You are on page 1of 8

Current Medical Research and Opinion Vol. 28, No.

12, 2012, 1873–1880

0300-7995 Article ST-0208.R1/744302


doi:10.1185/03007995.2012.744302 All rights reserved: reproduction in whole or part not permitted

Brief review
A review of intranasal ketorolac tromethamine
for the short-term management of moderate
to moderately severe pain that requires
analgesia at the opioid level

d
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

ite
l u nl n
na ow io

,
py us is Lim

d
se oa
so d ut
b
K
rp c i
fo ers tr
Abstract
aU
Andy He

er n
Post-Doctoral Fellow, College of Pharmacy and Health

a
m
Background:

D
Sciences, St. John’s University, Queens, New York;
American Regent, Inc., Shirley, New York, USA An intranasal (IN) formulation of ketorolac was recently FDA approved in adult patients for the short-term
or
le is al management of moderate to moderately severe pain that requires analgesia at the opioid level.
Elliot V. Hersh
ng or i
nf
For personal use only.

si th rc

Department of Pharmacology, University of


co ed
Scope:
2I

Pennsylvania School of Dental Medicine, Philadelphia,


t a . Au e

PA, USA The aim of this paper is to provide an overview of the clinical pharmacology, pharmacokinetics, efficacy, and
rin ed m
1

tolerability of IN ketorolac. Databases used for this literature search include PubMed, International
20
d p ibi m

Pharmaceutical Abstracts, Cochrane Library and ClinicalTrials.gov from January 1980 to January 2012.
Address for correspondence:
All primary papers on IN ketorolac were eligible, including pharmacologic, pharmacokinetic, clinical,
o

Andy He, PharmD, St. John’s University, St. Albert Hall


la d le ©

outcomes, and meta-analyses. The approved product labeling was a source of information, as well as
C

Room 114, 8000 Utopia Parkway, Queens,


the bibliographies of published articles which were reviewed for additional pertinent literature.
t

NY 11439, USA.
ht

ew p r

Tel.: þ1 631 924-4000 x228;


vi e o
an h
ig

Fax: þ1 631 924-1731; Findings:


o

hea@stjohns.edu
r

The search yielded six relevant studies all of which were selected for this review and included efficacy and
yr
sp ize a

safety trials, one pharmacokinetics study, and one preclinical study. IN ketorolac is a non-steroidal
di or S
y, us
No op

Keywords: inflammatory drug that exhibits its effect mainly by inhibiting cyclo-oxygenase (COX) 1 and 2 with high
r

Acute pain – Intranasal analgesic – Intranasal affinity for COX-1. Absorption of IN ketorolac displays a median tmax of 0.50–0.75 hours and has a t½ of
au fo
C

ketorolac – NSAIDs approximately 5–6 hours. Primary analyses included evaluation of morphine use and summed pain intensity
difference (SPID) which was assessed using a visual analog scale. In one of the two phase III studies, the
Un t

Accepted: 24 October 2012; published online: 9 November 2012


mean SPID6 score was 83.3 in the IN ketorolac group versus 37.2 in the placebo group, p ¼ 0.007. In
th

Citation: Curr Med Res Opin 2012; 28:1873–80


another phase III study, the mean SPID6 score was 117.4 in the IN ketorolac group versus 89.9 in the
placebo group, p ¼ 0.032. IN ketorolac was well-tolerated with most adverse events associated with the
route of administration.

Conclusion:
Based on the clinical trials reviewed, IN ketorolac was associated with significant pain reduction in patients
with various post-operative procedures, with good tolerability.

Introduction
Inadequate pain management is often a problem for the estimated 25 million
Americans experiencing acute pain due to injury or surgery yearly1. The most
common causes of acute pain include surgery, acute illness, trauma, labor/deliv-
ery and medical procedures1. In the majority of the cases, pain management
will rely on pharmacologic treatment, predominantly non-opioid and opioid

! 2012 Informa UK Ltd www.cmrojournal.com He & Hersh 1873


Current Medical Research and Opinion Volume 28, Number 12 December 2012

analgesic agents. A new formulation of ketorolac tro- O OH


methamine was recently approved for short-term manage-
ment of moderately to moderately severe pain that requires OH HO OH
N
analgesia at the opioid level2. Though not a new molecular NH2
O
entity, ketorolac tromethamine is highly water soluble,
which allows the preparation of a solution for intranasal Figure 1. Chemical structure of ketorolac tromethamine2.
use3. Other formulations of ketorolac that are available
include injectable, oral, and ophthalmic formulations,
nasal ketorolac, SPRIX, and ketorolac from PubMed,
approved in 1989, 1991, and 1992, respectively4.
International Pharmaceutical Abstracts, Cochrane
The parenteral formulation of ketorolac was the first
Library, and ClinicalTrials.gov. Data found from various
injectable non-steroidal inflammatory drug (NSAID)
papers were screened for relevancy for this review. All
approved in the United States for use as an analgesic5.
primary, original papers on intranasal ketorolac were eli-
Parenteral ketorolac is distinct from other NSAIDs due
gible, including pharmacologic, pharmacokinetic, clinical,
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

