GET THE APP

Clinical and Laboratory Hematologic Findings in Patients Receivin
Anesthesia & Clinical Research

Anesthesia & Clinical Research
Open Access

ISSN: 2155-6148

+44 1223 790975

Research Article - (2015) Volume 6, Issue 7

Clinical and Laboratory Hematologic Findings in Patients Receiving Repeated-Dose Injectable HPBCD-Diclofenac for Acute Postoperative Pain: Pooled Analysis of Two Randomized Controlled Phase III Clinical Trials

Tong J Gan1, Neil Singla2, Stephen E Daniels3, Douglas A Hamilton4,5, Peter G Lacouture6,7, Olufunmibi Johnson8, Christian RD Reyes8 and Daniel B Carr4,9*
1Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA
2Lotus Clinical Research, LLC, Pasadena, CA 91105, USA
3Premier Research, Austin, TX 78705, USA
4Javelin Pharmaceuticals, Inc., Cambridge, MA 02140, USA
5New Biology Ventures, LLC, San Francisco, CA 94133, USA
6Magidom Discovery, LLC, Lindenhurst, IL, 60046, USA
7Brown University School of Medicine, Providence, RI, 02912, USA
8Hospira, Inc, Lake Forest, IL 60045, USA
9Department of Anesthesiology, Tufts Medical Center, Boston, MA 02111, USA
*Corresponding Author: Daniel B Carr, Department of Anesthesiology, Tufts Medical Center, #298, 800 Washington Street, Boston, MA 02111, USA, Tel: 617-636-9710, Fax: 617-636-9709 Email:

Abstract

Objective: While the non-steroidal anti-inflammatory drugs (NSAIDs) represent an important option for the management of acute postoperative pain, their use can be limited due to potential safety concerns, including bleeding risks. This study examined the bleeding-related safety of injectable diclofenac formulated with hydroxypropyl-β-cyclodextrin (HPβCD-diclofenac) when used for postoperative pain management.

Methods: Data from two randomized, double-blind, placebo- and active comparator-controlled phase III trials were pooled. Patients in both studies received HPβCD-diclofenac, placebo, or the active comparator ketorolac via intravenous injection every 6 hours for ≤ 5 days following abdominal/pelvic or orthopedic surgery. Bleeding adverse events (AEs) were evaluated through the treatment period and follow-up (≤ 37 days), and relative bleeding AE risks (RR) were estimated. Changes in hematology laboratory values were also assessed.

Results: Overall, 608 surgical patients received ≥ 1 dose of study medication. Bleeding AEs occurred in n=9/318 (2.8%) patients receiving HPβCD-diclofenac, n=8/142 (5.6%) patients receiving ketorolac, and n=4/148 (2.7%) patients receiving placebo. Over the period examined, HPβCD-diclofenac was not associated with increased bleeding AE RR versus placebo (1.05 [0.33, 3.35]; p=0.93), nor was ketorolac (2.08 [0.64, 6.77]; p=0.22). Bleeding AEs were predominantly mild or moderate in severity. No treatment-related bleeding AEs occurred in the HPβCDdiclofenac group (1 in both the placebo and ketorolac groups). Among the subset of patients receiving concomitant anticoagulants, bleeding AEs occurred in n=3/60 (5.0%) patients receiving HPβCD-diclofenac, n=2/29 (6.9%) patients receiving ketorolac, and n=0/35 patients receiving placebo. In the HPβCD-diclofenac group, postsurgical shifts to low hematocrit and hemoglobin occurred in 35.7% and 28.3% of patients, respectively (versus 31.4% and 21.5%, respectively, with placebo). Postsurgical shifts in platelet count were uncommon (<3.0% across treatment groups).

Conclusions: While follow-up studies in larger populations are warranted, this analysis suggests that HPβCDdiclofenac may not present a significant incremental bleeding AE risk versus placebo when used for acute postoperative pain management.

Keywords: Postoperative pain; Non-steroidal anti-inflammatory drugs; Non-opioid analgesics; Safety; Orthopedic surgery; Bleeding; Multimodal analgesia

Introduction

Adequate treatment of acute pain following surgery is a key aspect of perioperative care, as under-treatment of acute pain can trigger greater use of healthcare resources and ultimately lead to poor outcomes, which can include the development of chronic postsurgical pain [1-5]. Intravenous (IV) non-steroidal anti-inflammatory drugs (NSAIDs) are considered an increasingly important element of postoperative pain management, either exclusively or as a component of a multimodal regimen [6-8]. Use of NSAIDs in combination with opioids in the postsurgical setting can contribute to reduced opioid consumption and lowered incidences of opioid-related adverse events (AEs) such as nausea and vomiting that can impede recovery, thereby helping to reduce hospital length of stay following surgery [9-15].

Though the efficacy of NSAIDs for the management of several painful conditions has been widely demonstrated, drugs in this class also carry potential concerns related to gastrointestinal (GI), hematologic, renal, and cardiovascular safety [16-20]. Owing to these concerns, NSAID-containing products include warnings outlining the GI, renal, and bleeding risks associated with NSAID use [21,22]. In addition, guidelines typically recommend limiting NSAID dosage and duration, as well as selection of NSAIDs based on individual patient risk assessments [8,20,23].

Specific bleeding risks related to NSAID use have largely been identified in the context of long-term or chronic use, which has been associated with increased risks of GI bleeding [23-27]. Factors such as advanced age, concomitant anticoagulant use, and longer-duration NSAID use have also been linked with increased bleeding-related AE risk in patients receiving NSAIDs [21,23-25,28-30]. Importantly, NSAID use has also been associated with potential bleeding-related complications when utilized postoperatively [31]. Thus, any new NSAID indicated for use in this setting must be carefully evaluated with respect to potential bleeding risks.

