-Blue signal /
Dextran-Fitc Signal.
Results
Under these test conditions in advanced subcutaneously grafted HCT116 human
KRAS/PI3KCA mutated colon carcinoma, Compound (1 ) and Compound (2a) used as single agents and the combination of Compound (2a)/Compound (1) did not significantly modify tumor permeability, showing -9%, -8% and 4% decrease, respectively, of the EvansBlue/Dextran-Fitc ratio compared to control. On the other hand, 3 days of treatment with Compound (2b) or the combination of Compound (2b)/Compound (1) induced clear modulation of Evans-Blue/Dextran Fitc ratio, producing a 50% decrease for the single agent and 45% decrease for the combination. See Figure 25.
Summarv
Compound (2b) used as a single agent or in combination with Compound (1) alters tumor vascular permeability after 3 days of treatment in advanced subcutaneously grafted HCT116 15 human KRAS/PI3KCA mutated colon carcinoma. This alteration in HCT116 tumor vascular permeability disrupts in vivo fluorescent-Annexin tumor distribution for FMT imaging and precludes apoptosis détection by this method.
-4216446
Table 1. Results of Compound (1) and Compound (2b) in vitro combination separated into 5 different groups
Cell line Vector sum Average HSA Cumulative z-score MSC6369 z-score MSC0765
Group I. Coll lines résistant to both MSC6369 and MSC0765 (z-score>1.0)
A673 -3.63 -0.18 -0.18 3.00 0.96
PANC1 -0.89 -0.11 -0.15 3.00 1.16
Group IL Cell lines with relative résistance to both agents (1.0<z-score<0)
BT20 -1.10 -0.14 -0.25 0.02 0.32
DLD1 -0.89 -0.12 -0.23 0.19 0.81
DU145 -0.81 -0.11 -0.17 0.73 -0.05
Group III. Cell lines very résistant to one of the agents (one z-score>0)
CASKI -0.89 -0.12 -0.19 1.55 -0.23
E.T28 -1.00 -0.13 -0.22 -0.68 1.25
HCT116 -1.19 -0.13 -0.19 -0.32 1.90
Group IV. CeL lines relativcly active for both agents
A549 -0.96 -0.11 -0.17 -0.11 -0.20
MCF7 -0.87 -0.12 -0.22 -0.33 0.30
Group V. Cell ines very sensitive to one of the agents
SKBR3 -0.85 -0.07 -0.08 0.69 -2.18
-4316446
Table 2. Antitumor activity of Compound (1) (5 mg/kg) in combination with Compound (2b) (30 mg/kg) or Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice
Agent (batch Route/Dosag e in mL/kg per administratio n Dosage in mg/kg per administrati on (total dose) Sched ule in days Dru g deat h (Da y of deat h Average body weight change in % per mouse at nadir (day of nadir Media n ΔΤ/ ΔΟ in % day 18 Régressions
Par tial Comp lete
Compoun d(1) (VAC.HA L1.166) PO 10 mL/kg 5(40) 11-18 0/7 -3.4(14) 70 0/7 0/7
Compoun d (2b) (T100738 8) PO 10 mL/kg 30(240) 11-18 0/7 -6.7(18) 77 1/7 0/7
Compoun d (2a) (2009015 0) PO 10 ml_/kg 75 (600) 11-18 0/7 -8.3(18) 80 0/7 0/7
50 (400) 0/7 -5.8(18) 62 0/7 0/7
-4416446
Agent (batch Route/Dosag e in mL/kg per admînistratio n Dosage in mg/kg per administrât! on (total dose) Sched ule în days Dru g deat h (Da y of deat h Average body weight change in % per mouse at nadir (day of nadir Media n ΔΤ/ AC in % day 18 Régressions
Par tial Comp lete
Compoun d(1) Compoun d (2b) PO 10 mL/kg 5(40) 30(240) 11-18 0/7 -7.4(15) 27 0/7 0/7
Compoun d(1) Compoun d (2a) PO 10 mL/kg 5 (40) 75 (600) 11-18 0/7 -7.4(18) 21 0/7 0/7
5(40) 50(400) 11-18 0/7 -7.4(16) 22 0/7 0/7
Control 0/7 -0.8(18) 100 0/7 0/7
Tumor size at start of therapy was 162-352 mm3, with a médian tumor burden per group of 198-221 mm3. Drug formulation: Compound (1 ) = carboxymethylcellulose 0.5%, tween 20 0.25% in water; Compound (2b) = water, pH3, Compound (2a) = water. Treatment duration: Compound (1), Compound (2b), Compound (2a) and combination = 8 days. Abbreviations used: BWL = body weight loss, DT/nC=Ratio of change in tumor volume from baseline médian between treated and control groups (TVday - TVO) / (CVday - CVO) * 100, HNTD = highest non toxic dose, HDT = highest dose tested.
