recent patents on calcium channel blockers: emphasis on cns diseases

19
1. Introduction 2. Molecular biology and pharmacology of VACC subtypes 3. Present and potential therapeutic indications of VACC ligands 4. New patents related to VACC ligands in the period 2011 -- 2013 5. Conclusion 6. Expert opinion Review Recent patents on calcium channel blockers: emphasis on CNS diseases Juan-Alberto Arranz-Tagarro, Cristo ´bal de los Rı ´os, Antonio G Garcı ´a & Juan-Fernando Padı ´n Servicio de Farmacologı´a Clı´nica, Hospital Universitario de La Princesa, Madrid, Spain Introduction: Altered homeostasis of cell calcium movement is a central stage in multiple diseases of CNS. This explains the great therapeutic interest in blockers for the various subtypes of voltage-activated calcium channels (VACCs) expressed in neurons. Mitigation of Ca 2+ entry excess elicited by those blockers may restore the altered synaptic transmission, synaptic plasticity and gene expression to normal parameters, ending the enhanced neuronal vulnerability. Areas covered: This review summarize 23 patents on ligands for L-, N- or T-type channels, claimed to have potential therapeutic interest in epilepsy, pain, migraine and neurodegenerative diseases. Expert opinion: Collections of compounds are generally screened in cell lines expressing a given subtype of VACCs. IC 50 to block such channels are often, but not always, provided. In few instances, compounds exhibiting the highest potency in in vitro experiments are also tested in animal models of pain, behavior, epilepsy or Alzheimer’s disease. Attempts to develop selectivity for a given VACC subtype with non-peptidic organic ligands have so far failed. Due to their wide tissue expression, such selectivity is crucial for minimizing possible side effects. However, the few data reported by patents does not allow prediction of selectivity of the new compounds in many cases. Keywords: calcium channel blockers, epilepsy, neurodegenerative diseases, pain, voltage-activated calcium channels Expert Opin. Ther. Patents (2014) 24(9):959-977 1. Introduction About 130 years ago, Sydney Ringer discovered that calcium was essential for both heart [1] and skeletal muscle contraction [2]. A few years later, Locke and Overton found that calcium was essential for impulse transmission between nerve and muscle [3,4]. Another crucial experiment demonstrated that the direct injection of calcium into muscle fibers caused their rapid and strong contraction, what was a direct proof that calcium acted as a contraction trigger [5]. Other additional relevant discovery for our current understanding of the role of calcium as an ubiquitous mes- senger was related to the coupling between calcium entry into the nerve terminal during action potential (AP) firing and neurotransmitter release at the muscle endplate [6]. In a myriad of subsequent studies, calcium was shown to play a role as second messenger in practically every cell of the body, from oocyte fertilization to programmed cell death, hormone secretion, neurotransmitter release, platelet aggregation, smooth muscle contraction, and so on. Our present detailed understanding on the regulation of cell calcium homeostasis is due to the development of sophisticated techniques to monitor calcium currents (i.e., patch-clamp) or the changes of calcium movements between the extracellular space, cytosol and organelles, occurring during cell activation (i.e., fluorescence probes, aequorins). This helped to identify and characterize the elements of the 10.1517/13543776.2014.940892 © 2014 Informa UK, Ltd. ISSN 1354-3776, e-ISSN 1744-7674 959 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Ther. Patents Downloaded from informahealthcare.com by University of Newcastle on 08/22/14 For personal use only.

Upload: juan-fernando

Post on 21-Feb-2017

215 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Recent patents on calcium channel blockers: emphasis on CNS diseases

1. Introduction

2. Molecular biology and

pharmacology of VACC

subtypes

3. Present and potential

therapeutic indications of

VACC ligands

4. New patents related to VACC

ligands in the period

2011 -- 2013

5. Conclusion

6. Expert opinion

Review

Recent patents on calcium channelblockers: emphasis on CNSdiseasesJuan-Alberto Arranz-Tagarro, Cristobal de los Rıos, Antonio G Garcıa† &Juan-Fernando Padın†Servicio de Farmacologıa Clınica, Hospital Universitario de La Princesa, Madrid, Spain

Introduction: Altered homeostasis of cell calciummovement is a central stage in

multiple diseases of CNS. This explains the great therapeutic interest in blockers

for the various subtypes of voltage-activated calcium channels (VACCs)

expressed in neurons. Mitigation of Ca2+ entry excess elicited by those blockers

may restore the altered synaptic transmission, synaptic plasticity and gene

expression to normal parameters, ending the enhanced neuronal vulnerability.

Areas covered: This review summarize 23 patents on ligands for L-, N- or

T-type channels, claimed to have potential therapeutic interest in epilepsy,

pain, migraine and neurodegenerative diseases.

Expert opinion: Collections of compounds are generally screened in cell lines

expressing a given subtype of VACCs. IC50 to block such channels are often,

but not always, provided. In few instances, compounds exhibiting the highest

potency in in vitro experiments are also tested in animal models of pain,

behavior, epilepsy or Alzheimer’s disease. Attempts to develop selectivity for

a given VACC subtype with non-peptidic organic ligands have so far failed.

Due to their wide tissue expression, such selectivity is crucial for minimizing

possible side effects. However, the few data reported by patents does not

allow prediction of selectivity of the new compounds in many cases.

Keywords: calcium channel blockers, epilepsy, neurodegenerative diseases, pain,

voltage-activated calcium channels

Expert Opin. Ther. Patents (2014) 24(9):959-977

1. Introduction

About 130 years ago, Sydney Ringer discovered that calcium was essential for bothheart [1] and skeletal muscle contraction [2]. A few years later, Locke and Overtonfound that calcium was essential for impulse transmission between nerve andmuscle [3,4]. Another crucial experiment demonstrated that the direct injection ofcalcium into muscle fibers caused their rapid and strong contraction, what was adirect proof that calcium acted as a contraction trigger [5]. Other additional relevantdiscovery for our current understanding of the role of calcium as an ubiquitous mes-senger was related to the coupling between calcium entry into the nerve terminalduring action potential (AP) firing and neurotransmitter release at the muscleendplate [6]. In a myriad of subsequent studies, calcium was shown to play a roleas second messenger in practically every cell of the body, from oocyte fertilizationto programmed cell death, hormone secretion, neurotransmitter release, plateletaggregation, smooth muscle contraction, and so on.

Our present detailed understanding on the regulation of cell calcium homeostasisis due to the development of sophisticated techniques to monitor calcium currents(i.e., patch-clamp) or the changes of calcium movements between the extracellularspace, cytosol and organelles, occurring during cell activation (i.e., fluorescenceprobes, aequorins). This helped to identify and characterize the elements of the

10.1517/13543776.2014.940892 © 2014 Informa UK, Ltd. ISSN 1354-3776, e-ISSN 1744-7674 959All rights reserved: reproduction in whole or in part not permitted

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 2: Recent patents on calcium channel blockers: emphasis on CNS diseases

complex machinery that regulates the transient elevations ofcytosolic calcium concentrations ([Ca2+]c) upon cell stimula-tion, in both physiological and pathological conditions. Theelements of such machinery include various channels; trans-porters, uniporters and antiporters; the endoplasmic andsarcoplasmic reticulums; mitochondria and Ca2+-bindingproteins. The regulation of calcium homeostasis under physi-ological and pathological conditions (e.g., neurodegenerativediseases or stroke) has been analyzed in various recent reviews[7-19].The calcium entry into cells via voltage-activated calcium

channels (VACCs), particularly relevant for this review, wasdiscovered through a pioneering experiment on AP recordingsin crayfish muscle fibers [20]. Since then, a large variety ofVACC subtypes has been identified and characterized invarious cell systems and tissues [21]. Also, many contributionshave been interested in selectively targeting the different chan-nel subtypes, giving rise to the new field of calcium antago-nists or L-type channel blockers, with indications mainly incardiovascular diseases [22]. Here, we review the new patentsfrom the years 2011 to 2013 on new compounds that, bytargeting a specific VACC subtype, may find a therapeuticapplication in the clinic. To place those patents in a physio-pharmacological and clinical context, we first summarize thecurrent understanding of the molecular and pharmacologicalproperties of the discovered VACC subtypes.

2. Molecular biology and pharmacology ofVACC subtypes

VACCs constitute a large family of multi-subunit complexescomposed of up to five distinct proteins, that is, a1, b, a2,d and g [23,24]. The a1 subunits are large proteins with amolecular weight between 212 and 273 kDa. Each subunit

is organized in four homologous repeats (I-IV) of six trans-membrane structures. Each repeat contains a S4 region thatacts as the voltage sensor, a P-loop that forms the selectivefilter and the S5-S6 segments that form the channel pore(Figure 1). The four domains are connected through cyto-plasmic linkers, and both C- and N-termini are cytoplasmic.These regions contain sites of interaction with auxiliarysubunits, various sites for activators and blockers, includingG proteins, as well as several phosphorylation sites [23,24].

The auxiliary intracellular b subunit in all high-VACCsbinds to a conserveda-interaction domain (AID) of thea1 sub-unit, thus modulating the channel gating properties and pro-moting cell surface trafficking (Figure 2). This interaction siteis located at the connector between domains I and II of the a1

subunit [25]. In addition to this regulatory function of the a1

subunit, a role for the b subunit in scaffolding multiple signal-ing pathways around the channel, have also been proposed; thisrole suggests that this subunit belongs to the membrane-associated guanylate kinase family [26]. Moreover, another studyreveals that b3 subunits directly reduce glucose-induced Ca2+

oscillations in pancreatic b cells [27]. Thus, two signaling path-ways are proposed in this b3-subunit-mediated effect: directregulation of the inositol trisphosphate receptor and indirectreduction of phospholipase Cb. Thus, b subunits are nowhypothesized to be independent regulatory proteins.

The a2/d subunit is translated as a single protein, butcleaved into d (a single transmembrane-spanning helix) anda2 (the extracellular domain), which are linked by a disulfidebond. The g subunit, characterized by four transmembranedomains, was formerly found in skeletal muscle and later onin the heart and brain VACCs [28]. Functional studies suggestthat some g subunits of VACCs may behave as regulators ofpostsynaptic membrane AMPA-type glutamate receptors [29].

On the basis of their biophysical and pharmacological prop-erties, several types of VACCs have been identified andclassified. They are primarily defined from the nature of theprincipal pore-forming a1 subunit; 10 different a1 subunitshave been characterized by cDNA cloning and functionalexpression in mammalian cells or Xenopus laevis oocytes [23].These subunits are classified into three structurally and func-tionally different a1 subtypes of VACCs that require strongdepolarizing pulses to open (high-VACCs) and others thatopen at lower voltages (low-VACCs) [30]. The first subfamily(Cav1.1 -- Cav1.4) belongs to the group of high-VACCsand includes channels containing a1 subunits that mediate L-type currents (a1S, a1C, a1D, a1F). The second subfamily(Cav2.1 -- Cav2.3) also belongs to the group of high-VACCsand comprises VACCs containing a1 subunits that mediateP/Q-type (a1A), N-type (a1B) and R-type (a1D) currents.Finally, the third subfamily (Cav3.1 -- Cav3.3) includes low-threshold-VACCs, containing a1 subunits (a1G, a1H, a1I)that mediate T-type currents. In contrast to the Cav3 channels,which express by themselves as typical T-type channels in heter-ologous systems, Cav1 and Cav2 channels act as oligomericcomplexes containing auxiliary subunits.

Article highlights.

. Altered calcium homeostasis is central stage in thepathogenesis of neurodegenerative diseases, stroke, painand epilepsies; thus, there is high interest in the searchof blockers of voltage-activated calcium channels (L-, N-,P/Q-, R-, T-type) that may have therapeutic indications inthose diseases.

. In the period 2011 -- 2013, most of the 23 patents herereviewed deal with N- and T-type channel blockers, withpotential therapeutic interest in pain, epilepsy, migraineand neurodegenerative diseases.

. Compounds blocking L-type channels focus thetreatment of Parkinson’s disease, those blocking N- andT-type channels address the treatment of epilepsy and,particularly, neuropathic pain.

. A major drawback to drug development in this field is thewide distribution of various calcium channel subtypes withsimilar molecular structure in the brain and peripheraltissues, which may give rise to intolerable side effects.

This box summarizes key points contained in the article.

