ArticlePDF Available

State of the Art in Topical Wound-Healing Products

Authors:

Abstract and Figures

Chronic wounds represent a significant medical burden. Such wounds fail to normally progress through the stages of healing, often complicated by a proinflammatory milieu caused by increased proteinases, hypoxia, and bacterial burden. As a result, several modalities, such as dressings, antimicrobials, growth factors, and human skin substitutes, have been devised in an attempt to correct the chronic wound environment. This review addresses these modalities with a focus on evidence and randomized controlled trials.
Content may be subject to copyright.
WOUND HEALING
State of the Art in Topical Wound-Healing Products
Kenneth Fan, B.S.
Jennifer Tang, B.S.
Julia Escandon, M.D.
Robert S. Kirsner, M.D.,
Ph.D.
Miami, Fla.
Summary: Chronic wounds represent a significant medical burden. Such
wounds fail to normally progress through the stages of healing, often compli-
cated by a proinflammatory milieu caused by increased proteinases, hypoxia,
and bacterial burden. As a result, several modalities, such as dressings, antimi-
crobials, growth factors, and human skin substitutes, have been devised in an
attempt to correct the chronic wound environment. This review addresses these
modalities with a focus on evidence and randomized controlled trials. (Plast.
Reconstr. Surg. 127 (Suppl.): 44S, 2011.)
RATIONALE FOR
TOPICAL TREATMENT
Chronic wounds are a medical challenge, cost-
ing the U.S. health system over $25 billion
dollars per year.1Leg ulcers alone affect up-
ward of 2.5 million Americans, with 2 million work-
days lost per year.2Two-thirds of patients report neg-
ative emotional impact directly attributable to their
ulcer, illustrating the necessity for proper wound
care.3Although the causes of chronic wounds are
numerous, diabetic, arterial, venous, and pressure
ulcers constitute the majority of chronic wounds.
Despite unique pathophysiology, factors contribut-
ing to chronicity of nonhealing wounds become sim-
ilar with time4(Fig. 1).
BIOLOGY OF CHRONIC WOUNDS
Wound healing is a dynamic process of three
overlapping phases: inflammation, proliferative
phase with granulation tissue formation and epi-
thelialization, and tissue remodeling.5,6 Inflamma-
tion lasts several days in the acute healing wound
but persists in the chronic wound.7Driven by
proinflammatory cytokines,8–10 the prolonged and
overactive neutrophil response leads to increased
protease activity, mainly matrix metalloproteinases.11,12
In some cases, protease activity has been found to be
over 100 times higher in chronic compared with
acute wounds.13 Increased metalloproteinases lead
to degradation of growth factors, their receptors,11
and adhesion proteins, such as fibronectin and
vitronectin,14 preventing cell adhesion for normal
wound closure.11,13,15 As a result, topical treatments
aimed at inflammation and excess proteases have
been developed.
Wounding damages the blood supply, leading
to hypoxia,16 along with subsequent decreased ox-
idative bursts and microbicidal activity by poly-
morphonuclear leukocytes.17,18 The uncontrolled
polymorphonuclear leukocytes respond to low ox-
ygen tension by releasing proteinases and toxic
oxygen metabolites, which damages endothelial
cells, leading to cellular destruction, deposition of
fibrin, and further decreased delivery of nutrients
and oxygen, propagating a vicious cycle.16 Sys-
temic disorders, such as decreased cardiac output,
smoking, peripheral vascular disease, past irradi-
ation, and chronic infection, all contribute to hyp-
oxia in the local environment.15 Reperfusion in-
jury plays a role. During ischemia, substances such
as hypoxanthine and xanthine oxidase are made.
Reperfusion causes oxygen to react with these sub-
stances, producing superoxide bursts that further
damage the endothelium.19
The inflammatory state is also prolonged by the
presence of bacteria,20 leading to increased meta-
bolic demand and protease levels in the wound.21
The mere presence of bacteria in chronic wounds
does not affect healing.22 Definitions used for overt,
clinical infection include microorganism density
greater than 105to 106colony-forming units/g, as
these levels are used as a threshold for delayed
wound healing and disease.23–25 Symptoms such as
From the University of Miami, Miller School of Medicine,
and the Department of Dermatology and Cutaneous Surgery,
University of Miami.
Received for publication April 7, 2010; accepted July 12,
2010.
Copyright ©2010 by the American Society of Plastic Surgeons
DOI: 10.1097/PRS.0b013e3181fbe275
Disclosures: Dr. Kirsner has received honoraria as
an advisor to Organogenesis and Molnlycke and as
a speaker for Advanced Biohealing and Organogen-
esis, and research grants from Healthpoint and
Advanced Biohealing. There are no other financial
disclosures to report.
www.PRSJournal.com44S
exudate, odor, pain, change in color and texture
of tissue, wound deterioration, and rapid onset of
slough tissue herald increasing bacterial burden.21
However, infection should be considered if the
wound simply fails to heal, as symptoms are not
always reliable.20
Corticosteroids, immunosuppression, diabetes,
liver cirrhosis, and undernourishment may serve to
suppress the immune system, increasing the risk of
infection.26 Malnourishment also deprives the pa-
tient of essential vitamins and proteins necessary
for proper healing.27 Insufficient blood flow, se-
roma, necrosis, hematoma, and other local factors
further increase the risk of infection.26 The pres-
ence of foreign materials and necrotic tissue
greatly decreases the amount of bacteria necessary
for wound infection.15,28 Dressings and surgical
techniques for de´bridement of these foreign ma-
terials are available to the surgeon.
