Skip to main content
Erschienen in: Cardiovascular Toxicology 2/2017

Open Access 28.03.2016

Anti-addiction Drug Ibogaine Prolongs the Action Potential in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes

verfasst von: Lena Rubi, Daniel Eckert, Stefan Boehm, Karlheinz Hilber, Xaver Koenig

Erschienen in: Cardiovascular Toxicology | Ausgabe 2/2017

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Ibogaine is a plant alkaloid used as anti-addiction drug in dozens of alternative medicine clinics worldwide. Recently, alarming reports of life-threatening cardiac arrhythmias and cases of sudden death associated with the ingestion of ibogaine have accumulated. Using whole-cell patch clamp recordings, we assessed the effects of ibogaine and its main metabolite noribogaine on action potentials in human ventricular-like cardiomyocytes derived from induced pluripotent stem cells. Therapeutic concentrations of ibogaine and its long-lived active metabolite noribogaine significantly retarded action potential repolarization in human cardiomyocytes. These findings represent the first experimental proof that ibogaine application entails a cardiac arrhythmia risk for humans. In addition, they explain the clinically observed delayed incidence of cardiac adverse events several days after ibogaine intake. We conclude that therapeutic concentrations of ibogaine retard action potential repolarization in the human heart. This may give rise to a prolongation of the QT interval in the electrocardiogram and cardiac arrhythmias.

Introduction

The plant alkaloid ibogaine exerts convincing anti-addictive properties, but has never been approved as anti-addiction medication [1, 2]. Largely because of ibogaine’s status as banned substance in the USA since 1970, the further development of the alkaloid’s use in addiction therapy took place outside conventional clinical and medical settings [1]. Ten years ago, Frank Vocci, at that time director of anti-addiction drug development at the National Institute on Drug Abuse, termed ibogaine therapy “a vast, uncontrolled experiment” [3]. Nevertheless, the ibogaine “medical subculture” has continued to grow, with dozens of alternative medicine clinics operating worldwide [1, 2].
Recently, alarming reports of life-threatening cardiac arrhythmias and sudden death cases [412], temporally associated with the ingestion of ibogaine, have accumulated. We [1316] and others [17] hypothesized that these were related to the drug’s propensity to block human ether-a-go-go-related gene (hERG) potassium channels in the heart, which can result in retardation of ventricular action potential (AP) repolarization and prolongation of the QT interval in the electrocardiogram (ECG) [18]. QT prolongation—indeed observed in people after ibogaine intake (e.g. [2, 4, 5])—is known to be associated with an increased risk of life-threatening torsade de pointes (TdP) arrhythmias [18].
Up to now, the inhibition of currents through heterologously expressed hERG channels by ibogaine (IC50 value, drug concentration needed for half-maximum inhibition = 3–4 µM [13, 14, 17]) remains the sole experimental evidence for the drug’s proarrhythmic potential in humans; data on human cardiomyocytes are still lacking. Moreover, two observations have cast reasonable doubts on ibogaine’s cardiotoxic effects: first, although hERG channel inhibition should retard AP repolarization, the drug significantly shortens AP duration in guinea pig ventricular cardiomyocytes [14]), a well-established model system for human myocytes. Secondly, cardiac adverse events in humans often occur many hours or even several days after ibogaine intake [2], albeit the drug has a half-life in human plasma of 4–7 h only [2, 19].

Methods

Details of our experimental procedures are described elsewhere [14]. Action potentials (APs) were recorded from “ventricular-like” human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CM, Cellectis, Sweden) in the current-clamp mode of the whole-cell patch clamp technique at room temperature (22 ± 1 °C). Cardiomyocytes were classified as ventricular-like if their APs showed a distinct “shoulder” (plateau or flat repolarization phase) prior to a final steep phase of repolarization. APs were elicited at 1 Hz by rectangular current pulses of 4 ms duration at 125 % threshold level. The pipette solution contained 10 mM NaCl, 140 mM KCl, 2 mM EGTA, 1 mM MgCl2, 0.1 mM Na-GTP, 5 mM Mg-ATP, 10 mM Hepes, and pH = 7.2 adjusted with KOH. The cells were bathed in 140 mM NaCl, 4 mM KCl, 2 mM CaCl2, 2 mM MgCl2, 5 mM HEPES, 5 mM glucose, and pH = 7.4 adjusted with NaOH.
Whole-cell currents through hERG potassium channels heterologously expressed in TSA-201 cells were recorded as in our previous studies [13, 14].