J.-A. Arranz-Tagarro et al.

960 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 3: Recent patents on calcium channel blockers: emphasis on CNS diseases

Table 1 summarizes the current and classical nomenclaturefor the diverse VACC subtypes, the type of current carried byeach channel, their blockers with their pIC50, their activatorsand their location in human tissues. L-type channels are themolecular targets of several organic ligands, including the1,4-dihydropyridine family (DHP), the phenylalkylaminesverapamil and D600, and the benzothiazepine diltiazem.Photoaffinity labeling and mutation analysis have revealedthat these three groups of blockers act at three separate, butallosterically coupled receptor sites (Figure 2) [31]. For instance,verapamil is thought to enter the pore from the cytoplasmicside to block it, where its receptor site is formed by amino-acid residues in the S6 segments of domains III and IV [32].

On the other hand, DHPs can be activators (e.g., BayK8644)or blockers (e.g., nifedipine); therefore, they are thought to actallosterically to shift the channel towards its open or closed state,

rather than occluding the ion-conducting pore. Their receptorsites consist of amino acids located in the S6 segments ofdomains III and IV, and the S5 segment of domain III [33].This DHP receptor site shares some common amino acidswith the binding site for verapamil. Finally, diltiazem binds toa third receptor site. Interestingly, the amino acids that arerequired for its interaction also overlap with those required forverapamil binding [34].

Members of the Cav2 family are relatively insensitive toDHPs but are specifically blocked by peptide toxins fromspiders and marine snails (Figure 2). For instance, N-type chan-nels are selectively blocked by w-conotoxin GVIA [35]. Thereceptor site for this toxin comprises amino-acids residues inthe extracellular loop between segments S5 and S6 of domainIII, consistent with a direct pore-blocking mechanism. P/Q-type channels are blocked by w-agatoxin IVA from the funnel

SS

SS

HOOC

S2S3

S4 S5S6

S1

S1S2

S3 S4S5

S6

S4S5

S6 S1S2

S3

S5S6

S1 S2S3

S4

NH2

NH2 COOH

H2N

HOOC

H2N

COOH

I

II III

IV

α2

γδ

NH2 COOH

β

Extracellular fluid

Cytosol

α1

Figure 1. Subunit arrangement for a typical VACC. VACCs are complex proteins composed of four or five different subunits

encoded by multiple genes. The largest a1 subunit incorporates the conduction pore, the voltage sensor and gating

apparatus. It is organized in four homologous domains (I -- IV) with six transmembrane segments (S1 -- S6) in each. The

S4 serves as the voltage sensor and the P-loop between S5 and S6 in each domain determines ion conductance and selectivity.

The a2 and b subunits are hydrophilic proteins located in the extra- and the intracellular space, respectively, whereas the

highly lipophilic g and d subunits are transmembrane proteins. a2 and d subunits are linked by disulfide bonds.P-loop: Pore loop; VACC: Voltage activated calcium channel.

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 961

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 4: Recent patents on calcium channel blockers: emphasis on CNS diseases

web spider venom and by w-conotoxin MVIIC from themarine snail Conus geographus [36,37]. Cav2.3 channels areblocked by the synthetic peptide toxin SNX-482 derivedfrom tarantula venom; the presence of domains III and IV arenecessary for toxin-mediated channel blockade [38].The Cav3 family of VACCs is insensitive to the blockers

mentioned above. Although there are no compounds thatspecifically target T-type channels, some clinically used drugsare able to block this channel subtype, in addition to otherpharmacological activities [39]. These Cav3 channel blockersinclude antiepileptics, such as ethosuximide, or antipsy-chotics, such as pimozide. In addition, Ni2+ is somewhatspecific for T-type versus other classes of calcium currents.Interestingly, the scorpion venomous kurtoxin binds to thea1G T-type channel with high affinity inhibiting the channelby modifying voltage-dependent gating [40].

3. Present and potential therapeuticindications of VACC ligands

The development of a defined pharmacology for the variousVACC subtypes has been intensely pursued during the last

two decades. Ligands for L-type channels have given riseto a new chapter of pharmacology with several therapeuticapproaches to treat cardiovascular diseases, such as arrhythmias,hypertension or angina. In contrast, selective non-peptide smallmolecules, which block or regulate N-, P/Q- or R-type chan-nels, are lacking. By contrast, recent contributions have affordedsome selective T-type channels ligands [41,42]. Hence, amajor goal in this field is the search for selective blockers ormodulators of these channels that could eventually be used astherapeutic tools in diseases.

There are several compounds that block the varioussubtypes of VACCs with poor selectivity. Such is the case ofthe piperazine derivatives flunarizine, R56865, lubeluzoleand dotarizine. These compounds have been grouped aswide-spectrum blockers of VACCs [43-45]. Fluspirilene, amember of the diphenylbutylpiperidine class of neurolepticdrugs (which also includes pimozide, clopimozide andpenfluridol) blocks N-type channels in PC12 cells [46]. Pen-fluridol also blocks T-type channels [47]. Thus, inhibition orfacilitation of a given neurotransmitter release by blockers oractivators of VACC subtypes may have interesting functionalconsequences. For instance, synthetic w-conotoxin MVIIA

H2N

COOH

Extracellular fluid

Cytosol

S1 S6S5S2 S3 S4 S1 S6S5S2 S3 S4 S1 S6S5S2 S3 S4

+ +

+ +

+ +

+ +

+ +

+ +

+ +

+ +

+ +

S1 S6

P-loop

P-loop

P-loop

P-loop

S5S2 S3 S4

I II III IV

P PP

H2N COOH

β

SNAREs

+ +

+ +

+ +

P

G protein

AID

NH2

α2

δ

HOOC

H2N

HOOCSS

SS

DHPs • BTZs• PAAs

• Gabapentin• Pregabalin

α2/δ

Toxins

α1

Figure 2. Membrane topology of a1 and a2/d subunits of the VACC illustrating proteins and blockers interaction sites. The a1

subunit incorporates most of the known sites of VACCs regulation by second messengers, drugs and toxins, except in the case

of gabapentin and pregabalin that interact with the a2/g subunits.AID: a interaction domain; BTZs: Benzothiazepines; DHPs: Dihydropyridines; PAAs: Phenylalkylamines; SNAREs: Soluble NSF attachment protein receptors VACC:

Voltage activated calcium channel.

J.-A. Arranz-Tagarro et al.

962 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 5: Recent patents on calcium channel blockers: emphasis on CNS diseases

Table 1. Characteristics of the various calcium channel subtypes according to their a1-containing subunit.

Calcium channel subtype Type of

current

Blockers

(affinity; pIC50*)

Activators Tissue location

(human)

Current

nomenclature

Traditional

nomenclature

Cav1.1 a1S L Nifedipine (6.3)NisoldipineNitrendipine (6)(+)-Isradipine (6.7 -- 8.2)(-)-Devapamil (8.2 -- 8.7)Cd2+ (4.3)

BayK8644FPL64176SZ(+)-(S)-202 -- 791

Skeletal muscleBasal gangliaCerebral cortexHippocampusSubstantia nigraSpinal cordLymphocytes

Cav1.2 a1C L Nifedipine (7 -- 8)Nimodipine (6.8)Nisoldipine (7.1)Nitrendipine (6)Kurtoxin (7.2)(+)-Isradipine (7.5)(-) -Devapamil (7 -- 8.4)Cd2+ (5.7 -- 6)Mibefradil (4.9)

BayK8644(-) -(S)-BayK8644FPL64176PCA50941SZ(+)-(S)-202 -- 791

HeartSmooth muscleBrainPituitaryAdrenal medulla

Cav1.3 a1D L NifedipineCalciseptineCalcicludineNisoldipine (6.4 -- 7)Nimodipine (5.7 -- 6.6)

BayK8644(-)-(S)-BayK8644FPL64176PCA50941

BrainPancreasAdrenal medullaCochleaKidneyOvary

Cav1.4 a1F L Nifedipine (6)Isradipine (6.7)

BayK8644FPL64176

RetinaLymphoid tissuesMast cells

Cav2.1 a1A P/Q w-aga-IIIA (9.3)w-aga-IVA (6.8 -- 8.7)w-aga-IVB (8.5)w-ctx-MVIIC (8 -- 9.5)w-ctx-MVIID (9)w-ctx-CVIB (8)w-ctx-CVIC (7.5)w-phonetoxin-IIA (9.2)DW13.3 (8.4)Kurtoxin (7.8)

Brain (hippocampus,entorhinal cortex,subiculum)CerebellumPituitaryCochleaAdrenal medulla

Cav2.2 a1B N w-ctx-GVIA (9.2 -- 10.4)w-ctx-MVIIA (7.7 -- 10.3)w-ctx-MVIIC (6.1 -- 8.5)w-ctx-CVIA (9.3)w-ctx-CVIB (8.1)w-ctx-CVIC (8.1)w-ctx-CVID (10.2)DW13.3 (7.7 -- 8.6)Huwertoxin I (7)w-ctx-SO-3 (6.8)

Glycerotoxin Brain (hippocampus andparahippocampal gyrus)Peripheral nervous systemAdrenal medulla

Peptide toxins from venom of marine snails: w-ctx-GVIA, w-conotoxin GVIA; w-ctx-MVIIA, w-conotoxin MVIIA; w-ctx-MVIIC, w-conotoxin MVIIC; w-ctx-MVIID,

w-conotoxin MVIID; w-ctx-CVIA, w-conotoxin CVIA; w- ctx-CVIB, w-conotoxin CVIB; w- ctx-CVIC, w-conotoxin CVIC; w- ctx-CVID, w-conotoxin CVID; w-ctx-SO-3,w-conotoxin SO-3.

Peptide toxins from different species of venom spiders: DW13.3 (74-amino acid peptide toxin); w-aga-IVA, w-agatoxin IVA; w-aga-IVB, w-agatoxin-IVB; w-aga-IIIA,w-agatoxin-IIIA; ProTx-I, Protoxin-I or b-theraphotoxin-Tp1a; SNX-482 (homologous to the spider peptides grammatoxin S1A and hanatoxin); w-PnTx3-3, w-ctenitoxin-Pn2a; w-PnTx3-6, PhTx-3 toxin fraction.

Drug from chemical synthesis: NNC55-0396, (1S,2S)-2-[2-[[3-(1H-Benzimidazol-2-yl)propyl]methylamino]ethyl]-6-fluoro-1,2,3,4-tetrahydro-1-(1-methylethyl)-

2-naphthalenyl cyclopropanecarboxylate dihydrochloride; ML 218, 3,5-Dichloro-N-[[(1a,5a,6-exo,6a)-3-(3,3-dimethylbutyl)-3-azabicyclo[3.1.0]hex-6-yl]methyl]-

benzamide hydrochloride.

Adapted from [148].

*pIC50 is the negative logarithm of the molar IC50 concentrations.

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 963

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 6: Recent patents on calcium channel blockers: emphasis on CNS diseases

Table 1. Characteristics of the various calcium channel subtypes according to their a1-containing subunit

(continued).

Calcium channel subtype Type of

current

Blockers

(affinity; pIC50*)

Activators Tissue location

(human)

Current

nomenclature

Traditional

nomenclature

Cav2.3 a1E R SNX-482 (7 -- 8.2)DW13.3 (7)w-PnTx3-3 (7.9)w-PnTx3-6 (6.9)w-phonetoxin-IIA (7.2)Pb2+ (7)Ni2+ (3.6 -- 4.7)

BrainCochleaRetinaHeartPituitaryAdrenal medulla

Cav3.1 a1G T ProTx-I (7,3)Kurtoxin (7.3 -- 7.8)Mibefradil (6 -- 6.6)NNC55-0396Pimozide (7.5)(-) -(R)-efonidipine (5 -- 7)Ni2+ (3.6 -- 3.8)Anandamide (5.4)

Brain(cerebellum>thalamus>n-eocortex > substantianigra >medulla > striatum)

Peripheral nervous systemAdrenal medulla

Cav3.2 a1H T Kurtoxin (7.3 -- 7.6)Mibefradil (5.9 -- 7.2)NNC55-0396ML 218 (6.5)Ni2+ (4.9 -- 5.2)Anandamide (6.5)Pimozine (7.3)

KidneyLiverHeartBrain(putamen > amygdale,caudate nucleus frontallobe, hippocampus,cerebellum, substantianigra>thalamus>medulla,spinal cord, occipital lobe,temporal lobe)LungSkeletal musclePancreasPlacentaAdrenal glomerulosaAdrenal medulla

Cav3.3 a1I T Mibefradil (5.8)NNC55-0396Pimozide (7.5)Anandamide (6)ML 218 (6.6)Ni2+ (3.7 -- 4.1)

Brain (cerebellum,occipital lobe, frontallobe, amygdale, caudatenucleus,hippocampus > temporallobe,putamen > substantianigra, medulla)

Peptide toxins from venom of marine snails: w-ctx-GVIA, w-conotoxin GVIA; w-ctx-MVIIA, w-conotoxin MVIIA; w-ctx-MVIIC, w-conotoxin MVIIC; w-ctx-MVIID,

w-conotoxin MVIID; w-ctx-CVIA, w-conotoxin CVIA; w- ctx-CVIB, w-conotoxin CVIB; w- ctx-CVIC, w-conotoxin CVIC; w- ctx-CVID, w-conotoxin CVID; w-ctx-SO-3,w-conotoxin SO-3.

