ArticlePDF AvailableLiterature Review

Abstract

The chapter discusses ibogaine, which is a naturally occurring plant alkaloid with a history of use as a medicinal and ceremonial agent in West Central Africa and has been alleged to be effective in the treatment of drug abuse. The National Institute on Drug Abuse (NIDA) has given significant support to animal research, and the U.S. Food and Drug Administration (FDA) has approved Phase I studies in humans. The chapter discusses the first International Conference on Ibogaine. A major focus of the Conference was the possible mechanism(s) of action of ibogaine. Another important focus of the Conference was to discuss human experience with ibogaine and preclinical and clinical evidence of efficacy and safety. The Conference also featured presentations related to the sociological and anthropological aspects of the sacramental context of the use of iboga in Africa and the distinctive ibogaine subculture of the U.S and Europe. Ibogaine is the most abundant alkaloid in the root bark of the Apocynaceous shrub Tabernanthe iboga, which grows in West Central Africa. The chapter presents a timeline that outlines the historical events relating to the development of ibogaine as a treatment for drug dependence. Ibogaine and serotonin both contain an indole ring in their structure, and ibogaine has been shown to bind to the serotonin transporter and to increase serotonin levels in the nucleus accumbens (NAc). Stereotypy is a methodologic issue that might explain some of the disparate results regarding ibogaine's interaction with the locomotor response to cocaine.
——Chapter 1——
IBOGAINE: A REVIEW
Kenneth R. Alper
Departments of Psychiatry and Neurology
New York University School of Medicine
New York, NY 10016
I. Introduction, Chemical Properties, and Historical Time Line ....................................
A. Introduction............................................................................................................
B. Chemical Structure and Properties ........................................................................
C. Historical Time Line ..............................................................................................
II. Mechanisms of Action.................................................................................................
A. Neurotransmitter Activities....................................................................................
B. Discrimination Studies...........................................................................................
C. Effects on Neuropeptides.......................................................................................
D. Possible Effects on Neuroadaptations Related to Drug Sensitization
or Tolerance ...........................................................................................................
III. Evidence of Efficacy in Animal Models.......................................................................
A. Drug Self-Administration ......................................................................................
B. Acute Opioid Withdrawal......................................................................................
C. Conditioned Place Preference................................................................................
D. Locomotor Activity................................................................................................
E. Dopamine Efflux....................................................................................................
IV Evidence of Efficacy and Subjective Effects in Humans............................................
A. Evidence of Efficacy..............................................................................................
B. Subjective Effects ..................................................................................................
V. Pharmacokinetics .........................................................................................................
A. Absorption..............................................................................................................
B. Distribution ............................................................................................................
C. Metabolism ............................................................................................................
D. Excretion................................................................................................................
VI. Safety ...........................................................................................................................
A. Neurotoxicity .........................................................................................................
B. Cardiovascular Effects ...........................................................................................
C. Fatalities.................................................................................................................
D. Abuse Liability ......................................................................................................
VII. Learning, Memory, and Neurophysiology...................................................................
A. Learning, Memory, and Addiction.........................................................................
B. Effects of Ibogaine on Learning and Memory ......................................................
C. Ibogaine and the EEG ............................................................................................
D. Goutarel’s Hypothesis............................................................................................
VIII. Anthropological and Sociological Perspectives ..........................................................
THE ALKALOIDS, Vol.56 Copyright © 2001 by Academic Press
0099-9598/01 $35.00 All rights of reproduction in any form reserved
1
IX. Economic and Political Perspectives...........................................................................
A. Economic Incentives and the Development of Ibogaine.......................................
B. Political Issues .......................................................................................................
X. Conclusions..................................................................................................................
References....................................................................................................................
I. Introduction and Historical Time Line
A. Introduction
Ibogaine, a naturally occurring plant alkaloid with a history of use as a
medicinal and ceremonial agent in West Central Africa, has been alleged to be
effective in the treatment of drug abuse. The National Institute on Drug Abuse
(NIDA) has given signicant support to animal research, and the U.S. Food and
Drug Administration (FDA) has approved Phase I studies in humans. Evidence
for ibogaines effectiveness includes a substantial preclinical literature on
reduced drug self-administration and withdrawal in animals, and case reports in
humans. There is relatively little nancial incentive for its development by the
pharmaceutical industry because ibogaine is isolated from a botanical source in
which it naturally occurs, and its chemical structure cannot be patented. This has
left the academic community and the public sector with a crucial role in research
on ibogaine, which was a major reason for organizing the First International
Conference on Ibogaine.
A major focus of the Conference was the possible mechanism(s) of action of
ibogaine. Ibogaine is of interest because it appears to have a novel mechanism of
action distinct from other existing pharmacotherapeutic approaches to addiction,
and it potentially could provide a paradigm for understanding the neurobiology
of addiction and the development of new treatments. Another important focus of
the Conference was to review human experience with ibogaine and preclinical
and clinical evidence of efcacy and safety. The Conference also featured presen-
tations related to the sociological and anthropological aspects of the sacramental
context of the use of iboga in Africa and the distinctive ibogaine subculture of the
United States and Europe.
B. Chemical Structure and Properties
Ibogaine (10-methoxyibogamine) (Figure 1) is an indole alkaloid with
molecular formula C20H26N20 and molecular weight 310.44. Ibogaine is the most
abundant alkaloid in the root bark of the Apocynaceous shrub Tabernanthe iboga,
which grows in West Central Africa. In the dried root bark, the part of the plant
2kenneth r. alper
in which alkaloid content is highest, total alkaloid content is reportedly 5 to 6%
(1).
Ibogaine has a melting point of 153°, a pKaof 8.1 in 80% methylcellosolve,
and it crystallizes as prismatic needles from ethanol. Ibogaine is levorotatory [α]D
53°(in 95% ethanol), soluble in ethanol, ether, chloroform, acetone and
benzene, but it is practically insoluble in water. Ibogaine is decomposed by the
action of heat and light. Ibogaine hydrochloride decomposes at 299°, is also
levorotatory [α]D63°(ethanol), [α]D49°(H2O), and is soluble in water,
methanol, and ethanol, slightly soluble in acetone and chloroform, and practically
insoluble in ether (2). The X-ray crystal analysis that conrmed the structure of
ibogaine has been described (3). The literature provides references to the mass
spectrum of ibogaine (4), and the proton (5,6) and the 13C (7-9) NMR spectra of
ibogaine and other iboga alkaloids. Analytic chemical methods for extraction,
derivatization, and detection of ibogaine utilizing combined gas chromatography-
mass spectometry have been described (10-13).
Ibogaine undergoes demethylation to form its principal metabolite,
noribogaine, also known as O-desmethylibogaine or 10-hydroxyibogamine. 18-
methoxycoronaridine (18-MC, see Glick et al. in this volume) is an ibogaine
congener that appears to have efcacy similar to ibogaine in animal models of
drug dependence with evidence of less potential toxicity.
3
1. ibogaine: a review
Alkaloid R1R2R3
Ibogaine OCH3HH
Noribogaine OH H H
(+)-18-Methoxycoronaridine H CO2CH3OCH3
Figure 1. Chemical Structures of Ibogaine, Noribogaine, and 18-Methoxycoronaridine.
The ibogamine skeleton above is numbered using the LeMen and Taylor system in which ibogaine is
designated as 10-methoxyibogamine and noribogaine as 10-hydroxyibogamine. Alternatively,
according to the Chemical Abstracts numbering system for the ibogamine skeleton which is
frequently encountered in the biological and medical literature, ibogaine and noribogaine have respec-
tively been referred to as 12-methoxyibogamine and 12-hydroxyibogamine.
C. Historical Time Line
The following timeline outlines the historical events relating to the
development of ibogaine as a treatment for drug dependence. Elsewhere in this
volume, Alper et al. provide a more detailed contemporary history of ibogaine in
the United States and Europe.
1864: The rst description of T. iboga is published. A specimen is brought to
France from Gabon. A published description of the ceremonial use of T. iboga in
Gabon appears in 1885 (14).
1901: Ibogaine is isolated and crystallized from T. iboga root bark (15-17).
1901-1905: The rst pharmacodynamic studies of ibogaine are performed.
During this period ibogaine is recommended as a treatment for asthenia at a
dosage range of 10 to 30 mg per day (14).
1939-1970: Ibogaine is sold in France as Lambarène, a neuromuscular
stimulant, in 8 mg tablets, recommended for indications that include fatigue,
depression, and recovery from infectious disease (14).
1955: Harris Isbell administers doses of ibogaine of up to 300 mg to eight
already detoxied morphine addicts at the U.S. Addiction Research Center in
Lexington, Kentucky (18).
1957: The description of the denitive chemical structure of ibogaine is
published. The total synthesis of ibogaine is reported in 1965 (19-21).
1962-1963: In the United States, Howard Lotsof administers ibogaine to 19
individuals at dosages of 6 to 19 mg/kg, including 7 with opioid dependence who
note an apparent effect on acute withdrawal symptomatology (22,23).
1967-1970: The World Health Assembly classies ibogaine with hallucinogens
and stimulants as a substance likely to cause dependency or endanger human
health. The U.S. Food and Drug Administration (FDA) assigns ibogaine
Schedule I classication. The International Olympic Committee bans ibogaine as
a potential doping agent. Sales of Lambarène cease in France (14).
1969: Dr. Claudio Naranjo, a psychiatrist, receives a French patent for the
psychotherapeutic use of ibogaine at a dosage of 4 to 5 mg/kg (24).
1985: Howard Lotsof receives a U.S. patent for the use of ibogaine in opioid
4kenneth r. alper
withdrawal (22). Additional patents follow for indications of dependence on
cocaine and other stimulants (23), alcohol (25), nicotine (26), and polysubstance
abuse (27).
1988-1994: U.S. and Dutch researchers publish initial ndings suggestive of
the efcacy of ibogaine in animal models of addiction, including diminished
opioid self-administration and withdrawal (28-30), as well as diminished cocaine
self-administration (31).
1989-1993: Treatments are conducted outside of conventional medical settings
in the Netherlands involving the International Coalition of Addict Self-Help
(ICASH), Dutch Addict Self Help (DASH), and NDA International (22,32-35).
1991: Based on case reports and preclinical evidence suggesting possible
efcacy, NIDA Medication Development Division (MDD) begins its ibogaine
project. The major objectives of the ibogaine project are preclinical toxicological
evaluation and development of a human protocol.
August 1993: FDA Advisory Panel meeting, chaired by Medical Review
Ofcer Curtis Wright, is held to formally consider Investigational New Drug
Application led by Dr. Deborah Mash, Professor of Neurology at the University
of Miami School of Medicine. Approval is given for human trials. The approved
ibogaine dosage levels are 1, 2, and 5 mg/kg. The Phase I dose escalation study
begins December 1993, but activity is eventually suspended (36).
October 1993-December 1994: The National Institute on Drug Abuse (NIDA)
holds a total of four Phase I/II protocol development meetings, which include
outside consultants. The resulting draft protocol calls for the single adminis-
tration of xed dosages of ibogaine of 150 and 300 mg versus placebo for the
indication of cocaine dependence (37).
March 1995: The NIDA Ibogaine Review Meeting is held in Rockville,
Maryland, chaired by the MDD Deputy Director, Dr. Frank Vocci. The possibility
of NIDA funding a human trial of the efcacy of ibogaine is considered. Opinions
of representatives of the pharmaceutical industry are mostly critical, and are a
signicant inuence in the decision not to fund the trial. NIDA ends its ibogaine
project, but it does continue to support some preclinical research on iboga
alkaloids.
Mid 1990s-2001: Ibogaine becomes increasingly available in alternative
settings, in view of the lack of approval in the Europe and the United States.
Treatments in settings based on a conventional medical model are conducted in
5
1. ibogaine: a review
Panama in 1994 and 1995 and in St. Kitts from 1996 to the present. Informal
scenes begin in the United States, Slovenia, Britain, the Netherlands, and the
Czech Republic. The Ibogaine Mailing List (38) begins in 1997 and heralds an
increasing utilization of the Internet within the ibogaine medical subculture.
II. Mechanisms of Action
A. Neurotransmitter Activities
1. General Comments
Elsewhere in this volume, Glick et al., Sershen et al., and Skolnick review the
mechanism of action of ibogaine. Popik and Skolnick (39) provide a recent,
detailed review of ibogaines receptor activities. Ibogaine appears to have a novel
mechanism of action that differs from other existing pharmacotherapies of
addiction, and its mechanism of action does not appear to be readily explained on
the basis of existing pharmacologic approaches to addiction. Ibogaines effects
may result from complex interactions between multiple neurotransmitter systems
rather than predominant activity within a single neurotransmitter system (39-42).
Several laboratories have reported on the results of pharmacological screens of
the receptor binding prole of ibogaine (40,43-45). Ibogaine has low micromolar
afnities for multiple binding sites within the central nervous system, including
N-methyl-D-aspartate (NMDA), kappa- and mu-opioid and sigma2receptors,
sodium channels, and the serotonin transporter. Although not apparent in binding
studies, functional studies indicate signicant activity of ibogaine as a noncom-
petitive antagonist at the nicotinic acetylcholine receptor (46-50).
Although in vitro activities in the micromolar range are often described as
ancillary in attempting to characterize a drugs in vivo mechanism of action,
micromolar activity may be pharmacologically important with regard to ibogaine
or noribogaine due to the relatively high concentrations reached in the brain
(40,44,51). Hough et al. (51) noted a brain level of ibogaine of 10 µM in female
rats at 1 hour after the administration of 40 mg/kg ibogaine i.p., which is the usual
dosage, animal, gender and route of administration used in that laboratory to
investigate ibogaines effects on drug self-administration and withdrawal. Brain
levels of ibogaine, and its major metabolite noribogaine, ranged from 1 to 17 µM
between 15 minutes and 2 hours in male rats following the oral administration
ibogaine at a dose of 50 mg/kg (44).
2. Glutamate
Elsewhere in this volume, Skolnick reviews the possible relevance of
6kenneth r. alper
ibogaines activity as a glutamate antagonist to its putative effects in drug
dependence. There is evidence that suggests that antagonists of the N-methyl-D-
aspartate (NMDA) subtype of glutamate receptor are a potentially promising
class of agents for the development of medications for addiction (52-54).
Ibogaines apparent activity as a noncompetitive NMDA antagonist has been
suggested to be a possible mechanism mediating its putative effects on drug
dependence (39,41,55-58).
Ibogaine competitively inhibits the binding of the NMDA antagonist MK801
to the NMDA receptor complex, with reported afnities in the range of 0.02 to
9.8 µM (40,45,55-57,59,60). Functional evidence supporting an antagonist action
of ibogaine at the NMDA receptor includes observations of reduced glutamate-
induced cell death in neuronal cultures, reduction of NMDA-activated currents in
hippocampal cultures (55,58), prevention of NMDA-mediated depolarization in
frog motoneurons (59), and protection against NMDA-induced convulsions (61).
Glycine, which acts as an NMDA co-agonist by binding at the NMDA receptor,
attenuates ibogaines effect of blocking naloxone-precipitated jumping (58).
MK801 and ibogaine do not produce identical effects, as evidenced by the
observation that in the rat brain ibogaine lowered the concentration of dopamine
while increasing the level of its metabolites, whereas MK801 did not have these
effects (62,63).
