SUBSTITUTED BENZAMIDES AND METHODS OF USE THEREOF

- GENENTECH, INC.

The invention provides compounds having the general formula (I): and pharmaceutically acceptable salts thereof, wherein the variables RA, RAA, n, ring A, X2, L, m, R1, R2, R3, R4, R5, and RN have the meaning as described herein, and compositions containing such compounds and methods for using such compounds and compositions.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
PRIORITY APPLICATION

This application is a divisional of U.S. patent application Ser. No. 15/779,065 filed 24 May 2018, which is a 35 U.S.C. § 371 application of International Application Serial No. PCT/US2016/063369, filed 22 Nov. 2016, which claims priority to U.S. Provisional Patent Application No. 62/260,113, filed 25 Nov. 2015. The entire contents of these patent applications are hereby incorporated by reference herein.

The present invention relates to organic compounds useful for therapy and/or prophylaxis in a mammal, and in particular to inhibitors of sodium channel (e.g., NAV1.7) that are useful for treating sodium channel-mediated diseases or conditions, such as pain, as well as other diseases and conditions associated with the mediation of sodium channels.

Voltage-gated sodium channels, transmembrane proteins that initiate action potentials in nerve, muscle and other electrically excitable cells, are a necessary component of normal sensation, emotions, thoughts and movements (Catterall, W. A., Nature (2001), Vol. 409, pp. 988-990). These channels consist of a highly processed alpha subunit that is associated with auxiliary beta subunits. The pore-forming alpha subunit is sufficient for channel function, but the kinetics and voltage dependence of channel gating are in part modified by the beta subunits (Goldin et al., Neuron (2000), Vol. 28, pp. 365-368). Electrophysiological recording, biochemical purification, and molecular cloning have identified ten different sodium channel alpha subunits and four beta subunits (Yu, F. H., et al., Sci. STKE (2004), 253; and Yu, F. H., et al., Neurosci. (2003), 20:7577-85).

The hallmarks of sodium channels include rapid activation and inactivation when the voltage across the plasma membrane of an excitable cell is depolarized (voltage-dependent gating), and efficient and selective conduction of sodium ions through conducting pores intrinsic to the structure of the protein (Sato, C., et al., Nature (2001), 409:1047-1051). At negative or hyperpolarized membrane potentials, sodium channels are closed. Following membrane depolarization, sodium channels open rapidly and then inactivate. Channels only conduct currents in the open state and, once inactivated, have to return to the resting state, favoured by membrane hyperpolarization, before they can reopen. Different sodium channel subtypes vary in the voltage range over which they activate and inactivate as well as their activation and inactivation kinetics.

The sodium channel family of proteins has been extensively studied and shown to be involved in a number of vital body functions. Research in this area has identified variants of the alpha subunits that result in major changes in channel function and activities, which can ultimately lead to major pathophysiological conditions. The members of this family of proteins are denoted NaV1.x, where x=1 to 9. NaV1.1 and NaV1.2 are highly expressed in the brain (Raymond, C. K., et al., J. Biol. Chem. (2004), 279(44):46234-41) and are vital to normal brain function. Some loss of function mutations in NaV1.1 in humans result in epilepsy, apparently because many of these channels are expressed in inhibitory neurons (Yu, F. H., et al., Nat Neurosci (2006), 9 (9), 1142-9). Thus, block of NaV1.1 in the CNS may be counter-productive because it can produce hyperexcitability. However, NaV1.1 is also expressed in the peripheral nervous system and block may afford analgesic activity.

NaV1.3 is expressed primarily in the fetal central nervous system. It is expressed at very low levels or not at all in the peripheral nervous system, but expression is upregulated in the dorsal horn sensory neurons of rats after nervous system injury (Hains, B. D., et al., J. Neurosci. (2003), 23(26):8881-92). Thus, it is an inducible target for treatment of pain following nerve injury.

NaV1.4 is expressed primarily in skeletal muscle (Raymond, C. K., et al., op. cit.). Mutations in this gene have been shown to have profound effects on muscle function including paralysis, (Tamaoka A., Intern. Med. (2003), (9):769-70).

NaV1.5, is expressed mainly in cardiac myocytes (Raymond, C. K., et al., op. cit.), including atria, ventricles, the sino-atrial node, atrio-ventricular node and cardiac Purkinje fibers. The rapid upstroke of the cardiac action potential and the rapid impulse conduction through cardiac tissue is due to the opening of NaV1.5. Abnormalities in the function of NaV1.5 can result in the genesis of a variety of cardiac arrhythmias. Mutations in human NaV1.5 result in multiple arrhythmic syndromes, including, for example, long QT3 (LQT3), Brugada syndrome (BS), an inherited cardiac conduction defect, sudden unexpected nocturnal death syndrome (SUNDS) and sudden infant death syndrome (SIDS) (Liu, H., et al., Am. J. Pharmacogenomics (2003), 3(3):173-9). Sodium channel blocker therapy has been used extensively in treating cardiac arrhythmias.

NaV1.6 is a widely distributed voltage-gated sodium channel found throughout the central and peripheral nervous systems. It is expressed at high density in the nodes of Ranvier of myelinated neurons (Caldwell, J. H., et al., Proc. Natl. Acad. Sci. USA (2000), 97(10): 5616-20).

NaV1.7 is a tetrodotoxin-sensitive voltage-gated sodium channel encoded by the gene SCN9A. Human NaV1.7 was first cloned from neuroendocrine cells (Klugbauer, N., et al., 1995 EMBO J., 14 (6): 1084-90.) and rat NaV1.7 was cloned from a pheochromocytoma PC12 cell line (Toledo-Aral, J. J., et al., Proc. Natl. Acad. Sci. USA (1997), 94:1527-1532) and from rat dorsal root ganglia (Sangameswaran, L., et al., (1997), J. Biol. Chem., 272 (23): 14805-9). NaV1.7 is expressed primarily in the peripheral nervous system, especially nocieptors and olfactory neurons and sympathetic neurons. The inhibition, or blocking, of NaV1.7 has been shown to result in analgesic activity. Knockout of NaV1.7 expression in a subset of sensory neurons that are predominantly nociceptive results in resistance to inflammatory pain (Nassar, et al., op. cit.). Likewise, loss of function mutations in humans results in congenital indifference to pain (CIP), in which the individuals are resistant to both inflammatory and neuropathic pain (Cox, J. J., et al., Nature (2006); 444:894-898; Goldberg, Y. P., et al., Clin. Genet. (2007); 71:311-319). Conversely, gain of function mutations in NaV1.7 have been established in two human heritable pain conditions, primary erythromelalgia and familial rectal pain, (Yang, Y., et al., J. Med. Genet. (2004), 41(3):171-4). In addition, a single nucleotide polymorphism (R1150W) that has very subtle effects on the time- and voltage-dependence of channel gating has large effects on pain perception (Estacion, M., et al., 2009. Ann Neurol 66: 862-6; Reimann, F., et al., Proc Natl Acad Sci USA (2010), 107: 5148-53). About 10% of the patients with a variety of pain conditions have the allele conferring greater sensitivity to pain and thus might be more likely to respond to block of NaV1.7. Because NaV1.7 is expressed in both sensory and sympathetic neurons, one might expect that enhanced pain perception would be accompanied by cardiovascular abnormalities such as hypertension, but no correlation has been reported. Thus, both the CIP mutations and SNP analysis suggest that human pain responses are more sensitive to changes in NaV1.7 currents than are perturbations of autonomic function.

NaV1.8 is expressed primarily in sensory ganglia of the peripheral nervous system, such as the dorsal root ganglia (Raymond, C. K., et al., op. cit.). There are no identified human mutations for NaV1.8 that produce altered pain responses. NaV1.8 differs from most neuronal NaV's in that it is insensitive to block by tetrodotoxin. Thus, one can isolate the current carried by this channel with tetrodotoxin. These studies have shown that a substantial portion of total sodium current is NaV1.8 in some dorsal root ganglion neurons (Blair, N. T., et al., J Neurosci (2002), 22: 10277-90). Knock-down of NaV1.8 in rats has been achieved by using antisense DNA or small interfering RNAs and virtually complete reversal of neuropathic pain was achieved in the spinal nerve ligation and chronic constriction injury models (Dong, X. W., et al., Neuroscience (2007),146: 812-21; Lai J., et al. Pain (2002), 95: 143-52). Thus, NaV1.8 is considered a promising target for analgesic agents based upon the limited tissue distribution of this NaV isoform and the analgesic activity produced by knock-down of channel expression.

NaV1.9 is also a tetrodotoxin insensitive, sodium channel expressed primarily in dorsal root ganglia neurons (Dib-Hajj, S. D., et al. (see Dib-Hajj, S. D., et al., Proc. Natl. Acad. Sci. USA (1998), 95(15):8963-8). It is also expressed in enteric neurons, especially the myenteric plexus (Rugiero, F., et al., J Neurosci (2003), 23: 2715-25). The limited tissue distribution of this NaV isoform suggests that it may be a useful target for analgesic agents (Lai, J., et al., op. cit.; Wood, J. N., et al., op. cit.; Chung, J. M., et al., op. cit.). Knock-out of NaV1.9 results in resistance to some forms of inflammatory pain (Amaya, F., et al., J Neurosci (2006), 26: 12852-60; Priest, B. T., et al., Proc Natl Acad Sci USA (2005), 102: 9382-7).

This closely related family of proteins has long been recognized as targets for therapeutic intervention. Sodium channels are targeted by a diverse array of pharmacological agents. These include neurotoxins, antiarrhythmics, anticonvulsants and local anesthetics (England, S., et al., Future Med Chem (2010), 2: 775-90; Termin, A., et al., Annual Reports in Medicinal Chemistry (2008), 43: 43-60). All of the current pharmacological agents that act on sodium channels have receptor sites on the alpha subunits. At least six distinct receptor sites for neurotoxins and one receptor site for local anesthetics and related drugs have been identified (Cestele, S., et al., Biochimie (2000), Vol. 82, pp. 883-892).

The small molecule sodium channel blockers or the local anesthetics and related antiepileptic and antiarrhythmic drugs interact with overlapping receptor sites located in the inner cavity of the pore of the sodium channel (Catterall, W. A., Neuron (2000), 26:13-25). Amino acid residues in the S6 segments from at least three of the four domains contribute to this complex drug receptor site, with the IVS6 segment playing the dominant role. These regions are highly conserved and as such most sodium channel blockers known to date interact with similar potency with all channel subtypes. Nevertheless, it has been possible to produce sodium channel blockers with therapeutic selectivity and a sufficient therapeutic window for the treatment of epilepsy (e.g., lamotrignine, phenytoin and carbamazepine) and certain cardiac arrhythmias (e.g., lignocaine, tocainide and mexiletine). However, the potency and therapeutic index of these blockers is not optimal and have limited the usefulness of these compounds in a variety of therapeutic areas where a sodium channel blocker would be ideally suited.

Sodium channel blockers have been shown to be useful in the treatment of pain, including acute, chronic, inflammatory and/or neuropathic pain (see, e.g., Wood, J. N., et al., J. Neurobiol. (2004), 61(1), 55-71. Preclinical evidence demonstrates that sodium channel blockers can suppress neuronal firing in peripheral and central sensory neurons, and it is via this mechanism that they are considered to be useful for relieving pain. In some instances, abnormal or ectopic firing can original from injured or otherwise sensitized neurons. For example, it has been shown that sodium channels can accumulate in peripheral nerves at sites of axonal injury and may function as generators of ectopic firing (Devor et al., J. Neurosci.(1993), 132: 1976). Changes in sodium channel expression and excitability have also been shown in animal models of inflammatory pain where treatment with proinflammatory materials (CFA, Carrageenan) promoted pain-related behaviors and correlated with increased expression of sodium channel subunits (Gould et al., Brain Res., (1999), 824(2): 296-99; Black et al., Pain (2004), 108(3): 237-47). Alterations in either the level of expression or distribution of sodium channels, therefore, may have a major influence on neuronal excitability and pain-related behaviors.

Controlled infusions of lidocaine, a known sodium channel blocker, indicate that the drug is efficacious against neuropathic pain, but has a narrow therapeutic index. Likewise, the orally available local anesthetic, mexiletine, has dose-limiting side effects (Wallace, M. S., et al., Reg. Anesth. Pain Med. (2000), 25: 459-67). A major focus of drug discovery targeting voltage-gated sodium channels has been on strategies for improving the therapeutic index. One of the leading strategies is to identify selective sodium channel blockers designed to preferentially block NaV1.7, NaV1.8, NaV1.9 and/or NaV1.3. These are the sodium channel isoforms preferentially expressed in sensory neurons and unlikely to be involved in generating any dose-limiting side effects. For example, there is concern that blocking of NaV1.5 would be arrhythmogenic, so that selectivity of a sodium channel blocker against NaV1.5 is viewed as highly desirable. Furthermore, nearly 700 mutations of the SCN1A gene that codes for NaV1.1 have been identified in patients with Severe Myoclonic Epilepsy of Infancy (SMEI), making this the most commonly mutated gene in human epilepsy. Half of these mutations result in protein truncation (Meisler, M. H., et al., The Journal of Physiology (2010), 588: 1841-8). Thus, selectivity of a sodium channel blocker against NaV1.1 is also desirable.

In addition to the strategies of identifying selective sodium channel blockers, there is the continuing strategy of identifying therapeutic agents for the treatment of neuropathic pain. There has been some degree of success in treating neuropathic pain symptoms by using medications originally approved as anticonvulsants, such as gabapentin, and more recently pregabalin. However, pharmacotherapy for neuropathic pain has generally had limited success for a variety of reasons: sedation, especially by drugs first developed as anticonvulsants or antidepressants, addiction or tachyphylaxis, especially by opiates, or lack of efficacy, especially by NSAIDs and anti-inflammatory agents. Consequently, there is still a considerable need to explore novel treatment modalities for neuropathic pain, which includes, but is not limited to, post-herpetic neuralgia, trigeminal neuralgia, diabetic neuropathy, chronic lower back pain, phantom limb pain, and pain resulting from cancer and chemotherapy, chronic pelvic pain, complex regional pain syndrome and related neuralgias.

There are a limited number of effective sodium channel blockers for the treatment of pain with a minimum of adverse side effects which are currently in the clinic. There is also an unmet medical need to treat neuropathic pain and other sodium channel associated pathological states effectively and without adverse side effects due to the blocking of sodium channels not involved in nociception. The present invention provides methods to meet these critical needs.

SUMMARY OF THE INVENTION

In one aspect the present invention provides for novel compounds. In a first embodiment of such compounds (Embodiment 1; abbreviated as “E1”) the invention provides for a compound of formula (I):

or a pharmaceutically acceptable salt thereof, wherein:

R1 is C1-8 alkyl, C2-8 alkenyl, C1-8 haloalkyl, C1-8 alkoxy, C3-8 carbocycle, C-linked 3-15 membered heterocyclyl, or —NR1AR1B, wherein R1A and R1B are each independently selected from the group consisting of hydrogen, C1-8 alkyl, C1-8 alkoxy, and wherein R1A and R1B are optionally combined to form a 3 to 8 membered heterocyclyl ring optionally comprising 1 additional heteroatom selected from N, O and S; and wherein R is optionally substituted with from 1 to 5 substituents selected from the group consisting of C1-4 alkyl, C1-4 haloalkyl, F, Cl, Br, I, —OH, —CN, —NO2, —NRR1aRR1b, —ORR1a, —SRR1a, —Si(RR1a)3 and C3-6 carbocycle; wherein RR1a and RR1b are independently selected from the group consisting of hydrogen, C1-8 alkyl, C1-8 haloalkyl;

RN is hydrogen, C1-4 alkyl or C1-4 haloalkyl;

R2 is selected from the group consisting of H, F, Cl, Br, I, —CN, C1-8 alkyl, C1-8 haloalkyl and C1-8 alkoxy;

R3 is selected from the group consisting of H, F, Cl, Br, I, —CN, C1-8 alkyl, C1-8 haloalkyl and C1-8 alkoxy;

R4 is selected from the group consisting of H, F, Cl, Br, I, —CN, C1-8 alkyl, C1-8 haloalkyl and C1-8 alkoxy;

R5 is selected from the group consisting of H, C1-8 alkyl, C1-8 haloalkyl, C1-8 alkoxy, and C3-8 cycloalkyl, wherein said C1-8 alkoxy, and C3-8 cycloalkyl is optionally substituted with 1-3 substituents selected from F, Cl, Br and I;

L is a linker selected from the group consisting of C1-4 alkylene, C2-4 alkenylene, and C2-4 alkynylene, wherein L is optionally substituted with from 1 to 3 substituents selected from the group consisting of ═O, C1-4 alkyl, halo, and C1-4 haloalkyl;

m is 0 or 1;

X2 is selected from the group consisting of absent, —O—, —S(O)—, —S(O)2— and —N(RX)— wherein Rx is H, C1-8 alkyl, C1-8 alkanoyl, or —S(O)2(C1-8 alkyl);

n is 0, 1, 2, 3, 4, or 5;

the ring A is a 3-15 membered carbocyclyl, a 6-12 membered aryl, a 5-12 membered heteroaryl, or a 3-15 membered heterocyclyl;

each RAA is independently selected from the group consisting of C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, CN, F, Cl, Br and I; and

RA is selected from the group consisting of H, —ORA1, —(XRA)-(6-12 membered aryl), —(XRA)-(5-12 membered heteroaryl), and —RA2, wherein said 6-12 membered aryl and 5-12 membered heteroaryl of RA is optionally substituted with from 1 to 5 substituents independently selected from the group consisting of F, Cl, Br, I, C1-4 alkyl, C1-4 haloalkyl, and C1-4(halo)alkoxy; RA1 is selected from the group consisting of hydrogen, C1-8 alkyl, C2-8 alkenyl, C1-8 haloalkyl, C3-8 cycloalkyl, phenyl and benzyl; RA2 is selected from the group consisting of C1-8 alkyl that is optionally substituted with one or more substituents selected from oxo (═O), fluoro, amino, C1-4 alkylamino and di(C1-4alkyl)amino; XRA is selected from the group consisting of absent, —C(═O)—, and C1-4 alkylene; wherein any C1-4 alkylene, of XRA is optionally substituted with 1 to 3 substituents selected from the group consisting of C1-4 alkyl, C1-4 haloalkyl, and phenyl that is optionally substituted with 1 to 5 substituents selected from, F, Cl, Br, I, —NH2, —OH, —CN, —NO2, C1-4 alkyl, C1-4 haloalkyl, C1-4 alkoxy, C1-4(halo)alkoxy, C1-4 alkylamino and C1-4 dialkylamino.

Further embodiments (E) of the first embodiment of compounds of the invention, are described below.

E2. The compound of E1 that has the formula (Ia):

or a pharmaceutically acceptable salt thereof.

E3. The compound of E1 that has the formula (Ib):

or a pharmaceutically acceptable salt thereof.

E4. The compound of E1, E2, or E3 wherein R2 is H.

E5. The compound of E1, E2, E3, or E4 wherein R3 is halo.

E6. The compound of E1, E2, E3, or E4 wherein R3 is F.

E7. The compound of E1, E2, E3, E4, E5, or E6 wherein R4 is H.

E8. The compound of E1, E2, E3, E4, E5, E6, or E7 wherein R5 is C1-4 alkyl or C1-4 haloalkyl.

E9. The compound of E1, E2, E3, E4, E5, E6, or E7 wherein R5 is methyl.

E10. The compound of E1 that has the formula (Ic):

or a pharmaceutically acceptable salt thereof.

E11. The compound of E1 that has the formula (Id):

or a pharmaceutically acceptable salt thereof.

E12. The compound of E1 that has the formula (Ie):

or a pharmaceutically acceptable salt thereof.

E13. The compound of E1, E2, E3, E4, E5, E6, E7, E8, E9, E10, E11, or E12 wherein R1 is methyl, cyclopropyl, or 1-azetidinyl.

E14. The compound of E1, E2, E3, E4, E5, E6, E7, E8, E9, E10, E11, E12, or E13 wherein A is an optionally substituted piperidine, optionally substituted pyrrolidine, optionally substituted azetidine, optionally substituted tetrahydronaphthlene, optionally substituted cyclohexane, optionally substituted tetrahydropyran, optionally substituted adamantly, or optionally substituted pyridine ring.

E15. The compound of E1, E2, E3, E4, E5, E6, E7, E8, E9, E10, E11, E12, or E13 wherein:

is:
and A is a 3-15 membered heterocyclyl.

E16. The compound of E1, E2, E3, E4, E5, E6, E7, E8, E9, E10, E11, E12, or E13 wherein:

is:
and A is a 3-15 membered heterocyclyl.

E17. The compound of E1, E2, E3, E4, E5, E6, E7, E8, E9, E10, E11, E12, E13, E14, E15, or E16 wherein each RAA is independently selected from the group consisting of F, Cl and C1-4 haloalkyl.

E18. The compound of E1, E4, E5, E6, E7, E8, E9, E10, or E13 wherein:

is selected from the group consisting of:

E19. The compound of E1, E2, E3, E4, E5, E6, E7, E8, E9, E10, E11, E12, E13, E14, E15, E16, or E17 wherein RA is selected from the group consisting of:

E20. The compound of E1 which is selected from:

and salts thereof.

In another aspect the present invention provides for a pharmaceutical composition comprising a compound of formula I or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

In another aspect the present invention provides for a method of treating a disease or condition in a mammal selected from the group consisting of pain, depression, cardiovascular diseases, respiratory diseases, and psychiatric diseases, and combinations thereof, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof. In another aspect of the present invention said disease or condition is selected from the group consisting of neuropathic pain, inflammatory pain, visceral pain, cancer pain, chemotherapy pain, trauma pain, surgical pain, post-surgical pain, childbirth pain, labor pain, neurogenic bladder, ulcerative colitis, chronic pain, persistent pain, peripherally mediated pain, centrally mediated pain, chronic headache, migraine headache, sinus headache, tension headache, phantom limb pain, dental pain, peripheral nerve injury or a combination thereof. In another aspect of the present invention said disease or condition is selected from the group consisting of pain associated with HIV, HIV treatment induced neuropathy, trigeminal neuralgia, post-herpetic neuralgia, eudynia, heat sensitivity, tosarcoidosis, irritable bowel syndrome, Crohns disease, pain associated with multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), diabetic neuropathy, peripheral neuropathy, arthritis, rheumatoid arthritis, osteoarthritis, atherosclerosis, paroxysmal dystonia, myasthenia syndromes, myotonia, malignant hyperthermia, cystic fibrosis, pseudoaldosteronism, rhabdomyolysis, hypothyroidism, bipolar depression, anxiety, schizophrenia, sodium channel toxi related illnesses, familial erythromelalgia, primary erythromelalgia, familial rectal pain, cancer, epilepsy, partial and general tonic seizures, restless leg syndrome, arrhythmias, fibromyalgia, neuroprotection under ischaemic conditions cause by stroke or neural trauma, tach-arrhythmias, atrial fibrillation and ventricular fibrillation.