-4516446
Table 3. Antitumor activity of Compound (1) (5 mg/kg) in combination with Compound (2b) (30 mg/kg) or Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice: Therapeutic synergy détermination
Group comparison Day Estimated Différence Between Groups Means T-test value Pa
Compound (1)5 mg/kg and Compound (2b) 30 mg/kg versus Compound (2b) at 30 mg/kg Global -72.6786 -1.91 0.0606
D11 1.7143 0.05 0.9572
D14 -91.4286 -1.31 0.2035
D16 -79.2857 -1.03 0.3134
D18 -121.71 -1.14 0.2653
Compound (1) at 5 mg/kg and Compound (2a) at 50 mg/kg versus Compound (2a) at 50 mg/kg Global -93.5357 -2.66 0.0091
D11 -4.5714 -0.14 0.8866
D14 -62.4286 -0.97 0.3394
D16 -128.29 -1.77 0.0853
D18 -178.86 -1.84 0.0735
Compound (1 ) at 5 mg/kg and Compound (2a) at 75 mg/kg versus Global -156.68 -4.45 <0001
D11 -4.1429 -0.13 0.8972
D14 -32.2857 -0.50 0.6196
-4616446
Group comparison Day Estimated Différence Between Groups Means T-test value Pa
Compound (2a) at 75 mg/kg D16 -249.14 -3.44 0.0015
D18 -341.14 -3.52 0.0012
a Each combination was compared to the best single agent using estimâtes obtained from a 2-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume (proc mixed of SAS 9.2 software). A probability less than 5% (p<0.05) was considered as significant
-4716446
Table 4. Antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) or Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice
Agent (batch) Route/Dosa ge in mL/kg per administrât! on Dosage in mg/kg per administrât ion (total dose) Sche dule in days Dr □9 de ath (Da y of de ath ) Average body weight change in % per mouse at nadir (day of nadir) Med ian ΔΤ/ AC in % day 18 Médian %of regressi on on day 18 Régressions
P ar tia I Comple te
Compou nd(1) (VAC.H AL1.166 ) PO 10 mL/kg 20(160) 11-18 0/7 -1.1 (15) 22 - 0/ 7 0/7
10 (90) a 0/7 -2.2(18) 20 - 0/ 7 0/7
Compou nd (2b) (T10073 88) PO 10 mL/kg 20 (160) 0/7 -3.7(15) 71 - 0/ 7 0/7
Compou nd(2a) PO 10 mL/kg 75 (600) 11-18 0/7 -6.7(18) 56 - 0/ 7 0/7
-4816446
Agent (batch) (200901 50) Route/Dosa ge in mL/kg per administrât! on Dosage in mg/kg per administrât ion (total dose) Sche dule in days Dr ug de ath (Da y of de ath ) Average body weight change in % per mouse at nadir (day of nadir) Med ian ΔΤ/ AC in % day 18 Médian %of regressi on on day 18 Régressions
P ar tia I Comple te
50(400) 0/7 -7.7(18) 52 - 0/ 7 0/7
Compou nd (1) Compou nd(2b) PO 10 mL/kg 20(160) 20(160) 0/7 -4.3(14) 0 - 0/ 7 0/7
Compou nd(1) Compou nd (2a) PO 10 mL/kg 20(160) 50 (400) 0/7 -5.3(18) -2 8 1/ 7 0/7
-4916446
Agent (batch) Route/Dosa ge in mL/kg per administrât! on Dosage in mg/kg per administrât ion (total dose) Sche dule in days Dr ug de ath (Da y of de ath ) Average body weight change in % per mouse at nadir (day of nadir) Med ian AT/ AC in % day 18 Médian %of regressi on on day 18 Régressions
P ar tia I Comple te
Compou nd(1) Compou nd (2b) PO 10 mL/kg 10(80) 20(160) 0/7 -6.3(18) 0 - 2/ 7 0/7
Compou nd (1) Compou nd (2a) PO 10 mL/kg 10(80) 75 (600) 11-18 0/7 -6.3(18) 5 - 1/ 7 0/7
10(80) 50(400) 0/7 -8.0(18) -4 8 1/ 7 0/7
Control 0/7 -2.4(13) 100
Tumor size at start of therapy was 126 - 294 mm3, with a médian tumor burden per group of 180-198 mm3. Drug formulation: Compound (1 ) = carboxymethylcellulose 0.5%, Tween 20 0.25% in water; Compound (2b) = water, pH3; Compound (2a) = water. Treatment duration: Compound (1), Compound (2b), Compound (2a) and combination = 8 days. Abbreviations used: BWL = body weight loss, OT/DC= Ratio of
-5016446 change in tumor volume from baseline médian between treated and control groups (TVday - TVO) / (CVday - CVO) * 100, HNTD = highest non toxic dose, HDT = highest dose tested.