to its strong analgesic activity where it has been shown


outcomes, as well as any meta-analyses. The approved
to have similar efficacy to injectable opioids6.
product labeling was a source of information, as well as
Premarketing clinical trials with approximately 5000
the bibliographies of published articles which were
patients demonstrated that the analgesic efficacy of paren-
reviewed for additional pertinent literature.
teral ketorolac was similar to that of parenteral opioids in
patients undergoing general and oral surgery5. The anal-
gesic efficacy of intramuscular ketorolac was shown to be
comparable to standard dosages of morphine and meperi- Results
dine in patients who had various surgical procedures6.
When compared to morphine, the efficacy of intramuscu- Six relevant articles were identified and were included
lar ketorolac 30 mg provided relief comparable to intra- in this review. Four clinical trials evaluated the efficacy
For personal use only.

muscular morphine 6–12 mg7. A recent study in oral and safety of intranasal ketorolac. The other two studies
surgery outpatients confirmed its efficacy at a 15 mg intra- included a pharmacokinetics evaluation in healthy volun-
venous dose8. A 10 mg oral formulation is also available, teers and a preclinical study in rats and rabbits. A numer-
but is only indicated as continuation therapy after paren- ical discrepancy with regard to doses was found in one of
teral treatment9. the articles identified compared with FDA approved doses.
Further supporting of ketorolac’s pronounced analgesic A clarification with the sponsor indicated that the doses
activity, is its well-described opioid-sparing effect10–12. studied were 15.75 mg and 31.5 mg and not 15 mg and
Opioid-induced adverse events are a concern as they 30 mg.
may reduce quality of life and further impair it by limiting
the dosage of opioid analgesic due to bothersome to
intolerable side-effects such as sedation, psychomotor Clinical pharmacology
impairment, nausea and constipation13. Opioid-induced
constipation can delay hospital discharge14. A 25–50% Ketorolac tromethamine is an NSAID indicated in adult
reduction in opioid requirements has been shown patients for the short-term (up to 5 days) management of
with combination therapy of ketorolac with opioids6. moderate to moderately severe pain that requires analgesia
This benefit of concomitant therapy may decrease at the opioid level2. It is a racemic mixture of [–] S- and [þ]
opioid-induced adverse events and accelerate hospital R-enantiomeric forms, with the S-form having analgesic
discharge6. activity2. The chemical name for ketorolac tromethamine
Ketorolac tromethamine nasal spray is an intranasal is ()-5-benzoyl-2,3-dihydro-1H-pyrrolizine-1-carboxylic
formulation of ketorolac developed for patients who acid, compound with 2-amino-2 -(hydroxymethyl)-1,3-
require pain relief at the opioid level. It was approved by propanediol (1:1), and the structural formula is shown in
the US Food and Drug Administration (FDA) in May the Figure 12.
2010. This paper reviews the clinical pharmacology,
pharmacokinetic properties, efficacy, and tolerability of Mechanism of action
ketorolac tromethamine nasal spray.
Behaving like other NSAIDs, ketorolac is a cyclo-oxyge-
nase (COX) inhibitor, inhibiting both COX-1 and COX-
2, although it has higher affinity for COX-1 than other
Methods representative NSAIDS3,16. Cyclo-oxygenase is an
A literature search (from January 1980 to January 2012) enzyme that metabolizes arachidonic acid to prostaglan-
was conducted using the search terms intranasal ketorolac, dins, prostacyclin, and thromboxanes3. Regarding the

1874 He & Hersh www.cmrojournal.com ! 2012 Informa UK Ltd


Current Medical Research and Opinion Volume 28, Number 12 December 2012

Table 1. Ketorolac pharmacokinetic parameters after single-dose administration.

Parameter Intranasal Intramuscular


15.75 mg 31.5 mg 15 mg 30 mg

Cmax, ng/mL 916 (293) 1806 (883) 1163 (280) 2382 (443)
tmax, h* 0.50 (0.25–1.00) 0.75 (0.50–2.00) 0.75 (0.25–1.50) 0.75 (0.23–1.03)
AUC0–t ng h/mL 3723 (1483) 7141 (3466) 4956 (1921) 10770 (3886)
AUC 0–1, ng h/mL 3907 (1569) 7477 (3654) 5196 (2077) 11153 (4260)
t½ , h 4.76 (1.38) 5.24 (1.33) 5.00 (1.72) 4.80 (1.11)

Values are expressed as mean (SD); n ¼ 15.


