ArticlePDF Available

The antiaddictive effects of ibogaine: A systematic literature review of human studies

Akadémiai Kiadó
Journal of Psychedelic Studies
Authors:
  • Ribeirão Preto Medical School, University of São Paulo, Brazil

Abstract and Figures

Background and aims Ibogaine is a naturally occurring hallucinogenic alkaloid with a therapeutic potential for reducing drug craving and withdrawal. To the best of our knowledge, no systematic review was previously performed assessing these effects. Thus, we conducted a systematic literature review of human studies assessing the antiaddictive effects of ibogaine. Methods Papers published up to July 2, 2016 were included from PubMed, LILACS, and SciELO databases following a comprehensive search strategy and a pre-determined set of criteria for article selection. Results Two hundred and fifty-nine studies were identified, of which eight met the established criteria. Seven studies were open-label case series with ibogaine and one study was a randomized, placebo-controlled clinical trial with noribogaine. Case series suggest that a single dose or a few treatments with ibogaine may significantly reduce drug withdrawal, craving, and self-administration in dependent individuals lasting from 24 h to weeks or months. No significant effects of noribogaine on opiate/opioid withdrawal were observed in the clinical trial. Conclusions Considering the necessity of new drugs that may produce fast-acting and sustained effects in opiate/opioid and cocaine dependence, the potential beneficial effects of ibogaine/noribogaine should be further investigated in controlled trials.
Content may be subject to copyright.
The antiaddictive effects of ibogaine: A systematic literature
review of human studies
RAFAEL G. DOS SANTOS
1,2,3
*, JOSÉ CARLOS BOUSO
2
and JAIME E. C. HALLAK
1,3
1
Department of Neurosciences and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
2
International Center for Ethnobotanical Education, Research and Service, Barcelona, Spain
3
National Institute of Science and Technology Translational Medicine, Ribeirão Preto, Brazil
(Received: August 22, 2016; revised manuscript received: October 15, 2016; accepted: October 19, 2016)
Background and aims: Ibogaine is a naturally occurring hallucinogenic alkaloid with a therapeutic potential for
reducing drug craving and withdrawal. To the best of our knowledge, no systematic review was previously performed
assessing these effects. Thus, we conducted a systematic literature review of human studies assessing the
antiaddictive effects of ibogaine. Methods: Papers published up to July 2, 2016 were included from PubMed,
LILACS, and SciELO databases following a comprehensive search strategy and a pre-determined set of criteria for
article selection. Results: Two hundred and fty-nine studies were identied, of which eight met the established
criteria. Seven studies were open-label case series with ibogaine and one study was a randomized, placebo-controlled
clinical trial with noribogaine. Case series suggest that a single dose or a few treatments with ibogaine may
signicantly reduce drug withdrawal, craving, and self-administration in dependent individuals lasting from 24 h to
weeks or months. No signicant effects of noribogaine on opiate/opioid withdrawal were observed in the clinical trial.
Conclusions: Considering the necessity of new drugs that may produce fast-acting and sustained effects in opiate/
opioid and cocaine dependence, the potential benecial effects of ibogaine/noribogaine should be further investigated
in controlled trials.
Keywords: ibogaine, noribogaine, substance use disorders, dependence, withdrawal
INTRODUCTION
Ibogaine is a naturally occurring indole alkaloid derived
from the root barks of Tabernanthe iboga, a plant native to
western Central African countries such as Gabon and
Cameroon, where it has been used traditionally by the
Pygmies and other African ethnic groups for several centu-
ries, and more recently, as a sacrament in initiatory rites and
for social cohesion in the Bwiti religion (Alper, 2001;Alper,
Lotsof, & Kaplan, 2008;Brown, 2013;Mash et al., 1998).
Although T. iboga contains several other alkaloids, ibogaine
is considered to be the main psychoactive substance present
in this plant. The mechanism of action of ibogaine and its
O-demethylated active metabolite noribogaine is poorly
understood and considered to involve antagonism of the
N-methyl-D-aspartate (NMDA) glutamate receptor and
α3β4 nicotinic acetylcholine receptor, inhibition of the
serotonin (5-HT) reuptake transporter and dopamine release,
agonism of the σ
2
receptor, κ- and μ-opioid receptors, M
1/2
muscarinic receptor, and 5-HT
2A
receptors [similar to classic
hallucinogens such as lysergic acid diethylamide (LSD),
psilocybin, and dimethyltryptamine (DMT)], and enhance-
ment of glial cell line-derived neurotrophic factor (GDNF)
(Alper, 2001;Alper et al., 2008;Brown, 2013;Donnelly,
2011;Lotsof & Alexander, 2001;Mash et al., 1998).
In the 1960s, Lotsof et al. showed that ibogaine could
reduce heroin withdrawal (Alper, 2001;Alper et al., 2008;
Brown, 2013;Donnelly, 2011;Lotsof & Alexander, 2001;
Mash et al., 1998). On the basis of the initial observations
and several treatments provided in non-medical contexts,
Lotsof et al. proposed ibogaine as a new method to treat
dependence to opiates/opioids (morphine, heroin, and meth-
adone), stimulants (nicotine, cocaine, amphetamine, and
methamphetamine), and ethanol and acquired several
patents for such uses (United States patents: US 4499096
1985, 4587243 1986, 4857523 1989, 5026697 1991, and
5152995 1992). It has been estimated that more than 3,400
people have been treated with ibogaine for drug dependence
in clinics around the world, mainly in countries such as The
Netherlands, the United States, Mexico, and Brazil (Alper,
2001;Alper et al., 2008;Brown, 2013;Donnelly, 2011).
In the context of drug dependence treatment, ibogaine is
usually ingested orally in the form of extracts/hydrochloride
(HCl) in doses ranging from 4 to 25 mg/kg (Alper, 2001;
Alper et al., 2008;Brown, 2013;Donnelly, 2011;Forsyth
et al., 2016;Glue, Lockhart, et al., 2015;Glue, Winter, et al.,
2015;Lotsof & Alexander, 2001;Mash et al., 1998). About
1 hr after oral administration, subjects experience decreased
muscular coordination, increased sensitivity to light and
sound, nausea and vomiting if they move, and visual effects
that the ibogaine supporters refer to them as oneirogenics
* Corresponding author: Rafael G. dos Santos, PhD; Departamento
de Neurociências e Ciências do Comportamento, Faculdade de
Medicina de Ribeirão Preto, Universidade de São Paulo, Hospital
das Clínicas, Terceiro Andar, Av. Bandeirantes, 3900, Ribeirão
Preto, São Paulo, Brazil; Phone/Fax: +55 16 3602 2703; E-mail:
banisteria@gmail.com
© 2016 The Author(s)
Journal of Psychedelic Studies 1(1), pp. 2028 (2017)
DOI: 10.1556/2054.01.2016.001
First published online December 19, 2016
(similar to dreams). These visual effects are sustained for
around 48 hr and are followed by a contemplative state of
1224 hr in which lucid dreaming may occur accompanied
by the emergence of autobiographical memories. Insomnia
and increased energy may be present for 72 hr following
ibogaine intake (Alper, 2001;Alper et al., 2008;Brown,
2013;Donnelly, 2011;Forsyth et al., 2016;Glue, Lockhart,
et al., 2015;Glue, Winter, et al., 2015;Lotsof & Alexander,
2001;Mash et al., 1998). Lower oral doses of ibogaine
(20 mg) and noribogaine (360 mg) were administered to
healthy male volunteers in recent open-label (ibogaine)
and double-blind, placebo-controlled (noribogaine) studies,
and both drugs were found to be safe and well tolerated
(Forsyth et al., 2016;Glue, Lockhart, et al., 2015;Glue,
Winter, et al., 2015). At this dose levels, subjects did not
experience the above-mentioned psychoactive effects,
reporting basically transient nausea, gastrointestinal symp-
toms, and dizziness.
Ibogaine administration has been associated with several
fatalities (>25 cases), which appear to involve increases in
cardiac arrhythmias, previous cardiovascular diseases, and
use of opiates/opioids or other drugs during the acute effects
of ibogaine (Alper, 2001;Alper, Staji´c, & Gill, 2012;
Brown, 2013;Koenig & Hilber, 2015;Litjens & Brunt,
2016;Meisner, Wilcox, & Richards, 2016). Ibogaine intake
has also been associated with psychosis (Houenou, Homri,
Leboyer, & Drancourt, 2011), mania (Marta, Ryan,
Kopelowicz, & Koek, 2015), and seizures (Breuer et al.,
2015). However, most of these cases happened in uncon-
trolled/non-medical settings, using unknown doses of
ibogaine of variable purity. When administered in more
controlled/supervised contexts, to individuals without pre-
vious cardiovascular diseases or under the acute effects of
drugs, ibogaine appears to be relatively safe (Alper, 2001;
Alper et al., 2008,2012;Brown, 2013;Donnelly, 2011;
Forsyth et al., 2016;Glue, Lockhart, et al., 2015;Glue,
Winter, et al., 2015;Koenig & Hilber, 2015;Lotsof &
Alexander, 2001;Mash et al., 1998;Meisner et al., 2016).
However, sudden deaths and fatalities with unknown causes
have also been reported, and ibogaine should be adminis-
tered only after a complete medical screening and with a
rigorous cardiovascular monitoring (Alper, 2001;Alper et al.,
2012;Brown, 2013;Koenig & Hilber, 2015;Litjens &
Brunt, 2016;Meisner et al., 2016).
Animal studies showed that ibogaine is neither reinfor-
cing nor aversive, and that this alkaloid reduces opiate/
opioid (morphine and heroin), cocaine, and ethanol self-
administration (Alper, 2001;Alper et al., 2008;Belgers
et al., 2016;Brown, 2013;Donnelly, 2011;Frenken, 2001;
Lotsof & Alexander, 2001;Mash et al., 1998). Animal
studies also reported that noribogaine reduces nicotine and
amphetamine self-administration (Alper, 2001;Alper et al.,
2008;Belgers et al., 2016;Brown, 2013;Donnelly, 2011;
Frenken, 2001;Lotsof & Alexander, 2001;Mash et al.,
1998). A recent meta-analysis of animal studies reported
that the most signicant effects of ibogaine in reducing drug
self-administration were observed in the rst 24 hr after its
administration, and these effects were sustained for more
than 72 hr (Belgers et al., 2016). Moreover, several case
reports described signicant reductions in drug (mostly
heroin, methadone, and cocaine) craving and withdrawal
symptoms within 12 hr after oral administration of single
or few doses of ibogaine, followed by complete cessation of
the opiate/opioid withdrawal syndrome within 2448 hr
and signicant reductions or even total cessation of
substance use weeks to months (or longer) following
ibogaine intake (Alper, 2001;Alper et al., 2008;
Brown, 2013;Donnelly, 2011;Frenken, 2001;Lotsof &
Alexander, 2001;Mash et al., 1998). These data from
human case reports are in line with animal studies (Belgers
et al., 2016).
