ArticlePDF AvailableLiterature Review

Dronabinol for chemotherapy-induced nausea and vomiting unresponsive to antiemetics

Taylor & Francis
Cancer Management and Research
Authors:

Abstract

Chemotherapy-induced nausea and vomiting (CINV) is one of the most common symptoms feared by patients, but may be prevented or lessened with appropriate medications. Several antiemetic options exist to manage CINV. Corticosteroids, serotonin receptor antagonists, and neurokinin receptor antagonists are the classes most commonly used in the prevention of CINV. There are many alternative drug classes utilized for the prevention and management of CINV such as antihistamines, benzodiazepines, anticonvulsants, cannabinoids, and dopamine receptor antagonists. Medications belonging to these classes generally have lower efficacy and are associated with more adverse effects. They are also not as well studied compared to the aforementioned agents. This review will focus on dronabinol, a member of the cannabinoid class, and its role in CINV. Cannabis sativa L. (also known as marijuana) contains naturally occurring delta-9-tetrahydrocannibinol (delta-9-THC). The synthetic version of delta-9-THC is the active ingredient in dronabinol that makes dronabinol an orally active cannabinoid. Evidence for clinical efficacy of dronabinol will be analyzed in this review as monotherapy, in combination with ondansetron, and in combination with prochlorperazine.
© 2016 May and Glode. This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.
php and incorporate the Creative Commons Attribution – Non Commercial (unported, v3.0) License (http://creativecommons.org/licenses/by-nc/3.0/). By accessing the work
you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For
permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms (https://www.dovepress.com/terms.php).
Cancer Management and Research 2016:8 49–55
Cancer Management and Research Dovepress
submit your manuscript | www.dovepress.com
Dovepress 49
REVIEW
open access to scientific and medical research
Open Access Full Text Article
http://dx.doi.org/10.2147/CMAR.S81425
Dronabinol for chemotherapy-induced nausea and
vomiting unresponsive to antiemetics
Megan Brafford May1
Ashley E Glode2
1Department of Pharmacy, Baptist
Health Lexington, Lexington, KY, USA;
2Department of Clinical Pharmacy,
Skaggs School of Pharmacy and
Pharmaceutical Sciences, University of
Colorado Anschutz Medical Campus,
Aurora, CO, USA
Abstract: Chemotherapy-induced nausea and vomiting (CINV) is one of the most common
symptoms feared by patients, but may be prevented or lessened with appropriate medications.
Several antiemetic options exist to manage CINV. Corticosteroids, serotonin receptor antagonists,
and neurokinin receptor antagonists are the classes most commonly used in the prevention of
CINV. There are many alternative drug classes utilized for the prevention and management of
CINV such as antihistamines, benzodiazepines, anticonvulsants, cannabinoids, and dopamine
receptor antagonists. Medications belonging to these classes generally have lower efficacy and
are associated with more adverse effects. They are also not as well studied compared to the
aforementioned agents. This review will focus on dronabinol, a member of the cannabinoid
class, and its role in CINV. Cannabis sativa L. (also known as marijuana) contains naturally
occurring delta-9-tetrahydrocannibinol (delta-9-THC). The synthetic version of delta-9-THC is
the active ingredient in dronabinol that makes dronabinol an orally active cannabinoid. Evidence
for clinical efficacy of dronabinol will be analyzed in this review as monotherapy, in combina-
tion with ondansetron, and in combination with prochlorperazine.
Keywords: dronabinol, cannabinoids, antiemetic, chemotherapy-induced nausea and vomiting
Introduction
Chemotherapy-induced nausea and vomiting (CINV) is one of the most common
symptoms feared by patients, but may be prevented or lessened with appropriate
medications.1 CINV is categorized as acute, delayed, anticipatory, breakthrough,
or refractory.2 Acute-onset CINV may occur within a few minutes to several hours
after treatment administration and typically resolves within the first 24 hours. Risk
factors for the development of this kind of nausea include female sex, age <50
years, environment of administration, lack of history of chronic alcoholism, his-
tory of motion sickness, previous incidence of nausea and vomiting, emetogenicity
of agent(s) administered, dose of emetogenic agent(s), infusion rate, duration of
therapy, number of cycles, and efficacy of preventive antiemetic regimen. Delayed-
onset CINV generally occurs in patients more than 24 hours after medication
administration and may last 6–7 days. This type of nausea unfortunately tends to be
more common, severe, and treatment resistant. Factors impacting the incidence of
delayed CINV include dose and emetogenicity of chemotherapy agent(s), incidence
of acute CINV, patient age, sex, and prophylactic antiemetics used. Anticipatory
nausea and/or vomiting is considered a conditioned response and may occur after a
negative prior experience with chemotherapy. This happens before a patient receives
Correspondence: Megan Brafford May
Baptist Health Lexington, 1740
Nicholasville Road, Lexington, KY,
40503, USA
Email megan.may@bhsi.com
Journal name: Cancer Management and Research
Article Designation: REVIEW
Year: 2016
Volume: 8
Running head verso: May and Glode
Running head recto: Dronabinol for CINV unresponsive to antiemetics
DOI: http://dx.doi.org/10.2147/CMAR.S81425
This article was published in the following Dove Press journal:
Cancer Management and Research
12 May 2016
Number of times this article has been viewed
Cancer Management and Research 2016:8
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
50
May and Glode
their next cycle of treatment. This type of CINV is more
common in younger patients due to the aggressive nature
of treatment. Breakthrough emesis is considered vomiting
that occurs even with prophylactic treatment and/or requires
the addition of “rescue” antiemetics. Refractory emesis is
classified as vomiting that occurs with subsequent treatment
when prophylactic antiemetics and/or rescue medications
have been ineffective in previous cycles.
CINV may have a significant negative impact on a
patient’s quality of life (QoL) as well as lead to increased
indirect and direct costs. When a patient’s QoL is impacted
in a negative manner, it may result in poor compliance
with future treatment. The development of CINV has been
reported to have a significant impact on QoL in 55%–60%
of chemotherapy cycles administered to patients.1 The inten-
sity of CINV and duration of CINV were the most notable
factors linked to a more significant influence on QoL. In a
study analyzing cost associated with the development of
CINV, it was noted that the average cost was nearly $800 per
patient for the first 5 days of the first cycle of chemotherapy.3
The cost analysis included overall direct and indirect costs:
direct medical, missed work, productivity loss, and cost of
any antiemetic medications taken on the day of treatment.
Indirect costs were greater for patients with more severe
CINV due to work absence for longer periods of time and
reduced productivity. Additional undesirable effects of CINV
to consider include metabolic imbalances, decline in self-care
and functional ability, nutrient depletion, anorexia, worsen-
ing of the patient’s performance status and mental status,
wound dehiscence, esophageal tears, and withdrawal from
potentially useful or curative treatment.2
It is reported that chemotherapy-induced vomiting can
be prevented in more than two-thirds of patients if antiemet-
ics are used correctly.1 Nausea remains difficult to prevent
and manage and is typically more commonly reported than
emesis: 54.9% versus 45.1% at any cycle. Several treatment
guidelines exist published by reliable institutions providing
recommendations to optimize CINV prevention and manage-
ment, yet CINV remains an issue. In a recent report, despite
prophylactic antiemetics, approximately 60% of patients who
received moderately and highly emetogenic chemotherapy
regimens still experienced some form of CINV; delayed being
more common (58% versus 34%).3 Improvements in CINV
management are needed.
Current antiemetic treatment
Several antiemetic options exist to manage CINV. Cortico-
steroids, serotonin receptor antagonists (5-HT3 RAs), and
neurokinin receptor antagonists (NK1 RAs) are the classes
most commonly used in the prevention of CINV.