DRESSINGS
Two general categories of wound dressing
exist.21 Passive wound dressings mainly control
wound moisture levels. The synthetic polymers
in dressings afford customization in terms of
absorbency, physical form, and gas permeability
(Table 1).29–37 Active dressings locally alter the
wound’s biochemical environment.21
Passive Dressings
The rationale for occlusive dressings is often
traced to 1962, when Winter observed that mois-
ture-retaining dressing speeds epithelization of
acute, superficial compared with air-exposed
wounds in pigs.38 The following year, Hinman
and Miabach published results in humans dem-
onstrating similar results.39
The benefits of occlusion in partial-thickness
wounds are numerous. Without the impediment
of the crust seen in dry wounds, the wet dermal
surface provides a superior medium for epidermal
cells to migrate.40 Eaglstein and Mertz demon-
strated that occlusion accelerates epithelialization
in the acute split-thickness wound by 40 percent.41
Acute wound fluid under the occlusive bandage
contains substances, such as growth factors, that
stimulate proliferation of fibroblasts and endothe-
lial cells,42,43 promoting granulation tissue formation.44
Fig. 1. Flowchart of the nonhealing, chronic wound. ECM, extracellular matrix.
Volume 127, Number 1S Topical Wound Healing
45S
Table 1. Available Passive Dressing Types*
Type Description
Discharge
Amount Indications Advantage Disadvantage
Examples of Available
Products
Alginates Produced from brown
seaweed; either 100%
calcium alginate or 80:20
calcium alginate to sodium
alginate; fibrous gel is
formed when calcium in
the dressing exchanges with
sodium from the wound29;
absorbs 20 times of weight;
nonocclusive; requires
secondary dressing; loose
rope or pad forms30
Heavy Moderate to heavy
exudate in
superficial to deep
wounds31; autolytic
de´bridement32;
rope form to pack
deep or tunneling
wounds; infected
wounds
Conformable; allows gas
exchange along with
protection from
contamination30; draws
out contaminates and
excess exudate in
heavily draining
wounds33
May dehydrate wounds
with minimal
exudate;
contraindicated in
third-degree burns31;
not recommended
for dry eschar or light
exudate,34 as it may
adhere and cause
pain on removal;
need to be changed
daily
Algicell Calcium Alginate
(Dermasciences,
Princeton, N.J.);
AlgiDERM (Bard
Medical, Covington,
Ga.); CURASORB
(Kendall, Dublin,
Ireland); KALTOSTAT
(ConvaTec, Skillman,
N.J.); Melgisorb
(Mo¨ lnlycke, Go¨teborg,
Sweden); Tegagen (3M,
St. Paul, Minn.)
Foams Air bubbles in a matrix of
polyurethane or silicone
capable of holding or
releasing fluids; highly
absorbent while
maintaining moisture;
provides thermal insulation;
dressings can be applied as
sheets or liquid30; some
require secondary dressings
Moderate Moderate to heavy
exudate in
superficial to deep
wounds31; autolytic
de´bridement;
padding and
protection in high
trauma areas31;
infected wounds;
under
compression
dressing
Silicone liquid form is
conformable to the
wound30; nonlinting
facilitates atraumatic
removal34; polyurethane
foam provides uniform
dispersion of exudate in
absorbant layer with
semipermeable backing
prevents strike-through,
which is when exudates
permeate dressings,
providing an entry point
for bacteria35
Does not conform well
to deep cavity or
sinus tracts; may
dehydrate wounds
with minimal
exudate; may
promote maceration
if saturated34
Allevyn Foam (Smith &
Nephew, London, United
Kingdom); Contreet
Foam (Coloplast,
Minneapolis, Minn.) ;
CURAFOAM (Kendall,
Dublin, Ireland);
Hydrocell Foam
(Dermasciences,
Princeton, N.J.); Invacare
Foam (Invacare Supply
Group, Elyria, Ohio);
LYOFOAM (Mo¨lnlycke,
Go¨teborg, Sweden);
Mepilex (Mo¨lnlycke,);
Versiva (ConvaTec,
Skillman, N.J.)
Transparent
film Thin sheet of polyurethane
film; semipermeable,
variable water vapor
transmission rate31;
impermeable to bacteria or
fluids
Scant Dry wounds to
maintain moisture;
IV site protection;
autolytic
de´bridement;
secondary dressing
Barrier against bacteria
and contaminants34;
binds tightly leading to
protection of high-
friction areas31,33;
flexible, can be placed
over joints
No absorbency; fluid
can accumulate,
break adhesive seal,
and cause infection
and maceration;
required border of
intact skin may be
torn by adhesives;
problematic with
fragile skin31;
adherent material can
come in contact with
wound and tear off
reepithelialized skin36
Comfeel Film (Coloplast,
Minneapolis, Minn.);
OpSite (Smith &
Nephew, London,
United Kingdom);
Polyskin (Kendall,
Dublin, Ireland);
ReliaMed Transparent
Film (ReliaMed, Fort
Worth, Texas);
Tegaderm (3M, St. Paul,
Minn.)