a On day 17, mice received 20 mg/kg instead of 10 mg/kg.
-51 16446
Table 5. Antitumor activity of Compound (1)(10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) or Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice: therapeutic synergy détermination
Group comparison Day Estimated Différence Between Groups Means T-test value Pa
Compound (1) at 10 mg/kg and Compound (2b) at 20 mg/kg versus Compound (1)at 10 mg/kg Global -107.10 -3.71 0.0004
D11 0.5714 0.02 0.9828
D14 -160.14 -2.91 0.0061
D18 -161.71 -2.62 0.0128
Compound (1 ) at 20 mg/kg and Compound (2b) at 20 mg/kg versus Compound (1 ) at 20 mg/kg Global -35.9524 -1.24 0.2169
D11 8.2857 0.32 0.7545
D14 -50.4286 -0.92 0.3648
D18 -65.7143 -1.07 0.2940
Compound (1)at 10 mg/kg and Compound (2a) at 50 mg/kg versus Compound (1)at 10 mg/kg Global -106.10 -3.21 0.0019
D11 4.1429 0.15 0.8784
D14 -139.29 -2.90 0.0056
D18 -183.14 -2.22 0.0315
Compound (1)at 10 mg/kg and Compound (2a) at 75 mg/kg Global -92.5238 -2.80 0.0063
D11 -5.1429 -0.19 0.8494
-5216446
Group comparison Day Estimated Différence Between Groups Means T-test value Pa
versus Compound (1) at 10 mg/kg D14 -158.86 -3.31 0.0018
D18 -113.57 -1.37 0.1758
a Each combination was compared to the best single agent using estimâtes obtained from a two-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume (proc mixed of SAS 9.2 software). A probability less than 5% (p<0.05) was considered as significant.
-5316446
Table 6. Antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against human HCT 116 bearing SCID female mice
Agent (batch) Rout e/ Dosa ge in mL/k 9 ΡθΓ injec tion Dosa ge in mg/k g per injecti on (total dose) Sched ule in days Drug death (Day of death ) Average body weight change in % per mouse at nadir (day of nadir) Med ian ΔΤ/ AC in % day 20 Médian % of regres sion on day 20 Régressions
Partial Complété
Compou nd(1) (VAC.H AL1.166 ) PO W mL/k 9 20 (200) 11-20 0/10 -3.6(19) 34 - 0/10 0/10
(27) 10 (100) 0/10 -4.9 (20) 43 - 0/10 0/10
Compou nd (2a) (200901 50) PO 10 mL/k g 75 (750) 11-20 0/10 -8.5 (20) 64 - 0/10 0/10
-5416446
50 (500) 0/10 -7.8(19) 66 - 0/10 0/10
Compou nd(1) Compou nd (2a) PO 10 mL/k g 20 (380) 75 (1425) 11-29b 0/10 -7.8(17) 9 - 0/10 0/10
20 (200) 50 (500) 11-20 0/10 -5.6 (20) 22 - 0/10 0/10
Compou nd(1) Compou nd (2a) PO 10 mL/k g 10 (100) 75 (750) 11-20 0/10 -7.5 (20) 18 - 0/10 0/10
10 (100) 50 (500) 11-20 0/10 -7.3 (20) 19 - 0/10 0/10
Control - 0/10 -1.4 (20) -
Vehicle 11-20 0/10 -3.1 (20) -
Tumor size at start of therapy was 112-319 mm3, with a médian tumor burden per group of 187-189 mm3. Drug formulation: Compound (1) =
-5516446 carboxymethylcellulose 0.5%, Tween 20 0.25% in water; Compound (2a) = water. Treatment duration: Compound (1), Compound (2a) and combination = 10 days. Abbreviations used: bwl = body weight loss, ΔΤ/Δ0- (TVday - TVO) / (CVday - CVO) * 100, HNTD = highest non toxic dose, HDT = highest dose tested.
