You are on page 1of 27

Pain Medicine 2017; 0: 1–27

doi: 10.1093/pm/pnx228

Review Article
Burn Pain: A Systematic and Critical Review of
Epidemiology, Pathophysiology, and Treatment

Michael Morgan, PhD,* Jennifer R. Deuis, PhD,* were systemically reviewed. In addition, a critical re-
Majbrit Frøsig-Jørgensen, MSc,* Richard J. Lewis, view was performed on the pathophysiology of
PhD,* Peter J. Cabot, PhD,† Paul D. Gray, MBBS, burn pain and animal models of burn pain.
PhD,‡,§ and Irina Vetter, PhD,*,†
Results. The search on the epidemiology of burn in-
jury yielded a total of 163 publications of interest,
*Centre for Pain Research, Institute for Molecular
72 of which fit the inclusion/exclusion criteria, with
Bioscience, The University of Queensland, St Lucia, no publications providing epidemiological data on
Queensland, Australia; †School of Pharmacy, The burn injury pain management outcomes. The search
University of Queensland, Wooloongabba, on the treatment of burn pain yielded a total of 213
Queensland, Australia; ‡Tess Cramond publications, 14 of which fit the inclusion/exclusion
Multidisciplinary Pain Centre, Royal Brisbane & criteria, highlighting the limited amount of evidence
Women’s Hospital, Metro North Health, Herston, available on the treatment of burn-induced pain.
Queensland, Australia; §School of Medicine, The
University of Queensland, Herston, Queensland, Conclusions. The pathophysiology of burn pain is
poorly understood, with limited clinical trials avail-
Australia
able to assess the effectiveness of analgesics in
Correspondence to: Irina Vetter, PhD, Institute for burn patients. Further studies are needed to identify
Molecular Bioscience, 306 Carmody Road, The new pharmacological targets and treatments for the
University of Queensland, St Lucia, Brisbane, effective management of burn injury pain.
Queensland 4072, Australia. Tel: þ61-7-3346-2660;
Fax: þ61-7-3346-2101; E-mail: i.vetter@uq.edu.au. Key Words. Burn Pain; Mechanisms; Analgesia;
Inflammation; Neuropathic Pain
Disclosures and conflicts of interest: The authors de-
clare no conflicts of interest. Dr. Vetter is supported
Introduction
by an Australian Research Council Future Fellowship,
and Prof. Lewis is supported by a National Health and Pain is a sensory response that normally helps the body
Medical Research Council Principle Investigator to respond to a source of danger or tissue damage and
fellowship. facilitates protection for tissue repair. Acute pain arises
Authors Michael Morgan and Jennifer R. Deuis con- from a range of noxious stimuli including heat, cold, me-
chanical stimuli, and chemicals. In the case of burn-
tributed equally to this work.
induced pain, damage to peripheral sensory neurons
and inflammatory processes initiated by the injury exac-
erbate this acute response and transform burn pain into
a complex symptom comprising multiple components,
including ongoing background pain and procedural pain
Abstract during surgical interventions and dressing changes. In
addition, burn-induced pain can persist beyond the ini-
Objective. This review aims to examine the avail- tial injury and develop into chronic pain with neuropathic
able literature on the epidemiology, pathophysiol- features.
ogy, and treatment of burn-induced pain.
Typically, the degree of pain is largely determined by
Methods. A search was conducted on the epidemi- how deep the burn penetrates, with areas of superficial
ology of burn injury and treatment of burn pain uti- burns resulting in greater pain than areas of full-
lizing the database Medline, and all relevant articles thickness burns, where the nerve endings are

C 2017 American Academy of Pain Medicine. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
V 1
Morgan et al.

Figure 1 The severity of a burn injury is determined by the depth of tissue injury. The skin is intensely innervated with
many morphologically and functionally distinct sensory nerve endings that respond to a multitude of non-noxious and
noxious stimuli. Noxious heat stimuli are generally conducted to the dorsal horn of the spinal cord via nociceptive Ad
and C fiber neurons. Only the epidermis is affected in epidermal or superficial epidermal burns while increasing dam-
age to the dermis occurs in mid-dermal, deep-dermal and full-thickness burns.

functionally compromised or lost. However, full- skin burnt [3] and is determined according to the “Rule
thickness burns are typically surrounded by areas of of Nines.” The TBSA, together with burn depth, the
more superficial injury, so that, clinically, patients with presence of inhalation burn, age, and gender, is used to
full-thickness burns report as much pain as those with a calculate the severity of a burn injury using scales such
superficial injury [1]. The duration of pain can last from as the Abbreviated Burn Severity Index (Table 1) [4] that
days to months. provide an estimate of prognosis for the patients and
the method of treatment.
The skin, the largest organ of the human body by
weight, plays many essential roles, including maintaining The aim of this review is to provide an overview of burn
body homeostasis, providing an immunological barrier, injury epidemiology, current treatments for burn injury,
and contributing to sensations of touch and pain. The and the mediators and mechanisms underlying burn in-
skin consists of two primary layers (Figure 1): the outer jury pain. We conducted a systematic evaluation of the
epidermis, which is predominantly made up of keratino- literature regarding the epidemiology of burn pain, dis-
cytes plus melanocytes, Merkel cells, and Langerhans cuss the advantages and limitations of various models
cells; and an inner dermis, comprising connective tis- of burn injury, summarize key burn injury pain mecha-
sue, fibroblasts, macrophages, and adipocytes. The nisms, and provide an overview of common approaches
skin is intensely innervated with many morphologically to treating burn pain.
and functionally distinct sensory nerve endings that re-
spond to a multitude of non-noxious and noxious stim- Methods
uli. Noxious heat stimuli are generally conducted to the
dorsal horn of the spinal cord via nociceptive Ad and C Epidemiology of Burn Injuries
fiber neurons.
A systematic search was performed on Medline
The classification of a burn injury is dependent both on (October 18–19, 2016) to retrieve studies related to the
the depth and area of the burn. The depth of burn inju- following search key word combinations: burn; burns;
ries was first classified by Peter Lowe in 1597 and mod- epidemiology; mortality; gender; age. Articles published
ified by Fabricius in 1610 to the terms “first-, second-, between January 2000 and October 2016 in English
and third-degree burns.” This remained largely were then selected from the list, along with selected
unchanged until recently, when a new system described references informing the results of the retrieved studies.
burn injuries as either epidermal, superficial epidermal, Articles were classified according the level of evidence,
mid-dermal, deep dermal, and full thickness [2]. The as detailed in Table 2. Searches for national registries
burn coverage, also called the total body surface area for burn injury were also included in the study. Articles
(TBSA), provides an estimate of the percentage of the were excluded if they focused on a particular

2
Burn Pain

Table 1 Abbreviated burn severity index


Variable Patient Characteristic Score

Sex Male 0
Female 1
Age, y 0–20 1
21–40 2
41–60 3
61–80 4
81–100 5
Inhalation injury No 0
Yes 1
Full-thickness burn No 0
Yes 1
% TBSA <10 1
11–20 2
21–30 3
31–40 4
41–50 5
51–60 6
61–70 7
71–80 8
81–90 9
91–100 10

Total score 2–3 4–5 6–7 8–9 10–11 12–13


Threat to Life Very Low Moderate Moderately Severe Serious Severe Maximum
Probability of survival 99 98 80–90 50–70 20–40 10

Table 2 Levels of evidence used to quantify hospitalized burn patients were included. The titles,
abstracts, and the full texts of these articles were
epidemiological studies screened for relevance. Studies assessing nonpharma-
Level 1 National trauma registry cological interventions were excluded. Studies assessing
Level 2 Systematic review or multicenter study topical and wound care interventions that were aimed
Level 3 Single-center study only at improving the healing process were excluded.
Level 4 Self-reporting
Results and Discussion

Epidemiology of Burn Injuries


demographic or subsection of a population, if the study
A number of epidemiology studies have been under-
contained fewer than 100 patients, or if a higher level of
taken to try to capture the current state of burn injury
evidence was available.
prevalence in different regions. These studies include
Treatment of Acute Burn Pain national burn and trauma registries, multicenter and sin-
gle-center studies, systematic reviews, and question-
A search was performed in Medline on October 18, naires. In this systematic review, we aimed to capture
2016, to retrieve included studies. The applied search the current best evidence for burn injury prevalence
strategy was as follows: (((burn[Title] OR burns[Title]) OR around the world, provide a critical analysis of the qual-
postburn[Title]) AND (“pain”[MeSH Terms] OR “pain”[All ity of the evidence, and describe where and how data
Fields])) AND (“therapy”[Subheading] OR “therapy”[All capture needs to improve.
Fields] OR “treatment”[All Fields] OR
“therapeutics”[MeSH Terms] OR “therapeutics”[All In total, 163 publications of interest were obtained, and
Fields]) AND Clinical Trial[ptyp]. Only randomized con- of those 63 fit the exclusion and inclusion criteria. Three
trolled studies assessing the efficacy of a pharmacologi- burn injury registries were identified; one from the
cal intervention in comparison with placebo or other United States, another from Australia and New Zealand,
analgesic(s) with a primary outcome measure of pain in and a third from England and Wales. These registries

3
Morgan et al.

and their epidemiological reports (annual or cumulative) In total, the publications examined in this systematic re-
added an additional nine references, totaling 72 referen- view covered 58 countries with HDI scores ranging from
ces. A burn registry in Iran that is not accessible online 0.465 to 0.944, along with regional studies from South
was identified through publications along with studies Asia and Europe. It is problematic that few countries
(eight in total) using data from national trauma registries, with lower HDI scores have high-quality (level 1 and 2)
and these were considered level 1 evidence. The major- evidence as burn injury disproportionately affects those
ity of studies (41) obtained were of level 3 evidence in- in developing countries. Globally, more than 11 million
volving single-center studies. This level of evidence was people experience burn injury requiring medical attention
deemed less reliable as comparisons between level 3 annually, with the majority of severe burn cases affecting
evidence studies within the same country often pro- people in emerging economies, including an estimated
duced variability in measures (Table 3). 50% of cases in Southeast Asia [11]. The World Health
Organization is currently testing a Global Burn Registry,
Burn victims are usually men, with only seven of the 72 which allows for standardization of data collection and
studies reporting greater female representation than will provide greater access to epidemiological data from
developing countries. A systematic review found a de-
male (Table 3). The vast majority of burns occur in the
crease in burn injury prevalence and injury severity in
home (73% USA; 76% Australia and New Zealand;
very high and highly developed countries over time, but
65.89% England and Wales), and the most common
with data under-reported for low- and middle-income
source of injury is flame and scald exposure (Table 3),
countries, where the majority of burn injuries occur, no
but injuries were also from contact burns, radiation, conclusions could be made on trends there [12].
chemicals, and electricity (Table 4), with these being ac- Databases for burn injury statistics established in highly
cidental, inflicted, or self-inflicted. No age group is im- developed countries provide high-quality data to draw
mune from this traumatic experience, but there is a conclusions on clinical outcomes and burn injury trends,
significant occurrence of burn injuries, predominantly but these registries have limitations also. While the
from scalds, in children age five years and younger International Burn Injury Database (England and Wales)
(Table 4) and for adults between 20 and 29 years. In provides data describing whether pain scores have
Europe, burn injuries affect between two and 29 people been taken from patients, measures of pain outcomes
per 100,000, with males representing 60% of cases [5], are omitted from all repository annual reports.
while in the United States males represent approxi- Epidemiological data for pain scores included in burn in-
mately 70% of all burn cases in the age group of 20 to jury repositories could provide ongoing measures de-
60 years (Table 5) [6]. In comparison, 3.98% (four peo- scribing how adequately burn injury is managed and
ple per 100) of those in Sierra Leone self-reported burn where improvements can be made.
injuries, although the extent of these injuries was not
specified [7]. Burns can lead to morbidity through Animal Models of Burn Injury Pain
wounds and inflammation, scarring, infection, amputa-
tions or surgical resections, psychological conditions, Animal models have provided some important insights
and ongoing pruritis and acute and chronic pain. into human pain associated with burn injury. There is a
limited number of randomized placebo-controlled clinical
Mortality varied throughout the world, with studies trials available to evaluate the effectiveness of analgesics
reporting between less than 1% and up to 60.8%. The in human burn patients (discussed in detail below), in
mortality rate correlated with the Human Development part because burn patients are a heterogeneous group
Index (HDI) (Figure 2A), with developing countries more with variable levels of burn severity and background
likely to report higher mortality than developed coun- demographics and are frequently quite unwell, with
tries. Mortality also correlated with the total burn surface multi-organ dysfunction. While a number of human
volunteer studies of burn pain have been
area, with greater surface area resulting in a higher likeli-
reported—including models of dry heat contact burn, ul-
hood of death (Figure 2, B and C). In highly developed
traviolet (UV) burn, and capsaicin-induced heat
countries, burn injuries commonly affect less than 10%
sensitization—these are for obvious reasons not numer-
of the total body surface area (84% Australia and New
ous [82–96]. Although all of these models have demon-
Zealand; 96.5% England and Wales; 77.9% USA); how- strated the development of secondary hyperalgesia,
ever, in countries with lower HDI scores, a greater likely due to central sensitization, such models are un-
TBSA was often reported presenting to hospitals (Table likely to address the need for ongoing research into the
3). The mortality of clinically presenting cases is reported pathophysiological mechanisms underlying burn pain.
as between 0.8% and 1.2% in Australia and New Given the importance of this area to improving patient
Zealand, between 1.4% and 18% in Europe [5], and outcomes, a number of animal models have been
3.14% in the United States [8]. Mortality resulting from established specifically to study the mechanisms of burn
burn injury was higher in females than males in England pain (Table 6). However, it is unclear how predictive ani-
and Wales (1.86% vs 1.31%, respectively) [9] and mal models of burn-induced pain are of the human con-
Australia and New Zealand (2.32 times more likely to dition as many other animal models of pain in the past
succumb to injury compared with males, adjusted for have failed to translate to the clinic [97]. Nevertheless,
confounding factors) [10]. improvements in experimental design and efficacy end

