OA16325A - Combination therapy for treating HCV infection. - Google Patents

Combination therapy for treating HCV infection. Download PDF

Info

Publication number
OA16325A
OA16325A OA1201300063 OA16325A OA 16325 A OA16325 A OA 16325A OA 1201300063 OA1201300063 OA 1201300063 OA 16325 A OA16325 A OA 16325A
Authority
OA
OAPI
Prior art keywords
compound
pharmaceutically acceptable
acceptable sait
day
sait
Prior art date
Application number
OA1201300063
Inventor
Wulf Boecher
Carla Haefner
George Kukolj
Original Assignee
Boehringer Ingelheim International 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 Boehringer Ingelheim International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Publication of OA16325A publication Critical patent/OA16325A/en

Links

Abstract

The present invention relates to therapeutic combinations comprising (a) Compound (1), or a pharmaceutically acceptable salt thereof, as herein described, (b) Compound (2), or a pharmaceutically acceptable salt thereof, as herein described, and optionally (c) ribavirin, and methods of using such therapeutic combinations for treating HCV infection or alleviating one or more symptoms thereof in a patient.

Description

The présent invention relates to therapeutic combinations comprising Compounds (1) and;
(2) as herein described and optionally ribavirin. The présent invention also relates to methods of using such therapeutic combinations for treating HCV infection or alleviatingi one or more symptoms thereof in a patient.i
BACKGROUND OF THE INVENTION
Hepatitis C virus (HCV) infection is a global human health problem with approximately!
150,000 new reported cases each year in the United States alone. HCV is a single strandedj
RNA virus, which is the etiological agent identified in most cases of non-A, non-B posttransfusion and post-transplant hepatitis and is a common cause of acute sporadic hepatitis.i
It îs estimated that more than 50% of patients infected with HCV become chronically infected and 20% of those develop cirrhosis of the liver within 20 years.
Several types of interferons, in particular, alfa-interferons are approved for the treatment of chronic HCV, e.g., interferon-alfa-2a (ROFERON®-A), interferon-alfa-2b (INTRON®20 A), consensus interferon (INFERGEN®), as well as pegylated forms of these and other interferons like pegylated interferon alfa-2a (PEGASYS®) and pegylated interferon alfa2b (PEG-INTRON®). Most patients are unresponsive to interferon-alfa treatment, however, and among the responders, there is a high récurrence rate within 6 months after cessation of treatment (Liang et al., J. Med. Viral. 40:69, 1993).
Ribavirin, a guanosine analog with broad spectrum activity against many RNA and DNA viruses, has been shown in clinical trials to be effective against chronic HCV infection when used in combination with interferon-alfas (see, e.g., Poynard et al., Lancet 352:14261432, 1998; Reîchard et al., Lancet 351:83-87, 1998), and this combination therapy has j 30 been approved for the treatment of HCV: REBETRON® (interferon alfa-2b plus ribavirin, j Schering-Plough); PEGASYS®RBV® (pegylated interferon alfa-2a plus ribavirin X >
; -ij i
i i
Ί
Ί ί ] i
ί
I i combination therapy, Roche); see also Manns et al, Lancet 358:958-965 (2001) and Fried et al., 2002, N. Engl. J. Med. 347:975-982, However, even with this combination therapy the virologie response rate is still at or below 50%.
i | 5 In May 2011, the first direct acting anti virais (DAA) hâve been approved in US by the
J
FDA: the protease inhibitors boceprevîr and telaprevir (Poordad et al., 2011 N Engl J Med 364:1195-2062; Jacobson et al.,2011 N Engl J Med 364:2405-2416; Zeuzem et al., 2011 N Engl J Med 364:2417-2428) Adding one of these drugs to pegylated interferon and ‘ ribavirin improves the cure rate from 50% to 70-75%. Approvals in Europe and other ’ 10 countries are expected to follow later in 2011 or 2012. This treatment is expected to i become the new standard of care for large patient populations.
i
There are significant side-effects typically associated with such thérapies. Ribavirin | suffers from disadvantages that include tératogénie activity, interférence with sperm development, haemolysis, fatigue, headache, insomnia, nausea and/or anorexia. Interferon i 15 alfa, with or without ribavirin, is associated with many side effects. During treatment, i patients must be monitored carefully for flu-like symptoms, dépréssion, rashes and abnormal blood counts. Patients treated with interferon alfa-2b plus ribavirin should not ! hâve complications of serious liver dysfunction and such subjects are only considered for treatment of hepatitis C in. carefully monitored studies. Telaprevir and boceprevir hâve the j 20 following additional side effects: anémia, rash, dysgeusia, neutropenia, anorectal symptoms and others.
-216325
The following Compound (1):
having the chemical name: l-{[4-[8-Bromo-2-(2-isopropylcarbamoyl-thiazol-4-yl)-7methoxy-quinolin-4-yloxy]-1 -(R.)-(2-cyclopentyloxycarbonyl amino-3,3 -(S)-dimethylbutyryl)-pyrrolîdine-(S)-2-carbonyl ]-amino}-2-(S)-vinyl-cyclopropane-(R)-carboxylic acid, is known as a sélective and potent inhibitor of the HCV NS3 serine protease and useful in the treatment of HCV infection. Compound (1) falls within the scope of the acyclic peptide sériés of HCV inhibitors disclosed in U.S. Patents 6,323,180, 7,514,557 and 7,585,845. Compound (1) is disclosed specifically as Compound # 1055 in U.S. Patent 7,585,845, and as Compound # 1008 in U.S. Patent 7,514,557. Compound (1), and pharmaceutical formulations thereof, can be prepared according to the general procedures found in the above-cited référencés, ail of which are herein incorporated by reference in their entirety. Preferred forms of Compound (1) include the crystalline forms, in particular the crystalline sodium sait form as described in U.S. Patent Application Publication No. 2010/0093792, also incorporated herein by reference.
Compound (1) may also be known by the following alternate depiction of its chemical structure, which is équivalent to the above-described structure: qX
-316325 wherein B is
| 5 A combination therapy regimen including administering Compound (1) with an interferonj alpha and ribavirin is described in U.S. Patent Application Publication No. 2010/0068182.
However, in view of the potentîal side-effects and overall inconvenience of treatment with an interferon (administered by injection), there is a continuing need in the field for alternative thérapies for the treatment and prévention of HCV infection which do not in volve the use of an interferon.
Applicants have discovered that excellent antiviral results can be achieved by combining Compound (1) with an HCV polymerase inhibitor Compound (2), as hereinafter described, and optionally ribavirin, as a combination therapy without the use of an interferon.
The following Compound (2):
i i
-416325
having the chemical name: (E)-3-[2-(l -{[2-(5-Bromo-pyrimîdin-2-yl)-3-cyclopentyl-1 methyl-1 H-indoIe-6-carbonyl]-amino}-cyclobutyl )-3-methyl-3 H-benzimidazol-5-yl]acrylic acîd, is known as a sélective and potent inhibitor of the HCV NS5B RNAdependent RNA polymerase and useful in the treatment of HCV infection. Compound (2) falls within the scope of HCV inhibitors disclosed in U.S. Patents 7,141,574 and 7,582,770, and US Application Publication 2009/0087409. Compound (2) is disclosed specifically as Compound # 3085 in U.S. Patent 7,582,770. Compound (2), and pharmaceutical formulations thereof, can be prepared according to the general procedures found in the above-cited référencés, ail of which are herein incorporated by reference in their entirety. Preferred forms of Compound (2) include the crystalline forms, in particular the crystalline sodium sait form which is prepared as herein described.
BRIEF SUMMARY OF THE INVENTION
The présent invention provides a method of treating HCV infection or alleviating one or more symptoms thereof in a patient comprising the step of administering to the patient an effective amount of a therapeutic combination comprising Compounds (1) and (2) as herein described, or a pharmaceutically acceptable sait thereof, and optionally ribavirin. The two or three actives of the combination can be administered simultaneously or separately, as part of a regimen. A'
-516325
The présent invention further provides for a packaged pharmaceutical composition comprising a Compound (1), which is accompanied by written instructions indicating administering Compound (1) with Compound (2) and optionally ribavirin for the treatment of HCV infection.
The présent invention further provides for a packaged pharmaceutical composition comprising a Compound (2), which is accompanied by written instructions indicating administering Compound (1) with Compound (2) and optionally ribavirin for the treatment ofHCV infection.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 depicts the change in HCV viral load in a group of treatment-naïve patients having chronic HCV genotype-1 infection and treated with Compound (1) sodium sait (120 mg/day), Compound (2) sodium sait (1200 mg/day) and ribavirin as combination therapy for 4 weeks, followed by combination therapy with Compound (1) sodium sait, pegylated interferon alfa-2a and ribavirin.
Figure 2 depicts the change in HCV viral load in a group of treatment-naïve patients having chronic HCV genotype-1 infection and treated with Compound (1) sodium sait (120 mg/day), Compound (2) sodium sait ( 1800 mg/day) and ribavirin as combination therapy for 4 weeks, followed by combination therapy with Compound (1) sodium sait, pegylated interferon alfa-2a and ribavirin.
DETAILED DESCRIPTION OF THE INVENTION
Defini tions “Compound (1)” and “Compound (2)” are as defined above.
Ribavirin refers to Ι-β-D-ribofuranosyl-lH-l,2,4-tnazole-3-carboxainidc, available from ICN Pharmaceuticals, Inc., Costa Mesa, Calif. and is described in the Merck Index,
-616325 compound No. 8199, Eleventh Edition. Its manufacture and formulation is described in U.S. Pat. No, 4,211,771. Preferred marketed ribavirin products include REBETOL® and COPEGUS®. The term further includes dérivatives or analogs thereof, such as those described in U.S· Pat Nos. 6,063,772, 6,403,564 and 6,277,830. For example, dérivatives or analogs include modified ribavirins such as 5'-amino esters, ICN Pharmaceutical’s Lenantiomer of ribavirin (ICN 17261), 2'-deoxy dérivatives of ribavirin and 3carboxamîdine dérivatives of ribavirin, viramidine (previously known as ribamidine) and the like.
The term pharmaceutically acceptable sait means a sait of a Compound of formula (1) which is, within the scope of Sound medical judgment, suitable for use in contact with the tissues of humans and lower animais without undue toxicity, irritation, allergie response, and the like, commensurate with a reasonable benefit/risk ratio, generally water or oilsoluble or dispersible, and effective for their intended use.
The term includes pharmaceutically-acceptable acid addition salts and pharmaceuticallyacceptable base addition salts. Lists of suitable salts are found in, e.g., S. M. Birge et al., J. Pharm. Sci., 1977, 66, pp. 1-19.
The term pharmaceutically-acceptable acid addition sait means those salts which rctain the biological effectiveness and properties of the free bases and which are not biologîcally or otherwise undesirable, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, phosphoric acid, and the like, and organic acids such as acetic acid, trifluoroacetic acid, adîpic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, butyric acid, camphoric acid, camphorsulfonic acid, cinnamic acid, citric acid, digluconic acid, ethanesulfonic acid, glutamic acid, glycolic acid, glycerophosphoric acid, hemisulfic acid, hexanoic acid, formic acid, fumaric acid, 2-hydroxyethane-sulfonic acid (isethionic acid), lactic acid, hydroxymaleic acid, malic acid, malonic acid, mandelic acid, mesitylenesulfonic acid, methanesulfonîc acid, naphthalenesulfonic acid, nicotinic acid, 2-naphthalenesulfonic acid,
-716325 oxalic acid, pamoic acid, pectinic acid, phenylacetic acid, 3-phenylpropionic acid, pivalic acid, propionic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, sulfanilic acid, tartane acid, p-toluenesulfonic acid, undecanoic acid, and the like.