Although some reviews analyzing the antiaddictive
effects of ibogaine in humans were published, these were
narrative and non-systematic reviews (Alper, 2001;Alper
et al., 2008;Brown, 2013;Donnelly, 2011;Mash et al.,
1998), and the most recent of them was published 3 years
ago (Brown, 2013). To the best of our knowledge, no
systematic review analyzing the antiaddictive effects of
ibogaine in humans was previously performed. Therefore,
considering the apparent increase in ibogaine use and its
possible toxic and therapeutic effects (Alper et al., 2008),
this study aimed to conduct a systematic literature review of
human studies that investigated the antiaddictive effects of
ibogaine or of its main active metabolite, noribogaine.
METHODS
Data for this systematic review were collected in accor-
dance with the Systematic Reviews and Meta-Analyses
guidelines (Moher, Liberati, Tetzlaff, Altman, & The
PRISMA Group, 2009).
Data acquisition
We attempted to identify all human studies available to
review up to July 2, 2016 in which the antiaddictive effect of
ibogaine or noribogaine was analyzed.
Search strategy
Electronic searches were performed using PubMed, LILACS,
and SciELO databases. The following keywords were used:
ibogaine OR noribogaine AND humans OR addiction OR
dependence. References were retrieved through searching
electronic databases and manual searches through reference
lists of identied literature. All the studies published up to
July 2, 2016 were included without any language restriction.
Eligibility criteria
The following inclusion and exclusion criteria were estab-
lished prior to the literature search.
Article type
Case reports and clinical studies published in peer-reviewed
journals were included. Books and book chapters indexed in
the above-cited databases were also included. Preclinical
studies (in vitro and in vivo), reviews, abstracts and letters,
comments, and editorials were excluded.
Journal of Psychedelic Studies 1(1), pp. 2028 (2017) |21
Antiaddictive effects of ibogaine
Study design
The review included case reports and clinical studies of
ibogaine administration that assessed dependence/abuse
symptoms.
Participants/sample
Subjects with a diagnosis of drug abuse/dependence were
included.
Interventions
All designs evaluating the effect of ibogaine on abuse/
dependence measures were included.
Comparisons
The main comparator considered was pre-treatment abuse/
dependence symptoms.
Outcomes
Studies investigating the effect of ibogaine on abuse/
dependence symptoms were included.
Data extraction
All studies were screened by two independent reviewers
with discrepancies resolved by a third reviewer. From the
articles included, we recorded the names of authors, year of
publication, study location (city and country), study design
(open label or controlled), characteristics of the participants
(sample size, age, and gender), response criteria (antiaddic-
tive effect), type of intervention (dose), and type of outcome
measure (abuse/dependence symptoms).
RESULTS
Study selection
Aow diagram illustrating the different phases of the
systematic review is presented in Figure 1.
The literature search yielded 259 separate references, all
from PubMed. These citations were reviewed for abstract
screening, and seven potentially relevant references were
identied (Alper, 2001;Alper, Lotsof, Frenken, Luciano, &
Bastiaans, 1999;Luciano, 1998;Mash et al., 2000,2001;
Schenberg, de Castro Comis, Chaves, & Da Silveira, 2014;
Sheppard, 1994). Full-text reports of these citations were
obtained for more detailed evaluation. One more citation
(Glue, Cape, Tunnicliff, Lockhart, Lam, Hung, et al., 2016)
was found after handsearching the bibliography of one of
the references used in the Introduction section (Forsyth
et al., 2016). This citation was not indexed at any of the
databases used at the time of preparation of the manuscript,
and the paper was obtained directly from the authors.
Following detailed examination of the reports, all citations
were included. The studies included comprised seven case
series involving the administration of ibogaine (Alper, 2001;
Alper et al., 1999;Luciano, 1998;Mash et al., 2000,2001;
Schenberg et al., 2014;Sheppard, 1994) and one random-
ized, placebo-controlled clinical trial with noribogaine
(Glue, Cape, Tunnicliff, Lockhart, Lam, Hung, et al.,
2016). Table 1shows the main information of each study.
Case series
In an open-label study performed in non-medical settings in
Amsterdam, The Netherlands, seven opiate-dependent indi-
viduals (ve men; mean age 29.28 years; six individuals
were dependent on heroin, two on ethanol, and one on
codeine) were treated with single oral doses of 7001800 mg
(11.725.0 mg/kg) ibogaine HCl (Sheppard, 1994). Treat-
ments occurred between October 1989 and August 1990,
and the researchers documented the immediate and long-
term (up to 14 weeks) effects of ibogaine. Subjects were
administered a trial dose of 100200 mg ibogaine, followed
12 hr later by the remainder of the dose. None of the
subjects showed signicant opiate withdrawal symptoms
2438 hr after ibogaine administration. Two subjects who
received the lowest dose (700 mg) relapsed to opiate usage
within 2 days after ibogaine administration, two subjects
who received >1,000 mg relapsed after a number of weeks,
one subject who received >1,000 mg reverted to intermittent
heroin use, and three subjects who received >1,000 mg
remained drug-free for >14 weeks after treatment. Ibogaine
administration was also associated with increased energy,
appetite, and reduced sleep for several weeks after drug
intake. Ibogaine was well tolerated, and adverse effects
reported included sensitivity to light and sound, ataxia,
diarrhea, and nausea and vomiting. Interestingly, ibogaine
Figure 1. Flow diagram illustrating the different phases of the
systematic review
22 |Journal of Psychedelic Studies 1(1), pp. 2028 (2017)
dos Santos et al.
Table 1. Open-label case series and clinical trials describing the antiaddictive effects of ibogaine
References Design/setting Subjects Dose Main results Safety
Sheppard
(1994)
a
Open-label 7 (heroin, methadone,
codeine, and ethanol)
5 men, mean age
29.28 years
Single doses
11.725 mg/kg
Absence of withdrawal symptoms after 2438 hr for
all subjects, two relapsed within 48 hr, two after
several weeks, one reverted to intermittent heroin
use, and three remained drug-free >14 weeks
No medical screening or monitoring
Non-medical No serious adverse reactions
Amsterdam, The Netherland
Luciano (1998) Open-label 3 (cocaine, opiates/
opioids, and ethanol)
Gender, age (?)
Single doses
2025 mg/kg
Absence of withdrawal and craving symptoms after
24 hr for all subjects
Medical screening and monitoring
No serious adverse reactions
Setting (?) Abstinence (?)
Alper et al.
(1999)
a
Open-label 33 (heroin. methadone,
and cocaine)
22 men, mean age
27.3 years
Single doses
629 mg/kg
Absence of withdrawal symptoms and drug use for
25 (76%) subjects after 72 hr, four did not report
symptoms but used drugs in <72 hr, two reported
attenuated symptoms and no drug use in <72 hr, and
one reported absence of effects
Medical screening and monitoring
one fatality, probably caused by
concomitant heroin use
Non-medical
United States and The
Netherlands
Mash et al.
(2000)
b
Open-label 27 (heroin, methadone [?]
and cocaine)
23 men, mean age 34.6
(opiates/opioids) and
37.5 (cocaine) years
Single doses
500, 600, and
800 mg
Signicant (p<.005) reductions in all HCQN-29
subscales
c
and in two CCQN-45 subscales
d
after
36 hr and 14 days, and signicant (p<.0005)
reductions in BDI scores after one month
Abstinence (?)
Medical screening and monitoring
Private clinic
St. Kitts, West Indies
No serious adverse reactions
Mash et al.
(2001)
b
Open-label 32 (heroin and methadone)
23 men, mean age
33.6 years
Single dose
800 mg
Signicant (p<.05) reductions in OOWS scores after
1224 hr and in OP-SCL scores after <72 hr and 69
days, and months of abstinence in many subjects
(although data were not presented)
Medical screening and monitoring
Private clinic
St. Kitts, West Indies
No serious adverse reactions
Alper (2001)
a
Open-label 41 (heroin. methadone,
cocaine, sedatives,
ethanol)
Gender, age (?)
Single doses
629 mg/kg
15 (29%) subjects reported abstinence for <2 months,
15 (29%) for 2 months/<6 months, 7 (13%) for
6 months/<1 year, 10 (19%) for >1 year, and
5 (10%) the outcomes could not be determined
Medical screening and monitoring
Non-medical No serious adverse reactions
United States and The
Netherlands
Schenberg
et al. (2014)
Open-label 75 (ethanol, cannabis,
cocaine, and crack-
cocaine)
67 men and
8 women, mean age
34.16 (men) and
29.50 (women) years
Single and multiple
doses; mean
number of
sessions 3.83
(men) and
5.40 (women)
1720 mg/kg
Signicant (p<.001) increases in abstinence
duration, and 61% of subjects were abstinent after
single (median 5.5 months) and multiple (median
8.4 months) doses
Medical screening and monitoring
Private hospital No serious adverse reactions
Santa Cruz do Rio Pardo, Brazil
Glue, Cape,
Tunnicliff,
Lockhart,
Lam, Gray,
et al. (2016)
Randomized, double-blind,
placebo-controlled, single
ascending-dose clinical trial
Research unit
27 (methadone)
21 men, mean age
41.2 years
Single doses
(noribogaine)
60, 120, and
180 mg
Non-signicant effects on SOWS, OOWS, COWS,
and time to resumption of MST
Medical screening and monitoring
No serious adverse reactions
Signicant QT interval
prolongation
Dunedin, New Zealand
Note. BDI, Beck Depression Inventory; CCQN-45, Cocaine Craving Questionnaire; COWS, Clinical Opioid Withdrawal Scale; HCQN-29, Heroin Craving Questionnaire; OOWS, Objective Opiate
Withdrawal Scale; OP-SCL, Opiate-Symptom Checklist; MST, methadone substitution treatment; SOWS, Subjective Opioid Withdrawal Scale. Interrogation: not informed.
a
Included subjects from the same sample.
b
Included subjects from the same sample.
c
Desire to Use, Intention to Use, Anticipation of Positive Outcomes, Relief of Negative States, and Lack of Control.
d
Relief of Negative States and Lack of Control.
Journal of Psychedelic Studies 1(1), pp. 2028 (2017) |23
Antiaddictive effects of ibogaine
administration was not associated with a reduction in can-
nabis use. Importantly, there is no control of drug use prior,
during, or after treatment was performed, and some subjects
used drugs (mostly heroin) in the days immediately before/
after treatment. Moreover, no medical or psychiatric screen-
ing was described in the report. Careful analysis of this
report, other citations, and handsearching the bibliography
of the references showed that this study described a full
sample of previous treatments published in non-scientic
reports (Frenken, 2001).
Areportfromanunidentied city/country (probably
Amsterdam, The Netherlands) and apparently of an open-
label design reported three cases of subjects treated with
ibogaine HCl (2025 mg/kg) for cocaine dependence (one
subject was also dependent on heroin and two were also
dependent on ethanol) (Luciano, 1998). No information
regarding age or gender of the participants was given in the
report. Subjects were included after medical and psychiatric
screening, laboratory exams, electrocardiogram (ECG),
electroencephalogram (EEG), and magnetic resonance.