(Continued)
Plastic and Reconstructive Surgery January Supplement 2011
46S
Table 1. Continued
Type Description
Discharge
Amount Indications Advantage Disadvantage
Examples of Available
Products
Hydrocolloids Mixture of materials such as
gelatin, pectin,
polycarboxymethylcellulose,
elastomers bonded to a
semipermeable film or a
foam sheet; highly
occlusive; brown, self-
adherent surface; usually
waterproof; absorbs
moisture slowly; available in
sheet form or
paste/granules to fill deep
wounds37
Minimal Light to moderate
exudate in shallow
full-thickness
defects31; wounds
requiring
moisture, such as
granulation tissue;
used under
compression
dressing; autolytic
de´bridement,
especially with
necrotic, dry
eschar
Available in ultrathin
conformable form;
reduce pain; highly
occlusive property allows
patient to continue daily
activity; molds well to
wound; some
transparent forms allow
visualization of wound;
barrier versus bacteria
and contaminants23, 31;
can be left for 7 days30
May leave residue or
adhere to wound
surfaces34;not
recommended with
heavy exudate, active
infection, or sinus
tracts34;contraindicated
in third-degree burns31;
injury of periwound
skin; highly occlusive
property can promote
anaerobic infection in
certain patients31;may
promote hypertrophic
granulation tissue; may
leave residue and odor
that can be mistaken
for infection
Allevyn (Smith & Nephew,
London, United
Kingdom); DuoDERM
(ConvaTec, Skillman,
N.J.); Hydrocol (Bertek,
Rockford, Ill.); Invacare
Hydrocolloid (Invacare
Supply Group, Elyria,
Ohio); Tegasorb (3M,
St. Paul, Minn.)
Hydrogels Glycerin- or water-based cross-
linked polymer gels or
sheet; up to 96%
water-based30;
semipermeable, transmits
vapor and water;
amorphous gels, sheet
dressings, or beads;
secondary dressing can
provide enhanced
absorption and
compression33; bead type
dressing can absorb
microorganisms, exudate,
or wound debris30
Minimal/
scant Minimal to
moderate exudate
in superficial to
deep wounds31;
rehydration of
wound bed; gel
form can pack
deep wounds and
conform to wound
defects; autolytic
de´bridement,
especially with
necrotic, dry
eschar; change
daily to 7 days
Cooling effect of sheet
hydrogels may provide
relief in burned or
excoriated wounds31;
can be used with
infection34
Minimal absorption;
desiccates quickly
without covering;
excess use can cause
wound maceration;
may promote yeast31;
sheet dressing
requires secondary
securement
Amorphous types include
Comfeel Triad (Coloplast,
Minneapolis, Minn.),
DuoDERM (ConvaTec,
Skillman, N.J.), and
Restore Hydrogel
(Hollister, Libertyville, Ill.);
Sheet types include
AQUAFLO (Kendall,
Dublin, Ireland), Aquasite
(Dermasciences,
Princeton, N.J.),
AQUASORB (DeRoyal,
Powell, Tenn.), and
CarraDres (Carrington
Laboratories, Irving,
Texas)
Contact
layers/low
adherent
Provides layer between
dressing and wound bed to
protect fragile healing
tissue34; single layer; reduces
adherence of wound to
dressing; woven or
nonwoven
Scant Granulation tissue
requiring some
moisture; apply
directly over the
wound or over
topical medication
Cheap; widely available Exudates will soak
through to above
dressing, not to be
used in viscous
exudates or third-
degree burns
Dermanet Wound Contact
Layer (DeRoyal, Powell,
Tenn.); Mepitel Soft
Silicone Wound Contact
Layer (Mo¨ lnlycke
Health Care, Go¨ teborg,
Sweden); Profore WCL
(Smith & Nephew);
ProGuide WCL (Smith
& Nephew); 3M
Tegapore (3M, St. Paul,
Minn.) (Continued)
Volume 127, Number 1S Topical Wound Healing
47S
Depending on the type, occlusive dressings can also
manage exudates (Table 1).45,46 Randomized trials
have demonstrated increased speed and less painful
healing in acute, superficial wounds compared with
nonocclusive modalities.47,48 An increased rate of
healing has been observed when dressings are
placed within 2 hours of injury and kept on for at
least 24 hours.49
Theoretically, a moist environment provides a
good environment for bacterial colonization and
possibly infection, which can be an impedance to
applying occlusion. This has not been shown to be
true: occlusion actually provides a barrier against
infection and reduces infection rates.23,34,36,50
Although partial-thickness healing is faster un-
der occlusion, it is less clear whether occlusive dress-
ings speed acute, full-thickness wound healing.48 In
the absence of wound site disease, full-thickness
wounds will eventually heal by means of secondary
intention.51 Although some experimental data in
pigs suggest complete healing is faster with occlusive
dressings in full-thickness wounds versus nonocclu-
sive therapy,52,53 the speed at which epithelialization
occurs and concomitant contractures are often not
optimal or desirable in patients. Most clinicians will
opt for grafts or flaps instead.