The terni pharmaceutically-acceptable base addition sait means those salts which retain the biological effectiveness and properties of the free acids and which are not biologically or otherwise undesirable, formed with inorganic bases such as ammonia or hydroxide, carbonate, or bicarbonate of ammonium or a métal cation such as sodium, potassium, lithium, calcium, magnésium, iron, zinc, copper, manganèse, aluminum, and the like. Particularly preferred are the ammonium, potassium, sodium, calcium, and magnésium salts. Salts derived from pharmaceutically-accepta- ble organic nontoxic bases include salts of primary, secondary, and tertiary amines, quatemary amine compounds, substituted amines including naturally occurring substituted amines, cyclic amines and basic ionexchange resins, such as methylamine, dimethylamine, trimethylamine, ethylamine, diethylamine, triethylamine, isopropylamine, tripropylamine, tributylamine, ethanolamine, diethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, hydrabamine, choline, betaine, ethylenedîamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, Nethylpiperidine, tétraméthylammonium compounds, tetraethylammonium compounds, pyridine, Ν,Ν-dimethylaniline, N-methylpiperidine, N-methylmorpholine, dicyclohexylamine, dibenzylamine, Ν,Ν-dibenzylphenethylamine, 1-ephenamine, N,N'dibenzylethylenediamine, polyamine resins, and the like. Particularly preferred organic nontoxic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
The term therapeutic combination as used herein means a combination of one or more active drug substances, i.e., compounds having a therapeutic utility. Typicalîy, each such compound in the therapeutic combinations of the présent invention will be présent in a pharmaceutical composition comprising that compound and a pharmaceutically acceptable carrier. The compounds in a therapeutic combination of the présent invention may be administered simultaneously or separately, as part of a regimen. qZ~
-816325
Embodiments of the Invention
According to a general embodiment, the présent invention provides for a method of treating HCV infection or alleviating one or more symptoms thereof in a patient comprising the step of administering to the patient an effective amount of a therapeutic combination comprising a Compound (1) as defined herein, or a pharmaceutically acceptable sait thereof, Compound (2) as defined herein, or a pharmaceutically acceptable sait thereof, optionally together with ribavirin. An additional embodiment is directed to the use of Compound (1), or a pharmaceutically acceptable sait thereof, and Compound (2) or a pharmaceutically acceptable sait thereof, for the manufacture of pharmaceutical compositions of each compound, for use together, optionally also with ribavirin, in the treatment of HCV infection.
Additional general embodiments include a packaged pharmaceutical composition comprising a packaging containing one or more doses of Compound (1) or a pharmaceutically acceptable sait thereof, or containing one or more doses of Compound (2) or a pharmaceutically acceptable sait thereof, together with written instructions directing the co-administration of Compound (1), Compound (2) and optionally ribavirin for the treatment of HCV infection. Another embodiment is directed to a kit for the treatment of HCV infection comprising: (a) one or more doses of Compound (1) or a pharmaceutically acceptable sait thereof, and (b) one or more doses of Compound (2) or a pharmaceutically acceptable sait thereof, and written instructions directing the coadministration of Compound (1), Compound (2) and optionally ribavirin for the treatment of HCV infection.
In administering the therapeutic combinations of the présent invention, each active agent can be administered together at the same time or separately at different times in separate dosage administrations. The présent invention contemplâtes and includes ali such dosage
-916325 regimens when admînistering the double or triple therapeutic combinations as defined herein.
Although this combination therapy is expected to be effective against ail HCV génotypes, it has been demonstrated to be particularly effective in treating HCV génotype 1 infection, including subgenotypes la and lb.
The patient population to be treated with the combination therapy of the présent invention can be further classified into “treatment-naïve” patients, i.e., those patients who hâve not received any prior treatment for HCV infection, including but not limited to interferonintolerant or contraindicated patients, and “treatment experienced” patients, i.e, those patients who hâve undergone prior treatment for HCV. Either of these classes of patients may be treated with the combination therapy of the présent invention. A particular class of patients that are preferably treated are those treatment experienced patients that hâve undergone prior interferon plus ribavirin therapy but are non-responsive to said therapy (herein “non-responders”). Such non-responders include three distinct groups ofpatients: (1) those who experienced < 2x log10 maximum réduction in HCV RNA levels during the first 12 weeks of treatment with interferon plus ribavirin (“null responders”), (2) those who experienced > 2x logio maximum réduction in HCV RNA levels during treatment with interferon plus ribavirin but never achieve HCV RNA levels below level of détection (“partial responders”), and (3) those who achieved a virologie response with and during interferon plus ribavirin therapy but had a viral load rebound either during treatment (other than due to patient non-compliance) or after treatment has completed (“relapser”).
According to an alternative embodiment, the présent invention provides a method of reducing HCV-RNA levels in a patient in need thereof, comprising the step of admînistering to said patient a therapeutic combination according to the présent invention. Preferably, the method of the présent invention reduces the HCV-RNA levels in a patient to a level below the lower limit of quantification (or “BLQ”). A BLQ level of HCV RNA as used in the présent invention means a level below 25 International Units (IU) per ml of sérum or plasma of a patient as measured by quantitative, multi-cycle reverse transcriptase ,
-1016325
È I ι
I i !
i t
i i
i
I
PCR methodology according to the WHO international standard (Saladanha J, Lelie N and Heath A, Establishment of the first international standard for nucleic acid amplification technology (NAT) assays for HCV RNA, WHO Collaborative Study Group. Vox Sang 76:149-158, 1999). Such methods are well known in the art. In a preferred embodiment, the method of the présent invention reduces the HCV-RNA leveis in a patient to less than
IU per ml of sérum or plasma. In another embodiment the method of the présent invention reduces the HCV-RNA leveis in a patient to less than a detectible level.
The usual duration of the treatment for standard interferon plus ribavirin therapy is at Ieast 48 weeks for HCV génotype 1 infection, and at Ieast 24 weeks for HCV génotypes 2 and 3.
However, with the triple combination therapy of the présent invention it may be possible to have a much shorter duration of treatment. With the triple combination therapy of the présent invention the contemplated durations of treatment include at Ieast 4 weeks, preferably at Ieast 12 weeks, e.g., from about 12 weeks to about 24 weeks, although treatment up to and even beyond 48 weeks is possible as well. Thus, further embodiments include treatment for at Ieast 24 weeks and for at Ieast 48 weeks. The time period for different HCV génotypes, e.g. HCV génotypes 2, 3, 4, 5 or 6 is expected to be similar, Also contemplated is an initial treatment regimen with the triple combination therapy of the présent invention, followed by a combination therapy of only Compound (1) with ribavirin (and with or without interferon) or followed by a combination therapy of only Compound (2) with ribavirin (and with or without interferon).
The first component of the therapeutîc combination, namely, Compound (1 ) or a pharmaceutically acceptable sait thereof is comprised in a composition. Such a composition comprises Compound (1), or a pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable adjuvant or carrier. Typical pharmaceutical compositions that may be used for Compound (l), or a pharmaceutically acceptable sait thereof, are as described in U.S. Patent 7,514,557. Further spécifie examples of compositions are as set forth in the examples section below. pA''
-1116325
In general, the Compound (l) or a pharmaceutically acceptable sait thereof may be administered at a dosage of at least 40 mg/day (in single or divided doses). Additional embodiments for dosage amounts and ranges may include (in single or divided doses):
(a) at least 100 mg/day (b) at least 120 mg/day (c) at least 200 mg/day (d) at least 240 mg/day (e) at least 360 mg/day (f) at least 480 mg/day (g) from about 40 mg/day to about 480 mg/day (h) from about 120 mg/day to about 240 mg/day (i) from about 240 mg/day to about 480 mg/day (j) about 120 mg/day (k) about 240 mg/day (l) about 360 mg/day (m) about 480 mg/day
Although Compound (1) or a pharmaceutically acceptable sait thereof may be administered in single or divided daily doses, once a day administration (QD) of the daily dose is preferred. As the skilled artisan will appreciate, however, lower or higher doses than those recited above may be required. Spécifie dosage and treatment regimens for any particular patient will dépend upon a variety of factors, including the âge, body weight, general health status, sex, diet, time of administration, rate of excrétion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physician. In general, the compound is most desirably administered at a concentration level that will generally afford antivirally effective results without causing any harmful or deleterious side effects.
In another embodiment according to the invention, a loading dose amount of Compound (1) is administered for the first administration dose ofthe treatment. The loading dose amount is higher than the dose amount administered for subséquent administrations in the
-1216325 treatment. Preferably, the loading dose amount is about double in quantity, by weight, of the amount in subséquent administrations in the treatment. For example, in one embodiment, the first dose of Compound (1) administered at dosage of about 240 mg and subséquent doses of Compound (1) are administered at a dosage of about 120 mg. In another embodiment, the first dose of Compound (1) administered at a dosage of about 480 mg and subséquent doses of Compound (1) are administered at a dosage of about 240 mg. In another embodiment, the first dose of Compound (1) administered is at a dosage of about 960 mg and subséquent doses of Compound (1) are administered at a dosage of about 480 mg.
B y using this loading dose concept, a clear advantage is that it is thereby possible to achieve steady state levels of active drug in the patient’s System earlier than would dtherwise be achieved. The blood level achieved by using a doubled loading dose is the same as would be achieved with a double dose but without the safety risk attendant to the 15 subséquent continuous administration of a double dose. By reaching the targeted steady state level of active drug earlier in therapy also means that there less possibility of insufficient drug pressure at the beginning of therapy so that résistant viral strains hâve a smaller chance of emerging.
The second component of the therapeutic combination, namely, Compound (2) or a pharmaceutically acceptable sait thereof is comprised in a composition. Such a composition comprises Compound (2), or a pharmaceutically acceptable sait thereof, and a pharmaceutically acceptable adjuvant or carrier. Typical pharmaceutical compositions that may be used for Compound (I), or a pharmaceutically acceptable sait thereof, are as described in U.S. Patent 7,582,770.
In general, the Compound (2) or a pharmaceutically acceptable sait thereof may be administered at dosage amounts and in dose ranges that may include (in single or divided doses):
(a) at least 800 mg/day
-1316325 (c) at least ISOOmg day (d) at least 2400 mg/day (e) from about 800 mg/day to about 2400 mg/day (f) from about 1200 mg/day to about 1800 mg/day (g) from about 1800 mg/day to about 2400 mg/day (h) from about 1200 mg/day to about 2400 mg/day (i) about 1200 mg/day (j) about 1800 mg/day (k) about 2400 mg/day
Although Compound (2) or a pharmaceutically acceptable sait thereof may be administered in single or divided daily doses, twice a day (BID) or thrice a day administration (TID) of the divided daily dose are preferred. As the skilled artisan will appreciate, however, lower or higher doses than those recited above may be required. Spécifie dosage and treatment 15 regimens for any particular patient will dépend upon a variety of factors, including the âge, body weight, general health status, sex, diet, time of administration, rate of excrétion, drug combination, the severity and course of the infection, the patient's disposition to the infection and the judgment of the treating physicîan. In general, the compound is most desirably administered at a concentration Ievel that will generally afford antîvirally effective results without causing any harmful or deleterious side effects.