Peptide toxins from different species of venom spiders: DW13.3 (74-amino acid peptide toxin); w-aga-IVA, w-agatoxin IVA; w-aga-IVB, w-agatoxin-IVB; w-aga-IIIA,w-agatoxin-IIIA; ProTx-I, Protoxin-I or b-theraphotoxin-Tp1a; SNX-482 (homologous to the spider peptides grammatoxin S1A and hanatoxin); w-PnTx3-3, w-ctenitoxin-Pn2a; w-PnTx3-6, PhTx-3 toxin fraction.

Drug from chemical synthesis: NNC55-0396, (1S,2S)-2-[2-[[3-(1H-Benzimidazol-2-yl)propyl]methylamino]ethyl]-6-fluoro-1,2,3,4-tetrahydro-1-(1-methylethyl)-

2-naphthalenyl cyclopropanecarboxylate dihydrochloride; ML 218, 3,5-Dichloro-N-[[(1a,5a,6-exo,6a)-3-(3,3-dimethylbutyl)-3-azabicyclo[3.1.0]hex-6-yl]methyl]-

benzamide hydrochloride.

Adapted from [148].

*pIC50 is the negative logarithm of the molar IC50 concentrations.

J.-A. Arranz-Tagarro et al.

964 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 7: Recent patents on calcium channel blockers: emphasis on CNS diseases

protects hippocampal CA1 pyramidal neurons from damagecaused by transient global forebrain ischemia in the rat [48].

Some medicines that are currently used to treat the epilep-sies are known to target some VACC subtypes. The linkbetween channel blockade and the antiepileptic effect couldbe based in the mitigation of excessive glutamate release partlycontrolled by P/Q-type channels [49], which are believed to beinvolved in idiopathic generalized epilepsies [50]. For instance,gabapentin and pregabalin bind to the a2/d subunit ofVACCs [51-53] and attenuate neurotransmitter release throughinhibition of P/Q-type channels [54,55]. In addition, gabapen-tin reduces excitatory and inhibitory postsynaptic currents inthe spinal cord through P/Q-type channel blockade [56]. Fur-thermore, levetiracetam blocks VACC currents in pyramidalneurons of hippocampal slices [57] and diminishes excitatorypostsynaptic potentials in granule cells through a P/Q-typechannel blockade [58]. In addition, levetiracetam attenuatesthe paroxysmal depolarization shift through N- and P/Q-type channel blockade [59]. Also, the antiepileptics lamotri-gine, ethosuximide, zonisamide and carbamazepine blockhigh-VACCs in cortical neurons [60-62].

Another therapeutic application of VACC ligands is pain.Such is the case of w-conotoxin MVIIA, also known asSNX-111 or ziconotide [63]. Thus, the approval of Prialt�, asynthetic version of w-conotoxin MVIIA, for the treatmentof severe chronic pain associated with cancer, AIDS and neu-ropathies represents a significant advance in analgesia. Itspotent analgesic effects, manifested even in patients resistantto opioids, are due to the inhibition of proprioceptive neuro-transmitters and neuromodulators from the nerve terminals ofprimary afferent neurons in the dorsal horn of the spinalcord [64,65]. Ziconotide has shown potent efficacy in a postsur-gical setting [66] as well as in patients suffering from a varietyof chronic, intractable severe pain syndromes [67].

T-type channel currents were first discovered and charac-terized by Carbone and Lux in dorsal root ganglion (DRG)neurons [68] T-type channels have been deeply studied forpain treatment [63,69-71]. So, T-type knockout mice are hyper-algesic in a model of visceral pain, which is likely related to aT-type channel-dependent antinociceptive mechanismoperating in the thalamus [72].

Some studies have implicated the T-channel isoformCav3.2 in the sensitization of pain responses by enhancingexcitability of nociceptors, in animal models of type 1 andtype 2 peripheral diabetic neuropathies (PDN). In thetype 1 diabetic model, Cav3.2 T-type currents are upregulatedthreefold [73-75]. Furthermore, the Cav3.2 channel is known toregulate presynaptic glutamate release in nociceptive dorsalhorn neurons of the spinal cord [76]. Due to the diabetichyperglycemia, it has been proposed that post-translationalmodification via glycosylation leading to upregulation of theCav3.2 channel in painful PDN, could become a useful target.Hence, by blocking the glycosylation of the Cav3.2 channelwith enzymes, the hyperexcitability of DRG neurons couldbe mitigated, thus preventing or reversing neuropathic pain

symptoms such as hyperalgesia and allodynia in PDN [71].Other state-independent blockers of the Cav3.2 channel,such as TTA-A2 or Z123212, which preferentially interactwith inactivated T-type channels, elicit analgesia in rodentmodels of pain. [77-79].

On the other hand, the antiepileptic ethosuximide blocksT-type currents [80] and reverses tactile allodynia and thermalhyperalgesia in nerve-ligated rats [81]. Other interestingtherapeutic target in analgesia is the auxiliary subunit a2/dof high-VACCs. Gabapentin binds to this subunit with highaffinity [82] and blocks the calcium current, particularly afterconstriction of the sciatic nerve in rats [83]. In clinical trials,gabapentin has shown moderate efficacy in postherpeticneuralgia, diabetic neuropathy, trigeminal neuralgia, lowback pain and cancer pain. More recently, pregabalin, anothera2/d ligand, has shown higher efficacy in animal models andin diseases developing neuropathic pain [84]. Gabapentin hasalso shown efficacy as a prophylactic drug in migraine [85],which agrees with its ability to block spreading depression [86].

L-type channels play multiple roles in the control of neuro-transmitter release. Spontaneous AP firing, low-thresholdexocytosis and compensatory/excess endocytosis are likelycontrolled by the Cav1.3 channel. The delayed inactivationof the Cav1.3 channel could be physiologically relevant forsustaining prolonged Ca2+ influxes that support the endocy-totic process [87,88]. The Cav1.3 channel is also critical forthe control of vital functions, such as heart beating [89], hear-ing [90] and dopamine release [91]. In this context, ongoingresearch focuses on the synthesis of new L-type channelligands with potential therapeutic interest in Parkinson’sdisease (PD), cardiac arrhythmias, chronic stress and otherneuro- and cardiovascular pathologies in which the Cav1.3channel is likely to be involved [92].

Concerning PD, it is worth to note that dopaminergicneurons of the substantia nigra pars compacta (SNc) utilizethe Cav1.3 channel to drive spontaneous pacemaking activity.The continuous Ca2+ influx creates an excessive metabolicload, rendering dopaminergic neurons particularly vulnerableto secondary insults on the mitochondrial function. Theseneurons become more vulnerable with the age, what couldexplain why ageing is a significant risk factor for developingPD. In this context, it is interesting that the L-type channelblocker isradipine restored the Ca2+-independent ‘juvenile’pacemaking activity in those neurons and the finding thatsubcutaneous delivery of isradipine-caused neuroprotection[93]. In fact, hypertensive patients treated with L-type channelblockers had a diminished risk of developing PD [94].

The fact that nimodipine reduced the infarct size in a ratmodel of focal ischemia indicated that the L-type channelblockade could be an effective neuroprotective strategy instroke [95]. Unfortunately, clinical trials have not providedpositive outcomes [96-98]. High-VACCs have also been linkedto Huntington’s disease. Huntingtin directly binds to thea2/d subunit of VACCs and to the pore-forming subunit ofN-type channels, suggesting their involvement in disease

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 965

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 8: Recent patents on calcium channel blockers: emphasis on CNS diseases

pathogenesis, together with the promising potential of thesechannel ligands to treat the disease [99]. Finally, amyloid bpeptide (A�) pathology has been associated to a gain-of-func-tion of P/Q-type channels, what could explain that pirace-tam [100] and levetiracetam [101] have shown efficacy inimproving cognitive performance and in slowing down thecognitive decline in patients of Alzheimer’s disease (AD).

4. New patents related to VACC ligands inthe period 2011 -- 2013

Patents that claim various potential therapeutic uses of differ-ent novel molecular entities targeting one or more of theVACC subtypes are summarized in this section. Out of58 patents reviewed dealing with VACC subtypes, but only23 of them included pharmacological tests to confirm theirVACC blocking activity, and these were that we selected fordiscussing in this review.

4.1 Blockers of L-type VACCsThree patents on chemistry and pharmacology of new ligandsfor L-type channels are reviewed. Their molecular structuresare shown in Figure 3.The patent by Delgado-Martın et al. from Consejo Supe-

rior de Investigaciones Cientıficas, Spain [102]. deals with adetailed procedure to extract and purify crambescidine800 and 816 (ratio 1:3), from the sponge Crambe crambe(Figure 3,1). The authors found that the mixture relaxed therat myometrium pre-contracted with high-K+, with an IC50

about 100 µM; veratridine-elicited contractions were notaltered by the mixture, nor those oxytocin or methacholine-induced contractions. Thus, the authors conclude that thecrambescidine mixture is not acting on sodium channels;rather, they attribute its relaxing effect to the blockade ofCa2+ entry through L-type channels that are expressed in therat myometrium. Although the potency of the crambescidinemixture is low, its molecular structure may inspire the synthe-sis and optimization of new more potent ligands for thesubtypes of L-type channels with potential therapeutic indica-tions in cardiovascular diseases.

A second patent focuses on new ligands for L-type channelswith therapeutic potential in PD [103]. The general structure ofthe claimed compounds is a pyrimidine trione core, where Natoms are substituted with alkyl groups, and the heterocycliccarbonyl groups facilitate the generation of either dipole orhydrogen bonds (Figure 3,2). The ligands were tested inHEK 293 cells expressing either human Cav1.3 or Cav1.2 L-type channels. The entry of Ca2+ through those channelswas monitored by using fluorescent imaging plate reader(FLIPR) technology using calcium-4 dye as indicator. Thehighest selectivity for the Cav1.3 channel was achieved withcompounds bearing cyclohexyl or cyclopentyl substituents atR1 and an m-substituted phenethyl moiety at R2 of the pyrim-idine trione core. The authors found that compoundSKP004C08 (Figure 3; 2) had an IC50 of 1.61 µM to blockCav1.3 channels, having > 1000-fold selectivity with respectto the Cav1.2 channel. With the patch-clamp technique,SKP004C08 blocks by 35% Cav1.3 currents at 1 µM, show-ing no effect on Cav1.2 currents. In addition, reportedcompounds presented a good blood--brain barrier (BBB) pen-etration, with blood:brain concentration ratios between10:1 and 1:1, when administered either intraperitoneally ororally. The Cav1.3 channel controls pacemaking in dopami-nergic neurons at the SNc. The fast AP firing of these neuronsgives rise to elevated intracellular Ca2+ trafficking, enhancedoxidative stress and greater neuronal vulnerability. Thus,blocking the Cav1.3 channel is expected to mitigate thoseevents, leading to neuronal protection. This is the basis forthe proposal of using L-type channel blockers to treat PD.To achieve this, the blocker should readily cross the BBBand displays selectivity for the dopaminergic Cav1.3 channelversus the Cav1.2 channel, widely expressed in cardiovasculartissues. For this reason, SKP004C08 could potentiallybecome a useful pharmacological tool to treat PD. Up tonow, however, all known L-type channel blockers exhibitcardiovascular effects.