Results and Discussion

By assessing the effects of ibogaine and its main metabolite noribogaine on APs in cardiomyocytes derived from human induced pluripotent stem cells, we provide the first experimental proof that ibogaine application does indeed entail a cardiac risk for humans, and we further unravel the mystery of the surprising longevity of the drug’s potential to cause arrhythmias. Thus, Fig. 1 shows that 3 µM ibogaine, a concentration at the IC50 value for hERG channel inhibition [13, 14, 17] and well within the free plasma concentration range reached in humans after drug intake (up to 11 µM) [14], significantly prolongs the AP and flattens its repolarization phase in human cardiomyocytes [Fig. 1a (top), b]. This effect was fully reversible after washout of the drug (data not shown). Similar to ibogaine, also its long-lived active metabolite noribogaine (plasma half-life, 28–49 h [20]) prolonged the human cardiac AP [Fig. 1a (bottom), b]. This observation goes along with noribogaine’s inhibitory action on hERG potassium channels (IC50 = 3 µM: Fig. 2; [17]), which very closely resembles hERG inhibition by ibogaine [13, 14, 17]. It further provides an explanation for the delayed incidence of cardiac adverse events, which may occur even several days after ibogaine intake.
Based on these unprecedented findings, we propose the following sequence to explain the cardiotoxicity associated with ibogaine ingestion (Fig. 1c): (1) inhibition of repolarizing hERG potassium channels by ibogaine and its metabolite noribogaine, followed by (2) retardation of ventricular AP repolarization, and finally (3) prolongation of the QT interval in the electrocardiogram (ECG), ultimately paving the way for life-threatening TdP arrhythmias. Because of its long half-life in human plasma [20], we consider noribogaine, rather than ibogaine itself, as the crucial molecule triggering this sequence of deleterious events.
Since drug-induced AP prolongation in ventricular cardiomyocytes directly correlates with prolonged QT intervals in the ECG [18], the actions of ibogaine and noribogaine reported herein can explain previously published case reports of QT interval prolongation triggered by single doses of ibogaine and lasting several days (e.g. [2, 4, 5]). hERG channel inhibition and concomitant retardation of AP repolarization caused by therapeutic plasma concentrations of any drug are well-known predictors of an increased risk for TdP arrhythmias and sudden cardiac death [18]. Moreover, formerly approved drugs with ibogaine-like effects on in vitro cardiac electrophysiology, e.g. cisapride and astemizole, have been withdrawn from the market because of the unbearable TdP arrhythmia risk associated with their application [18].
We therefore conclude that the use of ibogaine, at doses currently administered to drug addicts, must be considered a dangerous practice. Arrhythmias may emerge due to drug-induced QT interval prolongation resulting from retarded cardiac action potential repolarization.