In another embodiment according to the invention, an induction dose amount of Compound (2) is administered for the first administration dose of the treatment. The induction dose amount is higher than the dose amount administered for subséquent administrations in the treatment. Preferably, the induction dose amount is about double to triple in quantity, by weight, of the amount in subséquent administrations in the treatment.
For example, in one embodiment, the first dose of Compound (2) administered at dosage of about 1200 mg and subséquent doses of Compound (2) are administered at a dosage of about 600 mg. In another embodiment, the first dose of Compound (2) administered at a 30 dosage of about 1200 mg and subséquent doses of Compound (2) are administered at a dosage of about 400 mg.
-1416325
By using this induction dose concept, a clear advantage is that it is thereby possible to achieve a greater drop in initial viral load. Maximizing initial viral response with the first dose and then sustaining the drop with a subséquent lower dose also restricts the sélection of potential résistant variants.
The optional third component of the therapeutic combination, namely ribavirin, is comprised in a pharmaceutical composition. Typically, such compositions comprise ribavirin and a pharmaceutically acceptable adjuvant or carrier and are well known in the art, including in a number of marketed ribavirin formulations. Formulations comprising ribavirin are also disclosed, e.g., in US Patent 4,211,771.
The types of ribavirin that may be used in the combination are as outlined hereinabove in the définitions section. In one preferred embodiment, the ribavirin is either REBETOL® or COPEGUS® and they may be administered at their labeled dosage Ievels indicated for interferon plus ribavirin combination therapy for the treatment of HCV infection. Of course, with the triple combination therapy of the présent invention it may be possible to use a lower dosage of ribavirin, e.g., lower than is used the current standard interferon plus ribavirin therapy, while delivering the same or better efficacy than the current standard therapy with less side-effects usually associated with such therapy.
According to various embodiments, the ribavirin may be administered at dosages of (in single or divided doses):
(a) between 400 mg/day to about 1200 mg/day;
(b) between about 800 mg/day to about 1200 mg/day;
(c) between about 1000 mg/day to about 1200 mg/day;
(d) about 1000 mg/day (e) about 1200 mg/day (f) between about 300 mg/day to about 800 mg/day (g) between about 300 mg/day to about 700 mg/day (h) between 500 mg/day to about 700 mg/day (i) between 400 mg/day to about 600 mg/day m/
-1516325 (j) about 400 mg/day (k) about 600 mg/day (l) about 800 mg/day
According to one embodiment, the ribavirin composition comprises ribavirin in a formulation suitable for dosing once a day, twice daily, thrice daily, four times a day, five times a day, or six times a day. For example, if a therapeutic combination comprises about 1000 mg/day dosage of ribavirin, and a dosing of five times a day is desired, then the therapeutic combination will comprise ribavirin în a formulation, e.g., a tabiet, containing, e.g., about 200 mg of ribavirin.
For example, in one embodiment the présent invention contemplâtes a method of treating hepatitis C viral (HCV) infection or alleviating one or more symptoms thereof in a patient comprising the step of administering to the patient a therapeutic combination comprising:
(a) Compound (1) or a pharmaceutically acceptable sait thereof at a dosage between about 48 mg per day and about 480 mg per day;
(b) Compound (2) or a pharmaceutically acceptable sait thereof at a dosage between about 800 mg/day to about 2400 mg/day; and (c) optionally ribavirin at a dosage of between about 400 mg/day to about 1200 mg/day.
In another embodiment the présent invention contemplâtes a method of treating hepatitis C viral (HCV) infection or alleviating one or more symptoms thereof in a patient comprising the step of administering to the patient a therapeutic combination comprising:
(a) Compound (1) or a pharmaceutically acceptable sait thereof at a dosage between about 120 mg/day to about 240 mg/day;
(b) Compound (2) or a pharmaceutically acceptable sait thereof at a dosage between about 1200 mg/day to about 1800 mg/day; and (c) optionally ribavirin at a dosage of between about 1000 mg/day to about 1200 mg/day.
-1616325 j In another embodiment the présent invention contemplâtes a method of treating hepatitis C l viral (HCV) infection or alleviating one or more symptoms thereof in a patient comprising j the step of administering to the patient a therapeutic combination comprising:
I 5 j (a) Compound (1) or a pharmaceutically acceptable sait thereof at a dosage of
J ï about 120 mg/day;
i (b) Compound (2) or a pharmaceutically acceptable sait thereof at a dosage of about 1200 mg/day or about 1800 mg/day; and (c) optionally ribavirin at a dosage of between about 1000 mg/day to about 1200 mg/day.
! Further embodiments include any of the above-mentioned embodiments, and where:
(a) the therapy is a triple combination therapy including administration of
Compound (1) or a pharmaceutically acceptable sait thereof, Compound (2) ) or a pharmaceutically acceptable sait thereof and ribavirin; or j (b) the therapy is a double combination therapy including administration of i Compound (1) or a pharmaceutically acceptable sait thereof and Compound (2) ! or a pharmaceutically acceptable sait thereof, i.e., without any additional antij 20 HCV agents.
Further embodiments include any of the above-mentioned embodiments, and where:
P
I
I (a) the HCV infection is génotype 1 and the patient is a treatment-naïve patient; or (b) the HCV infection is génotype 1 and the patient is a treatment-experienced patient who is non-responsive to a combination therapy of interferon plus j ribavirin.
J i
Further embodiments include any of the above-mentioned embodiments, and where the
Compound (1) or a pharmaceutically acceptable sait thereof is administered once a day, the *
i t
I
Compound (2) or a pharmaceutically acceptable sait thereof is administered three times a day and the ribavirin, if included in the therapy, is administered twice a day.
Further embodiments include any of the above-mentioned embodiments and where the loadîng dose concept in used for Compound (l), e.g., the first dose of Compound (1) administered is double in quantity to the subséquent doses.
Further embodiments include any of the above-mentioned embodiments, and where the therapeutic regimen of the présent invention is administered to the patient for at least about
4 weeks, more preferably at least about 12 weeks, at least about 16 weeks, at least about weeks, at least about 28 weeks or at least about 40 weeks.
With respect to the double or triple combination thérapies of the présent invention, the présent invention contemplâtes and includes ail combinations of the various preferred embodiments and sub-embodiments as set forth herein.
An additional embodiment is directed to a packaged pharmaceutical composition comprising a packaging containing one or more doses of Compound (1) or a pharmaceutically acceptable sait thereof, or containing one or more doses of Compound (2) or a pharmaceutically acceptable sait thereof, each together with written instructions directing the co-administration of Compound (1), Compound (2) and optionally ribavirin for the treatment of HCV infection. In another embodiment, one or more doses of Compound (1) and one or more doses of Compound (2) are placed together in a single packaging forming a so-called “kit”, which includes written instructions directing the co25 administration of Compound (1), Compound (2) and optionally ribavirin for the treatment of HCV infection. In either case, the individual doses of Compound (1) or a pharmaceutically acceptable sait thereof, or Compound (2) or a pharmaceutically acceptable sait thereof, can be in the form of any of the standard pharmaceutical dosage forms, e.g. tablets, capsules, and packaged within any of the standard types of pharmaceutical packaging materials, e.g. bottles, blister-packs, etc., that may themselves be contaîned within an outer packaging material such as a paper/cardboard box. The written instructions will typically be provided either on the packaging material(s) itself or yd
-1816325 î
I I on a separate paper (a so-called “package insert”) that is provided together with the dosage forms within the outer packaging material. Ail such packaging embodiments and variations thereof are embraced by the présent invention.
j | 5 Additionally, surprising results hâve been seen in the excellent antiviral activity and ; suppression of HCV viral réplication and limited emergence of viral résistance during the > combination therapy treatment contemplated by the présent invention. Accordingly, in an.
I additional embodiment, there is limited or no emergence of viral résistance during the combination therapy of the présent invention. In a further embodiment, there is limited or no emergence of HCV variants that encode HCV NS3 protease amino acid substitutions at î one or more of RI 55 and/or D168 and/or A156 during the combination therapy of the présent invention In a further embodiement there is limited or no emergence of HCV variants that encode HCV NS5B polymerase amino acid substitutions at P495 during the combination therapy of the présent invention. In a more spécifie embodiement there is limited or no emergence of HCV variants that encode both HCV NS3 protease amino acid î substitutions (NS3R155 and/or NS3D168 and/or NS3A156) and HCV NS5B polymerase ( amino acid substitutions P495 during the combination therapy of the présent invention j
[ Further embodiments include any of the above-mentioned embodiments, and where either:
(a) the HCV infection is génotype la and the patient is a treatment-naïve patient; or i
(b) the HCV infection is génotype 1 a and the patient is a treatment-experienced patient who is non-responsive to a combination therapy of interferon plus ribavirin;
and wherein there is limited or no emergence of variants that encode substitutions at NS3 î protease amino acid R155 and substitutions at NS5B polymerase P495 during the t
( combination therapy of the présent invention,
-1916325 j
i ΐ Examples
I. Methods for Preparing Compound (!)
Methods for preparing amorphous Compound (1) and a general description of i pharmaceutically acceptable sait forms can be found in US Patents 6,323,180, 7,514,557 j
and 7,585,845. Methods for preparing additional forms of Compound (1), in particular the crystalline sodium sait form, can be found in U.S. Patent Application Publication No. 2010/0093792.
II. Formulations of Compound (1)
One example of a pharmaceutical formulation of Compound (1) include an oral solution formulation as disclosed in WO 2010/059667. Additional examples include capsules containing a lîpid-based liquid formulation, as disclosed in WO 2011/005646. Examples 15 of such capsule formulations are described below.
Example 1 - Softgel Capsule Formulation # 1
The composition of the liquid fill formulation:
Monograph Functionality % w/w
Ingrédient
Compound (1) Na sait API 15.0
Mono-, Diglycerides of Caprylic/Capric Acid (Capmul®MCM) Lipid 46.3
Polyoxyl 35 Castor Oil (Cremophor® EL) NF Surfactant 30.8
Propylene Glycol USP Solvent 7.7
DL-a-tocopherol USP Anti-oxidant 0.2
Total 100.0
Two spécifie soft-gel capsule drug product formulations were prepared according to the above general Formulation # 1, a 40 mg product and a 120 mg product:
-2016325
Ingrédient Function 40 mg 120 mg
mg/capsule mg/capsule
Compound (1) Na sait (milled) Drug substance 42.301 126.902
Mono/Diglycerides of Caprylic/Capric Acid Lipid phase 130.57 391.70
Polyoxyl 35 Castor Oil (NF) Macrogolglycerol Ricinoleate (Ph. Eur.) Surfactant 86.86 260.57
Propylene Glycol Solvent 21.71 65.14
Vitamin E (dl-alpha tocopherol) (USP) All-rac-alpha-tocopherol (Ph. Eur.) Antioxidant 0.56 1.69
Nitrogen3 Processing aid q.s. q.s.
Total Fill Weight 282.00 846.00
Soft Gelatin Capsule Shell Shell 28(4 59(4
Wet Total Capsule Weight 562 1436
Dry Total Capsule Weight 480 1250
ΐ ! 42.30 mg of Compound (1) Na sait is équivalent to 40.0 mg of the active moiety.
i 2 126.90 mg of Compound (1) Na sait is équivalent to 120.0 mg of the active moiety.
ί 3 Nitrogen is used as a processing aîd and does not appear in the final product.
4 The approximate weight of the capsule shell before drying and finishing is 280 mg. The approximate weight of the capsule shell after drying and finishing is 198 mg.
5 The approximate weight of the capsule shell before drying and finishing is 590 mg. The approximate weight of the capsule shell after drying and finishing is 404 mg.
Exampie 2 - Softgel Caspule Formulation # 2