4
Table 3 Epidemiological studies and results

Country Evidence Number of Study Age Predominant


Publication or Region Level HDI Score Patients Period Distribution Mortality TBSA  10% Male, % Source of Injury

[13] Afghanistan 3 0.465 532 2007–2008 Median age 19 y 28 Mean TBSA 36.5% 41.2 Flame
[14] Afghanistan 3 0.465 388 1996–2000 Median age 8 y 16 Median TBSA 15% 57 Scald
[15] Australia & 1 0.935 & 0.913 2,776 2014–2015 Greatest among 1 84% 67 Scald
New Zealand 1–2 y & 20–29 y
[16] Australia & 1 0.935 & 0.913 2,656 2013–2014 Greatest among 1 84% 68 Scald
New Zealand 1–2 y & 20–29 y
[17] Australia & 1 0.935 & 0.913 2,720 2012–2013 Greatest among 1.2 82% 68 Flame & scald
New Zealand 1–2 y & 20–29 y
[18] Australia & 1 0.935 & 0.913 2,772 2011–2012 Greatest among 1 82% 67 Flame & scald
New Zealand 1–2 y & 20–29 y
[19] Australia & 1 0.935 & 0.913 2,480 2010–2011 Greatest among 0.8 80% 67 Flame & scald
New Zealand 1–2 y & 20–29 y
[20] Australia & 1 0.935 & 0.913 2,103 2009–2010 Greatest among 20–29 y 1.6 74% 69 Flame & scald
New Zealand
[21] Brazil 3 0.755 921 1991–1997 Greatest among 20–39 y 8.4 NA 67.1 NA
[22] Bulgaria 3 0.782 2,627 2002–2011 Mean age 41 y 2.3–3.6 Median 10.6% 59 Scald
[23] Brunei 3 0.856 211 2007–2008 Mean age 19.6 y NA Mean TBSA 3.7% 52.6 Scald
[24] China 3 0.727 527 2002–2003 Median age 30 y 2 Median TBSA 10% 71 Flame
[25] China 2 0.727 1,974 2000–2008 Mean age 36 y 2.8 Mean TBSA 14.7% 70.7 Flame
[26] China 2 0.727 172,256 2001–2007 Median age 23 y 0.8 61.44% 70.1 Scald
[27] China 3 0.727 5,321 1980–1998 NA 0.9 68.47% NA NA
[28] Columbia 3 0.72 2,319 1994–2004 Mean age 18.1 y 7.4 Mean TBSA 26.9% 66.8 Scald
[29] Curacao 3 8.11 336 1992–2002 Mean age 24.3 y 3.3 Mean TBSA 13.6% 61.5 Scald
[30] England & 1 0.907 140,076 2003–2015 Greatest among 0–5 y NA 96.5% 58.5 Scald
Wales
[9] England & 1 0.907 81,181 2003–2011 Median age 21 y 1.5 Median TBSA 1.5 63 Contact & scald
Wales
[31] Ecuador 3 0.732 1,106 2005–2014 Mean age 33–34 y 10.2 Mean TBSA 20% 69.4 Heat
[5] Europe 2 NA 186,500 1985–2009 Greatest among 0–16 y 1.4–18 Mean TBSA 11–24% 55–75 Flame
[32] Finland 3 0.833 1,000 1994–2006 Mean age 39 y 4 78% 62.1 Scald
[33] Germany 3 0.916 695 2001–2010 Mean age 44 y 15 Mean TBSA 18% 70.9 NA
[34] Ghana 3 0.579 487 2009–2013 Greatest among 0–10 y 20.5 Mean TBSA 28.79 54 Scald
[35] India 3 0.609 101 2009 Mean age 32.03 y 40.9 Mean TBSA 45.8 45.4 Flame
[36] India 3 0.609 222 2006–2007 Mean age 24.58 y 60.8 Mean TBSA 48.75 37 Flame
[37] India 2 0.609 11,196 1993–2002 80% of cases between 51.8 Mean TBSA 50.35% 44.3 Flame
16–55 y
Burn Pain

5
(continued)
6
Table 3 Continued
Country Evidence Number of Study Age Predominant
Publication or Region Level HDI Score Patients Period Distribution Mortality TBSA  10% Male, % Source of Injury
Morgan et al.

[38] Iran 3 0.766 13,248 2010–2012 Mean age 27 y 1.6 Median TBSA 4% 50.5 Scald
[39] Iran 1 0.766 14,227 2010–2011 Mean age 28.85 y 8.9 Mean TBSA 23% 65.9 Flame
[40] Iran 3 0.766 713 2008–2010 Mean age 19.8 y 22.6 NA 54.5 Flame
[41] Iran 3 0.766 6,082 2003–2007 Greatest among 0–10 y 8 Mean 38.7 55.7 Flame
[42] Iraq 3 0.654 884 2009 Mean age 25.6 y 29.4 NA 38.2 Flame
[43] Israel 1 0.894 5,269 2004–2010 Greatest among 0–2 y 3.7 71% 67.3 Scald
[44] Israel 2 0.894 5,000 1997–2003 Greatest among 0–1 y 4.4 56% 68.9 Scald
[45] Italy 2 0.873 2,067 2008 Majority between 0–5 y 5.3 NA 59.3 Flame
[46] Japan 1 0.891 6,401 1983–2003 Mean age 40.4 y 15.4 Mean TBSA 18.8% 63 Flame
[47] Kenya 3 0.548 109 1984–2004 Mean age 14.4 y 12.0 Mean TBSA 22.3% 56 Scald
[48] Kenya 3 0.548 269 2006–2010 Majority less than 5 y 12 NA 59 Scald
[49] Kuwait 3 0.816 1,702 2006–2010 Greatest among 17–45 y 5.8 NA 71 Flame
[50] Kuwait 2&3 0.816 3,680 1982–1997 Greatest among 16–39 y 6.4 Mean TBSA 71% 47.9 Flame
[51] Lebanon 3 0.769 1,524 1992–2012 Mean age 34 (5–98) y 18 NA 61 Scald
[52] Lithuania 1&2 0.839 132,428 1991–2004 Greatest among 0–14 y 3.1 NA 67 NA
[53] Malawi 3 0.445 1,825 1994–1999 Majority less than 30 y 12 71% 52 Scald
[54] Malaysia 3 0.779 110 1991–2001 NA 6.3 Mean TBSA 19% 67 Scald
[55] Morocco 3 0.628 291 2004–2009 Majority between 20–59 y 5 Mean TBSA 21% 62 Flame
[56] Netherlands 3 0.922 9,031 2006–2011 Greatest among 0–4 y 4.1 Median TBSA 4–8% 65 Scald & flame
[57] Nigeria 3 0.514 407 2006–2010 Greatest among 20–29 y 30 16.32% 67.3 NA
[58] Norway 1 0.944 726 2007 Mean age 26.9 y 2.1 NA 65 NA
[59] Oman 1&2 0.793 3,531 1987–2011 Greatest among 0–10 y 8.2 49.4% 60.1 Flame & scald
[60] Pakistan 3 0.538 403 2010–2011 Median age 25 y 5.2 NA 67.2 Scald
[61] Pakistan 3 0.538 1,498 2007–2008 Majority 0–46 y NA NA 60 NA
[62] Pakistan 3 0.538 1,979 2010–2011 NA 36.1 17.50% 53.1 NA
[63] Portugal 1&2 0.83 14,797 1993–1999 Mean age 30 y 3.7 NA 59 NA
[64] Saudi Arabia 3 0.837 240 1997–2003 Majority 0–18 y 7 36.7% 59.2 Scald
[7] Sierra Leone 4 0.413 145 NA Greatest among 0–14 y NA NA 60 Scald
[65] Singapore 2 0.912 2,019 1997–2003 Mean age 32.5 y 4.6 70.5% 68.8 Scald
[66] Singapore 3 0.912 655 2011–2013 Mean age 42.6 y 2.7 Mean TBSA 9% 70.6 Scald
[67] Slovakia 2 0.844 20,865 1993–2004 NA 3.0 NA 59 Scald
[68] South Asia 2 NA 28 studies 1970–2001 NA 0–54 NA 37–88 Flame & scald
[69] South Korea 3 0.898 19,157 1986–2003 Greatest among 0–5 y 8.2 46.9% 66.2 Flame
[70] South Africa 2 0.666 NA NA Mean 4.3 y 27 Mean TBSA 48.5% 54.7 Scald
[71] South Africa 3 0.666 1,024 2001–2004 NA 7.9 NA NA NA
[72] Sri Lanka 3 0.757 345 1999–2001 Median age 22 y 27 Mean TBSA 16% 64.3 Flame
(continued)
Burn Pain

points are likely to increase the clinical validity of animal

Male, % Source of Injury


Predominant models of pain in the future.

Animal models of burn-induced pain are typically based

Flame
Scald

Scald
Scald
Scald
Scald
Scald
NA on a burn injury that is induced on the plantar skin of

NA
one hind paw in rodents, in contrast to animal models
43.1 used to study other aspects of burn injury, such as
46.1
65.2
67.8

67.6
wound healing, metabolic changes, and infection,
69
67

61
69
where a larger burn injury is commonly induced on the
dorsum or ventral skin of mice, rats, or pigs [119,120].
Mean TBSA 12–19%

The advantage of using the plantar skin of the hind


Mean TBSA 17.5%

Mean TBSA 4.82% paw is that it readily allows the quantification of pain
behaviors, including changes in mechanical and ther-
Mortality TBSA  10%

mal thresholds or weight-bearing behaviors in the ipsi-


lateral hind paw.
77.9%
76%

82%
NA

NA

NA

Dry Contact Heat


10.5

3.14
3.7
1.5
NA

0.4
1.5
3

Dry contact heat is the most commonly used method


for inducing burn injury in rodents and involves exposing
the plantar skin of one hind paw to a heated—often
Greatest among 0–4 y

Greatest among 0–4 y

Greatest among 0–5 y

metal—surface while the rodents are under anesthesia.


This is a clinically relevant model as contact with a dry
Median age 29.2 y

1–2 y & 20–29 y


Mean age 25.3 y
Mean age 17.0 y
Mean age 25.3 y

Mean age 22.5 y

2006–2015 Greatest among

heat source such as an iron, vehicle exhaust, or heater


accounts for 9–14% of burns cases [8,16]. To induce a
Distribution

mild burn injury or superficial burn in rats, the metal sur-


face is set to a temperature of 52–52.5  C and the skin
is exposed for 45 seconds, with constant pressure
Age

maintained by applying a sand pouch with a set weight


to the dorsal surface of the hind paw. This model leads
1987–2004
1997–1999

1996–2002
2012–2014
2010–2011
2003–2007

to the rapid development of mechanical and thermal


2010
2008
Period
Number of Study

allodynia in the injured hind paw, evident within


30 minutes of burn injury and persisting for at least
seven days [104,98,106]. This protocol has also been
205,033

modified for use in mice by reducing the exposure time


24,538
4,741
7,630

1,752
Patients

230
742
264

203

to 25 seconds [121]. In the mouse model, mechanical


and thermal allodynia develop rapidly and persist for at
least four days, along with changes in weight-bearing
behavior. The main advantage of mouse models of burn
HDI Score

injury pain is in the ability to study the specific effects of


0.907
0.822
0.822
0.761
0.761
0.761
0.747
0.835

0.915

NA ¼ not available; TBSA ¼ total body surface area.

gene deletion on the development of burn pain in


knockout mice.

To induce a partial-thickness injury (with blistering) in


Evidence

rats, the contact surface is set to a temperature of


1&2

1&2
Level

3
3
3
1
3

85  C and the skin is exposed for 15 seconds. This


model leads to the rapid development of mechanical
allodynia in the injured paw that persists for eight
United State of

weeks; however, unlike the mild burn injury model, lit-


United Arab
Emirates
Continued

America

tle to no thermal allodynia is observed [110]. The


Publication or Region

Sweden
Country

Ukraine

above protocol used in rats can be slightly modified


Taiwan
Taiwan
Turkey
Turkey
Turkey

to produce a partial-thickness injury model in mice,


using a temperature of 65  C and exposing the plantar
surface of one hind paw for 15 seconds [113]. This
Table 3

model leads to the rapid development of mechanical


and thermal allodynia that will persist for seven to
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]

[81]

14 days.

7
Morgan et al.

Table 4 Patient distribution by age and type of burn injury

Australia USA Europe


Infant, % Child, % Adult, % Infant, % Child, % Adult, % Pediatric, % Elderly, % All, %

Scald 71.6 54.8 27.7 51.5 48.9 23.3 65.9 33.9 36.6
Contact 16.4 19.9 14.0 19.8 14.0 5.3 9.3 10.8 9.6
Flame 4.3 12.1 43.9 7.6 23.0 47.1 13.7 49.1 42.5
Friction 7.8 9.6 3.7
Electrical 3.6 2.4 <1 1.8 4.1 4.1 1.8 7.6
Radiation 2.1 <1 <1 <1
Chemical 6.1 <1 1.2 3.8 2.4 2.2 3.9
Other <1 3 4 7.6
Unknown 16.2 6.9 8.5

Averaged percentages from three studies [5,8,82], with results shown for infants (0–12 mo), children (1–15 y), adults (16 y),
pediatric (0–16 y), and elderly (60 y).