In another aspect the present invention provides for a method of treating pain in a mammal by the inhibition of ion flux through a voltage-dependent sodium channel in the mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a method of decreasing ion flux through a voltage-dependent sodium channel in a cell in a mammal, wherein the method comprises contacting the cell with a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a method of treating pruritus in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a method of treating cancer in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a method of treating, but not preventing, pain in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof. In another aspect of the present invention the pain is selected from the group consisting of neuropathic pain, inflammatory pain, visceral pain, cancer pain, chemotherapy pain, trauma pain, surgical pain, post-surgical pain, childbirth pain, labor pain, neurogenic bladder, ulcerative colitis, chronic pain, persistent pain, peripherally mediated pain, centrally mediated pain, chronic headache, migraine headache, sinus headache, tension headache, phantom limb pain, dental pain, peripheral nerve injury or a combination thereof. In another aspect the present invention the pain is associated with a disease or condition selected from the group consisting of HIV, HIV treatment induced neuropathy, trigeminal neuralgia, post-herpetic neuralgia, eudynia, heat sensitivity, tosarcoidosis, irritable bowel syndrome, Crohns disease, pain associated with multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), diabetic neuropathy, peripheral neuropathy, arthritis, rheumatoid arthritis, osteoarthritis, atherosclerosis, paroxysmal dystonia, myasthenia syndromes, myotonia, malignant hyperthermia, cystic fibrosis, pseudoaldosteronism, rhabdomyolysis, hypothyroidism, bipolar depression, anxiety, schizophrenia, sodium channel toxi related illnesses, familial erythromelalgia, primary erythromelalgia, familial rectal pain, cancer, epilepsy, partial and general tonic seizures, restless leg syndrome, arrhythmias, fibromyalgia, neuroprotection under ischaemic conditions cause by stroke or neural trauma, tach-arrhythmias, atrial fibrillation and ventricular fibrillation.

In another aspect the present invention provides for a method of treating, but not preventing, acute pain or chronic pain in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a method of treating, but not preventing, neuropathic pain or inflammatory pain in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a method for the treatment or prophylaxis of pain, depression, cardiovascular disease, respiratory disease, or psychiatric disease, or a combinations thereof, in an animal which method comprises administering an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof.

In another aspect the present invention provides for a compound of formula I, or a pharmaceutically acceptable salt thereof for the use as a medicament for the treatment of diseases and disorders selected from the group consisting of pain, depression, cardiovascular diseases, respiratory diseases, and psychiatric diseases, or a combination thereof.

In another aspect the present invention provides for the use of a compound of formula I, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of diseases and disorders selected from the group consisting of pain, depression, cardiovascular diseases, respiratory diseases, and psychiatric diseases, or a combination thereof.

In another aspect the present invention provides for the invention as described herein.

DETAILED DESCRIPTION OF THE INVENTION Definitions

As used herein, the term “alkyl”, by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain hydrocarbon radical, having the number of carbon atoms designated (i.e., C1-8 means one to eight carbons). Examples of alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, t-butyl, iso-butyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. The term “alkenyl” refers to an unsaturated alkyl radical having one or more double bonds. Similarly, the term “alkynyl” refers to an unsaturated alkyl radical having one or more triple bonds. Examples of such unsaturated alkyl groups include vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.

As used herein, the term “alkanoyl” is a group alkyl-C(═O)—. For example the group C1-8 alkanoyl is a group C1-7 alkyl-C(═O)—.

The term “heteroalkyl,” by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain hydrocarbon radical, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms can optionally be oxidized and the nitrogen heteroatom can optionally be quaternized. The heteroatom(s) O, N and S can be placed at any interior position of the heteroalkyl group. The heteroatom Si can be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. A “heteroalkyl” can contain up to three units of unsaturation, and also include mono- and poly-halogenated variants, or combinations thereof. Examples include —CH2—CH2—O—CH3, —CH2—CH2—O—CF3, —CH2—CH2—NH—CH3, —CH2—CH2—N(CH3)—CH3, —CH2—S—CH2—CH3, —S(O)—CH3, —CH2—CH2—S(O)2—CH3, —CH═CH—O—CH3, —Si(CH3)3, —CH2—CH═N—OCH3, and —CH═CH═N(CH3)—CH3. Up to two heteroatoms can be consecutive, such as, for example, —CH2—NH—OCH3 and —CH2—O—Si(CH3)3.

The term “alkylene” by itself or as part of another substituent means a divalent radical derived from an alkane (including branched alkane), as exemplified by —CH2CH2CH2CH2— and —CH(CH2)CH2CH2—. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. “Alkenylene” and “alkynylene” refer to the unsaturated forms of “alkylene” having double or triple bonds, respectively.

The term “heteroalkylene” by itself or as part of another substituent means a divalent radical, saturated or unsaturated or polyunsaturated, derived from heteroalkyl, as exemplified by —CH2—CH2—S—CH2CH2— and —CH2—S—CH2—CH2—NH—CH2—, —CH2—CH═CH—, —CH2—CH═C(H)CH2—O—CH2— and —S—CH2—C≡C—. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). The terms “alkoxy,” “alkylamino” and “alkylthio”, are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom (“oxy”), an amino group (“amino”) or thio group. Additionally, for dialkylamino groups, the alkyl portions can be the same or different.

The terms “halo” or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. The term “haloalkyl,” is meant to include monohaloalkyl and polyhaloalkyl. For example, the term “C1-4 haloalkyl” is mean to include trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, difluoromethyl, and the like.

The term “aryl” as used herein refers to a single all carbon aromatic ring or a multiple condensed all carbon ring system wherein at least one of the rings is aromatic. For example, in certain embodiments, an aryl group has 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 12 carbon atoms. Aryl includes a phenyl radical. Aryl also includes multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) having about 9 to 20 carbon atoms in which at least one ring is aromatic and wherein the other rings may be aromatic or not aromatic (i.e., carbocycle). Such multiple condensed ring systems are optionally substituted with one or more (e.g., 1, 2 or 3) oxo groups on any carbocycle portion of the multiple condensed ring system. The rings of the multiple condensed ring system can be connected to each other via fused, spiro and bridged bonds when allowed by valency requirements. It is to be understood that the point of attachment of a multiple condensed ring system, as defined above, can be at any position of the ring system including an aromatic or a carbocycle portion of the ring. Non-limiting examples of aryl groups include, but are not limited to, phenyl, indenyl, naphthyl, 1,2,3,4-tetrahydronaphthyl, anthracenyl, and the like.

The term “carbocycle” or “carbocyclyl” refers to a single saturated (i.e., cycloalkyl) or a single partially unsaturated (e.g., cycloalkenyl, cycloalkadienyl, etc.) all carbon ring having 3 to 7 carbon atoms (i.e., (C3-C7)carbocycle). The term “carbocycle” or “carbocyclyl” also includes multiple condensed, saturated and partially unsaturated all carbon ring systems (e.g., ring systems comprising 2, 3 or 4 carbocyclic rings). Accordingly, carbocycle includes multicyclic carbocyles such as a bicyclic carbocycles (e.g., bicyclic carbocycles having about 6 to 12 carbon atoms such as bicyclo[3.1.0]hexane and bicyclo[2.1.1]hexane), and polycyclic carbocycles (e.g tricyclic and tetracyclic carbocycles with up to about 20 carbon atoms). The rings of the multiple condensed ring system can be connected to each other via fused, spiro and bridged bonds when allowed by valency requirements. For example, multicyclic carbocyles can be connected to each other via a single carbon atom to form a spiro connection (e.g., spiropentane, spiro[4,5]decane, etc), via two adjacent carbon atoms to form a fused connection (e.g., carbocycles such as decahydronaphthalene, norsabinane, norcarane) or via two non-adjacent carbon atoms to form a bridged connection (e.g., norbornane, bicyclo[2.2.2]octane, etc). The “carbocycle” or “carbocyclyl” can also be optionally substituted with one or more (e.g., 1, 2 or 3) oxo groups. In one embodiment the term carbocycle includes a 3-15 membered carbocycle.

In one embodiment the term carbocycle includes a 3-8 membered carbocycle. In one embodiment the term carbocycle includes a 3-6 membered carbocycle. In one embodiment the term carbocycle includes a 3-5 membered carbocycle. Non-limiting examples of carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, bicyclo[2.2.1]heptane, pinane, adamantane, norborene, spirocyclic C5-12 alkane, and 1-cyclohex-3-enyl.

The term “heteroaryl” as used herein refers to a single aromatic ring that has at least one atom other than carbon in the ring, wherein the atom is selected from the group consisting of oxygen, nitrogen and sulfur; “heteroaryl” also includes multiple condensed ring systems that have at least one such aromatic ring, which multiple condensed ring systems are further described below. In one embodiment the term heteroaryl includes a 5-12 membered heteroaryl. Thus, “heteroaryl” includes single aromatic rings of from about 1 to 6 carbon atoms and about 1-4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur. The sulfur and nitrogen atoms may also be present in an oxidized form provided the ring is aromatic. Exemplary heteroaryl ring systems include but are not limited to pyridyl, pyrimidinyl, oxazolyl or furyl. “Heteroaryl” also includes multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) wherein a heteroaryl group, as defined above, is condensed with one or more rings selected from heteroaryls (to form for example a naphthyridinyl such as 1,8-naphthyridinyl), heterocycles, (to form for example a 1,2,3,4-tetrahydronaphthyridinyl such as 1,2,3,4-tetrahydro-1,8-naphthyridinyl), carbocycles (to form for example 5,6,7,8-tetrahydroquinolyl) and aryls (to form for example indazolyl) to form the multiple condensed ring system. Thus, a heteroaryl (a single aromatic ring or multiple condensed ring system) has about 1-20 carbon atoms and about 1-6 heteroatoms within the heteroaryl ring. Such multiple condensed ring systems may be optionally substituted with one or more (e.g., 1, 2, 3 or 4) oxo groups on the carbocycle or heterocycle portions of the condensed ring. The rings of the multiple condensed ring system can be connected to each other via fused, spiro and bridged bonds when allowed by valency requirements. It is to be understood that the individual rings of the multiple condensed ring system may be connected in any order relative to one another. It is also to be understood that the point of attachment of a multiple condensed ring system (as defined above for a heteroaryl) can be at any position of the multiple condensed ring system including a heteroaryl, heterocycle, aryl or carbocycle portion of the multiple condensed ring system. It is also to be understood that the point of attachment for a heteroaryl or heteroaryl multiple condensed ring system can be at any suitable atom of the heteroaryl or heteroaryl multiple condensed ring system including a carbon atom and a heteroatom (e.g., a nitrogen). Exemplary heteroaryls include but are not limited to pyridyl, pyrrolyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrazolyl, thienyl, indolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, furyl, oxadiazolyl, thiadiazolyl, quinolyl, isoquinolyl, benzothiazolyl, benzoxazolyl, indazolyl, quinoxalyl, quinazolyl, 5,6,7,8-tetrahydroisoquinolinyl benzofuranyl, benzimidazolyl, thianaphthenyl, pyrrolo[2,3-b]pyridinyl, quinazolinyl-4(3H)-one, triazolyl, 4,5,6,7-tetrahydro-1H-indazole and 3b,4,4a,5-tetrahydro-1H-cyclopropa[3,4]cyclo-penta[1,2-c]pyrazole.

The term “heterocyclyl” or “heterocycle” as used herein refers to a single saturated or partially unsaturated ring that has at least one atom other than carbon in the ring, wherein the atom is selected from the group consisting of oxygen, nitrogen and sulfur; the term also includes multiple condensed ring systems that have at least one such saturated or partially unsaturated ring, which multiple condensed ring systems are further described below. In one embodiment the term heterocyclyl includes a 3-15 membered heterocyclyl. Thus, the term includes single saturated or partially unsaturated rings (e.g., 3, 4, 5, 6 or 7-membered rings) from about 1 to 6 carbon atoms and from about 1 to 3 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur in the ring. The ring may be substituted with one or more (e.g., 1, 2 or 3) oxo groups and the sulfur and nitrogen atoms may also be present in their oxidized forms. Exemplary heterocycles include but are not limited to azetidinyl, tetrahydrofuranyl and piperidinyl. The term “heterocycle” also includes multiple condensed ring systems (e.g., ring systems comprising 2, 3 or 4 rings) wherein a single heterocycle ring (as defined above) can be condensed with one or more groups selected from heterocycles (to form for example a 1,8-decahydronapthyridinyl), carbocycles (to form for example a decahydroquinolyl) and aryls to form the multiple condensed ring system. Thus, a heterocycle (a single saturated or single partially unsaturated ring or multiple condensed ring system) has about 2-20 carbon atoms and 1-6 heteroatoms within the heterocycle ring. Such multiple condensed ring systems may be optionally substituted with one or more (e.g., 1, 2, 3 or 4) oxo groups on the carbocycle or heterocycle portions of the multiple condensed ring. The rings of the multiple condensed ring system can be connected to each other via fused, spiro and bridged bonds when allowed by valency requirements. It is to be understood that the individual rings of the multiple condensed ring system may be connected in any order relative to one another. It is also to be understood that the point of attachment of a multiple condensed ring system (as defined above for a heterocycle) can be at any position of the multiple condensed ring system including a heterocycle, aryl and carbocycle portion of the ring. It is also to be understood that the point of attachment for a heterocycle or heterocycle multiple condensed ring system can be at any suitable atom of the heterocycle or heterocycle multiple condensed ring system including a carbon atom and a heteroatom (e.g., a nitrogen). In one embodiment the term heterocycle includes a C2-20 heterocycle. In one embodiment the term heterocycle includes a C2-7 heterocycle. In one embodiment the term heterocycle includes a C2-5 heterocycle. In one embodiment the term heterocycle includes a C2-4 heterocycle. Exemplary heterocycles include, but are not limited to aziridinyl, azetidinyl, pyrrolidinyl, piperidinyl, homopiperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, tetrahydrofuranyl, dihydrooxazolyl, tetrahydropyranyl, tetrahydrothiopyranyl, 1,2,3,4-tetrahydroquinolyl, benzoxazinyl, dihydrooxazolyl, chromanyl, 1,2-dihydropyridinyl, 2,3-dihydrobenzofuranyl, 1,3-benzodioxolyl, 1,4-benzodioxanyl, spiro[cyclopropane-1,1′-isoindolinyl]-3′-one, isoindolinyl-1-one, 2-oxa-6-azaspiro[3.3]heptanyl, imidazolidin-2-one N-methylpiperidine, imidazolidine, pyrazolidine, butyrolactam, valerolactam, imidazolidinone, hydantoin, dioxolane, phthalimide, 1,4-dioxane, thiomorpholine, thiomorpholine-S-oxide, thiomorpholine-S,S-oxide, pyran, 3-pyrroline, thiopyran, pyrone, tetrhydrothiophene, quinuclidine, tropane, 2-azaspiro[3.3]heptane, (1R,5S)-3-azabicyclo[3.2.1]octane, (1s,4s)-2-azabicyclo[2.2.2]octane, (1R,4R)-2-oxa-5-azabicyclo[2.2.2]octane and pyrrolidin-2-one.

The above terms (e.g., “alkyl,” “aryl” and “heteroaryl”), in some embodiments, will include both substituted and unsubstituted forms of the indicated radical. Preferred substituents for each type of radical are provided below.

Substituents for the alkyl radicals (including those groups often referred to as alkylene, alkenyl, alkynyl, heteroalkyl, carbocycle, and heterocyclyl) can be a variety of groups including but not limited to, -halogen, —OR′, —NR′R″, —SR′, —SiR′R″R′″, —OC(O)R′, —C(O)R′, —CO2R′, —CONR′R″, —OC(O)NR′R″, —NR″C(O)R′, —NR′″C(O)NR′R″, —NR″C(O)2R′, —NHC(NH2)═NH, —NR′C(NH2)═N H, —NHC(NH2)═NR′, —NR′″C(NR′R″)═N—CN, —NR′″C(NR′R″)═NOR′, —NHC(NH2)═NR′, —S(O)R′, —S(O)2R′, —S(O)2NR′R″, —NR'S(O)2R″, —NR′″S(O)2NR′R″, —CN, —NO2, —(CH2)1-4—OR′, —(CH2)1-4—NR′R″, —(CH2)1-4—SR′, —(CH2)1-4—SiR′R″R′″, —(CH2)1-4—OC(O)R′, —(CH2)1-4—C(O)R′, —(CH2)1-4—CO2R′, —(CH2)1-4CONR′R″, in a number ranging from zero to (2m′+1), where m′ is the total number of carbon atoms in such radical. R′, R″ and R′″ each independently refer groups including, for example, hydrogen, unsubstituted C1-6 alkyl, unsubstituted heteroalkyl, unsubstituted aryl, aryl substituted with 1-3 halogens, unsubstituted C1-6 alkyl, C1-6 alkoxy or C1-6 thioalkoxy groups, or unsubstituted aryl-C1-4 alkyl groups, unsubstituted heteroaryl, substituted heteroaryl, among others. When R′ and R″ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 3-, 4-, 5-, 6-, or 7-membered ring. For example, —NR′R″ is meant to include 1-pyrrolidinyl and 4-morpholinyl. Other substituents for alkyl radicals, including heteroalkyl, alkylene, include for example, ═O, ═NR′, ═N—OR′, ═N—CN, ═NH, wherein R′ include substituents as described above.

Similarly, substituents for the aryl and heteroaryl groups are varied and are generally selected from the group including, but not limited to halogen, —OR′, —OC(O)R′, —NR′R″, —SR′, —R′, —CN, —NO2, —CO2R′, —CONR′R″, —C(O)R′, —OC(O)NR′R″, —NR″C(O)R′, —NR″C(O)2R′, —NR′C(O)NR″R″ —NHC(NH2)═NH, —NR′C(NH2)═NH, —NHC(NH2)═NR′, —S(O)R′, —S(O)2R′, —S(O)2NR′R″, —NR'S(O)2R″, —N3, perfluoro-C1-4 alkoxy, and perfluoro-C1-4alkyl, —(CH2)1-4—OR′, —(CH2)1-4—NR′R″, —(CH2)1-4—SR′, —(CH2)1-4—SiR′R″R′″, —(CH2)1-4—OC(O)R′, —(CH2)1-4—C(O)R′, —(CH2)1-4—CO2R′, —(CH2)1-4CONR′R″, in a number ranging from zero to the total number of open valences on the aromatic ring system; and where R′, R″ and R′″ are independently selected from hydrogen, C1-6 alkyl, C3-6 carbocycle, C2-6 alkenyl, C2-6 alkynyl, unsubstituted aryl and heteroaryl, (unsubstituted aryl)-C1-4 alkyl, and unsubstituted aryloxy-C1-4 alkyl. Other suitable substituents include each of the above aryl substituents attached to a ring atom by an alkylene tether of from 1-4 carbon atoms. When a substituent for the aryl or heteroaryl group contains an alkylene linker (e.g., —(CH2)1-4—NR′R″), the alkylene linker includes halo variants as well. For example, the linker “—(CH2)1-4—” when used as part of a substituent is meant to include difluoromethylene, 1,2-difluoroethylene, etc.

As used herein, the term “heteroatom” is meant to include oxygen (O), nitrogen (N), sulfur (S) and silicon (Si).

As used herein, the term “chiral” refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.

As used herein, the term “stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.

As used herein a wavy line “” that intersects a bond in a chemical structure indicates the point of attachment of the bond that the wavy bond intersects in the chemical structure to the remainder of a molecule.

As used herein, the term “C-linked” means that the group that the term describes is attached the remainder of the molecule through a ring carbon atom.

As used herein, the term “N-linked” means that the group that the term describes is attached to the remainder of the molecule through a ring nitrogen atom.

“Diastereomer” refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers can separate under high resolution analytical procedures such as electrophoresis and chromatography.

“Enantiomers” refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.

Stereochemical definitions and conventions used herein generally follow S. P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York; and Eliel, E. and Wilen, S., “Stereochemistry of Organic Compounds”, John Wiley & Sons, Inc., New York, 1994. The compounds of the invention can contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the invention, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present invention. Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and l or (+) and (−) are employed to designate the sign of rotation of plane-polarized light by the compound, with (−) or l meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. For a given chemical structure, these stereoisomers are identical except that they are mirror images of one another. A specific stereoisomer can also be referred to as an enantiomer, and a mixture of such isomers is often called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which can occur where there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomeric species, devoid of optical activity.

When a bond in a compound formula herein is drawn in a non-stereochemical manner (e.g. flat), the atom to which the bond is attached includes all stereochemical possibilities. When a bond in a compound formula herein is drawn in a defined stereochemical manner (e.g. bold, bold-wedge, dashed or dashed-wedge), it is to be understood that the atom to which the stereochemical bond is attached is enriched in the absolute stereoisomer depicted unless otherwise noted. In one embodiment, the compound may be at least 51% the absolute stereoisomer depicted. In another embodiment, the compound may be at least 80% the absolute stereoisomer depicted. In another embodiment, the compound may be at least 90% the absolute stereoisomer depicted. In another embodiment, the compound may be at least 95% the absolute stereoisomer depicted. In another embodiment, the compound may be at least 97% the absolute stereoisomer depicted. In another embodiment, the compound may be at least 98% the absolute stereoisomer depicted. In another embodiment, the compound may be at least 99% the absolute stereoisomer depicted.

As used herein, the term “tautomer” or “tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons.

As used herein, the term “solvate” refers to an association or complex of one or more solvent molecules and a compound of the invention. Examples of solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term “hydrate” refers to the complex where the solvent molecule is water.

As used herein, the term “protecting group” refers to a substituent that is commonly employed to block or protect a particular functional group on a compound. For example, an “amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ) and 9-fluorenylmethylenoxycarbonyl (Fmoc). Similarly, a “hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable protecting groups include acetyl and silyl. A “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include phenylsulfonylethyl, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p-toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2-(diphenylphosphino)-ethyl, nitroethyl and the like. For a general description of protecting groups and their use, see P. G. M. Wuts and T. W. Greene, Greene's Protective Groups in Organic Synthesis 4th edition, Wiley-Interscience, New York, 2006.

As used herein, the term “mammal” includes, but is not limited to, humans, mice, rats, guinea pigs, monkeys, dogs, cats, horses, cows, pigs, and sheep

As used herein, the term “pharmaceutically acceptable salts” is meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds of the present invention contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of salts derived from pharmaceutically-acceptable inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like. Salts derived from pharmaceutically-acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally-occurring amines and the like, such as arginine, betaine, caffeine, choline, N,N′-dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like. When compounds of the present invention contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge, S. M., et al., “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19). Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.

The neutral forms of the compounds can be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.

In addition to salt forms, the present invention provides compounds which are in a prodrug form. As used herein the term “prodrug” refers to those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention. Additionally, prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.