The composition of the liquid fill formulation:
Monograph Functionality % w/w
Ingrédient
Compound (1) Na sait API 15.0
Mono-, Diglycerides of Caprylic/Capric Acid (Capmul®MCM) Lipid 42.4
Polyoxyl 35 Castor Oil (Cremophor® EL) NF Surfactant 33.9
] -21j
I i
i j
Propylene Glycol USP Solvent -
Oleic Acid Lipid 8.5
DL-a-tocopherol USP Anti-oxidant 0.2
Total 100.0
A spécifie 150 mg soft-gel capsule drug product formulation was prepared according to the above general formula.
Example 3 - Hard Shell Capsule Formulation # 3
The composition of the liquid fill formulation:
Monograph Functionaiity % w/w
Ingrédient
Compound (1) Na sait API 20.0
Mono-, Diglycerides of Caprylic/Capric Acid (Capmul® MCM) Lipid 53.8
Polyoxyl 35 Castor Oil (Cremophor® EL) NF Surfactant 23.0
Propylene Glycol USP Solvent 3.0
DL-a-tocopherol USP Anti-oxidant 0.2
Total 100.0
A spécifie 150 mg hard-shell capsule drug product formulation was prepared according to the above general formula.
Préparation of Formulations 1-3:
The drug substance is jet-milled to remove large aggregates so that the mixing time for the bulk fill manufacturing will be consistent and reasonably short. The target particle size distribution of the drug substance is to reduce the x90 (v/v) to no more than 10 micron and the x98 (v/v) to no more than 20 micron as measured by Sympatec. Ail the excipients in the fill formulation are combined in a mixing vessel and mixed until uniform prior to adding the drug substance. After addition of the drug substance, mixing continues until the fill solution is clear by visual inspection. A nitrogen blanket over the fill solution is used throughout the préparation as a standard practice. The fill solution is passed through a y/
-2216325 filter to remove any extraneous particles. Encapsulation of the filtered bulk fill material in capsules is performed utilizing standard soft gelatin or hard gelatin capsule technology and in-process controls. Filled capsules are dried and then washed with a finishing/wash solution prior to packaging resulting in shiny, pharmaceutically élégant capsules.
III. Methods for Preparing Compound (2)
Methods for preparing amorphous Compound (2) canbe found in U.S. Patents 7,141,574 and 7,582,770, and US Application Publication 2009/0087409.
The following Example provides the method for preparing an additional form of Compound (2), the sodium sait form, that may be used in the présent invention.
Example 4 - Préparation of Compound (2) Sodium Sait
Step 1. Synthesis of Isopropyl 3-CyclopentyI-l-methyl-17Z-mdole-6-carboxylate
Because of the instability of brominated product, methyl 3-cyclopentyl-l-methyl-1Hindole-6-carboxylate needed to be converted into the more stable isopropyl 3-cyclopentyl1 -methyl-1 H-indoIe-6-carboxylate via a simple and high yielding operation. The conversion worked the best with stoîchiometric amounts of solid lithium isopropoxide. Use of 0.1 eq lithium isopropoxide led to longer reaction times and as a resuit to more hydrolysis by-product, while lithium isopropoxide solution in THF caused a problematic isolation and required distillation of THF.
Procedure:
-2316325
The mixture of methyl 3-cyclopentyl-1 -methyl-lH-indole-6-carboxylate (50.0 g, 0.194 mol) and lithium îsopropoxide (16.2 g, 95%, 0.233 mol) in 2-propanol was stirred at 65±5 °C for at least 30 min for complété trans-esterification. The batch was cooled to 40±5 °C and water (600 g) was added at a rate to maintam the batch température at 40±5°C. After addition, the mixture was cooled to 20-25 °C over 2±0.5 h and held at 20-25 °C for at least 1 h. The batch was filtered and rinsed with 28 wt% 2-propanol in water (186 g), and water (500 g). The wet cake was dried in vacuo (< 200 Ton) at 40-45 °C until the water content was < 0.5% to give isopropyl 3-cyclopentyl-l-methyl-lH-indole-6-carboxyIate (52.7 g, 95% yield) in 99.2 A% (240 nm).
The starting material methyl 3-cyclopentyl-l -methyl-1 H-îndole-6-carboxyIate can be prepared as described in Example 12 of U.S, Patent 7,141,574, and in Example 12 of U.S. Patent 7,642,352, both herein incorporated by reference.
Step 2. Synthesis of Isopropyl 2-Bromo-3-cyclopentyl-l-methyI-l//-indole-6carboxylate
(1) Bf2, ch3cn (2) Na2S203, K2O
4-methy I morph of ine
This process identified the optimal conditions for the synthesis of 2-bromo-3-cyclopentyl1-methyl-lH-îndole-6-carboxylate via bromination ofthe corresponding 3-cyclopentyl-lmethyl-lH-indole-6-carboxylate with bromine. It’s very important to control the reaction température and to quench the reaction mixture with a mixture of aqueous sodium thiosulfate and 4-methylmorpholine to minimize the formation of the dibromo- and 2indolone impurities. Further neutralîzatîon of the crude product with NaOH in isopropanol greatly increases the stabiiity of the isolated product. yA'
-2416325
Procedure:
The mixture ofisopropyl 3-cyclopentyl-1 -methyl-lH-indole-6-carboxylate (50.0 g, 0.175 mol) and acetonitrile (393 g) was cooled to -6±3 °C. Bromine (33.6 g, 0.210 mol) was added while the batch was maintained at -6=3l,C. The resulting slurry was stirred at 5 6±3°C for at least 30 min. When HPLC showed > 94 % conversion (the HPLC sample must be quenched immediately with aqueous 4-methylmorpholine/sodium thiosulfate solution), the mixture was quenched with a solution of sodium thiosulfate (15.3 g) and 28.4 g 4-methylmorpholine in water (440 g) while the température was maintained at -5±5 °C.
After it was stirred at 0±5 °C for at least 2 h, the batch was fîltered and rinsed with 85 wt% methanol/water solution (415 g), followed by water (500 g), and dried until water content îs < 30%. The wet cake was suspended in 2-propanol (675 g), and heated to 75±5 °C. The resulting hazy solution was treated with 1.0 M aqueous sodium hydroxide solution (9.1 g) and then with 135.0 g water at a rate to maintain the batch at 75±5°C. The suspension was stirred at 75±5°C for at least 30 min, cooled to 15±2 °C over 30-40 min, and held at 15±2 °C for at least 1 h. The batch was fîltered, rinsed with 75 wt% 2-propanol/water solution (161 g), and dried in vacuo (<200 Ton) at 50-60 °C until the water content was < 0.4% to give isopropyl 2-bromo-3-cyclopentyl-l-methyl-177-indole-6-carboxylate as a solid (55.6 g, 87 % yield ) in 99.5 A% (240 nm) and 97.9 Wt%.
Alternative Procedure:
The mixture ofisopropyl 3-cyclopentyl-1 -methyl- 177-indole-6-carboxylate (84 g, 0.294 mol) and isopropyl acetate (1074 g) was cooled to between -10-0 °C. Bromine (50 g, 0.312 mol) was added while thebatch was maintained at-10- 0 °C. The resulting slurry 25 was stirred at the same température for additional 30 min and quenched with a pre-cooled solution of sodium thiosulfate pentahydrate (13 g) and triethylamîne (64.5 g) în water (240
g) while the température was maintained at 0-10 °C. The mixture was heated to 40 - 50 °C and charged with methanol (664 g). After it was stirred at the same température for at least 0.5 h, the batch was cooled to 0 - 10 °C and stirred for another 1 hr. The precipîtate was fîltered, rinsed with 56 wt% methanol/water solution (322 g), and dried in vacuo (<200
-2516325
Torr) at 50-60 °C until the water content was < 0.4% to give isopropyl 2-bromo-3cyclopentyl-l-methyl-l/Z-indole-6-carboxylate as a beige solid (90-95 g, 80-85 % yield ).
Step 3a,b. Préparation of compound I by one-pot Pd-catalyzed borylation-Suzuki coupling reaction
To a clean and dry reactor containing 20.04 g of isopropyl 2-bromo-3-cyclopentyl-1methyl-lZ/-indole-6-carboxylate, 1.06 gof Pd(TFP)2Cl2(3 mol%) and 0.76 g of tri(2furyl)phosphine (6 mol%) was charged 8.35 g of triethylamine (1.5 équivalent), 39.38 g of CHjCN at 23±10 °C under nitrogen or argon and started agitation for 10 min. 9.24 g of
4,4,5,5-tetramethyl-l,3,2-dioxaborolane was charged into the reactor. The mixture was heated to reflux (ca. 81 -83 °C) and stirred for 6h until the reaction completed. The batch was cooled to 30±5 °C and quenched with a mixture of 0.99 g of water in 7.86 g of CH3CN. 17.24 g of 5-bromo-2-iodopyrimidine and 166.7 g of degassed aqueous potassium phosphate solution (pre-prepared from 46.70 g of KaPC^and 120 g of H2O) was charged subsquently under argon or nitrogen. The content was heated to reflux (ca. 76-77 °C) for 2 <
-2616325 h until the réaction completed. 4.5 g of 1-methylimidazole was charged into the reactor at 70 °C. The batch was cooled to 2O±3 °C over 0.5h and hold at 2Q±3 °C for at least lh. The solid was collected by filtration. The wet cake was first rinsed with 62.8 g of 2-propanol, followed by 200 g of H?O. The solid was dried under vacuum at the température below 50 “C.
Into a dry and clean reactor was charged dried 1,10 wt% Norit SX Ultra and 5 V of THF. The content was heated at 60±5 °C for at least 1 h. After the content was cooled to 3 5±5 °C, the carbon was filtered off and rinsed with 3 V of THF. The filtrate was charged into a clean reactor containing 1-methylimidazole (10 wt % relative to I). After removal of 5 V of THF by distillation, the content was then cooled to 31±2 °C. After the agitation rate was adjusted to over 120 rpm, 2.5 V of water was charged over a period of at least 40 minutes while maintaining the content température at 31 ± 2 °C. After the content was agitated at 31 ± 2 °C for addîtional 20 min, 9.5 V of water was charged into the reactor over a period of at least 30 minutes at 31 ± 2 °C. The batch was then cooled to about 25 ± 3 °C and stirred for addîtional 30 minutes. The solid was collected and rinsed with 3 V of water. The wet product I was dried under vacuum at the température below 50 °C (19.5 g, 95 wt%, 76% yield).
Alternative Procedure:
To a clean and dry reactor containing 40 g of isopropyl 2-bromo-3-cyclopentyI-l-methylI//-indoie-6-carboxylate (0.110 mol), 0.74 g of Pd(OAc)2 (3.30 mmol, 3 mol% equiv.) and
3.2 g of tri(2-furyl)phosphine (13.78 mmol, 12.5 mol% equiv.) was charged 16.8 g of triethylamine (1.5 équivalent), 100 mL of acetonitrile at 25 °C under nitrogen or argon.
20.8 g of 4,4,5,5-tetramethyl-l,3,2-dioxaborolane was charged into the reactor within 30 min. The mixture was heated to reflux (ca. 81-83 °C) and stirred for over 5 hrs until the reaction completed. The batch was cooled to 20 °C and quenched with a mixture of 2.7 g of water in 50 mL of CH3CN. The batch was warmed to 30 °C, stirred for 1 hr and transferred to a second reactor containing 34.4 g of 5-bromo-2-iodopyrimidine in 100 mL of acetonitrile. The reactor was rinsed with 90 mL of acetonitrile. To the second reactor was charged with degassed aqueous potassium phosphate solution (pre-prepared from 93.2 g of K3PO4 and 100 g of H2O) under argon or nitrogen. The content was heated to reflux \[/~
-2716325 (ca. 80 °C) for over 3 h until the reaction completed, 9.2 g of l -methylimidazole was charged into the reactor at 70 °C and the mixture was stirred for at least 10 min. The aqueous phase was removed after phase séparation. 257 g of isopropanol was charged at 70 °C. The batch was cooled slowly to 0 °C and hold for at least 1 h. The solid was collected by filtration. The wet cake was rinsed twîce with 2-propanol (2 x 164 g) and dried under vacuum at the température below 50 °C to give I as a yellow to brown solid (26 g, 75% yield).
Step 4. Hydrolysis of I Éo II
+ f-PrOH + NaOAc
I (20 g) and 1-methyl-2-pyrrolîdinone (NMP) (113 g) were charged into a clean reactor under nitrogen. After the batch was heated to 50-53 °C with agitation, premixed aq. NaOH (5.4 g of 50% aq. NaOH and 14.3 g of water) was introduced into the reactor. The the resulting mixture was stirred at 50-53 °C for about 10 hrs until the reaction completed. A premixed aq. HOAc (60 g of water and 9.0 g of HOAc) was added over 0.5 h at 45 ±5 °C to reach pH 5.5- 7.5. The batch was cooled to 20±5 °C and then kept for at least 1.0 h. The solid product was collected and rinsed with 80 g of NMP/water (1:3 volume ratio) and then 60 g of water. The product was dried under vacuum at the température below 50 °C to give
II as a pale yellow powder (19 -20 g, purity > 99.0 A% and 88.4 wt%, containing 5.4 wt% NMP). The yield is about 93-98%.
Notes: The original procedure used for the hydrolysis of I was carried out with aq. NaOH (2.5 eq) in MeOH/THF at 60 °C. Although it has been applied to the préparation of II on several hundred grams scale, one disadvantage of this method is the formation of 5-MeO pyrimidine during hydrolysis (ca. 0.4 A%), which is extremely difficult to remove in the subséquent steps. In addition, careful control has to be exerted during crystallization. Otherwise, a thick slurry might form during acidification with HOAc. The use of NMP as solvent could overcome ail aforementioned issues and give the product with desired purity. *0
-2816325
Alternative Process