Table 5 Burn patient distribution by age and sex plantar surface of the hind paw to induce a burn injury
in rats [117,118]. This model results in a full-thickness
Australia, % USA, % burn injury that takes four weeks to resolve. Mechanical
and thermal allodynia is not present at the actual site of
Male Female Male Female the burn injury, consistent with local destruction of cuta-
neous nerve endings, but can be detected in the adja-
Infants 2.6 2.1 3.2 2.2
cent skin for two weeks after burn injury [118]. Although
Children 21.6 11.2 14.5 9.0
no changes in thermal hyperalgesia or mechanical allo-
Adults 45.2 17.4 50.7 20.5 dynia were detected in the contralateral paw, expression
of both calcitonin gene–related peptide (CGRP) and
Average percentages from two studies (N ¼ 197,063 patients,
Substance P was upregulated in the contralateral dorsal
including infants [0–12 mo], children [13 mo–15 y], and adults
horn, suggesting changes consistent with central sensi-
[>16 y]) [8,14].
tization. This method is associated with significant tissue
damage that necessitates wound management and an-
tibiotic treatment and may thus lead to ethical concerns
Scalding that would be justified predominantly for studies aiming
to determine effects on wound healing and systemic im-
Scalding is a method where burn injuries are induced pact of burn injuries. However, this method may be suit-
by immersing the dorsum or ventral skin of mice or rats able to model burn injury induced by direct contact with
in water (60–100  C) to induce burns of varying degree, a flame, the most common cause of burn injury in the
dependent on the time and temperature of exposure, clinic [8,16].
and is commonly used to study other aspects of burn
injury as opposed to the pain-related behavior [119].
However, modified scald methods that produce burn in- Ultraviolet Radiation
jury on the hind paws of rats have been described
[114,116]. This model is relevant to the clinic as scald- The UV method involves exposure of the plantar surface
ing is responsible for a third of burns requiring hospital- of the hind paw of rats to ultraviolet B radiation to in-
ization [8,16]. Immersing only the dorsal surface of the duce a burn injury. A dose of 1,000 mJ/cm2 causes a
hind paw in water at 85  C for 12 seconds induces a superficial burn injury without blistering, along with me-
third-degree burn injury, resulting in mechanical and chanical and thermal allodynia that develops rapidly and
thermal allodynia on the plantar surface of the hind paw persists for seven days [122,123]. While UV radiation
of rats that lasts for at least seven days, albeit this can be dosed more precisely than scalding or dry heat
method is associated with significant tissue damage contact, it is unclear whether the mechanisms underly-
and thus raises ethical concerns [116]. ing a burn injury induced by UV radiation are similar to
other causes of thermal injury. It is clear that UV radia-
tion is associated with DNA damage, and in humans
Soldering the long-term effects of UV burns include development
of skin cancers. Surprisingly, thermal burns are associ-
This recently described method utilizes a soldering iron ated with a significant increase in cancer burden, possi-
to apply a 100  C heat stimulus for 30 seconds to the bly due to systemic inflammatory processes [124,125].

8
Burn Pain

partial-thickness burn) and of short duration (days to


weeks). It is unclear if the severity of the burn has an
impact on the underlying mechanisms of pain, and thus
whether these milder pain models serve as appropriate
models for more severe burns typically necessitating
clinical intervention. In addition, the duration of these
animal studies is typically relatively short, thereby mod-
eling acute pain as opposed to chronic pain, which, as
described below, is clinically more difficult to manage
and can continue for months to years despite healing.

The animal models described thus far tend to focus on


stimulus-evoked rather than background pain as they
apply mechanical or thermal stimuli to the injured area
and quantify withdrawal responses. It could be argued
that stimulus-evoked pain is a surrogate measure of
procedural pain, which, as described below, can often
be severe and difficult to manage. Quantifying back-
ground pain in animals can be challenging, although
behavioral methods that quantify non-stimulus-evoked
pain are increasingly being developed [126–128]. Finally,
the majority of animal models of burn pain induce a
burn injury on glabrous or nonhairy skin, which in
humans is located on palms of the hands and soles of
the feet. The sensory innervations between hairy and
nonhairy skin differ [129,130], and it is unclear what im-
pact this has on the translation to pain from burns that
occur on hairy skin.

Burn Injury Pain Mechanisms

Both human and animal studies have provided signifi-


cant insight into the mechanisms underlying pain after a
burn injury, which has a complex pathology with both
peripheral (Figure 3) and central processes [131,132]
and combines features of acute nociceptive, inflamma-
tory, and neuropathic pain. However, the significant di-
Figure 2 Mortality was found to be correlated with the versity in the causes and severity of burn injuries make
(A) human development index score (HDI; R2 ¼ 0.335) it difficult to determine the relative contribution of differ-
and (B and C) total burn surface area (R2 ¼ 0.616 and ent pathways in individual cases. It should thus be
0.752, respectively). TBSA ¼ total body surface area. noted that, in the context of this review, it is not cur-
rently possible to attribute the relative importance of
those mechanisms to the individual pain experience or
analgesia.
It thus remains to be determined whether exposure to
UV radiation is a suitable model for assessing mecha-
nisms underlying burn-induced pain more generally.
Peripheral Nociceptive Mechanisms and
Adaptations in Burn Injuries

Clinical Applicability of Animal Models Acute Burn Injury Pain. The threshold temperature for
formation of edema in the skin in response to heat is
Researchers have an ethical obligation to refine 43  C, which is also the threshold temperature for acti-
approaches in studies involving animals to minimize the vation of the transient receptor potential vanilloid type 1
pain, suffering, and distress as much as possible. ion channel (TRPV1) [133,134]. Accordingly, TRPV1 ac-
Therefore, in the case of animal models of burn pain, tivation has been functionally linked to the development
careful consideration must be given to determine the of thermal allodynia in partial-thickness burns [112].
severity of burn injury and study duration necessary to Oxidized linoleic acid generated by thermal injuries also
achieve experimental outcomes. As such, most acts as an agonist at TRPV1 and activates the receptors
animal models of burn-induced pain reported in the liter- post–burn injury [112]. Stimuli above 52  C result in acti-
ature are of mild severity (equivalent to a superficial or vation of TRPV2 [135], while TRPV3 and TRPV4 are

9
Table 6 Animal models of burn-induced pain

10
Duration
Duration of Burn Mechanical Thermal of Pain
Model Species Location Temperature Exposure, s Severity Allodynia Allodynia Behaviors Reference
Morgan et al.

Dry contact heat Rat Hind paw (plantar) 52  C 45 Superficial Y Y 90 min [98–100]
Dry contact heat Rat Hind paw (plantar) 52.5  C 45 Superficial N/A Y 90 min [101]
Dry contact heat Rat Hind paw (plantar) 52  C 45 Superficial N/A Y 120 min [102]
Dry contact heat Rat Hind paw (plantar) 52  C 45 Superficial Y N/A 60 min [103]
Dry contact heat Rat Hind paw (plantar) 52 C 45 Partial thickness Y N/A 7d [104]
Hind paw (dorsal) 52  C 30–40 Partial thickness Y N/A 15 d
Dry contact heat Rat Hind paw (plantar) 52.5  C 45 Superficial Y N/A 120 min [105]
Dry contact heat Rat Hind paw (plantar) 52.5  C 45 Superficial Y N/A 90 min [106]
Dry contact heat Mouse Hind paw (plantar) 52.5  C 25 Superficial Y Y 4d [107]
Dry contact heat Rat Hind paw (plantar) 55  C 30 Not specified Y Y 120 min [108]
Dry contact heat Rat Hind paw (plantar) 85  C 15 Partial thickness Y N 4–8 wk [109–111]
Dry contact heat Rat Hind paw (plantar) 100  C 30 Partial thickness N/A Y 7d [112]
Dry contact heat Mouse Hind paw (plantar) 65  C 15 Partial thickness Y Y 14 d [113]
Scalding Rat Hind paw 60  C 20 Not specified Y N/A 180 min [114]
Scalding Rat Hind paw (dorsal) 85  C 12 Full thickness Y Y 7d [115,116]
Soldering Rat Hind paw (plantar) 100  C 30 Full thickness Y Y 2 wk [117,118]

NA ¼ not assessed.
Burn Pain

Figure 3 Burn pain mechanisms. A thermal insult to the skin initiates a cascade of mechanisms that are believed to
be involved in the signaling of pain. The group of transient receptor potential vanilloids (TRPV) 1–4 and transient re-
ceptor melastatin (TRPM) 3 are activated by different temperatures. Activation of the thermosensitive channels causes
the release of calcitonin gene–related peptide (CGRP) from the primary sensory nerves, which in turn results in the el-
evation of intracellular Ca2þ levels. The deteriorating cell membrane releases ATP, which targets P2X receptors on
primary sensory nerves. The voltage-gated sodium channel (NaV) 1.7 is essential for thermal allodynia. The trauma
caused to the cell membranes will result in the release of damage-associated molecular pattern molecules (DAMPs)
that will bind to Toll-like receptors, P2X receptors and AIM2-like receptors (ALRs). Following this trigger histamine,
catecholamines, TNF-a, IL-1b, IL-6, oxygen free radicals, nitric oxide (NO), bradykinin, and arachidonic acid cascade
products are released. Furthermore, leukocytes, eicosanoids (including prostaglandins [PGs]), and platelet-activating
factors (PAFs) are recruited. PGE2 activates prostanoid EP1–4 on mast cells, and subsequently histamine is released.
Prostanoid EP3–4, located on the primary afferent terminals and mast cells, furthermore results in release of IL-6.
Lastly, a catecholamine 5-hydroxytryptamine (5-HT) is also released at the site of injury and sensitizes the peripheral
nerves by acting at 5-HT2A.

11
Morgan et al.

thermosensitive channels that are gated by innocuous from wild-type mice but not from NaV1.7 knockout
temperatures above 33  C and 25  C, respectively. mice, suggesting that NaV1.7 is essential for thermal hy-
However, while thermal injury-induced keratinocyte persensitivity after burn injury [113]. These observations
death has been at least partially attributed to TRPV1- are consistent with the clinical phenotype of humans
TRPV4 isoforms, the contribution of these thermosensi- with a congenital mutation resulting in loss of function of
tive channels, specifically TRPV2-TRPV4, to burn- NaV1.7, who frequently present with painless burn inju-
induced pain has to-date not been systematically ries [145]. Given the intense drug discovery efforts di-
assessed [136]. Similarly, the transient receptor melasta- rected at identifying subtype-selective NaV1.7 inhibitors,
tin 3 ion channel (TRPM3), which is activated at rela- it appears likely that novel analgesics with efficacy in
tively low temperatures, has also been shown to play a acute burn pain will be developed in the future.
role in heat nociception, with TRPM3-deficient mice dis-
playing attenuated avoidance of noxious heat stimuli Inflammatory Burn Injury Pain. A burn results in an acute
[137]. Activation of these thermosensitive channels has inflammatory response that can be restricted to the site
been shown to evoke the release of the neuropeptide of injury or involve systemic inflammatory processes that
CGRP from the terminals of primary sensory nerves, trigger a syndrome associated with significant morbidity
and, accordingly, elevated circulating CGRP levels have and mortality. Trauma from a severe burn injury results
been found shortly after burn injury [138]. In addition, in denaturing of proteins and the leakage of cell plasma
the neuropeptide Substance P has been found to be due to the loss of cell membrane integrity. A necrotic
significantly higher in the blood of patients within trauma like this results in the release of damage-
12 hours of suffering a burn injury [138]. CGRP acts di- associated molecular pattern molecules (DAMPs) [146]
rectly on a subset of dorsal root ganglion (DRG) neu- from the cell plasma, which subsequently bind to vari-
rons, causing elevated intracellular Ca2þ and enhancing ous inflammatory receptors including Toll-like receptors
tetrodotoxin (TTX)-resistant sodium current density in [147–149], P2X receptors, and AIM2-like receptors
cultured sensory neurons [139]. Substance P also has (ALRs) [150], among others. Activation of DAMP recep-
direct effects on nociceptors, with lumbar DRGs super- tors and localized ischemia/reperfusion events result in
fused with Substance P displaying a prolonged depolar- activation of the alternative complement pathway [151],
ization [140]. Levels of both Substance P and CGRP in the release of histamine [152], catecholamines, TNF-a,
different layers of the skin have been correlated to the IL-1b, IL-6 [153], oxygen free radicals, nitric oxide, bra-
sensation of pain during wound healing and scarring in dykinin, and arachidonic acid cascade products, as well
burn patients [141], suggesting that they play a role in as the activation and recruitment of leukocytes.
the maintenance of pain after the initial heat insult. Histamine release results in early local vascular leakage
and peripheral edema, while reperfusion events result in
The loss of cell membrane integrity due to elevated heat localized cell apoptosis [154]. Accordingly, activation of
results in the release of cell contents, some of which peripheral sensory nerve terminals after burn injuries as
have also been shown to directly activate nociceptive well as sustained nociceptive input post-injury occurs
neurons. Specifically, ATP is released from the deterio- through a range of stimuli, including the initial thermal
rating cell membrane, with the ATP metabolite adeno- insult, vascular damage, hypoxia from ischemic events,
sine found in the fluid of burn blisters [142]. The DAMP receptor activation, and local release of pro-
pharmacological targets of ATP are the P2X receptors inflammatory and algogenic mediators and transmitters
on primary sensory fibers, where both P2X2 and P2X3 such as bradykinin, histamine, eicosanoids, platelet-
are thought to play a role in burn injury pain. The P2X3 activating factor (PAF), nerve growth factor (NGF),
receptor in particular is upregulated in peripheral nerve Substance P, and CGRP.
terminals of rats after burn injury, and P2X3 antagonists
also inhibited P2X-activated currents in isolated DRGs Bradykinin, an important mediator of peripheral vascular
from the same animals [143]. In addition, the release of permeability and pain, has been implicated in burn injury
protons from the acidified inflamed environment likely pain since a bradykinin-like substance was found in the
further enhances the effect of ATP on P2X2 receptors, urine of patients with burn injuries [155]. In addition, in-
resulting in enhanced perception of pain [144]. tradermal administration of the selective bradykinin 2 re-
ceptor antagonist, HOE-140, reduced the occurrence of
The voltage-gated sodium channels (NaV) play select edema in a model of burn injury [156] and reduced me-
roles in burn injury pain. The NaV blocker TTX was ef- chanical allodynia in a model of sterile inflammatory pain
fective at reducing thermal hyperalgesia and mechanical [157,158].
allodynia after full-thickness burn injury in rats [117],
suggesting that TTX-resistant isoforms, specifically the Similarly, histamine was detected in the urine of burn
sensory neuron-specific NaV1.8 and NaV1.9, play a lim- patients [157] and in blister fluid, with levels correlating
ited role in burn pain. Consistent with this observation, to edema formation [152]. Burn injury pruritus or itch is
the TTX-sensitive isoform NaV1.7 was essential for ther- also mediated by histamine, with 70% of patients
mal allodynia in a mouse model of a partial-thickness reporting effective relief with antihistamines [159].
burn injury, albeit mechanical allodynia was unaffected
in NaV1.7 knockout mice. In addition, burn injury A number of eicosanoids have been identified both at
resulted in reduced threshold of TTX-sensitive DRGs the site of burn injury and systemically, including