Medical monitoring was continuous, and neurologic/EEG
assessments were performed intermittently over 24 hr. All
subjects showed transient cerebellar dysfunction (nystagmus,
tremor, and ataxia) 2 hr after ibogaine intake, and these
symptoms improved 8 hr after drug administration. Visual
alterations with eyes closed were observed in only one
subject, reality testing remained normal in all of them, and
no anxiety symptoms or thought disorder were observed.
EEG assessments were normal in all participants during and
after treatment, and no medical or ECG abnormalities were
observed during the treatment. Twenty-four hours after
ibogaine administration, all subjects reported an absence of
subjective or objective signs of withdrawal or craving, and all
neurologic/EEG examinations were normal.
A larger retrospective report described a group of
33 subjects (22 men, mean age 27.3 ±4.7 years) that were
treated with ibogaine HCl (average dose: 19.3 ±6.9 mg/kg,
range 629 mg/kg) for opiate/opioid dependence (mostly
heroin, primarily by the i.v. route; all met DSM-IV criteria
for opiate/opioid dependence) in non-medical settings under
open-label conditions between 1962 and 1993 (Alper et al.,
1999). Eight subjects were also using methadone and eight
were using cocaine on a daily basis. Seven treatments were
carried out in the United States between 1962 and 1963, and
the other 26 treatments were carried out in The Netherlands
between 1989 and 1993 [including patients from a previous
report (Sheppard, 1994)]. Treatments occurred 810 hr after
the subjects made their last use of heroin (24 hr in the case of
the subjects using methadone), and they were monitored for
72 hr. Twenty-four hours after ibogaine administration,
25 subjects (76%) reported complete resolution of opiate/
opioid withdrawal without drug-seeking behavior, and these
effects were sustained for 72 hr. Four subjects did not report
withdrawal signs but used opiates/opioids within 72 hr after
ibogaine administration, and the other two subjects reported
attenuated signs of withdrawal but remained drug-free. Only
one participant reported an absence of effect of ibogaine on
withdrawal symptoms. But the report does not specify
whether the subjects with less effect of ibogaine on with-
drawal symptoms were the methadone users or not. One
fatality was reported: a 24-year-old female treated in The
Netherlands, in 1993, died 19 hr after ibogaine administra-
tion. Apparently, this fatality was due to heroin use in the
rst few hours after treatment.
In a conventional research setting (clinic) in St. Kitts,
West Indies, more than 150 drug-dependent subjects were
treated with single doses of 500800 mg ibogaine HCl
under open-label conditions (Mash et al., 2000,2001).
Baseline screening included physical and psychiatric exam-
inations, ECG, and blood/hematological tests, and adverse
effects were assessed by clinician ratings and self-reports.
Ibogaine was safely administered based on cardiorespiratory
measures, blood cell tests, and measures of hepatic
enzymes. The most frequent adverse effects were transient
nausea and mild tremor during the acute effects of ibogaine.
A subset of 27 opiate/opioid- and cocaine-dependent
(DSM-IV criteria) subjects (23 men, mean age 34.6 ±1.9
years for the opiate/opioid group and 37.5 ±2.9 years for
the cocaine group) participated in a 2-week inpatient Phase I
dose-escalation study to assess ibogaine safety and efcacy
for treating drug dependence symptoms (Mash et al., 2000).
Subjects were randomly assigned to receive single xed
doses of 500, 600, or 800 mg ibogaine HCl under open-label
conditions and completed structured self-reports relating to
depression symptoms (Beck Depression Inventory, BDI)
and craving [heroin (HCQN-29) or cocaine (Craving Ques-
tionnaire, CCQN-45); 1 hr before drug intake, and 36 hr and
14 days after drug intake]. The BDI was also applied
1 month after ibogaine administration. Ibogaine administra-
tion was associated with signicant reductions in all ve
subscales of the HCQN-29 (Desire to Use,Intention to Use,
Anticipation of Positive Outcomes,Relief of Negative States,
and Lack of Control; for all subscales p<.0001) 36 hr and
14 days after drug intake, suggesting decreased craving for
opiates/opioids. Ibogaine intake was also associated with
signicant reductions in two subscales of the CCQN-45
(Relief of Negative States,p<.0005; Lack of Control,
p<.002) 36 hr and 14 days after drug administration,
suggesting decreased craving for cocaine. Ibogaine admin-
istration was also associated with signicant reductions in
BDI scores (p<.0005) 1 month after drug intake, suggest-
ing sustained reductions in depressive symptoms. Again, the
study did not clarify if (or which) the subjects were under
methadone treatment and if this interfered somehow with
the results.
Another subset of 32 patients from the original sample
(23 men, mean age 33.6 years), all opiate/opioid dependent
(heroin or methadone, DSM-IV criteria), received a single
800 mg of dose, and withdrawal and craving symptoms were
assessed with the physician-rated Objective Opiate With-
drawal Scale (OOWS) and with the Opiate-Symptom Check-
list (OP-SCL, self-rated) (Mash et al., 2001). Data were
collected 1 hr before drug intake (12 hr after last opiate/
opioid dose) and 12 and 24 hr after drug intake (24 and 36 hr
after last opiate/opioid dose, respectively). The OP-SCL was
also used 69 days after ibogaine intake. Compared with
baseline values, OOWS scores were signicantly (p<.05)
reduced 12 and 24 hr after ibogaine administration, and
OP-SCL scores were signicantly (p<.05) decreased
<72 hr and 69 days after drug intake. The only informa-
tion provided regarding drug abstinence stated vaguely
that many subjects :::were able to maintain abstinence
24 |Journal of Psychedelic Studies 1(1), pp. 2028 (2017)
dos Santos et al.
from illicit opiates and methadone over the months follow-
ing detoxication (data not shown).Again, authors did not
differentiate subjects regarding previous methadone use.
A retrospective report assessed the long-term effects of
ibogaine in 41 individuals treated in non-medical settings
under open-label conditions between 1962 and 1993, in the
United States and in The Netherlands, mainly for opiate/
opioid or stimulant dependence (Alper, 2001), including
patients from previous studies (Alper et al., 1999;Sheppard,
1994). Fifteen (29%) reported abstinence of less than
2 months, 15 (29%) for at least 2 months and less than
6 months, seven (13%) for at least 6 months and less
than 1 year, and 10 (19%) for more than 1 year. In ve
cases (10%), the outcomes could not be determined.
Again, authors failed to identify subjects under methadone
treatment.
In another retrospective study conducted in a private
hospital in Santa Cruz do Rio Pardo, Brazil, the safety and
efcacy of an open-label ibogaine treatment combining the
administration of the drug with cognitive behavioral therapy
were analyzed using data from 75 drug-dependent (DSM-IV
criteria) patients (67 males and 8 females; mean ages 34.16
±8.33 and 29.50 ±5.31 years, respectively) (Schenberg
et al., 2014). Seventy-two percent of the subjects were
Brazilian polydrug users (ethanol, cannabis, cocaine, and
crack cocaine), with only one European patient having a
history of opiate/opioid use. Subjects underwent a total of
134 ibogaine sessions (mean number of sessions for male
and female participants was 3.83 ±3.31 and 5.40 ±0.91,
respectively). Patients were required to stay abstinent for
3060 days prior to ibogaine administration at their homes,
or for at least 24 hr as a residential patient at the clinic.
Subjects received oral doses of 1720 mg/kg ibogaine HCl,
preceded 3045 min before by 20 mg of the dopamine D
2/3
receptor antagonist domperidone, to reduce nausea. If a
patient reported a weak response to ibogaine after the initial
dose, an additional 100200 mg of dose was administered.
Blood pressure, cardiac frequency, and oxygen saturation
were measured every 2530 min for 10 hr, and subjects
were dismissed after 2448 hr, returning for psychological
therapy/follow-up. Single and multiple doses of ibogaine
were associated with signicant (p<.001) increases in
abstinence duration, and 61% of participants were abstinent
after ibogaine treatment (single dose, median 5.5 months;
multiple doses, median 8.4 months). Acute adverse effects
included transient nausea, ataxia, vomiting, tremors, head-
aches, and mental confusion. No serious adverse reactions
such as cardiac arrhythmias or fatalities were observed.
Clinical trials
In a randomized, double-blind, placebo-controlled, single
ascending-dose study, 27 patients (21 men, mean age 41.2
years) on methadone substitution therapy received noribo-
gaine doses of 60, 120, or 180 mg (Glue, Cape, Tunnicliff,
Lockhart, Lam, Hung, et al., 2016). Subjects were selected
based on medical history, physical examination, safety
laboratory tests, vital signs, ECG, and switched to morphine
treatment a week before study participation. The effects of
noribogaine on withdrawal symptoms and as a possible
agonist of μ-opioid receptors were assessed by pupillometry
(baseline to 144 hr later), and oximetry and capnography
(baseline to 72 hr later). Withdrawal symptoms were also
analyzed by measuring the time to resumption of methadone
treatment (mean time between last morphine dose given 2 hr
prior to treatment and time to resumption of methadone
treatment) and by the Subjective, Objective, and Clinical
Opioid Withdrawal Scales (SOWS, OOWS, and COWS;
baseline to 144 hr later, at the time of resumption of
methadone treatment, and 2 hr afterward). Safety and
tolerability measures (physical examinations, adverse
events, laboratory tests, vital signs, and continuous ECG
recordings) were assessed from baseline to 144 hr after
noribogaine/placebo administration, and outpatient and tele-
phone assessments were performed until 35 days later.
Noribogaine was well tolerated: no signicant changes
were observed in vital signs, safety laboratory tests, oxime-
try or capnography, respiratory rate, or physical examina-
tions, and there were no deaths or serious adverse effects.
The most frequent adverse effects were transient changes
in light perception (light brighter than usual), headache, and
nausea, and no hallucinogenic effects were reported.
However, noribogaine induced signicant dose- and
concentration-dependent QT interval prolongation (a risk
factor for cardiac arrhythmias and sudden death), which
reached clinically concerning levels with the 180 mg of
dose. Opiate/opioid withdrawal symptoms increased 12hr
before resumption of methadone treatment as well as pupil
diameter. Noribogaine did not produce signicant reduc-
tions on subjective and objective rating scales measuring
opiate/opioid withdrawal symptoms (SOWS, OOWS, and
COWS) and also failed to induce signicant increases in the
time to resumption of methadone treatment, which was very
similar between the placebo and noribogaine groups. Sur-
prisingly, the 120-mg dose group experienced the longest
time to resumption of methadone treatment, which was
supported with the lowest scores on opiate/opioid withdraw-
al symptoms.