Corticosteroids have been utilized as an effective anti-
emetic for over 30 years, with dexamethasone being the
most common agent.4 This class may be used for acute and
delayed CINV and are effective when given in combination
for prevention of CINV in moderately and highly emetogenic
regimens. A meta-analysis of over 5,000 patients receiving
highly or moderately emetogenic chemotherapy assessed
the efficacy of dexamethasone as prophylaxis for CINV. In
the majority of trials included, dexamethasone was given
in combination with other antiemetics, such as 5-HT3 RAs,
or metoclopramide. Dexamethasone was determined to be
superior to placebo or no treatment in terms of complete
protection (no vomiting or retching) for both acute (odds
ratio: 2.22, 95% confidence interval: 1.89–2.60) and delayed
(odds ratio: 2.04, 95% confidence interval: 1.63–2.56) vom-
iting. Corticosteroids are usually well tolerated when used
as a short-term antiemetic. Moderate-to-severe insomnia
(45%), gastrointestinal discomfort (27%), agitation (27%),
increased appetite (19%), weight gain (16%), and acne (15%)
have been reported by patients taking dexamethasone for the
prevention of delayed CINV.
Selective 5-HT3 RAs have been incorporated into the
management of CINV for over the last 20 years.4 In the
United States, the following agents are approved: dolasetron,
granisetron, ondansetron, and palonosetron, with palonose-
tron having a significantly longer half-life, making it excep-
tionally useful in the prevention of delayed CINV. Various
studies have shown this class of agents to be effective in the
prevention of acute and delayed CINV. One meta-analysis
including ten studies revealed an 8.2% absolute risk reduction
compared to placebo for the development of delayed CINV.
One of the more notable adverse effects of this class is the
potential development of electrocardiogram abnormalities,
including QT prolongation. Additional side effects reported
with these medications include headache, constipation, and
abdominal pain.
One of the newer classes of medications approved for the
prevention of CINV is the NK1 RAs. Agents from this class
should be administered in combination with dexamethasone
and a 5-HT3 RA to prevent acute and delayed CINV associ-
ated with moderately and highly emetogenic chemotherapy.2
A meta-analysis including nearly 9,000 patients receiving
moderately and highly emetogenic chemotherapy expe-
rienced a significant improvement in CINV in the acute,
delayed, and overall phases (P<0.001) when NK1 RAs were
added to 5-HT3 RAs and corticosteroids.4 Medications from
Cancer Management and Research 2016:8 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
51
Dronabinol for CINV unresponsive to antiemetics
this class tend to be well tolerated with minimal side effects.
It is important to note the potential for drug interactions
with medications metabolized through the cytochrome P450
enzyme system.
Olanzapine is the latest unique agent to be added to the
treatment guidelines.2 Olanzapine works on several neu-
rotransmitters involved in the development of CINV such
as dopamine, serotonin, histamine, and acetylcholine.4 Data
support its role for the prevention of acute and delayed CINV
as well as for the management of breakthrough CINV. Caution
should be employed when using this agent in elderly patients
with dementia-related psychosis as it may place them at an
increased risk of death.
There are many alternative drug classes utilized for the
prevention and management of CINV such as antihistamines,
benzodiazepines, anticonvulsants, cannabinoids, and dopa-
mine receptor antagonists. Medications belonging to these
classes generally have lower efficacy and are associated
with more adverse effects. They are also not as well studied
compared to the agents mentioned above. The remainder of
this review will focus on dronabinol, a member of the can-
nabinoid class, and its role in CINV.
Pharmacology of dronabinol
Cannabis sativa L. (also known as marijuana) contains
naturally occurring delta-9-tetrahydrocannibinol (delta-9-
THC). The synthetic version of delta-9-THC is the active
ingredient in dronabinol that makes dronabinol an orally
active cannabinoid.
There are at least two types of cannabinoid receptors, CB1
and CB2.5 CB1 receptors are located throughout the central
nervous system, whereas CB2 receptors are present on the
brainstem neurons but mostly concentrated in the periphery,
primarily on immunocytes and mast cells.6 These receptors
can be activated not only by cannabis-derived and synthetic
agonists, but also by endogenous cannabinoids produced in
mammalian tissues.5 The mediating effects of dronabinol
and other cannabinoids occur through these cannabinoid
receptors located in neural tissues.7
Dronabinol has an onset of action of approximately 0.5–1
hour, with a peak effect at 2–4 hours, lasting a total of 4–6
hours with the psychoactive effects. After a single dose of
dronabinol, 90%–95% of the medication is systemically
absorbed; however, only 10%–20% enters the systemic cir-
culation due to the high lipid solubility and first-pass hepatic
metabolism. Dronabinol has a large volume of distribution,
approximately 10 L/kg, which allows for the metabolites to
be released over a prolonged period of time at low levels. It
undergoes first-pass hepatic metabolism, leading to active
and inactive metabolites. The clearance for dronabinol varies
greatly, with an average of 0.2 L/kg/h. Dronabinol metabolites
have been detected after a single dose more than 5 weeks
after administration in the urine and feces. The major route
of elimination for dronabinol is through the feces, with
approximately 35%–50% removed by this route; however,
about 10%–15% is found in the urine.7
Evidence for clinical efcacy of
dronabinol in CINV
Dronabinol was approved by the US Food and Drug Admin-
istration (FDA) in 1985 for the treatment of CINV in patients
who have failed to respond adequately to conventional anti-
emetic treatment.7
The endogenous cannabinoid system is an important
pathway involved in the emetic response. Cannabinoids
can prevent chemotherapy-induced emesis by acting at
central CB1 receptors by preventing the proemetic effects of
endogenous compounds such as dopamine and serotonin.8 In
addition, by acting as an agonist to CB1, cannabinoids used
as a treatment results in an antiemetic effect.9 Cannabinoids
have been used effectively for treating CINV since 1985.10
Monotherapy
A meta-analysis evaluated a total of 1,366 patients with 30
randomized clinical trials between 1975 and 1996.10 Three dif-
ferent cannabinoids were evaluated among the 30 trials, with 16
trials investigating nabilone, 13 investigating dronabinol, and
one investigating intramuscular levonantradol. The controls
included prochlorperazine, metoclopramide, chlorproma-
zine, thiethylperazine, haloperidol, domperidone, alizapride,
and placebo. The data showed that cannabinoids were more
effective with completely controlling CINV than the active
comparators or placebo in all the trials (number needed to
treat [NNT] =6 for nausea and NNT =8 for nausea). In patients
receiving a low or highly emetogenic chemotherapy regimen,
cannabinoids were similar in efficacy of complete control of
CINV versus the control; however, in patients receiving mod-
erate emetogenic risk chemotherapy regimens, cannabinoids
performed better than the control.
The three cannabinoids resulted in greater adverse drug
effects compared to the controls.10 There was a significant
increase in the number of patients who withdrew from the
studies due to intolerable adverse drug effects in 19 of the
30 trials. The increased adverse drug effects were described
as being beneficial or harmful. The beneficial adverse effects
Cancer Management and Research 2016:8
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
52
May and Glode
included sensation of a “high”, euphoria, drowsiness, seda-
tion, and somnolence. The harmful adverse effects included
dysphoria, depression, hallucination, and paranoia. Can-
nabinoids also increased the risk of arterial hypotension
(>20% decrease in blood pressure from baseline) in patients.
This meta-analysis showed that one in eleven patients would
stop chemotherapy treatment if taking a cannabinoid for
antiemesis compared to no patients discontinuing treatment
if prescribed another antiemetic.
Rocha et al’s11 meta-analysis evaluated 13 randomized
clinical trials. Five trials included dronabinol, six included
nabilone, and two included levonantradol. Ten of the trials
used prochlorperazine as a comparator, with the remain-
ing using alizapride, chlorpromazine, or domperidone.
Dronabinol’s ability for antiemetic efficacy was determined
to show statistical significance over the comparator (P=0.03,
NNT =3.4). The difference in antiemetic efficacy with nabi-
lone or levonantradol compared to controls was not statisti-
cally significant (P=0.21 and P=0.60, respectively).