12
Burn Pain

prostaglandins (PG) E2, PGF1a, PGF2a, thromboxane anti-NGF serum resulted in the inhibition of hyperalgesia
B2 [160], and leukotrienes [161], and these are believed in burn injury [176]. NGF-induced systemic hyperalgesia
to contribute to burn pathology. PGE2 is a particularly is mediated by a reduced expression of lumbar l–opioid
important inflammatory mediator in burn pain and con- receptor expression and increased activation of gluta-
tributes to hyperalgesia in a number of ways. These in- mate receptors. Accordingly, administration of MK-801,
clude activation of the prostanoid receptors EP1–4, an NMDA receptor antagonist, inhibited systemic hyper-
which causes the release of histamine from mast cells algesia after thoracic burn injury, consistent with upre-
[162], activation of prostanoid EP3 and EP4 receptors gulation of NMDA receptor transcripts in DRG neurons
on primary afferent terminals and mast cells, causing from mice following burn inury [107,177]. It appears that
the release of IL-6 [162,163], sensitization of the activa- the high-affinity receptor for NGF, tropomyosin receptor
tion threshold of TTX-resistant NaV channels [164], inhi- kinase (TrkA), likely mediates these hyperalgesic effects
bition of voltage-gated potassium currents [164], and as intrathecal administration of antisense oligodeoxynu-
sensitization of TRPV1 through prostanoid EP1 recep- cleotides to TrkA resulted in dose-related decrease of
tors [165]. Accordingly, nonsteroidal anti-inflammatory burn-induced primary mechanical hyperalgesia. Thus,
drugs (NSAIDs) are a firstline treatment for burn pain anti-NGF treatment may provide substantial benefits to
and are discussed further in the Treatments section. burn patients, albeit this remains to be assessed clini-
cally [104].
Cytokines and inflammatory mediators released both
locally and from recruited leukocytes mediate inflamma- The catecholamine 5-hydroxytryptamine (5-HT) is also
tion and wound healing, but also contribute to burn-in- released at the site of burn injury and found systemically
duced allodynia and hyperalgesia. The cytokine and [178]. Administration of exogenous 5-HT results in noci-
immune modulators IL-1b, IL-6, IL-8, TNF-a, and TGF- ceptive behavior including mechanical and thermal
b, as well as platelet-derived growth factor, have all hyperalgesia and allodynia [179–181], which is mediated
been found in the blister fluid of burn patients [153]. by the 5-HT1A, 5-HT2A, and 5-HT3 receptors [180–182].
While IL-1b has been shown to be an important media- In an animal model of mild plantar thermal injury, a se-
tor of burn-induced inflammation, knockout of caspase- lective 5-HT2A receptor antagonist resulted in a reduc-
1, an enzyme that cleaves IL-1b into its active form, had tion in mechanical and thermal allodynia when
no effect on burn-induced mechanical or thermal allody- administered at the site of injury [183], demonstrating
nia in mice [166]. IL-6, on the other hand, has been that locally released 5-HT sensitizes peripheral nerves
shown to be an important mediator of burn injury pain by acting at 5-HT2A [183].
[146]. In rats subjected to a partial-thickness burn, IL-6
concentrations were elevated at the site of the burn cor- Activation of peripheral adrenoceptors is known to result
relating with the duration of pain, and treatment with in- in the production of cytokines and in nerve regeneration.
tradermal anti-IL-6 antibodies significantly reduced pain In the subsequent weeks after a full-thickness burn, the
[167]. In addition, subcutaneous administration of IL-1b, expression of the a1-adrenoreceptor is increased in pe-
IL-8, and TNF-a result in mechanical and thermal hyper- ripheral nerve terminals and in keratinocytes at the site
algesia, suggesting that it is likely that these cytokines of injury [184], albeit changes in expression of a1-adre-
contribute to burn injury hyperalgesia [168–170]. noreceptor subtypes were not systematically assessed.
While administration of the selective a1-adrenoceptor
PAF has been isolated from polymorphonuclear leuko- agonist phenylephrine resulted in increased hyperalgesia
cytes of burn injury patients [171], and administration of in a human study of mild burn that could be inhibited by
exogenous PAF results in hyperalgesia [172]. PAF a selective a1-adrenoceptor antagonist, analgesia after
receptor-deficient mice display normal thermal and me- administration of selective a1-adrenoceptor antagonists
chanical allodynia but show decreased persistent pain alone has not yet been shown [185].
in response to tissue injury, suggesting that PAF plays a
role in sensitizing peripheral sensory nerves [173]. Neuropathic Burn Injury Pain. By definition, neuropathic
Similarly, in animal models of peripheral nerve injury, in- pain occurs as a result of lesions or disease of the so-
trathecal administration of PAF receptor antagonists de- matosensory nervous system [186]. While the contribu-
creased mechanical and thermal allodynia mediated by tion of neuropathic components to burn pain is not
the glycine receptor 3a [174]. Thus, while the contribu- routinely considered, perhaps due to the parallel experi-
tion of PAF to burn-induced pain has not been system- ence of other forms of pain, it is not surprising that
atically assessed, modulation of this inflammatory patients can develop both acute and chronic neuro-
mediator may prove a viable option for treatment of in- pathic pain given the extensive damage to the somato-
flammatory pain associated with burn injury. sensory system in burn injury. Burn-induced damage to
cutaneous nociceptors and their superficial conducting
Given the substantial body of literature describing the fibers likely gives rise to ectopic firing and other patho-
role of NGF in pain, it is not surprising that this impor- logical processes that cause features of acute and
tant growth factor has also been ascribed a specific chronic neuropathic pain, which are often described as
contribution to burn-induced pain. NGF is released into “burning pain,” “on fire,” “pins and needles,” and
regenerating skin [175] and contributes to the develop- “stabbing” sensations [187]. These descriptors, com-
ment of systemic hyperalgesia as administration of bined with the underlying mechanism of injury, may

13
Morgan et al.

indicate that the patient is suffering from a form of acute inhibitory pathways to modulation of burn pain could be
neuropathic pain [188]. Indeed, this pain could very ap- expected, although this has not been systematically
propriately be referred to as “phantom skin pain”—a investigated.
phrase first coined by Atchison and colleagues in
1991 [1]. Treatment of Acute Burn Pain

While the mechanisms underlying burn-induced neuro- Acute pain occurs immediately following burn injury and
pathic pain have not been studied in detail, treatments can continue for several months until wound healing is
including gabapentin and pregabalin are reportedly ef- complete as patients undergo many return trips to the
fective in some patients with neuropathic pain symp- operating theater, repeated dressing changes and
toms [189,190], suggesting that the pathophysiology of baths, and regular sessions of rehabilitation that include
burn-associated neuropathic pain resembles that of stretching and physiotherapy. Acute burn pain has a
other types of neuropathic pain, at least to some complex pathology with central and peripheral pro-
degree. cesses, consisting of three dominant pain types: back-
ground pain, procedural pain, and neuropathic pain.
Effective treatment of each of these pain types, which
Central Nociceptive Mechanism and Neuronal will be discussed further below, requires the prescription
Adaptations to Burn Pain of regular multimodal analgesia with the addition of
carefully titrated breakthrough analgesia accompanied
Central sensitization appears to be a key contributor to by frequent review and assessment by experienced
burn-induced pain. Chronic, bilateral hyperexcitability of health professionals. Treatment is generally based on lo-
wide dynamic range neurons and microglial activation in cal burn center guidelines as limited randomized
the spinal cord accompany burn injuries [109]. Reversal placebo-controlled clinical trials are available to evaluate
of these changes by treatment with minocycline, a tetra- the effectiveness of analgesics specifically in the burns
cycline antibiotic that inhibits microglial activation and population.
prevents neuropathic pain in other models with central
components, was paralleled by a decrease in burn- A number of techniques have been described to assess
induced allodynia, lending further support to the impor- the level of pain experienced due to a burn injury, which
tance of central mechanisms. should take into account the intensity of the pain as well
as behavioral and physiological responses. In general,
Early spinal sensitization is thought to develop through adult patients are asked to describe their pain using a
the activation of the NMDA glutamate receptor and sub- numeric pain scale (0–10), visual analog scale (trans-
sequent calcium influx as intrathecal administration of ected line), or verbal scale (no pain to worst pain)
MK-801 has been shown to block both behavioral and [196,197]. Assessing the severity of pain in small chil-
electrophysiological measures of spinal sensitization dren and infants is more complex, with face scales
[191,192]. In addition, the development of secondary (e.g., Wong-Baker Faces Pain Rating Scale) and behavi-
hyperalgesia seen in burn injury is prevented by intrathe- oral observations by medical staff being a more com-
cal administration of AMPA receptor antagonists [103], mon measure [198].
suggesting that enhanced glutamatergic signaling plays
a key role in central sensitization after burn injury [193]. Our search strategy yielded a total of 213 articles de-
scribing treatment of burn pain, of which only 14 articles
In addition, due to systemic activation of DAMP recep- fitted our inclusion/exclusion criteria (Table 7), highlight-
tors and the direct effects of C5a, TNF-a, IL-1b, matrix ing the limited availability of randomized placebo-
metalloproteinases, and other pro-inflammatory media- controlled clinical trials to assess analgesic efficacy in
tors, the blood-brain barrier becomes compromised af- the burns population. Most studies (11/14) focused on
ter burn injury. DAMP receptor activation within the the treatment of procedural pain; two studies focused
brain results in cerebral inflammation characterized by on background pain, and one study focused on neuro-
increased levels of pro-inflammatory cytokines (IL-1b, pathic pain. Although topical interventions are likely to
IL-6, and TNF-a) in the brain and cerebral edema. have an impact on burn pain, studies assessing phar-
Central inflammation can be seen as a result of a scald- macological treatments alone are difficult to carry out as
ing burn. This can lead to states of delirium and dizzi- wound care interventions, such as dressing changes
ness. The upregulation of IL-1b within the brain and debridement, form the standard of care for partial-
activates cyclooxygenase and the subsequent produc- and full-thickness burns.
tion of prostaglandins, and this has been shown to con-
tribute to centrally mediated hyperalgesia [194]. TNF-a
in the brain contributes to the development of neuro- Background Pain
pathic pain [195].
Background pain is a continuous nociceptive inflamma-
Moreover, based on pathophysiological similarities to tory pain that is present while the patient is at rest. Pain
other types of pain, a contribution of descending management strategies that apply to other forms of pain

14
Table 7 Randomized double-blind clinical trials for the treatment of burn pain

Mean Double- Placebo- Primary Significant Mean Score


Study Pain type Population Size, N TBSA, % Randomized blinded controlled Outcome Comparison Difference Reduction

[119] Background Adult 43 6 Y Y Y NRS (0–10) Propranolol (p.o) vs placebo N N/A


[200] Background Adult 53 16 Y Y Y NRS (0–10) Gabapentin (p.o) vs placebo N N/A
[201] Procedural Adult 60 39 Y Y N VAS (0–10) Ketamine (p.o) vs dexmedeto- Y –1.2
midine (p.o)
[203] Procedural Adult 240 N.R Y Y N VAS (0–10) Nitrous oxide/oxygen vs Y –7.7
oxygen
[203] Procedural Adult 45 13 Y Y Y VRS (0–10) Lidocaine (i.v) vs placebo Y –0.4
[204] Procedural Adult 24 38 Y N N VAS (0–10) Ketamine (i.m) vs ketamine/ Y –2.3
dexmedetomidine/tramadol Y –2.1
(i.m)
Ketamine (i.m) vs ketamine/
midazolam/tramadol (i.m)
[205] Procedural Adult 26 7 Y Y Y NRS (0–10) Fentanyl (intranasal) vs N N/A
morphine (p.o)
[206] Procedural Pediatric 24 N.R Y Y Y NRS (0–10) Fentanyl (intranasal) vs N N/A
morphine (p.o)
[207] Procedural Pediatric 8 45 Y Y Y Face Pain Fentanyl (transmucosal) vs N N/A
Rating morphine (p.o)
Scale (0–5)
[208] Procedural Pediatric 14 20 Y Y Y VAS (0–10) Fentanyl (transmucosal) vs N N/A
hydromorphone (p.o)
[209] Procedural Pediatric 19 18 Y ? N Visual Ketamine (p.o) vs paracetamol/ Y –5.4
algometer codeine/diphenhydramine
(0–10) (p.o)
[210] Procedural Pediatric 60 5 Y Y N CHEOPS (4–13) Ketamine (p.o) vs acetamino- Y –1.5
phen/codeine (p.o)
[211] Procedural Adult 79 14 Y Y Y VAS (0–10) Lorazepam (p.o) vs placebo Y –1.6
[189] Neuropathic Adult 90 N.R Y Y Y NPS 2 (0–10) Pregabalin (p.o) vs placebo Y –0.7*
NPS 3 (0–10) –0.9*

CHEOPS ¼ Children’s Hospital of Eastern Ontario Pain Scale; NPS ¼ Neuropathic Pain Scale; NRS ¼ numerical rating scale; VAS ¼ visual analog scale; VRS ¼ verbal rating
scale.
*Week 1 values.