Similarly, the effect of occlusion on chronic
wounds has not been fully elucidated, as evidence
is mixed. It has been demonstrated that chronic
wound fluid under occlusive dressings actually in-
hibit cell proliferation in vitro54 by restricting entry
into the S phase and DNA production,55 and deg-
radation of adhesion proteins.14
Despite this, there are many reasons to justify the
use of occlusion in chronic wounds. It has been
shown to facilitate painless and effective autolytic
de´bridement,56 which is the use of enzymes within
the wound fluid to remove necrotic debris.36 Occlu-
sion reduces cost because of less frequent dressing
changes compared with standard gauze.57,58 Further-
more, the same principles of pain reduction,40,58 ex-
udate management, and safety59 with the use of oc-
clusive dressings apply in the chronic wound.
Currently, dressing modalities are chosen
mostly based on opinion, as high-level evidence is
conflicting.60,61 Although some meta-analyses have
demonstrated benefits of occlusive dressings in
the treatment of pressure ulcers62 and chronic
wounds,32,63 other authors have found insufficient
evidence to recommend modern dressings for
pressure ulcers,64,65 arterial leg ulcers,29 chronic
leg ulcers,66 venous ulcers,30,67 or surgical wounds
healing by secondary intention.61,68 The lack of
effective animal models available for study69 and
Table 1. Continued
Type Description
Discharge
Amount Indications Advantage Disadvantage
Examples of Available
Products
Composites Two or more distinct
dressings combined to take
advantage of each different
property; usually has an
absorptive layer, adhesive
border, and nonadherent/
semiadherent layer for
wound covering
Aquacel Hydrofiber
(ConvaTec, Skillman,
N.J.); Comfeel Plus
(Coloplast, Minneapolis,
Minn.); CovaDerm
(DeRoyal, Powell,
Tenn.); Tegaderm
Alginate (3M, St. Paul,
Minn.); Tegaderm Foam
(3M); Tegaderm
Hydrocolloid (3M);
Tegaderm with
Absorbent Pad (3M)
IV, intravenous.
*Data from Ovington L. The art and science of wound dressings in the twenty-first century. In: Falabella AF, Kirsner RS, eds. Wound Healing. Boca Raton, Fla.: Taylor & Francis Group; 2005:1–7;
Cuzzell J. Choosing a wound dressing. Geriatr Nurs. 1997;18:260–265; Hess CT. Wound Care. 5th ed. Philadelphia: Lippincott Williams & Wilkins; 2005; and Jones V, Grey JE, Harding KG.
Wound dressings. BMJ. 2006;332:777–780.
Plastic and Reconstructive Surgery January Supplement 2011
48S
level I evidence makes providing evidence-based
recommendations difficult.29,30,32,66
Awaiting better evidence, dressing choices should
be based on clinical experience.70,71 Ease of use, pa-
tient preference, amount of wound drainage, and
protection against bacteria should all be taken into
consideration (Table 1).30,64,72 Amoistyetabsorbent
environment needs to be maintained, avoiding
wound maceration or dehydration.30,70,71,73,74 Dress-
ings should minimize external forces such as friction
and sheer and should stay in place.70,71 Fear of mac-
eration and its potential complications in certain
wound types suggest completely occlusive and highly
adhesive dressings should be avoided in diabetic foot
ulcer,75 and hydrogels should be used instead.65
Active Dressings
Interest in development of active dressings stems
from the altered biology of the chronic wound (Ta-
ble 2).27,34,68,77 Potential targets include increased
bacterial load and excessive protease levels, which
lead to the development of antimicrobial, protease
inhibitor,76 and collagen dressings.21 The combina-
tion of antimicrobials with the dressings described
above affords bacterial reduction and maintains a
moist environment.
Collagen Dressings
Collagen has long been known to have a crit-
ical function in wound healing, providing platelet
aggregation; hemostasis; and chemotaxis of mac-
rophages, granulocytes, and fibroblasts.78 It was
rationalized that exogenous collagen matrices
might provide a scaffold for tissue ingrowth when
endogenous collagen is disrupted by the proteo-
lytic wound environment.21 In addition, dressings
containing oxidized regenerated cellulose bind
the high level of proteases seen in chronic wounds,
protecting growth factors from destruction and
neutralizing free radical damage.79,80
However, randomized controlled trials of 55
percent bovine collagen to 44 percent oxidized
regenerated cellulose dressing (Promogran wound
matrix; Systagenix, North Yorkshire, United
Kingdom) did not demonstrate significantly bet-
ter wound closure of Wagner grade I or II dia-
betic foot ulcers81 or venous leg ulcers,82 al-
though variability of loading (for the former
study) and dressing change procedure might
have negatively affected these results.
Another collagen dressing comprised of por-
cine-derived acellular small intestine submucosa
(Oasis Wound Matrix, Fort Worth, Texas) was found
to be equivalent to recombinant human platelet-
derived growth factor (rhPDGF)-BB in diabetic ul-
cers (see below) (becaplermin, Regranex; Systage-
nix, Commonwealth, Mass.).83 The dressing was
found to improve healing in full-thickness diabetic
ulcers84 and mixed vascular ulcers85 when compared
with standard-of-care treatment at 12 weeks and pe-
troleum gauze at 8 weeks, respectively.