Diclofenac is an NSAID with equivalent inhibition of cyclooxygenase-1 (COX-1) and COX-2 and established efficacy, and is widely used to treat a number of painful conditions [32-34]. An injectable formulation of diclofenac solubilized with hydroxypropyl-β-cyclodextrin (HPβCD-diclofenac) does not require dilution and is administered as a low-volume bolus injection [35]. The efficacy and overall safety of HPβCD-diclofenac have been demonstrated in clinical trials examining single- and repeated-dose regimens for the treatment of acute postsurgical pain [36-42], and HPβCD-diclofenac is indicated for the treatment of mild-to-moderate pain when given alone, and moderate-to-severe pain when given alone or in combination with opioids.

Given the importance of understanding the potential bleeding risks associated with any NSAID, the objective of the current study was to conduct an in-depth examination of bleeding safety in patients receiving HPβCD-diclofenac for ≤5 days for the treatment of acute moderate-to-severe pain following abdominal/pelvic or orthopedic surgery. The study comprised an analysis of pooled AE and hematology laboratory data from two randomized, double-blind, placebo- and active comparator-controlled phase III trials in patients requiring IV analgesia for acute postsurgical pain [36,37].

Methods

For the trials included in this analysis (clinicaltrials.gov identifiers NCT00448110, NCT00507026), all patients provided IRB-approved written informed consent. Detailed methods for the individual studies are presented in Gan et al. [37] and Daniels et al. [36]. For both studies, sample size was based on calculated values defining the number of patients required to detect a clinically significant difference in the study’s primary efficacy measure.

Patients

Per individual study protocols, patients were screened for inclusion if they were scheduled for abdominal/pelvic or orthopedic surgery requiring IV analgesia for the management of postoperative pain. Key patient inclusion criteria included age 18-65 years in the abdominal/pelvic surgery study and 18-85 years in the orthopedic surgery study, and the presence of moderate-to-severe patient-reported pain within 6 hours following surgery (pain intensity ≥ 50 mm on the 0-100 mm visual analog scale (VAS)). Females of childbearing age were required to have a negative pregnancy test at screening, as well as be practicing abstinence/using an approved contraception method. Patients were excluded from either study if they had a history of uncontrolled chronic disease contraindicating study participation, recent history (≤ 6 months) of cardiovascular events such as myocardial infarction (MI) or stroke, known allergy to diclofenac, NSAIDs, morphine, anesthetics, or any of the excipients of the study preparation, clinically significant lab or electrocardiography (ECG) result at baseline or screening, or had taken monoamine oxidase inhibitors, tryptophan, carbamazepine, or valproate ≤ 2 weeks prior to the study period. Aspirin (except for anti-platelet cardiac protection), NSAIDs, and other common analgesic drugs, centrally acting adjuvants, tranquilizers, or antihistamines were to be discontinued 24 h prior to study drug administration, and long acting NSAIDs or COX-2 inhibitors were to be discontinued 3 days prior to surgery.

Study design and outcomes

Both studies were multi-center, randomized, double-blind, placebo- and active comparator-controlled, repeated-dose, parallel-group phase III studies. In each study, patients were randomized to receive either HPβCD-diclofenac, ketorolac, or placebo, based on a computer-generated random number code. Clinical staff and patients were blinded to treatment group assignment (dose levels of individual study treatments in the orthopedic surgery population, however, were not blinded). All study medications were given as an IV bolus, with the first dose given ≤ 6 h following surgery and subsequent doses given every 6 hours until discharge or withdrawal/discontinuation (maximum 5 days post-first dose). Rescue medication (bolus IV morphine) was available upon patient request, up to once every 3 h after the initial study drug dose. If rescue medication did not provide adequate analgesia, the patient was withdrawn and given pain medication in accordance with the investigator’s usual practice.

In the abdominal/pelvic surgery study, patients were randomly assigned to receive either 18.75 mg or 37.5 mg HPβCD-diclofenac, 30 mg ketorolac, or placebo (saline). In the orthopedic surgery study, patients randomly assigned to the HPβCD-diclofenac group received a dosage of either 37.5 mg (standard dose), 18.75 mg (high-risk patients, based on age ≥ 65 years, preexisting renal insufficiency, or presence of NSAID-related GI risk factors), or 50 mg (high-weight (≥ 95 kg), no risk factors). Similarly, dosage in the ketorolac group was based on the absence or presence of risk factors (no risk factors and high-weight patients: 30 mg; high-risk patients: 15 mg).

Safety assessments

Overall safety assessments included physical examination, clinical laboratory tests, vital signs, 12-lead ECG, and AEs. Treatment-emergent bleeding AEs were recorded from baseline through a 30-37 day follow-up period, and were coded using the Medical Dictionary for Regulatory Activities (MedDRA), version 12.0. Hematology laboratory measurements (hemoglobin, hematocrit, erythrocytes, leukocytes, lymphocytes, neutrophils, platelets) were obtained at screening, 24 h post-first study drug dose, and at discharge or early termination (and in addition, at 5-9 days following first study drug dose in the abdominal/pelvic surgery study). For a given hematology measure, a shift to high was defined as a shift from a value within or below the normal range at screening to a value above the normal range during the study period. A shift to low was defined as a shift from a normal or high value at screening to a value below the normal range during the study period.

Statistical analysis

Safety analyses were performed on the pooled intent-to-treat (ITT) populations from both studies using SAS (Cary, NC, USA) version 9.1 or later. AE incidences were evaluated for all treatment groups, and relative risk (RR) ratios for active treatments versus placebo were calculated. RRs are presented as RR [95% confidence interval (CI)]. To further examine differences across treatment groups, p-values were calculated from ANOVA for numerical variables and Cochran–Mantel–Haenszel test for categorical variables. A p-value<0.05 was defined as statistically significant.

Results

Study population

In total, n=608 patients undergoing an orthopedic or abdominal/pelvic procedure were included in the analysis (n=318, 142, and 148 patients in the HPβCD-diclofenac, ketorolac, and placebo groups, respectively). The three treatment groups did not differ significantly with respect to mean patient age, procedure duration, or proportion of patients who were female, underwent an abdominal/pelvic or orthopedic procedure, or received concomitant anticoagulants (heparin, Coumadin/warfarin; Table 1). Similarly, when the proportion of patients receiving any medication with potential anticoagulant effects was examined, there were no significant differences across treatment groups.