3. Opioids
It has been suggested that ibogaines or noribogaines activity as a putative
agonist at mu-opioid receptors might explain ibogaines apparent efcacy in
opioid withdrawal (36,64,65). Ibogaine binds to mu-opioid receptors with
reported binding afnities in the range of 0.13 to 26 µM (40,45,64,66), with one
study reporting a result in excess of 100 µM (43). Ibogaine behaves as an agonist
in a functional assay for mu-opioid receptors, the binding of [35S]-GTPγS (65).
However, some observations are difcult to reconcile with a mu-agonist action of
ibogaine. Ibogaine did not behave as a mu-opioid agonist in assays with isolated
smooth muscle preparations (67). Unlike mu-opioid agonists, ibogaine (68-70)
and noribogaine (71) do not appear by themselves to have antinociceptive effects.
Some ndings suggest the intriguing possibility that ibogaine may act at the
level of second messenger signal transduction to enhance the functional activity
of mu-opioid receptors independently of any direct agonist interaction at opioid
receptors. Both ibogaine and noribogaine reportedly potentiated morphine-
induced inhibition of adenylate cyclase in vitro with opioid receptors already
occupied by the maximally effective concentration of morphine, but did not affect
adenylate cyclase in the absence of morphine (72). A similar interpretation might
also explain the nding that ibogaine inhibited the development of tolerance to
the antinociceptive effect of morphine in mice, without by itself affecting
nociception (73).
7
1. ibogaine: a review
Ibogaine binds to kappa-opioid receptors with reported binding afnities in the
range of 2.2 to 30 µM (43,45,56,66). Evidence consistent with a kappa-opioid
action of ibogaine includes the observation that the kappa-opioid antagonist,
norbinaltorphimine antagonized some of the effects of ibogaine in morphine-
treated rats (74,75). Kappa-opioid agonists reportedly can imitate certain effects
of ibogaine, such as reduced cocaine and morphine self-administration (76), and
reduction in locomotor activation to morphine accentuated by prior morphine
exposure (77). Sershen, on the other hand, attributes a kappa-opioid antagonist
action to ibogaine, based on the observation that stimulated dopamine efux from
mouse brain slices was decreased by a kappa opioid agonist, and the decrease was
offset by the addition of ibogaine (78). However, ibogaines interactions with
multiple neurotransmitter systems raises the possibility that the nding could be
accounted for by mechanisms that do not involve the kappa-opioid receptor, as
dopamine efux is modulated by multiple neurotransmitters.
4. Serotonin
Ibogaine and serotonin both contain an indole ring in their structure, and
ibogaine has been shown to bind to the serotonin transporter and to increase
serotonin levels in the nucleus accumbens (NAc) (41,79,80). The demonstration
that ibogaine blocks serotonin uptake (81) suggests that the effect of ibogaine on
extracellular serotonin levels may be mediated by uptake inhibition, in addition
to release (80). The reported afnity of ibogaine for the serotonin transporter
ranges from 0.55 to 10 µM (39,44,45,79,81), and the afnity of noribogaine for
the serotonin transporter is approximately 10-fold stronger (45,79). The
magnitude of the effect of ibogaine on serotonin release is reportedly large and is
comparable to that of the serotonin releasing agent fenfluramine, with
noribogaine having a lesser effect, and 18-MC no effect (80). Some authors
suggest a role for modulatory inuence of serotonin in ibogaines effects on
dampening dopamine efux in the NAc (41,80).
Ibogaines hallucinogenic effect has been suggested to involve altered
serotonergic neurotransmission (42,80). Ibogaine is reported in some studies to
bind the 5-HT2A receptor, which is thought to mediate the effects of classical
indolealkylamine and phenethylamine hallucinogens (82), with three studies
reporting afnities in the range of 4.1 to 12 µM (40,45,83), one reporting a value
of 92.5 µM (84), and with two other studies reporting no signicant afnity
(43,44). Drug discrimination studies provide some functional evidence for the
action of ibogaine as an agonist at the 5-HT2A receptor, which is apparently a
signicant, although nonessential, determinant of the ibogaine stimulus (84) (see
Section II.B, Discrimination Studies). Ibogaine binds to the 5-HT3receptor
with reported afnities of 2.6 and 3.9 µM (40,45), and it was without signicant
afnity in two other studies (43,83). The 5-HT3receptor is apparently not
involved in the ibogaine discriminative stimulus (85).
8kenneth r. alper
5. Dopamine
Ibogaine does not appear to signicantly affect radioligand binding to D1, D2,
D3, or D4receptors (40,43,44) and is a competitive blocker of dopamine uptake
at the dopamine transporter with afnities in the range of 1.5 to 20 µM (81).
Where afnities for the serotonin and dopamine transporter have been estimated
within the same study, the reported afnity of ibogaine for the serotonin
transporter has generally been 10 to 50 times stronger than its afnity for the
dopamine transporter (44,79,81). Ibogaine does not apparently affect norepi-
nephrine reuptake (44,45).
French et al. (86) studied the electrophysiological activity of dopamine
neurons in the ventral tegmental area (VTA) of rats given up to 7.5 mg/kg
ibogaine intravenously and found a signicant increase in ring rate. Ibogaine
given intraperitoneally (i.p.) at a dose of 40 mg/kg did not affect the spontaneous
ring of VTA dopamine neurons or the response of VTA dopamine neurons to
cocaine or morphine. Ibogaine reportedly lowers the concentration of dopamine,
while increasing the level of its metabolites, indicating diminished release of
dopamine in the brain of the rat (62,63) and the mouse (87). Decreased release of
dopamine could possibly explain the observation of increased prolactin release
following ibogaine administration (62,63,88). Staley et al. (44) have suggested
that ibogaine might act at the dopamine transporter to inhibit the translocation of
dopamine into synaptic vesicles, thereby redistributing dopamine from vesicular
to cytoplasmic pools. As a result, the metabolism of dopamine by monoamine
oxidase could explain the observation of decreased tissue dopamine content with
increased levels of its metabolites.
The effects of ibogaine on dopamine efux in response to the administration of
drugs of abuse are described in Section III.E, Dopamine Efux.
6. Acetylcholine
Ibogaine is a nonselective and weak inhibitor of binding to muscarinic receptor
subtypes. Reported afnities are 7.6 and 16 µM and 5.9 and 31 µM, respectively,
for the M1and M2muscarinic receptor subtypes (40,45), with another study
reporting no signicant afnity of ibogaine for muscarinic receptors (43).
Functional evidence consistent with a muscarinic cholinergic agonist effect of
ibogaine includes the observations of the elimination of ibogaine-induced EEG
dyssynchrony by atropine in cats (89), decreased heart rate following ibogaine
administration in rats (90), and the attribution of the effect of cholinesterase
inhibition to ibogaine in the older literature (1,91). The afnity of noribogaine for
muscarinic receptors is apparently similar to that of ibogaine (44,45).
Several laboratories have reported that ibogaine produces noncompetitive
functional inhibition of the nicotinic acetylcholine receptor, apparently involving
open channel blockade (46,48-50). As with a number of other channel blockers,
binding studies involving channels associated with nicotinic receptors have been
9
1. ibogaine: a review
limited by the lack of appropriate ligands, and investigations of the afnity of
ibogaine for the nicotinic acetylcholine receptor have mainly involved functional
assays. Utilizing 86Rb+efux assays, Fryer and Lukas (50) found that ibogaine
inhibited human ganglionic and muscle-type nicotinic acetylcholine receptors
with IC50 values of 1.06 and 22.3 µM, respectively. Badio et al. (48) found that
ibogaine inhibited 22Na+inux through rat ganglionic and human muscle-type
nicotinic acetylcholine receptors with IC50 values of 0.020 µM and 2.0 µM,
respectively. Noribogaine was 75-fold less active than ibogaine in the rat
ganglionic cell assay. In mice, ibogaine at a dose of 10 mg/kg completely blocked
the central antinociceptive nicotinic receptor-mediated response to epibatidine.
Ibogaine has been associated with decreased acetylcholine-stimulated nicotinic
receptor mediated catecholamine release in cultured cells (49) and decreased
dopamine release evoked by nicotine in the NAc of the rat (46,92).
7. Sigma Receptors
Elsewhere in this volume, Bowen discusses ibogaines action at the sigma
receptor. The afnity of ibogaine for the sigma2receptor is strong relative to other
known CNS receptors, and the reported range is 0.09 to 1.8 µM (45,60,93,94).
The afnity of ibogaine for the sigma1receptor is reportedly on the order of 2 to
100 times weaker than its afnity for the sigma2receptor (45,60,93,94). The
neurotoxic effects of ibogaine may involve activity at the sigma2receptor, which
reportedly potentiates the neuronal response to NMDA (95).
8. Sodium Channels
The reported afnity of ibogaine for sodium channels ranges from 3.6 to 9 µM
(40,43). There is apparently no experimental evidence regarding the functional
signicance of ibogaines action at sodium channels.
B. Discrimination Studies
Elsewhere in this volume, Helsley et al. discuss the topic of ibogaine and drug
discrimination. Drug discrimination studies offer a possible approach to the issue
of ibogaines mechanism of action and may help resolve the distinction between
ibogaines therapeutic and hallucinogenic effects. The 5-HT2A receptor appears to
be a signicant, but nonessential, determinant of the ibogaine stimulus (84,96).
The ibogaine stimulus is reportedly generalized to the indolealkylamine
hallucinogen D-lysergic acid diethylamide (LSD) and the phenethylamine
hallucinogen 2,5-dimethoxy-4-ethylamphetamine (DOM), and this general-
ization is abolished by the addition of a 5-HT2A receptor antagonist (96). The
addition of a 5-HT2A receptor antagonist did not attenuate stimulus control of
ibogaine itself in the ibogaine-trained animals, indicating that the 5-HT2A is not
essential to the ibogaine discriminative stimulus. The 5-HT2C receptor, which
10 kenneth r. alper
plays a modulatory role in hallucinogenesis, is also involved, but is not essential
to the ibogaine stimulus, and the 5-HT1A and 5-HT3receptors are apparently not
involved in the ibogaine stimulus (85). The ibogaine discriminative stimulus
reportedly is potentiated by the serotonin reuptake inhibitor uoxetine (85), and
has an insignicant degree of generalization to the serotonin releaser D-fenu-
ramine (97).
Ibogaine showed a lack of substitution for phencyclidine (98,99), and
substituted for MK 801 only at high (100 mg/kg) doses in mice (58,61), but not
at lower (10 mg/kg) doses in rats (99,100), suggesting that the NMDA receptor is
not a signicant determinant of the ibogaine stimulus. Sigma2, and mu- and
kappa-opioid activity may be involved in the ibogaine discriminative stimulus
(99). A high degree of stimulus generalization is reported between ibogaine and
some of the Harmala alkaloids, a group of hallucinogenic beta-carbolines that are
structurally related to ibogaine (101,102). While the discriminative stimulus for
both the Harmala alkaloids and ibogaine apparently involves the 5-HT2receptor
(84,85,103), it does not appear essential to generalization between ibogaine and
harmaline, as generalization to the harmaline stimulus was unaffected by the
addition of a 5-HT2antagonist in ibogaine-trained animals (84). Ibogaine-trained
rats generalize to noribogaine (100,104), which in one study was more potent
than ibogaine itself in eliciting ibogaine-appropriate responses (100).
C. Effects on Neuropeptides
Both ibogaine and cocaine given in multiple administrations over 4 days to
rats reportedly increase neurotensin-like immunoreactivity (NTLI) in the
striatum, substantia nigra, and NAc (105). However, unlike cocaine, which
increased NTLI in the frontal cortex, ibogaine had no effect on frontal cortical
NTLI. Ibogaine pretreatment prevented the increase of NTLI in striatum and
substantia nigra induced by a single dose of cocaine. Substance P, like NTLI,
was increased in the striatum and substantia nigra after either cocaine or
ibogaine, with an increase in frontal cortex with cocaine and no effect with
ibogaine (106). Ibogaineinduced increases in NTLI or substance P were
blocked by administration of a D1antagonist.
Unlike the NTLI or substance P responses, ibogaine alone had no effect on
dynorphin. However, ibogaine pretreatment dramatically enhanced cocaine-
induced increases in dynorphin, a kappa-opioid agonist (107). The authors
suggested that the increase in dynorphin related to cocaines interaction with
ibogaine could result in enhanced kappa-opioid activity. Kappa-opioid agonists
reportedly decrease cocaine intake in animal models (108,109).
11
1. ibogaine: a review
D. Possible Effects on Neuroadaptations Related to
Drug Sensitization or Tolerance
There is some evidence to suggest that ibogaine treatment might result in the
resetting or normalization of neuroadaptations related to drug sensitization or
tolerance (110 ). Ibogaine pretreatment blocked the expression of sensitization-
induced increases in the release of dopamine in the NAc shell in response to
cocaine in cocaine-sensitized rats (111). The effect of ibogaine on diminished
locomotor activity and dopamine efux in the NAc in response to morphine is
more evident in animals with prior exposure to morphine (112,113), which is
consistent with a relatively selective effect of ibogaine on neuroadaptations
acquired from drug exposure. Similarly, the observation that ibogaine inhibited
the development of tolerance in morphine-tolerant mice, but had no effect on
morphine nociception in morphine-naïve mice (114 ), suggests a selective effect
on acquired neuroadaptations related to repeated morphine exposure.
Ibogaine appears to have persistent effects not accounted for by a metabolite
with a long biological half-life (29,115). Ibogaines action could possibly involve
the opposition or reversal of persistent neuroadaptive changes thought to be
associated with drug tolerance or sensitization. Such an action could be related to
persistent effects on second messengers (72,116). For example, sensitization to
both opiates and cocaine is thought to involve enhanced stimulation of cyclic
AMP (117 ). Ibogaine has been reported to potentiate the inhibition of adenylyl
cyclase by serotonin (72), an effect that would be expected to oppose the
enhanced transduction of cyclic AMP that is reportedly associated with stimulant
sensitization (117 ).
III. Evidence of Efcacy in Animal Models
A. Drug Self-Administration
Evidence for ibogaines effectiveness in animal models of addiction includes
observations of reductions in self-administration of morphine or heroin
(29,31,118-120), cocaine (29,31,119,121), and alcohol (122), and reduced
nicotine preference (75). According to some reports, effects of ibogaine on drug
self-administration are apparently persistent. Sershen et al. (121) administered
ibogaine i.p. to mice as two 40 mg/kg dosages 6 hours apart, and found a
diminution of cocaine preference that was still evident after 5 days. Glick et al.
(29,119) noted reductions in cocaine and morphine self-administration that
persisted for at least 2 days and were dose dependent in the range of 2.5 to 80
mg/kg. ibogaine given i.p. The persistence of an effect beyond the rst day
12 kenneth r. alper
suggests a specic action of ibogaine on drug intake, as water intake was also
suppressed initially by ibogaine on the rst, but not the second day. Cappendijk
and Dzoljic (31) found reductions in cocaine self-administration that persisted for
more than 48 hours in rats treated with ibogaine at a dose of 40 mg/kg i.p., given
as a single administration, or repeatedly on 3 consecutive days or three
consecutive weeks.
In the studies by Glick et al. there was variation between results in individual
rats with some showing persistent decreases in morphine or cocaine intake for
several days or weeks after a single injection and others only after two or three
weekly injections. The authors noted evidence of a continuous range of individual
sensitivity to ibogaine among the experimental animals and that it appeared as if
adjustments of the dosage regimen could produce long-term reductions in drug
intake in most animals (29). Similarly, Cappendijk and Dzoljic (31) found the
largest effects on cocaine self-administration occurred when ibogaine was given
weekly for three consecutive weeks. This result suggests the possibility that the
optimal schedule of ibogaine administration to limit cocaine intake may involve
modication of the single dose regimen which has been used for opioid detoxi-
cation (32,123).