TOPICAL ANTIMICROBIALS
The ideal antimicrobial exists in an equilib-
rium whereby bacteria bioburden is reduced with-
out interfering with cellular processes by means of
cytotoxicities.16,20,86 Although antibiotics have spe-
cific sites of activity, antimicrobials have a lower
incidence of resistance and multiple sites of ac-
tivity, and target a wide variety of bacteria, proto-
zoa, fungi, viruses, and prions.16,20,87 The most
commonly used products include chlorhexidine,
povidone-iodine, cadexomer iodine, alcohol, ac-
etate, hydrogen peroxide, boric acid, silver sulfa-
diazine (an antibiotic), silver nitrate, and sodium
hypochlorite (Table 3).26,45,88–106
Iodophors, a class of antiseptic including po-
vidone-iodine and cadexomer iodine, are slow-
release carriers designed to curb elemental iodine
toxicity.87 Most in vitro studies suggest that povi-
done-iodine is toxic to the cells that participate in
wound healing. In vivo studies have not found that
it promotes good wound healing.91,107 However,
evidence exists suggesting cadexomer iodine,
compared with standard-of-care regimens, results
in significantly higher complete venous ulcer heal-
ing rates.96 It is also more cost effective, results in
less infection, and has fewer adverse effects than
hydrocolloids or paraffin gauze.97
Silver compounds, particularly silver nitrate
and silver sulfadiazine, have been used widely in
burns. The reactivity of silver compounds with
negative substances such as DNA, RNA, proteins,
and components of the electron transport system
is the heart of its antimicrobial effect.87 Such re-
activity requires a carrier for consistent slow de-
livery to balance the cellular toxicity against the
microbicidal activity.
Moyer et al. found that 0.5% silver nitrate
administered on gauze dressing achieved such
balance.102,108 However, silver nitrate darkens tis-
sue, causes hyponatremia and hypochloremia,
and has been shown to be systemically absorbed.103
The very frequent applications, up to 12 times per
day, results in a large excess of silver at the wound
site.109 Furthermore, nitrate is converted to nitrite,
an oxidant capable of cell damage.
The advent of silver sulfadiazine, created from
the combination of silver nitrate and sodium sulfa-
diazine, allowed for better delivery of silver, allowing
Volume 127, Number 1S Topical Wound Healing
49S
Table 2. Available Active Dressing Types*
Type Indications Comment Commercial Available Products
Antimicrobial
dressings Moderate to heavy exudate
depending on the passive
dressing27; may be
primary or secondary
depending on the
dressing chosen34
Silver-releasing foam shown to
decrease ulcer area, odor,
leakages, and maceration77;
no significance in terms of
achieving complete wound
healing, greater reduction in
chronic, infected, ulcer size
with silver-containing foam
dressings compared with
standard foam or best local
practice after 4 wk68
Acticoat (Smith & Nephew, London, United
Kingdom); Algicell Ag (Dermasciences, Princeton,
N.J.); Aquacel Ag (ConvaTec, Skillman, N.J.);
Contreet Foam/Biatain–Ag (Coloplast, Minneapolis,
Minn.); Contreet Hydrocolloid/Comfeel Ag
(Coloplast); Invacare Silver Alginate (Invacare
Supply Group, Elyria, Ohio); Iodosorb Gel and
Iodoflex Pad (Healthpoint, Fort Worth, Texas);
Maxorb Extra (Medline, Mundelein, Ill.); ReliaMed
Silver Alginate (ReliaMed, Fort Worth, Texas);
Restore Foam with Silver (Hollister, Libertyville,
Ill.); Silvasorb (Medline, Mundelein, Ill.); Silvercel
(Johnson & Johnson, New Brunswick, N.J.); Silverlon
(Silverlon Consumer Products, Geneva, Ill.)
Collagen
dressings Moderate to heavy exudate
in superficial to deep
wounds 27; can be use in
infected wounds27; can be
used in skin grafts, donor
sites, red or yellow
wounds27
Provides a scaffold for growth
of tissue; hydrophilic nature
allows cell attachment;
collagen encourages
hemostasis; chemotactic to
fibroblasts, granulocytes,
macrophages; oxidized
regenerated cellulose
inactivates excess protease
and protects growth factors
55% bovine collagen and 45% oxidized regenerated
cellulose (Promogran wound matrix; Systagenix,
North Yorkshire, United Kingdom); 90% bovine
collagen and 10% alginate (Fibracol plus collagen
dressing with alginate; Systagenix); 100% bovine
collagen (Medifill; Human BioSciences,
Gaithersburg, Md.); 100% porcine collagen (Oasis,
Cook Biotech, Fort Worth, Texas); collagen with
sodium alginate (Colactive; Smith & Nephew,
London, United Kingdom); type 1 hydrolyzed
bovine collagen (CellerateRX; Wound Care
Innovations, Fort Lauderdale, Fla.)
