Ziconotide

Identification

Summary

Ziconotide is an N-type calcium channel antagonist used to manage patients with severe chronic pain who cannot tolerate, or who have not responded adequately to other treatments such as intrathecal morphine and systemic analgesics.

Brand Names
Prialt
Generic Name
Ziconotide
DrugBank Accession Number
DB06283
Background

Ziconotide (also known as SNX-111) is a neurotoxic peptide derived from the cone snail Conus magus comprising 25 amino acids with three disulphide bonds.7,18 Other such peptides, collectively termed conotoxins, exist, and some have shown efficacy in binding specific subsets of calcium channels; ziconotide is used in part because it can be synthesized without loss of proper bond formation or structural elements.5,6 Ziconotide is used to manage severe chronic pain refractory to other methods, through its ability to inhibit N-type calcium channels involved in nociceptive signalling.5,7,8,9,12,14,18

Ziconotide was granted FDA approval on December 28, 2004 for marketing by TerSera therapeutics LLC. under the name Prialt.18 To date, ziconotide is the only calcium channel blocking peptide approved for use by the FDA.7

Type
Small Molecule
Groups
Approved
Structure
Weight
Average: 2639.14
Monoisotopic: 2637.09833574
Chemical Formula
C102H172N36O32S7
Synonyms
  • Ziconotida
  • Ziconotide
External IDs
  • SNX-111

Pharmacology

Indication

Ziconotide is indicated for the management of severe chronic pain in patients refractory to other treatments, and for whom intrathecal therapy is warranted.18

Reduce drug development failure rates
Build, train, & validate machine-learning models
with evidence-based and structured datasets.
See how
Build, train, & validate predictive machine-learning models with structured datasets.
See how
Associated Conditions
Indication TypeIndicationCombined Product DetailsApproval LevelAge GroupPatient CharacteristicsDose Form
Management ofSevere, chronic pain•••••••••••••••••••••• •• •• •••••••••• •• ••••• ••••••••••••••••••
Contraindications & Blackbox Warnings
Prevent Adverse Drug Events Today
Tap into our Clinical API for life-saving information on contraindications & blackbox warnings, population restrictions, harmful risks, & more.
Learn more
Avoid life-threatening adverse drug events with our Clinical API
Learn more
Pharmacodynamics

Ziconotide inhibits N-type calcium channels involved in nociceptive signalling, primarily in the dorsal horn of the spinal cord.5,7,8,9,12,14,18 Although binding is reversible, careful dosing is required to ensure therapeutic effects while minimizing adverse effects, and ziconotide has been described as possessing a narrow therapeutic window.13,18 Patients taking ziconontide may experience cognitive and neuropsychiatric symptoms, reduced levels of consciousness, and elevated serum creatine kinase levels. In addition, ziconotide may increase the risk of infection, including serious cases of meningitis. Patients who withdraw from opiates for ziconotide initiation are advised to taper off the dose.18

Mechanism of action

Nociceptive pain signalling is a complex processing pathway involving peripheral nociceptors, primary afferent nerve fibres, and downstream CNS neurons located in the spinal cord.8,14 Voltage-gated calcium channels (VGCCs) are important regulatory components of neural signalling and include the N-type (Cav2.2) heteromultimeric high-voltage type calcium channels.10 Chronic pain conditions, including inflammatory and neuropathic pain, often involve the aberrant upregulation of VGCC activity through various cellular mechanisms, which can lead to allodynia and hyperalgesia.14

Specifically, N-type channel activation in lightly myelinated Aδ- and C-fibres is known to mediate the release of neurotransmitters substance P (SP), calcitonin gene-related peptide (CGRP), and glutamate, which influence downstream neural activation and pain perception.8,14,15,16,17 In addition, SP and CGRP induce inflammation, potentially exacerbating pre-existing inflammatory chronic pain.14

Ziconotide belongs to the ω-conotoxin class of neurotoxic peptides derived from the cone snail Conus magus which are capable of inhibiting N-type VGCCs.4,5,7,9,18 Although the exact mechanism is yet to be elucidated, it is thought that ω-conotoxins function through direct occlusion of the ion pore to prevent calcium translocation across the membrane.9 Additional studies involving expression of chimeric subunits and molecular modelling suggest that insertion of the ziconotide Met12 residue into a hydrophobic pocket formed by Ile300, Phe302, and Leu305 of Cav2.2 increases binding and may be associated with toxic adverse effects.11