  HPβCD-diclofenac (n=318) Ketorolac (n=142) Placebo (n=148) p-valuea
Mean age, years (SD) 48.9 (14.1) 48.0 (14.7) 48.6 (14.1) 0.81
Gender
Female, n (%) 233 (73.3) 107 (75.4) 107 (72.3) 0.83
Male, n (%) 85 (26.7) 35 (24.6) 41 (27.7)  
Procedure typeb
Abdominal/pelvic, n (%) 173 (54.4) 82 (57.7) 76 (51.4) 0.55
Orthopedic, n (%) 145 (45.6) 60 (42.3) 72 (48.6)  
Mean procedure duration, h (SD) 1.17 (0.69)c 1.13 (0.65)d 1.19 (0.71) 0.76
Mean doses received (SD) 7.3 ( 3.4) 7.5 ( 3.5) 6.2 ( 3.7) --
1-6 doses, n (%) 101 (31.8) 43 (30.3) 71 (48.0) --
7-8 doses, n (%) 150 (47.2) 67 (47.2) 53 (35.8) --
>8 doses, n (%) 67 (21.1) 32 (22.5) 24 (16.2) --
Concomitant anticoagulant
No, n (%) 258 (81.1) 113 (79.6) 113 (76.4) --
Yes, n (%) 60 (18.9) 29 (20.4) 35 (23.6) 0.49
Heparin, n (%)e 53 (16.7) 25 (17.6) 33 (22.3) 0.33
Coumadin, n (%) 12 (3.8) 6 (4.2) 4 (2.7) 0.77
Warfarin, n (%) 1 (0.3) 0 0 --
Concomitant medication with potential anticoagulant effectsf
No, n (%) 180 (56.6) 71 (50.0) 81 (54.7) --
Yes, n (%) 138 (43.4) 71 (50.0) 67 (45.3) 0.42
aFrom ANOVA for numerical variables and Cochran–Mantel–Haenszel test for categorical variables;
bMost common procedures (>5% of subjects in all treatment groups): abdominal hysterectomy, abdominal surgery, bunionectomy/foot bone, inguinal hernia repair, knee replacement, vaginal hysterectomy, other;
cn=316;
dn=141;
eIncludes heparin, heparin sodium, enoxaparin, enoxaparin sodium, Lovenox;
fIncludes acetic acid derivatives and related substances, heparin, other antithrombotics, oxicams, platelet aggregation inhibitors, propionic acid derivatives, salicylic acid and derivatives, vitamin K antagonists (Coumadin, warfarin).

Table 1: Summary of baseline demographics and surgical characteristics. SD=standard deviation.

Bleeding adverse events and hematology laboratory findings

Bleeding AEs were relatively uncommon across the study population (Table 2). In total, 11 bleeding AEs occurred in n=9/318 patients (2.8%) in the HPβCD-diclofenac group, 8 bleeding AEs occurred in n=8/142 patients (5.6%) in the ketorolac group, and 6 bleeding AEs occurred in n=4/148 patients (2.7%) in the placebo group. This included two patients in the HPβCD-diclofenac group who experienced 2 bleeding AEs (one patient with two epistaxis events, one patient with two vaginal hemorrhage events), and one patient in the placebo group who experienced 3 bleeding AEs (hematemesis, hematocrit decreased, and hemoptysis). In the HPβCD-diclofenac group, all bleeding AEs occurred in patients who underwent a surgical procedure <2 h in duration, n=7/11 AEs were in orthopedic surgery patients, and n=9/11 were in patients ≤ 65 years old. Additionally, n=10/11 bleeding AEs in the HPβCD-diclofenac group occurred in patients receiving ≥ 7 doses (n=217 total patients). Similarly, n=5/8 bleeding AEs in the ketorolac group occurred in patients receiving ≥ 7 doses of ketorolac (n=99 total patients).

  HPβCD-diclofenac (n=318) Ketorolac (n=142) Placebo (n=148)
Total bleeding AEs 11 8 6
Patients with ≥1 treatment-emergent bleeding AE, n (%) 9 (2.8)a 8 (5.6) 4 (2.7)b
Risk ratio vs. placebo [95% CI] 1.05 [0.33, 3.35]; p=0.93c 2.08 [0.64, 6.77]; p=0.22c --
Bleeding AEs by preferred term, n (%)d
Rectal hemorrhage 2 (0.6) 2 (1.4) 0
Incision site hematoma 0 3 (2.1) 0
Vaginal hemorrhage 1 (0.3) 0 2 (1.4)
Epistaxis 2 (0.6) 0 0
Eye hemorrhage 0 1 (0.7) 0
Hemarthrosis 1 (0.3) 0 0
Hematemesis 0 0 1 (0.7)
Hematocrit decreased 0 0 1 (0.7)
Hematoma 1 (0.3) 0 0
Hematuria 1 (0.3) 0 0
Hemoptysis 0 0 1 (0.7)
Hemorrhagic anemia 1 (0.3) 0 0
Hemorrhagic ovarian cyst 0 0 1 (0.7)
Periorbital hematoma 0 1 (0.7) 0
Petechiae 0 1 (0.7) 0
Total postoperative anemia AEs 7 2 3
Patients with postoperative anemia AE, n (%) 7 (2.2) 2 (1.4) 3 (2.0)
Risk ratio vs. placebo [95% CI] 1.09 [0.28, 4.14]; p=0.83c 0.69 [0.12, 4.10]; p=0.76c --
aIncludes 1 patient with 2 epistaxis events and 1 patient with 2 vaginal hemorrhage events;
bIncludes 1 patient with 1 event each of hematemesis, hematocrit decreased, and hemoptysis;
cFrom Cochran–Mantel–Haenszel test comparing treatment and placebo;
dn (%) represents number and percentage of patients with a given bleeding AE.