Prodrugs of the invention include compounds wherein an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues, is covalently joined through an amide or ester bond to a free amino, hydroxy or carboxylic acid group of a compound of the present invention. The amino acid residues include but are not limited to the 20 naturally occurring amino acids commonly designated by three letter symbols and also includes phosphoserine, phosphothreonine, phosphotyrosine, 4-hydroxyproline, hydroxylysine, demosine, isodemosine, gamma-carboxyglutamate, hippuric acid, octahydroindole-2-carboxylic acid, statine, 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, penicillamine, ornithine, 3-methylhistidine, norvaline, beta-alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, methyl-alanine, para-benzoylphenylalanine, phenylglycine, propargylglycine, sarcosine, methionine sulfone and tert-butylglycine.

Additional types of prodrugs are also encompassed. For instance, a free carboxyl group of a compound of the invention can be derivatized as an amide or alkyl ester. As another example, compounds of this invention comprising free hydroxy groups can be derivatized as prodrugs by converting the hydroxy group into a group such as, but not limited to, a phosphate ester, hemisuccinate, dimethylaminoacetate, or phosphoryloxymethyloxycarbonyl group, as outlined in Fleisher, D. et al., (1996) Improved oral drug delivery: solubility limitations overcome by the use of prodrugs Advanced Drug Delivery Reviews, 19:115. Carbamate prodrugs of hydroxy and amino groups are also included, as are carbonate prodrugs, sulfonate esters and sulfate esters of hydroxy groups. Derivatization of hydroxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers, wherein the acyl group can be an alkyl ester optionally substituted with groups including, but not limited to, ether, amine and carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, are also encompassed. Prodrugs of this type are described in J. Med. Chem., (1996), 39:10. More specific examples include replacement of the hydrogen atom of the alcohol group with a group such as (C1-6)alkanoyloxymethyl, 1-((C1-6)alkanoyloxy)ethyl, 1-methyl-1-((C1-6)alkanoyloxy)ethyl, (C1-6)alkoxycarbonyloxymethyl, N—(C1-6)alkoxycarbonylaminomethyl, succinoyl, (C1-6)alkanoyl, alpha-amino(C1-4)alkanoyl, arylacyl and alpha-aminoacyl, or alpha-aminoacyl-alpha-aminoacyl, where each alpha-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(OH)2, —P(O)(O(C1-6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).

For additional examples of prodrug derivatives, see, for example, a) Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985) and Methods in Enzymology, Vol. 42, p. 309-396, edited by K. Widder, et al. (Academic Press, 1985); b) A Textbook of Drug Design and Development, edited by Krogsgaard-Larsen and H. Bundgaard, Chapter 5 “Design and Application of Prodrugs,” by H. Bundgaard p. 113-191 (1991); c) H. Bundgaard, Advanced Drug Delivery Reviews, 8:1-38 (1992); d) H. Bundgaard, et al., Journal of Pharmaceutical Sciences, 77:285 (1988); and e) N. Kakeya, et al., Chem. Pharm. Bull., 32:692 (1984), each of which is specifically incorporated herein by reference.

Additionally, the present invention provides for metabolites of compounds of the invention. As used herein, a “metabolite” refers to a product produced through metabolism in the body of a specified compound or salt thereof. Such products can result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound.

Metabolite products typically are identified by preparing a radiolabelled (e.g., 14C or 3H) isotope of a compound of the invention, administering it parenterally in a detectable dose (e.g., greater than about 0.5 mg/kg) to an animal such as rat, mouse, guinea pig, monkey, or to man, allowing sufficient time for metabolism to occur (typically about 30 seconds to 30 hours) and isolating its conversion products from the urine, blood or other biological samples. These products are easily isolated since they are labeled (others are isolated by the use of antibodies capable of binding epitopes surviving in the metabolite). The metabolite structures are determined in conventional fashion, e.g., by MS, LC/MS or NMR analysis. In general, analysis of metabolites is done in the same way as conventional drug metabolism studies well known to those skilled in the art. The metabolite products, so long as they are not otherwise found in vivo, are useful in diagnostic assays for therapeutic dosing of the compounds of the invention.

Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention can exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.

Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, diastereomers, geometric isomers, regioisomers and individual isomers (e.g., separate enantiomers) are all intended to be encompassed within the scope of the present invention.

The terms “treat” and “treatment” refer to both therapeutic treatment and/or prophylactic treatment or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological change or disorder, such as, for example, the development or spread of cancer. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease or disorder, stabilized (i.e., not worsening) state of disease or disorder, delay or slowing of disease progression, amelioration or palliation of the disease state or disorder, and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the disease or disorder as well as those prone to have the disease or disorder or those in which the disease or disorder is to be prevented.

The phrase “therapeutically effective amount” or “effective amount” means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. For cancer therapy, efficacy can, for example, be measured by assessing the time to disease progression (TTP) and/or determining the response rate (RR).

The term “bioavailability” refers to the systemic availability (i.e., blood/plasma levels) of a given amount of drug administered to a patient. Bioavailability is an absolute term that indicates measurement of both the time (rate) and total amount (extent) of drug that reaches the general circulation from an administered dosage form.

In another embodiment, the compound is selected from compounds of formula I as described in the Examples herein and salts thereof.

Synthesis of Compounds

Compounds of formula (I) may be prepared by the process illustrated in Scheme 1 below.

Compounds of formula (I) can be prepared from compounds of formulae (X) (V=Cl) according to reaction step (vii) by activation of the acid group of formulae (IX) with reagents such as oxalyl chloride, thionyl chloride, carbonyl di-imidazole (CDI), a uronium based amide coupling agent, propylphosphonic anhydride or a carbodiimide reagent followed by displacement with a suitable sulfonamide of formula (XI) in the presence of a nucleophilic base such as 4-dimethylaminopyridine, triethylamine, diethyliropropyl amine.

Alternatively, compounds of formula (I) can be prepared from compounds of formula (X) (V=NH2) according to reaction step (ix) by displacement of a sulfonyl chloride of formula (XII) under basic reaction conditions.

Compounds of formula (IX) can be prepared by hydrosis of the ester functional group in compounds of formula (VIII) by either acidic or basic methods according to step (vi) as required.

Compounds of formula (VIII) can be prepared from compounds of formula (VI) under suitable N-alkylation conditions in a polar aprotic solvent, such as, acetonitrile, dimethylformamide, tetrahydrofuran, dioxane or dimethoxyethane in the presence of a base, such as cesium carbonate, anhydrous potassium carbonate, sodium hydride, or calcium hydride with compounds of formula (VII) according to reaction step (v).

Compounds of formula (VI) can be prepared from compounds of formula (V) according to reaction step (iv) by deprotection of the protecting by either acidic conditions using acid such as trifluoroacetic acid or hydrochloric acid in a aprotic non-polar solvent such as dichlromethane, dichloroethane or hydrogenation with a variety of palladium catalysis such a Palladium on carcoal and palladium hydroxide in a polar protic solvent such a methanol, ethanol. Compounds of formula (V) can be prepared from compounds of formula (IV) according to the reaction step (iii) using either metal catalyzed carbonylation using palladium catalysis such as tetrakistriphenylphosphine palladium (O), palladium acetate in a polar aprotic solvent, such as dioxane, dimethoxyethane, tetrahydrofuran, or acetonitrile in the presence of carbon monoxide or molybdenumhexacarbonyl. Compounds of formula (IV) can be obtain from compounds of formula (III) by using protecting group such as benzyl chloride, di-tert-butyl dicarbonate, benzyl chloroformate, or para-methoxyl benzyl chloride. In a presence of a base such a sodium hydride, DMAP, potassium carbonate, DIPEA, triethyl amine, or pyridine in a solvent such as DMF, dioxane, or dichloromethane according to the reaction step (ii). Compounds of formula (III) can be prepared from commercially available compounds of formula (II) according to the reaction step (i) using hydrazine in refluxing conditions in a polar aprotic solvent such as toluene, benzene, dioxane.

Pharmaceutical Compositions and Administration

In addition to one or more of the compounds provided above (or stereoisomers, geometric isomers, tautomers, solvates, metabolites, isotopes, pharmaceutically acceptable salts, or prodrugs thereof), the invention also provides for compositions and medicaments comprising a compound of formula I or and embodiment thereof and at least one pharmaceutically acceptable carrier, diluent or excipient. The compositions of the invention can be used to selectively inhibit NaV1.7 in patients (e.g, humans).

The term “composition,” as used herein, is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. By “pharmaceutically acceptable” it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.

In one embodiment, the invention provides for pharmaceutical compositions (or medicaments) comprising a compound of formula I or an embodiment thereof, and its stereoisomers, geometric isomers, tautomers, solvates, metabolites, isotopes, pharmaceutically acceptable salts, or prodrugs thereof) and a pharmaceutically acceptable carrier, diluent or excipient. In another embodiment, the invention provides for preparing compositions (or medicaments) comprising compounds of the invention. In another embodiment, the invention provides for administering compounds of formula I or its embodiments and compositions comprising compounds of formula I or an embodiment thereof to a patient (e.g., a human patient) in need thereof.

Compositions are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. The effective amount of the compound to be administered will be governed by such considerations, and is the minimum amount necessary to inhibit NaV1.7 activity as required to prevent or treat the undesired disease or disorder, such as for example, pain. For example, such amount may be below the amount that is toxic to normal cells, or the mammal as a whole.

In one example, the therapeutically effective amount of the compound of the invention administered parenterally per dose will be in the range of about 0.01-100 mg/kg, alternatively about e.g., 0.1 to 20 mg/kg of patient body weight per day, with the typical initial range of compound used being 0.3 to 15 mg/kg/day. The daily does is, in certain embodiments, given as a single daily dose or in divided doses two to six times a day, or in sustained release form. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 mg to about 1,400 mg. This dosage regimen may be adjusted to provide the optimal therapeutic response. The compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.

The compounds of the present invention may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents.

The compounds of the invention may be administered by any suitable means, including oral, topical (including buccal and sublingual), rectal, vaginal, transdermal, parenteral, subcutaneous, intraperitoneal, intrapulmonary, intradermal, intrathecal and epidural and intranasal, and, if desired for local treatment, intralesional administration. Parenteral infusions include intramuscular, intravenous, intraarterial, intraperitoneal, intracerebral, intraocular, intralesional or subcutaneous administration.

The compositions comprising compounds of formula I or an embodiment thereof are normally formulated in accordance with standard pharmaceutical practice as a pharmaceutical composition. A typical formulation is prepared by mixing a compound of the present invention and a diluent, carrier or excipient. Suitable diluents, carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C., et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).

Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water and the like. The particular carrier, diluent or excipient used will depend upon the means and purpose for which a compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal. In general, safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG 400, PEG 300), etc. and mixtures thereof. The formulations can also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).

Acceptable diluents, carriers, excipients and stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG). A active pharmaceutical ingredient of the invention (e.g., compound of formula I or an embodiment thereof) can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Remington: The Science and Practice of Pharmacy: Remington the Science and Practice of Pharmacy (2005) 21st Edition, Lippincott Williams & Wilkins, Philadelphia, Pa.

Sustained-release preparations of a compound of the invention (e.g., compound of formula I or an embodiment thereof) can be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a compound of formula I or an embodiment thereof, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinyl alcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al., Biopolymers 22:547, 1983), non-degradable ethylene-vinyl acetate (Langer et al., J. Biomed. Mater. Res. 15:167, 1981), degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate) and poly-D-(−)-3-hydroxybutyric acid (EP 133,988A). Sustained release compositions also include liposomally entrapped compounds, which can be prepared by methods known per se (Epstein et al., Proc. Natl. Acad. Sci. U.S.A. 82:3688, 1985; Hwang et al., Proc. Natl. Acad. Sci. U.S.A. 77:4030, 1980; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324A). Ordinarily, the liposomes are of the small (about 200-800 Angstroms) unilamelar type in which the lipid content is greater than about 30 mol % cholesterol, the selected proportion being adjusted for the optimal therapy.

The formulations include those suitable for the administration routes detailed herein. The formulations can conveniently be presented in unit dosage form and can be prepared by any of the methods well known in the art of pharmacy. Techniques and formulations generally are found in Remington: The Science and Practice of Pharmacy: Remington the Science and Practice of Pharmacy (2005) 21st Edition, Lippincott Williams & Wilkins, Philadelphia, Pa. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.

In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers, diluents or excipients or finely divided solid carriers, diluents or excipients, or both, and then, if necessary, shaping the product. A typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient. The formulations can be prepared using conventional dissolution and mixing procedures. For example, the bulk drug substance (i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent) is dissolved in a suitable solvent in the presence of one or more of the excipients described above. A compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to enable patient compliance with the prescribed regimen.

In one example, compounds of formula I or an embodiment thereof may be formulated by mixing at ambient temperature at the appropriate pH, and at the desired degree of purity, with physiologically acceptable carriers, i.e., carriers that are non-toxic to recipients at the dosages and concentrations employed into a galenical administration form. The pH of the formulation depends mainly on the particular use and the concentration of compound, but preferably ranges anywhere from about 3 to about 8. In one example, a compound of formula I (or an embodiment thereof) is formulated in an acetate buffer, at pH 5. In another embodiment, the compounds of formula I or an embodiment thereof are sterile. The compound may be stored, for example, as a solid or amorphous composition, as a lyophilized formulation or as an aqueous solution.

Formulations of a compound of the invention (e.g., compound of formula I or an embodiment thereof) suitable for oral administration can be prepared as discrete units such as pills, capsules, cachets or tablets each containing a predetermined amount of a compound of the invention.

Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, preservative, surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered active ingredient moistened with an inert liquid diluent. The tablets can optionally be coated or scored and optionally are formulated so as to provide slow or controlled release of the active ingredient therefrom.

Tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, e.g., gelatin capsules, syrups or elixirs can be prepared for oral use. Formulations of a compound of the invention (e.g., compound of formula I or an embodiment thereof) intended for oral use can be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients can be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesium stearate, stearic acid or talc. Tablets can be uncoated or can be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax can be employed.

An example of a suitable oral administration form is a tablet containing about 1 mg, 5 mg, 10 mg, 25 mg, 30 mg, 50 mg, 80 mg, 100 mg, 150 mg, 250 mg, 300 mg and 500 mg of the compound of the invention compounded with about 90-30 mg anhydrous lactose, about 5-40 mg sodium croscarmellose, about 5-30 mg polyvinylpyrrolidone (PVP) K30, and about 1-10 mg magnesium stearate. The powdered ingredients are first mixed together and then mixed with a solution of the PVP. The resulting composition can be dried, granulated, mixed with the magnesium stearate and compressed to tablet form using conventional equipment. An example of an aerosol formulation can be prepared by dissolving the compound, for example 5-400 mg, of the invention in a suitable buffer solution, e.g. a phosphate buffer, adding a tonicifier, e.g. a salt such sodium chloride, if desired. The solution may be filtered, e.g., using a 0.2 micron filter, to remove impurities and contaminants.

For treatment of the eye or other external tissues, e.g., mouth and skin, the formulations are preferably applied as a topical ointment or cream containing the active ingredient(s) in an amount of, for example, 0.075 to 20% w/w. When formulated in an ointment, the active ingredient can be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients can be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase of the cream base can include a polyhydric alcohol, i.e., an alcohol having two or more hydroxyl groups such as propylene glycol, butane 1,3-diol, mannitol, sorbitol, glycerol and polyethylene glycol (including PEG 400) and mixtures thereof. The topical formulations can desirably include a compound which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include dimethyl sulfoxide and related analogs.

The oily phase of the emulsions of this invention can be constituted from known ingredients in a known manner. While the phase can comprise merely an emulsifier, it desirably comprises a mixture of at least one emulsifier with a fat or an oil or with both a fat and an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of the cream formulations. Emulsifiers and emulsion stabilizers suitable for use in the formulation of the invention include Tween® 60, Span® 80, cetostearyl alcohol, benzyl alcohol, myristyl alcohol, glyceryl mono-stearate and sodium lauryl sulfate.

In one aspect of topical applications, it is desired to administer an effective amount of a pharmaceutical composition according to the invention to target area, e.g., skin surfaces, mucous membranes, and the like, which are adjacent to peripheral neurons which are to be treated. This amount will generally range from about 0.0001 mg to about 1 g of a compound of the invention per application, depending upon the area to be treated, whether the use is diagnostic, prophylactic or therapeutic, the severity of the symptoms, and the nature of the topical vehicle employed. A preferred topical preparation is an ointment, wherein about 0.001 to about 50 mg of active ingredient is used per cc of ointment base. The pharmaceutical composition can be formulated as transdermal compositions or transdermal delivery devices (“patches”). Such compositions include, for example, a backing, active compound reservoir, a control membrane, liner and contact adhesive. Such transdermal patches may be used to provide continuous pulsatile, or on demand delivery of the compounds of the present invention as desired.

Aqueous suspensions of a compound of the invention (e.g., compound of formula I or an embodiment thereof) contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxymethylcellulose, croscarmellose, povidone, methylcellulose, hydroxypropyl methylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatide (e.g., lecithin), a condensation product of an alkylene oxide with a fatty acid (e.g., polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol (e.g., heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride (e.g., polyoxyethylene sorbitan monooleate). The aqueous suspension can also contain one or more preservatives such as ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents and one or more sweetening agents, such as sucrose or saccharin.

Formulations of a compound of the invention (e.g., compound of formula I or an embodiment thereof) can be in the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension can be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, such as a solution in 1,3-butanediol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that can be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile fixed oils can conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid can likewise be used in the preparation of injectables.

The amount of active ingredient that can be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release formulation intended for oral administration to humans can contain approximately 1 to 1000 mg of active material compounded with an appropriate and convenient amount of carrier material which can vary from about 5 to about 95% of the total compositions (weight:weight). The pharmaceutical composition can be prepared to provide easily measurable amounts for administration. For example, an aqueous solution intended for intravenous infusion can contain from about 3 to 500 g of the active ingredient per milliliter of solution in order that infusion of a suitable volume at a rate of about 30 mL/hr can occur.

Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which can contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which can include suspending agents and thickening agents.

Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredient is dissolved or suspended in a suitable carrier, especially an aqueous solvent for the active ingredient. The active ingredient is preferably present in such formulations in a concentration of about 0.5 to 20% w/w, for example about 0.5 to 10% w/w, for example about 1.5% w/w.

Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.

Formulations for rectal administration can be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.

Formulations suitable for intrapulmonary or nasal administration have a particle size for example in the range of 0.1 to 500 microns (including particle sizes in a range between 0.1 and 500 microns in increments microns such as 0.5, 1, 30 microns, 35 microns, etc.), which is administered by rapid inhalation through the nasal passage or by inhalation through the mouth so as to reach the alveolar sacs. Suitable formulations include aqueous or oily solutions of the active ingredient. Formulations suitable for aerosol or dry powder administration can be prepared according to conventional methods and can be delivered with other therapeutic agents such as compounds heretofore used in the treatment of disorders as described below.

The formulations can be packaged in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water, for injection immediately prior to use. Extemporaneous injection solutions and suspensions are prepared from sterile powders, granules and tablets of the kind previously described. Preferred unit dosage formulations are those containing a daily dose or unit daily sub-dose, as herein above recited, or an appropriate fraction thereof, of the active ingredient.

When the binding target is located in the brain, certain embodiments of the invention provide for a compound of formula I (or an embodiment thereof) to traverse the blood-brain barrier. Certain neurodegenerative diseases are associated with an increase in permeability of the blood-brain barrier, such that a compound of formula I (or an embodiment thereof) can be readily introduced to the brain. When the blood-brain barrier remains intact, several art-known approaches exist for transporting molecules across it, including, but not limited to, physical methods, lipid-based methods, and receptor and channel-based methods.

Physical methods of transporting a compound of formula I (or an embodiment thereof) across the blood-brain barrier include, but are not limited to, circumventing the blood-brain barrier entirely, or by creating openings in the blood-brain barrier.

Circumvention methods include, but are not limited to, direct injection into the brain (see, e.g., Papanastassiou et al., Gene Therapy 9:398-406, 2002), interstitial infusion/convection-enhanced delivery (see, e.g., Bobo et al., Proc. Natl. Acad. Sci. U.S.A. 91:2076-2080, 1994), and implanting a delivery device in the brain (see, e.g., Gill et al., Nature Med. 9:589-595, 2003; and Gliadel Wafers™, Guildford Pharmaceutical).

Methods of creating openings in the barrier include, but are not limited to, ultrasound (see, e.g., U.S. Patent Publication No. 2002/0038086), osmotic pressure (e.g., by administration of hypertonic mannitol (Neuwelt, E. A., Implication of the Blood-Brain Barrier and its Manipulation, Volumes 1 and 2, Plenum Press, N.Y., 1989)), and permeabilization by, e.g., bradykinin or permeabilizer A-7 (see, e.g., U.S. Pat. Nos. 5,112,596, 5,268,164, 5,506,206, and 5,686,416).

Lipid-based methods of transporting a compound of formula I (or an embodiment thereof) across the blood-brain barrier include, but are not limited to, encapsulating the a compound of formula I (or an embodiment thereof) in liposomes that are coupled to antibody binding fragments that bind to receptors on the vascular endothelium of the blood-brain barrier (see, e.g., U.S. Patent Application Publication No. 2002/0025313), and coating a compound of formula I (or an embodiment thereof) in low-density lipoprotein particles (see, e.g., U.S. Patent Application Publication No. 2004/0204354) or apolipoprotein E (see, e.g., U.S. Patent Application Publication No. 2004/0131692).

Receptor and channel-based methods of transporting a compound of formula I (or an embodiment thereof) across the blood-brain barrier include, but are not limited to, using glucocorticoid blockers to increase permeability of the blood-brain barrier (see, e.g., U.S. Patent Application Publication Nos. 2002/0065259, 2003/0162695, and 2005/0124533); activating potassium channels (see, e.g., U.S. Patent Application Publication No. 2005/0089473), inhibiting ABC drug transporters (see, e.g., U.S. Patent Application Publication No. 2003/0073713); coating a compound of formula I (or an embodiment thereof) with a transferrin and modulating activity of the one or more transferrin receptors (see, e.g., U.S. Patent Application Publication No. 2003/0129186), and cationizing the antibodies (see, e.g., U.S. Pat. No. 5,004,697).

For intracerebral use, in certain embodiments, the compounds can be administered continuously by infusion into the fluid reservoirs of the CNS, although bolus injection may be acceptable. The inhibitors can be administered into the ventricles of the brain or otherwise introduced into the CNS or spinal fluid. Administration can be performed by use of an indwelling catheter and a continuous administration means such as a pump, or it can be administered by implantation, e.g., intracerebral implantation of a sustained-release vehicle. More specifically, the inhibitors can be injected through chronically implanted cannulas or chronically infused with the help of osmotic minipumps. Subcutaneous pumps are available that deliver proteins through a small tubing to the cerebral ventricles. Highly sophisticated pumps can be refilled through the skin and their delivery rate can be set without surgical intervention. Examples of suitable administration protocols and delivery systems involving a subcutaneous pump device or continuous intracerebroventricular infusion through a totally implanted drug delivery system are those used for the administration of dopamine, dopamine agonists, and cholinergic agonists to Alzheimer's disease patients and animal models for Parkinson's disease, as described by Harbaugh, J. Neural Transm. Suppl. 24:271, 1987; and DeYebenes et al., Mov. Disord. 2: 143, 1987.