TargetActionsOrganism
AVoltage-dependent N-type calcium channel subunit alpha-1B
inhibitor
Humans
NVoltage-dependent P/Q-type calcium channel subunit alpha-1A
inhibitor
Humans
Absorption

Ziconotide administered intrathecally over one hour in doses between 1 and 10 mcg produced calculated AUC values between 83.6-608 ng*h/mL and Cmax between 16.4-132 ng/mL; these values are approximately dose-proportional. Given the intrathecal administration and low membrane permeability due to its size, ziconotide is expected to remain primarily in the CSF; plasma levels, where detected, remain constant up to nine months following administration.18

Volume of distribution

In patients administered 1-10 mcg intrathecal ziconotide over one hour, the apparent volume of distribution was calculated as 155 ± 263 mL; this value is roughly equivalent to the expected CSF volume.18 Although intravenous administration is not indicated, intravenous administration of between 0.3-10 mcg/kg/day ziconotide resulted in an apparent volume of distribution of 30,460 ± 6366 mL.18

Protein binding

Ziconotide is roughly 50% bound to human plasma proteins.18

Metabolism

Ziconotide is expected to be processed by various peptidases upon entering systemic circulation; no detailed information on ziconotide metabolism has been reported.18

Route of elimination

A small fraction of intravenous ziconotide (< 1%) is recovered in urine.18

Half-life

In patients administered 1-10 mcg intrathecal ziconotide over one hour, the elimination half-life was calculated as 4.6 ± 0.9 hr. Although intravenous administration is not indicated, intravenous administration of between 0.3-10 mcg/kg/day ziconotide resulted in an elimination half-life of 1.3 ± 0.3 hr.18

Clearance

Ziconotide CSF clearance is 0.38 ± 0.56 mL/min while plasma clearance is 270 ± 44 mL/min.18

Adverse Effects
Improve decision support & research outcomes
With structured adverse effects data, including: blackbox warnings, adverse reactions, warning & precautions, & incidence rates. View sample adverse effects data in our new Data Library!
See the data
Improve decision support & research outcomes with our structured adverse effects data.
See a data sample
Toxicity

Symptoms of overdose include neurological effects such as ataxia, nystagmus, stupor, sedation, speech difficulties, dizziness, nausea, and vomiting, and may also cause other effects such as hypotension; overdose is not associated with respiratory depression. In case of overdose, symptom-related supportive care up to and including hospitalization is recommended. Ziconotide has no known antidote, but the withdrawal of ziconotide generally allows patients to clear the drug and recover within 24 hours. As ziconotide does not bind to opiate receptors, opioid antagonists are not effective at ameliorating overdose effects.18

Pathways
Not Available
Pharmacogenomic Effects/ADRs
Not Available

Interactions

Drug Interactions
This information should not be interpreted without the help of a healthcare provider. If you believe you are experiencing an interaction, contact a healthcare provider immediately. The absence of an interaction does not necessarily mean no interactions exist.
DrugInteraction
1,2-BenzodiazepineThe risk or severity of CNS depression can be increased when Ziconotide is combined with 1,2-Benzodiazepine.
AcarboseThe risk or severity of hypoglycemia can be increased when Ziconotide is combined with Acarbose.
AcebutololAcebutolol may increase the arrhythmogenic activities of Ziconotide.
AcetazolamideThe risk or severity of CNS depression can be increased when Acetazolamide is combined with Ziconotide.
AcetohexamideThe risk or severity of hypoglycemia can be increased when Ziconotide is combined with Acetohexamide.
Food Interactions
  • Avoid excessive or chronic alcohol consumption. Ingesting alcohol may increase the CNS depressive effects of ziconotide.