*Median (range).

activation of peripheral nociceptors, prostaglandin E2 exposure of IN ketorolac was increased by 23% for adults
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

and prostacyclin (PGI2) appear to be key players17. 65 years as compared to subjects565 years2. Terminal t½
Prostaglandins play a function in pain as they are released was longer in the elderly population as compared to the
from injured tissues3. At the site of injury and inflamma- non-elderly, 4.5 vs. 3.3 hours, respectively. From a single-
tion, prostaglandins are thought to sensitize afferent nerve dose study, the mean t½ of IN ketorolac in renally impaired
endings to other mediators of pain (histamine, bradykinin, patients was found to be between 6 and 19 hours and is also
etc.), and thus, diminished prostaglandin concentrations dependent on the extent of impairment. The AUC1 of
at these nerve endings should ultimately mitigate the pain each enantiomer was increased by 100% in patients with
response3,18. renal disease as compared to healthy volunteers. Reducing
the dose by 50% is recommended in these patient popula-
tions. Hepatic impairment has also been studied; however,
Pharmacokinetics no significant difference in t½, AUC1, and Cmax was
For personal use only.

observed.
McAleer et al.15 conducted a pharmacokinetic and safety
study comparing a single-dose of intranasal (IN) and intra-
muscular (IM) ketorolac in 15 healthy volunteers aged
19–45 years and weighing 49–92 kg who were followed
Efficacy
for up to 24 hours post-dose. In this crossover, randomized Four clinical trials have assessed the efficacy of IN ketor-
study, 15.75, 31.5, and 47.25 mg of IN ketorolac were com- olac tromethamine using patient populations with moder-
pared to 15 and 30 mg of IM ketorolac. Based on this study ate-to-severe pain (i.e., abdominal, orthopedic and oral
and other literature, data have shown that ketorolac exhi- surgery)19–22. Assessments of pain intensity as measured
bits linear pharmacokinetics. Similar to the observed IM on a 100 mm visual analog scale (VAS), pain intensity
administration, IN ketorolac exhibited rapid absorption difference (PID), summed pain intensity difference
with a median tmax ranging from 0.50 to 0.75 hours, and (SPID), morphine dosage use, perceptible and meaningful
a t½ of approximately 5–6 hours. Relative bioavailability of relief, quality of pain relief, global assessment of pain con-
IN ketorolac was approximately 67–75% as compared to trol, and total pain relief scores (TOTPAR) have been
an absolute bioavailability of 100% in the IM formulation used as outcomes measures. The VAS evaluates pain
of ketorolac. Plasma concentrations of 31.5 mg IN intensity from 0 to 100, where 0 represents no pain and
ketorolac fell approximately between that of 15 and 100 is the worst pain imaginable. PID is calculated by sub-
30 mg IM ketorolac. A comparison of the pharmacokinetic tracting the post-treatment VAS score from the baseline
parameters among the treatment groups are displayed VAS score. SPID is calculated by adding the weighted PID
in Table 115. score over specific intervals. Therefore, a higher SPID
According to the package insert, the mean apparent score indicates greater overall pain relief for that time
volume of distribution is approximately 13 liters2. period. The use of stopwatch techniques is utilized for mea-
Ketorolac is a highly protein bound drug (approximately sures of perceptible and meaningful pain relief.
99%). Mainly metabolized by the liver, the metabolic Summarization of the clinical trials is found in Table 2.
products are hydroxylated and conjugated forms of the Moodie et al.19 reported the results from a randomized,
parent drug. After metabolism, ketorolac and its metabo- double-blind, placebo-controlled study that compared the
lites are eliminated renally, with around 92% of the given efficacy of IN ketorolac to placebo in patients (mean age
dose found in the urine and, to a minimal extent, in the 53 years) who underwent major surgery (i.e., abdominal
feces (6%). and orthopedic). A total of 127 patients were randomized
In a single-dose study (31.5 mg) that was conducted to to receive one of the three intranasal treatments
explore IN ketorolac’s pharmacokinetics in the elderly, the (i.e., 10 mg, 31.5 mg IN ketorolac, or IN placebo).

! 2012 Informa UK Ltd www.cmrojournal.com He & Hersh 1875


Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14
For personal use only.

1876
He & Hersh
Table 2. Summary of clinical trials of intranasal ketorolac.

Study Study design Patient population Study drug administration Primary efficacy endpoint Efficacy results Tolerability results
Current Medical Research and Opinion

Moodie et al. R, DB, PC 127 patients with 10 mg, 31.5 mg of IN Total morphine use by PCA Mean morphine use (mg) Top 3 adverse events
(2009)19 abdominal or orthope- ketorolac, or IN placebo (from start of dosing (SEM) 0–24 h Placebo: pyrexia (61.9%),
dic surgeries having an Q8H for 40 h through 24 h) Placebo: 56.5 mg (4.8) nausea (47.5%), tachycardia
PI score of 40 mm on Morphine sulfate (1 mg 10 mg IN ketorolac: (40.5%)
a 100 mm VAS of MS each actuation 54.3 mg (6.4) 10 mg IN ketorolac: nausea
with a lockout time of 31.5 mg IN ketorolac: (58.1%), pyrexia (55.8%),
6 min) administered via 37.8 mg (5.0) headache (34.9%)
PCA was available for (p = 0.0013 for 31.5 mg IN ketorolac:
Volume 28, Number 12

pain not relieved by 31.5 mg IN ketorolac nausea (45.2%), pyrexia


study drug vs. placebo) (33.3%), vomiting (28.6%)
Brown et al. R, DB, PC 300 patients with abdom- 31.5 mg of IN ketorolac or 6-h summed pain intensity Mean  SE SPID6 score Top 3 adverse events
(2009)20 Consisted of a inal or orthopedic sur- IN placebo TID for up to difference (SPID6) in 31.5 mg IN ketorolac: Placebo: pyrexia/intermittent
single-dose and geries having an PI 5 days the single-dose phase 83.3  10.6 pyrexia (67%), nausea (60%),
December 2012