A compound of formula I (or an embodiment thereof) used in the invention are formulated, dosed, and administered in a fashion consistent with good medical practice. Factors for consideration in this context include the particular disorder being treated, the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the site of delivery of the agent, the method of administration, the scheduling of administration, and other factors known to medical practitioners. A compound of formula I (or an embodiment thereof) need not be, but is optionally formulated with one or more agent currently used to prevent or treat the disorder in question. The effective amount of such other agents depends on the amount of a compound of the invention present in the formulation, the type of disorder or treatment, and other factors discussed above.

These are generally used in the same dosages and with administration routes as described herein, or about from 1 to 99% of the dosages described herein, or in any dosage and by any route that is empirically/clinically determined to be appropriate.

For the prevention or treatment of disease, the appropriate dosage of a compound of formula I (or an embodiment thereof) (when used alone or in combination with other agents) will depend on the type of disease to be treated, the properties of the compound, the severity and course of the disease, whether the compound is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the compound, and the discretion of the attending physician. The compound is suitably administered to the patient at one time or over a series of treatments. Depending on the type and severity of the disease, about 1 μg/kg to 15 mg/kg (e.g., 0.1 mg/kg-10 mg/kg) of compound can be an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion. One typical daily dosage might range from about 1 g kg to 100 mg/kg or more, depending on the factors mentioned above. For repeated administrations over several days or longer, depending on the condition, the treatment would generally be sustained until a desired suppression of disease symptoms occurs. One exemplary dosage of a compound of formula I (or an embodiment thereof) would be in the range from about 0.05 mg/kg to about 10 mg/kg. Thus, one or more doses of about 0.5 mg/kg, 2.0 mg/kg, 4.0 mg/kg, or 10 mg/kg (or any combination thereof) may be administered to the patient. Such doses may be administered intermittently, e.g., every week or every three weeks (e.g., such that the patient receives from about two to about twenty, or, e.g., about six doses of the antibody). An initial higher loading dose, followed by one or more lower doses may be administered. An exemplary dosing regimen comprises administering an initial loading dose of about 4 mg/kg, followed by a weekly maintenance dose of about 2 mg kg of the compound. However, other dosage regimens may be useful. The progress of this therapy is easily monitored by conventional techniques and assays.

Other typical daily dosages might range from, for example, about 1 g/kg to up to 100 mg/kg or more (e.g., about 1 g kg to 1 mg/kg, about 1 g/kg to about 5 mg/kg, about 1 mg kg to 10 mg/kg, about 5 mg/kg to about 200 mg/kg, about 50 mg/kg to about 150 mg/mg, about 100 mg/kg to about 500 mg/kg, about 100 mg/kg to about 400 mg/kg, and about 200 mg/kg to about 400 mg/kg), depending on the factors mentioned above. Typically, the clinician will administer a compound until a dosage is reached that results in improvement in or, optimally, elimination of, one or more symptoms of the treated disease or condition. The progress of this therapy is easily monitored by conventional assays. One or more agent provided herein may be administered together or at different times (e.g., one agent is administered prior to the administration of a second agent). One or more agent may be administered to a subject using different techniques (e.g., one agent may be administered orally, while a second agent is administered via intramuscular injection or intranasally). One or more agent may be administered such that the one or more agent has a pharmacologic effect in a subject at the same time. Alternatively, one or more agent may be administered, such that the pharmacological activity of the first administered agent is expired prior the administration of one or more secondarily administered agents (e.g., 1, 2, 3, or 4 secondarily administered agents).

Indications and Methods of Treatment

The compounds of the invention modulate, preferably inhibit, ion flux through a voltage-dependent sodium channel in a mammal, (e.g, a human). Any such modulation, whether it be partial or complete inhibition or prevention of ion flux, is sometimes referred to herein as “blocking” and corresponding compounds as “blockers” or “inhibitors”. In general, the compounds of the invention modulate the activity of a sodium channel downwards by inhibiting the voltage-dependent activity of the sodium channel, and/or reduce or prevent sodium ion flux across a cell membrane by preventing sodium channel activity such as ion flux.

The compounds of the invention inhibit the ion flux through a voltage-dependent sodium channel. In one aspect, the compounds are state or frequency dependent modifiers of the sodium channels, having a low affinity for the rested/closed state and a high affinity for the inactivated state. Without being bound by any particular theory, it is thought that these compounds are likely to interact with overlapping sites located in the inner cavity of the sodium conducting pore of the channel similar to that described for other state-dependent sodium channel blockers (Cestele, S., et al., op. cit.). These compounds may also be likely to interact with sites outside of the inner cavity and have allosteric effects on sodium ion conduction through the channel pore.

Any of these consequences may ultimately be responsible for the overall therapeutic benefit provided by these compounds.

Accordingly, the compounds of the invention are sodium channel blockers and are therefore useful for treating diseases and conditions in mammals, for example humans, and other organisms, including all those diseases and conditions which are the result of aberrant voltage-dependent sodium channel biological activity or which may be ameliorated by modulation of voltage-dependent sodium channel biological activity. In particular, the compounds of the invention, i.e., the compounds of formula (I) and embodiments and (or stereoisomers, geometric isomers, tautomers, solvates, metabolites, isotopes, pharmaceutically acceptable salts, or prodrugs thereof), are useful for treating diseases and conditions in mammals, for example humans, which are the result of aberrant voltage-dependent NaV1.7 biological activity or which may be ameliorated by the modulation, preferably the inhibition, of NaV1.7 biological activity. In certain aspects, the compounds of the invention selectively inhibit NaV1.7 over NaV1.5.

As defined herein, a sodium channel-mediated disease or condition refers to a disease or condition in a mammal, preferably a human, which is ameliorated upon modulation of the sodium channel and includes, but is not limited to, pain, central nervous conditions such as epilepsy, anxiety, depression and bipolar disease; cardiovascular conditions such as arrhythmias, atrial fibrillation and ventricular fibrillation; neuromuscular conditions such as restless leg syndrome and muscle paralysis or tetanus; neuroprotection against stroke, neural trauma and multiple sclerosis; and channelopathies such as erythromyalgia and familial rectal pain syndrome.

In one aspect, the present invention relates to compounds, pharmaceutical compositions and methods of using the compounds and pharmaceutical compositions for the treatment of sodium channel-mediated diseases in mammals, preferably humans and preferably diseases and conditions related to pain, central nervous conditions such as epilepsy, anxiety, depression and bipolar disease; cardiovascular conditions such as arrhythmias, atrial fibrillation and ventricular fibrillation; neuromuscular conditions such as restless leg syndrome and muscle paralysis or tetanus; neuroprotection against stroke, neural trauma and multiple sclerosis; and channelopathies such as erythromyalgia and familial rectal pain syndrome, by administering to a mammal, for example a human, in need of such treatment an effective amount of a sodium channel blocker modulating, especially inhibiting, agent.

A sodium channel-mediated disease or condition also includes pain associated with HIV, HIV treatment induced neuropathy, trigeminal neuralgia, glossopharyngeal neuralgia, neuropathy secondary to metastatic infiltration, adiposis dolorosa, thalamic lesions, hypertension, autoimmune disease, asthma, drug addiction (e.g., opiate, benzodiazepine, amphetamine, cocaine, alcohol, butane inhalation), Alzheimer, dementia, age-related memory impairment, Korsakoff syndrome, restenosis, urinary dysfunction, incontinence, Parkinson's disease, cerebrovascular ischemia, neurosis, gastrointestinal disease, sickle cell anemia, transplant rejection, heart failure, myocardial infarction, reperfusion injury, intermittant claudication, angina, convulsion, respiratory disorders, cerebral or myocardial ischemias, long-QT syndrome, Catecholeminergic polymorphic ventricular tachycardia, ophthalmic diseases, spasticity, spastic paraplegia, myopathies, myasthenia gravis, paramyotonia congentia, hyperkalemic periodic paralysis, hypokalemic periodic paralysis, alopecia, anxiety disorders, psychotic disorders, mania, paranoia, seasonal affective disorder, panic disorder, obsessive compulsive disorder (OCD), phobias, autism, Aspergers Syndrome, Retts syndrome, disintegrative disorder, attention deficit disorder, aggressivity, impulse control disorders, thrombosis, pre clampsia, congestive cardiac failure, cardiac arrest, Freidrich's ataxia, Spinocerebellear ataxia, myelopathy, radiculopathy, systemic lupus erythamatosis, granulomatous disease, olivo-ponto-cerebellar atrophy, spinocerebellar ataxia, episodic ataxia, myokymia, progressive pallidal atrophy, progressive supranuclear palsy and spasticity, traumatic brain injury, cerebral oedema, hydrocephalus injury, spinal cord injury, anorexia nervosa, bulimia, Prader-Willi syndrome, obesity, optic neuritis, cataract, retinal haemorrhage, ischaemic retinopathy, retinitis pigmentosa, acute and chronic glaucoma, macular degeneration, retinal artery occlusion, Chorea, Huntington's chorea, cerebral edema, proctitis, post-herpetic neuralgia, eudynia, heat sensitivity, sarcoidosis, irritable bowel syndrome, Tourette syndrome, Lesch-Nyhan Syndrome, Brugado syndrome, Liddle syndrome, Crohns disease, multiple sclerosis and the pain associated with multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), disseminated sclerosis, diabetic neuropathy, peripheral neuropathy, charcot marie tooth syndrome, arthritic, rheumatoid arthritis, osteoarthritis, chondrocalcinosis, atherosclerosis, paroxysmal dystonia, myasthenia syndromes, myotonia, myotonic dystrophy, muscular dystrophy, malignant hyperthermia, cystic fibrosis, pseudoaldosteronism, rhabdomyolysis, mental handicap, hypothyroidism, bipolar depression, anxiety, schizophrenia, sodium channel toxin related illnesses, familial erythromelalgia, primary erythromelalgia, rectal pain, cancer, epilepsy, partial and general tonic seizures, febrile seizures, absence seizures (petit mal), myoclonic seizures, atonic seizures, clonic seizures, Lennox Gastaut, West Syndome (infantile spasms), multiresistant seizures, seizure prophylaxis (anti-epileptogenic), familial Mediterranean fever syndrome, gout, restless leg syndrome, arrhythmias, fibromyalgia, neuroprotection under ischaemic conditions caused by stroke or neural trauma, tachy-arrhythmias, atrial fibrillation and ventricular fibrillation and as a general or local anesthetic.

As used herein, the term “pain” refers to all categories of pain and is recognized to include, but is not limited to, neuropathic pain, inflammatory pain, nociceptive pain, idiopathic pain, neuralgic pain, orofacial pain, burn pain, burning mouth syndrome, somatic pain, visceral pain, myofacial pain, dental pain, cancer pain, chemotherapy pain, trauma pain, surgical pain, post-surgical pain, childbirth pain, labor pain, chronic regional pain syndrome (CRPS), reflex sympathetic dystrophy, brachial plexus avulsion, neurogenic bladder, acute pain (e.g., musculoskeletal and post-operative pain), chronic pain, persistent pain, peripherally mediated pain, centrally mediated pain, chronic headache, migraine headache, familial hemiplegic migraine, conditions associated with cephalic pain, sinus headache, tension headache, phantom limb pain, peripheral nerve injury, pain following stroke, thalamic lesions, radiculopathy, HIV pain, post-herpetic pain, non-cardiac chest pain, irritable bowel syndrome and pain associated with bowel disorders and dyspepsia, and combinations thereof.

Furthermore, sodium channel blockers have clinical uses in addition to pain. The present invention therefore also relates to compounds, pharmaceutical compositions and methods of using the compounds and pharmaceutical compositions for the treatment of diseases or conditions such as cancer and pruritus (itch).

Pruritus, commonly known as itch, is a common dermatological condition. While the exact causes of pruritus are complex and incompletely understood, there has long been evidence that itch involves sensory neurons, especially C fibers, similar to those that mediate pain (Schmelz, M., et al., J. Neurosci. (1997), 17: 8003-8). In particular, it is believed that sodium influx through voltage-gated sodium channels is essential for the propagation of itch sensation from the skin. Transmission of the itch impulses results in the unpleasant sensation that elicits the desire or reflex to scratch.

Multiple causes and electrical pathways for eliciting itch are known. In humans, pruritus can be elicited by histamine or PAR-2 agonists such as mucunain that activate distinct populations of C fibers (Namer, B., et al., J. Neurophysiol. (2008),100: 2062-9). A variety of neurotrophic peptides are known to mediate itch in animal models (Wang, H., and Yosipovitch, G., International Journal of Dermatology (2010), 49: 1-11). Itch can also be elicited by opioids, evidence of distinct pharmacology from that of pain responses.

There exists a complex interaction between itch and pain responses that arises in part from the overlapping sensory input from the skin (Ikoma, A., et al., Arch. Dermatol. (2003),139: 1475-8) and also from the diverse etiology of both pain and pruritus. Pain responses can exacerbate itching by enhancing central sensitization or lead to inhibition of painful scratching. Particularly severe forms of chronic itch occur when pain responses are absent, as in the case of post-herpetic itch (Oaklander, A. L., et al., Pain (2002), 96: 9-12).

The compounds of the invention can also be useful for treating pruritus. The rationale for treating itch with inhibitors of voltage-gated sodium channels, especially NaV1.7, is as follows:

The propagation of electrical activity in the C fibers that sense pruritinergic stimulants requires sodium entry through voltage-gated sodium channels.

NaV1.7 is expressed in the C fibers and kerotinocytes in human skin (Zhao, P., et al., Pain (2008), 139: 90-105).

A gain of function mutation of NaV1.7 (L858F) that causes erythromelalgia also causes chronic itch (Li, Y., et al., Clinical and Experimental Dermatology (2009), 34: e313-e4).

Chronic itch can be alleviated with treatment by sodium channel blockers, such as the local anesthetic lidocaine (Oaklander, A. L., et al., Pain (2002), 96: 9-12; Villamil, A. G., et al., The American Journal of Medicine (2005), 118: 1160-3). In these reports, lidocaine was effective when administered either intravenously or topically (a Lidoderm patch). Lidocaine can have multiple activities at the plasma concentrations achieved when administered systemically, but when administered topically, the plasma concentrations are only about 1 μM (Center for Drug Evaluation and Research NDA 20-612). At these concentrations, lidocaine is selective for sodium channel block and inhibits spontaneous electrical activity in C fibers and pain responses in animal models (Xiao, W. H., and Bennett, G. J. Pain (2008), 137: 218-28). The types of itch or skin irritation, include, but are not limited to:

psoriatic pruritus, itch due to hemodyalisis, aguagenic pruritus, and itching caused by skin disorders (e.g., contact dermatitis), systemic disorders, neuropathy, psychogenic factors or a mixture thereof;

itch caused by allergic reactions, insect bites, hypersensitivity (e.g., dry skin, acne, eczema, psoriasis), inflammatory conditions or injury;

itch associated with vulvar vestibulitis; and

skin irritation or inflammatory effect from administration of another therapeutic such as, for example, antibiotics, antivirals and antihistamines.

The compounds of the invention are also useful in treating certain cancers, such as hormone sensitive cancers, such as prostate cancer (adenocarcinoma), breast cancer, ovarian cancer, testicular cancer and thyroid neoplasia, in a mammal, preferably a human. The voltage gated sodium channels have been demonstrated to be expressed in prostate and breast cancer cells. Up-regulation of neonatal NaV1.5 occurs as an integral part of the metastatic process in human breast cancer and could serve both as a novel marker of the metastatic phenotype and a therapeutic target (Clin. Cancer Res. (2005), August 1; 11(15): 5381-9). Functional expression of voltage-gated sodium channel alpha-subunits, specifically NaV1.7, is associated with strong metastatic potential in prostate cancer (CaP) in vitro. Voltage-gated sodium channel alpha-subunits immunostaining, using antibodies specific to the sodium channel alpha subunit was evident in prostatic tissues and markedly stronger in CaP vs non-CaP patients (Prostate Cancer Prostatic Dis., 2005; 8(3):266-73). See also Diss, J. K. J., et al., Mol. Cell. Neurosci. (2008), 37:537-547 and Kis-Toth, K., et al., The Journal of Immunology (2011), 187:1273-1280.

In consideration of the above, in one embodiment, the present invention provides a method for treating a mammal for, or protecting a mammal from developing, a sodium channel-mediated disease, especially pain, comprising administering to the mammal, especially a human, in need thereof, a therapeutically effective amount of a compound of the invention or a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention wherein the compound modulates the activity of one or more voltage-dependent sodium channels.

In another embodiment of the invention is a method of treating a disease or a condition in a mammal, preferably a human, wherein the disease or condition is selected from the group consisting of pain, depression, cardiovascular diseases, respiratory diseases, and psychiatric diseases, and combinations thereof, and wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of an embodiment of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.

One embodiment of this embodiment is wherein the disease or condition is selected from the group consisting of neuropathic pain, inflammatory pain, visceral pain, cancer pain, chemotherapy pain, trauma pain, surgical pain, post surgical pain, childbirth pain, labor pain, neurogenic bladder, ulcerative colitis, chronic pain, persistent pain, peripherally mediated pain, centrally mediated pain, chronic headache, migraine headache, sinus headache, tension headache, phantom limb pain, peripheral nerve injury, and combinations thereof.

Another embodiment of this embodiment is wherein the disease or condition is selected from the group consisting of pain associated with HIV, HIV treatment induced neuropathy, trigeminal neuralgia, post herpetic neuralgia, eudynia, heat sensitivity, tosarcoidosis, irritable bowel syndrome, Crohns disease, pain associated with multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), diabetic neuropathy, peripheral neuropathy, arthritic, rheumatoid arthritis, osteoarthritis, atherosclerosis, paroxysmal dystonia, myasthenia syndromes, myotonia, malignant hyperthermia, cystic fibrosis, pseudoaldosteronism, rhabdomyolysis, hypothyroidism, bipolar depression, anxiety, schizophrenia, sodium channel toxin related illnesses, familial erythromelalgia, primary erythromelalgia, familial rectal pain, cancer, epilepsy, partial and general tonic seizures, restless leg syndrome, arrhythmias, fibromyalgia, neuroprotection under ischaemic conditions caused by stroke or neural trauma, tachy arrhythmias, atrial fibrillation and ventricular fibrillation.

Another embodiment of the invention is a method of treating, but not preventing, pain in a mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.

One embodiment of this embodiment is a method wherein the pain is selected from the group consisting of neuropathic pain, inflammatory pain, visceral pain, cancer pain, chemotherapy pain, trauma pain, surgical pain, post surgical pain, childbirth pain, labor pain, dental pain, chronic pain, persistent pain, peripherally mediated pain, centrally mediated pain, chronic headache, migraine headache, sinus headache, tension headache, phantom limb pain, peripheral nerve injury, trigeminal neuralgia, post herpetic neuralgia, eudynia, familial erythromelalgia, primary erythromelalgia, familial rectal pain or fibromyalgia, and combinations thereof.

Another embodiment of this embodiment is a method wherein the pain is associated with a disease or condition selected from HIV, HIV treatment induced neuropathy, heat sensitivity, tosarcoidosis, irritable bowel syndrome, Crohns disease, multiple sclerosis, amyotrophic lateral sclerosis, diabetic neuropathy, peripheral neuropathy, rheumatoid arthritis, osteoarthritis, atherosclerosis, paroxysmal dystonia, myasthenia syndromes, myotonia, malignant hyperthermia, cystic fibrosis, pseudoaldosteronism, rhabdomyolysis, hypothyroidism, bipolar depression, anxiety, schizophrenia, sodium channel toxin related illnesses, neurogenic bladder, ulcerative colitis, cancer, epilepsy, partial and general tonic seizures, restless leg syndrome, arrhythmias, ischaemic conditions caused by stroke or neural trauma, tachy arrhythmias, atrial fibrillation and ventricular fibrillation.

Another embodiment of the invention is the method of treating pain in a mammal, preferably a human, by the inhibition of ion flux through a voltage dependent sodium channel in the mammal, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of an embodiment of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.

Another embodiment of the invention is the method of treating pruritus in a mammal, preferably a human, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of an embodiment of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.

Another embodiment of the invention is the method of treating cancer in a mammal, preferably a human, wherein the method comprises administering to the mammal in need thereof a therapeutically effective amount of an embodiment of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, or a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable excipient.

Another embodiment of the invention is the method of decreasing ion flux through a voltage dependent sodium channel in a cell in a mammal, wherein the method comprises contacting the cell with an embodiment of a compound of the invention, as set forth above, as a stereoisomer, enantiomer or tautomer thereof or mixtures thereof, or a pharmaceutically acceptable salt, solvate or prodrug thereof.

Another embodiment of the invention is the method of selectively inhibiting a first voltage-gated sodium channel over a second voltage-gated sodium channel in a mammal, wherein the method comprises administering to the mammal an inhibitory amount of a compound of formula (I), or an embodiment of a compound of formula (I).

Another embodiment of the invention is the method of selectively inhibiting NaV1.7 in a mammal or a mammalian cell as compared to NaV1.5, wherein the method comprises administering to the mammal in need thereof an inhibitory amount of a compound of formula (I) or an embodiment of an embodiment thereof.

For each of the above embodiments described related to treating diseases and conditions in a mammal, the present invention also contemplates relatedly a compound of formula I or an embodiment thereof for the use as a medicament in the treatment of such diseases and conditions.

For each of the above embodiments described related to treating diseases and conditions in a mammal, the present invention also contemplates relatedly the use of a compound of formula I or an embodiment thereof for the manufacture of a medicament for the treatment of such diseases and conditions.

Another embodiment of the invention is a method of using the compounds of formula (I) as standards or controls in in vitro or in vivo assays in determining the efficacy of test compounds in modulating voltage-dependent sodium channels.

In another embodiment of the invention, the compounds of formula (I) are isotopically-labeled by having one or more atoms therein replaced by an atom having a different atomic mass or mass number. Such isotopically-labeled (i.e., radiolabelled) compounds of formula (I) are considered to be within the scope of this invention. Examples of isotopes that can be incorporated into the compounds of formula (I) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine, and iodine, such as, but not limited to, 2H, 3H, 11C, 13C, 14C, 13N, 15N, 15O, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl, 123I, and 125I, respectively. These isotopically-labeled compounds would be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action on the sodium channels, or binding affinity to pharmacologically important site of action on the sodium channels, particularly NaV1.7. Certain isotopically-labeled compounds of formula (I), for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e., 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.

Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.

Substitution with positron emitting isotopes, such as 11C, 18F, 15O and 13N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.