Acknowledgments

This work was funded by the Austrian Science Fund (FWF, P23060 to K.H.). Ibogaine and noribogaine were kindly donated by Sacrament of Transition (Maribor, Slovenia). Open access funding provided by Austrian Science Fund (FWF).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Literatur
1.
Zurück zum Zitat Brown, T. K. (2013). Ibogaine in the treatment of substance dependence. Current Drug Abuse Reviews, 6, 3–16.CrossRefPubMed Brown, T. K. (2013). Ibogaine in the treatment of substance dependence. Current Drug Abuse Reviews, 6, 3–16.CrossRefPubMed
2.
Zurück zum Zitat Alper, K. R., Stajic, M., & Gill, J. R. (2012). Fatalities temporally associated with the ingestion of ibogaine. Journal of Forensic Sciences, 57, 398–412.CrossRefPubMed Alper, K. R., Stajic, M., & Gill, J. R. (2012). Fatalities temporally associated with the ingestion of ibogaine. Journal of Forensic Sciences, 57, 398–412.CrossRefPubMed
3.
Zurück zum Zitat Vastag, B. (2005). Addiction research. Ibogaine therapy: A ‘vast, uncontrolled experiment’. Science, 308, 345–346.CrossRefPubMed Vastag, B. (2005). Addiction research. Ibogaine therapy: A ‘vast, uncontrolled experiment’. Science, 308, 345–346.CrossRefPubMed
4.
Zurück zum Zitat Hoelen, D. W., Spiering, W., & Valk, G. D. (2009). Long-QT syndrome induced by the antiaddiction drug ibogaine. New England Journal of Medicine, 360, 308–309.CrossRefPubMed Hoelen, D. W., Spiering, W., & Valk, G. D. (2009). Long-QT syndrome induced by the antiaddiction drug ibogaine. New England Journal of Medicine, 360, 308–309.CrossRefPubMed
5.
Zurück zum Zitat Paling, F. P., Andrews, L. M., Valk, G. D., & Blom, H. J. (2012). Life-threatening complications of ibogaine: three case reports. Netherlands Journal of Medicine, 70, 422–424.PubMed Paling, F. P., Andrews, L. M., Valk, G. D., & Blom, H. J. (2012). Life-threatening complications of ibogaine: three case reports. Netherlands Journal of Medicine, 70, 422–424.PubMed
6.
Zurück zum Zitat Pleskovic, A., Gorjup, V., Brvar, M., & Kozelj, G. (2012). Ibogaine-associated ventricular tachyarrhythmias. Clinical Toxicology, 50, 157.CrossRefPubMed Pleskovic, A., Gorjup, V., Brvar, M., & Kozelj, G. (2012). Ibogaine-associated ventricular tachyarrhythmias. Clinical Toxicology, 50, 157.CrossRefPubMed
7.
Zurück zum Zitat Papadodima, S. A., Dona, A., Evaggelakos, C. I., Goutas, N., & Athanaselis, S. A. (2013). Ibogaine related sudden death: A case report. Journal of Forensic and Legal Medicine, 20, 809–811.CrossRefPubMed Papadodima, S. A., Dona, A., Evaggelakos, C. I., Goutas, N., & Athanaselis, S. A. (2013). Ibogaine related sudden death: A case report. Journal of Forensic and Legal Medicine, 20, 809–811.CrossRefPubMed
8.
Zurück zum Zitat Mazoyer, C., Carlier, J., Boucher, A., Peoc’h, M., Lemeur, C., & Gaillard, Y. (2013). Fatal case of a 27-year-old male after taking iboga in withdrawal treatment: GC-MS/MS determination of ibogaine and ibogamine in iboga roots and postmortem biological material. Journal of Forensic Sciences, 58, 1666–1672.CrossRefPubMed Mazoyer, C., Carlier, J., Boucher, A., Peoc’h, M., Lemeur, C., & Gaillard, Y. (2013). Fatal case of a 27-year-old male after taking iboga in withdrawal treatment: GC-MS/MS determination of ibogaine and ibogamine in iboga roots and postmortem biological material. Journal of Forensic Sciences, 58, 1666–1672.CrossRefPubMed
9.
Zurück zum Zitat Jalal, S., Daher, E., & Hilu, R. (2013). A case of death due to ibogaine use for heroin addiction: Case report. The American Journal on Addictions, 22, 302.CrossRefPubMed Jalal, S., Daher, E., & Hilu, R. (2013). A case of death due to ibogaine use for heroin addiction: Case report. The American Journal on Addictions, 22, 302.CrossRefPubMed
10.
Zurück zum Zitat Asua, I. (2013). Growing menace of ibogaine toxicity. British Journal of Anaesthesia, 111, 1029–1030.CrossRefPubMed Asua, I. (2013). Growing menace of ibogaine toxicity. British Journal of Anaesthesia, 111, 1029–1030.CrossRefPubMed
11.
Zurück zum Zitat Vlaanderen, L., Martial, L. C., Franssen, E. J., van der Voort, P. H., Oosterwerff, E., & Somsen, G. A. (2014). Cardiac arrest after ibogaine ingestion. Clinical Toxicology, 52, 642–643.CrossRefPubMed Vlaanderen, L., Martial, L. C., Franssen, E. J., van der Voort, P. H., Oosterwerff, E., & Somsen, G. A. (2014). Cardiac arrest after ibogaine ingestion. Clinical Toxicology, 52, 642–643.CrossRefPubMed
13.
Zurück zum Zitat Koenig, X., Kovar, M., Boehm, S., Sandtner, W., & Hilber, K. (2014). Anti-addiction drug ibogaine inhibits hERG channels: A cardiac arrhythmia risk. Addiction Biology, 19, 237–239.CrossRefPubMed Koenig, X., Kovar, M., Boehm, S., Sandtner, W., & Hilber, K. (2014). Anti-addiction drug ibogaine inhibits hERG channels: A cardiac arrhythmia risk. Addiction Biology, 19, 237–239.CrossRefPubMed