a multi-dose score of 40 mm on a Back-up analgesia is Placebo: 37.2  12.9 constipation (36%)
phase 100 mm VAS available with MS as (p = 0.007) 31.5 mg IN ketorolac: nausea
PCA (not available at (58%), pyrexia/intermittent
the single-dose phase) pyrexia (44%), constipation
(29%)
Grant and R, DB, PC 80 patients who had third 31.5 mg of IN ketorolac or 8-h summed pain intensity Mean  SE SPID8 score Adverse events
Mehlisch molar extraction sur- IN placebo difference (SPID8) 31.5 mg IN ketorolac: Placebo: 10 events in 8
(2010)21 gery with bony impac- Rescue analgesic 136.7  33.0 patients
tions having an PI score medication was Placebo: IN ketorolac: 3 events in 3
of 450 on a 100 mm provided for pain not 105.2  29.1 patients
VAS relieved by study drug (p50.001)
Singla et al. R, DB, PC, MC 321 patients who had 31.5 mg of IN ketorolac or 6-h summed pain intensity Least square means  SE Top 3 adverse events
(2010)22 abdominal surgery IN placebo Q6H for the difference (SPID6) SPID6 score Placebo: nausea (62%), pyrexia
having an PI score of first 48 h, then up to 31.5 mg IN ketorolac: (34%) constipation (33%)
40 on a 100 mm VAS 4 times/day for up to 117.4  7.7 31.5 mg IN ketorolac: nausea
5 days Placebo: 89.9  10.6 (57%), constipation (28%),
Access to MS via PCA (p = 0.032) vomiting (24%)
was available for first
48 h. Hydrocodone/
APAP was available
when PCA was no
longer required.

R, randomized; DB, double-blind; PC, placebo controlled; PI, pain intensity; VAS, visual analog scale; IN, intranasal; Q8H, every 8 hours; MS, morphine sulfate; PCA, patient-controlled analgesia; SEM, standard error of the
mean; SPID6, 6-hour summed pain intensity difference; SE, standard error; SPID8, 8-hour summed pain intensity difference; MC, multi-center; Q6H, every 6 hours; h, hours; min, minutes; APAP, acetaminophen.

www.cmrojournal.com ! 2012 Informa UK Ltd


Current Medical Research and Opinion Volume 28, Number 12 December 2012

These patients received the study drug if they had a pain they requested additional analgesia. Patients whose pain
intensity (PI) rating of at least 40 mm on a VAS. Study intensity (as assessed via VAS) never reached 40 after
drug was administered every 8 hours through 40 hours with 5 hours of withdrawing PCA on day 1 did not enter the
assessment before the study drug, at 30 minutes, and at 1, 2, single-dose phase but returned to the multi-dose phase.
3, 4, 5, 6, 8, 12, 16, 20, 24, 28, 32, 36, 40, 44, and 48 hours Patients in the multi-dose phase continued to receive
after the first study dose. If completed, the patient would study drug as randomized on day 0 (IN ketorolac or pla-
have received six doses of the study drug. Patients that had cebo) given three times a day for up to 5 days with PCA as
pain despite administration of the study drug had access to an option for pain not relieved by the study drug. The
patient-controlled analgesia (PCA), i.e. morphine sulfate. primary efficacy endpoint was the 6-hour summed pain
The primary efficacy endpoint was total morphine use intensity difference (SPID6) score for patients in the
(mg) by PCA from the start of dosing through 24 hours. single-dose phase. Other endpoints that were examined
Other endpoints that were examined include morphine include PID scores, duration of analgesia, quality of anal-
use 24–48 hour, 0–48 hours, PID, and SPID. gesia, meaningful pain relief, total morphine use and global
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

At baseline, PI ratings by VAS were similar among the assessment of pain control. In addition, a categorical mea-
three groups in addition to the patient demographics, sure termed quality of analgesia was assessed by patients
except for a trend for older age and a greater proportion on a 5-point scale where 0 ¼ poor and 4 ¼ excellent.
of men in the placebo group. The surgical procedures Similarly, global assessments of pain control was defined
included were orthopedic (77/127, 61%) and abdominal by patients on a 5-point scale where 0 ¼ poor and
(50/127, 39%). The mean morphine consumption for the 4 ¼ excellent based on a question regarding overall pain
first 24 hours was 56.6 mg in the placebo group, 54.3 mg in control.
the 10 mg IN ketorolac group, and 37.8 mg in the 31.5 mg A total of 300 patients were enrolled in the study with
IN ketorolac group, (p ¼ 0.0013 between 31.5 mg IN 189 patients in the single-dose phase and 210 patients in
ketorolac and placebo and p ¼ 0.0332 between the two the multi-dose phase. The mean age of the patients was
IN ketorolac groups). This study data suggest that IN around 52 years with majority of them being female and
For personal use only.