Testing Compounds

The assessment of the compounds of the invention in mediating, especially inhibiting, the sodium channel ion flux can be determined using the assays described hereinbelow. Alternatively, the assessment of the compounds in treating conditions and diseases in humans may be established in industry standard animal models for demonstrating the efficacy of compounds in treating pain. Animal models of human neuropathic pain conditions have been developed that result in reproducible sensory deficits (allodynia, hyperalgesia, and spontaneous pain) over a sustained period of time that can be evaluated by sensory testing. By establishing the degree of mechanical, chemical, and temperature induced allodynia and hyperalgesia present, several physiopathological conditions observed in humans can be modeled allowing the evaluation of pharmacotherapies.

In rat models of peripheral nerve injury, ectopic activity in the injured nerve corresponds to the behavioural signs of pain. In these models, intravenous application of the sodium channel blocker and local anesthetic lidocaine can suppress the ectopic activity and reverse the tactile allodynia at concentrations that do not affect general behavior and motor function (Mao, J. and Chen, L. L, Pain (2000), 87:7-17). Allometric scaling of the doses effective in these rat models, translates into doses similar to those shown to be efficacious in humans (Tanelian, D. L. and Brose, W. G., Anesthesiology (1991), 74(5):949-951). Furthermore, Lidoderm®, lidocaine applied in the form of a dermal patch, is currently an FDA approved treatment for post-herpetic neuralgia (Devers, A. and Glaler, B. S., Clin. J. Pain (2000), 16(3):205-8).

The present invention readily affords many different means for identification of sodium channel modulating agents that are useful as therapeutic agents. Identification of modulators of sodium channel can be assessed using a variety of in vitro and in vivo assays, e.g., measuring current, measuring membrane potential, measuring ion flux, (e.g., sodium or guanidinium), measuring sodium concentration, measuring second messengers and transcription levels, and using e.g., voltage-sensitive dyes, radioactive tracers, and patch-clamp electrophysiology.

One such protocol involves the screening of chemical agents for ability to modulate the activity of a sodium channel thereby identifying it as a modulating agent.

A typical assay described in Bean et al., J. General Physiology (1983), 83:613-642, and Leuwer, M., et al., Br. J. Pharmacol (2004), 141(1):47-54, uses patch-clamp techniques to study the behavior of channels. Such techniques are known to those skilled in the art, and may be developed, using current technologies, into low or medium throughput assays for evaluating compounds for their ability to modulate sodium channel behavior.

Throughput of test compounds is an important consideration in the choice of screening assay to be used. In some strategies, where hundreds of thousands of compounds are to be tested, it is not desirable to use low throughput means. In other cases, however, low throughput is satisfactory to identify important differences between a limited number of compounds. Often it will be necessary to combine assay types to identify specific sodium channel modulating compounds.

Electrophysiological assays using patch clamp techniques is accepted as a gold standard for detailed characterization of sodium channel compound interactions, and as described in Bean et al., op. cit. and Leuwer, M., et al., op. cit. There is a manual low-throughput screening (LTS) method which can compare 2-10 compounds per day; a recently developed system for automated medium-throughput screening (MTS) at 20-50 patches (i.e. compounds) per day; and a technology from Molecular Devices Corporation (Sunnyvale, Calif.) which permits automated high-throughput screening (HTS) at 1000-3000 patches (i.e. compounds) per day.

One automated patch-clamp system utilizes planar electrode technology to accelerate the rate of drug discovery. Planar electrodes are capable of achieving high-resistance, cells-attached seals followed by stable, low-noise whole-cell recordings that are comparable to conventional recordings. A suitable instrument is the PatchXpress 7000A (Axon Instruments Inc, Union City, Calif.). A variety of cell lines and culture techniques, which include adherent cells as well as cells growing spontaneously in suspension are ranked for seal success rate and stability. Immortalized cells (e.g. HEK and CHO) stably expressing high levels of the relevant sodium ion channel can be adapted into high-density suspension cultures.

Other assays can be selected which allow the investigator to identify compounds which block specific states of the channel, such as the open state, closed state or the resting state, or which block transition from open to closed, closed to resting or resting to open. Those skilled in the art are generally familiar with such assays.

Binding assays are also available. Designs include traditional radioactive filter based binding assays or the confocal based fluorescent system available from Evotec OAI group of companies (Hamburg, Germany), both of which are HTS.

Radioactive flux assays can also be used. In this assay, channels are stimulated to open with veratridine or aconitine and held in a stabilized open state with a toxin, and channel blockers are identified by their ability to prevent ion influx. The assay can use radioactive 22[Na] and 14[C] guanidinium ions as tracers. FlashPlate & Cytostar-T plates in living cells avoids separation steps and are suitable for HTS. Scintillation plate technology has also advanced this method to HTS suitability. Because of the functional aspects of the assay, the information content is reasonably good.

Yet another format measures the redistribution of membrane potential using the FLIPR system membrane potential kit (HTS) available from Molecular Dynamics (a division of Amersham Biosciences, Piscataway, N.J.). This method is limited to slow membrane potential changes. Some problems may result from the fluorescent background of compounds. Test compounds may also directly influence the fluidity of the cell membrane and lead to an increase in intracellular dye concentrations. Still, because of the functional aspects of the assay, the information content is reasonably good.

Sodium dyes can be used to measure the rate or amount of sodium ion influx through a channel. This type of assay provides a very high information content regarding potential channel blockers. The assay is functional and would measure Na+ influx directly. CoroNa Red, SBFI and/or sodium green (Molecular Probes, Inc. Eugene Oreg.) can be used to measure Na influx; all are Na responsive dyes. They can be used in combination with the FLIPR instrument. The use of these dyes in a screen has not been previously described in the literature. Calcium dyes may also have potential in this format.

In another assay, FRET based voltage sensors are used to measure the ability of a test compound to directly block Na influx. Commercially available HTS systems include the VIPR™ II FRET system (Life Technologies, or Aurora Biosciences Corporation, San Diego, Calif., a division of Vertex Pharmaceuticals, Inc.) which may be used in conjunction with FRET dyes, also available from Aurora Biosciences. This assay measures sub-second responses to voltage changes. There is no requirement for a modifier of channel function. The assay measures depolarization and hyperpolarizations, and provides ratiometric outputs for quantification. A somewhat less expensive MTS version of this assay employs the FLEXstation™ (Molecular Devices Corporation) in conjunction with FRET dyes from Aurora Biosciences. Other methods of testing the compounds disclosed herein are also readily known and available to those skilled in the art.

Modulating agents so identified are then tested in a variety of in vivo models so as to determine if they alleviate pain, especially chronic pain or other conditions such as cancer and pruritus (itch) with minimal adverse events. The assays described below in the Biological Assays Section are useful in assessing the biological activity of the instant compounds.

Typically, the efficacy of a compound of the invention is expressed by its IC50 value (“Inhibitory Concentration—50%”), which is the measure of the amount of compound required to achieve 50% inhibition of the activity of the target sodium channel over a specific time period. For example, representative compounds of the present invention have demonstrated IC50's ranging from less than 100 nanomolar to less than 10 micromolar in the patch voltage clamp NaV1.7 electrophysiology assay described herein.

In another aspect of the invention, the compounds of the invention can be used in in vitro or in vivo studies as exemplary agents for comparative purposes to find other compounds also useful in treatment of, or protection from, the various diseases disclosed herein.

Another aspect of the invention relates to inhibiting NaV1.1, NaV1.2, NaV1.3, NaV1.4, NaV1.5, NaV1.6, NaV1.7, NaV1.8, or NaV1.9 activity, preferably NaV1.7 activity, in a biological sample or a mammal, preferably a human, which method comprises administering to the mammal, preferably a human, or contacting said biological sample with a compound of formula (I) or a pharmaceutical composition comprising a compound of formula (I). The term “biological sample”, as used herein, includes, without limitation, cell cultures or extracts thereof, biopsied material obtained from a mammal or extracts thereof, and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.

Inhibition of NaV1.1, NaV1.2, NaV1.3, NaV1.4, NaV1.5, NaV1.6, NaV1.7, NaV1.8, or NaV1.9 activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, the study of sodium ion channels in biological and pathological phenomena; and the comparative evaluation of new sodium ion channel inhibitors.

The compounds of the invention (or stereoisomers, geometric isomers, tautomers, solvates, metabolites, isotopes, pharmaceutically acceptable salts, or prodrugs thereof) and/or the pharmaceutical compositions described herein which comprise a pharmaceutically acceptable excipient and one or more compounds of the invention, can be used in the preparation of a medicament for the treatment of sodium channel-mediated disease or condition in a mammal.

Combination Therapy

The compounds of the invention may be usefully combined with one or more other compounds of the invention or one or more other therapeutic agent or as any combination thereof, in the treatment of sodium channel-mediated diseases and conditions. For example, a compound of the invention may be administered simultaneously, sequentially or separately in combination with other therapeutic agents, including, but not limited to:

opiates analgesics, e.g., morphine, heroin, cocaine, oxymorphine, levorphanol, levallorphan, oxycodone, codeine, dihydrocodeine, propoxyphene, nalmefene, fentanyl, hydrocodone, hydromorphone, meripidine, methadone, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine and pentazocine;

non-opiate analgesics, e.g., acetomeniphen, salicylates (e.g., aspirin);

nonsteroidal antiinflammatory drugs (NSAIDs), e.g., ibuprofen, naproxen, fenoprofen, ketoprofen, celecoxib, diclofenac, diflusinal, etodolac, fenbufen, fenoprofen, flufenisal, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofenamic acid, mefenamic acid, meloxicam, nabumetone, naproxen, nimesulide, nitroflurbiprofen, olsalazine, oxaprozin, phenylbutazone, piroxicam, sulfasalazine, sulindac, tolmetin and zomepirac;

anticonvulsants, e.g., carbamazepine, oxcarbazepine, lamotrigine, valproate, topiramate, gabapentin and pregabalin;

antidepressants such as tricyclic antidepressants, e.g., amitriptyline, clomipramine, despramine, imipramine and nortriptyline;

COX-2 selective inhibitors, e.g., celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib, and lumiracoxib;

alpha-adrenergics, e.g., doxazosin, tamsulosin, clonidine, guanfacine, dexmetatomidine, modafinil, and 4-amino-6,7-dimethoxy-2-(5-methane sulfonamido-1,2,3,4-tetrahydroisoquinol-2-yl)-5-(2-pyridyl) quinazoline;

barbiturate sedatives, e.g., amobarbital, aprobarbital, butabarbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, phenobartital, secobarbital, talbutal, theamylal and thiopental;

tachykinin (NK) antagonist, particularly an NK-3, NK-2 or NK-1 antagonist, e.g., (aR, 9R)-7-[3,5-bis(trifluoromethyl)benzyl)]-8,9,10,11-tetrahydro-9-methyl-5-(4-methylphenyl)-7H-[1,4]diazocino[2,1-g][1,7]-naphthyridine-6-13-dione (TAK-637), 5-[[2R,3S)-2-[(1R)-1-[3,5-bis(trifluoromethylphenyl]ethoxy-3-(4-fluorophenyl)-4-morpholinyl]-methyl]-1,2-dihydro-3H-1,2,4-triazol-3-one (MK-869), aprepitant, lanepitant, dapitant or 3-[[2-methoxy5-(trifluoromethoxy)phenyl]-methylamino]-2-phenylpiperidine (2S,3S);

coal-tar analgesics, in particular paracetamol;

serotonin reuptake inhibitors, e.g., paroxetine, sertraline, norfluoxetine (fluoxetine desmethyl metabolite), metabolite demethylsertraline, 3 fluvoxamine, paroxetine, citalopram, citalopram metabolite desmethylcitalopram, escitalopram, d, l-fenfluramine, femoxetine, ifoxetine, cyanodothiepin, litoxetine, dapoxetine, nefazodone, cericlamine, trazodone and fluoxetine;

noradrenaline (norepinephrine) reuptake inhibitors, e.g., maprotiline, lofepramine, mirtazepine, oxaprotiline, fezolamine, tomoxetine, mianserin, buproprion, buproprion metabolite hydroxybuproprion, nomifensine and viloxazine (Vivalan®)), especially a selective noradrenaline reuptake inhibitor such as reboxetine, in particular (S,S)-reboxetine, and venlafaxine duloxetine neuroleptics sedative/anxiolytics;

dual serotonin-noradrenaline reuptake inhibitors, such as venlafaxine, venlafaxine metabolite O-desmethylvenlafaxine, clomipramine, clomipramine metabolite desmethylclomipramine, duloxetine, milnacipran and imipramine;

acetylcholinesterase inhibitors such as donepezil;

5-HT3 antagonists such as ondansetron;

metabotropic glutamate receptor (mGluR) antagonists;

local anaesthetic such as mexiletine and lidocaine;

corticosteroid such as dexamethasone;

antiarrhythimics, e.g., mexiletine and phenytoin;

muscarinic antagonists, e.g., tolterodine, propiverine, tropsium t chloride, darifenacin, solifenacin, temiverine and ipratropium;

cannabinoids;

vanilloid receptor agonists (e.g., resinferatoxin) or antagonists (e.g., capsazepine);

sedatives, e.g., glutethimide, meprobamate, methaqualone, and dichloralphenazone;

anxiolytics such as benzodiazepines,

antidepressants such as mirtazapine,

topical agents (e.g., lidocaine, capsacin and resiniferotoxin);

muscle relaxants such as benzodiazepines, baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine, methocarbamol and orphrenadine;

anti-histamines or H1 antagonists;

NMDA receptor antagonists;

5-HT receptor agonists/antagonists;

PDEV inhibitors;

Tramadol®;

cholinergic (nicotinc) analgesics;

alpha-2-delta ligands;

prostaglandin E2 subtype antagonists;

leukotriene B4 antagonists;

5-lipoxygenase inhibitors; and

5-HT3 antagonists.

Sodium channel-mediated diseases and conditions that may be treated and/or prevented using such combinations include but not limited to, pain, central and peripherally mediated, acute, chronic, neuropathic as well as other diseases with associated pain and other central nervous disorders such as epilepsy, anxiety, depression and bipolar disease; or cardiovascular disorders such as arrhythmias, atrial fibrillation and ventricular fibrillation; neuromuscular disorders such as restless leg syndrome and muscle paralysis or tetanus; neuroprotection against stroke, neural trauma and multiple sclerosis; and channelopathies such as erythromyalgia and familial rectal pain syndrome.

As used herein “combination” refers to any mixture or permutation of one or more compounds of the invention and one or more other compounds of the invention or one or more additional therapeutic agent. Unless the context makes clear otherwise, “combination” may include simultaneous or sequentially delivery of a compound of the invention with one or more therapeutic agents. Unless the context makes clear otherwise, “combination” may include dosage forms of a compound of the invention with another therapeutic agent. Unless the context makes clear otherwise, “combination” may include routes of administration of a compound of the invention with another therapeutic agent. Unless the context makes clear otherwise, “combination” may include formulations of a compound of the invention with another therapeutic agent. Dosage forms, routes of administration and pharmaceutical compositions include, but are not limited to, those described herein.

The invention will be more fully understood by reference to the following examples. They should not, however, be construed as limiting the scope of the invention.

EXAMPLES

These examples serve to provide guidance to a skilled artisan to prepare and use the compounds, compositions and methods of the invention. While particular embodiments of the present invention are described, the skilled artisan will appreciate that various changes and modifications can be made without departing from the spirit and scope of the inventions.

The chemical reactions in the examples described can be readily adapted to prepare a number of other compounds of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention. For example, the synthesis of non-exemplified compounds according to the invention can be successfully performed by modifications apparent to those skilled in the art, for example, by appropriately protecting interfering group, by utilizing other suitable reagents known in the art, for example, by appropriately protecting interfering groups by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions.

In the examples below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Commercially available reagents were purchased from suppliers such as Aldrich Chemical Company, Lancaster, TCI or Maybridge and were used without further purification unless otherwise indicated. The reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried. 1H NMR spectra were obtained in deuterated CDCl3, d6-DMSO, CH3OD or d6-acetone solvent solutions (reported in ppm) using or trimethylsilane (TMS) or residual non-deuterated solvent peaks as the reference standard. When peak multiplicities are reported, the following abbreviates are used: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet, br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, ar reported in

Hz (Hertz).

All abbreviations used to describe reagents, reaction conditions or equipment are intended to be consistent with the definitions set forth in the “List of standard abbreviates and acronyms”. The chemical names of discrete compounds of the invention were obtained using the structure naming feature of ChemDraw naming program.

LCMS Analytical Methods

Final compounds were analyzed using three different LC/MS conditions, with UV detector monitoring at 214 nm and 254 nm, and mass spectrometry scanning 110-800 amu in ESI+ ionization mode.
LC/MS Method A (8.0 min LC-MS Run): XBridge C18 column (4.6×50 mm, 3.5 μm, 40° C.); mobile phase: A=10 mM ammonium hydrogen carbonate in water, B=acetonitrile; gradient: 0.0-8.0 min, 5%-95% B; flow rate=1.2 mL/min.
LC/MS Method B (8.0 min LC-MS Run): XBridge C18 column (4.6×50 mm, 3.5 μm, 40° C.); mobile phase: A=0.1% ammonia in water, B=acetonitrile; gradient: 0.0-8.0 min, 5%-95% B; flow rate=1.2 mL/min.
LC/MS Method C (8.0 min LC-MS Run): XBridge C18 column (4.6×50 mm, 3.5 μm, 40° C.); mobile phase: A=0.1% TFA in water, B=acetonitrile; gradient: 0.0-8.0 min, 5%-95% B; flow rate=1.2 mL/min.
LC/MS Method D: Agilent SB C18, 2.1×30 mm, 1.8 μm; mobile phase: A water (0.05% TFA), B CH3CN (0.05% TFA); gradient: 3% B (0.3 min), followed by 3-95% B (6.5 min), 95% B (1.5 min); flow rate: 0.4 mL/min; oven temperature 25° C.
LC/MS Method E: Acquity BEH C18, 2.1×50 mm, 1.8 μm; mobile phase: A water (0.1% FA), B CH3CN (0.1% FA); gradient: 3% B (0.4 min), followed by 3-95% B (7.5 min), 95% B (0.5 min); flow rate: 0.5 mL/min; oven temperature 25° C.
LC/MS Method F: Agilent SB C18, 2.1×30 mm, 1.8 μm; mobile phase: A water (0.05% TFA), B CH3CN (0.05% TFA); gradient: 3% B (0.3 min), followed by 3-95% B (6.5 min), 95% B (1.5 min); flow rate: 0.4 mL/min; oven temperature 25° C.
LC/MS Method G: Acquity BEH C18, 2.1×50 mm, 1.8 μm; mobile phase: A water (0.1% FA), B CH3CN (0.1% FA); gradient: 3% B (0.4 min), followed by 3-95% B (7.5 min), 95% B (0.5 min); flow rate: 0.5 mL/min; oven temperature 25° C.

Abbreviations

  • MeCN Acetonitrile
  • EtOAc Ethyl acetate
  • DCE Dichloroethane
  • DCM Dichloromethane
  • DIPEA Diisopropylethylamine
  • DEA Diethylamine
  • DMAP 4-dimethylaminopyridine
  • DMF N,N-Dimethylformamide
  • DMSO Dimethylsulfoxide
  • FA Formic acid
  • IPA Isopropyl alcohol
  • TFA Trifluoroacetic acid
  • EDCI 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride
  • HCl Hydrochloric acid
  • HPLC High Pressure Liquid Chromatography
  • LCMS Liquid Chromatography Mass Spectrometry
  • MeOH Methanol
  • NMP N-methyl-2-pyrrolidone
  • RPHPLC Reverse phase high pressure liquid chromatography
  • RT Retention time
  • THF Tetrahydrofuran

Example 1 Synthesis of 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate Step 1. Preparation of 6-fluoro-3-methyl-1H-indazole-5-carbonitrile

To a solution of 5-bromo-6-fluoro-3-methyl-1H-indazole (9.0 g, 39.3 mmol) in N,N-dimethylformamide (50 mL) was added zinc cyanide (10.0 g 85.0 mmol). The flask was degassed under nitrogen and tetrakis(triphenylphosphine)palladium(O) (9.00 g, 7.80 mmol) was added. The reaction mixture was heated at 100° C. for 16 h, cooled to ambient temperature and poured onto mixture of water (100 mL) and saturated sodium chloride (100 mL). The mixture was extracted with dichloromethane (3×100 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo and the residue was purified on a silica column eluting with gradient of 0-100% ethyl acetate containing 10% ammonia in hexanes to afford the title compound as yellow solid (7.00 g, quant. yield): MS (ES+) m/z 177.1 (M+1).

Step 2. Preparation of 6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

A mixture of 6-fluoro-3-methyl-1H-indazole-5-carbonitrile (0.84 g, 4.7 mmol), concentrated hydrochloric acid (10 mL, 37% in water), and dimethyl sulfoxide (50 mL) was heated at 100° C. for 16 h. After cooling to ambient temperature, the precipitate was filtered off and dried under vacuum to afford the title compound as a yellow solid (0.56 g, 61% yield): MS (ES−) m/z 193.2 (M−1).

Step 3. Preparation of methyl 6-fluoro-3-methyl-1H-indazole-5-carboxylate

To a 0° C. mixture of 6-fluoro-3-methyl-1H-indazole-5-carboxylic acid (0.56 g, 2.9 mmol) in methanol (20 mL) was added thionyl chloride (1 mL). The reaction mixture was heated to reflux for 16 h and cooled to ambient temperature. The solvent was concentrated in vacuo. The residue was stirred with diethyl ether and the precipitate was filtered to afford the title compound as a beige solid (0.48 g, 80% yield): MS (ES+) m/z 209.3 (M+1).

Step 4. Preparation of methyl 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

A mixture of methyl 6-fluoro-3-methyl-1H-indazole-5-carboxylate (0.50 g, 2.41 mmol), tert-butyl 4-((methylsulfonyl)oxy)piperidine-1-carboxylate (0.84 g, 2.89 mmol) and cesium carbonate (1.60 g, 4.91 mmol) in N,N-dimethylformamide (10 mL) was heated at 80° C. for 16 h. The reaction was cooled to ambient temperature then water was added (10 mL) and the mixture was extracted with ethyl acetate (3×10 mL). The combined organic layers were dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo and the residue was purified by column chromatography eluting with ethyl acetate in hexanes to afford the title compound as a colorless gum (0.38 g, 40 yield): 1H NMR (300 MHz, CDCl3) δ 8.29 (d, J=6.9 Hz, 1H), 7.65 (d, J=12.2 Hz, 1H), 4.80-4.68 (in, 1H), 4.11-4.06 (m, 2H), 3.81 (s, 3H), 3.19-2.79 (m, 2H), 2.51 (s, 3H), 1.91-1.78 (m, 4H), 1.39 (s, 9H); MS (ES+) m/z 392.4 (M+1).