15
Burn Pain
Morgan et al.

also apply to burn injury pain—that is, the utilization of combination with other analgesics, most likely due to
well-directed, carefully titrated, and regularly reviewed their sedative and anxiolytic effects [215]. This has been
multimodal analgesia to cover a myriad of receptors and demonstrated in a randomized, placebo-controlled trial
pain pathophysiologies. This typically includes regular in burn patients, which found lorazepam to be effective
acetaminophen (paracetamol), regular NSAIDs where in reducing pain scores compared with placebo when
appropriate, and the use of long-acting opioid prepara- administered in combination with opioid analgesics
tions, with specific recommendations varying between [216]. Benzodiazepines with a short to moderate dura-
burns centers [212]. tion of action, such as midazolam and lorazepam, are
preferred to longer-acting drugs.
The use of ketamine in the burn-injured patient is largely
reserved for the facilitation of procedures such as dress- Nitrous oxide is an inhalant anesthetic that has modest
ing changes, which will be discussed in the next sec- analgesic properties when delivered at subanesthetic
tion. A constant ketamine infusion to assist with doses [216]. Inhaled nitrous oxide and oxygen mixtures
background pain management is utilized in some burns can be a very effective form of analgesia for short pro-
centers. In addition to assisting with analgesia, ketamine cedures, such as dressing changes [202], as it has a
is thought to produce benefits through effects on the rapid onset (within seconds) and short duration of ac-
NMDA receptor, which may reduce central sensitization, tion. Although it is generally well tolerated, nitrous oxide
as discussed above. However, the psychomimetic can cause nausea and vomiting, precluding its use in
effects of ketamine may be distressing for some some patients. Repeated or prolonged administration of
patients, which limits its use [213]. nitrous oxide can interfere with vitamin B12 metabolism,
causing serious hematological and neurological adverse
effects and necessitating monitoring and vitamin B12
Procedural Pain supplementation as required [217].

An especially distressing aspect of burn care for Ketamine is an intravenous anesthetic that can have po-
patients is procedural pain from interventions including tent analgesic effects when administered at sub-
physiotherapy, baths, and dressing changes. Due to the anesthetic doses. For the management of procedural
pain, ketamine is generally administered by the intrave-
constraints of hospital services, many of these interven-
nous or intramuscular route, with a fast onset of action
tions are done on the ward rather than in the operating
(1–5 minutes) and relatively short duration of action (10–
theater under general anesthesia. The pain during these
30 minutes) [218]. Ketamine induces a dissociative state,
interventions naturally reaches quite intense levels for a
where patients appear awake but are nonresponsive,
brief period of time and is often difficult to manage.
and requires close monitoring by an experienced health
Options to manage procedural pain include combina-
professional for potential adverse effects, including air-
tions of short-acting opioids, benzodiazepines, and sub-
way obstruction due to hypersalivation and transient re-
anesthetic doses of nitrous oxide/oxygen or ketamine. spiratory depression [219]. Upon recovery, patients can
Short-acting parenteral or immediate-release opioids, experience distressing psychomimetic symptoms, in-
such as morphine, oxycodone, or fentanyl, can be used cluding delirium, agitation, and hallucinations. The inci-
for procedural pain. These can be given in addition to dence of psychomimetic effects is much lower in
the sustained-release oral or transdermal opioids that children compared with adults and can be reduced by
are providing assistance with the constant background co-administration with a benzodiazepine [218]. Despite
pain. The choice of opioid, dose, and route of adminis- these adverse effects, ketamine has been demonstrated
tration depend on the patient and type of procedure be- to provide effective analgesia for procedural pain in both
ing performed. Although they are effective analgesics, adult and pediatric burn patients [201,209,210]. Some
opioids have many dose-limiting adverse effects, includ- burns centers use an intravenous patient-controlled se-
ing sedation, respiratory depression, nausea, vomiting, dation device where patients can provide their own se-
and constipation. Depending on the duration of treat- dation using a solution containing ketamine and
ment, dose escalation may also be required as toler- midazolam [220].
ance to the analgesic effects of opioids develops over
time. However, the risk of opioid-induced hyperalgesia Regional anesthesia, where a large part of the body,
must always be considered if there is escalating use of such as an arm or leg, is anesthetized, can also be
opioids in the presence of poorly controlled pain. This used. This can include using a fascia iliaca block to pro-
effect may also be seen acutely when higher doses of vide anesthesia to the antero-lateral thigh, a common
remifentanil are used intraoperatively with subsequent site used to acquire donor skin, or a brachial plexus
hyperalgesia and difficult-to-control pain during the re- block, which provides anesthesia for procedures involv-
covery phase after surgery. Polymorphisms in the l-opi- ing the arm. On occasion, patients may require a major
oid receptor may also result in variable responses to procedure, which for various reasons cannot be per-
opioid analgesics [214]. formed in the operating theater under general
anesthesia. Therefore, a deeper level of analgesia and
Although not considered analgesic, benzodiazepines sedation, using a combination of the above analgesic/
can be effective in alleviating pain symptoms in anesthetics may be required on the burns ward.

16
Burn Pain

Staff appropriately trained with skills in pharmacotherapy following burn injury is reported to be between 25% and
and airway management are required to perform this 36%, and it is correlated with the severity of the initial
task. burn injury [226–228]. To date, there is no evidence that
any one intervention is able to reduce the incidence of
In addition to pharmacological interventions, the power chronic pain in the burn-injured population. However,
of nonpharmacological approaches cannot be underes- this lack of evidence does not negate the absolute need
timated. Hypnosis with post-hypnotic suggestion and for effective multimodal analgesia in the acute period fol-
virtual reality, a technologically advanced method of dis- lowing the injury and the requirement for this therapy to
traction where patients are actively engaged in another be reviewed and titrated on a regular basis. General
task while undergoing procedural interventions, are principles of chronic pain management should be
emerging as important nonpharmacologic options with employed and should include multimodal analgesia in
strong evidence and support [221,222]. the setting of a supportive interdisciplinary team with
psychological intervention as required. The use of long-
term opioid therapy for the management of chronic pain
Neuropathic Pain following burn injury should, as for all forms of chronic
noncancer pain, be diligently monitored for ongoing
Many of the signs and symptoms in patients with burn benefit and adverse outcomes.
injuries resemble features that are often present in those
with neuropathic pain, which is not surprising given ex- Conclusions
tensive damage to cutaneous nociceptors and conduct-
ing nerve fibers following burn injury. This neuropathic A burn injury is one of the most severe forms of trauma
pain can occur acutely, being present only at the time that a person may experience and is associated with
of burn injury, and may become chronic despite healing significant pain and numerous physical, psychological,
of the burn injury. As is generally the case with burn- material, and social losses. The impact of a burn injury
induced pain, limited randomized placebo-controlled on the patient is a result of a multitude of factors and
trials assessing the effectiveness of treatments for neu- complex processes that together drive the experience of
ropathic pain in the burn population are available. pain. First and foremost, the physical damage to the
Systematic reviews have concluded that gabapentin skin not only determines the acute nociceptive re-
and pregabalin are effective in the treatment of other sponse, but also contributes to the transition to chronic
neuropathic pain states, such as diabetic neuropathy, pain states. Pain after a burn encompasses nociceptive,
post-herpetic neuralgia, and fibromyalgia [223,224], and inflammatory, and neuropathic components and may
their use in burn pain has therefore been investigated. persist well beyond healing of the injury, with central
Gabapentin was shown to be ineffective in the treat- sensitization being a key contributor to ongoing pain.
ment of acute burn pain compared with placebo in a The complex pathophysiological mechanisms underlying
small randomized trial; however, this result could be burn pain contribute in part to the difficulties in effective
explained by a large proportion (70%) of patients with- clinical management, but also present significant oppor-
out neuropathic pain being included in the study [200]. tunities for synergistic analgesic approaches, albeit
In contrast, pregabalin was demonstrated to be effective mechanism-based treatments specific to burn pain are
in alleviating acute neuropathic symptoms following not widely used clinically as yet. Current treatment se-
burn injury in a study that only included patients with lection is driven by patient symptoms and is based on
neuropathic pain symptoms [189]. Use of systemic analgesics that are efficacious for other pain types with
lignocaine has also been investigated, although no con- similar symptoms, with the assumption that the underly-
clusions can be drawn about effectiveness in burn injury ing mechanisms are comparable.
based on current evidence [225]. While one study found
a statistically significant reduction in pain scores follow- Given the high incidence of burn injuries and the signifi-
ing intravenous administration of lignocaine, the pain cant morbidity associated even with minor burns, im-
score reduction (–0.4 on a 10-point pain scale) was un- proved pain management is essential to improve the
likely to be clinically relevant [203]. The use of tricyclic quality of life of burn patients. However, relatively few
antidepressants and other anticonvulsants that are usu- human clinical studies have been reported as burn
ally used to treat neuropathic pain has not been sys- patients typically belong to a very heterogeneous popu-
tematically investigated in the burn population, although lation and frequently present with significant
they are used in some burn centers. comorbidities. Accordingly, animal models of burn pain
have provided considerable insight into the underlying
mechanisms of pain from burn injuries and may contrib-
Treatment of Chronic Pain Following Burn Injury ute to the development of novel analgesic strategies.
With a better characterization of the underlying mecha-
Chronic pain following healing of a burn injury can have nisms that drive burn pain, improved mechanism-based
a debilitating impact on the function and quality of life of treatments promise to deliver analgesia with fewer side
patients, and finding effective management remains a effects and ultimately improve quality of life for these
significant challenge. The incidence of chronic pain patients.

17
Morgan et al.

References June 2014-June 2015. Melbourne, Victoria: Monash


1 Atchison NE, Osgood PF, Carr DB, Szyfelbein SK. University; 2015.
Pain during burn dressing change in children:
Relationship to burn area, depth and analgesic regi- 16 de Silva H, Gabbe B, Callaghan J, Liman J. Burns
mens. Pain 1991;47(1):41–5. Registry of Australia and New Zealand (BRANZ)
Annual Report June 2013-June 2014. Melbourne
2 Jackson DM. [The diagnosis of the depth of burn- Victoria: Monash University; 2014.
ing]. Br J Surg 1953;40(164):588–96.
17 de Silva H, Gabbe B, Callaghan J, Liman J. Burns
3 Lund C, Browder N. The estimation of areas of Registry of Australia and New Zealand (BRANZ)
burns. Surg Gynecol Obstet 1944;79:352. Annual Report June 2012-June 2013. Melbourne,
Victoria: Monash University; 2013.
4 Tobiasen J, Hiebert JM, Edlich RF. The abbreviated
burn severity index. Ann Emerg Med 1982;11(5):260–2. 18 Gabbe B, Picton N, Loh I, Watterson D. Bi-National
Burns Registry Annual Report June 2011-June
5 Brusselaers N, Monstrey S, Vogelaers D, Hoste E, 2012. Melbourne, Victoria: Monash University; 2012.
Blot S. Severe burn injury in Europe: A systematic
review of the incidence, etiology, morbidity, and 19 Gabbe B, Picton N, Watterson D, Morgan M,
mortality. Crit Care 2010;14(5):R188. Jennings P. Bi-National Burns Registry Annual
Report June 2010-June 2011. Melbourne, Victoria:
6 Repository NB. Report of Data from 2000–2009. Monash University; 2011.
Chicago IL, U.S.A: American Burn Association; 2010.
20 Cameron P, Gabbe B, Watterson D, Picton N,
7 Wong EG, Groen RS, Kamara TB, et al. Burns in Hannaford A. Bi-National Burns Registry Annual
Sierra Leone: A population-based assessment. Report June 2009-June 2010. Melbourne, Victoria:
Burns 2014;40(8):1748–53. Monash University; 2010.

8 American Burn Association. National Burn 21 De-Souza DA, Manco AR, Marchesan WG, Greene LJ.
Repository 2014 Report. Chicago IL, U.S.A: Epidemiological data of patients hospitalized with burns
American Burn Association; 2014. and other traumas in some cities in the southeast of
Brazil from 1991 to 1997. Burns 2002;28(2):107–14.
9 Stylianou N, Buchan I, Dunn KW. A review of the in-
ternational Burn Injury Database (iBID) for England 22 Zayakova Y, Vajarov I, Stanev A, Nenkova N,
and Wales: Descriptive analysis of burn injuries Hristov H. Epidemiological analysis of burn patients
2003-2011. BMJ Open 2015;5(2):e006184. in east Bulgaria. Burns 2014;40(4):683–8.

10 Moore EC, Pilcher D, Bailey M, Cleland H. Women 23 Pande KC, Ishak HL. Epidemiology of burns in a
are more than twice as likely to die from burns as major referral hospital in Brunei Darussalam.
men in Australia and New Zealand: An unexpected Singapore Med J 2012;53(2):124–7.
finding of the Burns Evaluation And Mortality (BEAM)
Study. J Crit Care 2014;29(4):594–8. 24 Tang K, Jian L, Qin Z, et al. Characteristics of burn
patients at a major burn center in Shanghai. Burns
11 WHO. Burns. Fact Sheet No. 365 2014. Geneva 2006;32(8):1037–43.
Switzerland: World Health Organisation; 2016.
25 Cheng W, Yan-hua R, Fang-gang N, Wei-li D, Guo-
12 Smolle C, Cambiaso-Daniel J, Forbes AA, et al. an Z. Epidemiology of 1974 burn patients at a major
Recent trends in burn epidemiology worldwide: A burn center in Beijing: A nine-year study. J Burn
systematic review. Burns 2016;43(2):249–57. Care Res 2012;33(5):e228–33.

13 Padovese V, De Martino R, Eshan MA, Racalbuto V, 26 Yao Y, Liu Y, Zhou J, et al. The epidemiology of ci-
Oryakhail MA. Epidemiology and outcome of burns vilian inpatients’ burns in Chinese military hospitals,
in Esteqlal Hospital of Kabul, Afghanistan. Burns 2001-2007. Burns 2011;37(6):1023–32.
2010;36(7):1101–6.
27 Jie X, Baoren C. Mortality rates among 5321
14 Calder F. Four years of burn injuries in a Red Cross patients with burns admitted to a burn unit in China:
hospital in Afghanistan. Burns 2002;28(6):563–8. 1980-1998. Burns 2003;29(3):239–45.