Dworkin et al. (11 8) found that pretreatment with ibogaine at a dose of 80
mg/kg i.p. diminished the response for heroin and cocaine, and also for food,
suggesting a nonspecic confound. A 40 mg/kg intraperitoneal dose of ibogaine
sharply reduced heroin self-administration in the absence of a signicant effect
on food response, although the effect did not persist beyond 24 hours (118 ).
Dworkin et al. cited methodologic factors relating to differences in gender, strain,
and reinforcement schedule to explain the apparent discrepancy between their
results and other studies that reported persistent effects (29,31,119,121).
Noribogaine has also been reported to reduce cocaine and morphine self-
administration (124). The effect of noribogaine on drug self-administration
persisted for 2 days, after the response for water, which was initially suppressed
on the rst day, had returned to baseline. Other iboga alkaloids have also been
reported to reduce morphine and cocaine self-administration in rats for a period
of a day or longer following a single i.p dose (119 ). Some of the iboga alkaloids
tested in this study produced tremors, which typically occurred for a period of 2
to 3 hours, and were independent of persistent effects of drug self-administration.
An ibogaine congener, 18-methoxycoronaridine (18-MC) (45), reportedly
reduces in rats the self-administration of cocaine (120), morphine and alcohol
(125), and nicotine preference (75) without any apparent reduction in the
response for water.
B. Acute Opioid Withdrawal
Dzoljic et al. (28) administered ibogaine in a dose range of 4 to 16 µg intra-
13
1. ibogaine: a review
cerebroventricularly to rats and observed a dose-dependent attenuation of
naloxone-precipitated withdrawal signs. This same group also found an
attenuation of morphine withdrawal signs in rats with 40 mg/kg ibogaine
administered i.p., and also norharman, an endogenously occurring hallucinogenic
beta-carboline and a structural relative of ibogaine (126). Glick et al. have
reported dose-dependent reduction of the signs of naltrexone-precipitated
morphine withdrawal in rats administered ibogaine at doses of 20, 40, or 80
mg/kg i.p (127) or 18-MC (128) at doses of 20 and 40 mg/kg i.p. Attenuation of
withdrawal signs was reported in morphine-dependent monkeys given 2 or 8
mg/kg ibogaine subcutaneously (129). In their chapter in this volume, Parker and
Siegel report that 40 mg/kg ibogaine administered i.p attenuated naloxone-
precipitated morphine withdrawal in rats, as well as withdrawal-induced place
aversion.
Sharpe and Jaffe (130) reported that ibogaine in dosages ranging between 5
and 40 mg/kg administered subcutaneously failed to attenuate naloxone-precip-
itated withdrawal in rats, although they did nd that one sign (grooming) was
reduced, and noted the possible effect of methodological issues such as morphine
exposure and withdrawal procedures, or the route of administration of ibogaine.
Popik et al. (58) and Layer et al. (56) found that ibogaine at doses ranging from
40 to 80 mg/kg i.p. reduced naloxone-precipitated jumping in morphine
dependent mice, although Francés et al. (69) found the opposite effect of 30
mg/kg ibogaine administered i.p. in mice. As pointed out by Popik and Skolnik
(39), the divergent results in morphine dependent mice might relate to ibogaine
having been given prior to the administration of naloxone in the studies by Popik
et al. (58) and Layer et al. (56), whereas ibogaine was administered after
naloxone in the study by Francés et al.
C. Conditioned Place Preference
Parker and Siegel review ibogaine and place preference in this volume.
Ibogaine is reported to prevent the acquisition of place preference when given 24
hours before amphetamine (131) or morphine (132). The effect of ibogaine on
blocking the acquisition of place preference was diminished across multiple
conditioning trials. Ibogaine given after morphine did not apparently attenuate
the expression of previously established morphine place preference (133).
D. Locomotor Activity
Pretreatment with ibogaine and its principal metabolite, noribogaine reportedly
diminishes locomotor activation in response to morphine (74,112,113,124,134-
136). The effect of ibogaine in reducing locomotor activity in response to
morphine is reportedly greater in female than in male rats, probably reecting the
14 kenneth r. alper
relatively greater bioavailability of ibogaine in females (135). The literature on
cocaine appears to be less consistent, with some reports of decreased locomotor
activation (87,137-139), and others reporting increases (127,137,140,141). This
apparent disparity may be related in part to the species of experimental animal
that was used, as Sershen et al. (137) report increased locomotor activity in
response to cocaine in the rat, with the opposite result in the mouse.
Stereotypy is a methodologic issue that might explain some of the disparate
results regarding ibogaines interaction with the locomotor response to cocaine.
Higher doses of stimulants can produce strereotypy, which could decrease the
amount of measured locomotion relative to an animal that is experiencing less
locomotor stimulation at a lower stimulant dose. Thus, the potentiation by
ibogaine of locomotor activity related to cocaine administration can result in less
measured movement in animals experiencing locomotor stimulation to the point
of stereotypy (110 ). Ibogaine pretreatment reportedly potentiates stereotypy in
rats receiving cocaine or methamphetamine (111,142).
E. Dopamine Efflux
Reductions in dopamine efux in the NAc in response to morphine have been
reported in animals pretreated with ibogaine (113,115,134), noribogaine (124), or
18-MC (120,143). Similarly, reductions in dopamine efux in the NAc in
response to nicotine have been reported in animals pretreated with ibogaine
(46,92) and 18-MC (42).
As with locomotor stimulation, methodological issues may have played a part
in apparently divergent results regarding ibogaines effect on dopamine efux in
the NAc in response to cocaine or amphetamine, which is reportedly increased as
measured by microdialysis (134), although the opposite result was observed in a
study on cocaine using microvoltammetry (139). Dosage is an additional consid-
eration that might inuence ibogaines effect on dopamine efux in the NAc in
response to cocaine, with a larger ibogaine dose reportedly producing an increase
and a smaller dose producing a decrease (144).
Dopamine efux in response to cocaine may also depend on whether dopamine
measurements are made in the NAc core versus shell. Szumlinski et al. (111)
found that ibogaine pretreatment (given 19 hours earlier) abolished the sensitized
dopamine efux in response to cocaine in the NAc shell in rats that had been
sensitized by repeated prior exposure to cocaine. The same ibogaine pretreatment
had no apparent effect on dopamine efux in the NAc shell in response to acute
(administered without prior cocaine exposure) cocaine. The authors noted a prior
study in their laboratory that found a potentiation by ibogaine pretreatment of
dopamine efux in response to acute cocaine in which the position of the
recording probe spanned both the core and shell regions of the NAc (134). These
results indicate the possibility of a differential effect of ibogaine on dopamine
15
1. ibogaine: a review
efux in response to cocaine between the NAc shell, which is thought to play a
relatively greater role in the motivational aspects of drugs of abuse, and the NAc
core, which, in turn, is thought to play a relatively greater role in motor behavior
(145). The authors suggested that the effect of ibogaine on reduced cocaine self-
administration may be mediated by the observed reduction in dopamine efux in
response to cocaine in the NAc shell in cocaine-sensitized animals (111). On the
other hand, the enhancement by ibogaine preatreatment of locomotor activity
seen in response to acute or chronic cocaine administration may be mediated by
increased dopamine efux in the NAc core. The observed increase in dopamine
efux with ibogaine pretreatment in the NAc core in response to acute cocaine
(134) is consistent with such a formulation, although this group has yet to report
on the effect in cocaine-sensitized animals.
Ibogaine and 18-MC reportedly decrease dopamine release evoked by nicotine
in the NAc of the rat (46,92). In the study by Benwell et al. (46), the decreased
NAc dopamine release following ibogaine was independent of any change in
locomotor activity, which was viewed as notable given the usual association
between NAc dopamine efux and locomotor activity in response to nicotine.
The authors cited previous work in which a similar dissociation between NAc
dopamine efux and locomotor activity in response to nicotine was produced by
treatment with NMDA antagonists, and they suggested that their ndings might
be related to ibogaines NMDA antagonist activity.
IV. Evidence of Efcacy and Subjective Effects in Humans
A. Evidence Of Efcacy
1. Acute Opioid Withdrawal
One line of clinical evidence suggesting ibogaines possible efcacy are the
accounts of the addicts themselves, whose demand has led to the existence of an
informal treatment network in Europe and the United States. Opioid
dependence is the most common indication for which addicts have sought
ibogaine treatment, which has been typically administered as a single dose.
Common reported features of case reports describing ibogaine treatment
(35,36,146-149) are reductions in drug craving and opiate withdrawal signs and
symptoms within 1 to 2 hours, and sustained, complete resolution of the opioid
withdrawal syndrome after the ingestion of ibogaine. These case studies appear
consistent with general descriptions of ibogaine treatment (33,34,150).
Alper et al. (32) summarized 33 cases treated for the indication of opioid
detoxication in nonmedical settings under open label conditions. These cases
16 kenneth r. alper
are a subset of those presented at the NIDA Ibogaine Review Meeting held in
March, 1995 (151). A focus on acute opioid withdrawal may offset some of the
methodological limitations of the informal treatment context because the acute
opioid withdrawal syndrome is a clinically robust phenomenon that occurs within
a relatively limited time frame and yields reasonably clear outcome measures.
Despite the unconventional setting and the lack of structured clinical rating
instruments, the lay treatment guides who reported on the case series might
reasonably be expected to be able to assess the presence or absence of the
relatively clinically obvious and unambiguous features of opioid withdrawal.
The subjects in this series of cases reported an average daily use of heroin of
0.64 ± 0.50 g, primarily by the intravenous route, and received an average dose
of ibogaine of 19.3 ± 6.9 mg/kg (range of 6 to 29 mg/kg). Resolution of the signs
of opioid withdrawal without further drug seeking behavior was observed in 25
patients. Other outcomes included drug seeking behavior without withdrawal
signs (four patients), drug abstinence with attenuated withdrawal signs (two
patients), drug seeking behavior with continued withdrawal signs (one patient),
and one fatality, possibly involving surreptitious heroin use (see Section VI,
Safety). The reported effectiveness of ibogaine in this series suggests the need
for a systematic investigation in a conventional clinical research setting.
In their chapter in this volume, Mash et al. report having treated more than 150
subjects for substance dependence in a clinic located in St. Kitts, West Indies. A
subset of 32 of these subjects was treated with a xed dose of ibogaine of 800 mg
for the indication of opioid withdrawal. Physician ratings utilizing structured
instruments for signs and symptoms of opioid withdrawal indicated resolution of
withdrawal signs and symptoms at time points corresponding to 12 hours
following ibogaine administration and 24 hours after the last use of opiates, and
at 24 hours following ibogaine administration and 36 hours after the last use of
opiates. The resolution of withdrawal signs and symptoms was sustained during
subsequent observations over an interval of approximately one week following
ibogaine administration. Reductions of measures of depression and craving
remained signicantly reduced one month after treatment (123). The authors
noted that ibogaine appeared to be equally efcacious in achieving detoxication
from either methadone or heroin. The reported efcacy of ibogaine for the opioid
withdrawal syndrome observed in the St. Kitts facility appears to conrm the
earlier impressions of the case study literature (32-36,146-150).
2. Long-Term Outcomes
There is very little data regarding the long-term outcomes in patients treated
with ibogaine. Lotsof (151) presented a summary of 41 individuals treated
between 1962 and 1993 at the NIDA Ibogaine Review Meeting held in March
1995. The data consisted of self-reports obtained retrospectively, which are
essentially anecdotal, but apparently represent the only formal presentation of a
17
1. ibogaine: a review
systematic attempt to determine long-term outcomes in patients treated with
ibogaine. Thirty-eight of the 41 individuals presented in the summary reported
some opioid use, with approximately 10 of these apparently additionally
dependent on other drugs, mainly cocaine, alcohol, or sedative-hypnotics. The
use of tobacco or cannabis was not apparently assessed. Across the sample of 41
individuals, nine individuals were treated twice and one was treated three times
for a total of 52 treatments. The interval of time following treatment was recorded
for which patients reported cessation of use of the drug or drugs on which they
were dependent. Fifteen (29%) of the treatments were reportedly followed by
cessation drug use for less than 2 months, 15 (29%) for at least 2 months and less
than 6 months, 7 (13%) for at least 6 months and less than one year, 10 (19%) for
a period of greater than one year, and in 5 (10%) outcomes could not be
determined.
B. Subjective Effects
There appear to be common elements to experiences generally described by
patients treated with ibogaine. The stages of the subjective ibogaine experience
presented below are a composite derived by the author from interviews with
patients and treatment guides, and general descriptions and case studies provided
by the literature (33-35,146,150). Ibogaine has been typically given in a non-
hospital setting as a single dose in the morning. Vomiting is reportedly common
and usually occurred relatively suddenly as a single episode in the rst several
hours of treatment. Patients generally lie still in a quiet darkened room
throughout their treatment, a practice that is possibly related to the cerebellar
effects of ibogaine, and because vomiting tends to be more frequent with
movement. Patients later in treatment often experience muscle soreness, possibly
due to reduced motor activity earlier in treatment, that resolves with motion,
stretching, or massage.
1. Acute
The onset of this phase is within 1 to 3 hours of ingestion, with a duration on
the order of 4 to 8 hours. The predominant reported experiences appear to involve
a panoramic readout of long-term memory (152), particularly in the visual
modality, and visions or waking dream states featuring archetypal
experiences such as contact with transcendent beings, passage along a lengthy
path, or oating. Descriptions of this state appear more consistent with the
experience of dreams than of hallucinations. Informants appear to emphasize the
experience of being placed in, entering, and exiting entire visual landscapes,
rather than the intrusion of visual or auditory hallucinations on an otherwise
continuous waking experience of reality. Ibogaine-related visual experiences are
reported to be strongly associated with eye closure and suppressed by eye
18 kenneth r. alper
opening. The term oneiric (Greek, oneiros, dream) has been preferred to the
term hallucinogenic in describing the subjective experience of the acute state.
Not all subjects experience visual phenomena from ibogaine, which may be
related to dose, bioavailability, and interindividual variation.
2. Evaluative
The onset of this phase is approximately 4 to 8 hours after ingestion, with a
duration on the order of 8 to 20 hours. The volume of material recalled slows. The
emotional tone of this phase is generally described as neutral and reective.
Attention is still focused on inner subjective experience rather than the external
environment, and it is directed at evaluating the experiences of the acute phase.
Patients in this and the acute phase above are apparently easily distracted and
annoyed by ambient environmental stimuli and prefer as little environmental
sensory stimulation as possible in order to maintain an attentional focus on inner
experience.
3. Residual Stimulation
The onset of this phase is approximately 12 to 24 hours after ingestion, with a
duration in the range of 24 to 72 hours or longer. There is a reported return of
normal allocation of attention to the external environment. The intensity of the
subjective psychoactive experience lessens, with mild residual subjective arousal
or vigilance. Some patients report reduced need for sleep for several days to
weeks following treatment. It is not clear to what extent such reports might reect
a persistent effect of ibogaine on sleep or a dyssomnia due to another cause.