Step 5. Preparation of methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate

To a solution of methyl 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate (0.38 g, 0.97 mmol) in dichloromethane (2 mL) was added trifluoroacetic acid (1 mL). The reaction solution was stirred at ambient temperature under a nitrogen atmosphere for 20 h. The solution was concentrated in vacuo to afford the title compound as a colorless solid (0.41 g, quant. yield): 1H NMR (300 MHz, DMSO-d6) δ 8.96 (br s, 1H), 8.74 (br s, 1H), 8.33 (d, J=7.0 Hz, 1H), 7.62 (d, J=12.1 Hz, 1H), 4.87-4.79 (m, 1H), 3.83 (s, 3H), 3.42-3.39 (m, 2H), 3.05-2.96 (m, 2H), 2.49 (s, 3H), 2.30-2.16 (m, 2H), 2.05-2.00 (m, 2H); 19F NMR (282 MHz, DMSO-d6) δ −113.5 (s, 3F); MS (ES+) m/z 292.2 (M+1).

Step 6. Preparation of methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

To a 0° C. solution of (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol (0.50 g, 2.20 mmol) and triethylamine (0.6 mL, 4.40 mmol) in dichloromethane (10 mL) was added para-toluenesulfonyl chloride (0.50 g, 2.60 mmol). The reaction solution was stirred for 2 hours at ambient temperature then water (20 mL) was added and the organic layer was separated. The organic layer was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo and the residue was redissolved in N,N-dimethylformamide (3 mL) then methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate (0.29 g, 1.00 mmol) and potassium carbonate (0.55 g, 4.00 mmol) were added. The reaction mixture was heated at 80° C. for 16 h. The reaction was cooled to ambient temperature, then water (10 mL) was added and the mixture was extracted with ethyl acetate (3×10 mL). The combined organic layers were dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo and the residue was purified by column chromatography eluting with a 0-20% gradient of ethyl acetate with 10% isopropanol and 10% triethylamine in hexanes to afford the title compound as a colorless gum (0.25 g, 50% yield): MS (ES+) m/z 502.2, 504.0 (M+1).

Step 7. Preparation of 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

A mixture of methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate (0.25 g, 0.5 mmol) and lithium hydroxide (0.40 g, 5.00 mmol) in tetrahydrofuran (3 mL) and water (3 mL) was heated to reflux for 5 h. After cooling to ambient temperature, the solution was neutralized with 1M hydrochloric acid and extracted with ethyl acetate (3×5 mL). The combined extracts were dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to afford the title compound as a colorless gum (0.08 g, 31% yield): MS (ES+) m/z 488.2, 490.1 (M+1).

Step 8. Preparation of 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

A mixture of 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid (0.0.8 g, 0.15 mmol), 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (0.055 g, 0.35 mmol), 4-dimethylaminopyridine (0.043 g, 0.35 mmol) and methanesulfonamide (0.022 g, 0.23 mmol) in dichloromethane (3 mL) was stirred at a ambient temperature for 16 h. The reaction mixture was diluted with ethyl acetate (5 mL), washed with brine (10 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo and the residue was purified by reverse-phase HPLC, eluting with a 20-80% gradient of acetonitrile in water with 0.1% trifluoroacetic acid to afford the title compound as a colorless solid (0.050 g, 58% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.16 (br s, 1H), 10.10 (br s, 1H), 8.37-8.28 (m, 1H), 8.20 (d, J=9.0 Hz, 1H), 8.13-8.07 (m, 1H), 7.61 (d, J=9.0 Hz, 1H), 4.81 (br s, 1H), 4.54 (br s, 2H), 3.62 (br s, 2H), 3.39 (s, 3H), 3.28-3.12 (m, 2H), 2.51 (s, 3H), 2.44-2.23 (m, 2H), 2.20-2.08 (m, 2H); MS (ES+) m/z 565.1, 567.2 (M+1).

Example 2 Synthesis of 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.056 g, 63% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.10 (br s, 1H), 10.03 (br s, 1H), 8.34-8.28 (m, 1H), 8.19 (d, J=9.0 Hz, 1H), 8.13-8.06 (m, 1H), 7.62 (d, J=12.0 Hz, 1H), 4.80 (br s, 2H), 4.53 (br s, 2H), 3.62 (br s, 3H), 3.28-3.05 (m, 3H), 2.44-2.03 (m, 5H), 1.22-1.06 (m, 4H); MS (ES+) m/z 592.1, 594.1 (M+1).

Example 3 Synthesis of N-(azetidin-1-ylsulfonyl)-1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace methanesulfonamide with azetidine-1-sulfonamide, the title compound was obtained as a colorless solid (0.040 g, 44% yield): 1H NMR (300 MHz, DMSO-d6) δ 11.88 (br s, 1H), 9.99 (br s, 1H), 8.35-8.24 (m, 1H), 8.19 (d, J=6.0 Hz, 1H), 8.13-8.06 (m, 1H), 7.62 (d, J=12.0 Hz, 1H), 4.79 (br s, 1H), 4.53 (br s, 2H), 4.09 (t, J=6.0 Hz, 4H), 3.34-2.95 (m, 5H), 2.41-2.04 (m, 8H); MS (ES+) m/z 607.1 (M+1).

Example 4 Synthesis of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate Step 1. Preparation of methyl 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (3-chloro-5-(trifluoromethoxy)phenyl)methanol, the title compound was obtained as a colorless gum (0.07 g, 45% yield): MS (ES+) m/z 500.2, 501.9 (M+1).

Step 2. Preparation of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid (16)

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)-piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.050 g, 73% yield): MS (ES+) m/z 486.1, 488.2 (M+1).

Step 3. Preparation of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, the title compound was obtained as a colorless solid (0.006 g, 11% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.10 (br s, 1H), 10.01 (br s, 1H), 8.15 (d, J=6.0 Hz, 1H), 7.68-7.66 (m, 3H), 7.63 (d, J=12.0 Hz, 1H), 4.81 (br s, 1H), 4.54 (br s, 2H), 3.62 (br s, 2H), 3.39 (s, 3H), 3.28-3.12 (m, 2H), 2.51 (s, 3H), 2.44-2.23 (m, 2H), 2.20-2.08 (m, 2H); MS (ES+) m/z 563.0, 565.0 (M+1).

Example 5 Synthesis of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.12 g, 65% yield): 1H NMR (300 MHz, DMSO-d6) δ 8.14 (d, J=6.0 Hz, 1H), 7.67 (d, J=12.0 Hz, 1H), 7.53 (s, 1H), 7.49 (s, 1H), 7.40 (s, 1H), 7.07-6.83 (m, 1H), 4.65-4.49 (m, 1H), 3.73 (s, 2H), 3.15-2.93 (m, 3H), 2.51 (s, 3H), 2.40-2.20 (m, 2H), 2.22-2.05 (m, 2H), 1.96-1.82 (m, 2H), 1.18-1.05 (m, 4H); MS (ES+) m/z 590.0, 592.0 (M+1).

Example 6 Synthesis of N-(cyclopropylsulfonyl)-1-(1-(3,5-dichlorobenzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate Step 1. Preparation of methyl 1-(1-(3,5-dichlorobenzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (3,5-dichlorophenyl)methanol, the title compound was obtained as a colorless gum (0.097 g, 80% yield): MS (ES+) m/z 452.2, 450.2 (M+1).

Step 2. Preparation of 1-(1-(3,5-dichlorobenzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(3,5-dichlorobenzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.097 g, 81% yield): MS (ES+) m/z 437.1, 439.1 (M+1).

Step 3. Preparation of N-(cyclopropylsulfonyl)-1-(1-(3,5-dichlorobenzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(3,5-dichlorobenzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.012 g, 13% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.07 (br s, 1H), 9.87 (br s, 1H), 8.15 (d, J=9.0 Hz, 1H), 7.75 (s, 1H), 7.70-7.60 (m, 2H), 7.06 (d, J=12.0 Hz, 1H), 4.80 (br s, 2H), 4.31 (br s, 3H), 3.60-3.40 (m, 2H), 3.15-2.87 (m, 4H), 2.38-2.01 (m, 4H), 1.21-1.04 (m, 4H); MS (ES+) m/z 539.1, 541.1 (M+1).

Example 7 Synthesis of 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate Step 1. Preparation of methyl 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (2-bromo-5-(trifluoromethoxy)phenyl)methanol, the title compound was obtained as a colorless gum (0.32 g, 60% yield): MS (ES+) m/z 544.1, 546.1 (M+1).

Step 2. Preparation of 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.10 g, 31% yield): MS (ES+) m/z 530.1, 531.2 (M+1).

Step 3. Preparation of 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.03 g, 25% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.06 (br s, 1H), 9.80 (br s, 1H), 8.15 (d, J=6.0 Hz, 1H), 7.93-7.85 (m, 1H), 7.83-7.72 (m, 1H), 7.60 (d, J=12.0 Hz, 1H), 7.48-7.41 (m, 1H), 4.82 (br s, 2H), 4.50 (br s, 3H), 3.44-2.92 (m, 5H), 2.39-1.90 (m, 5H), 1.18-1.03 (m, 4H); MS (ES+) m/z 633.0, 635.0 (M+1).

Example 8 Synthesis of 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide Step 1. Preparation of methyl 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

The Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (5-chloro-2-(trifluoromethyl)phenyl)methanol, the title compound was obtained as a colorless gum (0.19 g, 80% yield): MS (ES+) m/z 483.2, 485.1 (M+1).

Step 2. Preparation of 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(2-bromo-5-(trifluoromethoxy)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.12 g, 67% yield): MS (ES+) m/z 470.2, 472.1 (M+1).

Step 3. Preparation of 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.17 g, 58% yield): 1H NMR (300 MHz, DMSO-d6) δ 11.75 (s, 1H), 8.16 (d, J=6.9 Hz, 1H), 7.89 (s, 1H), 7.79-7.66 (m, 2H), 7.67-7.54 (m, 1H), 4.68-4.52 (m, 1H), 3.71 (s, 2H), 3.17-3.04 (m, 1H), 2.53 (s, 3H), 2.41-2.22 (m, 2H), 2.22-2.05 (m, 2H), 1.95-1.82 (m, 2H), 1.17-1.06 (m, 3H), 0.94-0.83 (m, 3H); MS (ES+) m/z 573.2, 575.1 (M+1).

Example 9 Synthesis of (R)-N-(cyclopropylsulfonyl)-1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide Step 1. Preparation of (S)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate

To a solution of borane dimethyl sulfide complex (1.2 mL, 13.0 mmol) in anhydrous tetrahydrofuran (60 mL) was added a 1 M solution of (S)-2-methyl-CBS-oxazaborolidine in toluene (2.1 mL, 2.1 mmol). After stirring at ambient temperature for 45 minutes a solution of 1-(3,5-dichlorophenyl)ethanone (2.0 g, 10.6 mmol) in anhydrous tetrahydrofuran (10 mL) was slowly added. After stirring at ambient temperature for 16 h, the reaction mixture was quenched with methanol (20 mL), concentrated to 20% of its initial volume, diluted with water (100 mL) and extracted with dichloromethane (3×25 mL). The combined organic layers were washed with 1 M hydrochloric acid (80 mL), water (80 mL), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to give the corresponding alcohol which was used directly for the next step. To a solution of the alcohol (0.41 g, 2.12 mmol) in anhydrous dichloromethane (10 mL) was added para-toluenesulfonyl chloride (0.82 g, 4.27 mmol) and triethylamine (1.5 mL, 11.0 mmol). The reaction mixture was stirred at ambient temperature for 4 h then diluted with dichloromethane (100 mL), washed with water (100 mL) and brine (100 mL). The organic layer was dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography eluting with a 0-10% gradient of ethyl acetate in hexanes to afford the title compound as a colorless solid (0.19 g, 27% yield): MS (ES+) m/z 173.0, 175.0 (M−171).

Step 2. Preparation of (R)-methyl 1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

To a solution of methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate hydrochloride (0.14 g, 0.41 mmol) and (S)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate (0.17 g, 0.49 mmol) in anhydrous N,N-dimethylformamide (5 mL) was added potassium carbonate (0.20 g, 1.4 mmol). The reaction mixture was heated to 90° C. for 16 h, then a further portion of (S)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate (0.061 g, 0.18 mmol) was added. After heating at 90° C. for 5 h, the mixture was cooled to ambient temperature, diluted with water (100 mL), and extracted with ethyl acetate (2×100 mL). The combined organic layers were dried over anhydrous magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified by column chromatography, eluting with a 0-20% gradient of ethyl acetate with 10% isopropanol and 10% triethylamine in hexanes to afford the title compound as a light yellow syrup (0.15 g, 80% yield): 1H NMR (300 MHz, CDCl3) δ 8.30 (d, J=6.8 Hz, 1H), 7.26-7.22 (m, 3H), 7.03 (d, J=11.5 Hz, 1H), 4.20-4.11 (m, 1H), 3.93 (s, 3H), 3.44 (q, J=6.7, 6.7, 6.7 Hz, 1H), 3.17-3.12 (m, 1H), 2.95-2.90 (m, 1H), 2.55 (s, 3H), 2.38-2.08 (m, 4H), 1.98-1.85 (m, 2H) 1.33 (d, J=6.7 Hz, 3H); 19F NMR (282 MHz, CDCl3) δ −112.8 (s, 1F); MS (ES+) m/z 464.1, 466.1 (M+1).

Step 3. Preparation of (R)-N-(cyclopropylsulfonyl)-1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

To a solution of (R)-methyl 1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate (0.15 g, 0.33 mmol) in tetrahydrofuran (10 mL) was added water (5 mL) and lithium hydroxide (0.084 g, 3.5 mmol). The reaction mixture was heated to reflux for 18 h, then cooled to ambient temperature. The reaction mixture was acidified with 1 M hydrochloric acid (5 mL) and diluted with a mixture of saturated aqueous ammonium chloride and water (100 mL). The mixture was extracted with ethyl acetate (3×75 mL) and the combined organic extracts were dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The residue was dissolved in anhydrous dichloromethane (10 mL) and anhydrous tetrahydrofuran (5 mL) then N-(3-dimethylaminopropyl)-N-ethylcarbodiimide hydrochloride (0.20 g, 1.0 mmol), 4-(dimethylamino)pyridine (0.12 g, 0.97 mmol) and cyclopropanesulfonamide (0.10 g, 0.83 mmol) were added. The solution was stirred at ambient temperature for 23 h then diluted with ethyl acetate (100 mL), washed with saturated aqueous ammonium chloride (100 mL), brine (2×100 mL), dried over anhydrous magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by reverse-phase HPLC, eluting with a 20-80% gradient of acetonitrile in water with 0.1% trifluoroacetic acid to afford the title compound as a colorless solid (0.014 g, 6% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.03 (br s, 1H), 9.73 (br s, 1H), 8.14 (d, J=6.8 Hz, 1H), 7.76-7.68 (m, 3H), 7.54 (d, J=11.4 Hz, 1H), 4.77-4.59 (m, 2H), 3.74-3.64 (m, 1H), 3.53-3.42 (m, 1H), 3.11-3.03 (m, 1H), 2.92-2.76 (m, 2H), 2.46 (s, 3H), 2.34-2.06 (m, 4H), 1.64 (br s, 3H), 1.12-1.09 (m, 4H); 19F NMR (282 MHz, DMSO-d6) δ −73.5 (s, 3F), −115.6 (s, 1F); MS (ES+) m/z 553.0, 555.0 (M+1).

Example 10 Synthesis of (S)—N-(cyclopropylsulfonyl)-1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide Step 1. Preparation of (S)-methyl 1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 9 (Step 2) and making non-critical variations as required to replace (S)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate with (R)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate, the title compound was obtained as light yellow syrup (0.17 g, 90% yield): 1H NMR (300 MHz, CDCl3) δ 8.30 (d, J=6.8 Hz, 1H), 7.25-7.22 (m, 3H), 7.03 (d, J=11.5 Hz, 1H), 4.21-4.12 (m, 1H), 3.93 (s, 3H), 3.44 (q, J=6.7, 6.7, 6.7 Hz, 1H), 3.16-3.11 (m, 1H), 2.94-2.90 (m, 1H), 2.55 (s, 3H), 2.38-2.08 (m, 4H), 1.98-1.85 (m, 2H) 1.32 (d, J=6.7 Hz, 3H); 19F NMR (282 MHz, CDCl3) δ −112.8 (s, 1F); MS (ES+) m/z 464.1, 466.1 (M+1).

Step 2. Preparation of (S)—N-(cyclopropylsulfonyl)-1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the procedure as described in EXAMPLE 9 (Step 3) and making non-critical variations as required to replace (R)-methyl 1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with (S)-methyl 1-(1-(1-(3,5-dichlorophenyl)ethyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as a colorless solid as a trifluoroacetic acid salt (0.020 g, 8% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.02 (br s, 1H), 9.84 (br s, 1H), 8.14 (d, J=6.8 Hz, 1H), 7.75-7.68 (m, 3H), 7.54 (d, J=11.5 Hz, 1H), 4.77-4.56 (m, 2H), 3.70-3.63 (m, 1H), 3.51-3.44 (m, 1H), 3.11-3.03 (m, 1H), 2.88-2.76 (m, 2H), 2.46 (s, 3H), 2.34-2.07 (m, 4H), 1.64 (d, J=5.5 Hz, 3H), 1.12-1.09 (m, 4H); 19F NMR (282 MHz, DMSO-d6) δ −73.5 (s, 3F), −115.7 (s, 1F); MS (ES+) m/z 553.0, 555.0 (M+1).

Example 11 Synthesis of (R)-1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate (26) Step 1. Preparation of (R)-methyl 1-(1-(tert-butoxycarbonyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 4) and making non-critical variations as required to replace tert-butyl 4-((methylsulfonyl)oxy)piperidine-1-carboxylate with tert-butyl 3-((methylsulfonyl)oxy)pyrrolidine-1-carboxylate, the title compound was obtained as a colorless gum (0.50 g, 21% yield): MS (ES+) m/z 378.2 (M+1).

Step 2. Preparation of (R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 5) and making non-critical variations as required to replace methyl 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with (R)-methyl 1-(1-(tert-butoxycarbonyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as a yellow oil (2.51 g, quant. yield): 1H NMR (300 MHz, CDCl3) δ 9.61 (s, 1H), 8.39 (d, J=6.7 Hz, 1H), 7.82 (s, 1H), 7.10 (d, J=10.8 Hz, 1H), 5.33-5.26 (m, 1H), 3.99 (s, 3H), 3.96-3.69 (m, 4H), 2.78-2.62 (m, 1H), 2.56 (s, 3H), 2.38-2.26 (m, 1H); MS (ES+) m/z 278.2 (M+1).

Step 3. Preparation of (R)-methyl 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (3-chloro-5-(trifluoromethoxy)phenyl)methanol and to replace methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate with (R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate, the title compound was obtained as a colorless gum (0.10 g, 29% yield): MS (ES+) m/z 486.1, 488.2 (M+1).

Step 4. Preparation of (R)-1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with (R)-methyl 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.075 g, 80% yield): MS (ES+) m/z 472.0, 474.1 (M+1).

Step 5. Preparation of (R)-1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with (R)-1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.030 g, 33% yield): 1H NMR (300 MHz, DMSO-d6) δ 11.97 (br s, 1H), 9.72 (br s, 1H), 8.15, (d, J=9.0 Hz, 1H), 7.79 (s, 1H), 7.72-7.59 (m, 2H), 7.34 (s, 1H), 5.50 (br s, 2H), 4.50 (br s, 3H), 3.82-3.18 (m, 6H), 2.87-2.17 (m, 2H), 1.22-1.04 (m, 4H); MS (ES+) m/z 575.1, 577.2 (M+1).

Example 12 Synthesis of (R)-1-(1-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate Step 1. Preparation of (R)-methyl 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate with (R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate and to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (5-chloro-2-(trifluoromethyl)phenyl)methanol, the title compound was obtained as a colorless solid (0.14 g, 29% yield): MS (ES+) m/z 470.1, 472.1 (M+1).

Step 2. Preparation of (R)-1-(1-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with (R)-methyl 1-(1-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.12 g, 88% yield): MS (ES+) m/z 456.2, 458.1 (M+1).

Step 3. Preparation of (R)-1-(1-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with (R)-1-(1-(5-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.025 g, 17% yield): 1H NMR (300 MHz, DMSO-d6) δ 11.99 (br s, 1H), 8.15 (d, J=6.0 Hz, 1H), 7.97 (br s, 1H), 7.80-7.73 (m, 1H), 7.67 (d, J=12.0 Hz, 1H), 7.63-7.58 (m, 1H), 5.38 (br s, 1H), 4.10 (br s, 2H), 3.20-2.80 (m, 5H), 2.52 (s, 3H), 2.17 (br s, 2H), 1.18-1.02 (m, 5H); MS (ES+) m/z 559.1, 561.1 (M+1).

Example 13 Synthesis of 1-(1-(5-chloro-6-(trifluoromethyl)pyridin-2-yl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate Step 1. Preparation of methyl 1-(1-(5-chloro-6-(trifluoromethyl)pyridin-2-yl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

In a 20 mL microwave vial methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate (0.29 g, 1.00 mmol), 3,6-dichloro-2-(trifluoromethyl)pyridine (0.32 g, 1.50 mmol), cesium carbonate (1.30 g, 4.00 mmol) and toluene (10 mL) were added. The reaction mixture was degassed with nitrogen then (2,2′-bis(diphenylphosphino)-1,1′-binaphthyl) (BINAP) (0.12 g, 0.20 mmol) and tris(dibenzylideneacetone)dipalladium(O) (0.12 g, 0.20 mmol) were added. The reaction mixture was heated at 100° C. for 16 h, cooled to ambient temperature, filtered through a pad of diatomaceous earth and the filtrate was concentrated in vacuo. The residue was purified by column chromatography eluting with ethyl acetate in hexanes to afford the title compound as pale yellow solid (0.15 g, 32% yield): MS (ES+) m/z 471.1, 473.1 (M+1).

Step 2. Preparation of 1-(1-(5-chloro-6-(trifluoromethyl)pyridin-2-yl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic Acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(5-chloro-6-(trifluoromethyl)pyridin-2-yl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as a colorless solid (0.10 g, 69% yield): MS (ES+) m/z 456.2, 458.1 (M+1).

Step 3. Preparation of 1-(1-(5-chloro-6-(trifluoromethyl)pyridin-2-yl)piperidin-4-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(5-chloro-6-(trifluoromethyl)pyridin-2-yl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.050 g, 40% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.06 (br s, 1H), 8.16 (d, J=6.0 Hz, 1H), 7.85 (d, J=9.0 Hz, 1H), 7.74 (d, J=12.0 Hz, 1H), 7.27 (d, J=9.0 Hz, 1H), 5.24 (br s, 2H), 4.99-4.82 (m, 2H), 3.22-3.05 (m, 3H), 2.49 (s, 3H), 2.08-1.88 (m, 4H), 1.20-1.09 (m, 4H); MS (ES+) m/z 560.0, 562.0 (M+1).