To a reactor was charged I (71 g), isopropanol (332 g), aqueous NaOH (22 g, 45 wt%) and water (140 g) at ambient température. The mixture was heated to reflux (80 °C) and stirred for at least 3 hrs until the reaction completed. The batch was cooled to 70 °C and charged a suspension of charcoal (3.7 g) in isopropanol (31 g). The mixture was stirred at the same température for over 10 min and filtered. The residue was rinsed with isopropanol (154 g). Water (40 g) was charged to the filtrate at at 70 - 80 °C, followed by slow addition of 36% HCl solution (20 g) to reach pH 5- 6. The batch was stirred for over 30 min at 70 °C, then cooled to 20 °C over 1 hr and kept for at least 1.0 h. The solid product was collected and rinsed with 407 g of isopropanol/water (229 g IPA, 178 g H2O). The product was dried under vacuum at 80 °C for over 5 hrs to give II as a white powder (61 g, 95% yield).
Notes on Steps 5 to 8 below:
A concise and scalable 4-step process for the préparation of the benzimidazole intermediate V was developed. The first step was the préparation of 4-chloro-2-(methyl)aminonitrobenzene starting from 2,4-dichloronitrobenzene using aqueous methyl amine in DMSO at 65 °C. Then, a ligandless Heck reaction with n-butyl acrylate in the presence of Pd(OAc)2, ’P^NEt, LiCl, and DMAc at 110 °C was discovered.
Step 5: SNAr reaction of (5-chloro-2-nitrophenyl)-methylamiiie
2M MeNH2/THF
NEt3, DMSO, 65°C
90%
To a solution of (5-chloro-2-nitrophenyl)-methylamine (40 g, 208.3 mmol, 1 equiv) in DMSO (160 mL) was added 40% MeNH2 solution in water (100 mL, 1145. 6 mmol, 5.5 eq) slowly keeping the température below 35 °C. The reaction was stirred at r.t. until the complété consumption of the starting material (>10 h). Water (400 mL) was added to the resulting orange slurry and stirred at r.t. for additional 2 h. The solid was filtered, rinsed γ</ with water (200 mL) and dried under reduced pressure at 40 °C. (5-chloro-2-nitrophenyl)methylamine (36.2 g, 93% yield, 94 A% purity) was isolated as a solid.
Step 6: Ileck Reaction of (5-chloro-2-nitrophenyl)-methylamine î
i j i
Pd(OAc)2 (0.5%)
Pr^NEt (1.2 eq)
LiCi (1 eq) DMAc (5 vol), 110 °C, 7-22 h
To a mixture of 4-chloro-2-methylaminonitrobenzene (50.0 g, 268.0 mmol, 1.0 eq), Pd(OAc)2 (0.30 g, 1.3 mmol, 0.005 eq) and LiCl (11.4 g 268.0 mmol, 1.0 eq) in DMAc (250 mL) was added 'Pr2NEt (56 mL, 321.5 mmol, 1.2 eq) followed by n-butyl acrylate (40 mL, 281.4 mmol, 1.05 eq) under nitrogen. The reaction mixture was stirred at 110 °C for 12 h, then cooled to 50 °C. 1-methylimidazole (10.6 mL, 134.0 mmol, 0.5 eq) was added and the mixture was stirred for 30 min before filtering and adding water (250 mL). The resulting mixture was cooled to r.t. over 1 h. The resulting solid was filtered and washed with water and dried to yield n-butyl 3-methylamino-4-nitrocînnamate (71.8 g, 96 %, 99.2 A% purity).
L
Step 7: Réduction of n-butyl (3-methylamino-4-nitro)-cinnamate [ i
[
H 2 (4 bar) O
MeHN. OnBu Raney Ni MeHN. OBu
J M Toluene-MeOH h2n^ M
O2N 20- 25 °C III
To areactor was charged n-butyl 3-methylamino-4-nitrocinnamate (70.0 g, mmol, 1.0 eq) , Raney Ni (4.9 g, ~20wt% H2O), charcoal “Norit SX Ultra” (3.5 g), toluene (476 mL) and
-3016325
MeOH (224 mL). The reactor was charged with hydrogen (4 bar) and the mixture was stirred at 20- 25 °C for about 2 hrs until the reaction was completed. The reaction mixture was filtered and rinsed the filter residue with toluene (70 mL). To the combined filtrâtes were added “Norit SX Ultra” charcoal (3.5 g). The mixture was stirred at 50 °C for 1.0 hr and filtered. The filtrate was concentrated under reduced pressure to remove solvents to 50% of the original volume. The remained content was heated to 70 °C and charged slowly methyl cyclohexane (335 mL) at the same température. The mixture was cooled to about 30 - 40 °C and seeded with III seed crystals, then slowly cooled the suspension to - - 10 °C. The solid was filtered and rinsed with methyl cyclohexane in three portions (3 x 46 mL). The wet cake was dried in vacuo at 40 °C to give III (53.3 g, 215 mmol, 86%).
Step 8: Préparation of benzimidazole V
DCC
HCl, 70-80 ’C λ-BuOH, T0-80 ‘C
V
To reactor-1 was charged III (35 g, 140.95 mmol) in toluene (140 g). The mixture was heated to 50 °C to obtain a clear solution. To a second reactor was charged IV (36.4 g, 169.10 mmol) and toluene (300 g), followed by addition of a solution of dicyclohexyl carbodimîde (11.6 g, in 50% toluene, 28.11 mmol) at 0 - 10 °C- The mixture was stirred at the same température for 15 min, then charged parallelly with the content of reactor-1 and the solution of dicyclohexyl carbodimîde (52.4 g, in 50% toluene, 126.98 mmol) within 1 hr while maintaining the batch température at 0 - 10 °C. The mixture was agitated at the same température for 3 hrs, and warmed to 25 °C for another 1 hr. Once III was consumed, toluene (-300 mL) was distilled off under reduced pressure at 70 - 80 °C. «-Butanol (200 g) was added, followed by 3 M HCl solution in n-butanol (188 g) while maintaining the température at 70 - 80 °C (Gas évolution, productprécipitâtes). After stirring for over 30 min. at 70 - 80 °C, the mixture was cooled to 20 - 30 °C over 1 hr. The precipitate was ai/
-3116325 filtered and washed with acetone (172 g) and toluene (88 g). The wet cake was dried in vacuo at ~60 °C to give V toluene solvaté as off white solid (60 - 72 g, 85 - 95% yield). Compound V could be used directly for the next step or basified prior to next step to obtain the free base compound VI used in the next step.
Step 9. Synthesis of (E)-Butyl 3-(2-(l-(2-(5-Bromopyrimidin-2-yl)-3-cyclopentyI-lhydroxy-l//-indole-6-carboxamido)cyclobutvl)-l-methyl-l/7-benzo|d|iniidazol-6yl)acryiate VII
5) MeOH/H2O
Notes:
The conversion of the acid into acid chloride was achieved using inexpensive thionyl chloride in the presence of catalytic amount of NMP or DMF. An efficient crystallization was developed for the isolation of the desired product in high yield and purity.
Procedure (using free base VI):
To the suspension of 2-(5-bromopyrimidin-2-yl)-3-cyclopentyl-1-methyl-17Z-indole-6carboxylic acid II (see Step 4) (33.36 g, 90.0 wt %, containing -0.2 equiv of NMP from previous step,75.00mmol) in THF (133.4 g) was added thionyl chloride (10.71 g). The mixture was stirred at 25±5 °C for at least 1 h. After the conversion was completed as determined by HPLC (as derivative of diethylamine), the mixture was cooled to 10±5 °C and N,N-diisopropylethylamine (378.77 g, 300 mmol) below 25 °C. A solution of (E)-butyl
3-(2-(l-aminocyclobutyl)-l-methyl-l/f-benzo[<7]imidazol-6-yl)acrylate VI (25.86 g, 97.8 Wt%, 77.25 mmol) dissolved in THF (106.7 g) was added at a rate to maintain the température of the content < 25 °C. The mixture was stirred at 25±5 °C for at least 30 min for completion of the amide formation. The mixture was distilled at normal pressure to remove ca. 197 mL (171.5 g) of volatiles (Note: the distillation can also be done under reduced pressure). The batch was adjusted to 40±5 °C, and MeOH (118.6 g) was added. Water (15.0 g) was added and the mixture was stirred at 40±5 °C until crystallization occurred (typîcally in 30 min), and held for another 1 h. Water (90 g) was charged at 40±5 °C over 1 h, and the batch was cooled to 25±5 °C in 0.5 h, and held for at least 1 h. The solid was filtered, rinsed with a mixture of MeOH (39.5 g), water (100 g), and dried in vacuo (< 200 Torr) at 50±5 °C to give (E)-butyl 3-(2-(1-(2-(5-bromopyrimidin-2-yl)-3cyclopentyl-1 -methyl-1 77-indole-6-carboxamido)cyclobutyl)-1 -methyl-1 Hbcnzo[Y|imKiazol-6-yl)acrylate VII (51.82 g, 96.6 % yield) with a HPLC purity of 98.0 A% (240 nm) and 99.0 Wt%.
Alternative Process (using compound V from Step 8)
To reactor 1 was charged 2-(5-bromopyrimidin-2-yI)-3-cyclopentyl-l-methyl- l//-indole-6carboxylic acid II (33.6 g), toluene (214 g) and N-methylpyrrolidone (1.37 g). The mixture was heated to 40 °C, then added a solution of thionyl chloride (13 g) in toluene (17 g). The mixture was stirred at 40 °C for at least 0.5 h and cooled to 30 °C. To a second reactor was charged with compound V (the bis-HCl sait toluene solvaté from Step 8) (39.4 g), toluene (206 g) and N,N-diisopropylethylamine (70.8 g) at 25 °C. The content of reactor 1 was transferred to reactor 2 at 30 °C and rinsed with toluene (50 g). The mixture was stirred at 30 °C for another 0.5 h, then charged with isopropanol (84 g) and water (108 g) while maintained the température at 25 °C. After stirring for 10 min, remove the aqueous phase after phase cutting. To the organic phase was charged isopropanol (43 g), water (54 g) and stirred for 10 min. The aqueous phase was removed after phase cutting. The mixture was distilled under reduced pressure to remove ca. 250 mL of volatiles, followed by addition of methyl tert-butyl ether (MTBE, 238 g). The batch was was stirred at 65 °C for over 1 hr, then cooled to 20 C over I hr and held for another 1 hr at the same température. The solid was filtered, rinsed with MTBE (95 g), and dried in vacuo at 80 °C to give (E)-butyl 3-(2(l-(2-(5-bromopyrimidin-2-yl)-3-cyclopentyl-l-methyl-17/-indole-6carboxamido)cyclobutyl)-l-methyl-177-benzo[(f|imidazol-6-yl)acrylate VII as a beige solid (50 g, 90 % yield). qZ
-3316325
Step 10. Synthesis of (E)-3-(2-(l-(2-(5-Broinopyrimi(lin-2-yl)-3-cycJopentyl-l-niethvllZr-indole-6-carboxamido)cyclobutyI)-l-methyI-lff-benzo[rf]iniidazol-6-yI)acrylic acid (Compound (1))
Notes:
In this process, hydrolysis of (E)-butyl 3-(2-(l-(2-(5-bromopyrimidîn-2-yl)-3-cyclopentyl- l-methyl-lH-indole-6-carboxamido)cycIobutyI)-l-methyl-lH-benzo[d]imidazol-6yl)acrylate was carried out in mixture of THF/MeOH and aq NaOH. Controlled acidification of the corresponding sodium sait with acetic acid is very critical to obtain easy-fïltering crystalline product in high yield and purity.
Procedure:
To the suspension of (E)-butyl 3-(2-(1-(2-(5-bromopyrimidin-2-yl)-3-cyclopentyl-lmethyl-17f-indole-6-carboxamido)cyclobutyl)-1 -methyl-1 //-benzo [c?] imidazol-6yl)acrylate VII (489.0 g, 91.9 Wt%, 633.3 mmol) in THF (1298 g) and MeOH (387 g) was added 50% NaOH (82.7 g, 949.9 mmol), followed byrinse with water (978 g). The mixture was stirred between 65-68 C for about 1 h for complété hydrolysis. The resulting solution was cooled to 35 C, and filtered through an in-line filter (0.5 micron), and rinsed with a pre-mixed solution of water (978 g) and MeOH (387 g). The solution was heated to 60 ±4 °C, and acetic acid (41.4 g, 689 mmol) was added over 1 h while the mixture was well agitated. The resulting suspension was stirred at 60 ±4 C for 0.5 h. Another portion of acetic acid (41.4 g, 689 mmol) was charged in 0.5 h, and batch was stirred at 60 ±4 C o
for additional 0.5 h. The batch was cooled to 26 ±4 C over 1 h and held for 1 h. The batch was filtered, rinsed with a premixed solution of water (1956 g) and MeOH (773.6 g), dried at 50 °C under vacuum to give (E)-3-(2-(l-(2-(5-bromopyrimidin-2-yl)-3-cyclopentyl-Imethyl-l/f-indole-6-carboxamido)cyclobutyl)-l -methyl- 17/-benzo[J]imidazol-6-yl)acrylic acid (1) (419.0 g, 95 % yield) with > 99.0 A% (240 nm) and 94.1 Wt% by HPLC.
-3416325
Step IL Formation of Compound (1) Sodium Sait (Type A)
Step 11
NaOH (1.0 eq)
THF-MEK-H2O
95% (1) Na sait Type A
To a reactor were charged Compound (1) (150 g, mmol), THF (492 mL), H2O (51 mL) and 45% aqueous NaOH solution (20.4 g, mmol). The mixture was stirred for >1 hr at -25 °C to form a clear solution (pH = 9-11). To the solution was charged a suspension of Charcoal (1.5 g) and H2O (27 mL). The mixture was stirred at -35 °C for >30 min and filtered. The filter was rinsed with THF (108 mL) and H2O (21 mL). The filtrate was heated to 50 °C and charged with methyl ethylketone (MEK) (300 mL). The mixture was seeded with Compound (1) sodium sait MEK solvaté (Type A) seeds (0.5 g) and stirred for another 1 hr at 50 °C. To the mixture was charged additional MEK (600 mL). The résultant mixture was stirred for another 1 hr at 50 °C and then cooled to 25 °C. The precîpitate was filtered and rinsed with MEK twice (2 x 300 mL). The wet cake was dried in vacuum at 80 °C to give Compound (1) sodium sait (Type A) (145.6 g, 94%).
The Compound (1) sodium sait (Type A) MEK solvaté seeds used in the above process step can be manufactured by the above process except without using seeds and without drying of the solvaté.
-3516325
Notes Regarding Crystallization Step 11
Process Optimization for Producing Higher Bulk Densîty Material
Observation of lab experiments showed that the seeding température should be reduced from 60 °C to 50 °C to prevent the dissolution of seed crystals. The crystallization kinetics in the THF/MEK/H2O System was found to be slow, and oil / émulsion could be observed when anti-solvent MEK was added too fast after seeding. Thus experiments were performed to optimize the MEK addition time and aging time to minimize oiling. This improved process produced agglomerated granular crystals consistently that resulted in the desired high bulk density.