Patients in the cannabinoids group reported paranoid
delusions, hallucinations, dysphoria, and depression.11 These
adverse drug effects occurred exclusively in the cannabinoids
group. Other adverse drug effects such as a “high” sensation,
sleepiness, sedation, and euphoria occurred more frequently
and more intensely in the cannabinoids group. However,
only 30% of the 400 patient dropouts were due to toxicities.
Dronabinol in combination with
ondansetron
Meiri et al8 conducted a randomized, double-blind, placebo-
controlled, parallel group trial that enrolled 64 patients receiv-
ing moderate or highly emetic chemotherapy.8 The objective
of this study was to determine if 4 days after chemotherapy,
adding dronabinol to a prophylactic regimen for acute CINV
and continuing treatment either alone or in combination with
ondansetron can help prevent delayed CINV. The four parallel
groups were dronabinol monotherapy (D; n=17), ondansetron
monotherapy (O; n=17), combination with dronabinol and
ondansetron (D + O; n=16), and placebo (n=14). Patients
in all four groups received dexamethasone 20 mg PO (by
mouth) and ondansetron 16 mg intravenous prechemotherapy
on day 1. In the three active treatment groups, patients also
received dronabinol 2.5 mg PO both prechemotherapy and
postchemotherapy on day 1. Day 2 consisted of fixed doses
with dronabinol 2.5 mg PO four times daily and/or ondanse-
tron 8 mg PO twice daily in the respective groups. Days 3–5
were flexible dosing days, with patients being allowed to take
dronabinol 2.5–5 mg PO four times a day and/or ondansetron
4–8 mg PO twice daily depending on tolerability. In the pla-
cebo group, a placebo was matched with the doses of dronabi-
nol and/or ondansetron. In all groups, patients were provided
with rescue medications consisting of metoclopramide PO,
prochlorperazine PO, and prochlorperazine suppository to
be used on days 1–8 for intolerable nausea and vomiting or
retching after the maximum prescribed study doses.
In this trial, there were more females (37 out of 61
patients, 61%) compared to males (24 out of 61 patients,
39%).8 The two most common cancer diagnoses were breast
cancer (26 out of 64 patients, 41%) and non-small-cell lung
cancer (14 out of 64 patients, 22%). Among all four groups,
29 patients (45%) took all of the appropriate study medica-
tions in the correct dosages over the 5-day trial period. A
total response was defined as no vomiting and/or retching,
intensity of nausea less than 5 mm on a 100 mm visual analog
scale (VAS), and no rescue medications. VAS is ranked from
0 to 100 mm, with 0 mm meaning no nausea and 100 mm
meaning intractable nausea. For day 1 results, the three active
treatment groups were combined and compared to the placebo
group. The total response during active treatment on day 1
for the combined active treatment (CAT) group was 79%
compared to 40% for the placebo group. For days 2–5, the
total response for the D, O, D + O, and placebo groups were
54%, 58%, 47%, and 20%, respectively. The percentage of
patients without nausea on day 1 in CAT group was 79% and
in the placebo group 38%. For days 2–5, the percentages were
71%, 64%, 53%, and 15% in the D, O, D + O, and placebo
groups, respectively. The reported VAS for nausea intensity
was 7.65 in the CAT group and 30.67 in the placebo group.
For days 2–5, the VAS was 10.1 in the D group, 24 in the
O group, 14.3 in the D + O group, and 48.4 in the placebo
group. Overall, the complete response rate was 62% in the
D group, 58% in the O group, 60% in the D + O group, and
20% in the placebo group. In addition, the active treatment
groups reduced the number of vomiting episodes to zero and
decreased the duration of vomiting and retching to 0 hours
by days 4 and 5. The duration of nausea was comparable
between all four groups. On the flexible dosage days, the
median dosage in the D group was 20 mg/d of dronabinol,
16 mg/d of ondansetron in the O group, and 17.5–20 mg/d
of dronabinol and 12–16 mg/d of ondansetron in the D + O
group. Rescue medications were used in all four groups (24%
in D, 31% in O, 12% in D + O, and 43% in placebo groups).
Treatment-emergent adverse drug effects were reported
in all four groups, with the largest percentage in the ondan-
setron group.8 Adverse drug effects were reported in 82% in
Cancer Management and Research 2016:8 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
53
Dronabinol for CINV unresponsive to antiemetics
the D group, 88% in the O group, 71% in the D + O group,
and 50% in the placebo group. The percentage of patients
who permanently discontinued a study medication because
of a treatment-emergent adverse drug effect was 6% in the
D group, 13% in the O group, 18% in the D + O group, and
no patients in the placebo group.
Dronabinol in combination with
prochlorperazine
Lane et al12 conducted a multicentered, randomized, paral-
lel group, double-blind trial that enrolled 62 patients. All
the patients had previously received chemotherapy and
antiemetics. Patients were eligible for the trial if they were
receiving any chemotherapy agents except high-dose cisplatin
(>60 mg/m2). The most common chemotherapy agents were
doxorubicin and cyclophosphamide (n=26), 5-fluorouracil
(n=14), vincristine (n=13), and etoposide (n=10). Two or three
drug chemotherapy regimens were given to 79% of patients.
Patients were randomized to one of three arms: dronabinol 10
mg PO every 6 hours with placebo (n=21), prochlorperazine
10 mg PO every 6 hours with placebo (n=21), or dronabinol
10 mg PO every 6 hours with prochlorperazine 10 mg PO
every 6 hours (n=20). Patients started the prescribed anti-
emetic 24 hours prior to starting chemotherapy and continued
it until 24 hours after the last dose of chemotherapy.
The overall complete response rate was defined as no
episodes of nausea or vomiting and occurred in 41% in the
dronabinol group and 30% in the prochlorperazine group
(P<0.0001).12 No complaints of nausea or vomiting occurred
in 41% in the dronabinol group, 30% in the prochlorperazine
group, and 47% in the combination group. Two or fewer
nausea or vomiting episodes occurred in 71%, 45%, and
65% in the dronabinol, prochlorperazine, and combination
groups, respectively. The median duration per episode of
nausea or vomiting was 2 minutes in the combination group
compared to 5 minutes in the dronabinol and prochlorpera-
zine groups (P<0.001). In addition, the severity of the nausea
was less in the combination group versus the dronabinol
and prochlorperazine groups (P<0.001). The duration per
episode of nausea or vomiting in the dronabinol group was
2 and 10 minutes versus 4 and 15 minutes in the prochlor-
perazine group. However, the total duration of nausea and
vomiting episodes did not differ between the three treatment
groups. Eleven patients reported anticipatory nausea (30%
in dronabinol group, 0% in the prochlorperazine group, and
26% in the combination group).
Thirty-four patients reported adverse drug effects in the
dronabinol (n=16), prochlorperazine (n=7), and combination
(n=11) groups.12 The difference of reported adverse drug
effects was statistically significant between the dronabinol
and prochlorperazine groups (P<0.01). Most of the adverse
drug effects reported were mild to moderate. The most
common type of adverse effects reported were neuropsycho-
tropic, seen in a total of 48% of patients. The incidence of
neuropsychotropic adverse drug effects that was reported in
the dronabinol and prochlorperazine groups was 62% versus
29% (P=0.06). In the combination group, it was reported in
55% of patients. Overall, 14 patients withdrew (ten patients
in dronabinol group and four in combination group) due to
adverse drug effects, with neuropsychotropic effects being
the most common reason and beginning during the preche-
motherapy phase.
Indicated dronabinol dosage
Dronabinol is typically prescribed at a dosage of 5 mg PO
three or four times daily to control CINV.7 On the basis of
the patient’s response after each chemotherapy cycle, the
dose may be increased or decreased as tolerated. Another
option for dosing dronabinol is 5 mg/m2 PO every 1–3 hours
prechemotherapy and then every 2–4 hours for a total of 4–6
doses/d. The maximum individual dose is 15 mg/m2.