Example 14 Synthesis of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide Step 1. Preparation of methyl 1-(1-(tert-butoxycarbonyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 4) and making non-critical variations as required to replace tert-butyl 4-((methylsulfonyl)oxy)piperidine-1-carboxylate with tert-butyl 3-iodoazetidine-1-carboxylate, the title compound was obtained as a pale yellow solid (2.34 g, 66% yield): 1H NMR (300 MHz, CDCl3) δ 8.31 (d, J=6.8 Hz, 1H), 7.04 (d, J=11.1 Hz, 1H), 5.23-5.14 (m, 1H), 4.48-4.36 (m, 4H), 3.93 (s, 3H), 2.56 (s, 3H), 1.46 (s, 9H); 19F NMR (282 MHz, CDCl3) δ −111.4 (s, 1F); MS (ES+) m/z 364.1 (M+1).

Step 2. Preparation of methyl 1-(azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate hydrochloride

Following the procedure as described in EXAMPLE 1 (Step 5) and making non-critical variations as required to replace methyl 1-(1-(tert-butoxycarbonyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(tert-butoxycarbonyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate and to replace trifluoroacetic acid with 4 M solution of hydrochloric acid in 1,4-dioxane, the title compound was obtained as a colorless solid (2.26 g, quant. yield): 1H NMR (300 MHz, DMSO-d6) δ□9.67 (br s, 1H), 9.37 (br s, 1H), 8.30 (d, J=6.9 Hz, 1H), 7.40 (d, J=12.0 Hz, 1H), 5.80-5.70 (m, 1H), 4.19-3.92 (m, 4H), 3.53 (s, 3H), 2.56 (s, 3H); 19F NMR (282 MHz, DMSO-d6) δ −112.7 (s, 1F); MS (ES+) m/z 264.1 (M+1).

Step 3. Preparation of methyl 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 1 (step 6) and making non-critical variations as required to replace methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate with methyl 1-(azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate hydrochloride and to replace (3-chloro-2-fluoro-5-(trifluoromethyl)phenyl)methanol with (3-chloro-5-(trifluoromethoxy)phenyl)methanol, the title compound as a colorless syrup (1.16 g, 33% yield): MS (ES+) m/z 472.0, 474.0 (M+1).

Step 4. Preparation of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.48 g, 42% yield): MS (ES+) m/z 458.0, 460.0 (M+1).

Step 5. Preparation of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, the title compound as a colorless solid (0.11 g, 31% yield): 1H NMR (300 MHz, DMSO-d6) □□11.73 (br s, 1H), 10.93 (br s, 1H), 8.20 (d, J=6.8 Hz, 1H), 7.72 (s, 1H), 7.68 (s, 1H), 7.60-7.58 (m, 2H), 5.66 (br s, 1H), 4.55-4.48 (m, 6H), 3.36 (s, 3H), 2.56 (s, 3H); 19F NMR (282 MHz, DMSO-d6) δ −56.9 (s, 3F), −73.8 (s, 3F), −115.0 (s, 1F); MS (ES+) m/z 534.9, 536.9 (M+1).

Example 15 Synthesis of 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-(1-(3-chloro-5-(trifluoromethoxy)benzyl)azetidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid as a trifluoroacetic acid salt (0.10 g, 29% yield): 1H NMR (300 MHz, DMSO-d6) □□11.71 (br s, 1H), 10.96 (br s, 1H), 8.19 (d, J=6.8 Hz, 1H), 7.71 (s, 1H), 7.66 (s, 1H), 7.62-7.59 (m, 2H), 5.66 (br s, 1H), 4.52-4.49 (m, 6H), 3.13-3.04 (m, 1H), 2.57 (s, 3H), 1.13-1.10 (m, 4H); 19F NMR (282 MHz, DMSO-d6) δ −56.9 (s, 3F), −73.7 (s, 3F), −114.9 (s, 1F); MS (ES+) m/z 560.9, 562.9 (M+1).

Example 16 Synthesis of 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide, trifluoroacetic acid salt Step 1. Preparation of methyl 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 9 (step 2) and making non-critical variations as required to replace methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate hydrochloride with (R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate 2,2,2-trifluoroacetate, the title compound was obtained as a colorless oil (0.33 g, 62% yield): MS (ES+) m/z 450.0, 452.0 (M+1).

Step 2. Preparation of 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as a colorless solid (0.32 g, 99% yield): MS (ES+) m/z 436.0, 438.0 (M+1).

Step 3. Preparation of 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide, trifluoroacetic acid salt

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, the title compound was obtained as a colorless solid (0.082 g, 43% yield): 1H NMR (300 MHz, DMSO-d6+1% D2O) δ 11.90 (s, 1H), 8.21 (d, J=6.8 Hz, 1H), 7.72-7.61 (m, 4H), 5.58-5.44 (m, 1H), 4.67-4.56 (m, 1H), 4.02-3.67 (m, 4H), 3.50-3.42 (m, 1H), 3.39 (s, 3H), 2.55 (s, 3H), 2.43-2.35 (m, 1H), 1.65 (d, J=6.7 Hz, 3H); MS (ES+) m/z 513.0, 515.0 (M+1).

Example 17 Synthesis of N-(cyclopropylsulfonyl)-1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide 2,2,2-trifluoroacetate

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-((R)-1-((R)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.070 g, 35% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.13 (s, 1H), 10.88 (s, 1H), 8.21 (d, J=6.8 Hz, 1H), 7.73-7.62 (m, 4H), 5.68-5.41 (m, 1H), 4.72-4.50 (m, 1H), 4.13-3.30 (m, 5H), 3.16-3.07 (m, 1H), 2.56 (s, 3H), 2.43-2.27 (m, 1H), 1.65 (d, J=6.8 Hz, 3H), 1.17-1.12 (m, 4H); MS (ES+) m/z 539.0, 541.0 (M+1).

Example 18 Synthesis of 1-((R)-1-((S)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide, trifluoroacetic acid salt

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-((R)-1-((S)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, the title compound was obtained as a colorless solid (0.058 g, 32% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.22 (s, 1H), 10.81 (s, 1H), 8.21 (d, J=6.5 Hz, 1H), 7.74-7.69 (m, 4H), 5.56-5.47 (m, 1H), 4.70-4.56 (m, 1H), 3.87-3.56 (m, 5H), 3.39 (s, 3H), 2.55 (s, 3H), 2.44-2.32 (m, 1H), 1.65 (d, J=6.1 Hz, 3H); MS (ES+) m/z 513.1, 515.0 (M+1).

Example 19 Synthesis of N-(cyclopropylsulfonyl)-1-((R)-1-((S)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide, trifluoroacetic acid salt

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-((R)-1-((S)-1-(3,5-dichlorophenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.064 g, 33% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.10 (s, 1H), 10.58 (s, 1H), 8.17 (d, J=6.9 Hz, 1H), 7.70-7.65 (m, 4H), 5.54-5.43 (m, 1H), 4.64-4.51 (m, 1H), 3.75-3.43 (m, 5H), 3.13-3.04 (m, 1H), 2.52 (s, 3H), 2.40-2.26 (m, 1H), 1.65-1.57 (m, 3H), 1.12-1.10 (m, 4H); MS (ES+) m/z 539.1, 541.0 (M+1).

Example 20 Synthesis of (R)-1-(1-(4-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide, trifluoroacetic acid salt Step 1. Preparation of (R)-methyl 1-(1-(4-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 9 (step 2) and making non-critical variations as required to replace methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate hydrochloride with ((R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate, trifluoroacetic acid salt and to replace (S)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate with (4-chloro-1-(chloromethyl)-2-(trifluoromethyl)benzene, the title compound was obtained as a yellow oil (0.07 g, 19% yield): 1H NMR (300 MHz, CDCl3) δ 8.30 (d, J=6.8 Hz, 1H), 7.79 (d, J=8.4 Hz, 1H), 7.60 (d, J=2.0 Hz, 1H), 7.50 (dd, J=1.9, 8.3 Hz, 1H), 7.36 (d, J=11.7 Hz, 1H), 5.19-5.03 (m, 1H), 3.94 (s, 3H), 3.89 (d, J=14.9 Hz, 1H), 3.80 (d, J=14.6 Hz, 1H), 3.13-2.90 (m, 3H), 2.77-2.63 (m, 1H), 2.55 (s, 3H), 2.51-2.23 (m, 2H); MS m/z 472.1, 470.0 (M+1).

Step 2. Preparation of (R)-1-(1-(4-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, hydrochloric acid salt

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl R)-methyl 1-(1-(4-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as a colorless solid (0.07 g, quant. yield): MS (ES+) m/z 456.1, 458.0 (M+1)

Step 3. Preparation of (R)-1-(1-(4-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide, trifluoroacetic acid salt

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with (R)-1-(1-(4-chloro-2-(trifluoromethyl)benzyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, hydrochloric acid salt and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.008 g, 8% yield): 1H NMR (300 MHz, DMSO-d6) δ 8.21 (d, J=6.8 Hz, 1H), 7.99-7.90 (m, 3H), 7.66 (d, J=11.5 Hz, 1H), 5.56 (br, s, 1H), 4.69 (s, 2H), 3.91 (s, 1H), 3.43-3.29 (m, 1H), 3.80-3.52 (m, 2H), 3.45-3.37 (m, 1H), 3.18-3.03 (m, 1H), 2.56 (s, 3H), 2.38-2.22 (m, 1H), 1.24-1.08 (m, 4H); MS (ES+) m/z 559.1, 561.1 (M+1).

Example 21 Synthesis of (R)-N-(cyclopropylsulfonyl)-1-(1-(3,5-dichlorobenzoyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide Step 1. Preparation of (R)-methyl 1-(1-(3,5-dichlorobenzoyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

To a solution of ((R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate, trifluoroacetic acid salt (0.89 g, 2.27 mmol) in tetrahydrofuran (4.5 mL) and saturated aqueous sodium bicarbonate (4.5 mL) was added 3,5-dichlorobenzoyl chloride (0.52 g, 2.50 mmol) at 0° C. The reaction mixture was warmed to ambient temperature and stirred for 16 h. The reaction was extracted with dichloromethane (3×15 mL) and the combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography eluting with a 10-50% gradient of ethyl acetate in hexanes to afford the title compound as a colorless solid (0.80 g, 78% yield): 1H NMR (300 MHz, CDCl3) δ 8.41-8.28 (m, 1H), 7.49-7.32 (m, 3H), 7.09-6.95 (m, 1H), 5.15-4.91 (m, 1H), 4.19-4.05 (m, 1H), 4.04-3.98 (m, 1H), 3.98-3.92 (m, 3H), 3.91-3.54 (m, 2H), 2.60-2.55 (m, 3H), 2.55-2.33 (m, 2H); MS m/z 452.1, 450.1 (M+1).

Step 2. Preparation of (R)-1-(1-(3,5-dichlorobenzoyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with (R)-methyl 1-(1-(3,5-dichlorobenzoyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.70 g, 100%): MS (ES−) m/z 436.0, 434.3 (M+1).

Step 3. Preparation of (R)-N-(cyclopropylsulfonyl)-1-(1-(3,5-dichlorobenzoyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.084 g, 9% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.16-12.02 (m, 1H), 8.23-8.12 (m, 1H), 7.80-7.60 (m, 2H), 7.60-7.53 (m, 2H), 5.49-5.25 (m, 1H), 4.02-3.51 (m, 4H), 3.17-3.05 (m, 1H), 2.56-2.52 (m, 3H), 2.46-2.31 (m, 2H), 1.24-1.08 (m, 4H); MS (ES−) m/z 537.2, 539.2 (M−1).

Example 22 Synthesis of 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide, trifluoroacetic acid salt Step 1. Preparation of methyl 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate

Following the procedure as described in EXAMPLE 9 (step 1) and making non-critical variations as required to replace methyl 6-fluoro-3-methyl-1-(piperidin-4-yl)-1H-indazole-5-carboxylate hydrochloride with ((R)-methyl 6-fluoro-3-methyl-1-(pyrrolidin-3-yl)-1H-indazole-5-carboxylate, trifluoroacetic acid salt and to replace (S)-1-(3,5-dichlorophenyl)ethyl 4-methylbenzenesulfonate with (S)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl 4-methylbenzenesulfonate, the title compound was obtained as a yellow oil (0.06 g, 40% yield): MS (ES+) m/z 499.8, 502.1 (M+1).

Step 2. Preparation of 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid

Following the procedure as described in EXAMPLE 1 (step 7) and making non-critical variations as required to replace methyl 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate with methyl 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylate, the title compound was obtained as colorless solid (0.06 g, 100% yield): MS (ES+) m/z 486.1, 488.0 (M+1).

Step 3. Preparation of 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-N-(cyclopropylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide, trifluoroacetic acid salt

Following the procedure as described in EXAMPLE 1 (step 8) and making non-critical variations as required to replace 1-(1-(3-chloro-2-fluoro-5-(trifluoromethyl)benzyl)piperidin-4-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid with 1-((R)-1-((R)-1-(3-chloro-5-(trifluoromethoxy)phenyl)ethyl)pyrrolidin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxylic acid, hydrochloric acid salt and to replace methanesulfonamide with cyclopropanesulfonamide, the title compound was obtained as a colorless solid (0.004 g, 6% yield): 1H NMR (300 MHz, DMSO-d6) δ 12.16-11.95 (m, 2H), 8.11 (d, J=6.9 Hz, 1H), 7.61 (d, J=11.9 Hz, 1H), 7.53 (s, 1H), 7.40 (d, J=9.4 Hz, 2H), 5.35-5.18 (m, 2H), 3.51 (d, J=6.8 Hz, 1H), 3.10-3.00 (m, 2H), 2.89-2.83 (m, 1H), 2.74-2.69 (m, 1H), 2.45-2.25 (m, 3H), 2.21-2.05 (m, 2H), 1.33 (d, J=6.6 Hz, 3H), 1.11-0.89 (m, 4H); MS (ES+) m/z 589.1, 591.1 (M+1).

Example 23 Synthesis of 1-((1-benzhydrylazetidin-3-yl)methyl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 507.0.

Example 24 Synthesis of 1-(1-benzhydrylazetidin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1, the title compound was synthesized along with EXAMPLE 24 and separated by prep-HPLC (30-40% MeCN in 0.1% NH4HCO3); the title compound was obtained as a white solid. MS (ES+) m/z 493.1.

Example 25 Synthesis of N-(azetidin-1-ylsulfonyl)-6-fluoro-3-methyl-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 443.1.

Example 26 Synthesis of 6-fluoro-3-methyl-N-(methylsulfonyl)-1-(5,6,7,8-tetrahydronaphthalen-2-yl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 402.0.

Example 27 Synthesis of N-(azetidin-1-ylsulfonyl)-1-(4-(tert-butyl)cyclohexyl)-6-fluoro-3-methyl-H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 451.3.

Example 28 Synthesis of 1-(4-(tert-butyl)cyclohexyl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 410.1.

Example 29 Synthesis of N-(azetidin-1-ylsulfonyl)-6-fluoro-3-methyl-1-(4-methylcyclohexyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 409.2.

Example 30 Synthesis of N-(azetidin-1-ylsulfonyl)-6-fluoro-1-(4-isopropylcyclohexyl)-3-methyl-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 437.2.

Example 31 Synthesis of 6-fluoro-3-methyl-1-(4-methylcyclohexyl)-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 368.1.

Example 32 Synthesis of N-(azetidin-1-ylsulfonyl)-6-fluoro-3-methyl-1-(tetrahydro-2H-pyran-4-yl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 397.1.

Example 33 Synthesis of 6-fluoro-1-(4-isopropylcyclohexyl)-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 396.1.

Example 34 Synthesis of 6-fluoro-3-methyl-N-(methylsulfonyl)-1-(tetrahydro-2H-pyran-4-yl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 356.1.

Example 35 Synthesis of 1-((3s,5s,7s)-adamantan-1-yl)-N-(azetidin-1-ylsulfonyl)-6-fluoro-3-methyl-H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 447.1.

Example 36 Synthesis of 1-((3s,5s,7s)-adamantan-1-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 406.1.

Example 37 Synthesis of N-(azetidin-1-ylsulfonyl)-1-cyclohexyl-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 395.1.

Example 38 Synthesis of 1-cyclohexyl-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 354.1.

Example 39 Synthesis of 1-(5-chloro-6-isobutoxypyridin-3-yl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 455.0.

Example 40 Synthesis of 1-(((s,3s)-adamantan-1-yl)methyl)-6-fluoro-3-methyl-N-(methylsulfonyl)-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 420.0.

Example 41 Synthesis of 1-(((1s,3s)-adamantan-1-yl)methyl)-N-(azetidin-1-ylsulfonyl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 1 the title compound was obtained as a white solid. MS (ES+) m/z 461.0.

Example 42 Synthesis of N-(azetidin-1-ylsulfonyl)-1-(5-chloro-6-isobutoxypyridin-3-yl)-6-fluoro-3-methyl-1H-indazole-5-carboxamide

Following the same procedure as described in EXAMPLE 3 the title compound was obtained as a white solid. MS (ES+) m/z 496.0.

Example 43 Electrophysiological Assay (EP) (In Vitro Assay)

Patch voltage clamp electrophysiology allows for the direct measurement and quantification of block of voltage-gated sodium channels (NaV's), and allows the determination of the time- and voltage-dependence of block which has been interpreted as differential binding to the resting, open, and inactivated states of the sodium channel (Hille, B., Journal of General Physiology (1977), 69: 497-515).

The following patch voltage clamp electrophysiology studies were performed on representative compounds of the invention using human embryonic kidney cells (HEK), permanently transfected with an expression vector containing the full-length cDNA coding for the desired human sodium channel α-subunit, grown in culture media containing 10% FBS, 1% PSG, and 0.5 mg/mL G418 at 37° C. with 5% CO2. HEK cells used for the electrophysiology (EP) recordings had a passage number of less than 40 for all studies and were used within three days from the time of plating. NaV1.7 and NaV1.5 cDNAs (NM_002977 and AC137587; SCN5A, respectively) were stably expressed in HEK-293 cells. The 1 subunit was coexpressed in both the NaV1.7 and NaV1.5 cell lines.

Sodium currents were measured using the patch clamp technique in the whole-cell configuration using either a PatchXpress automated voltage clamp or manually using an Axopatch 200B (Axon Instruments) or Model 2400 (A-M systems) amplifier. The manual voltage clamp protocol was as follows: Borosilicate glass micropipettes were fire-polished to a tip diameter yielding a resistance of 2-4 Mohms in the working solutions. The pipette was filled with a solution comprised of: 5 mM NaCl, 10 mM CsCl, 120 mM CsF, 0.1 mM CaCl2), 2 mM MgCl2, 10 mM HEPES, 10 mM EGTA; and adjusted to pH 7.2 with CsOH. The external solution had the following composition: 140 mM NaCl, 5 mM KCl, 2 mM CaCl2), 1 mM MgCl2, 10 mM HEPES; and adjusted to pH 7.4 with NaOH. In some studies, the external sodium was reduced by equimolar replacement with choline. Osmolarity in the CsF internal and NaCl external solutions was adjusted to 300 mOsm/kg and 310 mOsm/kg with glucose, respectively. All recordings were performed at ambient temperature in a bath chamber with a volume of 150 μL. Control sodium currents were measured in 0.5% DMSO. Controls and representative compounds of the invention were applied to the recording chamber through a 4-pinch or 8-pinch valve bath perfusion system manufactured by ALA Scientific Instruments.

Currents were recorded at 40 kHz sampling frequency, filtered at 5 Hz, and stored using a Digidata-1322A analogue/digital interface with the pClamp software (Axon Instruments). Series resistance compensation was applied (60-80%). Cells were rejected if currents showed inadequate voltage control (as judged by the IV relationship during stepwise activation). All statistics in this study are given as mean SD.

The membrane potential was maintained at a voltage where inactivation of the channel is complete (which was −60 mV for both NaV1.7 and NaV1.5). The voltage is then stepped back to a very negative (Vhold=150 mV) voltage for 20 ms and then a test pulse is applied to quantify the compound block. The 20 ms brief repolarization was long enough for compound-free channels to completely recover from fast inactivation, but the compound-bound channels recovered more slowly such that negligible recovery could occur during this interval. The percent decrease in sodium current following wash-on of compound was taken as the percent block of sodium channels. Data for representative compounds of formula (I) is provided in Table 1.

Example 44 Tritiated Sulfonamide Binding to Membranes Isolated from Cells that Heterologously Express hNav1.7 and the β1 Subunit

Preparation of membranes containing recombinantly expressed sodium channels: Frozen recombinant cell pellets were thawed on ice and diluted to 4 times the cell pellet weight with ice cold 50 mM Tris HCl, pH 7.4 buffer. The cell suspensions were homogenized on ice using a motorized glass dounce homogeniser. Homogenates were further diluted 8.4 times with ice cold 50 mM Tris HCl, pH 7.4 buffer and then centrifuged at 200×g at 4° C. for 15 min. The supernatants were collected and centrifuged at 10000×g at 4° C. for 50 min. The pellets were then re-suspended in 100 mM NaCl, 20 mM Tris HCl, pH 7.4 buffer containing 1% v/v protease inhibitors (Calbiochem) and re-homogenized on ice. The homogenized membranes were then processed through a syringe equipped with a 26 gauge needle. Protein concentrations were determined by Bradford Assay and the membranes were stored at −80° C.

Radioligand Binding Studies: Saturation experiments. A competitive NaV1.7 inhibitor having a methyl group was tritiated. Three tritiums were incorporated in place of methyl hydrogens to generate [3H]compound. Binding of this radioligand was preformed in 5 mL borosilicate glass test tubes at room temperature. Binding was initiated by adding membranes to increasing concentrations of [3H]compound in 100 mM NaCl, 20 mM Tris HCl, pH 7.4 buffer containing 0.01% w/v bovine serum albumin (BSA) for 18 h. Non-specific binding was determined in the presence of 1 μM unlabelled compound. After 18 h, the reactants were filtered through GF/C glass fiber filters presoaked in 0.5% w/v polyethylene imine. Filters were washed with 15 mL ice cold 100 mM NaCl, 20 mM Tris HCl, pH7.4 buffer containing 0.25% BSA to separate bound from free ligand. [3H]compound bound to filters was quantified by liquid scintillation counting.

Competitive binding experiments: Binding reactions were performed in 96-well polypropylene plates at room temperature for 18 h. In 360 μL, membranes were incubated with 100 pM [3H]compound and increasing concentrations of Test Compound. Non-specific binding was defined in the presence of 1 μM unlabelled compound. Reactions were transferred and filtered through 96-well glass fiber/C filter plates presoaked with 0.5% polyethylene imine. The filtered reactions were washed 5 times with 200 μL ice cold buffer containing 0.25% BSA. Bound radioactivity was determined by liquid scintillation counting.