V. Pharmacokinetics
A. Absorption
Jeffcoat et al. (153) administered single oral doses of ibogaine of 5 mg/kg and
50 mg/kg to rats, and estimated oral bioavailabilities of 16 and 71% at the two
dosages, respectively, in females, and 7 and 43% in males. The dose-dependent
bioavailability was interpreted as suggesting that ibogaine absorption, and/or rst
pass elimination, is nonlinear, and the greater bioavailability in females was
viewed as consistent with gender-related differences in absorption kinetics. Pearl
et al. (135) administered ibogaine at a dose of 40 mg/kg i.p. and found whole
brain levels at 1, 5, and 19 hours post-administration of 10, 1, and 0.7 µM in
female rats, and 6, 0.9, and 0.2 µM in male rats, respectively. In the same study,
brain levels of noribogaine at 1, 5, and 19 hours post-administration were 20, 10,
19
1. ibogaine: a review
and 0.8 µM in female rats, and 13, 7, and 0.1 µM and male rats respectively. In
addition to gender differences in bioavailability, the data also provide evidence
for the pharmacologic relevance of micromolar activities of ibogaine and
noribogaine measured in vitro (40,44).
Upton (154) reported on observations in rats given ibogaine in the form of oral
suspension, oral solution, or via IV or intraperitoneal routes, and also reviewed
data obtained in beagle dogs, cynomologous monkeys, and human subjects.
Absorption of the oral suspension in rats was noted to be variable and incomplete.
As in the study cited above by Jeffcoat (153), peak levels and bioavailability were
greater in female than in male rats.
B. Distribution
Hough et al. (51) administered 40 mg/kg ibogaine by the intraperitoneal and
subcutaneous routes and evaluated its distribution in plasma, brain, kidney, liver,
and fat at 1 and 12 hours post-administration. Ibogaine levels were higher
following subcutaneous versus intraperitoneal administration, suggesting a
substantial “first pass effect involving hepatic extraction. The results were
consistent with the highly lipophilic nature of ibogaine; ibogaine concentrations
at 1 hour postadministration were 100 times greater in fat, and 30 times greater
in brain, than in plasma. These authors suggested that the prolonged actions of
ibogaine could relate to adipose tissue serving as a reservoir with release and
metabolism to noribogaine over an extended period of time (51). The apparently
greater levels of ibogaine in whole blood versus plasma suggests the possibility
that platelets might constitute a depot in which ibogaine is sequestered (42). If
there is conversion of ibogaine to noribogaine in the brain, then the signicantly
greater polarity of noribogaine relative to ibogaine could prolong the presence of
the more polar metabolite in the CNS after conversion from ibogaine (42).
C. Metabolism
The major metabolite of ibogaine, noribogaine, is formed through demethy-
lation, apparently via the cytochrome P-450 2D6 (CYP2D6) isoform (155).
Consistent with rst pass metabolism of the parent drug, noribogaine is
reportedly detectable in brain tissue within 15 minutes after oral administration
of 50 mg/kg ibogaine (44). Noribogaine is itself pharmacologically active and is
discussed in this volume by Baumann et al.
In pooled human liver microsomes, Pablo et al. identied two kinetically
distinguishable ibogaine O-demethylase activities which corresponded, respec-
tively, to high and low values of the apparent Michaelis constant (Kmapp) (155).
The low Kmapp ibogaine O-demethylase activity was attributable to CYP2D6 and
accounted for greater than 95% of the total intrinsic clearance in pooled human
20 kenneth r. alper
liver microsomes. The authors noted that the apparent involvement of the
CYP2D6 suggests possible human pharmacogenetic differences in the
metabolism of ibogaine. Poor metabolizers who lack a copy of the CYP2D6
gene (156) would be expected to have relatively less CYP2D6-catalyzed activity
to metabolize ibogaine to noribogaine. Consistent with such an expectation, a
subject identied as a phenotypic CYP2D6 poor metabolizer possessed only the
high Kmapp ibogaine O-demethylase activity, which had accounted for only a
small fraction of the intrinsic clearance. In another study, analysis of ibogaine and
noribogaine levels in human subjects yielded a distribution interpreted as
indicating three groups of rapid, intermediate, and poor metabolizers (157), a
pattern consistent with the observed pharmacogenetic polymorphism of CYP2D6
in human populations (156).
D. Excretion
Ibogaine has an estimated half-life on the order of 1 hour in rodents (158), and
7.5 hours in man (Mash et al., this volume). Ibogaine and its principal metabolite,
noribogaine, are excreted via the renal and gastrointestinal tracts. In rats, Jeffcoat
et al. (153) noted 60 to 70% elimination in urine and feces within 24 hours, and
Hough et al. (51) found plasma and tissue levels to be 10 to 20-fold lower at 12
hours versus 1 hour post dose.
Upton and colleagues (154) cited several pharmacokinetic issues of potential
concern based on their analysis of data obtained from rats. These include
evidence for presystemic clearance potentially resulting in low bioavailability
and interpatient variability, and saturable rst pass clearance, which could also
generate intrapatient variability. The possibility of saturable systemic clearance
was also noted. Mash et al. (36) suggested the possibility of species or strain
differences in ibogaine metabolism and clearance rates and cited the rapid
elimination of ibogaine from the blood of primates, as opposed to rats or humans,
as an example.
In human subjects, 90% of a 20 mg/kg dose of ibogaine was reportedly
eliminated within 24 hours (36). Noribogaine is apparently eliminated signi-
cantly more slowly than ibogaine, and observations in human subjects indicate
persistently high levels of noribogaine at 24 hours (36,79,123, Mash et al. in this
volume). The sequestration and slow release from tissues of ibogaine or
noribogaine and the slow elimination of noribogaine have been suggested to
account for the apparently persistent effects of ibogaine.
21
1. ibogaine: a review
VI. Safety
A. Neurotoxicity
1. Neuropathology
Multiple laboratories have reported on the degeneration of cerebellar Purkinje
cells in rats given ibogaine at a dose of 100 mg/kg i.p. (159,160). However, the
available evidence suggests that the neurotoxic effects of ibogaine may occur at
levels higher than those observed to have effects on opioid withdrawal and self-
administration. Molinari et al. (161) found no evidence of cerebellar Purkinje cell
degeneration with 40 mg/kg i.p. administered as a single dose, which is reported
to reduce morphine or cocaine self-administration or morphine withdrawal in rats
(29,119,126,161). Xu et al. (162) evaluated biomarkers of cerebellar
neurotoxicity in rats treated with single doses of ibogaine of 25, 50, 75, and 100
mg/kg i.p. The biomarkers used in this study included the specic labeling of
degenerating neurons with silver, and Purkinje neurons with antisera to calbindin.
Astrocytes were identied with antisera to glial brillary acidic protein (GFAP),
a marker of reactive gliosis, a general response of astrocytes to CNS injury. The
25 mg/kg dosage was found to correspond to a no-observable-adverse-effect-
level (NOAEL). Helsley et al. (102) treated rats with 10 mg/kg ibogaine every
other day for 60 days and observed no evidence of neurotoxicity.
Regarding the question of neurotoxicity in brain areas outside the cerebellum,
OHearn and Molliver (163) have stated, Evidence of neuronal injury following
ibogaine administration in rats appears to be almost entirely limited to the
cerebellum. While the cerebellum appears to be the brain region most vulnerable
to neurotoxic effects of ibogaine, some research has addressed the issue of
neurotoxicity in other brain regions. OCallaghan et al. (164) examined GFAP in
male and female rats exposed to either an acute regimen of ibogaine
administered at doses of 50, 100, or 150 mg/kg i.p. daily for 3 days or a chronic
regimen of daily oral administration of 25, 75, or 150 mg/kg for 14 days. The
acute i.p. regimen produced elevations of GFAP in animals of either gender that
were not restricted to the cerebellum, and were observed in the cerebellum and
hippocampus at the 50 mg/kg dosage level, and in the cortex, hippocampus,
olfactory bulb, brain stem, and striatum at the 100 mg/kg level. The effect of the
acute ibogaine regimen on GFAP was no longer evident at 14 days with either
dosage in male rats, and was restricted to the cerebellum with the 100 mg/kg dose
in female rats. GFAPlevels were examined at 17 days after the completion of the
chronic dosing regimen. No elevations of GFAP were found in any of the brain
regions examined at any of the dosages administered utilizing the chronic
regimen in males, and elevations of GFAP were found only in females, which
were restricted to the hippocampus with the 25 mg/kg dosage regimen and were
22 kenneth r. alper
present in the hippocampus, olfactory bulb, striatum, and brain stem with the 150
mg/kg dosage regimen.
OHearn et al. (159) found GFAP elevations in the cerebellum only, and not the
forebrain of male rats administered 100 mg/kg doses i.p on up to 3 consecutive
days. Elevations of GFAP are relatively sensitive, but not specic to, neuronal
degeneration (162). Using a silver degeneration-selective stain as a histologic
marker of neurodegeneration, Scallet et al. (165) examined diverse brain regions
in rats and mice treated with single 100 mg/kg doses of ibogaine administered i.p.
and found evidence of neurodegeneration only in the cerebellum in rats, whereas
mice showed no evidence of neurodegeneration. In rats that received a dose of
ibogaine of 100 mg/kg i.p., neuronal degeneration was conned to the cerebellum
as revealed by staining with Fluoro-Jade, a recently developed sensitive and
denitive marker of neuronal degeneration (166,167).
Sensitivity to ibogaine neurotoxicity appears to vary signicantly between
species. The monkey appears to be less sensitive to potential ibogaine
neurotoxicity than the rat (36). Mash et al. observed no evidence of neurotoxicity
in monkeys treated for 5 days with repeated oral doses of ibogaine of 5 to 25
mg/kg, or subcutaneously administered doses of 100 mg/kg (36). Another species
difference in sensitivity is the mouse, which unlike the rat shows no evidence of
cerebellar degeneration at a 100 mg/kg i.p. dose of ibogaine (165).
2. Mechanisms of Neurotoxicity
Ibogaines cerebellar toxicity could be related to excitatory effects mediated by
sigma2receptors in the olivocerebellar projection, which sends glutaminergic
excitatory input to cerebellar Purkinje cells, whose synaptic redundancy makes
them particularly vulnerable to excitotoxic injury (160). Sigma2agonists are
reported to potentiate the neuronal response to NMDA (95), and potentiation of
glutamatergic responses at Purkinje cells might lead to the observed
neurotoxicity. Sigma2agonists have also been shown to induce apoptosis, and
activation of sigma2receptors by ibogaine results in direct neurotoxicity via
induction of apoptosis in in vitro cell culture systems (168,169). Elsewhere in this
volume, Bowen discusses the effects of iboga alkaloids at sigma2receptors. It is
possible therefore that ibogaines neurotoxic effect on the highly sensitive
Purkinje neurons is the result of combined direct neurotoxicity and excitotoxicity
due to the enhancement of glutamatergic activity, both effects being mediated by
sigma2receptors. The agonist activity of ibogaine at the sigma2receptor might
explain the apparent paradox of ibogaine-induced excitotoxicity, despite its
properties as an NMDA antagonist (42). The neurotoxic effects of iboga alkaloids
can apparently be dissociated from their putative effects on addiction, since
sigma2receptors appear not to be involved in the suppression of drug self-
administration. 18-MC, an ibogaine congener with relatively much less sigma2
afnity, reportedly produces effects similar to ibogaine on morphine and cocaine
23
1. ibogaine: a review
administration in rats, but has shown no evidence of neurotoxicity, even at high
dosages (42,75,120).
Ibogaines NMDA antagonist activity has been cited as a rationale for a patent
for its use as a neuroprotective agent to minimize excitotoxic damage in stroke
and anoxic brain injury (170). In methamphetamine-treated mice, ibogaine is
reported to protect against hyperthermia and the induction of heat shock protein,
which are possible mediators of methamphetamine neurotoxicity (171). Binienda
et al. in this volume report an accentuation of delta amplitude in ibogaine
pretreated animals given cocaine, and they suggest a paradoxical proconvulsant
effect resulting from the interaction of cocaine and ibogaine, similar to
interactions reported between cocaine and other noncompetitive NMDA
antagonists. However, ibogaine is reported to protect against convulsions
produced by electroshock (61), or the administration of NMDA (55). Luciano et
al. (148) did not observe EEG abnormalities in ve human subjects during
treatment with ibogaine in the dosage range of 20 to 25 mg/kg. There is
apparently no reported human data on possible differences between the pre- and
post-ibogaine treatment EEG, or effects persisting into extended periods of time
after treatment.
3. Tremor
Ibogaine has been noted to produce tremor at dosages of 10 mg/kg i.p. in rats
(172) and 12 mg/kg s.c. in mice (173). Glick et al. (119 ) evaluated ibogaine and
several other iboga alkaloids and found that their effects on drug self-adminis-
tration and tendency to produce tremor were independent from one another.
Studies of structure-activity relationships of the iboga alkaloids indicate that the
tendency to cause tremor is enhanced by the presence of a methoxy group at
position 10 or 11 and is diminished or eliminated by the presence of a
carbomethoxy group at position 16 (173,174). Accordingly, tremors were not
produced in rats administered noribogaine, which differs from ibogaine with
respect to the absence of a methoxy group at position 10, at a dosage of 40 mg/kg
i.p. (124). Likewise, tremors were not observed in rats administered a dosage of
18-MC as high as 100 mg/kg. 18-MC differs from ibogaine with respect to the
absence of a methoxy group at position 10 and the presence of a carbomethoxy
group at position 16 (120).
4. Observations in Humans
Concern over possible neurotoxicity led Mash et al. to quantitatively
investigate ibogaines effects on postural stability, body tremor, and appendicular
tremor in humans (36). In U.S. FDA safety trials, nine subjects receiving 1 and 2
mg/kg of ibogaine showed only a statistically insignicant increase in body sway
6 hours after taking ibogaine. Ten patients evaluated 5 to 7 days after receiving
doses of ibogaine ranging from 10 to 30 mg/kg showed no evidence of
24 kenneth r. alper
abnormality on quantitative measures of static or dynamic posturography or hand
accelometry, or on clinical neurologic exam.
A woman died in the United States in 1994 who had been previously treated
with ibogaine 25 days earlier (36). This woman had undergone four separate
treatments with ibogaine in dosages ranging from 10 to 30 mg/kg in the 15
months prior to her death. The cause of death was concluded to have been a
mesenteric arterial thrombosis related to chronic cellulitis, and a role for ibogaine
in causing the fatality was not suspected. Of interest with regard to concerns over
potential neurotoxicity, was the absence of any neuropathological abnormality
not associated with chronic IV drug use. Neuropathological examination revealed
only slight medullary neuroaxonal dystrophy and an old focal meningeal brosis,
which were explainable on the basis of chronic IV drug use (36). There was no
evidence of cytopathology or neurodegenerative changes in the cerebellum or any
other brain area, nor was there evidence of astrocytosis or microglial activation.
B. Cardiovascular Effects
Glick et al. (45) found no changes in resting heart rate or blood pressure in rats
at the dose of 40 mg/kg of ibogaine, which was often used in that laboratory in
drug withdrawal or self-administration studies. Higher doses of ibogaine (100
and 200 mg/kg) decreased the heart rate without an effect on blood pressure, and
18-MC had no apparent effect on heart rate or blood pressure at any of the above
doses. Binieda et al. (90) found a signicantly decreased heart rate in rats given
50 mg/kg of ibogaine.