The third patent deals with the synthesis of dihydropyri-dine-benzodiazepine tetra- and tricyclic derivatives, for thetreatment of both central and vascular diseases (Figure 3,3)disclosed by Verdecia-Reyes et al., from Centro de

X = H, Crambescidine 800X = OH, Crambescidine 816

2 31

SKP004C08

NH

NH

N O N

OO

XH O O

NH2

NH2

OH

14 N N

OO

O

Cl

NH

N

NR1

R2

R3

R4

Ar

Figure 3. Chemical structures of compounds (1) and (2), and general formula (3), from patents on L-type channel ligands,

discussed in Section 4.1. SKP004C08 (2) is the best compound of a pyrimidine-derived family disclosed by Surmeier’s group,

where N atoms are substituted with alkyl groups. General formula (3) exemplifies a juxtaposition of both dihydropyridine and

benzodiazepine moieties, to target VACCs and GABAA receptors, respectively.VACC: Voltage activated calcium channel.

J.-A. Arranz-Tagarro et al.

966 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 9: Recent patents on calcium channel blockers: emphasis on CNS diseases

Investigacion y Desarrollo de Medicamentos [104]. The com-pounds block both GABAA receptors and VACCs. Thispharmacological profile makes Verdecia-Reyes et al. suggesttheir potential therapeutic use for cardiovascular, cerebrovas-cular, neurodegenerative and neurologic diseases. Somein vivo experiments are commented, such as the open-fieldtests to monitor the sedative effects of compounds in AlbinoSwiss mice at the dose of 4 mg/kg, observing a differential neu-rosedative behavior, depending on the structural modificationproposed.

4.2 Blockers of N-type VACCsThe discovery of new ligands for N-type channels (Cav2.2)has attracted much attention and efforts, as they are nowproposed as good targets for pain relief. This action is likelydue to the fact that, at the dorsal horn of the spinal cord,N-type channels modulate the release of key pronociceptiveneurotransmitters, such as glutamate, substance P and calcito-nin-gene-related peptide. Seven patents dealing with newN-type channel blockers are reviewed in this section.

Searle et al. from Abbot Laboratories, synthesizedcyclopenta-fused pyrroles with a general formula showing anamino substitution at C4 on the fully hydrogenated bicyclicring (Figure 4,4) [105]. Near 800 compounds were tested onIMR32 cells that endogenously expressed human N-typechannels, using FLIPR technology to monitor [Ca2+]c eleva-tions elicited by K+. Some hit compounds with IC50 below1 µM were found. Mechanical hyperalgesia elicited by capsa-icin was inhibited 90% by two compounds administeredorally at 30 mg/kg. Furthermore, in the Bennett model ofneuropathic pain-related allodynia, the blockade was 80%.These small molecules may have a better activity than thew-conotoxins peptides for the treatment of the intractablepain, due to their blockade of N-type channels.

Convergence Pharmaceuticals Ltd. has synthesized200 compunds showing a general formula defined by a piper-azinylsulfone, when the aryl group is a differently substitutedbenzene (Figure 4,5). Using the patch-clamp technique in

HEK 293 cells expressing human N-type channels, the bestcompounds caused use-dependent inhibition of calcium cur-rents when administered at 30 µM. However, no in vivodata have been reported [106]. Use-dependent channel block-ade is interesting because, in spite of low potency, thecompounds may efficiently target highly active channelsunder conditions of increased neuronal excitability, whichare contributing to the pathophysiology of chronic pain.

In another series of N-aryltetrazoles, where R2 and R3 canbe part of an aza-heterocycle nucleus (Figure 4,6), Beswicket al., from the same company, found that the use-dependentchannel blockade was achieved at concentrations 10-foldlower than in the previous family [107].

Li et al., from Abbvie, Inc., synthesized 212 octahydropyr-rolo[1,2-a]pyrazine derivatives. Compounds were screened inIMR32 cells expressing human N-type channels, monitoringCa2+ influx through FLIPR technology. For instance, example47 had an IC50 of 0.29 µM (Figure 5,7). Ten compounds wereselected to test in the Bennett model of neurophatic pain.Example 186 (Figure 5,7) exhibited 91% of maximum possi-ble analgesic effect [108]. More recently, the same group hasreported the synthesis of 231 pyrrolo[1,2-a]pyrazine sulfona-mides. In the case of compounds 203 and 223 (Figure 5,8),IC50 to block N-type channels were even lower (0.13 µM).However, the analgesic effect for compound 21 (Figure 5;8)was only 43% of the maximum posible analgesic effect [109].

Khanna et al., from Indiana University Research and Tech-nology Corp., have found an original strategy to block N-typechannels with peptides based on transactivator of transcription(TAT) CB3. The collapsin response mediator protein2 (CRMP-2) upregulates N-type currents. Thus, those peptidesuncouple the CRMP-2-Cav2.2 complex and elicit analgesia inanimal models of pain [110]. A more recent patent of the samegroup reports new TAT-conjugated peptides with the sequenceYGRKKRRQRRRARSRLAELRGVPRGL for compoundST1-104. In the compound ST1-106, arginine-9 is replacedby leucine. These peptides were studied in several pain animalmodels including diabetic neuropathy or retroviral-inducedneuropathy. Interestingly, ST1-106 blocked R-type currentsin addition to the blockade of N-type currents, whereasST1-104 did not target R-type currents [111].

4.3 Blockers of T-type VACCsT-type channels (Cav3) are involved in the regulation of anincreasing number of physiological functions, as well as inthe pathophysiology of diseases like pain, epilepsy, cancer,AD, essential tremor or hypertension. Although severalcompounds have been synthesized and tested in the last twodecades, a selective T-type channel blocker has not yet reacheda clinically useful status. However, much efforts and invest-ment are being devoted to the discovery and development ofnew ligands for T-type channels. The nine patents herereviewed exemplify such effort.

Zalicus Pharmaceuticals Ltd. has synthesized about 600 com-pounds derivates of N-piperidinylacetamide. The compounds

4

N

NR3R2

R1

5

N

N

S

Ar

OO

NO

6

N N

NN

R1

N

R2

R3

Figure 4. General formula (4) to (6), from patents on N-type

channel ligands, discussed in Section 4.2, are grouped.

Piperazinylsulfones disclosed by Convergence Pharmaceuti-

cals present different fusion with pyridine nuclei, and ‘Ar’

defines a differentially substituted benzene, as shown in

the general formula (5). R2 and R3 can be part of an

aza-heterocycle nucleus in the general formula (6).

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 967

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 10: Recent patents on calcium channel blockers: emphasis on CNS diseases

present general Markush structures where ‘Ar’ can be eitherphenyl or heterocyclic rings (Figure 6,9). Compound screeningwas performed in HEK 293 cells expressing human T-typechannels, using the patch-clamp technique. Some compoundshad IC50 of 0.1 µM, indicating a high potency to blockT-type channels. Two lead compounds were tested in a neuro-pathic painmodel consisting in L5/L6 spinal nerve ligation, andin two epilepsy models: the electroconvulsive shock thresholdtest and the genetic absence epilepsy rats from Strasbourg(GAERS). Their efficacy led the authors to claim theirtherapeutic use in both pain and epilepsy [112].Gray and Haverstick, from University of Virginia, have

synthesized a series of arylalkylpyrroles analogues to TH-1177, shown in Figure 6,10. This compound inhibitedCav3.2 channel currents with an IC50 of 0.8 µM in Jurkatcancer cells expressing the Cav3.2 channel. Because T-typechannels are associated with prostate cancer, cell proliferationand inflammation [113,114], the authors claim the use ofTH-1177 to treat androgen-sensitive human prostate adeno-carcinoma. In fact, the compound inhibited PC13 humanprostate cancer cell proliferation, with an IC50 of 14 µM [115].Merck Sharp & Dohme Corp. has disclosed the synthesis

of pyrazynilphenylamide derivatives with the general formulashown in Figure 6,11. For some of the compounds, blockadeof T-type currents in HEK 293 cells expressing human T-type channels, using the patch-clamp technique, showedIC50 of < 1 µM, indicating a high potency to block T-typechannels [116]. At this company, Schlegel et al. have patentedthe synthesis and use of heterocycle derivatives having acentral amide group with a general formula, where R1 canbe alkyl or hydrogen. In a few examples, triazole nuclei arereplaced by tetrazole, imidazole or oxazole (Figure 6,12).Some compounds exhibited IC50 lower than 1 µM to block

T-type currents [117]. Another patent from the same companyreports the synthesis of imidazolylmethylpiperidine deriva-tives (Figure 6,13). Again, some of these compounds hadactivities with IC50 below 1 µM to block T-type currents [118].

Tung et al., from Concert Pharmaceuticals, Inc., havepatented a family of deuterated mibefradil derivatives with atetrahydronaphtalene structure. Replacement of hydrogen bydeuterium is aimed to improve the metabolic stability of themibefradil derivatives. Mibefradil is a T-type channel blockerthat inhibits hepatic CYP, more specifically CYP3A4, pre-sumably by some of its oxidized metabolites. This gave riseto fatal drug interactions that caused its withdrawal from themarket. The lead deuterated derivative 500 (Figure 6,14) didnot inhibit CYP3A4 [119]. This effect is due to a slower rateof its enzymatic metabolism by either CYP3A4-catalyzed oxi-dation or ester hydrolysis by esterases. The physicochemicalfundament of this slowing lies in the so-called kinetic isotopeeffect. Basically, the rate of the chemical reaction to oxidize acarbon-deuterium bond is much higher than that one tooxidize a regular carbon-hydrogen bond. Thus, CYP3A4-ad-dressed metabolism over deuterated analogues of mibefradilwill be quite slower than over mibefradil.

Weber et al., from Zenyaku Kogyo Kabushikikaisha, havepatented the use of new heterocyclic compounds, analogues ofST101, that contain a central imidazol-4-one nucleus(Figure 6,15). In HEK 293 cells expressing human T-type chan-nel subtypes, compound ST101 blocked the Cav3.1 channelwith an IC50 of 42.9 nM, while it was unable to blockCav3.2 and Cav3.3 currents with IC50 below 100 µM [120]. Itis important to note that the same compound has been recentlydescribed to show agonist activity of this channel at 0.1 nM[121]. Otherwise, Weber et al. discuss that T-type currents havebeen linked to the activation of A-type potassium currents

CF3

HN

N

NO

N N

OH

SO

ONH

CF3

N

NH

N

NSO

OF3C

O

N N

H

N

NO

N N

SO

ON

H

N

N

O

SO

OF3C

CF3H

7

Example 47 Example 186

8

Example 21 Example 203 Example 223

Figure 5. Chemical structures of (7) to (8), showing the most active compounds from patents of Abbvie corp. on N-type

channels ligands, discussed in Section 4.2, are grouped. on N-type channels ligands, discussed in Section 4.2, are grouped.

This company focuses on pyrrolopyrazine derivatives, mainly showing arylsulfonamide moieties.

J.-A. Arranz-Tagarro et al.

968 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 11: Recent patents on calcium channel blockers: emphasis on CNS diseases

through Kv4 channels [122]. Thus, authors hypothesize that theblockade of T-type channels would increase the firing of APsto indirectly increase neurotransmitter release. Low-thresholdneurotransmitter release may also contribute to an enhancedsynaptic transmission [39]. The parent compound inhibits A�deposition and improves cognition, implying that T-type chan-nel blockade could be beneficial in AD, too [120].

Bourinet et al., from the Universit�e de Nice Sophia Antip-olis, have synthesized a peptide with the following amino-acidsequence: YCQKFLWTCDSERPCCEGLVCRLWCKIN.