Patient perspectives of dronabinol
CB1/CB2 receptor agonists can produce adverse effects in
patients, and many of these are likely caused by the activa-
tion of central CB1 receptors rather than CB2 or peripheral
CB1 receptors.5
In 18 cross-over trials included in the Tramer et al10 meta-
analysis, when patients were questioned which antiemetic
was preferred, 38%–90% of patients preferred cannabinoids.
Four trials compared cannabinoids to placebo. Out of 202
patients, 153 patients preferred cannabinoids versus 27
patients preferring the placebo (NNT =1.6). In ten additional
trials comparing cannabinoids to an active control, 371 out
of 604 (61%) preferred cannabinoids versus 156 patients
(26%) preferring the active control.
In the Rocha et al’s11 meta-analysis, 18 double-blind
and cross-over trials (n=1,138) included an analysis of the
patient’s preference of cannabinoids, and it resulted in a
statistically significant difference in favor of cannabinoids
(NNT =1.8, P<0.00001). The cannabinoids included in
the different trials included dronabinol, nabilone, and
levonantradol compared to the controls of prochlorperazine,
chlorpromazine, domperidone, haloperidol, alizapride, meto-
clopramide, and placebo. A relationship was determined with
the control drug that was utilized.
Cancer Management and Research 2016:8
submit your manuscript | www.dovepress.com
Dovepress
Dovepress
54
May and Glode
It is suggested that patients prefer the “beneficial” adverse
drug effects associated with dronabinol, such as, sedation,
sensation of a “high”, and somnolence.10,11 These adverse
drug effects might provide relief while receiving chemo-
therapy.11 Another view is since CINV has such a major
impact on the patient’s QoL and can cause such discomfort
to the patient, the patients prefer cannabinoids’ adverse drug
effects instead of the conventional medications that might be
less effective in preventing and relieving the CINV.11
Medical marijuana
Many states have legalized marijuana for medical use,
including the condition of cancer.13 The legal limit of the
amount allowed for a patient to possess varies by state.
Technically marijuana contains more than 60 pharma-
cologically active cannabinoids, with the primary active
cannabinoids being THC and cannabidiol. The therapeutic
effect depends on the concentration of THC in addition to
the THC-to-cannabidiol ratio. Various strains of marijuana
are engineered to have different concentrations and ratios to
achieve desired pharmacologic effects. Medical marijuana
may be purchased from dispensaries in various dosage forms
as well, and is most commonly dispensed in the form used
to smoke as “cigarettes” or with a water pipe.14 It may also
be inhaled through a vaporizer, eaten in food, or applied
topically as a lotion.
It is important to note that adverse effects on the cardio-
vascular, respiratory, and central nervous system have been
associated with marijuana use. Marijuana smoke has been
reported to contain more carcinogens than cigarette smoke,
and may lead to head and neck cancer, lung cancer, as well as
bronchitis.9 The development of atrial fibrillation, myocardial
infarction, stroke, drowsiness, dizziness, nightmares, diffi-
culty sleeping, acute toxic psychosis, anxiety, and depression
among many other adverse effects has been associated with
marijuana use.9,14 Another concern in cancer patients is the
reported immunosuppressive properties of marijuana.
Few studies have evaluated medical marijuana alone or
in combination to treat nausea and vomiting related to che-
motherapy. The published studies that have been conducted
have mixed results. One study of 15 patients receiving
adjuvant high-dose methotrexate for sarcoma were random-
ized to three paired trials of placebo-THC or THC-placebo
with the patients serving as their own control.15 THC was
administered as capsules and cigarettes or matching placebos.
Fourteen of the 15 patients reported a reduction in nausea and
vomiting when using THC. Another study conducted by the
same group in eight patients receiving adjuvant doxorubicin
and cyclophosphamide for sarcoma were randomized to
THC-placebo or placebo-THC in paired trials to serve as
their own control.16 THC was administered as oral capsules
and cigarettes compared to matching placebos. Although the
investigators used the same trial design and THC regimen, a
beneficial effect of THC was not seen in this patient popula-
tion. The lack of benefit in this trial is thought to potentially
be from lower THC concentrations achieved in this group of
patients and presence of anticipatory nausea and vomiting
in half of the patients.
The largest account of patients receiving marijuana for
CINV was published by Musty and Rossi in 2001.17 This
group compiled a report of state-run clinical trials that had
been conducted evaluating Cannabis sativa (smoked mari-
juana and/or delta-9-tetrahydrocannabinol capsules) efficacy
in reducing CINV. Six trials were included in the analysis
comprising 748 patients who smoked marijuana prior to and/
or after chemotherapy and 345 patients who used oral THC
capsules. For the patients who smoked marijuana, 70%–100%
experienced relief from nausea and vomiting, and 76%–88%
of those who ingested the oral THC capsules experienced
relief. Short-term side effects reported included sedation, a
“high”, and smoke intolerance. More studies are needed in
this area to evaluate the efficacy and safety as additional states
approve medical marijuana and more patients gain access to
this management option.
Future directions
Dronabinol is utilized for breakthrough CINV based upon
available evidence already discussed and recommendations
by major oncology guidelines. Dronabinol is currently
being investigated in a few studies for additional roles in the
management of CINV. A completed, not published study has
investigated the use of dronabinol versus standard ondanse-
tron antiemetic therapy in the prevention of delayed CINV or
retching after moderate-to-high emetogenic chemotherapy.18
Patients were randomized to ondansetron, dronabinol,
combination therapy, or placebo. The study outcome was to
measure response of nausea and vomiting/retching, intensity,
and use of rescue medication. An additional study completed,
but yet to be published, has investigated palonosetron and
dexamethasone with or without dronabinol in the preven-
tion of CINV with moderately emetogenic chemotherapy.19
Patients were assessed for protection against the develop-
ment of vomiting, nausea, and use of rescue therapy. In an
ongoing study of patients with primary glioma receiving
chemotherapy, dronabinol is being administered to assess
its tolerability, toxicity, and impact on QoL.20 Assessment
Cancer Management and Research 2016:8 submit your manuscript | www.dovepress.com
Dovepress
Dovepress
Cancer Management and Research
Publish your work in this journal
Submit your manuscript here: https://www.dovepress.com/cancer-management-and-research-journal
Cancer Management and Research is an international, peer-reviewed
open access journal focusing on cancer research and the optimal use of
preventative and integrated treatment interventions to achieve improved
outcomes, enhanced survival and quality of life for the cancer patient.
The manuscript management system is completely online and includes
a very quick and fair peer-review system, which is all easy to use. Visit
http://www.dovepress.com/testimonials.php to read real quotes from
published authors.
Dovepress
55
Dronabinol for CINV unresponsive to antiemetics
of emesis will be completed by patients filling out a daily
appetite and nausea/vomiting log as well as a QoL functional
living index emesis scale.
With medical marijuana remaining a schedule I substance
on a federal level, no studies are registered with clnicaltri-
als.gov to investigate its role in the management of CINV.
Anecdotally patients report benefit, but more research is
needed to identify the most appropriate dose, dosage form,
drug–drug interactions, and safety concerns with its use
before a role for medical marijuana can be elucidated in the
management of CINV.
Conclusion
Current guidelines as well as the FDA-approved indications
consider dronabinol’s role to be in the management of break-
through CINV. Dronabinol has a unique mechanism of action
and adverse effect profile that should be considered when
treating a patient with this medication. Unfortunately, there
are few ongoing studies evaluating the role of dronabinol in
the management of CINV. Two completed, yet unpublished,
studies have evaluated dronabinol in combination with a
5-HT3 RA as a prophylactic strategy. At this time, there is
insufficient data to support the routine use of dronabinol as an
antiemetic in all chemotherapeutic regimens. Data do support
the beneficial effects of dronabinol in the breakthrough CINV
setting. Further study of the scope of dronabinol’s potential
efficacy is warranted.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Fernandez-Ortega P, Caloto MT, Chirveches E, et al. Chemotherapy-
induced nausea and vomiting in clinical practice: impact on patient’s
quality of life. Support Care Cancer. 2012;20:3141–3148.