15 Dyson K, Gabbe B, Thomas T. Burns Registry of 28 Franco MA, Gonzales NC, Diaz ME, Pardo SV,
Australia and New Zealand (BRANZ) Annual Report Ospina S. Epidemiological and clinical profile of burn

18
Burn Pain

victims Hospital Universitario San Vicente de Paul, Taleghani Hospital during 2003-2007. Bratisl Lek
Medellin, 1994-2004. Burns 2006;32(8):1044–51. Listy 2010;111(7):384–8.

29 Frans FA, Keli SO, Maduro AE. The epidemiology of 42 Qader AR. Burn mortality in Iraq. Burns 2012;38
burns in a medical center in the Caribbean. Burns (5):772–5.
2008;34(8):1142–8.
43 Harats M, Peleg K, Givon A, et al. Burns in Israel,
30 Dunn K, Reade C, R Dudley-southern. International comparative study: Demographic, etiologic and clini-
Burn Injury Database Annual Report (2014/2015). cal trends 1997-2003 vs. 2004-2010. Burns 2016;
International Burn Injury Database. United Kingdom: 42(3):500–7.
National Health Service; 2010.
44 Haik J, Liran A, Tessone A, et al. Burns in Israel:
31 Ortiz-Prado E, Armijos L, Iturralde AL. A population- Demographic, etiologic and clinical trends, 1997-
based study of the epidemiology of acute adult 2003. Isr Med Assoc J 2007;9(9):659–62.
burns in Ecuador from 2005 to 2014. Burns 2015;
41(3):582–9. 45 Lancerotto L, Sferrazza R, Amabile A, Azzena B.
Burn care in relation to burn epidemiology in Italy.
32 Papp A. The first 1000 patients treated in Kuopio Burns 2011;37(5):835–41.
University Hospital Burn Unit in Finland. Burns 2009;
35(4):565–71. 46 Kobayashi K, Ikeda H, Higuchi R, et al.
Epidemiological and outcome characteristics of ma-
33 Theodorou P, Xu W, Weinand C, et al. Incidence and jor burns in Tokyo. Burns 2005;31(suppl 1):S3–11.
treatment of burns: A twenty-year experience from a
single center in Germany. Burns 2013;39(1):49–54. 47 Ndiritu S, Ngumi ZW, Nyaim O. Burns: The epidemi-
ological pattern, risk and safety awareness at
34 Agbenorku P, Aboah K, Akpaloo J, et al. Kenyatta National Hospital, Nairobi. East Afr Med J
Epidemiological studies of burn patients in a burn 2006;83(8):455–60.
center in Ghana: Any clues for prevention? Burns
Trauma 2016;4:21. 48 Otteni CR, Saruni SI, Duron VP, Hedges JP, White
RE. Baseline assessment of inpatient burn care at
35 Kumar N, Kanchan T, Unnikrishnan B, et al. Clinico- Tenwek Hospital, Bomet, Kenya. World J Surg
epidemiological profile of burn patients admitted in a 2013;37(7):1530–5.
tertiary care hospital in coastal South India. J Burn
Care Res 2012;33(5):660–7. 49 Khashaba HA, Al-Fadhli AN, Al-Tarrah KS, Wilson
YT, Moiemen N. Epidemiology and outcome of
36 Ganesamoni S, Kate V, Sadasivan J. Epidemiology burns at the Saud Al Babtain Burns, Plastic Surgery
of hospitalized burn patients in a tertiary care hospi- and Reconstructive Center, Kuwait: Our experience
tal in South India. Burns 2010;36(3):422–9. over five years (from 2006 to 2010). Ann Burns Fire
Disasters 2012;25(4):178–87.
37 Ahuja RB, Bhattacharya S. An analysis of 11,196
burn admissions and evaluation of conservative 50 Bang RL, Sharma PN, Gang RK, Ghoneim IE,
management techniques. Burns 2002;28(6):555–61. Ebrahim MK. Burn mortality during 1982 to 1997 in
Kuwait. Eur J Epidemiol 2000;16(8):731–9.
38 Ahmadijouybari T, Najafi F, Moradinazar M, et al.
Two-year hospital records of burns from a referral 51 Ghanime G, Rizkallah N, Said JM. Epidemiology of
center in Western Iran: March 2010-March 2012. J major burns at the Lebanese Burn Center in Geitawi,
Inj Violence Res 2014;6(1):31–6. Lebanon. Ann Burns Fire Disasters 2013;26(2):59–62.

39 Karimi H, Momeni M, Motevalian A, et al. The burn 52 Rimdeika R, Kazanavicius M, Kubilius D.


registry program in Iran—First report. Ann Burns Fire Epidemiology of burns in Lithuania during 1991-
Disasters 2014;27(3):154–9. 2004. Medicina (Kaunas) 2008;44(7):541–7.

40 Ansari-Moghaddam A, Baghbanian A, Dogoonchi 53 Komolafe OO, James J, Makoka M, Kalongeolera L.


M, et al. Epidemiology of burn injuries in south- Epidemiology and mortality of burns at the Queen
eastern Iran: A retrospective study. J Pak Med Elizabeth Central Hospital Blantyre, Malawi. Cent Afr
Assoc 2013;63(12):1476–81. J Med 2003;49(11–12):130–4.

41 Ekrami A, Hemadi A, Latifi M, Kalantar E. 54 Chan KY, Hairol O, Imtiaz H, et al. A review of burns
Epidemiology of hospitalized burn patients in patients admitted to the Burns Unit of Hospital

19
Morgan et al.

Universiti Kebangsaan Malaysia. Med J Malaysia 67 Babik J, Sopko K, Orsag J, Koller J. Epidemiology
2002;57(4):418–25. and therapeutic aspects of burn injuries in Slovakia
(1993-2003). Acta Chir Plast 2006;48(2):39–42.
55 Elkafssaoui S, Tourabi K, Bouaiti E, et al.
Epidemiological analysis of burn patients in the mili- 68 Golshan A, Patel C, Hyder AA. A systematic review
tary hospital, Rabat, Morocco. Ann Burns Fire of the epidemiology of unintentional burn injuries in
Disasters 2011;24(3):115–9. South Asia. J Public Health (Oxf) 2013;35
(3):384–96.
56 Dokter J, Vloemans AF, Beerthuizen GI, et al.
Epidemiology and trends in severe burns in the 69 Han TH, Kim JH, Yang MS, et al. A retrospective
Netherlands. Burns 2014;40(7):1406–14. analysis of 19,157 burns patients: 18-year experi-
ence from Hallym Burn Center in Seoul, Korea.
57 Adejumo PO, Akese MI. A five-year prevalence Burns 2005;31(4):465–70.
study of burn injury in a Nigerian teaching hospital.
World Hosp Health Serv 2012;48(1):31–4. 70 Rode H, Cox SG, Numanoglu A, Berg AM. Burn
care in South Africa: A micro cosmos of Africa.
58 Onarheim H, Jensen SA, Rosenberg BE, Pediatr Surg Int 2014;30(7):699–706.
Guttormsen AB. The epidemiology of patients with
burn injuries admitted to Norwegian hospitals in 71 Van Niekerk A, Laubscher R, Laflamme L.
2007. Burns 2009;35(8):1142–6. Demographic and circumstantial accounts of burn
mortality in Cape Town, South Africa, 2001-2004:
59 Al-Shaqsi S, Al-Kashmiri A, Al-Bulushi T. An observational register based study. BMC Public
Epidemiology of burns undergoing hospitalization to Health 2009;9:374.
the National Burns Unit in the Sultanate of Oman: A
72 Laloe V. Epidemiology and mortality of burns in a
25-year review. Burns 2013;39(8):1606–11.
general hospital of Eastern Sri Lanka. Burns 2002;
60 Siddiqui E, Zia N, Feroze A, et al. Burn injury char- 28(8):778–81.
acteristics: Findings from Pakistan National
73 Akerlund E, Huss FR, Sjoberg F. Burns in Sweden:
Emergency Department Surveillance Study. BMC
An analysis of 24,538 cases during the period
Emerg Med 2015;15(suppl 2):S5.
1987-2004. Burns 2007;33(1):31–6.
61 Farooq U, Nasrullah M, Bhatti JA, et al. Incidence
74 Chien WC, Pai L, Lin CC, Chen HC. Epidemiology
of burns and factors associated with their
of hospitalized burns patients in Taiwan. Burns
hospitalisation in Rawalpindi, Pakistan. Burns 2011;37
2003;29(6):582–8.
(3):535–40.
75 Chen SH, Chen YC, Chen TJ, Ma H. Epidemiology
62 Al Ibran E, Mirza FH, Memon AA, Farooq MZ, of burns in Taiwan: A nationwide report including
Hassan M. Mortality associated with burn injury—a inpatients and outpatients. Burns 2014;40
cross sectional study from Karachi, Pakistan. BMC (7):1397–405.
Res Notes 2013;6:545.
76 Aksoy N, Arli S, Yigit O. A retrospective analysis of
63 da Silva PN, Amarante J, Costa-Ferreira A, Silva A, the burn injury patients records in the emergency
Reis J. Burn patients in Portugal: Analysis of department, an epidemiologic study. Emerg (Tehran)
14,797 cases during 1993-1999. Burns 2003;29 2014;2(3):115–20.
(3):265–9.
77 Coruh A, Gunay GK, Esmaoglu A. A seven-year
64 Al-Hoqail RA, Fadaak H, Wafa AW. Burn injuries at burn unit experience in Kayseri, Turkey: 1996 to
a university hospital in Saudi Arabia: An audit and 2002. J Burn Care Rehabil 2005;26(1):79–84.
concept of total quality management, 1997-2003. J
Craniofac Surg 2011;22(2):404–8. 78 Eser T, Kavalci C, Aydogan C, Kayipmaz AE.
Epidemiological and cost analysis of burn injuries
65 Song C, Chua A. Epidemiology of burn injuries in admitted to the emergency department of a tertiary
Singapore from 1997 to 2003. Burns 2005;31(suppl burn center. Springerplus 2016;5(1):1411.
1):S18–26.
79 Fuzaylov G, Murthy S, Dunaev A, et al. Improving
66 Hwee J, Song C, Tan KC, Tan BK, Chong SJ. The burn care and preventing burns by establishing a
trends of burns epidemiology in a tropical regional burn database in Ukraine. Burns 2014;40
burns centre. Burns 2016;42(3):682–6. (5):1007–12.

20
Burn Pain

80 Grivna M, Eid HO, Abu-Zidan FM. Epidemiology of 92 LaMotte RH, Shain CN, Simone DA, Tsai EF.
burns in the United Arab Emirates: Lessons for pre- Neurogenic hyperalgesia: Psychophysical studies of
vention. Burns 2014;40(3):500–5. underlying mechanisms. J Neurophysiol 1991;66
(1):190–211.
81 Mosier MJ, Bernal N, Faraklas IH, Kahn SA,
Kemalyan N, Mian MA, et al. 2016 National Burn 93 Koltzenburg M, Lundberg LE, Torebjork HE.
Repository: Report of Data from 2005-2015. Dynamic and static components of mechanical
Chicago IL, U.S.A: American Burn Association hyperalgesia in human hairy skin. Pain 1992;51
National Repository; 2016. (2):207–19.

82 Schulte H, Sollevi A, Segerdahl M. The synergistic 94 Gustorff B, Anzenhofer S, Sycha T, Lehr S, Kress
effect of combined treatment with systemic keta- HG. The sunburn pain model: The stability of pri-
mine and morphine on experimentally induced mary and secondary hyperalgesia over 10 hours in
windup-like pain in humans. Anesth Analg 2004;98 a crossover setting. Anesth Analg 2004;98(1):
(6):1574–80. 173–7.

83 Raja SN, Campbell JN, Meyer RA. Evidence for dif- 95 Hoffmann RT, Schmelz M. Time course of UVA-
ferent mechanisms of primary and secondary hyper- and UVB-induced inflammation and hyperalgesia in
algesia following heat injury to the glabrous skin. human skin. Eur J Pain 1999;3:131–9.
Brain 1984;107(pt 4):1179–88.
96 Sycha T, Gustorff B, Lehr S, et al. A simple pain
84 Dahl JB, Brennum J, Arendt Nielsen L, Jensen TS, model for the evaluation of analgesic effects of
Kehlet H. The effect of pre- versus postinjury infiltra- NSAIDs in healthy subjects. Br J Clin Pharmacol
tion with lidocaine on thermal and mechanical 2003;56(2):165–72.
hyperalgesia after heat injury to the skin. Pain 1993;
53(1):43–51. 97 Mogil JS. Animal models of pain: Progress and chal-
lenges. Nat Rev Neurosci 2009;10(4):283–94.
85 Brennum J, Dahl JB, Moiniche S, Arendt Nielsen L.
Quantitative sensory examination of epidural anaes- 98 Nozaki-Taguchi N, Yaksh TL. A novel model of pri-
thesia and analgesia in man: Effects of pre- and mary and secondary hyperalgesia after mild thermal
post-traumatic morphine on hyperalgesia. Pain injury in the rat. Neurosci Lett 1998;254(1):25–8.
1994;59(2):261–71.
99 Nozaki-Taguchi N, Yaksh TL. Characterization of the
86 Petersen KL, Brennum J, Dahl JB. Experimental antihyperalgesic action of a novel peripheral mu-
evaluation of the analgesic effect of ibuprofen on pri- opioid receptor agonist—loperamide. Anesthesiology
mary and secondary hyperalgesia. Pain 1997;70(2– 1999;90(1):225–34.
3):167–74.
100 Nozaki-Taguchi N, Yaksh TL. Spinal and peripheral
87 Pedersen JL, Andersen OK, Arendt Nielsen L, mu opioids and the development of secondary tac-
Kehlet H. Hyperalgesia and temporal summation of tile allodynia after thermal injury. Anesth Analg
pain after heat injury in man. Pain 1998;74(2– 2002;94(4):968–74.
3):189–97.
101 Jun JH, Yaksh TL. The effect of intrathecal
88 Werner MU, Lassen B, Pedersen JL, Kehlet H. gabapentin and 3-isobutyl gamma-aminobutyric
Local cooling does not prevent hyperalgesia follow- acid on the hyperalgesia observed after thermal
ing burn injury in humans. Pain 2002;98(3):297–303. injury in the rat. Anesth Analg 1998;86(2):
348–54.
89 Pedersen JL, Kehlet H. Secondary hyperalgesia to
heat stimuli after burn injury in man. Pain 1998;76 102 Oatway M, Reid A, Sawynok J. Peripheral antihy-
(3):377–84. peralgesic and analgesic actions of ketamine and
amitriptyline in a model of mild thermal injury in the
90 Schulte H, Sollevi A, Segerdahl M. The distribution rat. Anesth Analg 2003;97(1):168–73.
of hyperaemia induced by skin burn injury is not
correlated with the development of secondary punc- 103 Jones TL, Sorkin LS. Calcium-permeable alpha-
tate hyperalgesia. J Pain 2004;5(4):212–7. amino-3-hydroxy-5-methyl-4-isoxazolepropionic
acid/kainate receptors mediate development, but
91 Werner MU, Lassen B, Kehlet H. Analgesic effects not maintenance, of secondary allodynia evoked
of dexamethasone in burn injury. Reg Anesth Pain by first-degree burn in the rat. J Pharmacol Exp
Med 2002;27(3):254–60. Ther 2004;310(1):223–9.