DISCUSSION
In this systematic review, eight studies suitable for inclusion
regarding the investigation of the antiaddictive effects of
ibogaine in humans were identied. Despite the small
number of studies, the open-label nature of most citations
(seven), the high degree of heterogeneity among them, and
the results reported in the case series suggest that ibogaine/
noribogaine signicantly reduced opiate/opioid withdrawal
symptoms and that many subjects remained drug-free for
several days after treatment. However, the only clinical trial
performed, using noribogaine, failed to nd signicant
reductions on opiate/opioid withdrawal symptoms. There-
fore, the antiaddictive effects of ibogaine/noribogaine
should be interpreted with caution.
The absence of effects of noribogaine could be related to
several factors. First, the equivalence between therapeutic
doses of ibogaine and noribogaine is not well known, so the
lack of effects could be related to the administration of low
doses of noribogaine, which would not be enough to achieve
anti-withdrawal effects. In fact, based on their studies
involving the administration of ibogaine and noribogaine
Journal of Psychedelic Studies 1(1), pp. 2028 (2017) |25
Antiaddictive effects of ibogaine
to healthy volunteers (Glue, Lockhart, et al., 2015;Glue,
Winter, et al., 2015), the authors predicted that an ibogaine
dose of 286 mg would lead to noribogaine C
max
values
comparable to a 180-mg dose of noribogaine, and there are
no reports in the bibliography, where such a low dose of
ibogaine has been administered to opiate-/opioid-dependent
subjects. Second, the case series described the treatments of
subjects mostly dependent on heroin, with fewer patients
reporting the use of methadone, and most of the reports did
not differentiate between heroin/methadone users.
Methadone has a longer half-life than heroin (Argoff &
Silvershein, 2009). In another paper, Glue, Cape, Tunnicliff,
Lockhart, Lam, Gray, et al. (2016) detailed how they did the
patients switching from methadone to morphine in 6 days
and explained that at that time 91% of methadone was
cleared. They also found a mean elimination half-life of
59 hr. Furthermore, subjective reports of methadone depen-
dents suggest that they may suffer withdrawal symptoms for
more than a month. Thus, the absence of signicant reduc-
tions in opiate/opioid withdrawal, as the authors acknowl-
edge, may need repetitive doses of ibogaine/noribogaine if
the propose is to detoxify from methadone.
The most important limitation of the reviewed studies is
that seven of the eight citations are open-label case series.
The lack of control groups and placebo in these studies does
not allow to suggest causation, especially considering that
most treatments were performed in non-medical and unsu-
pervised contexts with no standardized protocols. Therefore,
it is not possible to afrm that ibogaine/noribogaine are
effective treatments for drug dependence. Moreover, the
only clinical trial performed with noribogaine did not nd
signicant reductions on opiate/opioid withdrawal symp-
toms, suggesting that the results reporting antiaddictive
effects of ibogaine/noribogaine should be interpreted with
caution.
However, animal studies consistently show that ibogaine
signicantly reduces opiate/opioid and cocaine self-
administration (Alper, 2001;Alper et al., 2008;Belgers
et al., 2016;Brown, 2013;Donnelly, 2011;Frenken, 2001;
Lotsof & Alexander, 2001;Mash et al., 1998), and the
pattern of results observed in all case series including the
duration of the therapeutic effects of ibogaine is very
similar (from 24 hr to days/weeks). However, as exposed
above, the only clinical trial found in the review reported
that noribogaine failed to reduce opiate/opioid withdrawal
symptoms (Glue, Cape, Tunnicliff, Lockhart, Lam, Hung,
et al., 2016).
Some researchers have been speculated that the thera-
peutic effects of ibogaine seem to be related to the pharma-
cokinetics of its main metabolite, noribogaine, which is
longer lasting, and to the multiple receptor prole of both
compounds (Alper, 2001;Alper et al., 2008;Brown, 2013;
Donnelly, 2011;Frenken, 2001;Kolp et al., 2007;Lotsof &
Alexander, 2001;Mash et al., 1998,2000,2001). The
neurochemical effects of ibogaine/noribogaine are not
completely understood and may include antagonism of the
NMDA glutamatergic and α3β4 adrenergic receptors, inhi-
bition of 5-HT reuptake transporter and of dopamine release
in the nucleus accumbens and other brain areas, agonism of
the σ
2
,κ-opioid, and 5-HT
2A
receptors, and enhancement of
GDNF (Alper, 2001;Alper et al., 2008;Brown, 2013;Mash
et al., 1998). Antagonism of the NMDA receptor is shared
with the anesthetic hallucinogen ketamine, which has been
reported to show anxiolytic (Kolp et al., 2007), antidepres-
sive (Luckenbaugh et al., 2014;Sos et al., 2013), and
antiaddictive (Dakwar, Levin, Foltin, Nunes, & Hart,
2014;Krupitsky et al., 2007) effects. Agonism of the
κ-opioid receptor is shared with the plant hallucinogen
salvinorin A, which has potential effects in the treatment
of stimulant-related disorders (Dos Santos, Crippa,
Machado-de-Sousa, & Hallak, 2014). Agonism of the
5-HT
2A
receptor is shared with classic serotonergic hallu-
cinogens such as LSD, psilocybin, and ayahuasca/DMT,
which has anxiolytic, antidepressive, and antiaddictive
properties (Dos Santos, Os´orio, Crippa, & Hallak, 2016;
Dos Santos, Os´orio, Crippa, Riba, et al., 2016;Nunes et al.,
2016).
An important limitation to the clinical use of ibogaine/
noribogaine is the possible toxicity of these alkaloids, which
include fatalities, cardiac arrhythmias, psychosis, mania,
and seizures (Alper, 2001;Alper et al., 2012;Breuer
et al., 2015;Brown, 2013;Forsyth et al., 2016;Glue,
Lockhart, et al., 2015;Glue, Winter, et al., 2015;Houenou
et al., 2011;Koenig & Hilber, 2015;Litjens & Brunt, 2016;
Marta et al., 2015;Meisner et al., 2016). Moreover, the
clinical trial with noribogaine reported a signicant
dose-dependent increase in QT interval prolongation, which
is a risk factor for cardiac arrhythmias and sudden death.
Thus, a clear and rm warning of the cardiovascular dangers
of ibogaine/noribogaine should be provided to patients, who
must also be informed of the risks of using drugs in
particular opiates/opioids during or immediately after ibo-
gaine/noribogaine intake, which may potentiate the effects
of these alkaloids or of the opiates/opioids, thus increasing
the risk of an overdose. This is especially important con-
sidering that many ibogaine treatments are performed under
non-medical/unsupervised conditions using extracts of un-
known purity sold in the black market. The absence of
proper medical screening and monitoring increases the
possibility of hazardous situations (Alper, 2001;Alper
et al., 2012;Breuer et al., 2015;Brown, 2013;Forsyth
et al., 2016;Glue, Lockhart et al., 2015;Glue, Winter,
et al., 2015;Houenou et al., 2011;Koenig & Hilber, 2015;
Litjens & Brunt, 2016;Marta et al., 2015;Meisner et al.,
2016).
In summary, the results of this systematic review suggest
that ibogaine/noribogaine have antiaddictive properties.
Although these results are supported by animal studies, no
controlled clinical trials have been performed with ibogaine,
and the only clinical trial assessing the antiaddictive poten-
tials of noribogaine failed to nd signicant results. These
results suggest a more cautious interpretation of the anti-
addictive effects of ibogaine/noribogaine.
The use of heroin and synthetic opiates has reached an
alarming level in Europe (EMCDDA, 2016),andonlyinthe
United States, 44 people die every day from overdose of
prescription painkillers (SAMHSA, 2016). Moreover, there
are no proper medications for treating withdrawal symptoms
associated with methadone use in the case of patients that use
this drug for treating dependence on other opiates/opioids,
such as heroin. In this sense, the investigation of ibogaine/
noribogaine could provide new pharmacological treatments
26 |Journal of Psychedelic Studies 1(1), pp. 2028 (2017)
dos Santos et al.
with fast-acting and sustained benecial effects for patients
suffering drug dependence, especially to opiates/opioids and
cocaine. Controlled studies are needed to replicate these
ndings.
Acknowledgements: RGDS is the Fellow of the Brazilian
National Post-Doctorate Program (PNPD/CAPES) and the
member of the ICEERS Advisory Board. JCB is the Scien-
tic Investigations Director of ICEERS. ICEERS is a non-
prot organization that promotes the scientic research of
ibogaine. JECH receives a CNPq (Brazil) Productivity
Fellowship Award. None of the authors received any spe-
cic funding for participating in this investigation. All
authors had full access to all the data and had nal respon-
sibility for the decision to submit for publication.
Conict of interest: The authors declare that they have no
conict of interest.
REFERENCES
Alper, K. R. (2001). Ibogaine: A review. In K. Alper & G. A.
Cordell (Eds.), The alkaloids: Chemistry and biology (1st ed.,
Vol. 56, pp. 138). London: Academic Press/Elsevier.
Alper, K. R., Lotsof, H. S., Frenken, G. M., Luciano, D. J., &
Bastiaans, J. (1999). Treatment of acute opioid withdrawal
with ibogaine. American Journal of Addictions,8(3), 234242.
doi:10.1080/105504999305848
Alper, K. R., Lotsof, H. S., & Kaplan, C. D. (2008). The ibogaine
medical subculture. Journal of Ethnopharmacology, 115(1),
924. doi:10.1016/j.jep.2007.08.034
Alper, K. R., Staji´c, M., & Gill, J. R. (2012). Fatalities temporally
associated with the ingestion of ibogaine. Journal of Forensic
Sciences, 57(2), 398412. doi:10.1111/j.1556-4029.2011.
02008.x
Argoff, C. E., & Silvershein, D. I. (2009). A comparison of long-
and short-acting opioids for the treatment of chronic noncancer
pain: Tailoring therapy to meet patient needs. Mayo Clinic
Proceedings, 84(7), 602612. doi:10.1016/S0025-6196(11)
60749-0
Belgers, M., Leenaars, M., Homberg, J. R., Ritskes-Hoitinga, M.,
Schellekens, A. F., & Hooijmans, C. R. (2016). Ibogaine and
addiction in the animal model, a systematic review and meta-
analysis. Translational Psychiatry, 6(5), e826. doi:10.1038/
tp.2016.71
Breuer, L., Kasper, B. S., Schwarze, B., Gschossmann, J. M.,
Kornhuber, J., & Müller, H. H. (2015). Herbal seizures”–
Atypical symptoms after ibogaine intoxication: A case report.
Journal of Medical Case Reports, 9(1), 243. doi:10.1186/
s13256-015-0731-4
Brown, T. K. (2013). Ibogaine in the treatment of substance
dependence. Current Drug Abuse Reviews, 6(1), 316.
doi:10.2174/15672050113109990001
Dakwar, E., Levin, F., Foltin, R., Nunes, E., & Hart, C. (2014). The
effects of subanesthetic ketamine infusions on motivation to
quit and cue-induced craving in cocaine-dependent research
volunteers. Biological Psychiatry, 76(1), 4046. doi:10.1016/
j.biopsych.2013.08.009
Donnelly, J. R. (2011). The need for ibogaine in drug and alcohol
addiction treatment. Journal of Legal Medicine, 32(1), 93114.
doi:10.1080/01947648.2011.550832
Dos Santos, R. G., Crippa, J. A., Machado-de-Sousa, J. P., &
Hallak, J. E. (2014). Salvinorin A and related compounds as
therapeutic drugs for psychostimulant-related disorders.