Products

Drug product information from 10+ global regions
Our datasets provide approved product information including:
dosage, form, labeller, route of administration, and marketing period.
Access now
Access drug product information from over 10 global regions.
Access now
Product Ingredients
IngredientUNIICASInChI Key
Ziconotide acetateT2I226K69M914454-03-8Not applicable
Brand Name Prescription Products
NameDosageStrengthRouteLabellerMarketing StartMarketing EndRegionImage
PrialtInjection, solution100 ug/1mLIntrathecalElan Pharmaceuticals2004-12-282013-04-01US flag
PrialtInjection, solution100 μg/mlIntrathecalEsteve Pharmaceuticals Gmb H2016-09-08Not applicableEU flag
PrialtInjection, solution25 ug/1mLIntrathecalTersera Therapeutics Llc2018-10-15Not applicableUS flag
PrialtInjection, solution25 ug/1mLIntrathecalJazz Pharmaceuticals, Inc.2004-12-282022-07-31US flag
PrialtInjection, solution25 ug/1mLIntrathecalElan Pharmaceuticals2004-12-282012-01-01US flag

Categories

ATC Codes
N02BG08 — Ziconotide
Drug Categories
Classification
Not classified
Affected organisms
  • Humans and other mammals

Chemical Identifiers

UNII
7I64C51O16
CAS number
107452-89-1
InChI Key
BPKIMPVREBSLAJ-QTBYCLKRSA-N
InChI
InChI=1S/C102H172N36O32S7/c1-50(2)34-63-91(161)127-62(26-33-171-5)90(160)129-64(35-53-22-24-54(143)25-23-53)92(162)130-65(36-78(148)149)93(163)135-72-48-175-173-45-69(80(108)150)133-86(156)58(18-8-12-29-105)121-76(146)39-117-85(155)66(41-139)131-88(158)61(21-15-32-114-102(111)112)126-96(166)70-46-176-177-47-71(97(167)132-68(43-141)95(165)125-60(87(157)128-63)20-14-31-113-101(109)110)134-89(159)59(19-9-13-30-106)123-81(151)51(3)119-74(144)37-115-83(153)56(16-6-10-27-103)120-75(145)38-116-84(154)57(17-7-11-28-104)124-82(152)55(107)44-172-174-49-73(137-98(72)168)99(169)138-79(52(4)142)100(170)118-40-77(147)122-67(42-140)94(164)136-70/h22-25,50-52,55-73,79,139-143H,6-21,26-49,103-107H2,1-5H3,(H2,108,150)(H,115,153)(H,116,154)(H,117,155)(H,118,170)(H,119,144)(H,120,145)(H,121,146)(H,122,147)(H,123,151)(H,124,152)(H,125,165)(H,126,166)(H,127,161)(H,128,157)(H,129,160)(H,130,162)(H,131,158)(H,132,167)(H,133,156)(H,134,159)(H,135,163)(H,136,164)(H,137,168)(H,138,169)(H,148,149)(H4,109,110,113)(H4,111,112,114)/t51-,52+,55-,56-,57-,58-,59-,60-,61-,62-,63-,64-,65-,66-,67-,68-,69-,70-,71-,72-,73-,79-/m0/s1
IUPAC Name
2-[(1R,4S,7S,13S,16R,21R,24S,27S,30S,33S,36S,39S,42R,45S,48S,54S,60S,63R,68R,71S,77S)-63-amino-13,45,54,60-tetrakis(4-aminobutyl)-4,36-bis(3-carbamimidamidopropyl)-16-carbamoyl-71-[(1R)-1-hydroxyethyl]-7,39,77-tris(hydroxymethyl)-27-[(4-hydroxyphenyl)methyl]-48-methyl-33-(2-methylpropyl)-30-[2-(methylsulfanyl)ethyl]-2,5,8,11,14,23,26,29,32,35,38,41,44,47,50,53,56,59,62,69,72,75,78,85-tetracosaoxo-18,19,65,66,81,82-hexathia-3,6,9,12,15,22,25,28,31,34,37,40,43,46,49,52,55,58,61,70,73,76,79,84-tetracosaazatricyclo[40.37.4.2^{21,68}]pentaoctacontan-24-yl]acetic acid
SMILES
[H][C@]12CSSC[C@]3([H])NC(=O)[C@H](CCCCN)NC(=O)[C@H](C)NC(=O)CNC(=O)[C@H](CCCCN)NC(=O)CNC(=O)[C@H](CCCCN)NC(=O)[C@@H](N)CSSC[C@]([H])(NC(=O)[C@]([H])(CSSC[C@H](NC(=O)[C@H](CCCCN)NC(=O)CNC(=O)[C@H](CO)NC(=O)[C@H](CCCNC(N)=N)NC1=O)C(N)=O)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC1=CC=C(O)C=C1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CO)NC3=O)C(=O)N[C@@]([H])([C@@H](C)O)C(=O)NCC(=O)N[C@@H](CO)C(=O)N2

References

Synthesis Reference

Avi Tovi, Chaim Eidelman, Shimon Shushan, Shai Elster, Alon Hagi, Alexander Ivchenko, Gabriel-Marcus Butilca, Gil Zaoui, Eleonora Alterman, Leah Bar-Oz, Tehila Gadi. "Methods for the production of peptide having a c-terminal amide." Patent WO2006119388A2, issued November 09, 2006.