ketorolac in 31.5 mg doses significantly reduced morphine Caucasian. No significant differences in baseline charac-
consumption as compared to the 10 mg IN ketorolac and teristics were found among the two groups. Abdominal
placebo groups. There were no statistically significant dif- (52%) and orthopedic (46%) surgeries were the most
ferences between the 10 mg IN ketorolac group and pla- common types of procedure in this patient population.
cebo group and the two IN ketorolac groups at 24–48 hours Results from the study showed that the mean SPID6
and 0–48 hours with regard to morphine use. Mean SPID score was significantly higher in the IN ketorolac group
scores at 4 and 6 hours were higher in the 31.5 mg IN compared with the placebo group (83.3 vs. 37.2,
ketorolac group compared to placebo. The 4-hour SPID p ¼ 0.007). With regard to SPID48, a statistically signifi-
score was 120.1 in the 31.5 mg IN ketorolac group and 89.6 cant greater reduction in pain was demonstrated in
in placebo group (p ¼ 0.0017). The 6-hour SPID score was patients who received IN ketorolac compared to placebo2.
195.5 in the 31.5 mg IN ketorolac group and 130.6 in the For the mean PID scores, the IN ketorolac group had sta-
placebo group (p ¼ 0.0015). Higher SPID scores in the tistically significantly higher scores at 0.5, 1, 2, 3, and 4
31.5 mg IN ketorolac group indicated better overall pain hours20. Efficacy analysis of the single-dose phase indi-
relief as compared to the other two groups. cated that patients reported better quality of analgesia
In a randomized, double-blind, placebo-controlled, with IN ketorolac at each measurement between 0.5
phase III clinical trial, by Brown et al.20, patients who hours and 5 hours (p50.05). At 6 hours, there was a
underwent various surgical procedures were randomized trend toward better quality of analgesia with IN ketorolac
in a 2:1 ratio to either 31.5 mg IN ketorolac or placebo, (p ¼ 0.068). Duration of analgesia was longer in the IN
respectively. This study consisted of two phases, a single- ketorolac group (median 3.0 hours) than the placebo
dose phase and a multi-dose phase that evaluated the effi- group (1.3 hours, p50.04), which is characterized by the
cacy and tolerability of IN ketorolac. On the day of surgery time to restarting PCA or requesting rescue medication in
(day 0), patients were randomized to IN ketorolac or pla- the single-dose segment. In assessing acute pain relief, a
cebo when their pain intensity (as assessed via VAS) greater proportion of patients in the IN ketorolac group
reached 40 and began to receive study drug with access reported meaningful pain relief compared to placebo
to PCA morphine. The day after surgery (day 1), PCA within 1 hour, (84 vs. 70%, p50.05). At 2 hours, it still
was withdrawn and patients entered the single-dose favored the IN ketorolac group but was not statistically
phase if they had a VAS score 40. These patients significant (84 vs. 73%, p ¼ 0.058). Reduced morphine
received a single dose of IN ketorolac or placebo with use was associated with IN ketorolac reduction which
assessment at 30 minutes and hourly for 6 hours without ranged between 20 and 50% for the first 24 hours.
access to PCA. Patients in the single-dose phase rejoined During the multi-dose phase, the quality of analgesia was
the multi-dose phase when pain returned to baseline or if significantly higher at 8 hours for IN ketorolac compared

! 2012 Informa UK Ltd www.cmrojournal.com He & Hersh 1877


Current Medical Research and Opinion Volume 28, Number 12 December 2012

with placebo, however, it was not statistically significant drug were permitted an oral analgesic (e.g., hydrocodone/
at 32, 40, and at 48 hours. acetaminophen). Following an intent-to-treat analysis,
Efficacy of IN ketorolac was also reported by Grant and the primary efficacy outcome evaluated was the 6-hour
Mehlischin in patients who had received third molar bony summed pain intensity difference (SPID6). Other end-
impaction surgery21. This was a randomized, double-blind, points included morphine usage, SPID4, PID, quality of
placebo-controlled study that compared 31.5 mg IN ketor- pain relief, and global assessment of pain control.
olac to a matching IN placebo. Patients were randomized Baseline characteristics of both groups did not show any
to a single dose of IN ketorolac or placebo when their VAS apparent disparities (mean age of 46, predominantly
score was 50 subsequent to the dental procedure for up female population of 96%, and mostly white). The major-
to 8 hours. Backup analgesia was permitted for pain not ity of the abdominal surgical procedures were hysterecto-
relieved by the study drug. The primary efficacy endpoint mies (69.5%). IN ketorolac was associated with better pain
was SPID8. Assessment of other outcomes included PID, relief when compared to placebo, as shown by the SPID6
total pain relief (TOTPAR) scores, global pain control, score (least square means 117.4 vs. 89.9, respectively,
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

and the time to backup analgesia. Pain relief was assessed p ¼ 0.032). Efficacy was also demonstrated by SPID48
on a 5-point scale where 0 ¼ none and 4 ¼ complete. with a statistically significant greater reduction in pain
TOTPAR is calculated from taking a weighted sum of associated with IN ketorolac when compared to placebo2.
pain relief scores. Mean PID scores were statistically higher at 20 minutes, 1,
There were 40 patients in each study arm; patients in 2, and 3 hours, but were not statistically significant at 4, 5,
the two arms had similar baseline demographics as well as and 6 hours22. Both the IN ketorolac and placebo groups
pain intensity prior to administration of the study drug. showed decreased pain intensity (as assessed via mean
Mean SPID8 score for the IN ketorolac group was signif- VAS scores) at 20 minutes through 30 hours, but the
icantly higher compared with the placebo group (136.7 vs. decreases were statistically significantly greater in the IN
–105.2, p50.001). The negative mean SPID score in the ketorolac group. No statistically significant differences
placebo group indicates that during the 8-hour evaluation
For personal use only.