Optimization of anti-solvent addition and aging time
An experiment was designed to optimize the aging time following the MEK anti-solvent addition at 50 °C. The data indicated that ail solids crystallized out of solution within 3 hours of aging. Following aging, the slurry was cooled linearly over 2 hours to 20 °C. The extended aging time did not significantly improve yield losses in the mother liquor. The crystallization resulted în a 92.4% yield.
Immediately after the completion of the MEK addition, a milky oily solution was observed along with a large amount of crystals. The oily solution dissipated within one hour. A separate experiment determined that a slower addition rate of MEK can avoid the formation of oil.
The XRPD pattern on the wet cake confirmed the MEK solvated phase.
Another experiment was carried out to adapt the process for the slow crystallization kinetics observed in the current crystallization System. A 1/2 hour aging tîme was included after seeding and the MEK anti-solvent addition time was increased from 2 to 4 hours at 50
O,
-3616325
Ali solids were found to hâve crystallized out of solution within 2 hours of aging. Following aging, the slurry was cooled linearly over 2 hours to 20 °C and held overnight. This did not improve on the mother liquor losses significantly.
In conclusion, the slurry at the end of the MEK addition was found to produce clear mother lîquors without an oil phase; whereas previously in the 2-hour MEK addition, a milky oily mother liquor was observed. The recommendation is for a 4-hour anti-solvent addition to prevent the oiling.
Drying Time Study
A study was conducted to détermine the required drying time at 80 °C to meet the ICH limits of residual solvents of MEK and THF. The results showed that drying for a minimum of 5 hours is required to meet the ICH limît on THF.
Effects of Water Content on Yield and Crystallization
The effect of water content on crystallization was evaluated. The water content was varied from the 5.6% (w/w) level specified in the existing procedure. The study was done using 50% more and 50% less water in the crystallization. The data indicated that 5.6% water content is near optimum for good yield and operability.
IV. Formulations of Compound (2)
Examples of pharmaceutical formulations containing Compound (2) include the tablet formulations described below.
-3716325
Exampie 5: Solid Oral Formulation # 1
The composition of the solid oral formulation:
Monograph Functionality % w/w
Compound (2) Na sait Active 34.45
Meglumine USP / Ph. Eur. Basifier 7.00
Sodium Lauryl Sulfate NF / Ph. Eur. Surfactant 3.50
Polyethylene Glycol 6000 NF / Ph. Eur. Solubilizer/ Binder 10.33
Mannitol USP / Ph. Eur. Filler 43.72
Colloïdal Silicon Dioxide NF/ Ph. Eur. Glîdant 0.75
Magnésium Stéarate NF/ Ph. Eur. Lubricant 0.75
Two spécifie solid oral drug product formulations were prepared according to the above general Formulation # 1, a 50 mg product and a 200 mg product.
Ingrédient Function 200 mg 50 mg
mg/tablet mg/tablet
Compound (2) Na sait1 Drug Substance 206.71 51.71
Meglumine Basifier 42.0 10.5
Sodium Lauryl Sulfate Surfactant 21.0 5.3
Polyethylene Glycol 6000 Solubilizer Binder 62.0 15.5
Mannitol (powdered) Filler 262.3 65.6
Purified Water2 Granulating agent q.s. q.s.
Colloïdal Silicon Dioxide Glidant 3.0 0.8
Magnésium Stéarate3 Lubricant 3.0 0.8
Total 600.0 150.0
Ir 206.7 mg and 51.7 mg Compound (2) Na sait (sodium sait) is équivalent to 200 mg and 50 mg of the active moiety, Compound (2) (free acid), respectively.
Purified water is used as a granulating agent; it does not appear in the final product. 1 Vegetable origin
Example 6: Solid Oral Formulation # 2
The composition of the solid oral formulation:
Monograph Functionality % w/w
Compound (2) Na sait Active 40.00
Arginine USP / Ph. Eur. Basifier 8.00
Sodium Lauryl Sulfate NF / Ph. Eur. Surfactant 4.00
-3816325
Polyethylene Glycol 8000 NF / Ph. Eur. Solubilizer/ Binder 12.00
Mannitol USP / Ph. Eur. Filler 35.00
Colloïdal Sîlicon Dioxide NF/Ph. Eur. Glidanf 0.50
Magnésium Stéarate NF/ Ph. Eur. Lubricant 0.50
Two spécifie solid oral drug product formulations were prepared according to the above general Formulation # 1, a 200 mg product and a 400 mg product.
Ingrédient Function 200 mg 400 mg
mg/tablet mg/tablet
Compound (2) Na sait1 Drug Substance 206.7’ 413.41
Arginine Basifier 41.4 82.7
Sodium Lauryl Sulfate Surfactant 20.7 41.3
Polyethylene Glycol 8000 Solubilizer/ Binder 62.0 124.0
Mannitol (powdered) Filler 180.9 361.8
Purified Water2 Granulating agent q.s. q.s.
Colloïdal Sîlicon Dioxide Glidant 2.6 5.2
Magnésium Stéarate Lubricant 2.6 5.2
Total 516.8 1033.6
206.7 mg and 413.4 mg Compound (2) Na sait (sodium sait) is équivalent to
200 mg and 400 mg of the active moiety, Compound (2) (ffee acid), respectively. Purified water is used as a granulating agent; it does not appear in the final product.
3 Vegetable origin
Préparation of Formulations 1-2
The drug substance along with the intragranular excipients including the basifier, surfactant, solubilizer/binder, filler are mixed in a dry state in a high shear granulator prior to addition of water. The drug substance and the excipients may be screened prior to milling to remove large agglomérâtes if necessary. After mixing is complété, the mixture is granulated using purified water as a granulating agent in the high shear granulator till a suitable end point is achieved. The wet granules are removed and dried at appropriate drying températures either in a tray dryer or a fluid bed dryer. The dried granules are mîlled by passing through a high speed mill, such as a Comill. Milled granules are then blended with the extragranular excipients, glidant and lubricant and then tableted in a tablet press.
V. Clinical Results
For the clinical trials described below, the Compound (1) drug product was administered as a softgel capsule lipîd-based formulation containing Compound (1) sodium sait.
Compound (2) drug product was administered as a tablet formulation containing Compound (2) sodium sait.
Examplc 7 ~ Clinical Study with Treatment-Naïve Patients
Safety and antiviral activity of interferon-sparing treatment with the protease inhibitor Compound (1) sodium sait, the HCV polymerase inhibitor Compound (2) sodium sait and ribavirin in treatment-naive patients with chronic hepatitis C génotype-1 infection.
Background: Compound (1) and Compound (2) are potent and spécifie inhibitors of the HCV NS3/4A protease and NS5B RNA-dependent RNA polymerase, respectively. An interferon-free combination of both antîvirals with ribavirin (RBV) to eradicate HCV infection would create a major paradigm shift in HCV treatment.
Methods: In this randomized open-label trial, 32 treatment-naïve HCV génotype-1 patients were treated over 4 weeks with Compound (2) sodium sait given at 400mg or 600mg three times a day (TID), Compound (1) sodium sait given at 120mg daily (QD) and RBV (weight based dosage at lOOOmg or 1200mg daily in two doses). Plasma HCV RNA virus 25 load (VL) was measured by Roche COBAS® TaqMan® assay with a lower limît of quantification of 25 IU/ml.
Results: At baseline, mean âge was 51 + 11 years, mean BMI 23.8 + 3.5 kg/m2, mean VL 6.48 LOGiq. Ail patients had a rapid and sharp VL décliné during the first two days, followed by a slower second phase décliné in ail except 2 patients. One patient experienced VL breakthrough (increase by >1 LOGio from nadir during treatment) and one other μ_Γ '
-4016325 experienced a 0.7 LOGio VL increase. Both were in the lower dose group (400 mg TID Compound (2)) and were génotype la patients (confirmed by NS5B sequencing) with high baseline VL. On day 29, all patients were switched per protocol to treatment with Compound (l) sodium sait, and pegylated interferon(PegIFN )/RBV. Virological response 5 rates (VL < 25 lU/ml) after l, 2,3 and 4 weeks of oral treatment are shown in the table.
Table: Proportion of patients with viral load <25 lü/ml
Day 8
Day 15
Day 22 Day 29
400mg TID Cmpd (2) + Cmpd (1) + RBV 600mg TID Cmpd (2) + Cmpd (1) + RBV
7/15 10/15 11/15
14/17 17/17 17/17
Below is a more detailed présentation of the same data showing virologie response by dose group and by subgenotype (where BLQ = VL < 25 lU/ml) where a génotype 1 was more precisely characterized as génotype 6e in the 600 mg TID dose group:
Day 8 BLQ Day 15 BLQ Day 22 BLQ Day 29 BLQ
4 7 10 11
400mg TID 27% 47% 67% 73%
N=15 la: 2/10 la: 5/10 la: 6/10 la: 6/10
Ib: 2/5 lb: 2/5 lb: 4/5 lb: 5/5
3 14 17 17
600mg TID 18% 82% 100% 100%
N=17 la: 2/8 la: 8/8 la: 8/8 la: 8/8
Ib: 1/8 lb: 5/8 lb: 8/8 lb: 8/8
6e: 0/1 6e: 1/1 6e: 1/1 6e: 1/1
-4116325
Total N=32 7 22% la: 4/18 lb: 3/13 6e: 0/1 21 66% la: 13/18 lb: 7/13 6e: 1/1 27 84% la: 14/18 lb:12/13 6e: 1/1 28 88% la: 14/18 lb: 13/13 6e: 1/1
The results demonstrate robust anti viral activity at 400 mg TID (overall 73% RVR; 100 % for GTlb and 60% for GTla), and excellent antiviral activity at 600 mg TID (100 % RVR for GTla and lb). The change in VL over time for the 400 mg TID dose group is graphically depicted in FIG 1, and the change in VL over time for the 600 mg TID dose group is graphically depicted in FIG 2.
At the higher dose level, there was no différence between génotype (GT) 1 a and lb, while
GT1 a patients at 400mg TID had a lower response rate than those with GTlb, and one
GTla patient in the 400 mgTID dose group demonstrated a viral rebound (increaseof> 1logio from nadir) during the 28-day treatment. The PegIFN sparing treatment was well tolerated. The most common adverse events (AEs) were mostly mild gastro-intestinal effects (diarrhea, nausea, vomiting), rashes or photosensitivity. There were no severe AEs, <
SAEs or treatment discontinuations within the 4-week study period. Laboratory parametersj did not indicate any relevant changes from baseline, except for a continuous drop in ALT<
in ail patients, a decrease of haemoglobin (médian -2.5 and -3.6 g/dL) and increase of;
unconjugated bilirubin (médian +9.8 and +11.5 umol/1).i i
[
I
In addition, based on feedback from investigators, the combination therapy was found asi having improved tolerability as compared to other HCV treatments. A questionaire to compare tolerability of the triple combination therapy of the présent invention vs. other’
HCV regimens was sent to ail 14 investigators. Tolerability was rated on a scale from -5 to +5 (with “0” indicating comparable tolerability,“-5” much worse tolerability and “+5” much better tolerability).|
-4216325
The results are shown in the table below:
Trial Description vs. Other HCV Treatment n médian min max
Compound (1) vs. SOC (PegIFN/RBV) 8 +3.5 +2 +5
Ή Compound (2) + vs. Compound (1) + SOC 8 +3.5 0 +5
vs. Compound (2) + SOC 5 +3 0 +5
RBV vs. Telaprevir + SOC 7 +4 +3 +5
vs. other PegIFN-sparing regimen 2 +2.5 0 +5
Conclusions: PegIFN sparing treatment with the the NS3/4A inhibitor Compound (1), NS5B inhibitor Compound (2), and RBV demonstrated strong early antiviral activity against HCV génotype 1 with good safety and tolerability. A phase Ilb trial testing different dose regimens of this combination, with longer durations, is planned to evaluate sustained virologie response rates.
Only one GTla patient in the 400 mg TID dose group demonstrated a rebound in viral load during the 28-day treatment period. Sequencing of the viral nucleic acid from the rebound sample, showed that the virus contained sequence changes, relative to the baseline pre-treatment sample, in both the NS3 protease and NS5B polymerase régions. The nucléotide changes encoded for amino acid subsutitions RI 55K in NS3 and P495L in NS5B and represent virus that is dually résistant to compound (1) and compound (2). The low frequency of virologie rebound (only one out of 15 patients in the 400 mg TID dose group, and no patients out of 17 in the 600 mg TID dose group) during treatment is a surprising and unexpected resuit based on earlier assessment of compound (1) as a monotherapy in a PegIFN sparing regimen for 14-days where the vast majority of both la and lb patients demonstrated virologie rebound during the treatment period (see the monotherapy data for Compound (1) presented in U.S. Application Publication 2010/0068182).
-4316325
Defmitely surprising results include the fact that the PegIFN sparing regimen comprised of compound (1), compound (2) and ribavirin effectively suppresses the emergence of drug résistant variants that hâve been commonly observed in clinical trials of compound 1 or other protease inhibitors in monotherapy. The low frequency of virologie failure (1/15) in 5 the 400 mg TID dose group and the lack of any virology failures (0/17) in the 600 mg TID dose group suggest that the combination of compounds 1 and 2 hâve the potential to achieve significantly improved efficacies in a novel and more tolérable therapeutic regimen.