-5616446
Table 7. Antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2a) (50 and 75 mg/kg) against human HCT116 bearing SCID female mice: therapeutic synergy détermination
Group comparison Day Estimated Différence Between Groups Means I T-test value Pa
Combination of Compound (1) at 20 mg/kg and Compound (2a) at 75 mg/kg versus Compound (1) at 20 mg/kg alone Global -186.00 | -3.80 0.0003
D14 -117.00 I -2.60 0.0110
D18 -183.70 -3.09 0.0027
D20 -257.30 -3.02 0.0034
Combination of Compound (1) at 20 mg/kg and Compound (2a) at 50 mg/kg versus Compound (1 ) at 20 mg/kg alone Global -87.7667 -1.79 0.0764
D14 -95.3000 -2.12 0.0372
D18 -73.1000 I -1.23 0.2219
D20 -94.9000 -1.11 0.2692
Combination of Compound (1) at 10 mg/kg and Compound (2a) at 75 mg/kg versus Global -127.30 -2.60 0.0109
D14 -68.9000 -1.53 0.1297
-5716446
Group comparison Day Estimated Différence Between Groups Means T-test value Pa
Compound (1) at 10 mg/kg alone D18 -99.6000 -1.68 0.0974
D20 -213.40 -2.50 0.0143
Combination of Compound (1) at 10 mg/kg and Compound (2a) at 50 mg/kg versus Compound (1)at 10 mg/kg Global -131.30 -2.68 0.0088
D14 -104.60 -2.32 0.0226
D18 -140.30 -2.36 0.0206
D20 -149.00 -1.75 0.0844
a Each combination was compared to the best single agent using estimâtes obtained from a 2-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume (proc mixed of SAS 9.2 software). A probabîlity less than 5% (p<0.05) was considered as significant.
-5816446
Table 8. Antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) against human HCT 116 bearing SCID female mice
Agent (batch) Route / Dosa ge in mL/kg per injecti on Dosa ge in mg/k 9 ΡθΓ inject ion (total dose) Sched ule in days Dru g deat h (Day of deat h) Average body weight change in % per mouse at nadir (day of nadir) Media n ΔΤ/Δ C in % day 20 Médian %of regressi on on day 20 Régressions
Parti al Compl ete
Compoun d(1) (VAC.HA L1.166) PO 10 mL/kg 20 (200) 11-20 0/10 -4,1 (18) 41 - 0/10 0/10
(27) 10 (100) 0/10 -2.3(13) 53 - 0/10 0/10
Compoun d (2b) (T100738 8) PO 10 mL/kg 20 (200) 11-20 0/10 -4,8 (20) 83 - 0/10 0/10
-5916446
Compoun d(1) Compoun d (2b) PO 10 mL/kg 20 (200) 20 (200) 11-20 0/10 -3,3(13) 15 - 0/10 0/10
10 (100) 20 (200) 0/10 -3,7(13) 30 - 0/10 0/10
Control 0/10 -4,3(20) -
Vehicle 11-20 0/10 -2,1 (16) -
Tumor size at start of therapy was144-294 mm3, with a médian tumor burden per group of 189-196 mm3. Drug formulation: Compound (1) = carboxymethylcellulose 0.5%, Tween 20 0.25% in water ; Compound (2b) = water, pH3. Treatment duration: Compound (1), Compound (2b) and combination = 10 days . Abbreviations used: bwl - body weight loss, AT/AC= (TVday - TVO) / (CVday - CVO) * 100, HNTD = highest non toxic dose, HDT = highest dose tested
-6016446
Table 9. Antitumor activity of Compound (1) (10 and 20 mg/kg) in combination with Compound (2b) (20 mg/kg) against human HCT 116 bearing SCID female mice: therapeutic synergy détermination
Group comparison Day Estimated Différence Between Groups Means T-test value Pa
Combination of Compound (1) at 20 mg/kg and Compound (2b) at 20 mg/kg versus Compound (1) at 20 mg/kg alone Global -180.40 -3.53 0.0008
D13 -95.2000 -2.25 0.0281
D15 -168.90 -3.09 0.0032
D18 -194.70 -2.45 0.0172
D20 -262.80 -2.78 0.0072
Combination of Compound (1) at 10 mg/kg and Compound (2b) at 20 mg/kg versus Compound (1) at 10 mg/kg alone Global -202.72 -3.97 0.0002
D13 -51.3000 -1.22 0.2295
D15 -212.90 -3.89 0.0003
D18 -272.10 -3.43 0.0011
D20 -274.60 -2.91 0.0051
-61 16446 a Each combination was compared to the best single agent using estimâtes obtained from a 2-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume (proc mixed of SAS 9.2 software). A probability less than 5% (p<0.05) was considered as significant.