Mash et al. (175) reported on intensive cardiac monitoring in 39 human
subjects dependent on cocaine and/or heroin who received xed doses of
ibogaine of 500, 600, 800, or 1000 mg. Six subjects exhibited some signicant
decrease of resting pulse rate relative to baseline, one of whom evidenced a
signicant decrease in blood pressure, which was attributed to a transient
vasovagal response. Monitoring revealed no evidence of EKG abnormalities
appearing or intensifying during ibogaine treatment. No signicant adverse
events were seen under the study conditions, and it was concluded that the single
dose of ibogaine was apparently well tolerated. In their chapter in this volume,
Mash et al. comment further that random regression of vital signs showed no
changes across time or by dosage in opiate-dependent subjects. They did however
observe the occurrence of a hypotensive response to ibogaine in some cocaine-
dependent subjects, which was responsive to volume repletion.
C. Fatalities
The LD50 of ibogaine is reportedly 145 mg/kg i.p. and 327 mg/kg intragas-
trically in the rat, and 175 mg/kg i.p. in the mouse (158).
25
1. ibogaine: a review
In June 1990, a 44 year-old woman died in France approximately 4 hours after
receiving a dose of ibogaine of about 4.5 mg/kg. The cause of death was
concluded to have been acute heart failure in an autopsy carried out at the
Forensic-Medical Institute in Zurich (176). Autopsy revealed evidence of a prior
myocardial infarction of the left ventricle, severe atherosclerotic changes, and 70
to 80% stenosis of all three major coronary artery branches. This patient had a
history of hypertension, and inverted T waves were noted on EKG three months
prior to the patients death. The autopsy report concluded that the patients
preexisting heart disease was likely to have caused the patients death, and it
specically excluded the possibility of a direct toxic effect of ibogaine. The report
acknowledged the possibility that an interaction between ibogaine and the
patients preexisting heart condition could have been a contributing factor in the
fatal outcome.
The autopsy report, which included information obtained from the patients
family physician, and the psychiatrist who administered ibogaine, makes
reference to the possibility that the patient might have taken other drugs. The
autopsy report noted the presence of amphetamine in the enzyme immunocyto-
chemical (EMIT) assay of a dialysate of the kidney tissue (urine was reported not
to be obtainable). This nding, however, was regarded as artifactual and possibly
attributable to a false positive EMIT result due to the presence of phenylethy-
lamine.
A fatality occurred during a heroin detoxication treatment of a 24-year-old
female in the Netherlands in June 1993. This incident was a signicant factor in
the NIDA decision not to fund a clinical trial of ibogaine in 1995. The patient
received a total ibogaine dose of 29 mg/kg and suffered a respiratory arrest and
died 19 hours after the start of the treatment. Forensic pathological examination
revealed no denitive conclusion regarding the probable cause of death (177) and
cited the general lack of information correlating ibogaine concentrations with
possible toxic effects in humans. The high levels of noribogaine found in the
deceased patient were possibly consistent with saturation of elimination kinetics.
However, the higher levels of noribogaine in heart, relative to femoral blood, also
suggested signicant postmortem redistribution of noribogaine. The potential
artifact associated with a high volume of distribution and postmortem release of
drug previously sequestered in tissue (51,139,158) limits the interpretability of
postmortem levels of noribogaine.
Some evidence suggested the possibility of surreptitious opioid use in this
case, which was noted in the Dutch inquiry (178) and which is another source of
uncertainty in this fatality. There is evidence suggesting that the interaction of
opioids and ibogaine potentiates opioid toxicity (68,179). Analysis of gastric
contents for heroin or morphine, which might have conrmed recent heroin
smoking, and analysis of blood for 6-monoacetyl morphine, a heroin metabolite
whose presence indicates recent use (180), were not performed. This incident
26 kenneth r. alper
underscores the need for the security and medical supervision available in a
conventional medical setting, and for completion of dose escalation studies to
allow systematic collection of pharmacokinetic and safety data.
In London, in January 2000, a 40-year-old heroin addict died after having
allegedly taken 5 g of iboga alkaloid extract 40 hours prior to his death (38, see
the chapter by Alper et al. in this volume). The extract was said to have contained
approximately ve times the alkaloid content of the dried rootbark. The ofcial
British inquest regarding this matter is still in progress as of the time of the
writing of this book.
D. Abuse Liability
The available evidence does not appear to suggest that ibogaine has signicant
potential for abuse. The 5-HT2A receptor, the primary mediator of responding for
LSD and other commonly abused drugs classied as hallucinogenic or
psychedelic, does not appear to be essential to discriminability of the ibogaine
stimulus (84,96). Ibogaine is reportedly neither rewarding or aversive in the
conditioned place preference paradigm (132). Rats given either 10 or 40 mg/kg
ibogaine daily for 6 consecutive days did not show withdrawal signs (129).
Animals do not self-administer 18-MC, an ibogaine analog, in paradigms in
which they self-administer drugs of abuse (45). None of the consultants to NIDA
in the 1995 Ibogaine Review Meeting identied the possible abuse of ibogaine as
a potential safety concern.
VII. Learning, Memory, and Neurophysiology
A. Learning, Memory, and Addiction
Drug abusers may be viewed as having a disorder involving excess attribution
of salience to drugs and drug-related stimuli (181), which suggests the possibility
of a role of processes subserving learning and memory in the acquisition of the
pathological motivational focus in addiction (182-185). Learning, in the most
general sense, can be viewed as the modication of future brain activity, of which
thought, motivation, consciousness, or sensory experience are emergent
properties, on the basis of prior experience. This broad denition subsumes
everything from social behavior to learning to read, to the neuroadaptations of
drug tolerance and dependence.
Addiction can be argued to involve the pathological acquisition or learning
of associations of drug related stimuli with motivational states corresponding to
27
1. ibogaine: a review
valuation and importance (181,183,184). The pathological learning of addiction
differs from that of normal learning in at least two important respects. First, the
acquisition of drug salience in addiction does not involve learned associations
between drug-related external cues or internal representations, and the experience
of external events as they actually occur. Instead, the imprinting or stamping
in of drug incentives appears to involve alterations of neural plasticity in
processes that subserve motivation, memory and learning, resulting in neural
behavior that to a signicant extent has escaped the constraint of validation by
experience with external reality (183-186). Dopamine and glutamate
transmission are thought to be involved in the modulation of neural plasticity of
both normal learning and the neuroadaptations of drug salience (184). Second,
drug-related learning does not apparently habituate (184). Unlike normal
learning, the drug stimulus appears to be experienced as perpetually novel and
continues to command attention and be attributed with salience unattenuated by
habituation (53,182).
B. Effects of Ibogaine on Learning and Memory
Ibogaine appears to have signicant effects on brain events involved in
learning and the encoding of drug salience. Ibogaine interacts signicantly with
the NMDA receptor (39,58,179), which is involved in long term potentiation
(LTP), a process thought to be important in neural plasticity, memory, and
learning (182,184,187). Experiences apparently involving memory, such as
panoramic recall, are prominent in descriptions by individuals who have taken
ibogaine (14).
The observation of an effect of ibogaine on the expression of behavioral
sensitization to amphetamine, but not a conditioned place preference (188), raises
the interesting possibility of a relatively selective effect of ibogaine on the
pathological encoding of drug salience, distinguished from learning involving
non-drug incentives. Ibogaine reportedly attenuates the acquisition of place
preference for morphine or amphetamine (131,132). A general effect of
interference with learning has been suggested (189), but studies on spatial
learning show an actual enhancement by ibogaine (102,190). Consistent with a
selective effect on neuroadaptations acquired from drug exposure are ibogaines
effects on locomotor activity and dopamine efux in the NAc, which are
relatively more evident in animals with prior experience with morphine (112,113)
or cocaine (111).
C. Ibogaine and the EEG
Studies of animals treated acutely with ibogaine report a desynchronized EEG
with fast low amplitude activity, a state described as activated or aroused
28 kenneth r. alper
(89,90,191). Binienda et al. (90) noted a decline in delta amplitude and
interpreted this as consistent with activation of dopaminergic receptors. However,
observations on the interaction of atropine and ibogaine with respect to the EEG
suggest the involvement of ascending cholinergic input. Depoortere (191) found
that ibogaine enhanced an atropine-sensitive theta frequency EEG rhythm in rats.
Schneider and Sigg (89) observed a shift toward high-frequency low-voltage
EEG activity following the administration of ibogaine to cats, and they noted that
this effect was blocked by the administration of atropine. Luciano et al. (148)
observed no changes in the visually evaluated EEG in humans administered 20 to
25 mg/kg ibogaine.
D. Goutarels Hypothesis
The French chemist Robert Goutarel (14) hypothesized that ibogaine treatment
involves a state with functional aspects shared by the brain states of REM sleep,
with important effects on learning and memory. During the REM state, there is
believed to be reconsolidation of learned information in a state of heightened
neural plasticity, with the reprocessing of previously learned information and the
formation of new associations (192,193). Goutarel suggested that a REM-like
state may be induced by ibogaine, which corresponds to a window of heightened
neural plasticity, during which there may be weakening of the pathological
linkages between cues and representations of the drug incentive and the motiva-
tional states with which they have become paired (14). Analogous to the
reconsolidation of learned information that is thought to occur during the REM
state (192,193), Goutarel theorized that the pathological learning of addiction was
modied during ibogaine treatment. He appears to have based his theoretical
formulation mainly on reports of the phenomenological experiences of awake
ibogaine-treated subjects that share features in common with dreams. Goutarels
hypothesis is speculative, but nonetheless has an interesting apparent consistency
with the literature on the relationship of learning and addiction and the
physiologic function of the REM EEG state with regard to the consolidation of
learned information.
There is some evidence that may be viewed as consistent with Goutarels
hypothesis. Goutarels belief in a relationship of the ibogaine-treated EEG state
to that of REM is supported by studies in animals treated with ibogaine that report
an apparently activated or desynchronized EEG state consistent with arousal,
vigilance, or REM sleep (90,191). The observation that ibogaine enhanced an
atropine-sensitive theta frequency rhythm (191) suggests the possible
involvement of ascending cholinergic input, which is an essential determinant of
EEG desynchronization during REM sleep (192). The possible reconsolidation of
learned information due to heightened plasticity during both the REM and
ibogaine-induced desynchronized EEG states is suggested by the observation that
29
1. ibogaine: a review
EEG dyssynchrony is associated with an increased facilitation of Hebbian
covariance (194), which is believed to be an important determinant of the neural
plasticity involved in consolidation of learning and memory. Also, with regard to
a possible analogy of the REM and ibogaine induced brain states, some ibogaine
treatment guides have anecdotally mentioned that they have observed REM-like
eye movements in awake patients during treatments (195,196).
VIII. Anthropological and Sociological Perspectives
As discussed in various aspects by this volume by the Fernandezes, Frenken,
and Lotsof and Alexander, ibogaines use appears to involve distinctive
interactions of psychopharmacologic effects with set and setting in both the
subcultures of the United States and Europe, and the centuries older, sacramental
context of the use of iboga in Bwiti, the religious movement in West Central
Africa. In the Bwiti religious subculture, and arguably to some extent in the
European ibogaine subculture, there is the common attribute of a group of
initiates that seek to facilitate healing through the afliation of the collective with
the individual. In both the African and U.S./European contexts, the ibogaine
experience has been attributed to serving the objective of facilitating personal
growth and change. Use of ibogaine in both contexts has been criticized as
involving the use of an addictive or hallucinogenic agent, and it appears to
some extent to involve the formation of a subculture among individuals
confronted with marginal social circumstances such as colonialism, or the state of
addiction (197-199, see also Fernandez and Fernandez in this volume).
Galanter (200) identies three important psychological features that he regards
as descriptive of the process of charismatic groups or zealous self-help
movements such as 12-step programs that appear to also be relevant to Bwiti.
These three processes are group cohesiveness, shared belief, and altered
consciousness, such as that of religious ecstasy or insight to which the group can
attribute a new construction of reality in their life. An understanding of these
powerful behavioral inuences could be useful in optimizing the clinical milieu
and interpersonal dynamics of present conventional treatment settings, or of
future treatment settings, if ibogaine or a congener should receive ofcial
approval.
The application of ethnographic techniques to the analysis of the phenomeno-
logical features of the acute treatment experience could be informative from a
neuropsychiatric, as well as from a cultural perspective. For example, similar
subjective phenomena are frequently described in both ibogaine treatment and
near death experiences (NDEs) (14,152,199,201) such as panoramic memory;
30 kenneth r. alper
calm, detached emotional tone; specic experiences, such as passage along a long
path or oating; visions or waking dream states featuring archetypal
experiences such as contact with transcendent beings; and the frequent attribution
of transcendent signicance to the experience. Such shared features between
ibogaine and NDEs suggest a common transcultural phenomenology of
transcendent or religious experience or, alternatively, the possibility of a similar
subjective experience due to the inuence of a common underlying neurobio-
logical mechanism such as NMDA transmission (202).
IX. Economic and Political Perspectives
A. Economic Incentives and the Development of Ibogaine
The academic research community working in the public sector has a crucial
role in studying ibogaine as a paradigm for the development of new treatment
approaches. The strategy of relying on the pharmaceutical industry to underwrite
the cost of drug development works extremely well in many instances, but
appears to present some limitations with regard to the development of pharma-
cotherapy for addiction in general, and specically ibogaine.
In the public sector, the major economic incentives for the development of
addiction treatment are the saved costs associated with preventing lost economic
productivity, medical morbidity, or crime. In the private sector, decisions are
based on weighing the expense of development against the expected prot, and
not the magnitude of saved economic or social costs. Owing to limited nancial
incentives in the form of insurance reimbursements and a perceived lack of
breakthrough compounds, the U.S. pharmaceutical industry has not generally
viewed addiction as an attractive area for development (203), and expenditures
for the development of medications for addiction are small relative to those to
develop drugs for other indications. Ibogaine is particularly unattractive to
industry for several reasons: its mechanism of action is apparently complex and
incompletely understood, it may present signicant safety issues, it is a naturally
occurring alkaloid whose structure cannot itself be patented, and some of its use
patent are close to expiration.
There is arguably an important role for academic/public-sector development in
the case of a theoretically interesting drug with a limited prot potential and
signicant developmental expense such as ibogaine. However, the entire annual
expenditures for medications development in NIDA, which accounts to about
90% of U.S. public sector spending on developing addiction pharmacotherapy, is
on the order of approximately $60 million, a fraction of the average cost of
31
1. ibogaine: a review
successfully developing a drug to market, which is estimated to exceed $300
million (204). Opportunities to fund research on ibogaine are limited by factors
that generally affect the development of other drugs to treat addiction: a limited
public sector budget in the presence of disproportionately low private-sector
expenditures on the development of pharmacotherapies for addiction relative to
other indications (203).
B. Political Issues
The chapter by Alper et al. in this volume describes the medical subculture of
the informal ibogaine treatment scene and the political subculture of advocacy for
the development and availability of ibogaine. These scenes are a distinctive and
signicant aspect of ibogaines history, which arguably have impacted on
decisions regarding its development. From a clinical standpoint, the informal
treatment subculture has been an important source of information on human
experience with ibogaine (32).
From a political or historical standpoint, the informal treatment subculture has
viewed itself as a form of activism or civil disobedience on the part of its partic-
ipants seeking a treatment, despite a lack of ofcial approval (34). Ibogaine has
been associated with a vocal activist subculture, which views its mission as
making controversial treatments available to a stigmatized minority group of
patients suffering from a life-threatening illness, and has utilized tactics intended
to engage the attention of the press (34). These confrontational media-oriented
tactics may well have provoked negative reactions at times, but may also have
inuenced Curtis Wright, the former FDA ibogaine project ofcer, to write in
1995 that . . . a signicant portion of the public we serve believes the drug merits
investigation (205).