21
Morgan et al.

104 Summer GJ, Puntillo KA, Miaskowski C, et al. TrkA rightwards shift of the morphine dose-response
and PKC-epsilon in thermal burn-induced mechan- curve. Pain 2005;116(1–2):87–95.
ical hyperalgesia in the rat. J Pain 2006;7
(12):884–91. 116 Wang S, Zhang L, Ma Y, et al. Nociceptive behav-
ior following hindpaw burn injury in young rats:
105 Sorkin LS, Doom CM, Maruyama KP, Nanigian Response to systemic morphine. Pain Med 2011;
DB. Secondary hyperalgesia in the rat first degree 12(1):87–98.
burn model is independent of spinal cyclooxygen-
ase and nitric oxide synthase. Eur J Pharmacol 117 Salas MM, McIntyre MK, Petz LN, et al.
2008;587(1–3):118–23. Tetrodotoxin suppresses thermal hyperalgesia and
mechanical allodynia in a rat full thickness thermal
106 Kim HJ, Seol TK, Lee HJ, Yaksh TL, Jun JH. The injury pain model. Neurosci Lett 2015;607:108–13.
effect of intrathecal mu, delta, kappa, and alpha-2
agonists on thermal hyperalgesia induced by mild 118 Fowler M, Clifford JL, Garza TH, et al. A rat model
burn on hind paw in rats. J Anesth 2011;25 of full thickness thermal injury characterized by
(6):884–91. thermal hyperalgesia, mechanical allodynia, prono-
ciceptive peptide release and tramadol analgesia.
107 Yin K, Deuis JR, Lewis RJ, Vetter I. Transcriptomic Burns 2014;40(4):759–71.
and behavioural characterisation of a mouse model
of burn pain identify the cholecystokinin 2 receptor 119 Abdullahi A, Amini-Nik S, Jeschke MG. Animal
as an analgesic target. Mol Pain 2016;12:1–13. models in burn research. Cell Mol Life Sci 2014;71
(17):3241–55.
108 Johanek LM, Simone DA. Activation of peripheral
cannabinoid receptors attenuates cutaneous 120 Calum H, Hoiby N, Moser C. Burn mouse models.
hyperalgesia produced by a heat injury. Pain 2004; Methods Mol Biol 2014;1149:793–802.
109(3):432–42.
121 Deuis JR, Vetter I. The thermal probe test: A novel
109 Chang YW, Waxman SG. Minocycline attenuates behavioural assay to quantify thermal heat thresh-
mechanical allodynia and central sensitization fol- olds in mice. Temperature (Austin) 2016;3
lowing peripheral second-degree burn injury. J (2):199–207.
Pain 2010;11(11):1146–54.
122 Bishop T, Hewson DW, Yip PK, et al.
110 Chang YW, Tan A, Saab C, Waxman S. Unilateral Characterisation of ultraviolet-B-induced inflamma-
focal burn injury is followed by long-lasting bilateral tion as a model of hyperalgesia in the rat. Pain
allodynia and neuronal hyperexcitability in spinal 2007;131(1–2):70–82.
cord dorsal horn. J Pain 2010;11(2):119–30.
123 Davies EK, Boyle Y, Chizh BA, Lumb BM, Murrell
111 Tan AM, Samad OA, Liu S, et al. Burn injury- JC. Ultraviolet B-induced inflammation in the rat: A
induced mechanical allodynia is maintained by model of secondary hyperalgesia? Pain 2011;152
Rac1-regulated dendritic spine dysgenesis. Exp (12):2844–51.
Neurol 2013;248:509–19.
124 Duke JM, Bauer J, Fear MW, et al. Burn injury,
112 Green DP, Ruparel S, Roman L, Henry MA, gender and cancer risk: Population-based cohort
Hargreaves KM. Role of endogenous TRPV1 ago- study using data from Scotland and Western
nists in a postburn pain model of partial-thickness Australia. BMJ Open 2014;4(1):e003845.
injury. Pain 2013;154(11):2512–20.
125 Lindelof B, Krynitz B, Granath F, Ekbom A. Burn
113 Shields SD, Cheng X, Uceyler N, et al. Sodium injuries and skin cancer: A population-based co-
channel Na(v)1.7 is essential for lowering heat pain hort study. Acta Derm Venereol 2008;88(1):20–2.
threshold after burn injury. J Neurosci 2012;32
(32):10819–32. 126 Andrews N, Harper S, Issop Y, Rice AS. Novel,
nonreflex tests detect analgesic action in rodents
114 Lofgren O, Qi Y, Lundeberg T. Inhibitory effects of at clinically relevant concentrations. Ann N Y Acad
tachykinin receptor antagonists on thermally in- Sci 2011;1245:11–3.
duced inflammatory reactions in a rat model. Burns
1999;25(2):125–9. 127 Brodkin J, Frank D, Grippo R, et al. Validation and
implementation of a novel high-throughput behav-
115 Wang S, Lim G, Yang L, et al. A rat model of uni- ioral phenotyping instrument for mice. J Neurosci
lateral hindpaw burn injury: Slowly developing Methods 2014;224:48–57.

22
Burn Pain

128 Sotocinal SG, Sorge RE, Zaloum A, et al. The Rat ganglion neurons of young rats. J Peripher Nerv
Grimace Scale: A partially automated method for Syst 1999;4(3–4):270–8.
quantifying pain in the laboratory rat via facial
expressions. Mol Pain 2011;7:55. 141 Kwak IS, Choi YH, Jang YC, Lee YK.
Immunohistochemical analysis of neuropeptides
129 Granovsky Y, Matre D, Sokolik A, Lorenz J, Casey (protein gene product 9.5, substance P and calci-
KL. Thermoreceptive innervation of human gla- tonin gene-related peptide) in hypertrophic burn
brous and hairy skin: A contact heat evoked po- scar with pain and itching. Burns 2014;40
tential analysis. Pain 2005;115(3):238–47. (8):1661–7.

130 Abraira VE, Ginty DD. The sensory neurons of 142 Shaked G, Gurfinkel R, Czeiger D, Douvdevani A,
touch. Neuron 2013;79(4):618–39. Sufaro Y. Adenosine in burn blister fluid. Burns
2007;33(3):352–4.
131 Coderre TJ, Melzack R. Increased pain sensitivity
following heat injury involves a central mechanism. 143 Gao Y, Xu C, Yu K, et al. Effect of tetramethylpyra-
Behav Brain Res 1985;15(3):259–62. zine on DRG neuron P2X3 receptor involved in
transmitting pain after burn. Burns 2010;36(1):
132 Coderre TJ, Melzack R. Central neural mediators 127–34.
of secondary hyperalgesia following heat injury in
rats: Neuropeptides and excitatory amino acids. 144 King BF, Wildman SS, Ziganshina LE, Pintor J,
Neurosci Lett 1991;131(1):71–4. Burnstock G. Effects of extracellular pH on
agonism and antagonism at a recombinant
133 Caterina MJ, Schumacher MA, Tominaga M, et al. P2X2 receptor. Br J Pharmacol 1997;121
The capsaicin receptor: A heat-activated ion chan- (7):1445–53.
nel in the pain pathway. Nature 1997;389
(6653):816–24. 145 Goldberg YP, MacFarlane J, MacDonald ML,
et al. Loss-of-function mutations in the Nav1.7
134 Nagy I, Rang HP. Similarities and differences be- gene underlie congenital indifference to pain in
tween the responses of rat sensory neurons to multiple human populations. Clin Genet 2007;71(4):
noxious heat and capsaicin. J Neurosci 1999;19 311–9.
(24):10647–55.
146 Zhang Q, Raoof M, Chen Y, et al. Circulating mito-
135 Caterina MJ, Rosen TA, Tominaga M, Brake AJ, chondrial DAMPs cause inflammatory responses to
Julius D. A capsaicin-receptor homologue with a injury. Nature 2010;464(7285):104–7.
high threshold for noxious heat. Nature 1999;398
(6726):436–41. 147 Liu T, Gao YJ, Ji RR. Emerging role of Toll-like
receptors in the control of pain and itch. Neurosci
136 Radtke C, Sinis N, Sauter M, et al. TRPV channel Bull 2012;28(2):131–44.
expression in human skin and possible role in ther-
mally induced cell death. J Burn Care Res 2011;32 148 Liu T, Ji RR. Toll-like receptors and itch. In:
(1):150–9. Carstens E, Akiyama T, eds. Itch: Mechanisms and
Treatment. Boca Raton, FL: Frontiers in
137 Vriens J, Owsianik G, Hofmann T, et al. TRPM3 is Neuroscience; 2014.
a nociceptor channel involved in the detection of
noxious heat. Neuron 2011;70(3):482–94. 149 Schwacha MG, Zhang Q, Rani M, Craig T, Oppeltz
RF. Burn enhances toll-like receptor induced
138 Onuoha GN, Alpar EK. Levels of vasodilators (SP, responses by circulating leukocytes. Int J Clin Exp
CGRP) and vasoconstrictor (NPY) peptides in early Med 2012;5(2):136–44.
human burns. Eur J Clin Invest 2001;31(3):253–7.
150 Kigerl KA, de Rivero Vaccari JP, Dietrich WD,
139 Natura G, von Banchet GS, Schaible HG. Popovich PG, Keane RW. Pattern recognition
Calcitonin gene-related peptide enhances TTX- receptors and central nervous system repair. Exp
resistant sodium currents in cultured dorsal root Neurol 2014;258:5–16.
ganglion neurons from adult rats. Pain 2005;116
(3):194–204. 151 Gelfand JA, Donelan M, Hawiger A, Burke JF.
Alternative complement pathway activation
140 Szucs P, Polgar E, Spigelman I, Porszasz R, Nagy increases mortality in a model of burn injury in
I. Neurokinin-1 receptor expression in dorsal root mice. J Clin Invest 1982;70(6):1170–6.

23
Morgan et al.

152 Horakova Z, Beaven MA. Time course of histamine neonatal rat dorsal root ganglion neurones via the
release and edema formation in the rat paw after cyclic AMP-protein kinase A cascade. J Physiol
thermal injury. Eur J Pharmacol 1974;27 1996;495(pt 2):429–40.
(3):305–12.
165 Moriyama T, Higashi T, Togashi K, et al.
153 Ono I, Gunji H, Zhang JZ, Maruyama K, Kaneko F. Sensitization of TRPV1 by EP1 and IP reveals pe-
A study of cytokines in burn blister fluid related to ripheral nociceptive mechanism of prostaglandins.
wound healing. Burns 1995;21(5):352–5. Mol Pain 2005;1:3.

154 Zawacki BE. Reversal of capillary stasis and pre- 166 Deuis JR, Yin K, Cooper MA, Schroder K, Vetter I.
vention of necrosis in burns. Ann Surg 1974;180 Role of the NLRP3 inflammasome in a model of
(1):98–102. acute burn-induced pain. Burns 2017;43(2):304–9.

155 Goodwin LG, Jones CR, Richards WH, Kohn J. 167 Summer GJ, Romero-Sandoval EA, Bogen O,
Pharmacologically active substances in the urine of et al. Proinflammatory cytokines mediating burn-
burned patients. Br J Exp Pathol 1963;44:551–9. injury pain. Pain 2008;135(1–2):98–107.

156 Wirth KJ, Alpermann HG, Satoh R, Inazu M. The 168 Sachs D, Cunha FQ, Poole S, Ferreira SH. Tumour
bradykinin antagonist Hoe 140 inhibits carra- necrosis factor-alpha, interleukin-1beta and
geenan- and thermically induced paw oedema in interleukin-8 induce persistent mechanical nocicep-
rats. Agents Actions Suppl 1992;38(pt 3):428–31. tor hypersensitivity. Pain 2002;96(1–2):89–97.