Claims (2)

  1. (a) a compound of the following formula (1 ) or a pharmaceutically acceptable sait thereof:
    (b) a compound of the following formula (2) or a pharmaceutically acceptable sait thereof:
    -4516325
    -i
    In the manufacture of a médicament for treating hepatitis C viral (HCV) infection or alleviating one or more symptoms thereof in a patient
    2. The use according to claim I, wherein the HCV infection is génotype 1.
    3. The use according to claim 1 or 2, wherein said patient is a treatment-naive patient.
    4. The use according to any of the preceding claims, wherein said patient is nonresponsive to a combination therapy using ribavirin and an interferon alpha.
    5. The use according to any of the preceding claims, wherein the HCV-RNA levels of said patient are reduced to a level below 25 International Unîts (IU) per ml of sérum or plasma as a resuit of the treatment.
    6. The use according to any of the preceding claims, wherein compound (1) or a pharmaceutically acceptable sait thereof is administered at a dosage between about 40 mg per day and about 480 mg per day.
    7. The use according to any of the preceding claims, wherein compound ( 1 ) is administered in the form of its sodium sait.
    8. The use according to any of the preceding claims, wherein compound (2) or a pharmaceutically acceptable sait thereof is administered at a dosage between about 800 mg per day and about 2400 mg per day.
    9. The use according to any of the preceding claims, wherein compound (2) is administered in the form of its sodium sait.
    10. The use according to any of the preceding claims, wherein said ribavirin is administered at a dosage between about 400 mg/day and about 1200 mg/day.
    -4616325
    11. The use according to any of the preceding claims, wherein the therapeutic combination administered is a triple combination therapy including administration of Compound (1) or a pharmaceutically acceptable sait thereof, Compound (2) or a pharmaceutically acceptable sait thereof and ribavirin.
    12. The use according to any of claims 1 to 12, wherein the therapeutic combination administered is a double combination therapy including administration of Compound (1) or a pharmaceutically acceptable sait thereof and Compound (2) or a pharmaceutically acceptable sait thereof without the administration of ribavirin.
    13. A packaged pharmaceutical composition comprising a packaging containing:
    (a) one or more doses ofthe following Compound (1) or a pharmaceutically acceptable sait thereof:
    or (b) one or more doses of the following Compound (2) or a pharmaceutically acceptable sait thereof:
    -4716325 ί
    i and written instructions directing the co-administration of Compound (1), or a pharmaceutically acceptable sait thereof, and Compound (2), or a pharmaceutically acceptable sait thereof, and optionally ribavirin, for the treatment of HCV infection.
    10 14. A kit for the treatment of HCV infection comprising:
    (a) one or more doses of the following Compound (1) or a pharmaceutically acceptable sait thereof:
    -481 and (b) one or more doses of the following Compound (2) or a pharmaceutically acceptable sait thereof:
    and written instructions directing the co-administration of Compound (1), or a pharmaceutically acceptable sait thereof, and Compound (2), or a pharmaceutically 10 acceptable sait thereof, and optionally ribavirin, for the treatment of HCV infection.
    15. A compound of the following formula (1) or a pharmaceutically acceptable sait thereof:
    -4915 cr° <*>
    for use together with a compound of the following formula (2) or a pharmaceutically acceptable sait thereof:
  2. (2), and optionally ribavirin, in a method for the treatment of HCV infection.
    16. A compound of the following formula (2) or a pharmaceutically acceptable sait thereof:
    for use together with a compound of the following formula (1 ) or a pharmaceutically acceptable sait thereof:
    -501 i
    -5116325 ε03α æ/eo ιε^α si^a ol/bq s^a
    HCVRNAfILVmL)
    100000000
    Da/2 Da/4 Da/8 Da/10 Da/15 Da/22 Da/29 Da/31 Da/36 Da/43 Day57
    W5 FEV 201&
    -5416325
    Planche de l'abrégé
    HCVFMA(lUmQ
OA1201300063 2010-09-30 2011-09-23 Combination therapy for treating HCV infection. OA16325A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US61/388,253 2010-09-30