The peptide was synthesized either by expression of theencoding DNA into a plasmid or by solid phase chemicalsynthesis. In HEK 293 cells expressing the human Cav3.2channel, the peptide blocked T-type currents with an IC50

in the low micromolar range. In a mouse model of pain eli-cited by thermal stimuli, treatment with the peptide showedmuch longer reaction time to heat, what was not observedin Cav3.2 channel-knockout mice, demonstrating its selectiv-ity of action. Authors also show in vivo studies to testboth antihypertensive and antiproliferative effects, as the

9

NHN

OR

R

NNH

Ar

R

O

10

NO

Cl

O

11

N

N O

HN

N

O CF3

R1 R2

12

O

HN

X

N NNAlkyl

R

R1

13

NN

NH

O

R3

R4

R2

R1

14

N

NHN

O

OO

FD D

DD

15

N

N

O

ST101

16

HN NN

O

O

Figure 6. Chemical structures and general formula (9) to (16), of the patents on T-type Ca2+ channels ligands described in

Section 4.3, are grouped. Acetamides disclosed by Zalicus (9) can present as ‘Ar’ phenyl or heterocyclic rings, while those

patented by Merck are limited to the general formula (11) and (12), where R1 and R2 can be alkyl or hydrogen. In the case of

acetamide lactams disclosed by Abbott, compound (16) showed the best profile in the reduction of the secondary mechanical

hyperalgesia exerted by capsaicin. Structure (10) represents the unique compound disclosed by University of Virginia Patent

Foundation, proposed to block T-type Ca2+ channels in adenocarcinoma cells, thus possessing antiproliferative activity. Merck

has also patented T-type Ca2+ channel ligands with piperidine-imidazole double core, as represented by general formula (13).

Compound (14) showed the best pharmacokinetic profile of all the deuterated analogues of mibefradil disclosed by Concert.

Finally, ST101 (15) is the compound selected by Zeniaku from a one-hundred piridoimidazole-derived family, to carry out

in vivo experiments related to the T-type Ca2+ channel blockade.

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 969

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 12: Recent patents on calcium channel blockers: emphasis on CNS diseases

Cav3.2 channel is also involved in cell proliferation. Its anti-proliferative effects suggest this peptide could be useful inthe treatment of cancer [123].Abbott Laboratories has disclosed a series of acetamide

lactams, with similar formula of the selected compound dis-played in Figure 6,16. In IMR32 cells expressing the humanCav3.2 channel, compounds showed IC50 between 10 and15 µM to block [Ca2+]c transients. However, monitoring T-type currents, some compounds exhibited IC50 in the submi-cromolar range. In an animal model of hyperalgesia inducedby capsaicin, compound 16 (Figure 6) reduced it by 74% [124].

4.4 Blockers of L- and T-type VACCsHilpert has reported the synthesis of 25 bridged tetrahydro-naphthalene derivatives (Figure 7,17). In HEK 293 cellsexpressing the human Cav1.2 channel, IC50 to block [Ca2+]ctransients were in the range 1.02 -- 8.7 µM. In HEK 293 cellsexpressing human Cav3.1, Cav3.2 or Cav3.3 channels, IC50

were in the range 0.19 -- 2.9 µM. In the perfused mouse heart,IC50 values to depress contractility ranged between 6 and42 µM. Thus, the compounds are claimed to be useful inthe treatment of cardiovascular diseases [125].

4.5 Blockers of N- and T-type VACCsNon-selective blockers of N- and T-type channels have beenreported in three patents, where the claimed therapeuticindications were inflammatory pain, migraine and epilepsy.Merck Sharp & Dohme Corp. reports a family of N-

substituted oxindoline derivatives with the general structuregiven in Figure 7,18. ‘Ar’ was either a substituted phenyl ringor a pyrimidine heterocycle; X substituents were mostly halogenatoms and the N-anchored heterocycle was 5- and 6-memberedaza-heterocycles. The 59 compounds were tested in HEK293 cells expressing human Cav2.2, Cav3.1 or Cav3.2 channels.Authors used recordings of calcium currents and monitored

[Ca2+]c transients with fluorescent probes. To block N-typecurrents (Cav2.2), preferred compounds exhibited IC50 below1 µM. This was also the case for T-type currents. The bestcompounds were tested in an acute inflammatory pain modelin rats treated with complete Freund’s adjuvant. Based on theirefficacy to inhibit pain in this model, the authors claim theycould be useful in the treatment of acute pain [126].

Pajouhesh et al., from Zalicus Pharmaceuticals Ltd., havepatented 200 bis-aryl and alkylarylsulfones. Most compoundsare exemplified by the Markush structures indicated inFigure 7,19. In cells expressing N- or T-type channel subtypesusing [Ca2+]c transient monitoring, bis-arylsulfones-derivedcompounds inhibited both channel types with IC50 in thesubmicromolar range but with some selectivity for N-typechannels. However, alkylarylsulfones blocked selectively N-type channels with IC50 in the range 0.1 -- 1 µM. The authorsclaim these compounds could be useful in the treatment ofpain and epilepsy [127].

Heer et al., from Convergence Pharmaceuticals Ltd., havereported the synthesis of piperazine derivatives with the gen-eral molecular structure shown in Figure 7,20, where R1 iseither hydrogen or a methyl group and the phenyl group iscommonly mono-substituted with either CF3 or CN groups.The compounds were tested in HEK 293 cells expressinghuman Cav2.2 or Cav3.2 channels, using the patch-clamptechnique. Several compounds blocked both N- and T-typecurrents with IC50 in a range between 3.5 and 30 µM. Onthe basis of these results, the authors claim that these com-pounds may have therapeutic interest in neuropathic pain,inflammatory pain and migraine [128].

5. Conclusion

In these 23 recent patents reviewed, a few thousand newlysynthesized compounds have been screened to assess their

17

OO

CF3

N

N

HN

O 18

NO

Ar

Het

X

19

SO O

NH

OArR

SO O

RHN

O

R

20

R2

SN

NQuinoline

OO

O

R1

Figure 7. Chemical structure of (17) from the patent on L- and T-type channel ligands described in Section 4.4, and chemical

structures (18) to (20) from patents on N- and T-type channels in Section 4.5, are grouped.

J.-A. Arranz-Tagarro et al.

970 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 13: Recent patents on calcium channel blockers: emphasis on CNS diseases

ability to target a given subtype of VACC expressed in celllines. It is highlighted the increasing interest in T-type chan-nels (nine patents), particularly of its isoform Cav3.2 thathas been implicated in the transmission of neuropathic painat the spinal cord. Great efforts are also being made in block-ers of N-type channels (seven patents) and ligands for thea2/d subunit of VACCs, also focused on pain. Intractablepain of peripheral diabetes neuropathy, herpes, cancer, tri-geminal neuralgia or post-surgery is of particular relevance.Thus, pain is the main focus of most patents (16 out of 23).T-type channel blockers mitigating increased neuronal excit-ability in the epilepsy is another area of interest. Finally,some patents on ligands for L-type channels are also discussed,where blockers of the Cav1.3 isoform are being considered forthe treatment of PD. Also, some T-type channel ligands arebeing tested to mitigate Ab burden in AD. Information onselectivity for a given VACC subtype, on pharmacokinetics,particularly the ability of selected ligands to cross the BBB,and to interact with other drugs at the CYP system is requiredbefore positioning compounds as drug-able potential medi-cines indicated in the treatment of those diseases. Because ofthe widespread expression of various of the VACCs in differ-ent brain areas, it is anticipated that unless highly selectivecompounds were found, intolerable side effects may seriouslylimit the clinical use of the compounds here reviewed.

6. Expert opinion

In this review, 23 patents claiming selectivity of newly synthe-sized compounds for L-type channels (three patents), N-typechannels (seven patents) or T-type channels (nine patents)have been selected. Some additional patents have beenreviewed on compounds that target L- and T-type channels(one patent) and others targeting N- and T-type channels(three patents). The most common therapeutic claim isneuropathic pain; occasionally, inflammatory pain has alsobeen claimed. The success of peptidic w-conotoxin MVIIA(ziconotide) in intractable pain has stimulated the search ofnon-peptide compounds that, by targeting N- and/or T-typechannels or both together, could be therapeutically useful totreat severe pain. The second most often claimed indicationis epilepsy, particularly with T-type channel blockers. It isimportant to highlight that Cav3.1 ligands may enhance cog-nition in AD patients by augmenting neurotransmitterrelease. Another interesting claim is that made for ligands ofL-type channels in PD, that could protect dopaminergicneurons from calcium overload, secondary to fast AP firing.We found also some therapeutic claims for these L-type chan-nel ligands in cerebrovascular and neurodegenerative diseases.

Three patents on L-type channel ligands are presented. Inthe patent of Delgado-Martın et al. [102], the original aspect isthe extraction procedure for marine crambescidines. However,the low potency of the crambescidine mixture should beenhanced with new semisynthetic compounds. On theother hand, the patent covering DHP-benzodiazepine hybrid

structures are interesting from a multitarget point of view;although some in vivo experiments are commented, no biolog-ical data are reported [104]. Finally, the patent of new ligandsfor the L-type channel isoform Cav1.3 aims the importantgoal of slowing down PD progression [103]. Channel selectivityis critical here to avoid intolerable side effects. Also, the abilityof readily crossing the BBB is imperative in this type of drug.

The seven patents covering N-type channel ligands focusmainly on the treatment of neuropathic pain. Some patentsare more valuable because they include data obtained in ani-mal models of neuropathic pain. This is the case of the patentsby Searle et al. [105], Khanna et al. [110] and Li et al. [108]. Someselected compounds from these patents exhibited notable effi-cacy when administered orally to murine models of neuro-pathic pain. An interesting new target is that on the N-typechannel regulatory protein CRMP-2. However, the limitationof this strategy is the peptidic nature of the ligands for suchprotein, thus requiring parenteral administration [111].

In some of the nine patents on T-type channel ligandsreviewed, in vivo experiments are given. This is the case for apatent from Zalicus Pharmaceuticals [112] that reports experi-ments on animal models of epilepsy and pain, showing goodefficacy. Blockade of the Cav3.2 channel by compounds synthe-sized by Abbott [124] is also effective in animal models of hyper-algesia induced by capsaicin, indicating the relationshipbetween those channels and neuropathic pain. The patent ofWeber et al. [120] is interesting taking into account the inhibitionof Ab deposition and improved cognition in animal models ofAD. This is interesting because presynaptic T-type channelsare known to enhance transmitter release and thus blockerscould decrease neurotoxicity linked to excess glutamate release.However, other authors have described that the hit compoundof this patent, ST101, is a T-type Ca2+ channel agonist at sub-nanomolar concentrations. A possibility exists that the in vivooutcomes with ST101 would come from an indirect pharmaco-logical effect on several kinase enzymes, as hypothesized byMoriguchi et al. [121], a Ca2+ promoting action or the expressedCa2+ antagonist activity proposed by Weber et al. Otherwise,the physiological relevance of this compound derives from itscognition enhancer activity.

The three patents reporting ligands for both N- and T-typechannels are interesting because these compounds may exhibitsynergy in treating painful conditions in inflammatory pain,neuropathic pain and migraine. This is the case of the patentof Merck Sharp & Dohme Corp. [126]. that shows in vitro andin vivo data. A major limitation of this multitarget strategy isthat the simultaneous blockade of N- and T-type channelsmay give rise to greater effectiveness but also to additive tox-icity. In fact, the great challenge for future research is tofind out more selective ligands not only for the different iso-forms of VACC subtypes L, N, P/Q, R and T, but also forisoforms that may proof to be tissue specific. In fact, marketedCa2+ antagonists present side effects mainly due to their lackof selectivity. One can realize the opposite is also true, as ahuge inhibition of a specific isoform can derive in severe