General References
  1. Skov MJ, Beck JC, de Kater AW, Shopp GM: Nonclinical safety of ziconotide: an intrathecal analgesic of a new pharmaceutical class. Int J Toxicol. 2007 Sep-Oct;26(5):411-21. [Article]
  2. Miljanich GP: Ziconotide: neuronal calcium channel blocker for treating severe chronic pain. Curr Med Chem. 2004 Dec;11(23):3029-40. [Article]
  3. McGivern JG: Ziconotide: a review of its pharmacology and use in the treatment of pain. Neuropsychiatr Dis Treat. 2007 Feb;3(1):69-85. [Article]
  4. Olivera BM, Gray WR, Zeikus R, McIntosh JM, Varga J, Rivier J, de Santos V, Cruz LJ: Peptide neurotoxins from fish-hunting cone snails. Science. 1985 Dec 20;230(4732):1338-43. doi: 10.1126/science.4071055. [Article]
  5. Olivera BM, Cruz LJ, de Santos V, LeCheminant GW, Griffin D, Zeikus R, McIntosh JM, Galyean R, Varga J, Gray WR, et al.: Neuronal calcium channel antagonists. Discrimination between calcium channel subtypes using omega-conotoxin from Conus magus venom. Biochemistry. 1987 Apr 21;26(8):2086-90. doi: 10.1021/bi00382a004. [Article]
  6. Valentino K, Newcomb R, Gadbois T, Singh T, Bowersox S, Bitner S, Justice A, Yamashiro D, Hoffman BB, Ciaranello R, et al.: A selective N-type calcium channel antagonist protects against neuronal loss after global cerebral ischemia. Proc Natl Acad Sci U S A. 1993 Aug 15;90(16):7894-7. doi: 10.1073/pnas.90.16.7894. [Article]
  7. Bourinet E, Zamponi GW: Block of voltage-gated calcium channels by peptide toxins. Neuropharmacology. 2017 Dec;127:109-115. doi: 10.1016/j.neuropharm.2016.10.016. Epub 2016 Oct 15. [Article]
  8. Park J, Luo ZD: Calcium channel functions in pain processing. Channels (Austin). 2010 Nov-Dec;4(6):510-7. doi: 10.4161/chan.4.6.12869. Epub 2010 Nov 1. [Article]
  9. Patel R, Montagut-Bordas C, Dickenson AH: Calcium channel modulation as a target in chronic pain control. Br J Pharmacol. 2018 Jun;175(12):2173-2184. doi: 10.1111/bph.13789. Epub 2017 Apr 26. [Article]
  10. Simms BA, Zamponi GW: Neuronal voltage-gated calcium channels: structure, function, and dysfunction. Neuron. 2014 Apr 2;82(1):24-45. doi: 10.1016/j.neuron.2014.03.016. [Article]
  11. Wang F, Yan Z, Liu Z, Wang S, Wu Q, Yu S, Ding J, Dai Q: Molecular basis of toxicity of N-type calcium channel inhibitor MVIIA. Neuropharmacology. 2016 Feb;101:137-45. doi: 10.1016/j.neuropharm.2015.08.047. Epub 2015 Sep 4. [Article]
  12. Deer TR, Pope JE, Hanes MC, McDowell GC: Intrathecal Therapy for Chronic Pain: A Review of Morphine and Ziconotide as Firstline Options. Pain Med. 2019 Apr 1;20(4):784-798. doi: 10.1093/pm/pny132. [Article]
  13. Sanford M: Intrathecal ziconotide: a review of its use in patients with chronic pain refractory to other systemic or intrathecal analgesics. CNS Drugs. 2013 Nov;27(11):989-1002. doi: 10.1007/s40263-013-0107-5. [Article]
  14. Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW: Calcium-permeable ion channels in pain signaling. Physiol Rev. 2014 Jan;94(1):81-140. doi: 10.1152/physrev.00023.2013. [Article]
  15. Evans AR, Nicol GD, Vasko MR: Differential regulation of evoked peptide release by voltage-sensitive calcium channels in rat sensory neurons. Brain Res. 1996 Mar 18;712(2):265-73. doi: 10.1016/0006-8993(95)01447-0. [Article]
  16. Maggi CA, Tramontana M, Cecconi R, Santicioli P: Neurochemical evidence for the involvement of N-type calcium channels in transmitter secretion from peripheral endings of sensory nerves in guinea pigs. Neurosci Lett. 1990 Jul 3;114(2):203-6. doi: 10.1016/0304-3940(90)90072-h. [Article]
  17. Smith MT, Cabot PJ, Ross FB, Robertson AD, Lewis RJ: 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 Mar;96(1-2):119-27. doi: 10.1016/s0304-3959(01)00436-5. [Article]
  18. FDA Approved Drug Products: Prialt (ziconotide) solution [Link]
KEGG Drug
D06363
PubChem Substance
347910345
ChemSpider
17291932
RxNav
68503
ChEMBL
CHEMBL4594214
Wikipedia
Ziconotide