Table 2: Summary of treatment-emergent bleeding-related adverse events in the study population. AE = adverse event; CI = confidence interval.

The overall bleeding AE risk was not significantly increased or decreased with HPβCD-diclofenac versus placebo (RR: 1.05 [0.33, 3.35]; p=0.93). Ketorolac was likewise not associated with a significant difference in bleeding AE risk versus placebo (RR: 2.08 [0.64, 6.77]; p=0.22). The most common bleeding AEs in the pooled population were rectal hemorrhage (n=2 patients receiving HPβCD-diclofenac and n=2 patients receiving ketorolac) and incision site hematoma (n=3 patients in the ketorolac group). In addition to bleeding AEs, the incidence of postoperative anemia was likewise low (n=7/318 patients (2.2%), n=2/142 patients (1.4%), and n=3/148 patients (2.0%) in the HPβCD-diclofenac, ketorolac, and placebo groups, respectively).

The majority of bleeding AEs were mild or moderate in severity, with only 1 AE judged to be severe (hematoma in a female patient who underwent abdominal hysterectomy and received HPβCD-diclofenac; Table 3). Similarly, only two bleeding AEs were judged to be treatment-related (incision site hematoma in a patient in the ketorolac group; hematemesis in a patient who received placebo) and both were mild in severity.

  HPβCD-diclofenac (n=318) Ketorolac (n=142) Placebo (n=148)
Bleeding AE severity
Number of patients with AE that was:
Mild, n (%) 6 (1.9) 7 (4.9) 2 (1.4)
Moderate, n (%) 2 (0.6) 1 (0.7) 2 (1.4)
Severe, n (%) 1 (0.3)a 0 0
Relationship to treatment
Not related to treatment, n (%) 9 (2.8) 7 (4.9) 3 (2.0)
Treatment-related, n (%) 0 1 (0.7)b 1 (0.7)c
aHematoma in a patient who underwent abdominal hysterectomy;
bIncision site hematoma in a patient who underwent partial colectomy;
cHematemesis in a patient who underwent bunionectomy/foot bone procedure.

Table 3: Summary of bleeding adverse events by severity and relationship to treatment. AE = Adverse Event.

Among patients receiving concomitant anticoagulants, bleeding AEs occurred in n=3/60 patients (5.0%) receiving HPβCD-diclofenac, n=2/29 patients (6.9%) receiving ketorolac, and n=0/35 patients receiving placebo (Table 4; p=0.19 and p=0.11 for HPβCD-diclofenac and ketorolac versus placebo, respectively). On expansion of this analysis to include all concomitant medications with potential anticoagulant effects, bleeding AE incidences were 3.6% (n=5/138), 4.2% (n=3/71), and 1.5% (n=1/67) for the HPβCD-diclofenac, ketorolac, and placebo groups, respectively (p=0.39 and p=0.35 for HPβCD-diclofenac and ketorolac versus placebo, respectively).

  HPβCD-diclofenac (n=60) Ketorolac (n=29) Placebo (n=35)
Patients receiving anticoagulantsa,b
Total bleeding AEs 4 2 0
Patients with ≥1 treatment-emergent bleeding AE, n (%) 3 (5.0) 2 (6.9) 0
Bleeding AEs by preferred term, n (%)
Epistaxis 1 (1.7)c 0 0
Hematuria 1 (1.7) 0 0
Incision site hematoma 0 1 (3.4)c 0
Rectal hemorrhage 1 (1.7) 1 (3.4) 0
aIncludes heparin and vitamin K antagonists (Coumadin/warfarin);
bFor patients receiving any medication with potential anticoagulant effects (includes acetic acid derivatives and related substances, heparin, other antithrombotics, oxicams, platelet aggregation inhibitors, propionic acid derivatives, salicylic acid and derivatives, vitamin K antagonists), incidences of bleeding AEs were 3.6% (n=5/138), 4.2% (n=3/71), and 1.5% (n=1/67) for the HPβCD-diclofenac, ketorolac, and placebo groups, respectively;
cIncludes 1 patient with two separate AEs of epistaxis;
dAE judged by investigator to be related to treatment

Table 4: Summary of bleeding adverse events in patients receiving anticoagulants. AE = adverse event.

With respect to timing relative to anticoagulant use, all bleeding AEs but one appeared to be concurrent with anticoagulant use, i.e., AE onset was on the same day as the anticoagulant was first received or prior to the last day of treatment with the anticoagulant (Supplemental Table 1). The lone exception was an incidence of incision site hematoma in a patient in the ketorolac treatment group, for whom the AE occurred on postsurgical day 8, one day following the end of the patient’s enoxaparin sodium regimen for DVT prophylaxis; this AE was judged by the investigator to be treatment-related.

Among the most common hematology laboratory parameter shifts occurring in the pooled study population were shifts to low hematocrit and hemoglobin levels. Among patients receiving HPβCD-diclofenac, a shift to a low hematocrit or hemoglobin level during the study period occurred in 35.7% and 28.3% of patients, respectively (versus 31.4% and 21.5%, respectively for placebo and 47.6% and 38.4%, respectively for ketorolac; Figure 1). Shifts in platelet counts were relatively uncommon (<3.0% in all treatment groups). Neutrophil and leukocyte counts were the most common laboratory parameters to demonstrate a shift to high values during the study period (Supplemental Figure 1). Stratification of patients revealed a general trend toward more frequent shifts to low hemoglobin , low erythrocytes, and low lymphocytes with increasing age for all treatment groups, and a trend toward more frequent shifts to low hemoglobin and low erythrocytes with longer surgery duration.

anesthesia-clinical-research-hematology-laboratory-shifts

Figure 1: Summary of hematology laboratory shifts from baseline in the study population. The number of patients with a given shift tended to be lowest for patients receiving HPβCD-diclofenac and placebo. Shift to high = shift from normal or low value at screening to a high value during study. Shift to low = shift from normal or high value at screening to a low value during study. Hematocrit was measured in %, hemoglobin in g/L, and platelet count in 109/L. For each measurement, the total number of patients (HPβCD-diclofenac, ketorolac, placebo) was as follows: hematocrit (277, 124, 121); hemoglobin (279, 125, 121); platelets (274, 124, 120).