X. Conclusions
Evidence that supports the possible efcacy of ibogaine as a treatment for
addiction includes case reports in humans, and effects in preclinical models of
drug dependence. The case report evidence has mainly involved the indication of
acute opioid withdrawal, and there appears to be consistency between earlier
observations derived from informal treatment contexts (32-36,146-150) and more
recent work from a setting that appears to conform to a conventional medical
model (123, Mash et al. in this volume). The continued existence of informal
treatment scenes parallels case report evidence indicating possible efcacy.
Animal work has provided observations of attenuation of opiate withdrawal signs
32 kenneth r. alper
and reductions of self-administration of a variety of drugs including morphine,
cocaine, alcohol, and nicotine. Preclinical models have also yielded evidence that
with respect to certain abused drugs, ibogaine may dampen responses that may
be associated with dependence, such as dopamine efux in the NAc or locomotor
activation.
Ibogaines pharmacologic profile includes interactions with multiple
neurotransmitter systems that could plausibly be related to addiction, including
NMDA, nicotinic, mu- and kappa-opioid, and serotonergic systems. The putative
efcacy of ibogaine does not appear fully explainable on the basis of interactions
with any single neurotransmitter system, or on the basis of currently utilized
pharmacologic strategies such as substitution therapies, or monoamine reuptake
inhibition. Ibogaines effects may result from interactions between multiple
neurotransmitter systems, and might not be attributable to actions at any single
type of receptor. The apparently persistent effect of ibogaine has been suggested
to involve a long-lived metabolite. Some evidence suggests effects on second
messenger signal transduction, an interesting possibility that could conceivably
result from interactions between multiple neurotransmitter systems and produce
persistent effects lasting beyond the duration of occupancy at receptor sites. Work
with ibogaine congeners suggests that other iboga alkaloids can be developed that
might minimize unwanted toxic, or possibly behavioral effects, while retaining
apparent efcacy in drug dependence. In summary, the available evidence
suggests that ibogaine and the iboga alkaloids may have efcacy in addiction on
the basis of mechanisms that are not yet known and which can possibly be
dissociated from toxic effects, and may present signicant promise as a paradigm
for the study and development of pharmacotherapy for addiction.
References
1. H. Pope, Econ. Bot. 23, 174 (1969).
2. S. Budvari and M.J. ONeil, Merck Index (S. Budvari and M.J. ONeil, eds.), Chapman and
Hall, New York, NY, 1996.
3. G. Arai, J. Coppola and C.A. Jeffrey, Acta Cryst. 13, 553 (1960).
4. K. Biemann and Friedmann-Spiteller, J. Am. Chem. Soc. 83, 4805 (1961).
5. G.V. Binst, C. Danhoux, C. Hootele, J. Pecher and R.H. Martin, Tetrahedron Lett. 973 (1964).
6. J. Pecher, R.H. Martin, N. Defay, M. Kaisin, J. Peeters, G.V. Binst, N. Verzele, and F.
Alderweireldt, Tetrahedron Lett. 270 (1961).
7. M. Damak, C. Poupat, and A. Ahond, Tetrahedron Lett. 39, 3531 (1976).
8. F. Ladhar, N. Gorbel, and M. Damak, J. de la Societé Chimique de Tunisie 5, 43 (1981).
9. E. Wenkert, D.W. Cochran, H.E. Gottlieb, and E.W. Hagaman, Helv. Chim. Acta. 59, 2437
(1976).
10. M.E. Alburges, R.L. Foltz, and D.E. Moody, J Anal. Toxicol. 19, 381 (1995).
11. C.A. Gallagher, L.B. Hough, S.M. Keefner, A. Syed-Mozaffari, S. Archer, and S.D. Glick,
33
1. ibogaine: a review
Biochem. Pharmacol. 49, 73 (1995).
12. W.L. Hearn, J. Pablo, G.W. Hime, and D.C. Mash, J Anal. Toxicol. 19, 427 (1995).
13. F.R. Ley, A.R. Jeffcoat, and B.F. Thomas, J. Chromatogr. 723, 101 (1996).
14. R. Goutarel, O. Gollnhofer, and R. Sillans, Psyched. Mono. Essays 6, 70 (1993).
15. J. Dybowsky and E. Landrin, Compt. Rend. Acad. Sci. 133, 748 (1901).
16. A. Haller and E. Heckel, Compt. Rend. Acad. Sci. 133, 850 (1901).
17. M. Lambert and E. Heckel, Compt. Rend. Acad. Sci. 133, 1236 (1901).
18. H. Isbell, Ciba Ibogaine File Document No. AB0491-492 410, 1955.
19. W.I. Taylor, J. Am. Chem. Soc. 79, 3298 (1957).
20. D.F. Dickel, C.I. Holden, R.C. Maxeld, L.E. Paszak, and W.I. Taylor, J. Am. Chem. Soc. 80,
123 (1958).
21. G. Büchi, D.L. Coffen, K. Kocsis, P.E. Sonnet, and F.E. Ziegler, J. Am. Chem. Soc. 88, 3099
(1966).
22. H.S. Lotsof, U.S. Patent 4,499,096; Chem. Abstr. 102,160426w (1985).
23. H.S. Lotsof, U.S. Patent 4,587,243; Chem. Abstr. 106,12967r (1986).
24. D.P. Bocher and C. Naranjo, French Special Drug Patent No. 138.081;081713m, Inst. Class
A61k (1969).
25. H.S. Lotsof, U.S. Patent 4,857,523; Chem. Abstr. 112,32041m (1989).
26. H.S. Lotsof, U.S. Patent 5,026,697; Chem. Abstr. 116,17031x (1991).
27. H.S. Lotsof, U.S. Patent 5,152,994; Chem. Abstr. 116,100980b (1992).
28. E.D. Dzoljic, C.D. Kaplan, and M.R. Dzoljic, Arch. Int. Pharmacodyn. Ther. 294, 64 (1988).
29. S.D. Glick, K. Rossman, S. Steindorf, I.M. Maisonneuve, and J.N. Carlson, Eur. J. Pharmacol.
195, 341 (1991).
30. S.D. Glick, C.A. Gallagher, L.B. Hough, K.L. Rossman, and I.M. Maisonneuve, Brain Res.
588, 173 (1992).
31. T.S.L. Cappendijk and M.R. Dzoljic, Eur. J. Pharmacol. 14, 261 (1993).
32. K.R. Alper, H.S. Lotsof, G.M.N. Frenken, D.J. Luciano, and J. Bastiaans, Am. J. Addict. 8, 234
(1999).
33. H.S. Lotsof, Multidiscip. Assoc. Psyched. Stud. 5, 16 (1995).
34. P. DeRienzo and D. Beal, The Ibogaine Story, Autonomedia, Brooklyn, New York, 1997.
35. B. Sisko, Multidiscip. Assoc. Psyched. Stud. 4, 15 (1993).
36. D.C. Mash, C.A. Kovera, B.E. Buck, M.D. Norenberg, P. Shapshak, W.L. Hearn, and J.
Sanchez-Ramos, Ann. N. Y. Acad. Sci. 844, 274 (1998).
37. MDD-NIDA, Draft Protocol: Rising Dose Tolerance Study using Single Administration to
Assess the Safety and Preliminary Efcacy of Ibogaine for the Treatment of Cocaine and/or
Heroin Dependence, NIDA, Rockville, MD, 1995.
38. egroups, http://www.egroups.com/group/ibogaine, 2000.
39. P. Popik and P. Skolnick, in The Alkaloids (G.A. Cordell, ed.), p. 197. Academic Press, New
York, 1999.
40. P.M. Sweetnam, J. Lancaster, A. Snowman, J.L. Collins, S. Perschke, C. Bauer, and J.
Ferkany, Psychopharmacology 118, 369 (1995).
41. H. Sershen, A. Hashim, and A. Lajtha, Brain Res. Bull. 42, 161 (1997).
42. S.D. Glick and I.M. Maisonneuve, Ann. N. Y. Acad. Sci. 844, 214 (1998).
43. D.C. Deecher, M. Teitler, D.M. Soderlund, W.G. Bornmann, M.E. Kuehne, and S.D. Glick,
Brain Res. 571, 242 (1992).
44. J.K. Staley, Q. Ouyang, J. Pablo, W.L. Hearn, D.D. Flynn, R.B. Rothman, K.C. Rice, and D.C.
Mash, Psychopharmacology 127, 10 (1996).
45. S.D. Glick, I.M. Maisonneuve, M.E. Kuehne and U.K. Bandarage, CNS Drug Rev. 5, 27
(1999).
46. M.E.M. Benwell, P.E. Holton, R.J. Moran, and D.J.K. Balfour, Br. J. Pharmacol. 117, 743
(1996).
47. A.S. Schneider, J.E. Nagel, and S.J. Mah, Eur. J. Pharmacol. 317, R1 (1996).
48. B. Badio, W.L. Padgett and J.W. Daly, Mol. Pharmacol. 51, 1 (1997).
34 kenneth r. alper
49. S.J. Mah, Y. Tang, P.E. Liauw, J.E. Nagel, and A.S. Schneider, Brain Res. 797, 173 (1998).
50. J.D. Fryer and R.J. Lukas, J. Pharmacol. Exp. Ther. 288, 88 (1999).
51. L.B. Hough, S.M. Pearl, and S.D. Glick, Life Sci. 58, L119 (1996).
52. A. Bisaga and P. Popik, Drug & Alcohol Depend. 59, 1 (2000).
53. B.H. Herman, F. Vocci, and P. Bridge, Neuropsychopharmacology 13, 269 (1995).
54. B.H. Herman and C.P. OBrien, Seminars Neurosci. 9, 158 (1997).
55. K. Chen, T.G. Kokate, S.D. Donevan, F.I. Carroll, and M.A. Rogawski, Neuropharmacology
35, 423 (1996).
56. R.T. Layer, P. Skolnick, C.M. Bertha, U.K. Bandarage, M.E. Kuehne, and P. Popik, Eur. J.
Pharmacol. 309, 159 (1996).
57. P. Popik, R.T. Layer, and P. Skolnick, Psychopharmacology 114, 672 (1994).
58. P. Popik, R.T. Layer, L.H. Fossom, M. Benveniste, B. Geter-Douglass, J.M. Witkin, and P.
Skolnick, J. Pharmacol. Exp. Ther. 275, 753 (1995).
59. D.C. Mash, J.K. Staley, J. Pablo, A.M. Holohean, J.C. Hackman, and R.A. Davidoff, Neurosci.
Lett. 192, 53 (1995).
60. Y. Itzhak and S.F. Ali, Ann. N. Y. Acad. Sci. 844, 245 (1998).
61. B. Geter-Douglass and J.M. Witkin, Psychopharmacology 146, 280 (1999).
62. S.F. Ali, G.D. Newport, W. Slikker, Jr., R.B. Rothman, and M.H. Baumann, Brain Res. 737,
215 (1996).
63. M.H. Baumann, R.B. Rothman, and S.F. Ali, Ann. N. Y. Acad. Sci. 844, 252 (1998).
64. E.E. Codd, Pharmacol. Lett. 57, 315 (1995).
65. J.P. Pablo and D.C. Mash, Neuroreport 9, 109 (1998).
66. S.M. Pearl, K. Herrick-Davis, M. Teitler, and S.D. Glick, Brain Res. 675, 342 (1995).
67. M.K. Mundey, N.A. Blaylock, R. Mason, S.D. Glick, I.M. Maisonneuve, and V.G. Wilson, Br.
J. Pharmacol. 129, 1561 (2000).
68. J.A. Schneider and M. McArthur, Experientia 12, 323 (1956).
69. B. Frances, R. Gout, J. Cros, and J.M. Zajac, Fundament. Clin. Pharmacol. 6, 327 (1992).
70. H.N. Bhargava, Y.J. Cao, and G.M. Zhao, Brain Res. 752, 234 (1997).
71. S.D. Glick, Personal Communication, October 19, 2000.
72. R.A. Rabin and J.C. Winter, Eur. J. Pharmacol. 316, 343 (1996).
73. Y.J. Cao and H.N. Bhargava, Brain Res. 752, 250 (1997).
74. S.D. Glick, I.M. Maisonneuve, and S.M. Pearl, Brain Res. 749, 340 (1997).
75. S.D. Glick, I.M. Maisonneuve, K.E. Visker, K.A. Fritz, U.K. Bandarage, and M.E. Kuehne,
Psychopharmacology 139, 274 (1998).
76. S.D. Glick, I.M. Maisonneuve, J. Raucci, and S. Archer, Brain Res 681, 147 (1995).
77. S.M. Pearl and S.D. Glick, Neurosci Lett. 213, 5 (1996).
78. H. Sershen, A. Hashim, and A. Lajtha, Brain Res. Bull. 36, 587 (1995).
79. D.C. Mash, J.K. Staley, M.H. Baumann, R.B. Rothman, and W.L. Hearn, Life Sci. 57, L45
(1995).
80. D. Wei, I.M. Maisonneuve, M.E. Kuehne, and S.D. Glick, Brain Res. 800, 260 (1998).
81. G.B. Wells, M.C. Lopez, and J.C. Tanaka, Brain Res. Bull. 48, 641 (1999).
82. R. Glennon, Neuropsychopharmacology 3, 509 (1990).
83. D. Repke, D. Artis, and E.H.Z. Wong, J. Org. Chem. 59, 2164 (1994).
84. S. Helsley, D. Fiorella, R.A. Rabin, and J.C. Winter, Pharmacol. Biochem. Behav. 59, 419
(1998).
85. S. Helsley, R.A. Rabin, and J.C. Winter, Prog. Neuro-Psychopharmacol. Biol. Psychiat. 23,
317 (1999).
86. E.D. French, K. Dillon, and S. Ali, Life Sci. 59, PL 199 (1996).
87. H. Sershen, A. Hashim, L. Harsing, and A. Lajtha, Life Sci. 50, 1079 (1992).
88. M.H. Baumann, R.B. Rothman, and S.F. Ali, Drug & Alcohol Dependence 59, 143 (2000).
89. J.A. Schneider and E.B. Sigg, Ann NY Acad Sci 66, 765 (1957).
90. Z. Binienda, M.A. Beaudoin, B.T. Thorn, D.R. Papurna, R.A. Johnson, C.M. Fogle, W. Jr.
Slikker, and S.F. Ali, Ann. N. Y. Acad. Sci. 844, 265 (1998).
35
1. ibogaine: a review
91. E. Vincent and M. Serro, Compt. Rend Soc. Biol. 136, 612 (1942).
92. I.M. Maisonneuve, G.L. Mann, C.R. Deibel, and S.D. Glick, Psychopharmacology 129, 249
(1997).
93. R.H. Mach, C.R. Smith, and S.R. Childers, Life Sci. 57, L57 (1995).
94. W.D. Bowen, B.J. Vilner, W. Williams, C.M. Bertha, M.E. Kuehne, and A.E. Jacobson, Eur. J.
Pharmacol. 279, R1 (1995).
95. S. Courtre and G. Debonnel, Synapse 29, 62 (1998).
96. S. Helsley, D. Fiorella, R.A. Rabin, and J.C. Winter, Prog. Neuro-Psychopharmacol. Biol.
Psychiat. 22, 649 (1998).