*Data from Ovington L. The art and science of wound dressings in the twenty-first century. In: Falabella AF, Kirsner RS, eds. Wound Healing. Boca Raton, Fla.: Taylor & Francis Group; 2005:1–7;
Cuzzell J. Choosing a wound dressing. Geriatr Nurs. 1997;18:260–265; Hess CT. Wound Care, 5th ed. Philadelphia: Lippincott Williams & Wilkins; 2005; and Jones V, Grey JE, Harding KG.
Wound dressings. BMJ. 2006;332:777–780.
Plastic and Reconstructive Surgery January Supplement 2011
50S
Table 3. Commonly Used Antiseptics*
Type Composition Indications Comments
Povidone-iodine Iodophor; complex of
polyvinyl/pyrrolidone
and 1–10% iodine;
available as a solution,
cream, scrub, ointment
Approved for short-term
treatment of
superficial and acute
wounds89
FDA has concluded no evidence of improved or slowed
wound healing89; irrigation with povidone-iodine in 12-hr-
old wounds colonized by Staphylococcus aureus in guinea pigs
showed no significant decrease in bacterial count90; 1%
povidone-iodine decreased wound infection in surgical
wounds, whereas 5% povidone-iodine aerosol inhibited
leukocyte migration and resulted in increased infection26;
1% povidone-iodine was nonsignificant in decreasing
bacterial counts vs. normal saline in acute traumatic wounds
after 10-min soak91; 1% povidone-iodine no more effective
than saline in chronic pressure ulcers92; no effect on wound
healing in 1% formulations26; no resistance has been
demonstrated93
Cadexomer iodine Iodophor; microspheres
0.1–0.3 mm in diameter
in a cadexomer-starch
with iodine physically
fixed within the sphere;
iodine performs its
antimicrobial activity as
bacteria and exudates
are absorbed within the
sphere45; available as
ointment or cream
Short-term use in
healable, superficial
wounds with high
bacterial burden94;
silver dressings, silver
sulfadiazine, and
cadexomer effective
against MRSA95
Found to generate higher rates of complete healing at 4–6 wk
vs. control when all patients receive standard of care in
venous leg ulcers96; cadexomer iodine paste more cost
effective vs. hydrocolloid or paraffin dressings in venous
ulcers with compression therapy97; increased epidermal
regeneration in full-thickness wounds compared with
cadexomer-starch ointment or saline treatment,98 and
partial-thickness wounds88; avoid use in in patients with
thyroid abnormalities97; good absorptive properties; 1 g of
cadexomer can absorb 7 ml of liquid35
Hydrogen peroxide 3% hydrogen peroxide is
most common; 6% in
some medical
formulations
Effective debrider by
loosening necrotic
debris88
No statistical difference in wound infection rates in surgical
wounds in 3% hydrogen peroxide vs. control99; may cause
tissue embolism in open wounds95; gas-producing property
may cause oxygen bubbles in newly formed skin100
Acetic acid 0.5–1% acetic acid (white
vinegar) Decreases pH to inhibit
Pseudomonas growth Possible cytotocitity,101 although other studies have shown no
effect on the wound healing environment in vitro100
Chlorhexidine Available in 0.5–4.0% Surgical irrigation,94
hand washing Persisting activity within the stratum corneum95; no definitive
evidence of cytotoxicity88; not enough evidence to draw
conclusion regarding chlorhexidine use in open wounds88
Silver compounds Silver nitrate; silver
sulfadiazine
(antibiotic); silver
charcoal;
nanocrystalline silver–
sustained silver-
releasing systems
(dressings); silver
calcium sodium
carboxymethylcellulose
dressing
Silver sulfadiazine is
commonly used as a
topical burn wound
treatment; silver
dressings, silver
sulfadiazine, and
cadexomer effective
against MRSA95
Silver nitrate stains objects on contact, causes hypochloremia
and hyponatremia, and is deposited within kidney, spleen,
liver, and muscles102,103; delay in healing of superficial and
partial-thickness burn wounds treated with silver
sulfadiazine for the full duration of treatment104,105; slow
emergence of silver-resistant bacteria with prolonged use106;
silver sulfadiazine rarely causes temporary leukopenia102
FDA, U.S. Food and Drug Administration; MRSA, methicillin-resistant Staphylococcus aureus.
*Data from Orsted H, Sibbald RG. Topical treatment of wound infection. In: Falabella AF, Kirsner RS, eds. Wound Healing. Boca Raton, Fla: Taylor & Francis Group; 2005:599 615.
Volume 127, Number 1S Topical Wound Healing
51S
application two times per day. Fox demonstrated the
elimination of Pseudomonas in burn wounds, and
reduction in mortality and destruction in muscle
and skin compared with silver nitrate and mafenide
acetate.110,111 Since then, silver sulfadiazine gauze has
been used widely as an antibiotic in the topical treat-
ment of second- to third-degree burns. Nanocrystal-
line silver dressings represent further modification
in prolongation of silver delivery.109 With sustained
release of silver in a less rapidly deactivated form,
daily or weekly dressing change with steady silver
release can be achieved.