Discussion

Bleeding risks are a key concern both with NSAID use and in the context of surgery. As a surgical complication, bleeding can be associated with increased morbidity and mortality [43]. The results of this pooled analysis of data from two phase III placebo- and active comparator-controlled clinical trials reveal a relatively low incidence of bleeding AEs among patients receiving HPβCD-diclofenac for ≤ 5 days for the management of acute postsurgical pain. During this acute treatment timeframe, there was no significant difference in bleeding AE risk with HPβCD-diclofenac versus placebo. The low incidence of treatment-emergent bleeding AEs (2.8%) and lack of treatment-related bleeding AEs in patients receiving HPβCD-diclofenac are in line with data from an open-label phase III safety study in which n=32/971 surgical patients (3.3%) receiving HPβCD-diclofenac had a bleeding-related AE and a total of 2 treatment-related bleeding AEs occurred [38]. Notably, the open-label safety study included large proportions of at-risk patients (35% of patients were ≥ 65 years old, and >60% received concomitant anticoagulants). Furthermore, the reported bleeding-related AE rates in the present study are consistent with rates reported for similar surgical groups in large retrospective studies [43,44].

In addition to factors such as age and preexisting medical conditions, postoperative or concomitant use of NSAIDs and anticoagulants has been identified as a risk factor for clinically relevant bleeding AEs [45,46]. In light of this important consideration, the present study examined bleeding AE incidences in patients receiving anticoagulants (heparin, coumadin/warfarin), as well as all drugs with potential anticoagulant effects. Though the overall number of patients receiving anticoagulants in each treatment group was relatively limited, none of the AEs occurring in patients receiving a concomitant anticoagulant was judged to be severe by the study investigator. All but one bleeding AE in this group appeared to occur concurrently with anticoagulant treatment, suggesting a potential contribution of anticoagulant treatment to the observed AEs. It is important to note, however, that an open-label large phase III HPβCD-diclofenac safety study found no difference in bleeding-related AE incidence between patients receiving and not receiving concomitant anticoagulants [38].

NSAIDs have also been reported to be associated with platelet function inhibition and increased perioperative bleeding [47]. However, data from healthy subjects have demonstrated that therapeutic dosages of diclofenac produce less GI damage and bleeding than aspirin, indomethacin, or naproxen, and have little effect on platelet aggregation or bleeding time [48]. Recently, a single-dose study demonstrated significantly less disruption of platelet function following administration of HPβCD-diclofenac than ketorolac or acetylsalicylic acid (ASA) [42]. This reduced effect of HPβCD-diclofenac on platelet function could be related to diclofenac’s balanced inhibition of COX-1 and -2, as selective COX-1 inhibition is associated with GI and platelet adverse effects [42,49,50].

In addition to findings in line with previous studies of HPβCD-diclofenac, the bleeding AE rates observed in the present study compare favorably with those reported in previous studies of other IV NSAIDs and acetaminophen [18,21,46,51-58]. In one study of patients undergoing major surgery, a 1.1% incidence of GI bleeding was reported in patients receiving ketorolac postsurgically (1.0% for the comparators diclofenac and ketoprofen) [46]. Similarly, a study in patients undergoing elective orthopedic and abdominal/pelvic surgical procedures found no increased risk of bleeding AEs in patients receiving IV ibuprofen versus placebo [57].

Further, the results of the present analysis support previous studies that suggest that diclofenac is not associated with added risk of postoperative bleeding complications versus placebo in the postsurgical setting [59-61]. Similarly, a study in patients undergoing transurethral prostate resection reported no added bleeding risk with the use of intramuscular diclofenac versus IV paracetamol [62]. It is important to note, however, that higher postoperative blood loss has been reported with administration of diclofenac versus placebo following breast surgery [63].

In addition to bleeding AEs, the prevalence of shifts in common laboratory hematology parameters was relatively low across treatment groups in the present study, and the shifts that were observed (e.g. an initial decrease in hemoglobin, increases in leukocyte and neutrophil counts) were in line with typical hematological shifts that can occur following surgical procedures [64-66].

In summary, the data from the present pooled analysis suggest that short-term postoperative use of HPβCD-diclofenac was not associated with significant bleeding risk versus placebo over the time window examined, with a similar overall finding for the active comparator ketorolac. The two studies included in the analysis were similar in terms of setting, drug dosage, dosing regimen, and safety analyses, which all lend themselves to an effective pooled analysis. Notably, the treatment regimen in the included studies was relatively brief (≤ 5 days, with most patients receiving ≤ 8 doses of study medication), and therefore does not represent long-term use, which is more typically associated with bleeding-related adverse events than acute postsurgical use [18,21,23,67-69]. The population size (608 total patients) represents a limitation of this analysis, such that conclusions about the relative safety of HPβCD-diclofenac with respect to bleeding must be inferred with caution. Still, the findings are supportive of the safety profile of repeated-dose HPβCD-diclofenac when used for short-term management of acute postoperative pain. This safety profile, in addition to the demonstrated efficacy of HPβCD-diclofenac for acute moderate-to-severe pain, suggest that this diclofenac formulation can play an important role in managing postsurgical pain in appropriate patient populations, particularly in light of the potential importance of acute pain management in avoiding unfavorable postoperative outcomes such as the transition to chronic postsurgical pain [70]. Future studies, including retrospective analyses of large postsurgical populations using electronic health (EHR) data may be able to provide further insight into the relative safety of HPβCD-diclofenac versus other IV analgesics with respect to bleeding and hematology outcomes.