97. M.D. Schecter, Life Sci 60, 83 (1997).
98. H.E. Jones, H. Li, and R.L. Balster, Pharmacol. Biochem. Behav. 59, 413 (1998).
99. S. Helsley, R.A. Filipink, W.D. Bowen, R.A. Rabin, and J.C. Winter, Pharmacol. Biochem.
Behav. 59, 495 (1998).
100. C. Zubaran, M. Shoaib, I.P. Stolerman, J. Pablo, and D.C. Mash, Neuropsychopharmacology
21, 119 (1999).
101. S. Helsley, R.A. Rabin and J.C. Winter, Eur. J. Pharmacol. 345, 139 (1998).
102. S. Helsley, C.A. Dlugos, R.J. Pentney, R.A. Rabin, and J.C. Winter, Brain Res. 759, 306
(1997).
103. B. Grella, M. Dukat, R. Young, M. Teitler, K. Herrick-Davis, C.B. Gauthier, G and R.A.
Glennon, Drug & Alcohol Depend. 50, 99 (1998).
104. S. Helsley, R.A. Rabin, and J.C. Winter, Life Sci. 60, L147 (1997).
105. M.E. Alburges and G.R. Hanson, Brain Res. 844, 96 (1999).
106. M.E. Alburges, B.P. Ramos, L. Bush, and G.R. Hanson, Eur. J. Pharmacol. 390, 119 (2000).
107. M.E. Alburges and G.R. Hanson, Brain Res. 847, 139 (1999).
108. S.D. Glick, K.E. Visker, and I.M. Maisonneuve, Eur. J. Pharmacol. 357, 9 (1998).
109. S. Schenk, B. Partridge, and T.S. Shippenberg, Psychopharmacol. (Berl.) 144, 339 (1999).
110. K.K. Szumlinski and I.M. Maisonneuve, Toxicon 39, 75 (2001).
111. K.K. Szumlinski, I.M. Maisonneuve, and S.D. Glick, Eur. J. Pharmacol. 398, 259 (2000).
112. S.M. Pearl, D.W. Johnson, and S.D. Glick, Psychopharmacology 121, 470 (1995).
113. S.M. Pearl, I.M. Maisonneuve, and S.D. Glick, Neuropharmacology 35, 1779 (1996).
114. S.S. Sharma and H.N. Bhargava, Pharmacol. 57, 1079 (1998).
115. I.M. Maisonneuve, R.W.J. Keller, and S.D. Glick, Eur. J. Pharmacol. 199, 35 (1991).
116. R.A. Rabin and J.C. Winter, Brain Res. 731, 226 (1996).
117. F.J. White and P.W. Kalivas, Drug & Alcohol Dependence 51, 141 (1998).
118. S.I. Dworkin, S. Gleeson, D. Meloni, T.R. Koves, and T.J. Martin, Psychopharmacology 117,
257 (1995).
119. S.D. Glick, M.E. Kuehne, J. Raucci, T.E. Wilson, D. Larson, J.R.W. Keller, and J.N. Carlson,
Brain Res. 657, 14 (1994).
120. S.D. Glick, M.E. Kuehne, I.M. Maisonneuve, U.K. Bandarage, and H.H. Molinari, Brain Res.
719, 29 (1996).
121. H. Sershen, A. Hashim, and A. Lajtha, Pharmacol. Biochem. Behav. 47, 13 (1994).
122. A.H. Rezvani, D.H. Overstreet, Y. Yang, I.M. Maisonneuve, U.K. Bandarage, M.E. Kuehne,
and S.D. Glick, Pharmacol. Biochem. Behav. 58, 615 (1995).
123. D.C. Mash and C.A. Kovera, in College of Problems on Drug Dependence (CPDD) 62nd
Annual Scientic Meeting, p. 121. San Juan, Puerto Rico, 2000.
124. S.D. Glick, S.M. Pearl, J. Cai, and I.M. Maisonneuve, Brain Res. 713, 294 (1996).
125. A.H. Rezvani, D.H. Overstreet, Y. Yang, I.M. Maisonneuve, U.K. Bandarage, M.E. Kuehne,
and S.D. Glick, Pharmacol. Biochem. Behav. 58, 615 (1997).
126. T.S.L. Cappendijk, D. Fekkes, and M.R. Dzoljic, Behav. Brain Res. 65, 117 (1994).
127. S.D. Glick, K. Rossman, N.C. Rao, I.M. Maisonneuve, and J.N. Carlson, Neuropharmacology
31, 497 (1992).
128. B. Rho and S.D. Glick, Neuroreport 9, 1283 (1998).
129. M.D. Aceto, E.R. Bowman, and L.S. Harris, NIDA Research Monograph 95, 576 (1990).
36 kenneth r. alper
130. L.G. Sharpe and J.H. Jaffe, Neuroreport 1, 17 (1990).
131. I. Moroz, L.A. Parker, and S. Siegel, Exp. Clin. Psychopharmacol. 5, 119 (1997).
132. L.A. Parker, S. Siegel, and T. Luxton, Learning & Memory 3, 344 (1995).
133. T. Luxton, L.A. Parker, and S. Siegel, Prog. Neuro-Psychopharmacol. Biol. Psychiat. 20, 857
(1996).
134. I.M. Maisonneuve and S.D. Glick, Eur. J. Pharmacol. 212, 263 (1992).
135. S.M. Pearl, L.B. Hough, D.L. Boyd, and S.D. Glick, Pharmacol. Biochem. Behav. 57, 809
(1997).
136. I.M. Maisonneuve, K.L. Rossman, R.W.J. Keller, and S.D. Glick, Brain Res. 575, 69 (1992).
137. H. Sershen, L.G.J. Harsing, A. Hashim, and A. Lajtha, Life Sci. 51, 1003 (1992).
138. H. Sershen, A. Hashim, and A. Lajtha, Pharmacol. Biochem. Behav. 53, 863 (1996).
139. P.A. Broderick, F.T. Phelan, F. Eng, and R.T. Wechsler, Pharmacol. Biochem. Behav. 49, 771
(1994).
140. K.K. Szumlinski, I.M. Maisonneuve, and S.D. Glick, Pharmacol. Biochem. Behav. 63, 457
(1999).
141. K.K. Szumlinski, I.M. Maisonneuve, and S.D. Glick, Psychopharmacology 145, 227 (1999).
142. K.K. Szumlinski, M.Y. Balogun, I.M. Maisonneuve, and S.D. Glick, Psychopharmacol. 151,
234 (2000).
143. I.M. Maisonneuve and S.D. Glick, Eur. J. Pharmacol. 383, 15 (1999).
144. M.S. Reid, K.J. Hsu, K.H. Souza, P.A. Broderick, and S.P. Berger, J. Neural Transm.
(Budapest) 103, 967 (1996).
145. S. Ikemoto and J. Panksepp, Brain Res. Rev. 31, 6 (1999).
146. M. Cantor, The Truth Seeker 117, 23 (1990).
147. D. Luciano, Am. J. Addict. 7, 89 (1998).
148. D. Luciano, E.A. Della Sera, and E.G. Jethmal, Multidiscip. Assoc. Psyched. Stud. 9, 27
(2000).
149. S.G. Sheppard, J. Subst. Abuse Treat. 11, 379 (1994).
150. C.D. Kaplan, E. Ketzer, J. DeJong, and M. DeVires, Social Neurosci. Bull. 6, 6 (1993).
151. H.S. Lotsof, Presented at the NIDA Ibogaine Review Meeting, Rockville, MD, 1995.
152. G. Roberts and J. Owen, Br. J. Psychiat. 153, 607 (1988).
153. A.R. Jeffcoat, C.E. Cook, J.M. Hill, D.P. Coleman, and G.M. Pollack, NIDA Research
Monograph 141, 309 (1994).
154. R.A. Upton, Presented at the NIDA Ibogaine Review Meeting, Rockville, MD, 1995.
155. R.S. Obach, J. Pablo, and D.C. Mash, Drug Metab. Disp. 26, 764 (1998).
156. C.R. Wolf and G. Smith, Br. Med. Bull. 55, 366 (1999).
157. J. Pablo, R. Tyndale, S. Obach, W.L. Hearn, and D.C. Mash, Abstracts, Sixtieth Annual
Meeting of the College of Problems on Drug Dependence, p. 108. Scottsdale, AZ, 1998.
158. H.I. Dhahir, A Comparative Study of the Toxicity of Ibogaine and Serotonin. Doctoral Thesis,
71-25-341, University Microlm International, Ann Arbor, MI, 1971.
159. E. OHearn and M.E. Molliver, Neuroscience 55, 303 (1993).
160. E. OHearn and M.E. Molliver, J. Neurosci. 17, 8828 (1997).
161. H.H. Molinari, I.M. Maisonneuve, and S.D. Glick, Brain Res. 737, 255 (1996).
162. Z. Xu, L.W. Chang, Jr., W. Slikker, S.F. Ali, R.L. Rountree, and A.C. Scallet, Toxicol. Sci. 57,
95 (2000).
163. E. OHearn and M.E. Molliver, in Cell Death and Diseases of the Nervous System (V.E.
Koliatos and R.R. Ratan, eds.), p. 221. Humana Press, Totowa, NJ, 1998.
164. J.P. OCallaghan, T.S. Rogers, L.E. Rodman, and J.G. Page, Ann. NY Acad. Sci. 801, 205
(1996).
165. A.C. Scallet, X. Ye, R. Rountree, P. Nony, and S.F. Ali, Ann. NY Acad. Sci. 801, 227 (1996).
166. L.C. Schmued, C. Albertson, and W. Slikker, Jr., Brain Res. 751, 37 (1997).
167. L.C. Schmued and K.J. Hopkins, Toxicol. Pathol. 28, 91 (2000).
168. B.J. Vilner, U.K. Bandarage, M.E. Kuehne, C.M. Bertha, and W.D. Bowen, NIDA Research
Monograph 178, 235, (1998).
37
1. ibogaine: a review
169. W.D. Bowen, B.J. Vilner, W. Williams, U.K. Bandarage, and M.E. Kuehne, Soc. Neurosci.
Abstr. 23, 2319, #905.7 (1997).
170. J.W. Olney, U.S. Patent 5,629,307; Chem. Abstr. 127, 819q (1997).
171. X. Yu, S.Z. Imam, G.D. Newport, W. Slikker, Jr., and S.F. Ali, Brain Res. 823, 213 (1999).
172. S. Helsley, D. Fiorella, R.A. Rabin, and J.C. Winter, Pharmacol., Biochem. Behav. 58, 37
(1997).
173. G. Zetler, G. Singbartl, and L. Schlosser, Pharmacology 7, 237 (1972).
174. G. Singbartl, G. Zetler, and L. Schlosser, Neuropharmacology 12, 239 (1973).
175. D.C. Mash, K. Allen-Ferdinand, M. Mayor, C.A. Kovera, J.F. Ayafor, I.C. Williams, and F.R.
Ervin, NIDA Research Monograph 179, 294 (1999).
176. W. Baer, Forensic Subsequent Autopsy/Report Case # N-138 1991. University of Zurich,
Switzerland.
177. G. van Ingen and C.J. Meijer, Ned. Tijdsch. Geneeskd. 138, 767 (1994).
178. C. Court of Appeal at the Hague, Order of the Court. Cause List Number: 997179K09, 1999.
179. P. Popik, R.T. Layer, and P. Skolnick, Pharmacol. Rev. 47, 235 (1995).
180. P. Kintz, P. Mangin, A.A. Lugnier, and A.J. Chaumont, Human Toxicol. 8, 487 (1989).
181. T.E. Robinson and K.C. Berridge, Brain Res. Rev. 18, 247 (1993).
182. I. Wickelgren, Science 280, 2045 (1998).
183. S. Siegel, Addiction 94, 1113 (1999).
184. G. DiChiara, Eur. J. Pharmacol. 375, 13 (1999).
185. V. Deroche, M. Le Moal, and P.V. Piazza, Eur. J. Neurosci. 11, 2731 (1999).
186. N.M. White, Addiction 91, 921 (1996).
187. X. Nogues, Prog. Neuro-Psychopharmacol. Biol. Psychiat. 21, 507 (1997).
188. J.R. Blackburn and K.K. Szumlinski, Behav. Brain. Res. 89, 99 (1997).
189. R.P. Kesner, P.Jackson-Smith, C. Henry, and K. Amann, Pharmacol., Biochem. Behav. 51, 103
(1995).
190. P. Popik, Life Sci. 59, L379 (1996).
191. H. Depoortere, Neuropsychobiology 18, 160 (1987).
192. E. Hennevin, B. Hars, C. Maho, and V. Bloch, Behav. Brain. Res. 69, 125 (1995).
193. M. Jouvet, J. Sleep Res. 7, 1 (1998).
194. S.J. Cruikshank and N.M. Weinberger, J. Neurosci. 16, 861 (1996).
195. G.M. Frenken, Personal Communication, 2000.
196. E. Taub, Personal Communication, August 23, 2000
197. S.J. Blatt, B. Rounsaville, S.L. Eyre, and C. Wilber, J. Nerv. Ment. Dis. 172, 342 (1984).
198. C.J. Acker, J. Psychoactive Drugs 25, 193 (1993).
199. J.W. Fernandez, in Flesh of the Gods: The Ritual Use of Hallucinogens (P.T. Furst, ed.),
Waveland Press, Prospect Heights, IL, 237, 1990.
200. M. Galanter, Am. J. Psychiat. 147, 543 (1990).
201. J.W. Fernandez, Bwiti: An Ethnography of Religious Imagination in Africa, Princeton
University Press, Princeton, NJ, 1982.
202. K.L. Jansen, Med. Hypoth. 31, 25 (1990).
203. W.K. Schmidt, C.W. Gorodetzky, N.L. Teti, F. Vocci, S.A. Grossman, and R.S. Mansbach, in
Proceedings of the 60th Annual Meeting, The College of Problems on Drug Dependence, p.
20. U.S. Department of Health & Human Services, Bethesda, MD, 1999.
204. H. Grabowski, Pharmacoeconomics 11, 389 (1997).
205. C.R. Wright, Memorandum to Franck Vocci: Ibogaine. March 10, 1995.
38 kenneth r. alper
... Ibogaine is one of several alkaloids present in the iboga shrub (Tabernanthe iboga), a plant native to Central Africa that has been traditionally used for centuries in traditional medicine by people in countries like Gabon and Cameroon. Iboga is used by members of the Bwiti religion in initiation and religious rituals, usually by chewing or scraping the bark of its roots, and, at lower doses, its consumption is believed to have stimulating properties, used to mitigate sensations of fatigue, thirst, and hunger [24,25]. ...
... In 1957, Ciba Pharmaceutical (now Novartis) secured a patent for the utilization of ibogaine to reduce tolerance to opioid analgesics. In the late 1950s and during the 1960s, ibogaine was used as an auxiliary tool in psychotherapeutic processes [24,25]. Between the years of 1962 and 1963, a New York psychedelic enthusiast named Howard Lotsof first reported the anti-addictive properties of ibogaine on heroin dependence during recreational use [24][25][26][27]. ...
... In the late 1950s and during the 1960s, ibogaine was used as an auxiliary tool in psychotherapeutic processes [24,25]. Between the years of 1962 and 1963, a New York psychedelic enthusiast named Howard Lotsof first reported the anti-addictive properties of ibogaine on heroin dependence during recreational use [24][25][26][27]. Based on the possible efficacy of ibogaine in the treatment of drug addiction, the Medication Development Division of the National Institute on Drug Abuse (NIDA) in the United States supported preclinical and clinical research on ibogaine, and a phase I/II study with ibogaine on cocaine users was authorized by the Food and Drug Administration (FDA) between 1993 and 1995. ...