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 971

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 14: Recent patents on calcium channel blockers: emphasis on CNS diseases

adverse effects. We emphasize that, for obtaining selective Ca2+

channel ligands, it is not necessary to reach nanomolar activi-ties, but simply that IC50 data for the inhibition of otherisoforms were 10-fold higher. An emerging discussion iswhether a therapeutically useful Ca2+ antagonist has to be asmuch selective as possible, or possessing a broad-spectrumblocking activity. The only way to resolve this argument is todevelop a real highly selective Ca2+ channel blocker, with theaim to figure out the real contribution of any specific isoformto the pathology object of study, and its therapeutic range.Should a dual, or multitargeted, blocking activity on severalCav isoforms the cause of the therapeutic activity of a certainCa2+ antagonist, this has to be empirically characterized bystudying the contribution of each isoform to blockade. Toachieve this difficult goal, it could be interesting to pay moreattention to structural molecular studies that compare severalof the natural toxins that target with high selectivity some ofthe VACC subtypes. These studies would help to the develop-ment of future peptidomimetic drugs and, in addition, to findthe key pharmacophoric groups in the binding with the biolog-ical target, which in turn would be essential for the develop-ment of small drugs, through an epitope-based chemicalconstruction. Sparse information on structure--activity rela-tionships and limited high-throughput electrophysiologicalmethods have hampered the development of the appropriatelead molecule for a given channel type. The knowledge of thethree-dimensional structure in solution of w-toxins is criticalto study the specificity of their interactions with the varioussubtypes of VACCs as well as to define active sites that canserve as models to design and synthesize non-peptide blockers.The w-conotoxins are small peptides containing 24 -- 29amino-acid residues. A surprising finding is that the amino-acid sequence of w-conotoxin MVIIA (a reversible N-typechannel blocker) is closer to that of w-conotoxin MVIIC (anN- and P/Q-type channel blocker) than to the w-conotoxinGVIA (an irreversible N-type channel blocker). These differ-ences are important in order to define the toxin selectivity forN- or P/Q-type channels. The three-dimensional structuresof w-conotoxin GVIA [129], w-conotoxin MVIIA [130] andw-conotoxinMVIIC [131] have been elucidated. The definitionof the structure of other toxins will facilitate their comparisonsand the definition of structural determinants for specific bind-ing to VACC subtypes. This will allow the identification ofpharmacophores to facilitate the synthesis of non-peptidechannel blockers of therapeutic interest [132].Some recent examples may illustrate the challenge to

develop a highly specific molecule to target one singleVACC type. For instance, compound A-1048400 (AbbotLaboratories) is a small molecule with high potency for theN-, P/Q- and T-type channels, lacking L-type channel activ-ity, with no hemodynamic effects, and having antinocipeptiveactions [133]. Also, Neuromed has described a number of com-pounds that show some selectivity for the N-type channelsversus the P/Q-type channels, and with potential therapeuticindications in pain and stroke [134]. The aliphatic monoamine

dodecylamine has also been suggested to be largely selectivefor P/Q-type channels [135]. A major limitation of thesestudies, however, is that authors do not provide a clear struc-ture--activity relationship for a further lead optimizationfocusing on small P/Q-type channel blockers.

A similar strategy to that followed to develop the N-typechannel blocker ziconotide for severe pain treatment [136] couldbe applied to the w-toxin P/Q-type channel blockers withpotential indications in migraine and AD. Thus, peptide cycli-zation has improved the biophysical properties and the activityof w-agatoxins [137]. However, good bioavailability of peptidetoxins is still a challenge, and these molecules do not penetratethe BBB. Furthermore, even if given intrathecally, like the caseof ziconotide, w-agatoxin IVA may have strong adverse effects,as it irreversibly blocks P/Q-type channels. However, structuralinformation from the binding domain of w-agatoxin IVA andw-agatoxin IVB to the a1 subunit of P/Q channels may inspirethe development of selective and more appropriate peptideanalogues. The strategy of narrowing down the amino-acidsequence of the pharmacophore has met with some success.For instance, the analogue compound 4a of w-conotoxinGVIA, which mimics three of its side chains, potently blocksN-type channels in the micromolar range [138]. Additionally,Pfizer designed a small-molecule mimicking three residues ofw-conotoxin MIIA that blocks N-type channels [139]. Furtherdevelopment resulted in the generation of an orally availablesmall molecule that blocked N-type channels, displayingimproved physicochemical properties [140].

A major limitation to the development of selective modula-tors of VACC subtypes is their wide distribution in the CNS.For instance, the P/Q-type channels are expressed in all brainareas, especially in the cerebellum. This could be a challengefor drug development, as P/Q-type channel blockade in thecerebellum may cause gait and movement disorders. In fact,P/Q-type channel knock-out mice exhibit symptoms of ataxiaand dystonia [141,142]. Addressing a particular splice variantexpressed in the brain region of interest could become anapproach to bypass the effects on the cerebellum of such P/Q-type channel ligands [143,144]. Another interesting approachis the development of state-dependent ligands that preferen-tially bind to the inactivated state of the channel. In this man-ner, the ligand will target channels at overactive synapsesunder pathological conditions, thus sparing normal synapses.This kind of compounds could be indicated in migraine,pain and epilepsy, where the pathophysiology involves pro-longed depolarizations of the membrane over seconds orminutes [145]. Conversely, some compounds bind to theopen state of the channel and thereby delay its deactivation.This leads to a facilitation of Ca2+ influx that can be beneficialin certain types of ataxia. These limitations have precludedthe development of non-toxin, small molecules to targetselectively P/Q-type channels for CNS diseases [146].

The hypothesis on a potential therapeutic application ofVACC agonists deserves attention. In a recent study, Arm-strong and Drupeau [147] have expressed the human gene

J.-A. Arranz-Tagarro et al.

972 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 15: Recent patents on calcium channel blockers: emphasis on CNS diseases

encoding TDP43 (TAR DNA-binding protein) in zebrafishlarvae, with the mutation G348C found in patients of amyo-trophic lateral sclerosis (ALS). These larvae showed impairedswimming and increased motor neuron vulnerability, withreduced synaptic fidelity, reduced quantal transmission andmore orphaned presynaptic and postsynaptic structures atthe neuromuscular junction. Remarkably, all behavioral andcellular features were stabilized by chronic treatment witheither L-type VACC agonists FPL64176 or BayK8644, sug-gesting that these compounds could be a novel therapeutic

approach for ALS. A major limitation of these compoundscould be their arrhythmogenic cardiac effects. Thus, VACCagonists, selective for Cav1.3 channels, could be an interestingnovel approach to treat ALS patients.

Declaration of interest

The authors declare no conflict of interest and have receivedno payment in preparation of this manuscript.

BibliographyPapers of special note have been highlighted as

either of interest (�) or of considerable interest(��) to readers.

1. Ringer S. A further contribution

regarding the influence of the. different

constituents of the blood on the

contraction of the heart. J Physiol

1883;4:29-42

2. Ringer S. Further experiments regarding

the influence of small quantities of lime

potassium and other salts on muscular

tissue. J Physiol 1886;7:291-308

3. Locke F. Notiz uber den einfluss,

physiologischer kochsalzl€osung auf die

erregbarkeit von muscel und nerve.

Zentralbl Physiol 1894;8:166-7

4. Overton E. Beitrage zur allgemeinen

muskel- und nerven physiologie III.

Mittheilung. Studien uber die wirkung

der alkali- und erdkali-salze auf

skeletalmuskeln und nerven.

Pflugers Arch 1904;105:176-290

5. Heilbrunn LV, Wiercinski FJ. The action

of various cations on muscle protoplasm.

J Cell Physiol 1947;29:15-32

6. Katz B, Miledi R. The timing of calcium

action during neuromuscular

transmission. J Physiol 1967;189:535-44

7. Garcia AG, Garcia-De-Diego AM,

Gandia L, et al. Calcium signaling and

exocytosis in adrenal chromaffin cells.

Physiol Rev 2006;86:1093-131

8. Garcia AG, Padin F,

Fernandez-Morales JC, et al. Cytosolic

organelles shape calcium signals and

exo-endocytotic responses of chromaffin

cells. Cell Calcium 2012;51:309-20

9. Garcia-Sancho J, de Diego AM,

Garcia AG. Mitochondria and

chromaffin cell function. Pflugers Arch

2012;464:33-41

10. Catterall WA, Leal K, Nanou E.

Calcium channels and short-term

synaptic plasticity. J Biol Chem

2013;288:10742-9

.. A nice review on calcium channels,

calcium ions and synaptic plasticity.

11. Fernandez-Morales JC,

Arranz-Tagarro JA, Calvo-Gallardo E,

et al. Stabilizers of neuronal and

mitochondrial calcium cycling as a

strategy for developing a medicine for

alzheimer’s disease. ACS Chem Neurosci

2012;3:873-83

. A review on the concept of

multi-target ligands including L-type

channels, to treat AD.

12. Yu JT, Chang RC, Tan L. Calcium

dysregulation in alzheimer’s disease: from

mechanisms to therapeutic opportunities.

Prog Neurobiol 2009;89:240-55

13. Chan CS, Gertler TS, Surmeier DJ.

Calcium homeostasis, selective

vulnerability and parkinson’s disease.

Trends Neurosci 2009;32:249-56

14. Grosskreutz J, Van Den Bosch L,

Keller BU. Calcium dysregulation in

amyotrophic lateral sclerosis.

Cell Calcium 2010;47:165-74

15. Berridge MJ. Calcium regulation of

neural rhythms, memory and alzheimer’s

disease. J Physiol 2014;592(Pt 2):281-93

16. Carafoli E. Calcium signaling: a tale for

all seasons. Proc Natl Acad Sci USA

2002;99:1115-22

17. MacDonald JF, Xiong ZG, Jackson MF.

Paradox of ca2+ signaling, cell death and

stroke. Trends Neurosci 2006;29:75-81

18. Triggle DJ. Calcium channel antagonists:

clinical uses -- past, present and future.

Biochem Pharmacol 2007;74:1-9

19. de Diego AM, Lorrio S,

Hernandez-Guijo JM, et al. Multitarget

drugs for stabilization of calcium cycling

and neuroprotection in neurodegenerative

diseases and stroke. In: Botana LM,

Loza MI, editors, Therapeutic targets:

modulation, inhibition, and activation.

John Wiley & Soons, Inc., Hoboken NJ,

USA; 2012. p. 123-97

20. Fatt P, Ginsborg BL. The ionic

requirements for the production of action

potentials in crustacean muscle fibres.

J Physiol 1958;142:516-43

21. Dolphin AC. A short history of voltage-

gated calcium channels. Br J Pharmacol

2006;147(Suppl 1):S56-62

22. Fleckenstein A. History of calcium

antagonists. Circ Res 1983;52:I3-16

23. Catterall WA. Voltage-gated calcium

channels. Cold Spring Harb

Perspect Biol 2011;3:a003947

24. De Diego AM, Gandıa L, Padın F, et al.

Calcium channels for exocytosis and

endocytosis: Pharmacological modulation.

In: Botana LM, editor, Seafood and

freshwater toxins: Pharmacology,

physiology, and detection. Taylor &

Francis Group, USA; 2014. p. 1215

25. Pragnell M, De Waard M, Mori Y, et al.

Calcium channel beta-subunit binds to a

conserved motif in the i-ii cytoplasmic

linker of the alpha 1-subunit. Nature

1994;368:67-70

26. Van Petegem F, Clark KA,

Chatelain FC, et al. Structure of a

complex between a voltage-gated calcium

channel beta-subunit and an alpha-

subunit domain. Nature 2004;429:671-5

27. Berggren PO, Yang SN, Murakami M,

et al. Removal of ca2+ channel

beta3 subunit enhances ca2+ oscillation

frequency and insulin exocytosis. Cell

2004;119:273-84

28. Kang MG, Campbell KP. Gamma

subunit of voltage-activated calcium

channels. J Biol Chem

2003;278:21315-18

29. Chen L, Chetkovich DM, Petralia RS,

et al. Stargazin regulates synaptic

targeting of ampa receptors by two

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 973

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 16: Recent patents on calcium channel blockers: emphasis on CNS diseases

distinct mechanisms. Nature

2000;408:936-43

30. Ertel EA, Campbell KP, Harpold MM,

et al. Nomenclature of voltage-gated

calcium channels. Neuron 2000;25:533-5

31. Doering CJ, Zamponi GW. Molecular

pharmacology of high voltage-activated

calcium channels. J Bioenerg Biomembr

2003;35:491-505

32. Lipkind GM, Fozzard HA. Molecular

modeling of interactions of

dihydropyridines and phenylalkylamines

with the inner pore of the l-type ca2+

channel. Mol Pharmacol

2003;63:499-511

33. Hockerman GH, Peterson BZ, Sharp E,

et al. Construction of a high-affinity

receptor site for dihydropyridine agonists

and antagonists by single amino acid

substitutions in a non-l-type ca2+

channel. Proc Natl Acad Sci USA

1997;94:14906-11

34. Hockerman GH, Dilmac N, Scheuer T,

et al. Molecular determinants of

diltiazem block in domains iiis6 and

ivs6 of l-type ca(2+) channels.

Mol Pharmacol 2000;58:1264-70

35. Boland LM, Morrill JA, Bean BP.

Omega-conotoxin block of n-type

calcium channels in frog and rat

sympathetic neurons. J Neurosci

1994;14:5011-27

36. Hillyard DR, Monje VD, Mintz IM,

et al. A new conus peptide ligand for

mammalian presynaptic ca2+ channels.