-6216446
Table 10. Percent ΔΤ/ΔΟ and statistical analysis in MiaPaCa-2 tumor-bearing mice treated with Compound (1), Compound (2a), and
Compound (2b) alone or in combination.
% ΔΤ/ΔΟ Vehicle AS703026 XL-147 XL-765 AS703026+ XL-147 AS703026 + XL-765
po QD 5mg/kg po QD 50mg/kg poQD 30mg/kg po QD po QD po QD
Vehicle po QD 100.0 <0.05 <0.05 <0.05 <0.05 <0.05
AS703026 5mg/kg po QD 56.2 <0.05 NS NS NS <0.05
XL-147 50mg/kg po QD 71.2 <0.05 NS NS NS <0.05
XL-765 30mg/kg po QD 77.0 <0.05 NS NS NS <0.05
AS703026+ XL-147 48.7 <0.05 NS NS NS NS
AS703026 + XL-765 27.3 <0.05 <0.05 <0.05 <0.05 NS
The mean percent of actual Miapaca-2 tumor growth inhibited by the treatments was calculated as follows: [%AT/AC= (TVf- Τν/Τν^- TVictri) x 100%], where TV=tumor volume, f= final, i = initial and Ctrl = control group.
-6316446
Table 11 A.
AT/AC (%) on d18
Compound (1) 5mpk 70
Compound (2b) 30mpk 77
Compound (2b) 30mpk Compound (1) 5mpk 27
-6416446
Table 11 B.
AT/AC (%) on d18
Compound (1) 5mpk 70
Compound (2a) 75mpk 80
Compound (2a) 50mpk 62
Compound (2a) 75mpk Compound (1)5mpk 21
Compound (2a) 50mpk Compound (1) 5mpk 22
-6516446
Table 12A.
ΔΤ/ΔΟ (%) on d18
Compound (1)20mpk 22
Compound (1) 10mpk 20
Compound (2b) 20mpk 71
Compound (2b) 20mpk Compound (1)20mpk 0
Compound (2b) 20mpk Compound (1) 10mpk 0
-6616446
Table 12B.
ΔΤ/AC (%) on d18
Compound (1) 10mpk 20
Compound (2a) 75mpk 56
Compound (2a) 50mpk 52
Compound (2a) 75mpk Compound (1) 10mpk 5
Compound (2a) 50mpk Compound (1) 10mpk -4
-6716446
Table 13.
ΔΤ/AC (%) on d20
Compound (1)20mpk 34
Compound (1) 10mpk 43
Compound (2a) 75mpk 64
Compound (2a) 50mpk 66
Compound (2a) 75mpk Compound (1) 20mpk 9
Compound (2a) 75mpk Compound (1) 10mpk 18
Compound (2a) 50mpk Compound (1) 20mpk 22
Compound 19
-6816446
(2a) 50mpk Compound (1) 10mpk
Table 14.