Publications (1)

Publication Number Publication Date
OA16325A true OA16325A (en) 2015-05-11

Family

ID=

Similar Documents

Publication Publication Date Title
US20120135949A1 (en) Combination therapy for treating hcv infection
AU2010333656B2 (en) HCV combination therapy
TWI389688B (en) Forms and formulations of vx-950 and production process and use thereof
ES2276045T3 (en) PHARMACEUTICAL COMPOSITIONS FOR HEPATITIS C VIRAL PROTEASE INHIBITORS.
CN1301158A (en) Compositions for the treatment of HIV and other viral infections
AU2013358897A1 (en) Pharmaceutical compositions
EP1104297B1 (en) Compositions for the treatment of chronic lymphocytic leukemia
TW201526899A (en) Pharmaceutical compositions
CN102438588A (en) Methods of treatment using combination therapy
TW200811180A (en) Novel pyridazine compound and use thereof
WO2011150162A1 (en) Methods for treating viral conditions
WO2010151487A1 (en) Combination of telaprevir with 5- ({6- [2,4-bis (trifluoromethyl) phenyl] pyridazin-3 -yl)methyl) -2- (2 -fluorophenyl) -5h- imidazo [4, 5-c]pyridine for the treatment of hcv
KR20050085681A (en) Use of a combination containing a non-nucleoside reverse transcriptase inhibitor(nnrti) with an inhibitor of cytochrome p450, such as protease inhibitors
FR3056108B1 (en) USE OF HARRINGTONINS IN THE TREATMENT OF BREAST CANCER, IN PARTICULAR TRIPLE-NEGATIVE
ZA200201466B (en) Pharmaceutical agent comprising a benzamide derivative as active ingredient.
OA16325A (en) Combination therapy for treating HCV infection.
US20170333460A1 (en) Long acting pharmaceutical compositions for hepatitis c
WO2006041120A1 (en) Pharmaceutical composition
KR20150046083A (en) Combination of a macrocyclic protease inhibitor of hcv, a non­nucleoside hcv inhibitor and ritonavir
WO2001098300A1 (en) Medicines for the prevention and treatment of neurodegenerative diseases
KR20150056853A (en) Use of a tetrasubstituted pyrazolo[4,3-d]pyrimidine compound for treating diabetic nephropathy
WO2013147750A1 (en) Oral combination therapy for treating hcv infection in specific patient sub-population
US20130288957A1 (en) Oral combination therapy for treating hcv infection in specific patient subgenotype populations
WO2013147749A1 (en) Oral combination therapy for treating hcv infection in specific patient subgenotype populations
JP2017514834A (en) Combination therapy for treating HCV infection