Neuron 1992;9:69-77

37. Gandia L, Lara B, Imperial JS, et al.

Analogies and differences between

omega-conotoxins mviic and mviid:

binding sites and functions in bovine

chromaffin cells. Pflugers Arch

1997;435:55-64

38. Bourinet E, Stotz SC, Spaetgens RL,

et al. Interaction of snx482 with domains

iii and iv inhibits activation gating of

alpha(1e) (ca(v)2.3) calcium channels.

Biophys J 2001;81:79-88

39. Carbone E, Calorio C, Vandael DH.

T-type channel-mediated

neurotransmitter release. Pflugers Arch

2014;466:677-87

40. Chuang RS, Jaffe H, Cribbs L, et al.

Inhibition of t-type voltage-gated calcium

channels by a new scorpion toxin.

Nat Neurosci 1998;1:668-74

41. Jarvis MF, Scott VE, McGaraughty S,

et al. A peripherally acting, selective t-

type calcium channel blocker, abt-639,

effectively reduces nociceptive and

neuropathic pain in rats.

Biochem Pharmacol 2014;89:536-44

42. Watanuki S, Matsuura K, Tomura Y,

et al. Synthesis and pharmacological

evaluation of 2-(1-alkylpiperidin-4-yl)-n-

[(1r)-1-(4-fluorophenyl)-2-methylpropyl]

acetamide derivatives as novel

antihypertensive agents. Chem Pharm

Bull (Tokyo) 2012;60:223-34

43. Garcez-Do-Carmo L, Albillos A,

Artalejo AR, et al. R56865 inhibits

catecholamine release from bovine

chromaffin cells by blocking calcium

channels. Br J Pharmacol

1993;110:1149-55

44. Villarroya M, Gandia L, Lara B, et al.

Dotarizine versus flunarizine as calcium

antagonists in chromaffin cells.

Br J Pharmacol 1995;114:369-76

45. Hernandez-Guijo JM, Gandia L,

de Pascual R, et al. Differential effects of

the neuroprotectant lubeluzole on bovine

and mouse chromaffin cell calcium

channel subtypes. Br J Pharmacol

1997;122:275-85

46. Grantham CJ, Main MJ, Cannell MB.

Fluspirilene block of n-type calcium

current in ngf-differentiated pc12 cells.

Br J Pharmacol 1994;111:483-8

47. Enyeart JJ, Biagi BA, Mlinar B.

Preferential block of t-type calcium

channels by neuroleptics in neural crest-

derived rat and human c cell lines.

Mol Pharmacol 1992;42:364-72

48. Valentino K, Newcomb R, Gadbois T,

et al. A selective n-type calcium channel

antagonist protects against neuronal loss

after global cerebral ischemia. Proc Natl

Acad Sci USA 1993;90:7894-7

49. Qian J, Noebels JL. Presynaptic ca2+

channels and neurotransmitter release at

the terminal of a mouse cortical neuron.

J Neurosci 2001;21:3721-8

50. Zamponi GW, Lory P, Perez-Reyes E.

Role of voltage-gated calcium channels in

epilepsy. Pflugers Arch

2010;460:395-403

51. Vohora D, Saraogi P, Yazdani MA, et al.

Recent advances in adjunctive therapy for

epilepsy: focus on sodium channel

blockers as third-generation antiepileptic

drugs. Drugs Today (Barc)

2010;46:265-77

52. Suman-Chauhan N, Webdale L,

Hill DR, et al. Characterisation of [3h]

gabapentin binding to a novel site in rat

brain: homogenate binding studies.

Eur J Pharmacol 1993;244:293-301

53. Gee NS, Brown JP, Dissanayake VU,

et al. The novel anticonvulsant drug,

gabapentin (neurontin), binds to the

alpha2delta subunit of a calcium channel.

J Biol Chem 1996;271:5768-76

54. Dooley DJ, Donovan CM, Meder WP,

et al. Preferential action of gabapentin

and pregabalin at p/q-type voltage-

sensitive calcium channels: inhibition of

k+-evoked [3h]-norepinephrine release

from rat neocortical slices. Synapse

2002;45:171-90

55. Cunningham MO, Woodhall GL,

Thompson SE, et al. Dual effects of

gabapentin and pregabalin on glutamate

release at rat entorhinal synapses in vitro.

Eur J Neurosci 2004;20:1566-76

56. Bayer K, Ahmadi S, Zeilhofer HU.

Gabapentin may inhibit synaptic

transmission in the mouse spinal cord

dorsal horn through a preferential block

of p/q-type ca2+ channels.

Neuropharmacology 2004;46:743-9

57. Niespodziany I, Klitgaard H,

Margineanu DG. Levetiracetam inhibits

the high-voltage-activated ca(2+) current

in pyramidal neurones of rat

hippocampal slices. Neurosci Lett

2001;306:5-8

58. Lee CY, Chen CC, Liou HH.

Levetiracetam inhibits glutamate

transmission through presynaptic p/q-

type calcium channels on the granule

cells of the dentate gyrus. Br J Pharmacol

2009;158:1753-62

59. Pisani A, Bonsi P, Martella G, et al.

Intracellular calcium increase in

epileptiform activity: modulation by

levetiracetam and lamotrigine. Epilepsia

2004;45:719-28

60. Stefani A, Spadoni F, Siniscalchi A, et al.

Lamotrigine inhibits ca2+ currents in

cortical neurons: functional implications.

Eur J Pharmacol 1996;307:113-16

61. Zhu G, Okada M, Murakami T, et al.

Interaction between carbamazepine,

zonisamide and voltage-sensitive ca2+

channel on acetylcholine release in rat

frontal cortex. Epilepsy Res

2002;49:49-60

62. Kostyuk PG, Molokanova EA,

Pronchuk NF, et al. Different action of

ethosuximide on low- and high-threshold

J.-A. Arranz-Tagarro et al.

974 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 17: Recent patents on calcium channel blockers: emphasis on CNS diseases

calcium currents in rat sensory neurons.

Neuroscience 1992;51:755-8

63. McGivern JG. Targeting n-type and t-

type calcium channels for the treatment

of pain. Drug Discov Today

2006;11:245-53

64. Santicioli P, Del Bianco E,

Tramontana M, et al. Release of

calcitonin gene-related peptide like-

immunoreactivity induced by electrical

field stimulation from rat spinal afferents

is mediated by conotoxin-sensitive

calcium channels. Neurosci Lett

1992;136:161-4

65. Smith MT, Cabot PJ, Ross FB, et al.

The novel n-type calcium channel

blocker, am336, produces potent dose-

dependent antinociception after

intrathecal dosing in rats and inhibits

substance p release in rat spinal cord

slices. Pain 2002;96:119-27

.. A contribution on the mechanism of

action of N-type channel ligands to

treat pain.

66. Atanassoff PG, Hartmannsgruber MW,

Thrasher J, et al. Ziconotide, a new n-

type calcium channel blocker,

administered intrathecally for acute

postoperative pain. Reg Anesth Pain Med

2000;25:274-8

67. Mathur VS. Ziconotide: a new

pharmacological class of drug for the

management of pain. Semin Anesth

Perioper Med Pain 2000;19:67-75

68. Carbone E, Lux HD. A low voltage-

activated, fully inactivating ca channel in

vertebrate sensory neurones. Nature

1984;310:501-2

69. McGivern JG, McDonough SI.

Voltage-gated calcium channels as targets

for the treatment of chronic pain.

Curr Drug Targets CNS Neurol Disord

2004;3:457-78

70. Francois A, Laffray S, Pizzoccaro A, et al.

T-type calcium channels in chronic pain:

mouse models and specific blockers.

Pflugers Arch 2014;466:707-17

71. Todorovic SM, Jevtovic-Todorovic V.

Targeting of cav3.2 t-type calcium

channels in peripheral sensory neurons

for the treatment of painful diabetic

neuropathy. Pflugers Arch

2014;466:701-6

72. Kim D, Park D, Choi S, et al. Thalamic

control of visceral nociception mediated

by t-type ca2+ channels. Science

2003;302:117-19

73. Hall KE, Sima AA, Wiley JW.

Voltage-dependent calcium currents are

enhanced in dorsal root ganglion

neurones from the bio bred/worchester

diabetic rat. J Physiol

1995;486(Pt 2):313-22

74. Jagodic MM, Pathirathna S, Nelson MT,

et al. Cell-specific alterations of t-type

calcium current in painful diabetic

neuropathy enhance excitability of

sensory neurons. J Neurosci

2007;27:3305-16

75. Khomula EV, Viatchenko-Karpinski VY,

Borisyuk AL, et al. Specific functioning

of cav3.2 t-type calcium and

trpv1 channels under different types of

stz-diabetic neuropathy.

Biochim Biophys Acta 2013;1832:636-49

76. Jacus MO, Uebele VN, Renger JJ, et al.

Presynaptic cav3.2 channels regulate

excitatory neurotransmission in

nociceptive dorsal horn neurons.

J Neurosci 2012;32:9374-82

77. Choe W, Messinger RB, Leach E, et al.

Tta-p2 is a potent and selective blocker

of t-type calcium channels in rat sensory

neurons and a novel antinociceptive

agent. Mol Pharmacol 2011;80:900-10

78. Hildebrand ME, Smith PL, Bladen C,

et al. A novel slow-inactivation-specific

ion channel modulator attenuates

neuropathic pain. Pain 2011;152:833-43

79. Delfini MC, Mantilleri A, Gaillard S,

et al. Tafa4, a chemokine-like protein,

modulates injury-induced mechanical and

chemical pain hypersensitivity in mice.

Cell Rep 2013;5:378-88

80. Coulter DA, Huguenard JR, Prince DA.

Differential effects of petit mal

anticonvulsants and convulsants on

thalamic neurones: calcium current

reduction. Br J Pharmacol

1990;100:800-6

81. Dogrul A, Gardell LR, Ossipov MH,

et al. Reversal of experimental

neuropathic pain by t-type calcium

channel blockers. Pain 2003;105:159-68

82. Marais E, Klugbauer N, Hofmann F.

Calcium channel alpha(2)delta subunits-

structure and gabapentin binding.

Mol Pharmacol 2001;59:1243-8

83. Sarantopoulos C, McCallum B,

Kwok WM, et al. Gabapentin decreases

membrane calcium currents in injured as

well as in control mammalian primary

afferent neurons. Reg Anesth Pain Med

2002;27:47-57

84. Horga de la Parte JF, Horga A.

[pregabalin: new therapeutic

contributions of calcium channel

alpha2delta protein ligands on epilepsy

and neuropathic pain]. Rev Neurol

2006;42:223-37

85. Di Trapani G, Mei D, Marra C, et al.

Gabapentin in the prophylaxis of

migraine: a double-blind randomized

placebo-controlled study. Clin Ter

2000;151:145-8

86. Hoffmann U, Dilekoz E, Kudo C, et al.

Gabapentin suppresses cortical spreading

depression susceptibility. J Cereb Blood

Flow Metab 2010;30:1588-92

87. Rosa JM, Nanclares C, Orozco A, et al.

Regulation by l-type calcium channels of

endocytosis: an overview. J Mol Neurosci

2012;48:360-7

88. Comunanza V, Marcantoni A,

Vandael DH, et al. Cav1.3 as pacemaker

channels in adrenal chromaffin cells:

specific role on exo- and endocytosis?

Channels (Austin) 2010;4:440-6

89. Mangoni ME, Couette B, Bourinet E,

et al. Functional role of l-type cav1.3 ca2

+ channels in cardiac pacemaker activity.

Proc Natl Acad Sci USA

2003;100:5543-8

90. Marcotti W, Johnson SL, Rusch A, et al.

Sodium and calcium currents shape

action potentials in immature mouse

inner hair cells. J Physiol

2003;552:743-61

91. Olson PA, Tkatch T,

Hernandez-Lopez S, et al.

G-protein-coupled receptor modulation

of striatal cav1.3 l-type ca2+ channels is

dependent on a shank-binding domain.