Acknowledgements

The authors thank Scott Paluszkiewicz, Ph.D., Tejaswi Worlikar, M.S. and Fred Peyerl, Ph.D. of Boston Strategic Partners, Inc. for editorial assistance. Hospira, Inc. provided support to Boston Strategic Partners, Inc. for editorial assistance.

Disclosures

This study was sponsored by Javelin Pharmaceuticals, Inc., Cambridge, MA (acquired in 2010 by Hospira, Inc., Lake Forest, IL) and Hospira, Inc. Hospira, Inc. provided support to Boston Strategic Partners, Inc. for editorial assistance. TJ Gan, N Singla, and SE Daniels were paid study investigators. PG Lacouture was an employee of Hospira, Inc. at the time that the study was initiated. O Johnson and CRD Reyes are employees of Hospira, Inc. DA Hamilton was full-time Chief Operating Officer for the study sponsor during the trials included in the present study, and served a consultant to Hospira, Inc. following its acquisition of Javelin Pharmaceuticals in 2010. DB Carr was the full-time Chief Medical Officer for the study sponsor during the trials included in the present study, and served as a consultant to Hospira, Inc. following its acquisition of Javelin Pharmaceuticals in 2010.

References

  1. Hutchison RW (2007) Challenges in acute post-operative pain management. Am J Health Syst Pharm 64: S2-5.
  2. Argoff CE (2014) Recent management advances in acute postoperative pain. Pain Pract 14: 477-487.
  3. Joshi GP, Ogunnaike BO (2005) Consequences of inadequate postoperative pain relief and chronic persistent postoperative pain. AnesthesiolClin North America 23: 21-36.
  4. Kehlet H, Jensen TS, Woolf CJ (2006) Persistent postsurgical pain: risk factors and prevention. Lancet 367: 1618-1625.
  5. Lacouture PG (2010) Persistent post-operative pain. In: Sinatra RS, Jahr JS, Watkins-Pitchford JM (eds) Essence of Analgesia and Analgesics. (1st edn), Cambridge University Press, London.
  6. Macintyre PE, Schug SA, Scott DA, Visser EJ, Walker S (2010) Acute Pain Management: Scientific Evidence. (3rdedn), Australian and New Zealand College of Anaesthetists and Faculty of Pain Medicine, Melbourne, Australia.
  7. Carr DB, Jacox AK, Chapman CR(1992) Acute Pain Management: Operative or Medical Procedures and Trauma. Clinical Practice Guideline Number 1. AHCPR Publication No. 92 00, Rockville, Maryland: Agency for Healthcare Research and Quality, Public Health Service, US Department of Health and Human Services.
  8. American Society of Anesthesiologists Task Force on Acute Pain Management (2012) Practice guidelines for acute pain management in the perioperative setting: an updated report by the American Society of Anesthesiologists Task Force on Acute Pain Management. Anesthesiology 116: 248-273.
  9. Bainbridge D, Cheng DC, Martin JE, Novick R; Evidence-Based Perioperative Clinical Outcomes Research (EPiCOR) Group (2006) NSAID-analgesia, pain control and morbidity in cardiothoracic surgery. Can J Anaesth 53: 46-59.
  10. National (US) Pharmaceutical Council and Joint Commission on Accreditation of Healthcare Organizations (2001) Pain: Current Understanding of Assessment, Management, and Treatments, National Pharmaceutical Council, Reston, VA.
  11. Maund E, McDaid C, Rice S, Wright K, Jenkins B, et al. (2011) Paracetamol and selective and non-selective non-steroidal anti-inflammatory drugs for the reduction in morphine-related side-effects after major surgery: a systematic review. Br J Anaesth 106: 292-297.
  12. Skinner HB (2004) Multimodal acute pain management. Am J Orthop (Belle Mead NJ) 33: 5-9.
  13. Cepeda MS, Carr DB, Miranda N, Diaz A, Silva C, et al. (2005) Comparison of morphine, ketorolac, and their combination for postoperative pain: results from a large, randomized, double-blind trial. Anesthesiology 103: 1225-1232.
  14. Elia N, Lysakowski C, Tramèr MR (2005) Does multimodal analgesia with acetaminophen, nonsteroidalantiinflammatory drugs, or selective cyclooxygenase-2 inhibitors and patient-controlled analgesia morphine offer advantages over morphine alone? Meta-analyses of randomized trials. Anesthesiology 103: 1296-1304.
  15. Marret E, Kurdi O, Zufferey P, Bonnet F (2005) Effects of nonsteroidalantiinflammatory drugs on patient-controlled analgesia morphine side effects: meta-analysis of randomized controlled trials. Anesthesiology 102: 1249-1260.
  16. Feldman HI, Kinman JL, Berlin JA, Hennessy S, Kimmel SE, et al. (1997) Parenteral ketorolac: the risk for acute renal failure. Ann Intern Med 126: 193-199.
  17. Souter AJ, Fredman B, White PF (1994) Controversies in the perioperative use of nonsterodialantiinflammatory drugs. AnesthAnalg 79: 1178-1190.
  18. Strom BL, Berlin JA, Kinman JL, Spitz PW, Hennessy S, et al. (1996) Parenteral ketorolac and risk of gastrointestinal and operative site bleeding. A postmarketing surveillance study. JAMA 275: 376-382.
  19. Whelton A1 (1999) Nephrotoxicity of nonsteroidal anti-inflammatory drugs: physiologic foundations and clinical implications. Am J Med 106: 13S-24S.
  20. Sostres C, Gargallo CJ, Arroyo MT, Lanas A (2010) Adverse effects of non-steroidal anti-inflammatory drugs (NSAIDs, aspirin and coxibs) on upper gastrointestinal tract. Best Pract Res ClinGastroenterol 24: 121-132.