AT/AC (%) on d20
Compound (1) 20mpk 41
Compound (1) 10mpk 53
Compound (2b) 20mpk 83
Compound (2b) 20mpk Compound d) 20mpk 15
Compound (2b) 20mpk Compound (1)10mpk 30
-6916446
Table 15. Antitumor activity of Compound (1 ) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) or Compound (2a) (75 mg/kg) against human primary colon CR-LRB-009C tumors bearing SCID female mice
Avera
Régressions
Agent (batch)
Dr ug de
Route/Dosa Dosage in at
ge in ml_/kg mg/kg per Sched h
per administrati ule in (D
administrati on (total days ay
on dose) of
de at
h) ge body weigh t chang e in % per mous e at nadir (day of
Medi
an ΔΤ/
AC Pa Com
in % day 21 rtia I plete
nadir)
Compound (1) PO (VAC.HAL1.166) 10mL/kg
20(220)
-7.7
11-21 0/7 (20)
0/7 0/7
Compound (2b) PO (T1007388) 10mL/kg (220)
-7.4 11-21 0/7 (19)
0/7 0/7
-7016446
Route/Dosa Dosage in
Agent (batch) ge in mL/kg mg/kg per
per administrati
administrati on (total
on dose)
Avera Régressions
Dr ge
ug body weigh Medi
de
t
at chang an AT/
Sched h
ule in (D e in % AC Pa Com
days ay per mous in % rtia plete I
of e at day
de at h) nadir (day 21
of
nadir)
Compound (2a) (T1007032 M022906)
PO mL/kg
75(825)
Compound (1) PO
20(220)
Compound (2b) 10mL7kg
20(220)
Compound (1) PO (220)
Compound (2a) mL/kg
75(825)
11-21 0/7 15.8(2 39 0/70/7
1)
-13.7
11-21 0/7 4 1/70/7 (21)
-14.0
11-21 0/7 21 0/70/7 (21)
Control
-7.8
0/7 100 (20)
Tumor size at start of therapy was 100-221 mm3, with a médian tumor burden per group of 126-144 mm3. Drug formulation: Compound (1) = carboxymethylcellulose 0.5%, tween 20 0.25% in water; Compound (2b) and Compound (2a) = water, pH3. Treatment duration: Compound (1), Compound (2a) and Compound (2b) and combination = 11 days. Abbreviations used: BWL = body weight loss, AT/AC=Ratio of change in tumor volume from baseline médian between treated and control groups (TVday - TV0) / (CVday - CVO) * 100, HDT = highest dose tested.
-7216446
Table 16. Antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) or Compound (2a) (75 mg/kg) against human primary colon CR-LRB-009C tumors bearing SCID female mice: Therapeutic synergy détermination
Day Estimated Différence between Groups Means T-test value Pa
Combination of Compound (1) at 20 mg/kg and Compound (2a) at 75 mg/kg versus Compound (2a) at 75 mg/kg Global -21.3214 -2.13 | 0.0386
D13 3.5714 0.27 0.7891
D15 -22.7143 -1.71 0.0912
D18 -34.5714 -2.60 0.0109
D21 -31.5714 -2.37 | 0.0197
Combination of Compound (1) at 20 mg/kg and Compound (2b) at 20 mg/kg versus Compound (2b) at 20 mg/kg Global -56.3016 -5.61 | <.0001
D13 -5.1429 | -0.39 | 0.7001
D15 -57.2857 -4.30 | <.0001
D18 I -82.2143 -6.18 | <.0001
.. D21 -80.5635 -5.89 <.0001
-7316446 a Each combination was compared to the best single agent using estimâtes obtained from a 2-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume (proc mixed of SAS 9.2 software). A probability less than 5% (p<0.05) was considered as significant.
Table 17.
ΔΤ/AC (%) on d21
Compound (1) 20mg/kg 53
Compound (2a) 75mg/kg 39
Compound (2b) 20mg/kg 51
Compound (2a) 75mg/kg Compound (1) 20mg/kg 21
Compound (2b) 20mg/kg Compound (1) 20mg/kg 4
-7416446
Table 18. Antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) or Compound (2a) (75 mg/kg) against human primary colon CR-LRB-013P tumors bearing SCID female mice
Régressions
Dru
Med ian
Average body
Agent (batch) Route/Dosage in mL/kg per administration Dosage in mg/kg per administration (total dose) Schedul e in days dea th (Da y of dea th) change in % per mouse at nadir (day of nadir) ΔΤ/ ΔΟ in % day 50 P
ar tia I Corn p lete
Compound (1) (VAC.HAL1.166) PO 10 mL/kg 20 (360) 33-50 0/7 -4.5 (50) 30 0/ 7 0/7
Compound (2b) (T1007388) PO 10 mL/kg 20(360) 33-50 0/7 -5.2 (50) 83 0/ 7 0/7
Compound (2a) (20090150) PO 10 mL/kg 75(1350) 33-50 0/7 -9.2 (50) 53 0/ 7 0/7
Compound (1) Compound (2b) PO 10 mL/kg 20 (360) 20 (360) 33-50 0/7 -3.7 (43) 26 1/ 7 0/7
Compound (1) Compound (2a) PO 10 mL/kg 20 (360) 75(1350) 33-50 0/7 -10.2 (38) -5 5/ 7 0/7
-7516446
Dru
Agent (batch)
Route/Dosage in mL/kg per administration
Dosage in mg/kg per administration (total dose) g
dea Schedul th e in (Da days y of dea th)
Average body weight change in % per mouse at nadir (day of nadir)
Régressions
Med ian
ΔΤ/
AC in % day ar tia
Comp lete
Control
0/7 -3.5(50) 100
Tumor size at start of therapy was 108-245 mm3, with a médian tumor burden per group of 144-162 mm3. Drug formulation: Compound (1) = carboxymethylcellulose 0.5%, tween 20 0.25% in water; Compound (2b) and Compound (2a) = water, pH3. Treatment duration: Compound (1), Compound (2a) and Compound (2b) and combination = 18 days. Abbreviations used: BWL = body weight loss, AT/AC=Ratio of change in tumor volume from baseline médian between treated and control groups (TVday - TV0) / (CVday - CVO) * 100, HDT = highest dose tested.