Data Analysis: For saturation experiments, non-specific binding was subtracted from total binding to provide specific binding and these values were recalculated in terms of pmol ligand bound per mg protein. Saturation curves were constructed and dissociation constants were calculated using the single site ligand binding model: Beq=(Bmax*X)/(X+Kd), where Beq is the amount of ligand bound at equilibrium, Bmax is the maximum receptor density, Kd is the dissociation constant for the ligand, and X is the free ligand concentration. For competition studies percent inhibition was determined and IC50 values were calculated using a 4 parameter logistic model (% inhibition=(A+((B−A)/(1+((x/C){circumflex over ( )}D)))) using XLfit, where A and B are the maximal and minimum inhibition respectively, C is the IC50 concentration and D is the (Hill) slope.

Representative compounds, when tested in this model, demonstrated affinities as set forth in Table 1.

TABLE 1 Nav1.7 (LBA) Nav1.7 Nav1.5 Example Structure (□M) (□M) (□M) 1 0.034 0.68 22 2 0.013 0.086 4.9 3 0.033 0.25 7.4 4 0.024 0.42 26 5 0.006 0.051 2.4 6 0.014 0.072 7 7 0.005 0.009 0.63 8 0.005 0.026 0.61 9 0.006 0.046 8.3 10 0.01 0.033 14 11 0.013 0.023 0.8 12 0.005 0.032 0.51 13 0.015 0.25 2.4 14 0.032 0.4 13 15 0.011 0.088 3.2 16 0.017 0.1 13 17 0.006 0.045 0.88 18 0.04 0.15 17 19 0.012 0.041 6.3 20 0.01 0.062 0.58 21 1.5 4 30 22 0.02 0.11 4.8 23 0.28 0.55 30 24 0.048 0.087 8.6 25 0.26 0.044 ND 26 0.1 0.16 ND 27 0.023 0.14 ND 28 0.047 0.32 ND 29 0.35 1.00 ND 30 0.025 0.039 ND 31 0.34 2 ND 32 10 30.0 30.0 33 0.02 0.079 20.0 34 10 30 30 35 0.18 1.0 30 36 0.37 3.1 30 37 0.49 3.6 30 38 1 11 30 39 0.008 0.039 30 40 0.41 1.2 30 41 0.3 3.40 30 42 0.007 0.071 30

Example 45 Additional Assays Formalin Assay

The formalin test is used as an animal model of acute pain. In the formalin test, animals are briefly habituated to the Plexiglas test chamber on the day prior to experimental day for 20 minutes. On the test day, animals are randomly injected with the test articles. At 30 minutes after drug administration, 50 μL of 10% formalin is injected subcutaneously into the plantar surface of the left hind paw of the rats. Video data acquisition begins immediately after formalin administration, for duration of 90 minutes.

The images are captured using the Actimetrix Limelight software and the files are converted into the MPEG-4 coding. The videos are then analyzed using behavior analysis software “The Observer 5.1”, (Version 5.0, Noldus Information Technology, Wageningen, The Netherlands). The video analysis is conducted by watching the animal behavior and scoring each according to type, and defining the length of the behavior (Dubuisson and Dennis, 1977). Scored behaviors include: (1) normal behavior, (2) putting no weight on the paw, (3) raising the paw, (4) licking/biting or scratching the paw. Elevation, favoring, or excessive licking, biting and scratching of the injected paw indicate a pain response. Analgesic response or protection from compounds is indicated if both paws are resting on the floor with no obvious favoring, excessive licking, biting or scratching of the injected paw.

Analysis of the formalin test data is done according to two factors: (1) Percent Maximal Potential Inhibitory Effect (% MPIE) and (2) pain score. The % MPIEs is calculated by a series of steps, where the first is to sum the length of non-normal behaviors (behaviors 1,2,3) of each animal. A single value for the vehicle group is obtained by averaging all scores within the vehicle treatment group. The following calculation yields the MPIE value for each animal:


MPIE (%)=100−[(treatment sum/average vehicle value)×100%]

The pain score is calculated from a weighted scale as described above. The duration of the behavior is multiplied by the weight (rating of the severity of the response), and divided by the total length of observation to determine a pain rating for each animal. The calculation is represented by the following formula:


Pain rating=[0(To)+1(T1)+2(T2)+3(T3)]/(To+T1+T2+T3)

CFA Induced Chronic Inflammatory Pain

In this test, tactile allodynia is assessed with calibrated von Frey filaments. Following a full week of acclimatization to the vivarium facility, 150 μL of the “Complete Freund's Adjuvant” (CFA) emulsion (CFA suspended in an oil/saline (1:1) emulsion at a concentration of 0.5 mg/mL) is injected subcutaneously into the plantar surface of the left hind paw of rats under light isoflurane anaesthesia. Animals are allowed to recover from the anaesthesia and the baseline thermal and mechanical nociceptive thresholds of all animals are assessed one week after the administration of CFA. All animals are habituated to the experimental equipment for 20 minutes on the day prior to the start of the experiment. The test and control articles are administrated to the animals, and the nociceptive thresholds measured at defined time points after drug administration to determine the analgesic responses to each of the six available treatments. The time points used are previously determined to show the highest analgesic effect for each test compound.

Thermal nociceptive thresholds of the animals are assessed using the Hargreaves test. Animals are placed in a Plexiglas enclosure set on top of an elevated glass platform with heating units. The glass platform is thermostatically controlled at a temperature of approximately 30° C. for all test trials. Animals are allowed to accommodate for 20 minutes following placement into the enclosure until all exploration behavior ceases. The Model 226 Plantar/Tail Stimulator Analgesia Meter (IITC, Woodland Hills, Calif.) is used to apply a radiant heat beam from underneath the glass platform to the plantar surface of the hind paws. During all test trials, the idle intensity and active intensity of the heat source are set at 1 and 45 respectively, and a cut off time of 20 seconds is employed to prevent tissue damage.

The response thresholds of animals to tactile stimuli are measured using the Model 2290 Electrovonfrey anesthesiometer (IITC Life Science, Woodland Hills, Calif.) following the Hargreaves test. Animals are placed in an elevated Plexiglas enclosure set on a wire mesh surface. After 10 minutes of accommodation, pre-calibrated Von Frey hairs are applied perpendicularly to the plantar surface of both paws of the animals in an ascending order starting from the 0.1 g hair, with sufficient force to cause slight buckling of the hair against the paw. Testing continues until the hair with the lowest force to induce a rapid flicking of the paw is determined or when the cut off force of approximately 20 g is reached. This cut off force is used because it represent approximately 10% of the animals' body weight and it serves to prevent raising of the entire limb due to the use of stiffer hairs, which would change the nature of the stimulus.

Postoperative Models of Nociception

In this model, the hypealgesia caused by an intra-planar incision in the paw is measured by applying increased tactile stimuli to the paw until the animal withdraws its paw from the applied stimuli. While animals are anaesthetized under 3.5% isofluorane, which is delivered via a nose cone, a 1 cm longitudinal incision is made using a number 10 scalpel blade in the plantar aspect of the left hind paw through the skin and fascia, starting 0.5 cm from the proximal edge of the heel and extending towards the toes. Following the incision, the skin is apposed using 2, 3-0 sterilized silk sutures. The injured site is covered with Polysporin and Betadine. Animals are returned to their home cage for overnight recovery.

The withdrawal thresholds of animals to tactile stimuli for both operated (ipsilateral) and unoperated (contralateral) paws can be measured using the Model 2290 Electrovonfrey anesthesiometer (IITC Life Science, Woodland Hills, Calif.). Animals are placed in an elevated Plexiglas enclosure set on a wire mesh surface. After at least 10 minutes of acclimatization, pre-calibrated Von Frey hairs are applied perpendicularly to the plantar surface of both paws of the animals in an ascending order starting from the 10 g hair, with sufficient force to cause slight buckling of the hair against the paw. Testing continues until the hair with the lowest force to induce a rapid flicking of the paw is determined or when the cut off force of approximately 20 g is reached. This cut off force is used because it represent approximately 10% of the animals' body weight and it serves to prevent raising of the entire limb due to the use of stiffer hairs, which would change the nature of the stimulus.

Neuropathic Pain Model; Chronic Constriction Injury

Briefly, an approximately 3 cm incision is made through the skin and the fascia at the mid-thigh level of the animals' left hind leg using a no. 10 scalpel blade. The left sciatic nerve is exposed via blunt dissection through the biceps femoris with care to minimize haemorrhagia. Four loose ligatures are tied along the sciatic nerve using 4-0 non-degradable sterilized silk sutures at intervals of 1 to 2 mm apart. The tension of the loose ligatures is tight enough to induce slight constriction of the sciatic nerve when viewed under a dissection microscope at a magnification of 4 fold. In the sham-operated animal, the left sciatic nerve is exposed without further manipulation. Antibacterial ointment is applied directly into the wound, and the muscle is closed using sterilized sutures. Betadine is applied onto the muscle and its surroundings, followed by skin closure with surgical clips.

The response thresholds of animals to tactile stimuli are measured using the Model 2290 Electrovonfrey anesthesiometer (IITC Life Science, Woodland Hills, Calif.). Animals are placed in an elevated Plexiglas enclosure set on a wire mesh surface. After 10 minutes of accommodation, pre-calibrated Von Frey hairs are applied perpendicularly to the plantar surface of both paws of the animals in an ascending order starting from the 0.1 g hair, with sufficient force to cause slight buckling of the hair against the paw. Testing continues until the hair with the lowest force to induce a rapid flicking of the paw is determined or when the cut off force of approximately 20 g is reached. This cut off force is used because it represents approximately 10% of the animals' body weight and it serves to prevent raising of the entire limb due to the use of stiffer hairs, which would change the nature of the stimulus.

Thermal nociceptive thresholds of the animals are assessed using the Hargreaves test. Following the measurement of tactile thresholds, animals are placed in a Plexiglas enclosure set on top of an elevated glass platform with heating units. The glass platform is thermostatically controlled at a temperature of approximately 24 to 26° C. for all test trials. Animals are allowed to accommodate for 10 minutes following placement into the enclosure until all exploration behaviour ceases. The Model 226 Plantar/Tail Stimulator Analgesia Meter (IITC, Woodland Hills, Calif.) is used to apply a radiant heat beam from underneath the glass platform to the plantar surface of the hind paws. During all test trials, the idle intensity and active intensity of the heat source are set at 1 and 55 respectively, and a cut off time of 20 seconds is used to prevent tissue damage.

Neuropathic Pain Model: Spinal Nerve Ligation

The spinal nerve ligation (SNL) neuropathic pain model is used as an animal (i.e. rat) model of neuropathic pain. In the SNL test, the lumbar roots of spinal nerves L5 and L6 are tightly ligated to cause nerve injury, which results in the development of mechanical hyperalgesia, mechanical allodynia and thermal hypersensitivity. The surgery is performed two weeks before the test day in order for the pain state to fully develop in the animals. Several spinal nerve ligation variations are used to characterize the analgesic properties of a compound of the invention.

Ligation of the L5 spinal nerve;

Ligation of the L5 and L6 spinal nerves;

Ligation and transection of the L5 spinal nerve;

Ligation and transection of the L5 and L6 spinal nerves; or

Mild irritation of the L4 spinal nerve in combination with any one of the above (1)-(4).

While the animals are anaesthetized under 3.5% isofluorane delivered via a nose cone, an approximately 2.5 cm longitudinal incision is made using a number 10 scalpel blade in the skin just lateral to the dorsal midline, using the level of the posterior iliac crests as the midpoint of the incision. Following the incision, the isoflourane is readjusted to maintenance levels (1.5%-2.5%). At mid-sacral region, an incision is made with the scalpel blade, sliding the blade along the side of the vertebral column (in the saggital plane) until the blade hits the sacrum. Scissors tips are introduced through the incision and the muscle and ligaments are removed from the spine to expose 2-3 cm of the vertebral column. The muscle and fascia are cleared from the spinal vertebra in order to locate the point where the nerve exits from the vertebra. A small glass hook is placed medial to the spinal nerves and the spinal nerves are gently elevated from the surrounding tissues. Once the spinal nerves have been isolated, a small length of non-degradable 6-0 sterilized silk thread is wound twice around the ball at the tip of the glass hook and passed back under the nerve. The spinal nerves are then firmly ligated by tying a knot, ensuring that the nerve bulges on both sides of the ligature. The procedure may be repeated as needed. In some animals, the L4 spinal nerve may be lightly rubbed (up to 20 times) with the small glass hook to maximize the development of neuropathic pain. Antibacterial ointment is applied directly into the incision, and the muscle is closed using sterilized sutures. Betadine is applied onto the muscle and its surroundings, followed by skin closure with surgical staples or sterile non-absorbable monofilament 5-0 nylon sutures.

The analgesic effect produced by topical administration of a compound of the invention to the animals can then be observed by measuring the paw withdrawal threshold of animals to mechanical tactile stimuli. These may be measured using either the mechanical allodynia procedure or the mechanical hyperalgesia procedure as described below. After establishment of the appropriate baseline measurements by either method, topical formulation of a compound of the invention is applied on the ipsilateral ankle and foot. The animals are then placed in plastic tunnels for 15 minutes to prevent them from licking the treated area and removing the compound. Animals are placed in the acrylic enclosure for 15 minutes before testing the ipsilateral paw by either of the methods described below, and the responses are recorded at 0.5, 1.0 and 2.0 hour post treatment.

Mechanical Allodynia Method

The pain threshold of animals to mechanical alloydnia for both operated and control animals can be measured approximately 14 days post-surgery using manual calibrated von Frey filaments as follows. Animals are placed in an elevated Plexiglas enclosure set on a wire mesh surface. Animals are allowed to acclimate for 20-30 minutes. Pre-calibrated Von Frey hairs are applied perpendicularly to the plantar surface of the ipsilateral paw of the animals starting from the 2.0 g hair, with sufficient force to cause slight buckling of the hair against the paw to establish the baseline measurements. Stimuli are presented in a consecutive manner, either in an ascending or descending order until the first change in response is noted, after which four additional responses are recorded for a total of six responses. The six responses measured in grams are entered into a formula as described by Chaplan, S. R. et al., J. Neurosci. Methods, 1994 July; 53(1):55-63, and a 50% withdrawal threshold is calculated. This constitutes the mechanical allodynia value.

Mechanical Hyperalgesia Method

The response thresholds of animals to tactile stimuli are measured using the Model 2290 Electrovonfrey anesthesiometer (IITC Life Science, Woodland Hills, Calif.). Animals are placed in an elevated Plexiglas enclosure set on a wire mesh surface. After 15 minutes of accommodation in this enclosure, a von Frey hair is applied perpendicularly to the plantar surface of the ipsilateral hind paws of the animals, with sufficient force, measured in grams, to elicit a crisp response of the paw. A response indicates a withdrawal from the painful stimulus and constitutes the efficacy endpoint. The data are expressed as percent change from baseline threshold measured in grams.

Example 46 In Vivo Assay for Treatment of Pruritus

The compounds of the invention can be evaluated for their activity as antipruritic agents by in vivo test using rodent models. One established model for peripherally elicited pruritus is through the injection of serotonin into the rostral back area (neck) in hairless rats. Prior to serotonin injections (e.g., 2 mg/mL, 50 μL), a dose of a compound of the present invention can be applied systemically through oral, intravenous or intraperitoneal routes or topically to a circular area fixed diameter (e.g. 18 mm). Following dosing, the serotonin injections are given in the area of the topical dosing. After serotonin injection the animal behaviour is monitored by video recording for 20 min-1.5 h, and the number of scratches in this time compared to vehicle treated animals. Thus, application of a compound of the current invention could suppress serotonin-induced scratching in rats.

All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non patent publications referred to in this specification are incorporated herein by reference in their entireties.

Although the foregoing invention has been described in some detail to facilitate understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the appended claims. Accordingly, the described embodiments are to be considered as illustrative and not restrictive, and the invention is not to be limited to the details given herein, but may be modified within the scope and equivalents of the appended claims.

Claims

1. A method of treating pain in a mammal comprising, administering to the mammal in need thereof a therapeutically effective amount of a compound of formula (I):

or a pharmaceutically acceptable salt thereof, wherein:
R1 is C1-8 alkyl, C2-8 alkenyl, C1-8 haloalkyl, C1-8 alkoxy, C3-8 carbocycle, C-linked 3-15 membered heterocyclyl, or —NR1AR1B, wherein R1A and R1B are each independently selected from the group consisting of hydrogen, C1-8 alkyl, C1-8 alkoxy, and wherein R1A and R1B are optionally combined to form a 3 to 8 membered heterocyclyl ring optionally comprising 1 additional heteroatom selected from N, O and S; and wherein R1 is optionally substituted with from 1 to 5 substituents selected from the group consisting of C1-4 alkyl, C1-4 haloalkyl, F, Cl, Br, I, —OH, —CN, —NO2, —NRR1aRR1b, —ORR1a, —SRR1a, —Si(RR1a)3 and C3-6 carbocycle; wherein RR1a and RR1b are independently selected from the group consisting of hydrogen, C1-8 alkyl, C1-8 haloalkyl;
RN is hydrogen, C1-4 alkyl or C1-4 haloalkyl;
R2 is selected from the group consisting of H, F, Cl, Br, I, —CN, C1-8 alkyl, C1-8 haloalkyl and C1-8 alkoxy;
R3 is selected from the group consisting of H, F, Cl, Br, I, —CN, C1-8 alkyl, C1-8 haloalkyl and C1-8 alkoxy;
R4 is selected from the group consisting of H, F, Cl, Br, I, —CN, C1-8 alkyl, C1-8 haloalkyl and C1-8 alkoxy;
R5 is selected from the group consisting of H, C1-8 alkyl, C1-8 haloalkyl, C1-8 alkoxy, and C3-8 cycloalkyl, wherein said C1-8 alkoxy, and C3-8 cycloalkyl is optionally substituted with 1-3 substituents selected from F, Cl, Br and I;
L is a linker selected from the group consisting of C1-4 alkylene, C2-4 alkenylene, and C2-4 alkynylene, wherein L is optionally substituted with from 1 to 3 substituents selected from the group consisting of ═O, C1-4 alkyl, halo, and C1-4 haloalkyl;
m is 0 or 1;
X2 is selected from the group consisting of absent, —O—, —S(O)—, —S(O)2— and —N(RX)— wherein Rx is H, C1-8 alkyl, C1-8 alkanoyl, or —S(O)2(C1-8 alkyl);
n is 0, 1, 2, 3, 4, or 5;
the ring A is a 3-15 membered carbocyclyl, a 6-12 membered aryl, a 5-12 membered heteroaryl, or a 3-15 membered heterocyclyl;
each RAA is independently selected from the group consisting of C1-6 alkyl, C1-6 haloalkyl, C1-6 heteroalkyl, CN, F, Cl, Br and I; and
RA is selected from the group consisting of H, —ORA1, —(XRA)-(6-12 membered aryl), —(XRA)-(5-12 membered heteroaryl), and —RA2, wherein said 6-12 membered aryl and 5-12 membered heteroaryl of RA is optionally substituted with from 1 to 5 substituents independently selected from the group consisting of F, Cl, Br, I, C1-4 alkyl, C1-4 haloalkyl, and C1-4(halo)alkoxy; RA1 is selected from the group consisting of hydrogen, C1-8 alkyl, C2-8 alkenyl, C1-8 haloalkyl, C3-8 cycloalkyl, phenyl and benzyl; RA2 is selected from the group consisting of C1-8 alkyl that is optionally substituted with one or more substituents selected from the group consisting of oxo (═O), fluoro, amino, C1-4 alkylamino and di(C1-4alkyl)amino; XRA is selected from the group consisting of absent, —C(═O)—, and C1-4 alkylene; wherein any C1-4 alkylene, of XRA is optionally substituted with 1 to 3 substituents selected from the group consisting of C1-4 alkyl, C1-4 haloalkyl, and phenyl that is optionally substituted with 1 to 5 substituents selected from the group consisting of, F, Cl, Br, I, —NH2, —OH, —CN, —NO2, C1-4 alkyl, C1-4 haloalkyl, C1-4 alkoxy, C1-4(halo)alkoxy, C1-4 alkylamino and C1-4 dialkylamino.

2. The method of claim 1, wherein the compound of formula (I) has the formula (Ia):

3. The method of claim 1, wherein the compound of formula (I) has the formula (Ib):

4. The method of claim 1, wherein R2 is H.

5. The method of claim 1, wherein R3 is halo.

6. The method of claim 1, wherein R3 is F.

7. The method of claim 1, wherein R4 is H.

8. The method of claim 1, wherein R5 is C1-4 alkyl or C1-4 haloalkyl.

9. The method of claim 1, wherein R5 is methyl.

10. The method of claim 1, wherein the compound of formula (I) has the formula (Ic):

11. The method of claim 1, wherein the compound of formula (I) has the formula (Id):

12. The method of claim 1, wherein the compound of formula (I) has the formula (Ie):

13. The method of claim 1, wherein R is methyl, cyclopropyl, or 1-azetidinyl.

14. The method of claim 1, wherein A is an optionally substituted piperidine, optionally substituted pyrrolidine, optionally substituted azetidine, optionally substituted tetrahydronaphthlene, optionally substituted cyclohexane, optionally substituted tetrahydropyran, optionally substituted adamantly, or optionally substituted pyridine ring.

15. The method of claim 1, wherein:

is:
and A is a 3-15 membered heterocyclyl.

16. The method of claim 1, wherein:

is:
and A is a 3-15 membered heterocyclyl.

17. The method of claim 1, wherein each RAA is independently selected from the group consisting of F, Cl and C1-4 haloalkyl.

18. The method of claim 1, wherein:

is selected from the group consisting of:

19. The method of claim 1, wherein RA is selected from the group consisting of:

20. The method of claim 1, wherein the compound of formula (I) is selected from the group consisting of:

Patent History
Publication number: 20210253548
Type: Application
Filed: Jan 19, 2021
Publication Date: Aug 19, 2021
Applicants: GENENTECH, INC. (South San Francisco, CA), XENON PHARMACEUTICALS INC. (Burnaby)
Inventors: Chien-an CHEN (South San Francisco, CA), Sultan CHOWDHURY (Burnaby), Shannon Marie DECKER (Burnaby), Christoph Martin DEHNHARDT (Burnaby), Ivan William HEMEON (Burnaby), Steven MCKERRALL (South San Francisco, CA), Brian SAFINA (South San Francisco, CA), Tao SHENG (Burnaby), Dan SUTHERLIN (South San Francisco, CA)
Application Number: 17/152,185
Classifications
International Classification: C07D 401/04 (20060101); C07D 401/14 (20060101); C07D 405/14 (20060101); C07D 231/56 (20060101); C07D 403/04 (20060101); C07D 405/04 (20060101); C07D 403/06 (20060101); C07D 403/12 (20060101); A61P 29/00 (20060101);