Despite large bodies of research on the ben-
efits of silver therapeutics, much less evidence re-
garding the effect of silver ion on the wound bed
exists.109 In vitro, silver has been shown to delay
wound healing when compared with tulle gras
dressing by means of toxicity to keratinocytes and
fibroblasts.112 In vivo, a similar delay in healing
exists with partial-thickness burns treated with sil-
ver sulfadiazine.113 Two recent Cochrane reports
found little evidence to support use of silver sul-
fadiazine in burn wounds or silver-containing
dressing in wounds because of a lack of clinical
proof of wound infection prevention or increased
rate of healing.104,105
As with occlusive dressings, a lack of high-quality
evidence exists.68,114,115 Recommendations are often
based on expert opinion or personal preference.94
Topical antiseptics should be reserved for signs of
bacterial burden detrimental to healing as evi-
denced by clinical signs (described above) or by
failure to heal to avoid possible cytotoxicity.20,21,109
GROWTH FACTORS
In a series of elegant experiments, Cohen no-
ticed that the purification of submaxillary gland
extracts led to earlier eyelid separation and erup-
tion of the incisor in mice, which eventually led to
the isolation of the first growth factor, epidermal
growth factor, and the 1986 Nobel Prize in
Medicine.116 Since Cohen’s discovery, knowledge
of growth factors has increased. In the acute
wound, growth factors function in a paracrine,
autocrine, intercrine, or endocrine manner, stim-
ulating healing.15 However, in the chronic wound,
the balance between stimulation and inhibition is
lost, as growth factors and their receptors are
destroyed,15,117 trapped,118 or maldistributed in the
chronic wound. Fibroblasts have also been re-
ported to be less responsive to growth hormone,
likely secondary to senescence.119
Platelet-Derived Growth Factor
Currently, rhPDGF-BB is the only growth fac-
tor approved by the U.S. Food and Drug Admin-
istration for use in chronic wounds (Table 4).121–123
Becaplermin is rhPDGF-BB produced by the yeast
Saccharomyces cerevisiae. The BB isoform is the only
one of the three (PDGF-AA, PDGF-AB, and PDGF-BB)
that has been shown to bind to both alpha and
beta receptors.120 Prepared as a topical gel, becapler-
min has been approved for the treatment of lower
extremity diabetic neuropathic ulcers that extend
into the subcutaneous tissue or beyond and have an
adequate blood supply (stage III or IV).
A meta-analysis of 922 patients among four
trials of rhPDGF-BB in full-thickness, nonhealing,
lower extremity diabetic ulcers found that becapler-
min with good wound care leads to a significant
increase in complete healing when compared with
placebo.121 Among the studies included in the meta-
analysis, Steed found that patients (n!118) treated
with rhPDGF-BB compared with placebo achieved
significantly higher complete wound healing rates
when evaluated at 20 weeks. Although not statisti-
cally significant, there was a greater recurrence rate
in placebo (46 percent) compared with rhPDGF-BB
(26 percent).124 This trial was the first to demon-
strate that topical application of a single growth fac-
tor could speed healing of a chronic wound. In a
pivotal trial, Wieman et al. found that patients (n!
382) treated with 100
!
g/g of becaplermin gel ex-
perienced significantly increased complete diabetic
lower extremity ulcer closure compared with place-
bo-treated patients.125
Despite the impressive findings, becaplermin
has recently received a new black box warning
from the U.S. Food and Drug Administration. Al-
though no increase in incidence of cancer was
found, there was an increased mortality secondary
to malignancy in those using three or more
tubes.122 The U.S. Food and Drug Administration
does not recommend becaplermin for those with
known malignancies. However, longer follow-up
of those patients in the original report found that
the increase in mortality among the becaplermin-
treated group did not persist.126
Epidermal growth factor and macrophage col-
ony-stimulating factor are being examined for use
in wound healing.127,128 Although initial results
were promising, fibroblast growth factor-2 has had
its production discontinued.129
ENGINEERED LIVING SKIN
Despite use of standard of care and advanced
topical treatments, some wounds fail to improve.130
Plastic and Reconstructive Surgery January Supplement 2011
52S
Another topical therapy, engineered skin, is an
option (Table 5). Engineered skin was initially
envisioned to treat burn wounds when autologous
grafts were unavailable.131 However, skin substi-
tutes have received wider applications in chronic
venous132 and diabetic133,134 wounds, stimulating
healing through a variety of potential mecha-
nisms. Two major classes of cellular engineered
skin are currently used in the United States: der-
mal and bilayered constructs.132–144
Cellular Dermal Constructs
As a living dermal substitute, human fibroblast–
derived dermal substitute is created by seeding
fibroblasts derived from neonatal foreskin onto
abioabsorbablepolyglactinmatrix(Dermagraft;Ad-
vanced BioHealing, Inc., La Jolla, Calif.).146 The fi-
broblasts secrete adhesion molecules and growth
factors as epithelialization occurs over the dermal
bed replacement.143,147,148 Dermagraft was found to
result in a faster and higher percentage of com-
plete healing in full-thickness, chronic ("6 weeks)
diabetic ulcers compared with conventional therapy.133
Bilayered Constructs
The bilayered construct is composed of living
keratinocytes for epidermis and fibroblasts de-
rived from neonatal foreskin, placed within a bo-
vine type 1 collagen matrix for dermis.149 Apligraf
(Organogenesis, Canton, Mass.) is designed to re-
semble skin, with cells producing a milieu of
growth factors, collagen, and extracellular matrix
proteins to promote reepithelialization, granula-
tion tissue formation, and angiogenesis, while pro-
viding protection against infection.132,145,150,151 In
randomized controlled trials, Apligraf achieved a
significantly faster and higher rate of complete
healing in venous ulcers132,152 and diabetic foot
ulcers,134 with lower incidences of osteomyelitis
and amputations in the latter disease compared
with the standard of care.