were found after 30 hours. Patient rating on quality of


period, mean pain intensity continued to increase com- analgesia (on a scale of 0–4) were better with IN ketorolac
pared to baseline. PID scores were also higher at every compared to placebo from 20 minutes to 24 hours with
time point with statistical significance at 20 minutes statistically significant differences, except at 40 minutes
through 8 hours (p50.001). Results from the TOTPAR and 6 hours. Global assessment of pain control was signif-
score indicated better pain relief from the IN ketorolac icantly better in the IN ketorolac group compared to the
group than the placebo group, reaching statistical signifi- placebo group on day 1, but was not significantly different
cance for the 4 -, 6 -, and 8-hour area-under-the-curve on days 2, 3, and 4. Morphine utilization was evaluated
measures. The comparison of IN ketorolac with placebo at several time intervals (i.e., 0–24, 0–48, 24–48 and 0–72
indicated a faster median onset of perceptible pain relief hours). Mean morphine use from 0 to 24 hours was 42 mg
(21.5 vs. 480.0 minutes, p50.001), a faster median onset in the IN ketorolac group versus 54 mg in the placebo
of meaningful pain relief (66.0 vs. 480.0 minutes, group (p ¼ 0.003) representing a 22% decrease in opioid
p50.001), and a longer median time for rescue analgesic requirement. Mean morphine use from 24 to 48 hours was
medication (360.0 vs. 95.5 minutes, p50.001), respec- 23 mg in the IN ketorolac group versus 31 mg in the pla-
tively. As assessed from 20 minutes to 8 hours, a greater cebo group (p ¼ 0.041), indicating a 26% decrease in
percentage of patients in the IN ketorolac group reported opioid requirement. Statistical significance was not
‘some’ (rating of 2) to ‘complete’ (rating of 4) pain relief found between 0 and 72 hours even though a 32% decrease
compared to placebo. Global assessment of pain control on of morphine use was associated with the IN ketorolac
a 0–4 scale also favored IN ketorolac with mean scores of group compared to the placebo group.
1.9 for the IN ketorolac group and 0.5 for the placebo
group (p50.001).
Using a multicenter (United States and New Zealand),
Safety and tolerability
randomized, phase III, placebo-controlled design study,
Singla et al.22 studied the efficacy and tolerability of IN Among the two phase III clinical trials, there were similar
ketorolac in abdominal surgery patients. A total of 321 overall rates of adverse events between the study groups,
patients were enrolled in the study to receive 31.5 mg IN which were 97.5% in the IN ketorolac group and 98.0% in
ketorolac or placebo every 6 hours in a 2:1 ratio for up to 5 the placebo group in one study20 and 93% in the IN ketor-
days. Once the patients reached a VAS score 40, they olac group and 96% in the placebo group in another
received their respective study drug with access to mor- study22. Since this product is an intranasal product, it is
phine sulfate as PCA if their pain was not alleviated with not surprising that the most frequent adverse events
the study drug for the first 48 hours. Following the first 48 reported on the package insert are associated with the
hours, patients who had pain not controlled by the study route of administration. Based on the postoperative pain

1878 He & Hersh www.cmrojournal.com ! 2012 Informa UK Ltd


Current Medical Research and Opinion Volume 28, Number 12 December 2012

studies conducted with a population of 455 patients who Dosing and administration
received IN ketorolac, adverse reactions were observed at a
rate of 2% or more and at least twice the incidence seen Currently, intranasal ketorolac is available as single-day
in the placebo group and were as follows: nasal discomfort nasal spray bottles in the US. Each single-day nasal spray
(15%), rhinalgia (13%), increased lacrimation (5%), bottle contains a sufficient quantity of solution to admin-
throat irritation (4%), oliguria (3%), rash (3%), bradycar- ister 8  100 mL sprays after appropriate priming (instruc-
tions included with package insert). Each spray provides
dia (2%), urine output decreased (2%), ALT and/or AST
15.75 mg of ketorolac. For adult patients565 years of age,
increased (2%), hypertension (2%), and rhinitis (2%)2.
the dose is 31.5 mg (one 15.75 mg spray in each nostril)
Local nasal symptoms were reported as mild, transient
every 6–8 hours. For patients who are 65 years of age,
and did not increase in severity with repeated
renally impaired patients, and patients less than 50 kg,
dosing20,22. Conversely, IN ketorolac was associated with
the dose is 15.75 mg (one 15.75 mg spray in one nostril)
modestly less constipation, pyrexia and pruritus (events
every 6–8 hours2.
frequently associated with opioids) as compared to
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