2. National Comprehensive Cancer Network. NCCN Clinical Practice
Guidelines in Oncology (NCCN Guidelines)®: Antiemesis Version 2.2015.
Fort Washington, PA: National Comprehensive Cancer Network; 2015.
3. Haiderali A, Menditto L, Good M, Teitelbaum A, Wegner J. Impact on
daily functioning and indirect/direct costs associated with chemotherapy-
induced nausea and vomiting (CINV) in a US population. Support Care
Cancer. 2011;19:843–851.
4. Natale J. Reviewing current and emerging antiemetics for chemo-
therapy-induced nausea and vomiting prophylaxis. Hosp Pract.
2015;43(4):226–234.
5. Pertwee RG. Emerging strategies for exploiting cannabinoid receptor
agonists as medicines. Br J Pharmacol. 2009;156:397–411.
6. Slatkin NE. Cannabinoids in the treatment of chemotherapy-induced
nausea and vomiting: beyond prevention of acute emesis. J Support
Oncol. 2007;5(Suppl 3):1–9.
7. Marinol(R) [package insert]. High Point, NC: Unimed Pharmaceuticals,
Inc; 2004.
8. Meiri E, Jhangiani H, Vredenburgh, et al. Efficacy of dronabinol alone
and in combination with ondansetron versus ondansetrom alone for
delayed chemotherapy-induced nausea and vomiting. Curr Med Res
Opin. 2007;23(3):533–543.
9. Todaro B. Cannabinoids in the treatment of chemotherapy-induced
nausea and vomiting. JNCCN. 2012;10(4):487–492.
10. Tramer MR, Carroll D, Campbell FA, Reynolds JM, Moore RA, McQuay
HJ. Cannabinoids for control of chemotherapy induced nausea and
vomiting: quantitative systemic review. BMJ. 2001;323:16–21.
11. Rocha FM, Stefano SC, De Cassia Haiek R, Oliveira LR, Silveira DD.
Therapeutic use of Cannabis sativa on chemotherapy-induced nausea
and vomiting among cancer patients: systematic review and meta-
analysis. Eur J Cancer Care. 2008;17:431–443.
12. Lane M, Vogel CL, Ferguson J, et al. Dronabinol and prochlorperazine
in combination for treatment of cancer chemotherapy-induced nausea
and vomiting. J Pain Symptom Manage. 1991;6(6):352–359.
13. Hill KP. Medical marijuana for treatment of chronic pain and
other medical and psychiatric problems. A clinical review. JAMA.
2015;313(24):2474–2483.
14. Seamon MJ, Fass JA, Maniscalco-Feichtl M, Abu-Shraie NA. Medical
marijuana and the developing role of the pharmacist. Am J Health Syst
Pharm. 2007;64:1037–1044.
15. Chang AE, Shiling DJ, Stillman RC, et al. Delta-9-tetrahydrocannabinol
as an antiemetic in cancer patients receiving high-dose methotrexate. A
prospective, randomized evaluation. Ann Intern Med. 1979;91:819–824.
16. Chang AE, Shiling DJ, Stillman RC, et al. A prospective evaluation
of delta-9-tetrahydrocannabinol as an antiemetic in patients receiving
adriamycin and cytoxan chemotherapy. Cancer. 1981;47:1746–1751.
17. Musty RE, Rossi, R. Effects of smoked cannabis and oral 9-trahydro-
cannabinol on nausea and emesis after cancer chemotherapy: A review
of state clinical trials. J Cannabis Therapeutics. 2001;1(1):29–56.
18. Solvay Pharmaceuticals. Dronabinol versus standard ondansetron anti-
emetic therapy in preventing delayed-onset chemotherapy-induced nausea
and vomiting. NLM identifier: NCT00642512. Available from: https://
clinicaltrials.gov/ct2/show/NCT00642512. Accessed April 13, 2016.
19. National Cancer Institute and Solvay Pharmaceuticals; M.D. Anderson
Cancer Center. Palonosetron and dexamethasone with or without dronabi-
nol in preventing nausea and vomiting in patients receiving chemotherapy
for cancer. NLM Identifier: NCT00553059. Available from: https://
clinicaltrials.gov/ct2/show/NCT00553059 . Accessed April 13, 2016.
20. Solvay Pharmaceuticals; Duke University. A pilot study of dronabinol
for adult patients with primary gliomas. NLM Identifier: NCT00314808.
Available from: http://clinicaltrials.gov/show/NCT00314808. Accessed
April 13, 2016.
... Dronabinol is synthetic tetrahydrocannabinol (THC) that has been US Food and Drug Administration (FDA) approved since 1985 to treat HIV/AIDS-induced anorexia and to treat patients who do not respond to conventional anti-emetics from the nausea and vomiting of chemotherapy [1,2]. Dronabinol activates the cannabinoid CB 1 receptor, stimulating appetite and provoking an anti-emetic response by reducing the emetic effects of endogenous neurochemicals like dopamine and serotonin [1,2]. ...
... Dronabinol is synthetic tetrahydrocannabinol (THC) that has been US Food and Drug Administration (FDA) approved since 1985 to treat HIV/AIDS-induced anorexia and to treat patients who do not respond to conventional anti-emetics from the nausea and vomiting of chemotherapy [1,2]. Dronabinol activates the cannabinoid CB 1 receptor, stimulating appetite and provoking an anti-emetic response by reducing the emetic effects of endogenous neurochemicals like dopamine and serotonin [1,2]. Over half of chemotherapy cycles adversely affected the quality of life for patients by inducing undesirable effects including nausea, vomiting, anorexia, nutrient depletion, and metabolic imbalances. ...
... Over half of chemotherapy cycles adversely affected the quality of life for patients by inducing undesirable effects including nausea, vomiting, anorexia, nutrient depletion, and metabolic imbalances. By using dronabinol alone or in combination with conventional anti-emetics like ondansetron and prochlorperazine, randomized clinical trials concluded that dronabinol helped prevent nausea and vomiting episodes [2]. Prescription THC activates the CB 1 to combat weight loss and wasting syndrome from HIV by increasing appetite [1]. ...
Article
Full-text available
Introduction: Dronabinol is approved in the USA for chemotherapy-induced nausea as well as vomiting and HIV-induced anorexia, while cannabidiol is primarily approved for childhood epileptic disorders Lennox-Gastaut and Dravet syndrome. The use pattern for these prescription cannabinoids in the USA is unknown. This study examined Medicaid claims for two FDA-approved prescription cannabinoids, dronabinol and cannabidiol, approved in 1985 and 2018, respectively, from 2016-2020 to better understand the pharmacoepidemiologic trends and distribution of these drugs in US Medicaid amidst the increasing use of non-pharmaceutical formulations of cannabis. Methods: The longitudinal study analyzed Medicaid prescription claims that were calculated by extracting the prescriptions on a state level from 2016 to 2020 for two cannabinoids, dronabinol and cannabidiol, where outcomes over each year were calculated. Outcomes were (1) the number of prescriptions for each state corrected for the number of Medicaid enrollees and (2) dronabinol and cannabidiol spending. Spending refers to the amount reimbursed by the state Medicaid program. Results: Dronabinol prescriptions per state decreased by 25.3% from 2016 to 2020, while cannabidiol prescriptions increased by 16,272.99% from 2018 to 2020. The spending on these drugs parallels that of their prescription trend with a 66.3% decrease in reimbursement for dronabinol (5.7millionin2020),whereascannabidiolincreasedby+26,582.05.7 million in 2020), whereas cannabidiol increased by +26,582.0% (233.3 million in 2020). Dronabinol prescriptions, when corrected for the number of enrollees, in Connecticut were 136.4 times larger than in New Mexico, and seventeen states had zero prescriptions. Idaho's prescriptions of cannabidiol (27.8/10,000 enrollees) were significantly elevated relative to the national average and were 15.4-fold higher than Washington, DC (1.8/10K enrollees). Conclusions: The prescriptions of pharmaceutical-grade tetrahydrocannabinol decreased while those of cannabidiol increased. This study also identified pronounced state-level variation in cannabinoid prescribing to Medicaid patients. State formularies and prescription drug list variation may contribute to the drug reimbursements in Medicaid, though further research is needed to identify the health policy or pharmacoeconomic origins of these disparities.