-7616446
Table 19. Antitumor activity of Compound (1) (20 mg/kg) in combination with Compound (2b) (20 mg/kg) or Compound (2a) (75 mg/kg) against human primary colon CR-LRB-013P tumors bearing SCID female mice: Therapeutîc synergy détermination
-7716446
Day Estimated Différence between Groups Means t Value Pa
Combination of Compound (1) at 20 mg/kg and Compound (2a) at 75 mg/kg versus Compound (1) at 20 mg/kg Global -149.52 -4.88 <.0001
D35 -10.1429 -0.30 0.7639
D37 -67.5714 -2.18 | 0.0345
D40 -191.71 -6.68 <.0001
D43 -199.86 -4.96 <.0001
D47 -207.86 -3.51 0.0011
D50 -220.00 -3.18 0.0028
Global -68.5952 -2.24 0.0302
Combination of Compound (1) at 20 mg/kg and Compound (2b) at 20 mg/kg versus Compound (1) at 20 mg/kg D35 8.8571 | 0.26 0.7931
D37 49.0000 1.58 0.1205
D40 -115.71 -4.03 0.0002
D43 -129.71 -3.22 0.0026
D47 -122.43 -2.07 | 0.0450
D50 -101.57 —--———--— -1,47 0.1499
a Each combination was compared to the best single agent using estimâtes obtained from a 2-way analysis of variance with repeated measurements (Time factor) on parameter tumor volume (proc mixed of SAS 9.2 software). A probability less than 5% (p<0.05) was considered as significant.
-7916446
Table 20.
AT/AC (%) on d50
Compound (1) 20mg/kg 30
Compound (2a) 75mg/kg 53
Compound (2b) 20mg/kg 83
Compound (2a) 75mg/kg Compound (1) 20mg/kg -5
Compound (2b) 20mg/kg Compound (1) 20mg/kg 26
While there hâve been shown and described what are at présent considered the preferred embodiments of the invention, those skilled in the art may make various changes and modifications which remain within the scope of the appended daims.

Claims (13)

  1. WE CLAIM:
    1. A composition comprisîng a compound having the foliowing structural formula:
    or a pharmaceutically acceptable sait thereof, and a compound having a structural formula selected from the group consisting of and (2a) (2b) or a pharmaceutically acceptable sait thereof.
    -81 16446
  2. 2. The composition of claim 1, further comprising a pharmaceutically acceptable carrier.
  3. 3. The composition of claim 1, wherein said compound according to formula (1) and said compound according to formula (2a) or (2b) are in amounts that produce a synergistic effect in reducing tumor volume in a patient when said composition is administered to a patient.
  4. 4. A method of treating a patient with cancer, comprising administering to said patient a therapeutically effective amount of the compound of Formula (1), or a pharmaceutically acceptable sait thereof, in combination with the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof.
  5. 5. The method of claim 4, wherein the effective amount achieves a synergistic effect in reducing a tumor volume in said patient.
  6. 6. The method of claim 4, wherein the effective amount achieves tumor stasis in said patient.
  7. 7. The method of claim 4, wherein said cancer is selected from the group consisting of nonsmall cell lung cancer, breast cancer, pancreatic cancer, liver cancer, prostate cancer, bladder cancer, cervical cancer, thyroid cancer, colorectal cancer, liver cancer, muscle cancer, hematological malignancies, melanoma, endométrial cancer and pancreatic cancer.