  21. Kroll PB, Meadows L, Rock A, Pavliv L (2011) A multicenter, randomized, double-blind, placebo-controlled trial of intravenous ibuprofen (i.v.-ibuprofen) in the management of postoperative pain following abdominal hysterectomy. Pain Pract 11: 23-32.
  22. US FDA Department of Health and Human Services (2006) Decision memorandum: analysis and recommendations for agency action: COX-2 selective and nonselective NSAIDs.
  23. Vonkeman HE, van de Laar MA (2010) Nonsteroidal anti-inflammatory drugs: adverse effects and their prevention. Semin Arthritis Rheum 39: 294-312.
  24. Risser A, Donovan D, Heintzman J, Page T (2009) NSAID prescribing precautions. Am Fam Physician 80: 1371-1378.
  25. Frampton C, Quinlan J (2009) Evidence for the use of non-steroidal anti-inflammatory drugs for acute pain in the post anaesthesia care unit. J PerioperPract 19: 418-423.
  26. Wehling M1 (2014) Non-steroidal anti-inflammatory drug use in chronic pain conditions with special emphasis on the elderly and patients with relevant comorbidities: management and mitigation of risks and adverse effects. Eur J ClinPharmacol 70: 1159-1172.
  27. Langman MJ, Weil J, Wainwright P, Lawson DH, Rawlins MD, et al. (1994) Risks of bleeding peptic ulcer associated with individual non-steroidal anti-inflammatory drugs. Lancet 343: 1075-1078.
  28. Aubrun F, Marmion F (2007) The elderly patient and postoperative pain treatment. Best Pract Res ClinAnaesthesiol 21: 109-127.
  29. Hernández-Díaz S, García-Rodríguez LA (2001) Epidemiologic assessment of the safety of conventional nonsteroidal anti-inflammatory drugs. Am J Med 110 Suppl 3A: 20S-7S.
  30. Caradoc-Davies TH (1984) Nonsteroidal anti-inflammatory drugs, arthritis, and gastrointestinal bleeding in elderly in-patients. Age Ageing 13: 295-298.
  31. Møiniche S, Rømsing J, Dahl JB, Tramèr MR (2003) Nonsteroidalantiinflammatory drugs and the risk of operative site bleeding after tonsillectomy: a quantitative systematic review. AnesthAnalg 96: 68-77, table of contents.
  32. Todd PA, Sorkin EM (1988) Diclofenac sodium. A reappraisal of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy. Drugs 35: 244-285.
  33. Barden J, Edwards J, Moore RA, McQuay HJ (2004) Single dose oral diclofenac for postoperative pain. Cochrane Database Syst Rev CD004768.
  34. Gan TJ (2010) Diclofenac: an update on its mechanism of action and safety profile. Curr Med Res Opin 26: 1715-1731.
  35. Loftsson T, Hreinsdóttir D, Másson M (2005) Evaluation of cyclodextrinsolubilization of drugs. Int J Pharm 302: 18-28.
  36. Daniels S, Melson T, Hamilton DA, Lang E, Carr DB (2013) Analgesic Efficacy and Safety of a Novel Injectable Formulation of Diclofenac Compared with Intravenous Ketorolac and Placebo after Orthopedic Surgery: A Multicenter, Randomized, Double-Blinded, Multiple-Dose Trial. Clin J Pain 29: 655-663.
  37. Gan TJ, Daniels SE, Singla N, Hamilton DA, Carr DB (2012) A novel injectable formulation of diclofenac compared with intravenous ketorolac or placebo for acute moderate-to-severe pain after abdominal or pelvic surgery: a multicenter, double-blind, randomized, multiple-dose study. AnesthAnalg 115: 1212-1220.
  38. Chelly JE, Singla SK, Melson TI, Lacouture PG, Paadre S, et al. (2013) Safety of a novel parenteral formulation of diclofenac after major orthopedic or abdominal/pelvic surgery in a population including anticoagulated, elderly or renally insufficient patients: an open-label, multiday, repeated dose clinical trial. Pain Med 14: 749-761.
  39. Christensen K, Daniels S, Bandy D, Ernst CC, Hamilton DA, et al. (2011) A double-blind placebo-controlled comparison of a novel formulation of intravenous diclofenac and ketorolac for postoperative third molar extraction pain. AnesthProg 58: 73-81.
  40. Leeson RM, Harrison S, Ernst CC, Hamilton DA, Mermelstein FH, et al. (2007) Dyloject, a novel injectable diclofenac formulation, offers greater safety and efficacy than voltarol for postoperative dental pain. RegAnesth Pain Med 32: 303-310.
  41. Colucci RD, Wright C, Mermelstein FH, Gawarecki DG, Carr DB (2009) Dyloject®, a novel injectable diclofenacsolubilised with cyclodextrin: reduced incidence of thrombophlebitis compared to injectable diclofenacsolubilised with polyethylene glycol and benzyl alcohol. Acute Pain 11: 15-21.
  42. Bauer KA, Gerson W, Wright C 4th, Wang J, McNicol E, et al. (2010) Platelet function following administration of a novel formulation of intravenous diclofenac sodium versus active comparators: a randomized, single dose, crossover study in healthy male volunteers. J ClinAnesth 22: 510-518.
  43. Stokes ME, Ye X, Shah M, Mercaldi K, Reynolds MW, et al. (2011) Impact of bleeding-related complications and/or blood product transfusions on hospital costs in inpatient surgical patients. BMC Health Serv Res 11: 135.
  44. Ghaferi AA, Birkmeyer JD, Dimick JB (2009) Variation in hospital mortality associated with inpatient surgery. N Engl J Med 361: 1368-1375.
  45. Davidson BL, Verheijen S, Lensing AW, Gebel M, Brighton TA, et al. (2014) Bleeding risk of patients with acute venous thromboembolism taking nonsteroidal anti-inflammatory drugs or aspirin. JAMA Intern Med 174: 947-953.
  46. Forrest JB, Camu F, Greer IA, Kehlet H, Abdalla M, et al. (2002) Ketorolac, diclofenac, and ketoprofen are equally safe for pain relief after major surgery. Br J Anaesth 88: 227-233.