... However, both the FDA and EMA have approved the use of drugs that contain individual cannabinoids (Whiting et al., 2015). Some examples include Epidiolex, a purified form of CBD used for the treatment of seizures associated with Lennox-Gastaut syndrome or Dravet syndrome (Sekar and Pack, 2019) (FDA and EMA-approved) or Dronabinol (FDA-approved)-a synthetic THC used as an appetite stimulant in AIDS-related cachexia (Badowski and Yanful, 2018) and for alleviating chemotherapy-induced nausea and vomit in cancer patients (May and Glode, 2016). Another area of interest is the treatment of post-traumatic stress disorder (PTSD) in military personnel with THC or nabilone (Sholler et al., 2020). ...
... Oncology practice guidelines for clinicians, NCCN and ASCO include the indication of Nabilone and Dronabinol for nausea and vomiting refractory to conventional therapy. 2.5-5 mg oral solution Dronabinol should be used every 4-6 h 1-2 mg of Nabilone can be used twice daily as rescue therapy (May and Glode, 2016). ...
Article
Full-text available
Cannabis enjoyed a “golden age” as a medicinal product in the late 19th, early 20th century, but the increased risk of overdose and abuse led to its criminalization. However, the 21st century have witnessed a resurgence of interest and a large body of literature regarding the benefits of cannabinoids have emerged. As legalization and decriminalization have spread around the world, cancer patients are increasingly interested in the potential utility of cannabinoids. Although eager to discuss cannabis use with their oncologist, patients often find them to be reluctant, mainly because clinicians are still not convinced by the existing evidence-based data to guide their treatment plans. Physicians should prescribe cannabis only if a careful explanation can be provided and follow up response evaluation ensured, making it mandatory for them to be up to date with the positive and also negative aspects of the cannabis in the case of cancer patients. Consequently, this article aims to bring some clarifications to clinicians regarding the sometimes-confusing various nomenclature under which this plant is mentioned, current legislation and the existing evidence (both preclinical and clinical) for the utility of cannabinoids in cancer patients, for either palliation of the associated symptoms or even the potential antitumor effects that cannabinoids may have.
... One of the first recognized medicinal uses of cannabinoids was for treating chemotherapy-induced nausea and vomiting. 11 Cannabis and synthetic CB1 agonists in small amounts effectively reduce acute vomiting in humans, 85,86 suggesting ECS modulation of emesis. ...
Article
Full-text available
Background Classical antiemetics that target the serotonin system may not be effective in treating certain nausea and vomiting conditions like cyclic vomiting syndrome (CVS) and cannabinoid hyperemesis syndrome (CHS). As a result, there is a need for better therapies to manage the symptoms of these disorders, including nausea, vomiting, and anxiety. Cannabis is often used for its purported antiemetic and anxiolytic effects, given regulation of these processes by the endocannabinoid system (ECS). However, there is considerable evidence that cannabinoids can also produce nausea and vomiting and increase anxiety in certain instances, especially at higher doses. This paradoxical effect of cannabinoids on nausea, vomiting, and anxiety may be due to the dysregulation of the ECS, altering how it maintains these processes and contributing to the pathophysiology of CVS or CHS. Purpose The purpose of this review is to highlight the involvement of the ECS in the regulation of stress, nausea, and vomiting. We discuss how prolonged cannabis use, such as in the case of CHS or heightened stress, can dysregulate the ECS and affect its modulation of these functions. The review also examines the evidence for the roles of ECS and stress systems' dysfunction in CVS and CHS to better understand the underlying mechanisms of these conditions.
... Additionally, other medications such as NK-1 receptor antagonists and dopamine receptor antagonists may interact with cannabinoids. 21 Therefore, studies suggest the importance of ensuring proper dosing of these drugs when used alongside cannabis supplements, taking into account their potential interactions. Haloperidol, another antiemetic drug, relies on conversion by the enzyme CYP3A4. ...
Article
Full-text available
The utilization of cannabis in oncology presents a multifaceted approach to addressing the complex challenges associated with cancer treatment. With roots tracing back to ancient medicinal practices, cannabis contains numerous compounds, including cannabinoids like tetrahydrocannabinol (THC) and cannabidiol (CBD), that interact with the body's endocannabinoid system. These interactions offer potential therapeutic benefits, ranging from antiemetic and analgesic effects to the inhibition of cancer growth processes. While clinical evidence supports the efficacy of cannabinoids in managing symptoms and enhancing the quality of life for cancer patients, caution is warranted due to potential interactions with conventional medications. Understanding the mechanisms of action, optimal administration routes, and potential side effects is crucial for integrating cannabis into personalized cancer care plans. By navigating these considerations thoughtfully, medical practitioners can harness the therapeutic potential of cannabis to complement traditional cancer treatments and improve patient outcomes in the field of oncology.
... THC, the principal psychoactive constituent of Cannabis sativa, was recently licensed (i.e. dronabinol) for medical use to treat chemotherapy-induced nausea and vomiting [17,18]. This compound is a partial agonist for Cannabinoid receptor type 1 (CB1) and Cannabinoid receptor type 2 (CB2) but with a higher affinity for CB1. ...
Article
Full-text available
Cannabis has gained popularity in recent years as a substitute treatment for pain following the risks of typical treatments uncovered by the opioid crisis. The active ingredients frequently associated with pain-relieving effects are the phytocannabinoids Δ⁹-tetrahydrocannabinol (THC) and cannabidiol (CBD), but their effectiveness and mechanisms of action are still under research. In this study, we used Caenorhabditis elegans, an ideal model organism for the study of nociception that expresses mammal ortholog cannabinoid (NPR-19 and NPR-32) and vanilloid (OSM-9 and OCR-2) receptors. Here, we evaluated the antinociceptive activity of THC and CBD, identifying receptor targets and several metabolic pathways activated following exposure to these molecules. The thermal avoidance index was used to phenotype each tested C. elegans experimental group. The data revealed for the first time that THC and CBD decreases the nocifensive response of C. elegans to noxious heat (32–35 °C). The effect was reversed 6 h post- CBD exposure but not for THC. Further investigations using specific mutants revealed CBD and THC are targeting different systems, namely the vanilloid and cannabinoid systems, respectively. Proteomic analysis revealed differences following Reactome pathways and gene ontology biological process database enrichment analyses between CBD or THC-treated nematodes and provided insights into potential targets for future drug development.
... 4 Delayed phase is more common and severe than the other phases manner and is resistant to antiemetic treatment. 5 According to NCCN guidelines, standard antiemetic agents for patients receiving moderate and high emetogenic chemotherapy include 5-HT3 antagonists, dexamethasone, NK1 RAs, and olanzapine. However, NK1 RAs and olanzapine were added for patients receiving high emetic risk chemotherapy. ...