placebo, most likely due to the reduction in morphine


utilization with IN ketorolac administration
(Table 2)19,20,22. Discussion
Given that ketorolac is an NSAID that is highly
This review included four studies that reported the efficacy
selective for blocking COX-1, gastrointestinal adverse
and safety of intranasal ketorolac in adult patients with
events become of greater clinical concern with long-term
acute postoperative pain. These clinical trials were well-
use than other NSAIDs16,23. A large epidemiologic study
designed studies with well-defined outcomes and included
was conducted and investigated the risk of gastrointestinal
an adequate number of patients to provide information
and operative site bleeding associated with the use of
about efficacy and safety. Pain assessments included
parenteral ketorolac. This study concluded that the risk summed pain intensity difference scores, summed pain
For personal use only.

of both gastrointestinal bleeding and operative site relief scores, quality of analgesia, global evaluation of
bleeding are small, but the risk is higher when therapy pain control, duration of analgesia, and onset of analgesia.
was in higher doses, in older subjects, and for more than Measurements were based on VAS and categorical rating
5 consecutive days5. Currently, the oral, parenteral, and scales. To fully assess acute pain relief, the use of percep-
the nasal formulation of ketorolac have a 5-day limitation tible pain relief as well as meaningful pain relief was uti-
as part of their Black Box Warnings on the product lized as outcome measures. Evaluation of an opioid-sparing
label2,8,24. All four clinical trials had administered IN effect was investigated by an efficacy measure of morphine
ketorolac with a timeframe of 8 hours to 5 days19–22. use through various time periods. An additional endpoint
There were no reports of gastrointestinal bleeding that could have been investigated is the percentage of
associated with IN ketorolac in the two phase III stud- subjects who needed a rescue medication.
ies20,22. From the package insert information, seven Results from the two phase III trials, like the phase II
patients (n ¼ 455, 1.5%) treated with IN ketorolac trials, indicated better analgesic efficacy with intranasal
experienced adverse events of bleeding (four patients) or ketorolac as compared to placebo with statistical signifi-
hematoma (three patients) at the operative site versus one cance20,22. Furthermore, Brown et al. (one of the phase III
patient (n ¼ 245, 0.4%) treated with placebo (hema- trials) reported that patients who were treated with intra-
toma)2. Because of its potent antiplatelet effects, IN ketor- nasal ketorolac required 34% less morphine than patients
olac (similar to other ketorolac formulations) should not who were treated with placebo over 48 hours20. Likewise,
be used as a pre-emptive analgesic before any major Singla et al. (the other phase III trial) reported that
surgery2. patients needed 26% less morphine when treated with
Within the two phase III trials, there were no intranasal ketorolac than those with placebo22. These
clinically relevant differences among the treatment results support efficacy as well as reduced opioid use.
groups with reference to vital signs, hematology, or clinical Intranasal ketorolac proved to be well-tolerated with
chemistry measurements20,22. Rates of serious adverse local nasal symptoms being the most common adverse
events were similar in the two phase III trials with reactions among post-operative patients. Among the four
2.5%20 and 6%22 in the IN ketorolac arms and 1.9%20 clinical trials, there were no gastrointestinal bleeding
and 6%22 in the placebo arms. Serious adverse events events reported with intranasal ketorolac use.
that may have been related to IN ketorolac included Major advantages of intranasal ketorolac are the con-
wound complication and gastrointestinal events. venience afforded by its route of administration relative
Regarding treatment-emergent cardiac adverse events, to injectable forms and the ability to provide a level of
Singla et al. reported no differences between the two analgesia similar to that of opioids without side-effects
groups22. often associated with this class of analgesics

! 2012 Informa UK Ltd www.cmrojournal.com He & Hersh 1879


Current Medical Research and Opinion Volume 28, Number 12 December 2012

(e.g., drowsiness and constipation). This review was 3. Boyer KC, McDonald P, Zoetis T. A novel formulation of ketorolac trometha-
limited by the small number of randomized studies. In mine for intranasal administration: preclinical safety evaluation. Int J Toxicol
2010;29:467-78
addition, the use of supplement PCA morphine in some 4. Food and Drug Administration. Orange Book: Approved Drug Products with
studies can act as a potential confounding factor although Therapeutic Equivalence Evaluations. 32th ed. Silver Spring, MD; Food and
this represents a typical standard of care. Post-operative Drug Administration, 2012. http://www.accessdata.fda.gov/scripts/cder/ob/
procedures studied in this review include abdominal, gyne- eclink.cfm. Accessed January 2012
cologic, orthopedic, and oral surgery models. These proce- 5. Strom BL, Berlin JA, Kinman JL, et al. Parenteral ketorolac and risk of gas-
trointestinal and operative site bleeding. JAMA 1996;275:376-82
dures are frequently performed in clinical practice and they
6. Gillis JC, Brogden RN. Ketorolac. A reappraisal of its pharmacodynamic and
are representative of both inpatient and outpatient set- pharmacokinetics properties and therapeutic use in pain management. Drugs
tings. Other patient populations who require treatment 1997;53:139-88
for acute pain should be studied in the future. No studies 7. Brown CR, Mazzulla JP, Mok MS, et al. Comparison of repeat doses of
were available that compared intranasal ketorolac with intramuscular ketorolac tromethamine and morphine sulfate for analgesia
oral NSAIDs or opioids. Future research should include after major surgery. Pharmacotherapy 1990;10:45-50S
8. Newberg AB, Hersh EV, Levin LM, et al. Double-blind, placebo-controlled,
Curr Med Res Opin Downloaded from informahealthcare.com by McMaster University on 12/09/14