Clinical Trials

Clinical Trials
PhaseStatusPurposeConditionsCount
4CompletedNot AvailablePeripheral neuropathy1
4CompletedTreatmentNeuropathic Pain1
4WithdrawnTreatmentPainful Myelopathy / Painful Neuropathy1
3CompletedTreatmentPain2
2Active Not RecruitingTreatmentCancer / Intractable Pain / Neuropathic Pain / Pain1

Pharmacoeconomics

Manufacturers
Not Available
Packagers
Not Available
Dosage Forms
FormRouteStrength
Injection, solutionIntrathecal100 ug/1mL
Injection, solutionIntrathecal100 μg/ml
Injection, solutionIntrathecal100 MICROGRAMMI/ML
Injection, solutionIntrathecal25 μg/ml
Injection, solutionIntrathecal25 ug/1mL
Injection, solutionIntrathecal25 MICROGRAMMI/ML
Prices
Not Available
Patents
Patent NumberPediatric ExtensionApprovedExpires (estimated)Region
US5859186No1999-01-122011-12-30US flag
US5364842No1994-11-152016-12-30US flag
US8653033No2014-02-182024-10-01US flag
US8765680No2014-07-012024-10-01US flag
US9707270No2017-07-182024-10-01US flag

Properties

State
Solid
Experimental Properties
Not Available
Predicted Properties
PropertyValueSource
Water Solubility2.36 mg/mLALOGPS
logP-2ALOGPS
logP-23Chemaxon
logS-3ALOGPS
pKa (Strongest Acidic)3.41Chemaxon
pKa (Strongest Basic)12.18Chemaxon
Physiological Charge6Chemaxon
Hydrogen Acceptor Count43Chemaxon
Hydrogen Donor Count42Chemaxon
Polar Surface Area1133.84 Å2Chemaxon
Rotatable Bond Count38Chemaxon
Refractivity671.2 m3·mol-1Chemaxon
Polarizability265.03 Å3Chemaxon
Number of Rings4Chemaxon
Bioavailability0Chemaxon
Rule of FiveNoChemaxon
Ghose FilterNoChemaxon
Veber's RuleNoChemaxon
MDDR-like RuleYesChemaxon
Predicted ADMET Features
Not Available

Spectra

Mass Spec (NIST)
Not Available
Spectra
SpectrumSpectrum TypeSplash Key
Predicted MS/MS Spectrum - 10V, Positive (Annotated)Predicted LC-MS/MSsplash10-0fk9-0000009000-d847dc514df14a8b558a
Predicted MS/MS Spectrum - 10V, Negative (Annotated)Predicted LC-MS/MSsplash10-014i-1000039000-774efc06f84b43d3b999
Predicted MS/MS Spectrum - 20V, Positive (Annotated)Predicted LC-MS/MSsplash10-0fk9-1000019000-bd4ba34b1b6552dddee4
Predicted MS/MS Spectrum - 20V, Negative (Annotated)Predicted LC-MS/MSsplash10-00di-0000092000-598dea2bd89591d978db
Predicted MS/MS Spectrum - 40V, Negative (Annotated)Predicted LC-MS/MSsplash10-002f-4000092000-8518208fdd5886fa6f89
Predicted MS/MS Spectrum - 40V, Positive (Annotated)Predicted LC-MS/MSsplash10-03di-9000021000-fe48cddddf430af0eb08
Chromatographic Properties
Collision Cross Sections (CCS)
Not Available