Article
Full-text available
Objective To evaluate the effects of tetrahydrocannabinol (THC):cannabinoid (CBD) (1:1) oil in reducing chemotherapy-induced nausea and vomiting (CINV) in gynecologic cancer patients who received moderate-to-high emetogenic chemotherapy. Material and Method This was a randomized, double-blinded, crossover and placebo-controlled trial. The study was conducted at the Gynecologic Oncology Units, Bhumibol Adulyadej Hospital (BAH), Royal Thai Air Force, Bangkok, Thailand, between August and November 2022. Participants had gynecologic cancer and received moderate-to-high emetogenic chemotherapy. Subjects were randomized and divided into two groups (A and B) based on the block of four randomization method. In the first cycle, groups A and B received THC:CBD extract oil 1:1 (TCEO) and placebo before chemotherapy administration. In the second cycle, groups A and B received placebo and TCEO before chemotherapy administration. Both groups received per protocol antiemetic medication during chemotherapy. Nausea score and side effects were recorded. Results A total of 60 cases were recruited. After exclusion, 54 cases were included in the study. The mean age of participants was 54.4 years. The mean body mass index (BMI) was 26.5 kg/m². Fifty-nine (21/54) percent cases were the advanced stages of cancer. The nausea score of TCEO and placebo groups were 2.11 and 2.99, respectively (P < 0.05). More than half of the participants (36/54) reported dizziness and sedation side effects. Dry mouth, confusion, anxiety, and palpitation of both groups were comparable. Conclusion The cannabinoid extract (THC:CBD) was an appropriate adjuvant agent to reduce CINV in patients with gynecologic cancer who received high-emetogenic chemotherapy. Dizziness and sedation were the major side effects.
Article
Full-text available
Purpose of Review This document critically examines the role of cannabinoids in cancer care during an era marked by rapid advancements in oncology and changing perceptions on cannabis. It traces the historical context of cannabis in medicinal use, navigating its journey from widespread acceptance, subsequent criminalization, to its resurgence in modern therapeutic applications, particularly within the framework of Evidence-Based Medicine (EBM). Recent Findings Anchored in EBM principles, this study synthesizes current research from clinical trials, systematic reviews, and meta-analyses to evaluate the efficacy and safety of cannabinoids in oncology. The focus is on their palliative effects, considering the nuances of effectiveness, risk assessment, and challenges inherent in translating these findings into clinical guidelines. Summary The study seeks to bridge the gap between scientific research and clinical practice, offering insights to inform future oncological therapies and symptom management strategies involving cannabinoids. The potential benefits and risks of cannabinoid use in cancer treatment are assessed to guide clinicians and researchers in developing comprehensive, evidence-based approaches to patient care.
Article
Full-text available
Application of nitrogenous fertilizer to agricultural fields leads to excess availability of urea in a shorter period which is not only a wasteful expenditure but is also degradative to the...
Article
Full-text available
The ‘entourage effect’ term was originally coined in a pre-clinical study observing endogenous bio-inactive metabolites potentiating the activity of a bioactive endocannabinoid. As a hypothetical afterthought, this was proposed to hold general relevance to the usage of products based on Cannabis sativa L. The term was later juxtaposed to polypharmacy pertaining to full-spectrum medicinal Cannabis products exerting an overall higher effect than the single compounds. Since the emergence of the term, a discussion of its pharmacological foundation and relevance has been ongoing. Advocates suggest that the ‘entourage effect’ is the reason many patients experience an overall better effect from full-spectrum products. Critics state that the term is unfounded and used primarily for marketing purposes in the Cannabis industry. This scoping review aims to segregate the primary research claiming as well as disputing the existence of the ‘entourage effect’ from a pharmacological perspective. The literature on this topic is in its infancy. Existing pre-clinical and clinical studies are in general based on simplistic methodologies and show contradictory findings, with the clinical data mostly relying on anecdotal and real-world evidence. We propose that the ‘entourage effect’ is explained by traditional pharmacological terms pertaining to other plant-based medicinal products and polypharmacy in general (e.g., synergistic interactions and bioenhancement).
Article
Full-text available
This paper aims to evaluate the anti-emetic efficacy of cannabinoids in cancer patients receiving chemotherapy using a systematic review of literature searched within electronic databases such as PUBMED, EMBASE, PSYCINFO, LILACS, and ‘The Cochrane Collaboration Controlled Trials Register’. Studies chosen were randomized clinical trials comprising all publications of each database until December 2006. From 12 749 initially identified papers, 30 fulfilled the inclusion criteria for this review, with demonstration of superiority of the anti-emetic efficacy of cannabinoids compared with conventional drugs and placebo. The adverse effects were more intense and occurred more often among patients who used cannabinoids. Five meta-analyses were carried out: (1) dronabinol versus placebo [n = 185; relative risk (RR) = 0.47; confidence interval (CI) = 0.19–1.16]; (2) Dronabinol versus neuroleptics [n = 325; RR = 0.67; CI = 0.47–0.96; number needed to treat (NNT) = 3.4]; (3) nabilone versus neuroleptics (n = 277; RR = 0.88; CI = 0.72–1.08); (4) levonantradol versus neuroleptics (n = 194; RR = 0.94; CI = 0.75–1.18); and (5) patients' preference for cannabis or other drugs (n = 1138; RR = 0.33; CI = 0.24–0.44; NNT = 1.8). The superiority of the anti-emetic efficacy of cannabinoids was demonstrated through meta-analysis.
Article
Full-text available
Chemotherapy-induced nausea and vomiting (CINV) in cancer patients are common symptoms most feared by patients. The aim of this study was to analyze the impact of CINV associated to moderate/highly emetogenous chemotherapy regimens on patients' quality of life (QoL). Open, multicenter, prospective observational study was performed. Each patient filled out a patient diary for each cycle from the day before chemotherapy and for the next 5 days that included the number of emetic episodes, the intensity of nausea, and QoL evaluation (functional living index-emesis questionnaire). Data from 202 consecutive patients from nine university hospitals were collected, but only data from 160 were analyzed (79.2 %). Most of the participants (70 %) were women with a mean age of 50 years (SD 1.2 years). The most frequent cancer site was breast (44 %) followed by lung (16 %) and 76.3 % were receiving highly emetogenous chemotherapy. Despite the use of antiemetic prophylaxis, patients experienced significant nausea and vomiting during 31 % (3.2 % during acute, 15.0 % during delayed phase, and 13.2 % during both phases) and 45.1 % (5.1 % only during the acute phase, 23.5 % only during the delayed phase and 16.5 % during both phases) of the cycles, respectively, having 44.5 % (nausea) and 39.3 % (emesis) of the cycles an impact on patients' QoL. The results of the study confirm the detrimental effect of CINV on patients' QoL despite the use of antiemetic prophylaxis (5HT(3) receptor antagonist, steroids, and dopamine receptor antagonists). It is mandatory to intensify the detection of CINV in order to improve the management of these important, albeit frequent, side effects of cancer treatments.
Article
This review provides background information on chemotherapy-induced nausea and vomiting (CINV) classification and pathophysiology and reviews various antiemetic agents for CINV prophylaxis, including corticosteroids, serotonin receptor antagonists (5-HT3 RAs), tachykinin NK1 receptor antagonists (NK1 RAs), and olanzapine. Other less commonly used agents are briefly discussed. Practical considerations are reviewed as well, including emetogenicity of chemotherapeutic regimens, patient-specific risk factors for CINV, principles of CINV management, health economics outcome research, and quality of life. Available data on the newly FDA-approved antiemetic combination netupitant/palonosetron (NEPA) is also reviewed. Prevention of CINV is an important goal in managing patients with cancer and is especially difficult with respect to nausea and delayed CINV. Corticosteroids are a mainstay of CINV prophylaxis and are usually given in combination with other therapies. The 5-HT3 RA palonosetron has shown increased efficacy over other agents in the same class for prevention of delayed emesis with moderately emetogenic chemotherapy and NK1 RAs improve emesis prevention in combination with 5-HT3 RAs and dexamethasone. Olanzapine has shown efficacy for CINV prophylaxis and the treatment of breakthrough CINV. The new combination therapy, NEPA, has been shown to be efficacious for the prevention of acute, delayed, and overall CINV. Risk factors that have been identified for CINV include gender, age, and alcohol intake. It is important to assess the emetogenicity of chemotherapy regimens as well as the potential impact of patient risk factors in order to provide adequate prophylaxis. Acute and delayed CINV are severe, burdensome side effects of chemotherapy; however, new data on prevention and the discovery of new agents can further improve CINV control.