157 George J, Pulickal SJ, Singh A, et al. Locally medi- 169 Safieh-Garabedian B, Poole S, Allchorne A, Winter
ated analgesic effect of bradykinin type 2 receptor J, Woolf CJ. Contribution of interleukin-1 beta to
antagonist HOE 140 during acute inflammatory the inflammation-induced increase in nerve growth
pain in rats. J Burn Care Res 2014;35(6):e391–8. factor levels and inflammatory hyperalgesia. Br J
Pharmacol 1995;115(7):1265–75.
158 Chuang HH, Prescott ED, Kong H, et al.
Bradykinin and nerve growth factor release the 170 Opree A, Kress M. Involvement of the proinflam-
capsaicin receptor from PtdIns(4,5)P2-mediated in- matory cytokines tumor necrosis factor-alpha, IL-1
hibition. Nature 2001;411(6840):957–62. beta, and IL-6 but not IL-8 in the development of
heat hyperalgesia: Effects on heat-evoked calcito-
159 Goutos I, Eldardiri M, Khan AA, Dziewulski P, nin gene-related peptide release from rat skin. J
Richardson PM. Comparative evaluation of anti- Neurosci 2000;20(16):6289–93.
pruritic protocols in acute burns. The emerging
value of gabapentin in the treatment of burns pruri- 171 Braquet M, Lavaud P, Dormont D, et al.
tus. J Burn Care Res 2010;31(1):57–63. Leukocytic functions in burn-injured patients.
Prostaglandins 1985;29(5):747–64.
160 Heggers JP, Loy GL, Robson MC, Del Beccaro
EJ. Histological demonstration of prostaglandins 172 Dallob A, Guindon Y, Goldenberg MM.
and thromboxanes in burned tissue. J Surg Res Pharmacological evidence for a role of lipoxyge-
1980;28(2):110–7. nase products in platelet-activating factor (PAF)-in-
duced hyperalgesia. Biochem Pharmacol 1987;36
161 Denzlinger C, Rapp S, Hagmann W, Keppler D. (19):3201–4.
Leukotrienes as mediators in tissue trauma.
Science 1985;230(4723):330–2. 173 Tsuda M, Ishii S, Masuda T, et al. Reduced pain
behaviors and extracellular signal-related protein ki-
162 Wang XS, Lau HY. Prostaglandin E potentiates the nase activation in primary sensory neurons by periph-
immunologically stimulated histamine release from eral tissue injury in mice lacking platelet-activating
human peripheral blood-derived mast cells through factor receptor. J Neurochem 2007;102(5):1658–68.
EP1/EP3 receptors. Allergy 2006;61(4):503–6.
174 Motoyama N, Morita K, Kitayama T, et al. Pain-re-
163 St-Jacques B, Ma W. Role of prostaglandin E2 in leasing action of platelet-activating factor (PAF)
the synthesis of the pro-inflammatory cytokine antagonists in neuropathic pain animal models and
interleukin-6 in primary sensory neurons: An in vivo the mechanisms of action. Eur J Pain 2013;17
and in vitro study. J Neurochem 2011;118(5):841–54. (8):1156–67.

164 England S, Bevan S, Docherty RJ. PGE2 modu- 175 Matsuda H, Koyama H, Sato H, et al. Role of
lates the tetrodotoxin-resistant sodium current in nerve growth factor in cutaneous wound healing:

24
Burn Pain

Accelerating effects in normal and healing-impaired 188 Gray P. Acute neuropathic pain: Diagnosis and
diabetic mice. J Exp Med 1998;187(3):297–306. treatment. Curr Opin Anaesthesiol 2008;21
(5):590–5.
176 Ueda M, Hirose M, Takei N, et al. Nerve growth
factor induces systemic hyperalgesia after thoracic 189 Gray P, Kirby J, Smith MT, et al. Pregabalin in se-
burn injury in the rat. Neurosci Lett 2002;328 vere burn injury pain: A double-blind, randomised
(2):97–100. placebo-controlled trial. Pain 2011;152(6):1279–88.

177 Ueda M, Hirose M, Takei N, et al. Foot hyperalge- 190 Gray P, Williams B, Cramond T. Successful use of
sia after thoracic burn injury-histochemical, behav- gabapentin in acute pain management following
ioral and pharmacological studies. Acta burn injury: A case series. Pain Med 2008;9
Histochemica Et Cytochemica 2001;34(6):441–50. (3):371–6.

178 Samuelsson A, Abdiu A, Wackenfors A, Sjoberg F. 191 Woolf CJ, Thompson SW. The induction and main-
Serotonin kinetics in patients with burn injuries: A tenance of central sensitization is dependent on
comparison between the local and systemic N-methyl-D-aspartic acid receptor activation; impli-
responses measured by microdialysis-a pilot study. cations for the treatment of post-injury pain hyper-
Burns 2008;34(5):617–22. sensitivity states. Pain 1991;44(3):293–9.
179 Sufka KJ, Schomburg FM, Giordano J. Receptor 192 Dougherty PM, Palecek J, Paleckova V, Sorkin LS,
mediation of 5-HT-induced inflammation and noci- Willis WD. The role of NMDA and non-NMDA excit-
ception in rats. Pharmacol Biochem Behav 1992; atory amino acid receptors in the excitation of pri-
41(1):53–6. mate spinothalamic tract neurons by mechanical,
chemical, thermal, and electrical stimuli. J Neurosci
180 Taiwo YO, Levine JD. Serotonin is a directly-acting
1992;12(8):3025–41.
hyperalgesic agent in the rat. Neuroscience 1992;
48(2):485–90.
193 Wang H, Kohno T, Amaya F, et al. Bradykinin pro-
duces pain hypersensitivity by potentiating spinal
181 Tokunaga A, Saika M, Senba E. 5-HT2A receptor
cord glutamatergic synaptic transmission. J
subtype is involved in the thermal hyperalgesic
Neurosci 2005;25(35):7986–92.
mechanism of serotonin in the periphery. Pain
1998;76(3):349–55.
194 Samad TA, Moore KA, Sapirstein A, et al.
182 Giordano J, Dyche J. Differential analgesic actions Interleukin-1beta-mediated induction of Cox-2 in
of serotonin 5-HT3 receptor antagonists in the the CNS contributes to inflammatory pain hyper-
mouse. Neuropharmacology 1989;28(4):423–7. sensitivity. Nature 2001;410(6827):471–5.

183 Sasaki M, Obata H, Kawahara K, Saito S, Goto F. 195 Ignatowski TA, Covey WC, Knight PR, et al. Brain-
Peripheral 5-HT2A receptor antagonism attenuates derived TNFalpha mediates neuropathic pain. Brain
primary thermal hyperalgesia and secondary me- Res 1999;841(1–2):70–7.
chanical allodynia after thermal injury in rats. Pain
2006;122(1–2):130–6. 196 Gaston-Johansson F, Albert M, Fagan E,
Zimmerman L. Similarities in pain descriptions of
184 Drummond PD, Dawson LF, Finch PM, et al. Up- four different ethnic-culture groups. J Pain
regulation of cutaneous alpha1-adrenoceptors after Symptom Manage 1990;5(2):94–100.
a burn. Burns 2015;41(6):1227–34.
197 Wibbenmeyer L, Sevier A, Liao J, et al. Evaluation
185 Drummond PD. alpha(1)-Adrenoceptors augment of the usefulness of two established pain assess-
thermal hyperalgesia in mildly burnt skin. Eur J ment tools in a burn population. J Burn Care Res
Pain 2009;13(3):273–9. 2011;32(1):52–60.

186 Merskey H, Bogduk N, eds. Classification of 198 Tomlinson D, von Baeyer CL, Stinson JN, Sung L.
Chronic Pain: Descriptions of Chronic Pain A systematic review of faces scales for the self-
Syndromes and Definitions of Pain Terms, 2nd ed. report of pain intensity in children. Pediatrics 2010;
Seattle: IASP Press; 1994. 126(5):e1168–98.

187 Choiniere M, Melzack R, Rondeau J, Girard N, 199 Orrey DC, Halawa OI, Bortsov AV, et al. Results of
Paquin MJ. The pain of burns: Characteristics and a pilot multicenter genotype-based randomized
correlates. J Trauma 1989;29(11):1531–9. placebo-controlled trial of propranolol to reduce

25
Morgan et al.

pain after major thermal burn injury. Clin J Pain 210 Norambuena C, Yanez J, Flores V, et al. Oral keta-
2015;31(1):21–9. mine and midazolam for pediatric burn patients: A
prospective, randomized, double-blind study. J
200 Wibbenmeyer L, Eid A, Liao J, et al. Gabapentin is Pediatr Surg 2013;48(3):629–34.
ineffective as an analgesic adjunct in the immediate
postburn period. J Burn Care Res 2014;35 211 Patterson DR, Ptacek JT, Carrougher GJ, Sharar
(2):136–42. SR. Lorazepam as an adjunct to opioid analgesics
in the treatment of burn pain. Pain 1997;72
201 Kundra P, Velayudhan S, Krishnamachari S, Gupta (3):367–74.
SL. Oral ketamine and dexmedetomidine in adults’
burns wound dressing—a randomized double blind 212 Gamst-Jensen H, Vedel PN, Lindberg-Larsen VO,
cross over study. Burns 2013;39(6):1150–6. Egerod I. Acute pain management in burn patients:
Appraisal and thematic analysis of four clinical
202 Li YX, Han WJ, Tang HT, et al. Nitrous oxide- guidelines. Burns 2014;40(8):1463–9.
oxygen mixture during burn wound dressing: A
double-blind randomized controlled study. CNS 213 McGuinness SK, Wasiak J, Cleland H, et al. A sys-
Neurosci Ther 2013;19(4):278–9. tematic review of ketamine as an analgesic agent
in adult burn injuries. Pain Med 2011;12
203 Wasiak J, Spinks A, Costello V, et al. Adjuvant use (10):1551–8.
of intravenous lidocaine for procedural burn pain
relief: A randomized double-blind, placebo-con- 214 Ren ZY, Xu XQ, Bao YP, et al. The impact of ge-
trolled, cross-over trial. Burns 2011;37(6):951–7. netic variation on sensitivity to opioid analgesics in
patients with postoperative pain: A systematic re-
204 Zor F, Ozturk S, Bilgin F, Isik S, Cosar A. Pain relief view and meta-analysis. Pain Physician 2015;18
during dressing changes of major adult burns: (2):131–52.
Ideal analgesic combination with ketamine. Burns
2010;36(4):501–5. 215 Reddy S, Patt RB. The benzodiazepines as adju-
vant analgesics. J Pain Symptom Manage 1994;9
205 Finn J, Wright J, Fong J, et al. A randomised (8):510–4.
crossover trial of patient controlled intranasal fenta-
nyl and oral morphine for procedural wound care 216 Tomi K, Mashimo T, Tashiro C, et al. Alterations in
in adult patients with burns. Burns 2004;30 pain threshold and psychomotor response associ-
(3):262–8. ated with subanaesthetic concentrations of inhala-
tion anaesthetics in humans. Br J Anaesth 1993;
206 Borland ML, Bergesio R, Pascoe EM, Turner S, 70(6):684–6.
Woodger S. Intranasal fentanyl is an equivalent an-
algesic to oral morphine in paediatric burns 217 Sanders RD, Weimann J, Maze M. Biologic effects
patients for dressing changes: A randomised dou- of nitrous oxide: A mechanistic and toxicologic re-
ble blind crossover study. Burns 2005;31(7):831–7. view. Anesthesiology 2008;109(4):707–22.

207 Robert R, Brack A, Blakeney P, et al. A double- 218 Green SM, Roback MG, Kennedy RM, Krauss B.
blind study of the analgesic efficacy of oral trans- Clinical practice guideline for emergency depart-
mucosal fentanyl citrate and oral morphine in ment ketamine dissociative sedation: 2011 update.
pediatric patients undergoing burn dressing Ann Emerg Med 2011;57(5):449–61.
change and tubbing. J Burn Care Rehabil 2003;24
(6):351–5. 219 Haas DA, Harper DG. Ketamine: A review of its
pharmacologic properties and use in ambulatory
208 Sharar SR, Bratton SL, Carrougher GJ, et al. A anesthesia. Anesth Prog 1992;39(3):61–8.
comparison of oral transmucosal fentanyl citrate
and oral hydromorphone for inpatient pediatric 220 MacPherson RD, Woods D, Penfold J. Ketamine
burn wound care analgesia. J Burn Care Rehabil and midazolam delivered by patient-controlled an-
1998;19(6):516–21. algesia in relieving pain associated with burns
dressings. Clin J Pain 2008;24(7):568–71.
209 Humphries Y, Melson M, Gore D. Superiority of
oral ketamine as an analgesic and sedative for 221 Patterson DR, Wiechman SA, Jensen M, Sharar
wound care procedures in the pediatric patient SR. Hypnosis delivered through immersive virtual
with burns. J Burn Care Rehabil 1997;18(1 pt reality for burn pain: A clinical case series. Int J
1):34–6. Clin Exp Hypn 2006;54(2):130–42.

26
Burn Pain

222 Jeffs D, Dorman D, Brown S, et al. Effect of virtual background or procedural burn pain. Cochrane
reality on adolescent pain during burn wound care. Database Syst Rev 2014;10:CD005622.
J Burn Care Res 2014;35(5):395–408.
226 Choiniere M, Melzack R, Papillon J. Pain and par-
223 Moore RA, Straube S, Wiffen PJ, Derry S, McQuay esthesia in patients with healed burns: An explor-
HJ. Pregabalin for acute and chronic pain in atory study. J Pain Symptom Manage 1991;6
adults. Cochrane Database Syst Rev 2009;3 (7):437–44.
:CD007076.
227 Ward RS, Saffle JR, Schnebly WA, Hayes Lundy
224 Moore RA, Wiffen PJ, Derry S, Toelle T, Rice AS. C, Reddy R. Sensory loss over grafted areas in
Gabapentin for chronic neuropathic pain and fibro- patients with burns. J Burn Care Rehabil 1989;10
myalgia in adults. Cochrane Database Syst Rev (6):536–8.
2014;4:CD007938.
228 Malenfant A, Forget R, Papillon J, et al. Prevalence
225 Wasiak J, Mahar PD, McGuinness SK, et al. and characteristics of chronic sensory problems in
Intravenous lidocaine for the treatment of burn patients. Pain 1996;67(2–3):493–500.

27

You might also like