14.
Zurück zum Zitat Koenig, X., Kovar, M., Rubi, L., Mike, A. K., Lukacs, P., Gawali, V. S., et al. (2013). Anti-addiction drug ibogaine inhibits voltage-gated ionic currents: A study to assess the drug’s cardiac ion channel profile. Toxicology and Applied Pharmacology, 273, 259–268.CrossRefPubMedPubMedCentral Koenig, X., Kovar, M., Rubi, L., Mike, A. K., Lukacs, P., Gawali, V. S., et al. (2013). Anti-addiction drug ibogaine inhibits voltage-gated ionic currents: A study to assess the drug’s cardiac ion channel profile. Toxicology and Applied Pharmacology, 273, 259–268.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Thurner, P., Gafar, H., Gawali, V. S., Kudlacek, O., Zezula, J., Hilber, K., et al. (2014). Mechanism of hERG channel block by the psychoactive indole alkaloid ibogaine. Journal of Pharmacology and Experimental Therapeutics, 348, 346–358.CrossRefPubMed Thurner, P., Gafar, H., Gawali, V. S., Kudlacek, O., Zezula, J., Hilber, K., et al. (2014). Mechanism of hERG channel block by the psychoactive indole alkaloid ibogaine. Journal of Pharmacology and Experimental Therapeutics, 348, 346–358.CrossRefPubMed
17.
Zurück zum Zitat Alper, K., Bai, R., Liu, N., Fowler, S. J., Huang, X. P., Priori, S. G., & Ruan, Y. (2016). hERG blockade by Iboga alkaloids. Cardiovascular Toxicology, 16, 14–22.CrossRefPubMed Alper, K., Bai, R., Liu, N., Fowler, S. J., Huang, X. P., Priori, S. G., & Ruan, Y. (2016). hERG blockade by Iboga alkaloids. Cardiovascular Toxicology, 16, 14–22.CrossRefPubMed
18.
Zurück zum Zitat Redfern, W. S., Carlsson, L., Davis, A. S., Lynch, W. G., MacKenzie, I., Palethorpe, S., et al. (2003). Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: Evidence for a provisional safety margin in drug development. Cardiovascular Research, 58, 32–45.CrossRefPubMed Redfern, W. S., Carlsson, L., Davis, A. S., Lynch, W. G., MacKenzie, I., Palethorpe, S., et al. (2003). Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: Evidence for a provisional safety margin in drug development. Cardiovascular Research, 58, 32–45.CrossRefPubMed
19.
Zurück zum Zitat Mash, D. C., Kovera, C. A., Buck, B. E., Norenberg, M. D., Shapshak, P., Hearn, W. L., & Sanchez-Ramos, J. (1998). Medication development of ibogaine as a pharmacotherapy for drug dependence. Annals of the New York Academy of Sciences, 844, 274–292.CrossRefPubMed Mash, D. C., Kovera, C. A., Buck, B. E., Norenberg, M. D., Shapshak, P., Hearn, W. L., & Sanchez-Ramos, J. (1998). Medication development of ibogaine as a pharmacotherapy for drug dependence. Annals of the New York Academy of Sciences, 844, 274–292.CrossRefPubMed
20.
Zurück zum Zitat Glue, P., Lockhart, M., Lam, F., Hung, N., Hung, C. T., & Friedhoff, L. (2015). Ascending-dose study of noribogaine in healthy volunteers: pharmacokinetics, pharmacodynamics, safety, and tolerability. Journal of Clinical Pharmacology, 55, 189–194.CrossRefPubMed Glue, P., Lockhart, M., Lam, F., Hung, N., Hung, C. T., & Friedhoff, L. (2015). Ascending-dose study of noribogaine in healthy volunteers: pharmacokinetics, pharmacodynamics, safety, and tolerability. Journal of Clinical Pharmacology, 55, 189–194.CrossRefPubMed
21.
Zurück zum Zitat Obach, R. S., Pablo, J., & Mash, D. C. (1998). Cytochrome P4502D6 catalyzes the O-demethylation of the psychoactive alkaloid ibogaine to 12-hydroxyibogamine. Drug Metabolism and Disposition, 26, 764–768.PubMed Obach, R. S., Pablo, J., & Mash, D. C. (1998). Cytochrome P4502D6 catalyzes the O-demethylation of the psychoactive alkaloid ibogaine to 12-hydroxyibogamine. Drug Metabolism and Disposition, 26, 764–768.PubMed
22.
Zurück zum Zitat Mash, D. C., Kovera, C. A., Pablo, J., Tyndale, R., Ervin, F. R., Kamlet, J. D., & Hearn, W. L. (2001). Ibogaine in the treatment of heroin withdrawal. Alkaloids Chemistry Biology, 56, 155–171.CrossRefPubMed Mash, D. C., Kovera, C. A., Pablo, J., Tyndale, R., Ervin, F. R., Kamlet, J. D., & Hearn, W. L. (2001). Ibogaine in the treatment of heroin withdrawal. Alkaloids Chemistry Biology, 56, 155–171.CrossRefPubMed
Metadaten
Titel
Anti-addiction Drug Ibogaine Prolongs the Action Potential in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes
verfasst von
Lena Rubi
Daniel Eckert
Stefan Boehm
Karlheinz Hilber
Xaver Koenig
Publikationsdatum
28.03.2016
Verlag
Springer US
Erschienen in
Cardiovascular Toxicology / Ausgabe 2/2017
Print ISSN: 1530-7905
Elektronische ISSN: 1559-0259
DOI
https://doi.org/10.1007/s12012-016-9366-y

Weitere Artikel der Ausgabe 2/2017

Cardiovascular Toxicology 2/2017 Zur Ausgabe