comparative, economic, and post-marketing surveillance randomized pilot study of cerebral blood flow patterns employing SPECT
studies. imaging in dental postsurgical pain patients with and without pain relief.
Clin Ther 2011;33:1894-903
9. TORADOL (ketorolac tromethamine) tablet, film coated [package insert].
Nutley, NJ: Roche Laboratories Inc., Dec 2008
Conclusion 10. Chen JY, Ko TL, Wen YR, et al. Opioid-sparing effects of ketorolac and its
Based on the literature reviewed, intranasal ketorolac is correlation with the recovery of postoperative bowel function in colorectal
surgery patients: a prospective randomized double-blinded study. Clin J
efficacious in acute pain relief in adult patients with Pain 2009;25:485-9
good tolerability. 11. Pavy TJ, Paech MJ, Evans SF. The effect of intravenous ketorolac on opi-
oid requirement and pain after cesarean delivery. Anesth Analg
2001;92:1010-14
For personal use only.

12. Etches RC, Warriner CB, Badner N, et al. Continuous intravenous adminis-
Transparency tration of ketorolac reduces pain and morphine consumption after total hip or
Declaration of funding knee arthroplasty. Anesth Analg 1995;81:1175-80
The creation of this manuscript was requested by Luitpold 13. McNicol E, Horowicz-Mehler N, Fisk RA, et al. Management of opioid side
effects in cancer-related and chronic noncancer pain: a systematic review.
Pharmaceuticals. No funding was provided to the authors for
J Pain 2003;4:231-56
the preparation of this manuscript.
14. White PF. The role of non-opioid analgesic techniques in the management of
pain after ambulatory surgery. Anesth Analg 2002;94:577-85
Declaration of financial/other relationships 15. McAleer SD, Majid O, Venables E, et al. Pharmacokinetics and safety of
A.H. is an employee of St. John’s University and is currently in a ketorolac following single intranasal and intramuscular administration in
fellowship at American Regent, Inc. E.H. is an employee of healthy volunteers. J Clin Pharmacol 2007;47:13-18
University of Pennsylvania and receives research funding from 16. Hersh EV, Lally ET, Moore PA. Update on cyclooxygenase inhibitors: has a
Luitpold Pharmaceuticals. third COX isoform entered the fray? Curr Med Res Opin 2005;21:1217-26
CMRO peer reviewers may have received honoraria for their 17. Dray A. Inflammatory mediators of pain. Br J Anaesth 1995;75:125-31
review work. The peer reviewers of this manuscript have dis- 18. Ferreira SH, Moncada S, Vane JR. Prostaglandins and the mechanism of
closed that they have no relevant financial relationships. analgesia produced by aspirin-like drugs. Br J Pharmacol 1973;49:86-97
19. Moodie JE, Brown CR, Bisley EJ, et al. The safety and analgesic efficacy of
intranasal ketorolac in patients with postoperative pain. Anesth Analg
Acknowledgments
2008;107:2025-31
Employees of Luitpold Pharmaceuticals, Inc. reviewed the 20. Brown C, Moodie J, Bisley E, et al. Intranasal ketorolac for postoperative pain:
manuscript. a phase 3, double-blind, randomized study. Pain Med 2009;10:1106-14
21. Grant GM, Mehlisch DR. Intranasal ketorolac for pain secondary to third molar
impaction surgery: a randomized, double-blind, placebo-controlled trial.
J Oral Maxillofac Surg 2010;68:1025-31
References 22. Singla N, Singla S, Minkowitz HS, et al. Intranasal ketorolac for acute post-
1. Pain: Current Understanding of Assessment, Management, and Treatments. operative pain. Curr Med Res Opin 2010;26:1915-23
Editoral advisory board Berry PH, Chapman CR, Covington EC, Dahl JL, Katz JA, 23. Warner TD, Guiliano F, Vojnovic I, et al. Nonsteroid drug selectivities for cyclo-
Miakowski C. Continuing Education Sponsored by the American Pain Society oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gas-
and supported by unrestricted education grant from the National trointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci USA
Pharmaceutical Council, Inc., Dec 2001 1999;96:7563-8
2. SPRIX (ketorolac tromethamine) Nasal Spray [package insert]. Shirley, NY: 24. Ketorolac tromethamine injection, solution [package insert]. Lake Forest, IL:
Regency Therapeutics, 2011 Hospira, Inc., Mar 2007

1880 He & Hersh www.cmrojournal.com ! 2012 Informa UK Ltd

You might also like