  47. Hynes D, McCarroll M, Hiesse-Provost O (2006) Analgesic efficacy of parenteral paracetamol (propacetamol) and diclofenac in post-operative orthopaedic pain. ActaAnaesthesiolScand 50: 374-381.
  48. McCormack PL, Scott LJ (2008) Diclofenac sodium injection (Dyloject): in postoperative pain. Drugs 68: 123-130.
  49. Chaiamnuay S, Allison JJ, Curtis JR (2006) Risks versus benefits of cyclooxygenase-2-selective nonsteroidalantiinflammatory drugs. Am J Health Syst Pharm 63: 1837-1851.
  50. Warner TD, Giuliano F, Vojnovic I, Bukasa A, Mitchell JA, et al. (1999) Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. ProcNatlAcadSci U S A 96: 7563-7568.
  51. Candiotti KA, Bergese SD, Viscusi ER, Singla SK, Royal MA, et al. (2010) Safety of multiple-dose intravenous acetaminophen in adult inpatients. Pain Med 11: 1841-1848.
  52. Jahr JS, Breitmeyer JB, Pan C, Royal MA, Ang RY (2012) Safety and efficacy of intravenous acetaminophen in the elderly after major orthopedic surgery: subset data analysis from , randomized, placebo-controlled trials. Am J Ther 19: 66-75.
  53. Fransen M, Anderson C, Douglas J, MacMahon S, Neal B, et al. (2006) Safety and efficacy of routine postoperative ibuprofen for pain and disability related to ectopic bone formation after hip replacement surgery (HIPAID): randomised controlled trial. BMJ 333: 519.
  54. Marret E, Flahault A, Samama CM, Bonnet F (2003) Effects of postoperative, nonsteroidal, antiinflammatory drugs on bleeding risk after tonsillectomy: meta-analysis of randomized, controlled trials. Anesthesiology 98: 1497-1502.
  55. Morris PE, Promes JT, Guntupalli KK, Wright PE, Arons MM (2010) A multi-center, randomized, double-blind, parallel, placebo-controlled trial to evaluate the efficacy, safety, and pharmacokinetics of intravenous ibuprofen for the treatment of fever in critically ill and non-critically ill adults. Crit Care 14: R125.
  56. O'Hara DA, Fanciullo G, Hubbard L, Maneatis T, Seuffert P, et al. (1997) Evaluation of the safety and efficacy of ketorolac versus morphine by patient-controlled analgesia for postoperative pain. Pharmacotherapy 17: 891-899.
  57. Southworth S, Peters J, Rock A, Pavliv L (2009) A multicenter, randomized, double-blind, placebo-controlled trial of intravenous ibuprofen 400 and 800 mg every 6 hours in the management of postoperative pain. ClinTher 31: 1922-1935.
  58. Thwaites BK, Nigus DB, Bouska GW, Mongan PD, Ayala EF, et al. (1996) Intravenous ketorolac tromethamine worsens platelet function during knee arthroscopy under spinal anesthesia. AnesthAnalg 82: 1176-1181.
  59. Al-Waili NS (2001) Efficacy and safety of repeated postoperative administration of intramuscular diclofenac sodium in the treatment of post-cesarean section pain: a double-blind study. Arch Med Res 32: 148-154.
  60. Fayaz MK, Abel RJ, Pugh SC, Hall JE, Djaiani G, et al. (2004) Opioid-sparing effects of diclofenac and paracetamol lead to improved outcomes after cardiac surgery. J CardiothoracVascAnesth 18: 742-747.
  61. Laitinen J, Nuutinen L (1992) Intravenous diclofenac coupled with PCA fentanyl for pain relief after total hip replacement. Anesthesiology 76: 194-198.
  62. Kara C, Resorlu B, Cicekbilek I, Unsal A (2010) Analgesic efficacy and safety of nonsteroidal anti-inflammatory drugs after transurethral resection of prostate. IntBraz J Urol 36: 49-54.
  63. Legeby M, Sandelin K, Wickman M, Olofsson C (2005) Analgesic efficacy of diclofenac in combination with morphine and paracetamol after mastectomy and immediate breast reconstruction. ActaAnaesthesiolScand 49: 1360-1366.
  64. Hughes SF, Hendricks BD, Edwards DR, Maclean KM, Bastawrous SS, et al. (2010) Total hip and knee replacement surgery results in changes in leukocyte and endothelial markers. J Inflamm (Lond) 7: 2.
  65. Lee JH, Lee JH, Kim JB, Lee HS, Lee DY, et al. (2012) Normal range of the inflammation related laboratory findings and predictors of the postoperative infection in spinal posterior fusion surgery. ClinOrthopSurg 4: 269-277.
  66. Evans S, O'Loughlin E, Bruce J (2011) Retrospective audit of blood transfusion and comparison with haemoglobin concentration in patients undergoing elective primary and revision lower limb arthroplasty. Anaesth Intensive Care 39: 480-485.
  67. Derry P, Derry S, Moore RA, McQuay HJ (2009) Single dose oral diclofenac for acute postoperative pain in adults. Cochrane Database Syst Rev 2: CD004768.
  68. García Rodríguez LA, Jick H (1994) Risk of upper gastrointestinal bleeding and perforation associated with individual non-steroidal anti-inflammatory drugs. Lancet 343: 769-772.
  69. Gutthann SP, Garcia Rodriguez LA, Raiford DS (1997) Individual nonsteroidalantiinflammatory drugs and other risk factors for upper gastrointestinal bleeding and perforation. Epidemiology 8: 18-24.
  70. Shipton EA (2011) The transition from acute to chronic post surgical pain. Anaesth Intensive Care 39: 824-836.
Citation: Gan TJ, Singla N, Daniels SE, Hamilton DA, Lacouture PG, et al. (2015) Clinical and Laboratory Hematologic Findings in Patients Receiving Repeated-Dose Injectable HPβCD-Diclofenac for Acute Postoperative Pain: Pooled Analysis of Two Randomized Controlled Phase III Clinical Trials. J Anesth Clin Res 6:538.

Copyright: © 2015 Gan TJ, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top