Targets

Build, predict & validate machine-learning models
Use our structured and evidence-based datasets to unlock new
insights and accelerate drug research.
Learn more
Use our structured and evidence-based datasets to unlock new insights and accelerate drug research.
Learn more
Kind
Protein
Organism
Humans
Pharmacological action
Yes
Actions
Inhibitor
General Function
Voltage-gated calcium channel activity
Specific Function
Voltage-sensitive calcium channels (VSCC) mediate the entry of calcium ions into excitable cells and are also involved in a variety of calcium-dependent processes, including muscle contraction, hor...
Gene Name
CACNA1B
Uniprot ID
Q00975
Uniprot Name
Voltage-dependent N-type calcium channel subunit alpha-1B
Molecular Weight
262493.84 Da
References
  1. Olivera BM, Cruz LJ, de Santos V, LeCheminant GW, Griffin D, Zeikus R, McIntosh JM, Galyean R, Varga J, Gray WR, et al.: Neuronal calcium channel antagonists. Discrimination between calcium channel subtypes using omega-conotoxin from Conus magus venom. Biochemistry. 1987 Apr 21;26(8):2086-90. doi: 10.1021/bi00382a004. [Article]
  2. Valentino K, Newcomb R, Gadbois T, Singh T, Bowersox S, Bitner S, Justice A, Yamashiro D, Hoffman BB, Ciaranello R, et al.: A selective N-type calcium channel antagonist protects against neuronal loss after global cerebral ischemia. Proc Natl Acad Sci U S A. 1993 Aug 15;90(16):7894-7. doi: 10.1073/pnas.90.16.7894. [Article]
  3. Bourinet E, Zamponi GW: Block of voltage-gated calcium channels by peptide toxins. Neuropharmacology. 2017 Dec;127:109-115. doi: 10.1016/j.neuropharm.2016.10.016. Epub 2016 Oct 15. [Article]
  4. Park J, Luo ZD: Calcium channel functions in pain processing. Channels (Austin). 2010 Nov-Dec;4(6):510-7. doi: 10.4161/chan.4.6.12869. Epub 2010 Nov 1. [Article]
  5. Patel R, Montagut-Bordas C, Dickenson AH: Calcium channel modulation as a target in chronic pain control. Br J Pharmacol. 2018 Jun;175(12):2173-2184. doi: 10.1111/bph.13789. Epub 2017 Apr 26. [Article]
  6. Wang F, Yan Z, Liu Z, Wang S, Wu Q, Yu S, Ding J, Dai Q: Molecular basis of toxicity of N-type calcium channel inhibitor MVIIA. Neuropharmacology. 2016 Feb;101:137-45. doi: 10.1016/j.neuropharm.2015.08.047. Epub 2015 Sep 4. [Article]
  7. Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW: Calcium-permeable ion channels in pain signaling. Physiol Rev. 2014 Jan;94(1):81-140. doi: 10.1152/physrev.00023.2013. [Article]
  8. FDA Approved Drug Products: Prialt (ziconotide) solution [Link]
Kind
Protein
Organism
Humans
Pharmacological action
No
Actions
Inhibitor
Curator comments
The reported inhibition was low, even at relatively high peptide concentrations; this interaction is unlikely to play a substantial role in vivo.
General Function
Voltage-gated calcium channel activity
Specific Function
Voltage-sensitive calcium channels (VSCC) mediate the entry of calcium ions into excitable cells and are also involved in a variety of calcium-dependent processes, including muscle contraction, hor...
Gene Name
CACNA1A
Uniprot ID
O00555
Uniprot Name
Voltage-dependent P/Q-type calcium channel subunit alpha-1A
Molecular Weight
282362.39 Da
References
  1. Wang F, Yan Z, Liu Z, Wang S, Wu Q, Yu S, Ding J, Dai Q: Molecular basis of toxicity of N-type calcium channel inhibitor MVIIA. Neuropharmacology. 2016 Feb;101:137-45. doi: 10.1016/j.neuropharm.2015.08.047. Epub 2015 Sep 4. [Article]

Drug created at March 19, 2008 16:21 / Updated at February 02, 2024 22:55