Current Drug Abuse Reviews, 7(2), 128132. doi:10.2174/
1874473708666150107122741
Dos Santos, R. G., Os´orio, F. L., Crippa, J. A., & Hallak, J. E.
(2016). Antidepressive and anxiolytic effects of ayahuasca: A
systematic literature review of animal and human studies.
Revista Brasileira de Psiquiatria, 38(1), 6572. doi:10.1590/
1516-4446-2015-1701
Dos Santos, R. G., Os´orio, F. L., Crippa, J. A., Riba, J., Zuardi,
A. W., & Hallak, J. E. (2016). Antidepressive, anxiolytic, and
antiaddictive effects of ayahuasca, psilocybin and lysergic acid
diethylamide (LSD): A systematic review of clinical trials
published in the last 25 years. Therapeutic Advances in Psycho-
pharmacology, 6(3), 193213. doi:10.1177/2045125316638008
European Monitoring Centre for Drugs and Drug Addiction
(EMCDDA). (2016). European drug report 2016: Trends and
developments. Luxembourg: Publications Ofce of the
European Union.
Forsyth, B., Machado, L., Jowett, T., Jakobi, H., Garbe, K., Winter,
H., & Glue, P. (2016). Effects of low dose ibogaine on subjective
mood state and psychological performance. Journal of Ethno-
pharmacology, 189, 1013. doi:10.1016/j.jep.2016.05.022
Frenken, G. (2001). From the roots up: Ibogaine and addict self-
help. In K. Alper & G. A. Cordell (Eds.), The alkaloids:
Chemistry and biology (1st ed., Vol. 56, pp. 283292).
London: Academic Press/Elsevier.
Glue, P., Cape, G., Tunnicliff, D., Lockhart, M., Lam, F., Gray, A.,
Hung, N., Hung, C. T., Harland, S., Devane, J., Howes, J.,
Weis, H., & Friedhoff, L. (2016). Switching opioid-dependent
patients from methadone to morphine: Safety, tolerability, and
methadone pharmacokinetics. Journal of Clinical Pharmacol-
ogy, 56(8), 960965. doi:10.1002/jcph.704
Glue, P., Cape, G., Tunnicliff, D., Lockhart, M., Lam, F., Hung,
N., Hung, C. T., Harland, S., Devane, J., Crockett, R. S.,
Howes, J., Darpo, B., Zhou, M., Weis, H., & Friedhoff, L.
(2016). Ascending single-dose, double-blind, placebo-
controlled safety study of noribogaine in opioid-dependent
patients. Clinical Pharmacology in Drug Development, 5(6),
460468. doi:10.1002/cpdd.254
Glue, P., Lockhart, M., Lam, F., Hung, N., Hung, C. T., &
Friedhoff, L. (2015). Ascending-dose study of noribogaine in
healthy volunteers: Pharmacokinetics, pharmacodynamics,
safety, and tolerability. Journal of Clinical Pharmacology,
55(2), 189194. doi:10.1002/jcph.404
Glue, P., Winter, H., Garbe, K., Jakobi, H., Lyudin, A., Lenagh-
Glue, Z., & Hung, C. T. (2015). Inuence of CYP2D6 activity
on the pharmacokinetics and pharmacodynamics of a single
20 mg dose of ibogaine in healthy volunteers. Journal of
Clinical Pharmacology, 55(6), 680687. doi:10.1002/jcph.471
Houenou, J., Homri, W., Leboyer, M., & Drancourt, N. (2011).
Ibogaine-associated psychosis in schizophrenia: A case report.
Journal of Clinical Psychopharmacology, 31(5), 659.
doi:10.1097/JCP.0b013e31822c6509
Koenig, X., & Hilber, K. (2015). The anti-addiction drug ibogaine
and the heart: A delicate relation. Molecules, 20(2),
22082228. doi:10.3390/molecules20022208
Journal of Psychedelic Studies 1(1), pp. 2028 (2017) |27
Antiaddictive effects of ibogaine
Kolp, E., Young, M. S., Friedman, H., Krupitsky, E., Jansen, K., &
OConnor, L.-A. (2007). Ketamine-enhanced psychotherapy:
Preliminary clinical observations on its effectiveness in treating
death anxiety. International Journal of Transpersonal Studies,
26(1), 117.
Krupitsky, E., Burakov, A., Dunaevsky, I., Romanova, T., Slavina,
T., & Grinenko, A. (2007). Single versus repeated sessions of
ketamine-assisted psychotherapy for people with heroin de-
pendence. Journal of Psychoactive Drugs, 39(1), 1319.
doi:10.1080/02791072.2007.10399860
Litjens, R. P., & Brunt, T. M. (2016). How toxic is ibogaine?
Clinical Toxicology, 54(4), 297302. doi:10.3109/
15563650.2016.1138226
Lotsof, H. S., & Alexander, N. E. (2001). Case studies of ibogaine
treatment: Implications for patient management strategies. In
K. Alper & G. A. Cordell (Eds.), The alkaloids: Chemistry and
biology (1st ed., Vol. 56, pp. 293313). London: Academic
Press/Elsevier.
Luciano, D. (1998). Observations on treatment with ibogaine.
American Journal of Addictions, 7(1), 8989. doi:10.1111/
j.1521-0391.1998.tb00472.x
Luckenbaugh, D. A., Niciu, M. J., Ionescu, D. F., Nolan, N. M.,
Richards, E. M., Brutsche, N. E., Guevara, S., & Zarate, C. A.
(2014). Do the dissociative side effects of ketamine mediate its
antidepressant effects? Journal of Affective Disorders, 159,
5661. doi:10.1016/j.jad.2014.02.017
Marta, C. J., Ryan, W. C., Kopelowicz, A., & Koek, R. J. (2015).
Mania following use of ibogaine: A case series. American
Journal of Addictions, 24(3), 203205. doi:10.1111/ajad.12209
Mash, D. C., Kovera, C. A., Buck, B. E., Norenberg, M. D.,
Shapshak, P., Hearn, W. L., & Sanchez-Ramos, J. (1998).
Medication development of ibogaine as a pharmacotherapy for
drug dependence. Annals of the New York Academy of Sciences,
844, 274292. doi:10.1111/j.1749-6632.1998.tb08242.x
Mash, D. C., Kovera, C. A., Pablo, J., Tyndale, R., Ervin, F. R.,
Kamlet, J. D., & Hearn, W. L. (2001). Ibogaine in the treatment
of heroin withdrawal. In K. Alper, & G. A. Cordell (Eds.), The
alkaloids: Chemistry and biology (1st ed., Vol. 56, pp. 155
171). London: Academic Press/Elsevier.
Mash, D. C., Kovera, C. A., Pablo, J., Tyndale, R. F., Ervin, F. D.,
Williams, I. C., Singleton, E. G., & Mayor, M. (2000). Ibogaine:
Complex pharmacokinetics, concerns for safety, and preliminary
efcacy measures. Annals of the New York Academy of Sciences,
914(1), 394401. doi:10.1111/j.1749-6632.2000.tb05213.x
Meisner, J. A., Wilcox, S. R., & Richards, J. B. (2016). Ibogaine-
associated cardiac arrest and death: Case report and review of
the literature. Therapeutic Advances in Psychopharmacology,
6(2), 9598. doi:10.1177/2045125315626073
Moher, D., Liberati, A., Tetzlaff, J., Altman, D. G., & The
PRISMA Group. (2009). Preferred reporting items for system-
atic reviews and meta-analyses: The PRISMA statement. PLoS
Medicine, 6, e1000097. doi:10.1371/journal.pmed.1000097
Nunes, A. A., Dos Santos, R. G., Os´orio, F. L., Sanches, R. F.,
Crippa, J. A., & Hallak, J. E. (2016). Effects of ayahuasca and
its alkaloids on drug dependence: A systematic literature
review of quantitative studies in animals and humans. Journal
of Psychoactive Drugs, 48(3), 195205. doi:10.1080/
02791072.2016.1188225
Schenberg, E. E., de Castro Comis, M. A., Chaves, B. R., &
Da Silveira, D. X. (2014). Treating drug dependence with
the aid of ibogaine: A retrospective study. Journal of
Psychopharmacology, 28(11), 9931000. doi:10.1177/
0269881114552713
Sheppard, S. G. (1994). A preliminary investigation of ibogaine:
Case reports and recommendations for further study. Journal of
Substance Abuse Treatment, 11(4), 379385. doi:10.1016/
0740-5472(94)90049-3
Sos, P., Klirova, M., Novak, T., Kohutova, B., Horacek, J., &
Palenicek, T. (2013). Relationship of ketamines antidepres-
sant and psychotomimetic effects in unipolar depression.
Neuroendocrinology Letters, 34(4), 287293.
Substance Abuse and Mental Health Services Administration
(SAMHSA). (2016). Opioids. Retrieved from http://www.
samhsa.gov/atod/opioids
28 |Journal of Psychedelic Studies 1(1), pp. 2028 (2017)
dos Santos et al.
... 2. There is a persistent desire or unsuccessful efforts to cut down or control opioid use. 3. A great deal of time is spent on activities necessary to obtain the opioid, use the opioid, or recover from its effects. ...
... The plant Tabernanthe iboga has been used for centuries, playing a central role in various spiritual and healing practices such as Bwiti and other African equatorial traditions [1,2]. The main alkaloid of the plant, ibogaine (IBO), is a psychoactive compound with putative antiaddictive properties primarily found in the root bark [3]. IBO is subjected to extensive first-pass metabolism in the liver, primarily by the cytochrome P4502D6 (CYP2D6) enzyme. ...
... Apart from fatal outcomes, several health problems related to SUDs can be observed over the short-and long-term, including intoxication, misuse, heart disease, depression, and more [2]. Treating SUDs generally has poor adherence and high rates of relapse [3,4] which makes innovative approaches highly necessary. Ibogaine (IBO) is an alkaloid naturally found in the root bark of some plants belonging to the Apocynaceae family, including Tabernanthe iboga. ...