Article
As of March 2015, 23 states and the District of Columbia had medical marijuana laws in place. Physicians should know both the scientific rationale and the practical implications for medical marijuana laws. To review the pharmacology, indications, and laws related to medical marijuana use. The medical literature on medical marijuana was reviewed from 1948 to March 2015 via MEDLINE with an emphasis on 28 randomized clinical trials of cannabinoids as pharmacotherapy for indications other than those for which there are 2 US Food and Drug Administration-approved cannabinoids (dronabinol and nabilone), which include nausea and vomiting associated with chemotherapy and appetite stimulation in wasting illnesses. Use of marijuana for chronic pain, neuropathic pain, and spasticity due to multiple sclerosis is supported by high-quality evidence. Six trials that included 325 patients examined chronic pain, 6 trials that included 396 patients investigated neuropathic pain, and 12 trials that included 1600 patients focused on multiple sclerosis. Several of these trials had positive results, suggesting that marijuana or cannabinoids may be efficacious for these indications. Medical marijuana is used to treat a host of indications, a few of which have evidence to support treatment with marijuana and many that do not. Physicians should educate patients about medical marijuana to ensure that it is used appropriately and that patients will benefit from its use.
Article
Background. In 1999 the Institute of Medicine (IOM) issued a report entitled Marijuana and Medicine (Joy, Watson and Benson, 1999). It recommended the development of cannabinoid drug delivery systems which might be effective for nausea, vomiting and AIDS wasting syndrome, among other chronic disorders. The report went on to recognize that patients should be allowed to smoke marijua- na if they failed to achieve relief from approved symptoms that could be relieved by cannabinoid drugs with rapid onset. Recommended criteria of the report included: access to marijuana within 24 hours of submission by a physician, supervision that allows for assessment of treatment effectiveness, and an oversight strategy comparable to an institutional review board. In this context a review of previously un- published state-run clinical trials with Cannabis sativa (marijuana and/ or ∆9-tetrahydrocannabinol capsules) to test efficacy in reducing nau- sea and vomiting following cancer chemotherapy is warranted. The impetus for these studies came from individual state legislatures re- sponding to constituents' claims that smoking marijuana reduced or blocked nausea and vomiting. Methods. Technical reports were obtained from 6 states which had
Article
Before the introduction of the serotonin receptor antagonists (5-HT3 receptor antagonists) in the early 1990s, limited effective options were available to prevent and treat chemotherapy-induced nausea and vomiting (CINV). In 1985, the FDA approved 2 cannabinoid derivatives, dronabinol and nabilone, for the treatment of CINV not effectively treated by other agents. Today, the standard of care for prevention of CINV for highly and moderately emetogenic chemotherapy is a 5-HT3 receptor antagonist, dexamethasone, with or without aprepitant or fosaprepitant. With the approval of safer and more effective agents, cannabinoids are not recommended as first-line treatment for the prevention of CINV and are reserved for patients with breakthrough nausea and vomiting. Because of medical and legal concerns, the use of marijuana is not recommended for management of CINV and is not part of the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Antiemesis. Although patients may like to pursue this treatment option in states that have approved the use of marijuana for medical purposes, its use remains legally and therapeutically controversial.
Article
Chemotherapy-induced nausea and vomiting (CINV) is a debilitating side effect of chemotherapy, but it may be prevented or mitigated with medications. Uncontrolled CINV can lead to reduced quality of life and can result in increased costs (due to health care utilization and missed work). We prospectively assessed the prevalence and burden of CINV in a US population. Final analysis was performed on 178 patients, beginning chemotherapy during 2007-2008 at oncology specialty settings. Patients kept a diary recording use of antiemetic medications just before the start of chemotherapy and use of antiemetic medications, health care resources, and episodes of nausea and vomiting during the 5 days following. In addition, they completed a Functional Living Index-Emesis (FLIE) questionnaire and a Work Productivity and Assessment Inventory-Nausea and Vomiting assessment, to determine the impact of CINV on daily functioning and on work productivity, respectively. Physicians independently recorded prescribed medications and health care utilization. Of the patients, 61.2% reported experiencing CINV (34.3% with acute CINV and 58.4% with delayed CINV). Based on the FLIE assessment, 37.2% of all patients reported reduced daily functioning, and of those with poorly managed CINV, about 90% reported a significant impact on daily functioning. Total costs due to CINV were on average $778.58 per patient from the day of administration through the 5 days following the first cycle of chemotherapy; patients with more severe CINV typically had higher costs. CINV remains a significant problem among US patients, suggesting a need for more effective prophylaxis use in clinical practice.
Article
Mandarin translation of abstract Medicines that activate cannabinoid CB 1 and CB 2 receptor are already in the clinic. These are Cesamet ® (nabilone), Marinol ® (dronabinol; Δ ⁹ ‐tetrahydrocannabinol) and Sativex ® (Δ ⁹ ‐tetrahydrocannabinol with cannabidiol). The first two of these medicines can be prescribed to reduce chemotherapy‐induced nausea and vomiting. Marinol ® can also be prescribed to stimulate appetite, while Sativex ® is prescribed for the symptomatic relief of neuropathic pain in adults with multiple sclerosis and as an adjunctive analgesic treatment for adult patients with advanced cancer. One challenge now is to identify additional therapeutic targets for cannabinoid receptor agonists, and a number of potential clinical applications for such agonists are mentioned in this review. A second challenge is to develop strategies that will improve the efficacy and/or the benefit‐to‐risk ratio of a cannabinoid receptor agonist. This review focuses on five strategies that have the potential to meet either or both of these objectives. These are strategies that involve: (i) targeting cannabinoid receptors located outside the blood‐brain barrier; (ii) targeting cannabinoid receptors expressed by a particular tissue; (iii) targeting up‐regulated cannabinoid receptors; (iv) targeting cannabinoid CB 2 receptors; or (v) ‘multi‐targeting’. Preclinical data that justify additional research directed at evaluating the clinical importance of each of these strategies are also discussed. Mandarin translation of abstract
Article
Fifteen patients with osteogenic sarcoma receiving high-dose methotrexate chemotherapy were studied in a randomized, double-blind, placebo-controlled trial of oral and smoked delta-9-tetrahydrocannabinol (THC) as an antiemetic. Each patient served as his or her own control. Fourteen of 15 patients had a reduction in nausea and vomiting on THC as compared to placebo. Delta-9-tetrahydrocannabinol was significantly more effective than placebo in reducing the number of vomiting and retching episodes, degree of nausea, duration of nausea, and volume of emesis (P less than 0.001). There was a 72% incidence of nausea and vomiting on placebo. When plasma THC concentrations measured less than 5.0 ng/mL, 5.0 to 10.0 ng/mL, and greater than 10.0 ng/mL, the incidences of nausea and vomiting were 44%, 21%, and 6%, respectively. Delta-9-tetrahydrocannabinol appears to have significant antiemetic properties when compared with placebo in patients receiving high-dose methotrexate.
Article
Dronabinol (Marinol, Roxane Laboratories, Columbus, OH) and prochlorperazine were tested alone and in combination in a randomized, double-blind, parallel group, multicenter study. Patients were randomized to receive either 1) dronabinol 10 mg every 6 hr plus placebo; 2) placebo plus prochlorperazine 10 mg every 6 hr; or 3) dronabinol and prochlorperazine, each 10 mg every 6 hr. Antiemetic treatment was begun 24 hr prior to and continued for 24 hr after the last dose of chemotherapy; all was given orally. Only 29% of patients in group 3 versus 47% in group 1 and 60% in group 2 experienced nausea after chemotherapy. In addition, the median duration per episode and severity of nausea were significantly less with combination therapy. Vomiting occurred after chemotherapy in 41%, 55%, and 35% of patients in groups 1, 2, and 3, respectively. The median duration per episode of vomiting was 1 min in group 3 versus two in group 1 and four in group 2. Side effects, primarily CNS, were more common in group 1 than in group 2; addition of prochlorperazine to dronabinol appeared to decrease the frequency of dysphoric effects seen with the latter agent. The combination was significantly more effective than was either single agent in controlling chemotherapy-induced nausea and vomiting.