Article
Full-text available
As the research field with psychedelic substances grows, it is expected to encompass a more extensive cohort of individuals presenting a spectrum of medical conditions, comorbidities, and unique physiological traits, thereby increasing the likelihood of potential adverse events. Furthermore, it is worth noting that there is a scarcity of the specialized literature regarding procedures to ensure the safe management of clinical trials involving psychedelics. Acknowledging this, our research team designed a series of protocols to standardize the care and management of adverse scenarios, ensuring the safety and well-being of research volunteers included in clinical trials conducted by the LEAPS (Laboratory for Studies with Hallucinogens and Psychedelics in Mental Health, linked to the University of São Paulo). These guidelines have been meticulously crafted based on the established guideline philosophy of Hospital das Clínicas de Ribeirão Preto (the university hospital of Universidade de São Paulo), consultation with specialists in the field, and a thorough review of the existing literature. The process resulted in protocols that have been tailored to specifically address the unique requirements and particularities of clinical research with psychedelic substances (in this case, ayahuasca and ibogaine). As a result, these guidelines aim to cover a range of potential issues, encompassing both psychiatric manifestations (e.g., panic attacks, suicidal behavior, and psychotic episodes) and clinical manifestations (e.g., hypertensive crisis and hypoglycemia).
... Iboga is a shrub native from central Africa and its roots have been traditionally used for centuries by members of the Bwiti religion in countries such as Gabon and Cameroon. The roots are credited to have medicinal properties and are consumed by chewing low doses to combat fatigue, hunger and thirst and in high doses in the context of traditional spiritual ceremonies [1,2]. Ibogaine is the most abundant of the alkaloids present in the bush and is, to this date, the most studied. ...
... It was isolated for the first time in 1901 and has been researched as a possible treatment for asthenia. From 1939 to 1970 it was marketed in France under the name Lambarène ® as a neuromuscular stimulant to combat fatigue, depression, and infectious diseases, and in the late 1950s to 1960s, ibogaine has been used as a tool in psychotherapeutic processes, receiving a patent for this purpose in 1969 in France [1][2][3]. ...
... Running alongside this, in 1957, Ciba Pharmaceutical (now Novartis) patented the use of ibogaine to reduce tolerance to opioid analgesics. Between the years 1962 and 1963, its anti-addictive properties were discovered in an unpretentious way: Howard Lostof and other heroin users took ibogaine recreationally and noticed the next day that heroin-related withdrawal symptoms were not present [1][2][3][4]. This discovery had a great impact on investigations involving the alkaloid and since then, science has been trying to understand its anti-addictive properties. ...
Article
Full-text available
Ibogaine is a psychoactive alkaloid derived from the west-African shrub Tabernanthe iboga. Western cultures are increasing the interest for the substance due to its claimed anti addictive properties, although the evidence supporting this effect is still preliminary. The use of ibogaine often occurs with no medical supervision in uncontrolled settings, and its use has been associated with several reports of severe adverse events. This review aims to evaluate the clinical studies of ibogaine, with a focus on administration settings, to elucidate specific criteria that may promote safer contexts for ibogaine use. A systematic review of the literature was conducted based on PRISMA guidelines. PubMed, Scielo, ClinicalTrials.gov and Core.ac.uk electronic databases were searched, and clinical studies published until November 17, 2022, were retrieved. The final synthesis included 12 sources. Information about general characteristics of the studies, adverse effects, screening of participants and setting characteristics were summarized and discussed. It is concluded that the use of controlled settings, supported by trained professionals and equipment allowing for rigorous medical, psychiatric, and cardiac monitoring, are essential to promote the safety of patients receiving ibogaine.
... Ibogaine is an indole alkaloid often referred to as oneirogenic ("dream-generating") due to its complex effects on a range of neurotransmitter sites (4). It is most well-known as a putative therapeutic for mitigating withdrawal and cravings from opioids and other drugs (5), however it has also been studied experimentally for other conditions with a neurological basis (6). ...
Article
Full-text available
Brachial plexus nerve root avulsion results from complete separation of the nerve root from the spinal cord and is one of the most challenging types of neuropathic pain, coinciding with motor, sensory and autonomic deficits. The severe pain and typical impossibility of root reattachment often leads to requests for amputation. Ibogaine is an indole alkaloid producing psychoactive effects through reported actions upon multiple neurotransmitter systems, including NMDA, κ- and µ-opioid receptors and σ 2 receptor sites, along with stimulation of neurotrophic factors GDNF and BDNF. In this case report we describe a 53-year-old male with two decades of severe intractable pain due to brachial plexus nerve root avulsion from vehicular trauma who was successfully treated with both high dose inpatient and low dose outpatient administrations of ibogaine. Though promising for future study, the adverse effects of high dose ibogaine administrations may limit tolerability of this saturation protocol to the most refractory cases.
Article
Full-text available
Ibogaine and its main metabolite noribogaine provide important molecular prototypes for markedly different treatment of substance use disorders and co-morbid mental health illnesses. However, these compounds present a cardiac safety risk and a highly complex molecular mechanism. We introduce a class of iboga alkaloids – termed oxa-iboga – defined as benzofuran-containing iboga analogs and created via structural editing of the iboga skeleton. The oxa-iboga compounds lack the proarrhythmic adverse effects of ibogaine and noribogaine in primary human cardiomyocytes and show superior efficacy in animal models of opioid use disorder in male rats. They act as potent kappa opioid receptor agonists in vitro and in vivo, but exhibit atypical behavioral features compared to standard kappa opioid agonists. Oxa-noribogaine induces long-lasting suppression of morphine, heroin, and fentanyl intake after a single dose or a short treatment regimen, reversal of persistent opioid-induced hyperalgesia, and suppression of opioid drug seeking in rodent relapse models. As such, oxa-iboga compounds represent mechanistically distinct iboga analogs with therapeutic potential.
Article
Full-text available
Ibogaine is a psychedelic alkaloid being investigated as a possible treatment for opioid use disorder. Ibogaine has a multi-receptor profile with affinities for mu and kappa opioid as well as NMDA receptors amongst others. Due to the sparsity of research into ibogaine's effects on white matter integrity and given the growing evidence that opioid use disorder is characterized by white matter pathology, we set out to investigate ibogaine's effects on two markers of myelination, 2′, 3′-cyclic nucleotide 3′-phosphodiesterase (CNP) and myelin basic protein (MBP). Fifty Sprague Dawley rats were randomly assigned to five experimental groups of n = 10; (1) a saline control group received daily saline injections for 10 days, (2) a morphine control group received escalating morphine doses from 5 to 15 mg/kg over 10 days, (3) an ibogaine control group that received 10 days of saline followed by 50 mg/kg ibogaine hydrochloride, (4) a combination morphine and ibogaine group 1 that received the escalating morphine regime followed by 50 mg/kg ibogaine hydrochloride and (5) a second combination morphine and ibogaine group 2 which followed the same morphine and ibogaine regimen yet was terminated 72 h after administration compared to 24 h in the other groups. White matter from the internal capsule was dissected and qPCR and western blotting determined protein and gene expression of CNP and MBP. Morphine upregulated CNPase whereas ibogaine alone had no effect on CNP mRNA or protein expression. However, ibogaine administration following repeated morphine administration had an immediate effect by increasing CNP mRNA expression. This effect diminished after 72 h and resulted in a highly significant upregulation of CNPase protein at 72 h post administration. Ibogaine administration alone significantly upregulated protein expression yet downregulated MBP mRNA expression. Ibogaine administration following repeated morphine administration significantly upregulated MBP mRNA expression which increased at 72 h post administration resulting in a highly significant upregulation of MBP protein expression at 72 h post administration. These findings indicate that ibogaine is able to upregulate genes and proteins involved in the process of remyelination following opioid use and highlights an important mechanism of action of ibogaine's ability to treat substance use disorders.
Article
Full-text available
Mental health disorders and substance use disorders (SUDs) in particular, contribute greatly to the global burden of disease. Psychedelics, including entactogens and dissociative substances, are currently being explored for the treatment of SUDs, yet with less empirical clinical evidence than for other mental health disorders, such as depression or post-traumatic stress disorder (PTSD). In this narrative review, we discuss the current clinical research evidence, therapeutic potential and safety of psilocybin, lysergic acid diethylamide (LSD), ketamine, 3,4-methylenedioxymethamphetamine (MDMA) and ibogaine, particularly in the context of the SUD treatment. Our aim was to provide a balanced overview of the current research and findings on potential benefits and harms of psychedelics in clinical settings for SUD treatment. We highlight the need for more clinical research in this particular treatment area and point out some limitations and challenges to be addressed in future research.
Article
Full-text available
L-type calcium channels (LTCCs), the largest subfamily of voltage-gated calcium channels (VGCCs), are the main channels for Ca ²⁺ influx during extracellular excitation. LTCCs are widely present in excitable cells, especially cardiac and cardiovascular smooth muscle cells, and participate in various Ca ²⁺ -dependent processes. LTCCs have been considered as worthy drug target for cardiovascular, neurological and psychological diseases for decades. Natural products from Traditional Chinese medicine (TCM) have shown the potential as new drugs for the treatment of LTCCs related diseases. In this review, the basic structure, function of LTCCs, and the related human diseases caused by structural or functional abnormalities of LTCCs, and the natural LTCCs antagonist and their potential usages were summarized.
Article
Full-text available
Traumatic brain injury (TBI) is a leading cause of disability. Sequelae can include functional impairments and psychiatric syndromes such as post-traumatic stress disorder (PTSD), depression and anxiety. Special Operations Forces (SOF) veterans (SOVs) may be at an elevated risk for these complications, leading some to seek underexplored treatment alternatives such as the oneirogen ibogaine, a plant-derived compound known to interact with multiple neurotransmitter systems that has been studied primarily as a treatment for substance use disorders. Ibogaine has been associated with instances of fatal cardiac arrhythmia, but coadministration of magnesium may mitigate this concern. In the present study, we report a prospective observational study of the Magnesium–Ibogaine: the Stanford Traumatic Injury to the CNS protocol (MISTIC), provided together with complementary treatment modalities, in 30 male SOVs with predominantly mild TBI. We assessed changes in the World Health Organization Disability Assessment Schedule from baseline to immediately (primary outcome) and 1 month (secondary outcome) after treatment. Additional secondary outcomes included changes in PTSD (Clinician-Administered PTSD Scale for DSM-5), depression (Montgomery–Åsberg Depression Rating Scale) and anxiety (Hamilton Anxiety Rating Scale). MISTIC resulted in significant improvements in functioning both immediately (Pcorrected < 0.001, Cohen’s d = 0.74) and 1 month (Pcorrected < 0.001, d = 2.20) after treatment and in PTSD (Pcorrected < 0.001, d = 2.54), depression (Pcorrected < 0.001, d = 2.80) and anxiety (Pcorrected < 0.001, d = 2.13) at 1 month after treatment. There were no unexpected or serious adverse events. Controlled clinical trials to assess safety and efficacy are needed to validate these initial open-label findings. ClinicalTrials.gov registration: NCT04313712.
Article
The iboga alkaloids scaffold shows great potential as a pharmacophore in drug candidates for the treatment of neuropsychiatric disorders. Thus, the study of the reactivity of this type of motif is particularly useful for the generation of new analogs suitable for medicinal chemistry goals. In this article, we analyzed the oxidation pattern of ibogaine and voacangine using dioxygen, peroxo compounds, and iodine as oxidizing agents. Special focus was placed on the study of the regio- and stereochemistry of the oxidation processes according to the oxidative agent and starting material. We found that the C16-carboxymethyl ester present in voacangine stabilizes the whole molecule toward oxidation in comparison to ibogaine, especially in the indole ring, where 7-hydroxy- or 7-peroxy-indolenines can be obtained as oxidation products. Nevertheless, the ester moiety enhances the reactivity of the isoquinuclidinic nitrogen to afford C3-oxidized products through a regioselective iminium formation. This differential reactivity between ibogaine and voacangine was rationalized using computational DFT calculations. In addition, using qualitative and quantitative NMR experiments combined with theoretical calculations, the absolute stereochemistry at C7 in the 7-hydroxyindolenine of voacangine was revised to be S, which corrects previous reports proposing an R configuration.
Article
Introduction: Ibogaine is one of the alkaloids naturally found in plants such as Tabernanthe iboga, which has been traditionally used by members of the Bwiti culture. Since the discovery of its anti-addictive properties by Howard S. Lotsof in 1962, ibogaine has been used experimentally to treat substance use disorders (SUD), especially those involving opioids. We aim to provide a detailed understanding of the underlying psychological aspects of underground ibogaine use for the treatment of SUD. Methods: Semi-structured interviews were carried out with 13 participants with SUD, which motivated their self-treatment with ibogaine. The data were analysed using the grounded theory approach and considered the context of the treatment, and the nature of the occurring hallucinogenic and cognitive phenomena during the treatment experience. Results: We identified several psychological effects that the study respondents experienced , which seem to play a substantial role in the therapeutic process concerning SUD. The evoking of interpersonal and transpersonal experiences, autobiographical memories, and preparation, integration and motivation for a lifestyle change are important components that participants reported during and after ibogaine intake. Discussion and Conclusion: Ibogaine is increasingly being used for the treatment of SUD, due in part to the limited treatment options currently available. Its beneficial effects seem to be related not only to its complex pharmacology but also to the subjective experience that ibogaine induces. The main aspects of this experience are related to autobiographical memories and valuable personal insights, which together appear to help individuals cope with their SUD. K E Y W O R D S hallucinogenic, iboga, psychedelic, qualitative analysis, substance use disorders
Article
This paper presents a biopsychological theory of drug addiction, the 'Incentive-Sensitization Theory'. The theory addresses three fundamental questions. The first is: why do addicts crave drugs? That is, what is the psychological and neurobiological basis of drug craving? The second is: why does drug craving persist even after long periods of abstinence? The third is whether 'wanting' drugs (drug craving) is attributable to 'liking' drugs (to the subjective pleasurable effects of drugs)? The theory posits the following. (1) Addictive drugs share the ability to enhance mesotelencephalic dopamine neurotransmission. (2) One psychological function of this neural system is to attribute 'incentive salience' to the perception and mental representation of events associated with activation of the system. Incentive salience is a psychological process that transforms the perception of stimuli, imbuing them with salience, making them attractive, 'wanted', incentive stimuli. (3) In some individuals the repeated use of addictive drugs produces incremental neuroadaptations in this neural system, rendering it increasingly and perhaps permanently, hypersensitive ('sensitized') to drugs and drug-associated stimuli. The sensitization of dopamine systems is gated by associative learning, which causes excessive incentive salience to be attributed to the act of drug taking and to stimuli associated with drug taking. It is specifically the sensitization of incentive salience, therefore, that transforms ordinary 'wanting' into excessive drug craving. (4) It is further proposed that sensitization of the neural systems responsible for incentive salience ('for wanting') can occur independently of changes in neural systems that mediate the subjective pleasurable effects of drugs (drug 'liking') and of neural systems that mediate withdrawal. Thus, sensitization of incentive salience can produce addictive behavior (compulsive drug seeking and drug taking) even if the expectation of drug pleasure or the aversive properties of withdrawal are diminished and even in the face of strong disincentives, including the loss of reputation, job, home and family. We review evidence for this view of addiction and discuss its implications for understanding the psychology and neurobiology of addiction.