  8. 8. The method of claim 4, wherein the cancer is selected from the group consisting of colorectal cancer, endométrial cancer, hematological malignancies, thryoid cancer, breast cancer, melanoma, pancreatic cancer and prostate cancer.
  9. 9. The method of claim 4, wherein said method comprises administering the compound of Formula (2a).
  10. 10. The method of claim 4, wherein said method comprises administering the compound of Formula (2b).
  11. 11. A combination for use in treating cancer, the combination comprising a therapeutically effective amount of (A) the compound of Formula (1 ), or a pharmaceutically acceptable sait thereof, and (B) the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof.
  12. 12. A kit comprising: (A) the compound of Formula (1), or a pharmaceutically acceptable sait thereof; (B) the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof; and (C) instructions for use.
  13. 13. Use of a combination comprising a therapeutically effective amount of (A) the compound
    -8216446 of Formula (1), or a pharmaceutically acceptable sait thereof, and (B) the compound of Formula (2a) or Formula (2b), or a pharmaceutically acceptable sait thereof, for the préparation of a médicament for use in treatment of cancer.
    -8316446 l/40
OA1201300235 2010-12-09 2011-12-08 Compositions comprising a PI3K inhibitor and a MEK inhibitor and their use for treating cancer. OA16446A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US61/421,465 2010-12-09
US61/436,258 2011-01-26
US61/467,485 2011-03-25
FR1159940 2011-11-03

Publications (1)

Publication Number Publication Date
OA16446A true OA16446A (en) 2015-10-15

Family

ID=

Similar Documents

Publication Publication Date Title
US20140024653A1 (en) Compositions and methods for treating cancer using pi3k inhibitor and mek inhibitor
Boss et al. Clinical evaluation of AZD1152, an iv inhibitor of Aurora B kinase, in patients with solid malignant tumors
Heist et al. Phase I/II study of AT-101 with topotecan in relapsed and refractory small cell lung cancer
EP3342411B1 (en) Rapamycin derivative for treating pancreas cancer
JP2012500180A5 (en)
Reck et al. Sunitinib in combination with gemcitabine plus cisplatin for advanced non-small cell lung cancer: a phase I dose-escalation study
US20170071903A1 (en) Use of eribulin and mtor inhibitors as combination therapy for the treatment of cancer
JP2017530983A (en) Synergistic auristatin combination
EP2570127A1 (en) Compositions and methods for treating cancer using PI3KB beta inhibitor and MAPK pathway inhibitor, including MEK and RAF inhibitors
KR20180129917A (en) Combination therapy of Notch and PI3K / mTOR inhibitors for use in cancer therapy
TWI447113B (en) Pharmaceutical combination
EP2754441B1 (en) Composition for preventing and treating non-small cell lung cancer, containing pyrazino-triazine derivatives
KR20210010524A (en) Composition comprising a bisfluoroalkyl-1,4-benzodiazepinone compound for the treatment of adenocyst carcinoma
CA2985379C (en) Micronized pharmaceutical compositions for treatment of angiogenisis conditions
OA16446A (en) Compositions comprising a PI3K inhibitor and a MEK inhibitor and their use for treating cancer.
KR20150003786A (en) Methods for treating cancer using pi3k inhibitor and mek inhibitor
US20220071982A1 (en) Methods and uses for treating cancer
TW202337469A (en) Methods of treating small cell lung cancer
Reyderman et al. Eribulin and cytochrome P450 effectors: in vitro studies and population pharmacokinetic–pharmacodynamic analysis
AU2016259775A1 (en) Cabazitaxel and its use for treating cancer
Pouessel et al. Phase II Study of TLN-232*, a novel M2PK targeting agent, administered by CIV to patients with advanced renal cell carcinoma (RCC)
OA16757A (en) Compositions and methods for treating cancer using P13K beta inhibitor and MAPK pathway inhibitor, including MEK and RAF inhibitors.
WO2023281413A1 (en) Methods and dosing regimens comprising pf-06873600 for the treatment of cancer
WO2023196329A1 (en) Combination therapies comprising gdc-6036 and gdc-0077 for the treatment of cancer
TW201315471A (en) Compositions and methods for treating cancer using PI3K β inhibitor and mapk pathway inhibitor, including MEK and RAF inhibitors