Thesis
Full-text available
Substance use disorders remain one of the most challenging health problems to address. Specifically, opioid dependence has caused serious public health issues in countries such as the United States and Canada over the last decade, underscoring the need for innovative and effective treatments. Recently, mental health researchers have shown a renewed interest in psychedelic drugs. Substances such as lysergic acid diethylamide (LSD), psilocybin mushrooms, and ayahuasca have shown promising results in treating conditions including major depression and anxiety disorders. Among these, ibogaine, an alkaloid found naturally in the West African plant Tabernanthe iboga, appears particularly effective in treating substance use disorders. However, despite its widespread underground and unsupervised use, controlled trials evaluating the safety and efficacy of ibogaine are lacking, and its mechanisms of action remain largely unknown. In this thesis, we conducted both clinical and preclinical studies on ibogaine to provide more evidence about this molecule and to expand our understanding of it. Clinically, we performed a systematic review of adverse events in humans associated with ibogaine to collect updated safety data. Subsequently, we designed a Phase II, randomized, double-blind clinical trial. In this trial, low, single doses of ibogaine (100 mg) were administered in the context of methadone detoxification. Plasma samples from the trial were analyzed using a metabolomic approach. The systematic review and clinical trial data were complemented with a narrative review, which identified all potential ibogaine targets associated with its anti-addictive effect and provided updated mechanistic literature. Preclinically, we designed a study with mice to elucidate further mechanisms of action. Following acute administration of ibogaine, brain tissue was analyzed using transcriptomic analysis to determine the expression levels of a wide array of genes. The clinical results were highly promising. The systematic review highlighted the need for medical supervision during ibogaine treatments due to its potential to prolong the QT interval and its complex metabolism. In the clinical trial, which included 20 patients, we observed a significant decrease in both tolerance to methadone and opioid withdrawal syndrome (OWS). As a result, 17 out of 20 patients were able to halve their methadone dose over seven days without experiencing OWS symptoms and discontinue their daily methadone use for an average of 18.03 hours. No serious adverse events were reported. Results from the metabolomic analysis suggest that ibogaine can potentially reverse the effects of chronic opioid use on energy metabolism. These findings align with the multi-target profile of ibogaine identified in the narrative review. The preclinical study revealed new potential pathways associated with ibogaine's anti-addictive effects. Specifically, genes related to hormonal pathways and synaptogenesis showed increased expression after acute ibogaine administration. Additionally, gender differences were observed, with females exhibiting changes in 28 genes compared to eight in males. This thesis provides the first evidence of ibogaine's safety and efficacy in a Phase II study and delves deeper into its mechanisms of action through a review, a preclinical study, and an analysis of human plasma samples using innovative techniques. We conclude that ibogaine represents a promising candidate for the treatment of opioid use disorders, warranting further research.
... Ibogaine has high affinity for k-opioid, m-opioid, N-methyl-D-aspartate (NMDA), and sigma-2 receptors, and it also inhibits voltage-gated ion channels. 6,7,[16][17][18][19][20] Sigma receptors are a hypothesized active site for other neuropsychiatric agents, including fluvoxamine 21,22 and DMT (see the "DMT and Ayahuasca" chapter of this special edition). Ibogaine's NMDA antagonism and k-opioid agonism may mediate its efficacy for treating substance use disorder and withdrawal. ...
... 27 Ibogaine is also thought to have an effect on the expression of various neurotrophic factors (NFs) including brain-derived neurotrophic factor and glial-derived neurotrophic factor, which may further contribute to the enduring anti-addictive and antidepressant properties of ibogaine. 17,28 Additionally, ibogaine has been found to have anti-inflammatory and antioxidant effects. 29 ...
... (Abuse of ibogaine itself is not a concern. 3 ) Animal studies suggest that ibogaine significantly reduces the severity of withdrawal symptoms from opioids and decreases opioid and cocaine self-administration. 17,18 In humans, there has been one double-blind RCT (Table 1) and at least six observational studies that reported changes in OUD or cocaine use disorder symptoms from ibogaine treatment, and a number of retrospective surveys and case reports (see Ref. 65 for a systematic review). The first observational study examined 27 opioid-and/or cocaine-dependent participants in an ibogaine treatment clinic in St. Kitts and found that ibogaine significantly reduced opioid cravings. ...
Article
Full-text available
Background Ibogaine is a plant-derived alkaloid that has been used for thousands of years in rites of passage and spiritual ceremonies in West-Central Africa. In the West, it has primarily been used and studied for its anti-addictive properties and more recently for other neuropsychiatric indications, including post-traumatic stress disorder, depression, anxiety, and traumatic brain injury. Areas of Uncertainty Ibogaine requires careful patient screening and monitoring because of significant safety issues. There is potential for cardiotoxicity (prolonged QT interval); without rigorous screening, fatal arrhythmias may occur. However, preliminary research suggests that co-administration of ibogaine with magnesium may mitigate cardiotoxicity. Additionally, ibogaine may have dangerous interactions with opiates, so patients who receive ibogaine treatment for opioid use disorder must withdraw from long-acting opioids. Other potential concerning effects of ibogaine include rare incidences of mania or psychosis. Anticipated transient effects during ibogaine treatment can include ataxia, tremors, and gastrointestinal symptoms. Therapeutic Advances Robust effects after a single treatment with ibogaine have been reported. In open-label and randomized controlled trials (RCTs), ibogaine reduces heroin and opioid cravings by upwards of 50%, up to 24 weeks after the treatment. An observational study of 30 Special Operations Forces veterans with mild traumatic brain injury reported that 86% were in remission from post-traumatic stress disorder, 83% from depression, and 83% from anxiety, one month after a single-dose ibogaine treatment. Limitations Although there are several observational and open-label studies, there is only a single double-blind, placebo-controlled RCT on ibogaine. More RCTs with large sample sizes must be conducted to support ibogaine's safety and efficacy. Conclusions Given the promising preliminary findings, ibogaine could potentially fill a much-needed gap in treatments for challenging conditions, including opioid dependence. Ibogaine's remarkable effects in traditionally treatment-resistant, combat-exposed individuals hints at its potential in broader populations with physical and psychological trauma.
... Ibogaine has high affinity for k-opioid, m-opioid, N-methyl-D-aspartate (NMDA), and sigma-2 receptors, and it also inhibits voltage-gated ion channels. 6,7,[16][17][18][19][20] Sigma receptors are a hypothesized active site for other neuropsychiatric agents, including fluvoxamine 21,22 and DMT (see the "DMT and Ayahuasca" chapter of this special edition). Ibogaine's NMDA antagonism and k-opioid agonism may mediate its efficacy for treating substance use disorder and withdrawal. ...
... 27 Ibogaine is also thought to have an effect on the expression of various neurotrophic factors (NFs) including brain-derived neurotrophic factor and glial-derived neurotrophic factor, which may further contribute to the enduring anti-addictive and antidepressant properties of ibogaine. 17,28 Additionally, ibogaine has been found to have anti-inflammatory and antioxidant effects. 29 ...
... (Abuse of ibogaine itself is not a concern. 3 ) Animal studies suggest that ibogaine significantly reduces the severity of withdrawal symptoms from opioids and decreases opioid and cocaine self-administration. 17,18 In humans, there has been one double-blind RCT (Table 1) and at least six observational studies that reported changes in OUD or cocaine use disorder symptoms from ibogaine treatment, and a number of retrospective surveys and case reports (see Ref. 65 for a systematic review). The first observational study examined 27 opioid-and/or cocaine-dependent participants in an ibogaine treatment clinic in St. Kitts and found that ibogaine significantly reduced opioid cravings. ...
Preprint
Full-text available
Background: Ibogaine is a plant-derived alkaloid that has been used for thousands of years in rites of passage and spiritual ceremonies in West-Central Africa. In the West, it has primarily been used and studied for its anti-addictive properties and more recently for other neuropsychiatric indications, including post-traumatic stress disorder (PTSD), depression, anxiety, and traumatic brain injury (TBI). Areas of Uncertainty: Ibogaine requires careful patient screening and monitoring due to significant safety issues. There is potential for cardiotoxicity (prolonged QT interval); without rigorous screening, fatal arrhythmias may occur. Additionally, ibogaine may have dangerous interactions with opiates, so patients who receive ibogaine treatment for opioid use disorder must withdraw from long-acting opioids. Other potential concerning effects of ibogaine include rare incidents of mania or psychosis. Anticipated transient effects during ibogaine treatment can include ataxia, tremors, and gastrointestinal symptoms.Therapeutic Advances: Nonetheless, robust effects following a single treatment with ibogaine have been reported. In open-label and randomized controlled trials (RCTs), ibogaine reduces heroin and opioid cravings by upwards of 50%, up to 24 weeks after the treatment. Observational studies have shown that ibogaine, in combination with the psychedelic 5-methoxy-dimethyltryptamine (5-MeO-DMT), significantly diminishes PTSD (d = 0.414) and depression symptoms (d = 0.275). Treatment clinics have opened in countries where ibogaine is either legal or unregulated for people struggling with substance use disorders or refractory distress. Conclusions: Given the promising preliminary findings, ibogaine could potentially fill a much-needed gap in treatments for challenging conditions, including opioid dependence and combat-related symptom complexes. However, there is currently a lack of rigorous research, such as double-blind, placebo-controlled RCTs, to support ibogaine’s safety and efficacy. It is imperative to develop practices that reduce patient risk and improve treatment sustainability, given high rates of ibogaine trafficking.
... Ibogaine (IBO) is the main alkaloid of the shrub Tabernanthe iboga (Lavaud and Massiot, 2017). While traditionally part of West Central African medicine (Faura and Langlois, 2022), IBO is now gaining recognition as a potential treatment for Substance Use Disorders (SUDs) (Belgers et al., 2016;dos Santos et al., 2016). A substantial body of preclinical research supports IBO's effectiveness in reducing drug self-administration and decreasing the likelihood of drug relapse involving cocaine, ethanol, nicotine, and opioids (Belgers et al., 2016). ...
... This phenomenon has been described as a "vast, uncontrolled experiment" (Vastag, 2005). A systematic review of these studies supports IBO's therapeutic potential to significantly reduce drug withdrawals, cravings, and self-administration in patients with SUDs (dos Santos et al., 2016). The duration of the therapeutic effect varies highly between studies, ranging from 24 h to weeks or months (dos Santos et al., 2016). ...
... A systematic review of these studies supports IBO's therapeutic potential to significantly reduce drug withdrawals, cravings, and self-administration in patients with SUDs (dos Santos et al., 2016). The duration of the therapeutic effect varies highly between studies, ranging from 24 h to weeks or months (dos Santos et al., 2016). ...
Article
Full-text available
Background There is a growing interest in studying ibogaine (IBO) as a potential treatment for substance use disorders (SUDs). However, its clinical use has been hindered for mainly two reasons: First, the lack of randomized, controlled studies informing about its safety and efficacy. And second, IBO’s mechanisms of action remain obscure. It has been challenging to elucidate a predominant mechanism of action responsible for its anti-addictive effects. Objective To describe the main targets of IBO and its main metabolite, noribogaine (NOR), in relation to their putative anti-addictive effects, reviewing the updated literature available. Methods A comprehensive search involving MEDLINE and Google Scholar was undertaken, selecting papers published until July 2022. The inclusion criteria were both theoretical and experimental studies about the pharmacology of IBO. Additional publications were identified in the references of the initial papers. Results IBO and its main metabolite, NOR, can modulate several targets associated with SUDs. Instead of identifying key targets, the action of IBO should be understood as a complex modulation of multiple receptor systems, leading to potential synergies. The elucidation of IBO’s pharmacology could be enhanced through the application of methodologies rooted in the polypharmacology paradigm. Such approaches possess the capability to describe multifaceted patterns within multi-target drugs. Conclusion IBO displays complex effects through multiple targets. The information detailed here should guide future research on both mechanistic and therapeutic studies.