J Neurosci 2005;25:1050-62

92. Goswami C, Islam MS. Transient

receptor potential channels: what’s

happening? Reflections in the wake of

the 2009 trp meeting, karolinska

institutet, stockholm. Channels (Austin)

2010;4:124-35

93. Chan CS, Guzman JN, Ilijic E, et al.

’Rejuvenation’ protects neurons in mouse

models of parkinson’s disease. Nature

2007;447:1081-6

94. Becker C, Jick SS, Meier CR. Use of

antihypertensives and the risk of

parkinson disease. Neurology

2008;70:1438-44

95. Roda JM, Carceller F, Diez-Tejedor E,

et al. Reduction of infarct size by intra-

arterial nimodipine administered at

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 975

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 18: Recent patents on calcium channel blockers: emphasis on CNS diseases

reperfusion in a rat model of partially

reversible brain focal ischemia. Stroke

1995;26:1888-92

96. Pantoni L, del Ser T, Soglian AG, et al.

Efficacy and safety of nimodipine in

subcortical vascular dementia:

a randomized placebo-controlled trial.

Stroke 2005;36:619-24

97. Pantoni L, Bianchi C, Beneke M, et al.

The scandinavian multi-infarct dementia

trial: a double-blind, placebo-controlled

trial on nimodipine in multi-infarct

dementia. J Neurol Sci 2000;175:116-23

98. Parnetti L, Senin U, Carosi M, et al.

Mental deterioration in old age: results

of two multicenter, clinical trials with

nimodipine. The nimodipine study

group. Clin Ther 1993;15:394-406

99. Kaltenbach LS, Romero E, Becklin RR,

et al. Huntingtin interacting proteins are

genetic modifiers of neurodegeneration.

PLoS Genet 2007;3:e82

100. Croisile B, Trillet M, Fondarai J, et al.

Long-term and high-dose piracetam

treatment of alzheimer’s disease.

Neurology 1993;43:301-5

101. Cumbo E, Ligori LD. Levetiracetam,

lamotrigine, and phenobarbital in

patients with epileptic seizures and

alzheimer’s disease. Epilepsy Behav

2010;17:461-6

102. Consejo Superior de Investigaciones

Cientıficas. Compuestos. marinos

antagonistas de los canales de calcio para

el tratamiento de enfermedades

cardiovasculares. ES2383858; 2012

103. Surmeier DJ. Selective calcium channel

antagonists. US20120196883; 2012

.. Design and study of selective Cav1.3

channel blockers for Parkinson’s

disease.

104. Centro de Investigacion y Desarrollo de

Medicamentos. Tricyclic and tetracyclic

systems with activity on the central-

nervous and vascular systems.

WO2011041989; 2011

105. Abbott Laboratories. Novel substituted

octahydrocyclopenta[c]pyrrol-4-amines as

calcium channel blockers.

US20110281870; 2011

. Almost a thousand compounds

targeting Cav2.2 channels and assessed

in both in vitro and in vivo models,

focused on the treatment of pain.

106. Convergence Pharmaceuticals Ltd.

Piperazine derivatives as cav2.2 calcium

channel blockers. WO2012098400; 2012

107. Convergence Pharmaceuticals Ltd.

Tetrazole compounds as calcium channel

blockers. WO2012004604; 2012

108. Abbvie, Inc. Substituted

octahydropyrrolo[1,2-a]pyrazines as

calcium channel blockers.

WO2013049164; 2013

109. Abbvie, Inc. Substituted

octahydropyrrolo[1,2-a]pyrazine

sulfonamides as calcium channel

blockers. WO2013049174; 2013

110. Indiana University Research and

Technology Corporation. Materials and

method for suppresing inflamatory and

neuropathic pain. WO2012009075;

2012

111. Indiana University Research and

Technology Corp. Peptide conjugates for

treating pain. WO2013184614; 2013

.. Peptides binding VACCs through the

collapsin response mediator protein 2.

112. Zalicus Pharmaceuticals Ltd.

N-piperidinyl acetamide derivatives as

calcium channel blockers. US8377968;

2013

113. Buset Rios N, Rodriguez Esparragon F,

Fernandez-Andrade Rodriguez C, et al.

Vascular and metabolic properties of

manidipine. Nefrologia 2011;31:268-74

114. Mariot P, Vanoverberghe K, Lalevee N,

et al. Overexpression of an alpha 1h

(cav3.2) t-type calcium channel during

neuroendocrine differentiation of human

prostate cancer cells. J Biol Chem

2002;277:10824-33

115. University of Virginia Patent

Foundation. T type calcium channel

blockers and the treatment of diseases.

US20120264804; 2012

116. Merck Sharp & Dohme Corp. Pyrazinyl

phenyl amide t-type calcium channel

antagonists. WO2011022315; 2011

117. Merck Sharp & Dohme Corp.

Heterocycle amide t-type calcium

channel antagonists. WO2011053542;

2011

118. Merck Sharp & Dohme Corp.

Imidazolyl methyl piperidine t-type

calcium channel antagonists.

WO2013148640; 2013

119. Concert Pharmaceuticals, Inc.

Tetrahydronaphthalene derivatives as

t-type calcium channel blocker.

WO2013036434; 2013

. Modulation of pharmacokinetic

properties of mibefradil by

synthesising deuterated derivatives.

120. Zenyaku Kogyo Kabushikikaisha. Use of

cav3.1 selective t-type calcium channel

antagonists. WO2012094615; 2012

121. Moriguchi S, Shioda N, Yamamoto Y,

et al. The t-type voltage-gated calcium

channel as a molecular target of the

novel cognitive enhancer st101:

enhancement of long-term potentiation

and camkii autophosphorylation in rat

cortical slices. J Neurochem

2012;121:44-53

122. Anderson D, Rehak R, Hameed S, et al.

Regulation of the kv4.2 complex by

cav3.1 calcium channels.

Channels (Austin) 2010;4:163-7

123. Universite de Nice Sophia Antipolis.

Identification of novel antagonist toxins

of t-type calcium channel for analgesic

purposes. US20120040909; 2012

124. Abbott Laboratories. Lactam acetamides

as calcium channel blockers.

US20110230459; 2011

125. Hilpert K. Bridged

tetrahydronaphthalene derivatives.

US20110263648; 2011

126. Merck Sharp & Dohme Corp.

N-substituted oxindoline derivatives as

calcium channel blockers. US8273749;

2012

127. Zalicus Pharmaceuticals Ltd.

Bisarylsulfone and dialkylarylsulfone

compounds as calcium channel blockers.

WO2012061926; 2012

128. Convergence Pharmaceuticals Ltd.

Piperazine derivatives for blocking

cav2.2 calcium channels.

WO2011086377; 2011

129. Sevilla P, Bruix M, Santoro J, et al.

Three-dimensional structure of omega-

conotoxin gvia determined by 1h nmr.

Biochem Biophys Res Commun

1993;192:1238-44

130. Kohno T, Kim JI, Kobayashi K, et al.

Three-dimensional structure in solution

of the calcium channel blocker omega-

conotoxin mviia. Biochemistry

1995;34:10256-65

131. Nemoto N, Kubo S, Yoshida T, et al.

Solution structure of omega-conotoxin

mviic determined by nmr.

Biochem Biophys Res Commun

1995;207:695-700

132. Nielsen KJ, Schroeder T, Lewis R.

Structure-activity relationships of omega-

conotoxins at n-type voltage-sensitive

calcium channels. J Mol Recognit

2000;13:55-70

J.-A. Arranz-Tagarro et al.

976 Expert Opin. Ther. Patents (2014) 24(9)

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.

Page 19: Recent patents on calcium channel blockers: emphasis on CNS diseases

133. Scott VE, Vortherms TA, Niforatos W,

et al. A-1048400 is a novel, orally active,

state-dependent neuronal calcium

channel blocker that produces dose-

dependent antinociception without

altering hemodynamic function in rats.

Biochem Pharmacol 2012;83:406-18

134. Yamamoto T, Takahara A. Recent

updates of n-type calcium channel

blockers with therapeutic potential for

neuropathic pain and stroke. Curr Top

Med Chem 2009;9:377-95

135. Beedle AM, Zamponi GW. Block of

voltage-dependent calcium channels by

aliphatic monoamines. Biophys J

2000;79:260-70

136. Schmidtko A, Lotsch J, Freynhagen R,

et al. Ziconotide for treatment of severe

chronic pain. Lancet 2010;375:1569-77

137. Craik DJ, Adams DJ. Chemical

modification of conotoxins to improve

stability and activity. ACS Chem Biol

2007;2:457-68

138. Baell JB, Duggan PJ, Forsyth SA, et al.

Synthesis and biological evaluation of

nonpeptide mimetics of omega-conotoxin

gvia. Bioorg Med Chem

2004;12:4025-37

139. Menzler S, Bikker JA,

Suman-Chauhan N, et al. Design and

biological evaluation of non-peptide

analogues of omega-conotoxin mviia.

Bioorg Med Chem Lett 2000;10:345-7

140. Hu LY, Ryder TR, Rafferty MF, et al.

The discovery of [1-(4-dimethylamino-

benzyl)-piperidin-4-yl]-[4-(3,3-

dimethylbutyl)-phen yl]-(3-methyl-but-2-

enyl)-amine, an n-type ca+2 channel

blocker with oral activity for analgesia.

Bioorg Med Chem 2000;8:1203-12

.. First description of an orally active

analgesic compound acting on

N-type channels.

141. Jun K, Piedras-Renteria ES, Smith SM,

et al. Ablation of p/q-type ca(2+) channel

currents, altered synaptic transmission,

and progressive ataxia in mice lacking the

alpha(1a)-subunit. Proc Natl Acad

Sci USA 1999;96:15245-50

142. Fletcher CF, Tottene A, Lennon VA,

et al. Dystonia and cerebellar atrophy in

cacna1a null mice lacking p/q calcium

channel activity. FASEB J

2001;15:1288-90

143. Bourinet E, Soong TW, Sutton K, et al.

Splicing of alpha 1a subunit gene

generates phenotypic variants of p- and

q-type calcium channels. Nat Neurosci

1999;2:407-15

144. Soong TW, DeMaria CD, Alvania RS,

et al. Systematic identification of splice

variants in human p/q-type channel

alpha1(2.1) subunits: implications for

current density and ca2+-dependent

inactivation. J Neurosci

2002;22:10142-52

145. Mezler M, Hermann D, Swensen AM,

et al. Development and validation of a

fluorescence-based hts assay for the

identification of p/q-type calcium

channel blockers. Comb Chem High

Throughput Screen 2012;15:372-85

.. Analysis of the complexities involved

in the P/Q-type channel ligands to

treat CNS diseases.

146. Nimmrich V, Gross G. P/q-type calcium

channel modulators. Br J Pharmacol

2012;167:741-59

147. Armstrong GA, Drapeau P. Calcium

channel agonists protect against

neuromuscular dysfunction in a genetic

model of tdp-43 mutation in als.

J Neurosci 2013;33:1741-52

.. Innovative hypothesis on the potential

therapeutic application of L-type

channel agonists.

148. Voltage-gated calcium channels.

IUPHAR/BPS Guide to

PHARMACOLOGY. Available from:

http://www.guidetopharmacology.org/

GRAC/FamilyDisplayForward?

familyId=80 [Accessed on 15 June 2014]

AffiliationJuan-Alberto Arranz-Tagarro1,2,3,

Cristobal de los Rıos1,3,

Antonio G Garcıa†1,2,3,4 &

Juan-Fernando Padın1,2,3

†Author for correspondence1Instituto Teofilo Hernando, Universidad

Autonoma de Madrid, Madrid, Spain2Departamento de Farmacologıa y Terap�eutica,

Facultad de Medicina, Universidad Autonoma de

Madrid, Madrid, Spain3Servicio de Farmacologıa Clınica, Instituto de

Investigacion Sanitaria, Hospital Universitario de

La Princesa, Madrid, Spain4Professor,

Hospital Universitario de La Princesa, Spain

Tel: +34 914973120 3121;

Fax: +34 914975380;

E-mail: [email protected]

Recent patents on calcium channel blockers

Expert Opin. Ther. Patents (2014) 24(9) 977

Exp

ert O

pin.

The

r. P

aten

ts D

ownl

oade

d fr

om in

form

ahea

lthca

re.c

om b

y U

nive

rsity

of

New

cast

le o

n 08

/22/

14Fo

r pe

rson

al u

se o

nly.