... Along with this, the irreversible blockage of human ether-ago-go-related gene (hERG) potassium channel and prolonged QT interval and development of TdP arrhythmias remained as the other major risk factors associated with ibogaine when administered at low micromolar concentration [19][20][21][22][23]. The limited number of case studies (mainly anecdotal reports) and lack of extensive clinical research had raised questions about ibogaine's efficacy and safety as an antiaddictive and analgesic agent in humans [24][25][26][27]. This major shortcoming, however, motivated scientists to develop safer iboga congeners with fewer side effects, while retaining their anti-addictive properties and reducing the simultaneous targeting of multiple brain receptors [28][29][30][31]. ...
Preprint
Full-text available
The clinical importance of iboga alkaloids lies in their efficacy in reversing drug addiction and modulating drug tolerance. However, due to safety concerns, their use is restricted to appropriate medical supervision. These alkaloids often cause severe hallucinogenic effects due to differential binding to various brain receptors and cardiotoxicity by blocking the human ether-ago go related gene (hERG) potassium channel. To create safer analogs, our group previously synthesized various benzofuran-containing iboga analogs with good opioid binding selectivity and excellent antinociceptive property. However, the present manuscript disclosed a step-economical and cost-effective synthesis of modified ibogaine/ibogamine analogs (C1, C2, C3 & C4) with bio-isosteric replacement of the indole scaffold with a benzofuran moiety, and comparing their antinociceptive/anxiolytic activity with their natural counterparts. Among the synthesized iboga analogs, the Endo-iboga analogs (C2 & C4, epimers of C1 and C3, respectively) not only exhibited superior anti-inflammatory and oxidative stress-relieving activity, but also effectively improved restricted locomotor activity in a formalin-induced acute pain model in mice. These Endo-iboga analogs significantly elevated the levels of inhibitory neurotransmitters (GABA and dopamine) and brain-derived neurotrophic factor (BDNF) compared to their Exo-counterparts or previously published benzofuran-containing iboga analogs lacking the tetrahydroazepine ring. Amongst, C2 and C4, the latter exhibited superior cardiac safety profile in C2C12 cells (IC50 = 235 µM) and showed no adverse effects on rat hearts during in vivo ECG tests, indicated by no significant QTc prolongation. Overall, the development of bioisosteric iboga analogs, particularly C4, demonstrated significant potential for acute pain management without notable cardiotoxicity, representing a breakthrough in pain therapy innovation.
... Ibogaine, an iboga-type alkaloids, has shown promise as a medicinal drug. Several studies, both in animals and in humans, report long-term drug abstinence from various substances, including opioids, alcohol, and psychostimulants, and sustained reductions in depressive symptoms after ibogaine administration (Mash et al. 2000;Schenberg et al. 2014;Santos et al. 2017;Brown and Alper, 2018;Mash, 2018;Mash et al. 2018;Wasko et al. 2018). T. iboga is a plant native to Africa and is a limited resource. ...
Article
Full-text available
Coronaridine, a monoterpenoid indole alkaloid, is present in Tabernanthe iboga and the related species Tabernaemontana divaricata. Recent exhaustive analysis revealed its presence in Catharanthus roseus, though specific details remain unknown. We conducted a detailed analysis of coronaridine in C. roseus, detecting it in seedlings post-germination up to 8 weeks after sowing, with peak abundance at 3–4 weeks. Gradual decrease occurred from the flowering stage, and it was absent during seed formation. The accumulation varied dramatically with the plant's growth phase. LC–MS/MS analysis confirmed (−) coronaridine, consistent with T. iboga. Additionally, cultivating at 35 °C increased coronaridine accumulation over 10-fold. These findings hold potential for enhancing the stable production of iboga alkaloids for pharmaceutical use. Fullsize Image
... Strictosidine aglycones are the precursors of heteroyohimbane and yohimbane alkaloids such as the antihypertensive ajmalicine [36][37][38][39][40]. Geissoschizine is the precursor of sarpagane alkaloids such as the antiarrhythmic ajmaline [41,42]. Dihydroprecondylocarpine is the precursor of iboga alkaloids such as the antiaddictive ibogaine [43,44] and aspidosperma alkaloids such [45,46]. The identification of the MIA biosynthesis molecular actors in several plant species has progressively paved the way for the development of alternative biotechnological processes for MIA-derived pharmaceutical supply, notably by reconstructing the plant biosynthetic pathways in heterologous organisms [47][48][49]. ...
Article
Full-text available
Specialized metabolites possess diverse interesting biological activities and some cardenolides- and monoterpene indole alkaloids- (MIAs) derived pharmaceuticals are currently used to treat human diseases such as cancers or hypertension. While these two families of biocompounds are produced by specific subfamilies of Apocynaceae, one member of this medicinal plant family, the succulent tree Pachypodium lamerei Drake (also known as Madagascar palm), does not produce such specialized metabolites. To explore the evolutionary paths that have led to the emergence and loss of cardenolide and MIA biosynthesis in Apocynaceae, we sequenced and assembled the P. lamerei genome by combining Oxford Nanopore Technologies long-reads and Illumina short-reads. Phylogenomics revealed that, among the Apocynaceae whose genomes have been sequenced, the Madagascar palm is so far the species closest to the common ancestor between MIA producers/non-MIA producers. Transposable elements, constituting 72.48% of the genome, emerge as potential key players in shaping genomic architecture and influencing specialized metabolic pathways. The absence of crucial MIA biosynthetic genes such as strictosidine synthase in P. lamerei and non-Rauvolfioideae species hints at a transposon-mediated mechanism behind gene loss. Phylogenetic analysis not only showcases the evolutionary divergence of specialized metabolite biosynthesis within Apocynaceae but also underscores the role of transposable elements in this intricate process. Moreover, we shed light on the low conservation of enzymes involved in the final stages of MIA biosynthesis in the distinct MIA-producing plant families, inferring independent gains of these specialized enzymes along the evolution of these medicinal plant clades. Overall, this study marks a leap forward in understanding the genomic dynamics underpinning the evolution of specialized metabolites biosynthesis in the Apocynaceae family, with transposons emerging as potential architects of genomics restructuring and gene loss.
... After a period of more curbed and negative attention in the Global North, partially due to various governmental interventions (Giffort, 2020), the past two decades have witnessed a revival of scientific and public interest in the use of psychedelics for therapeutic purposes (Carhart-Harris & Goodwin, 2017). A fast-growing number of studies indicate that psychedelics, when used with certain conducts and frames, can alleviate a wide range of conditions such as major and treatment-resistant depression Gukasyan et al., 2022;Sanches et al., 2016;van Oorsouw et al., 2022), eating disorders (Spriggs et al., 2021), endof-life psychological distress (Gasser et al., 2014;Griffiths et al., 2016;Ross et al., 2016), alcohol, tobacco and opioid addiction (Bogenschutz et al., 2015;dos Santos et al., 2017;Johnson et al., 2014;Noorani et al., 2018), cluster headaches (Andersson et al., 2017) and improve general wellbeing (Griffiths et al., 2006;Haijen et al., 2018). These research results have been accompanied by a prosperous psychedelics industry and tourism and a rocketing amount of organisations, businesses, citizens' initiatives and media coverage trying to either ride, profit from or steer the wave (Chabloz, 2009;Psychedelic Invest, 2024;Steinhardt & Noorani, 2020). ...
Article
Full-text available
Within recent years, an increasing number of people and researchers in the Global North have become interested in psychedelic substances and their therapeutic application. While much of the current media attention and research effort mainly concentrate on the therapeutic potential and actions of the individual's acute psychedelic experience, this article explores the user‐perceived, therapeutic dynamics of psychedelics in a more long‐term perspective by charting the lived experiences and practices of ‘integration’ among psychedelic users in Denmark. Based on ethnographic fieldwork from November 2020 to June 2021, I offer a dual typology of self‐related integration as narrative and experiential‐somatic . Combining the two, I argue that psychedelic integration in contemporary Denmark can be viewed as a processual self‐transformation of the users' experiential orientation where understandings and/or modes of being from the acute psychedelic experience are woven into, prolonged, and/or embodied in their everyday existence .
Article
Full-text available
Accumulating psychedelic research has demonstrated a potential for improving mental health and wellbeing, yet studies in the context of eating disorders (EDs) are limited. This study aims to explore the subjective effects of psychedelic experiences to gain insight into the benefits and risks for people with EDs. Semi-structured interviews were conducted with eight adults aged 25–54 (mean age = 36.9), reporting to have had experiences with EDs and psychedelics in both naturalistic and clinical settings. Participants had multiple diagnoses and suffered chronic EDs, (mean age of onset = 13), diagnosed (N = 7) and undiagnosed (N = 1). Reports of cessation or the reduction of ED symptoms were unanimous and long-lasting for seven, with two participants reporting recovery attributed to psychedelic use. Two participants reported relapsing, attributed to environmental factors in the months following. Thematic analysis resulted in two superordinate themes, each comprising three subordinate themes. The first superordinate theme, ‘Exploring’ via the ‘gateway to healing’, illustrates mental, emotional, and transcendental elements of psychedelic experiences. The second superordinate theme, ‘Transformation’ and being ‘able to do the work’, illustrates cognitive and behavioural outcomes, with retrospective safety perceptions. These findings may provide more in-depth information on what benefits and experiences people with EDs can obtain from the use of psychedelic drugs and may inform more robust investigations of psychedelic-assisted therapy for the treatment of EDs.
Article
Full-text available
Introduction: Misuse of various new psychotropic substances such as ibogaine is increasing rapidly. Knowledge of their negative side effects is sparse. Case presentation: We present a case of intoxication with the herbal substance ibogaine in a 22-year-old white man. After taking a cumulative dose of 38 g (taken in two doses), he developed visual memories, nausea and vomiting. He developed a generalized tonic–clonic seizure with additional grand mal seizures. He was treated with midazolam and levetiracetam. Extended drug screenings and computed tomography and magnetic resonance imaging findings were all negative. Conclusions: Knowledge of the side effects of ibogaine has mainly come from reports of cardiovascular complications; seizures are rarely mentioned and experimental findings are inconsistent. It seems that ibogaine acts like a proconvulsive drug at high doses.
Article
Full-text available
Ibogaine is a naturally occurring substance which has been increasingly used in the lay-scene to reduce craving and relapse in patients with substance use disorders (SUDs). Although human clinical trials on the safety and efficacy of ibogaine are lacking, animal studies do support the efficacy of ibogaine. In this systematic review and meta-analysis (MA), we summarise these animal findings, addressing three questions: (1) does ibogaine reduce addictive behaviour in animal models of SUDs?; (2) what are the toxic effects of ibogaine on motor functioning, cerebellum and heart rhythm?; (3) what are neuropharmacological working mechanisms of ibogaine treatment in animal models of SUDs? MA of 27 studies showed that ibogaine reduced drug self-administration, particularly during the first 24 h after administration. Ibogaine had no effect on drug-induced conditioned place preference. Ibogaine administration resulted in motor impairment in the first 24 h after supplementation, and cerebral cell loss even weeks after administration. Data on ibogaines effect on cardiac rhythm, as well as on its neuropharmacological working mechanisms are limited. Our results warrant further studies into the clinical efficacy of ibogaine in SUD patients in reducing craving and substance use, but close monitoring of the patients is recommended because of the possible toxic effects. In addition, more work is needed to unravel the neuropharmacological working mechanisms of ibogaine and to investigate its effects on heart rhythm.
Article
Full-text available
To date, pharmacological treatments for mood and anxiety disorders and for drug dependence show limited efficacy, leaving a large number of patients suffering severe and persistent symptoms. Preliminary studies in animals and humans suggest that ayahuasca, psilocybin and lysergic acid diethylamide (LSD) may have antidepressive, anxiolytic, and antiaddictive properties. Thus, we conducted a systematic review of clinical trials published from 1990 until 2015, assessing these therapeutic properties. Electronic searches were performed using the PubMed, LILACS, and SciELO databases. Only clinical trials published in peer-reviewed journals were included. Of these, 151 studies were identified, of which six met the established criteria. Reviewed studies suggest beneficial effects for treatment-resistant depression, anxiety and depression associated with life-threatening diseases, and tobacco and alcohol dependence. All drugs were well tolerated. In conclusion, ayahuasca, psilocybin and LSD may be useful pharmacological tools for the treatment of drug dependence, and anxiety and mood disorders, especially in treatment-resistant patients. These drugs may also be useful pharmacological tools to understand psychiatric disorders and to develop new therapeutic agents. However, all studies reviewed had small sample sizes, and half of them were open-label, proof-of-concept studies. Randomized, double-blind, placebo-controlled studies with more patients are needed to replicate these preliminary findings.
Article
Full-text available
Objective: To conduct a systematic literature review of animal and human studies reporting anxiolytic or antidepressive effects of ayahuasca or some of its isolated alkaloids (dimethyltryptamine, harmine, tetrahydroharmine, and harmaline). Methods: Papers published until 3 April 2015 were retrieved from the PubMed, LILACS and SciELO databases following a comprehensive search strategy and using a predetermined set of criteria for article selection. Results: Five hundred and fourteen studies were identified, of which 21 met the established criteria. Studies in animals have shown anxiolytic and antidepressive effects of ayahuasca, harmine, and harmaline, and experimental studies in humans and mental health assessments of experienced ayahuasca consumers also suggest that ayahuasca is associated with reductions in anxiety and depressive symptoms. A pilot study reported rapid antidepressive effects of a single ayahuasca dose in six patients with recurrent depression. Conclusion: Considering the need for new drugs that produce fewer adverse effects and are more effective in reducing anxiety and depression symptomatology, the described effects of ayahuasca and its alkaloids should be further investigated.
Article
Full-text available
Context: Ibogaine is a psychoactive indole alkaloid found in the African rainforest shrub Tabernanthe Iboga. It is unlicensed but used in the treatment of drug and alcohol addiction. However, reports of ibogaine's toxicity are cause for concern. Objectives: To review ibogaine's pharmacokinetics and pharmacodynamics, mechanisms of action and reported toxicity. Methods: A search of the literature available on PubMed was done, using the keywords "ibogaine" and "noribogaine". The search criteria were "mechanism of action", "pharmacokinetics", "pharmacodynamics", "neurotransmitters", "toxicology", "toxicity", "cardiac", "neurotoxic", "human data", "animal data", "addiction", "anti-addictive", "withdrawal", "death" and "fatalities". The searches identified 382 unique references, of which 156 involved human data. Further research revealed 14 detailed toxicological case reports. Pharmacokinetics and pharmacodynamics: Ibogaine is metabolized mainly by CYP2D6 to the primary metabolite noribogaine (10-hydroxyibogamine). Noribogaine is present in clinically relevant concentrations for days, long after ibogaine has been cleared. Mechanisms of action: Ibogaine and noribogaine interact with multiple neurotransmitter systems. They show micromolar affinity for N-methyl-D-aspartate (NMDA), κ- and μ-opioid receptors and sigma-2 receptor sites. Furthermore, ibogaine has been shown to interact with the acetylcholine, serotonin and dopamine systems; it alters the expression of several proteins including substance P, brain-derived neurotrophic factor (BDNF), c-fos and egr-1. Neurotoxicity: Neurodegeneration was shown in rats, probably mediated by stimulation of the inferior olive, which has excitotoxic effects on Purkinje cells in the cerebellum. Neurotoxic effects of ibogaine may not be directly relevant to its anti-addictive properties, as no signs of neurotoxicity were found following doses lower than 25 mg/kg intra-peritoneal in rats. Noribogaine might be less neurotoxic than ibogaine. Cardiotoxicity: Ether-a-go-go-related gene (hERG) potassium channels in the heart might play a crucial role in ibogaine's cardiotoxicity, as hERG channels are vital in the repolarization phase of cardiac action potentials and blockade by ibogaine delays this repolarization, resulting in QT (time interval between the start of the Q wave and the end of the T wave in the electrical cycle of the heart) interval prolongation and, subsequently, in arrhythmias and sudden cardiac arrest. Twenty-seven fatalities have been reported following the ingestion of ibogaine, and pre-existing cardiovascular conditions have been implicated in the death of individuals for which post-mortem data were available. However, in this review, 8 case reports are presented which suggest that ibogaine caused ventricular tachyarrhythmias and prolongation of the QT interval in individuals without any pre-existing cardiovascular condition or family history. Noribogaine appears at least as harmful to cardiac functioning as ibogaine. Toxicity from drug-drug interaction: Polymorphism in the CYP2D6 enzyme can influence blood concentrations of both ibogaine and its primary metabolite, which may have implications when a patient is taking other medication that is subject to significant CYP2D6 metabolism. Conclusions: Alternative therapists and drug users are still using iboga extract, root scrapings, and ibogaine hydrochloride to treat drug addiction. With limited medical supervision, these are risky experiments and more ibogaine-related deaths are likely to occur, particularly in those with pre-existing cardiac conditions and those taking concurrent medications.
Article
Full-text available
A naturally occurring hallucinogenic plant alkaloid, ibogaine has been used as an adjuvant for opiate withdrawal for the past 50 years. In the setting of an escalating nationwide opiate epidemic, use of substances such as ibogaine may also increase. Therefore, familiarity with the mechanisms and potential adverse effects of ibogaine is important for clinicians. We present the case report of a man whose use of ibogaine resulted in cardiac arrest and death, complemented by a review of the literature regarding ibogaine’s clinical effects. A 40-year-old man who used ibogaine for symptoms of heroin withdrawal suffered acute cardiac arrest leading to cerebral edema and brain death. His presentation was consistent with ibogaine-induced cardiotoxicity and ibogaine-induced cardiac arrest, and a review of the literature regarding the history, mechanisms, risks and clinical outcomes associated with ibogaine is presented. The case presented underscores the significant potential clinical risks of ibogaine. It is important the healthcare community be aware of the possible effects of ibogaine such that clinicians can provide informed counseling to their patients regarding the risks of attempting detoxification with ibogaine.
Article
Recently, the anti-addictive potential of ayahuasca, a dimethyltryptamine(DMT)- and β-carboline-rich hallucinogenic beverage traditionally used by indigenous groups of the Northwest Amazon and currently by syncretic churches worldwide, has received increased attention. To better evaluate this topic, we performed a systematic literature review using the PubMed database to find quantitative studies (using statistical analysis) that assessed the effects of ayahuasca or its components in drug-related symptoms or disorders. We found five animal studies (using harmaline, harmine, or ayahuasca) and five observational studies of regular ayahuasca consumers. All animal studies showed improvement of biochemical or behavioral parameters related to drug-induced disorders. Of the five human studies, four reported significant reductions of dependence symptoms or substance use, while one did not report significant results. The mechanisms responsible for the anti-addictive properties of ayahuasca and its alkaloids are not clarified, apparently involving both peripheral MAO-A inhibition by the β-carbolines and central agonism of DMT at 5-HT2A receptors expressed in brain regions related to the regulation of mood and emotions. Although results are promising, controlled studies are needed to replicate these preliminary findings.
Article
Ethnopharmacological relevance: Root bark from Tabernanthe iboga has been used traditionally in West Africa as a psychoactive substance in religious rituals. In smaller doses it is reported anecdotally to have stimulant properties. Aim of the study: To evaluate the influence of a single 20mg ibogaine dose on psychological variables reflecting subjective mood state and a range of cognitive functions. Materials and methods: 21 healthy male volunteers received single 20mg doses of ibogaine after 6 days pretreatment with double-blind paroxetine or placebo. We compared responses to a battery of psychometric tests and subjective mood ratings performed before and 2h after ibogaine dosing, and assessed relationships between changes in test scores and concentrations of active moiety (the sum of molar noribogaine and ibogaine concentrations). Psychological tests were chosen based on responsiveness to opioid and serotonergic ligands. Results: Ibogaine had minimal influence on psychological tests and mood ratings. The ability to selectively ignore distracting spatial information showed some evidence of modulation; however because this effect was limited to the less challenging condition calls into question the reliability of this result. Conclusion: We were unable to identify stimulant effects after single 20mg doses of ibogaine. Future research is needed to confirm whether active moiety concentrations impact selective attention abilities while leaving other cognitive functions and mood state unaffected.
Article
Ibogaine is a psychoactive substance that may reduce opioid withdrawal symptoms. This was the first clinical trial of noribogaine, ibogaine's active metabolite, in patients established on methadone opioid substitution therapy (OST). In this randomized, double-blind, placebo-controlled, single ascending dose study, we evaluated the safety, tolerability, and pharmacokinetics of noribogaine in 27 patients seeking to discontinue methadone OST, who had been switched to morphine during the previous week. Noribogaine doses were 60, 120 or 180mg (n = 6/dose level) or matching placebo (n = 3/dose level). Noribogaine was well tolerated. The most frequent treatment-emergent adverse events were non-euphoric changes in light perception at ∼1h post dose, headache and nausea. Noribogaine had dose-linear increases for AUC and Cmax, and was slowly eliminated (mean t1/2 range 24–30h). There was a concentration-dependent increase in QTcI (0.17msec/ng/mL) with largest observed mean effect of ∼16msec, 28msec, and 42msec in the 60mg, 120mg, and 180mg groups, respectively. Noribogaine showed a non-statistically significant trend to decrease total scores in opioid withdrawal ratings, most notably at the 120mg dose, however the study design may have confounded evaluations of time to resumption of OST. Future exposure-controlled multiple-dose noribogaine studies are planned that will address these safety and design issues. This article is protected by copyright. All rights reserved