ENGINEERED ACELLULAR SKIN:
ACELLULAR DERMAL SUBSTITUTES
In addition to cellular constructs, acellular
constructs have been developed primarily for
burn wounds. One such product is AlloDerm
(LifeCell Corp., Branchburg, N.J.), a cadaveric
skin processed to remove cellular and antigenic
components, leaving an acellular dermal matrix
with an intact basement membrane complex.138
It has seen use in burns,139 abdominal wall
reconstruction,153 and breast reconstruction,154,155
to name a few.
Table 4. Available Growth Factors in Wound Healing*
Growth Factor Indication Advantages Disadvantages Comments
rhPDGF-BB/becaplermin
(Regranex; Systagenix,
Commonwealth,
Mass.)
Indicated for the treatment of
lower extremity diabetic
neuropathic ulcers that
extend into the
subcutaneous tissue or
beyond and have an
adequate blood supply
Adverse effects similar in
incidence across all
treatment groups,
including placebo121
New black box FDA warning: although
no increase in incidence of cancer
was found, there was an increased
mortality secondary to malignancy
in those using 3 or more tubes; the
FDA does not recommend
becaplermin for those with known
malignancies122; cost is $586 per 15-g
tube which, for a 2-cm2ulcer, will
last 4 wk; treatment typically takes at
least 10 wk, requiring 2–3 tubes123
rhPDGF-BB is produced by yeast;
moist saline changes every 12
hr121; daily application, thin
layer should be applied for 12
hr121; treatment should
continue for 10 wk or
complete ulcer healing
FDA, U.S. Food and Drug Administration.
*Data from Broughton G II, Janis JE, Attinger CE. Wound healing: An overview. Plast Reconstr Surg. 2006;117:1S–32S.
Volume 127, Number 1S Topical Wound Healing
53S
Table 5. Available Skin Substitutes*
Type FDA Indications Advantages Disadvantages
Dermal replacement
Human cryopreserved
allograft skin Temporary coverage for burns until permanent
coverage; supplied by tissue banks Long use has provided much experience Limited supply; disease transmission;
sloughing/rejection by 2 wk135,136;
difficult to remove off of wound
bed136; loss of viability and
performance with
cryopresservation137
Bioabsorbable membrane
impregnated with
human dermal
fibroblasts from
neonatal foreskin
(Dermagraft, Advanced
BioHealing, Inc., La
Jolla, Calif.)
FDA approval for Dermagraft in full-thickness
diabetic foot ulcers that have been present
for longer than 6 wk and ulcers that extend
deeper into the skin where the blood vessels
are, but do not involve tendon, muscle, joint
capsule, or bone (PMA/2001); FDA approval
for Dermagraft for ulcers secondary to
epidermolysis bullosa (HDE/2004)
Dermagraft allows the patient’s own
epithelial cells to close the wound:
biodegradable matrix allows absorption in
3–4 wk138; lower number of wound
infections, osteomyelitis, and cellulitis and
fewer surgical procedures involving the
diabetic foot ulcer133
Short shelf life unless cryopreserved;
contraindicated in ulcers with
infections, sinus tracts, or in
patients with bovine product
hypersensitivity
Human allograft skin
chemically treated for
decellularization, freeze-
dried for moisture
removal, and
stabilization of dermal
matrix (AlloDerm,
LifeCell Corp.,
Branchburg, N.J.)
Burns, abdominal wall repairs, breast
reconstruction, nasal septum reconstruction,
tympanic membrane grafting
Immediate wound coverage; acellular,
immunologically inert; allows host cell
repopulation and ultrathin split-thickness
grafts; case series reports reduction in
scarring139
Low risk of disease transmission;
contraindicated in infected or
nonvascular wounds; cost
Polymer of bovine
collagen type 1 collagen
and shark chondroitin-6-
sulfate with an overlying
silastic sheet140 (Integra;
Integra LifeSciences
Corp., Plainsboro, N.J.)
Postexcisional treatment of life-threatening full-
thickness or deep partial-thickness thermal
injury where sufficient autograft is not
available (PMA/1996)135; patients undergoing
reconstructive surgery for burn scar
contracture where there is a limited amount
of their own skin to use for autografts or
they are too ill to have more wound sites
created (2002)
Immediate wound coverage; allows ultrathin
split-thickness autograft; better cosmetic
outcome versus split-thickness autograft135;
histologic analysis shows good wound
healing with minimal scarring141;
immunologically well tolerated142; silicone
is transparent, allowing visualization
Complete wound excision before
grafting; study performed within 7
days135; learning curve of 10
procedures135; contraindications
include known hypersensitivity to
ingredients and patients with
hemochromatosis, hemosiderosis,
hemolytic anemias, and pernicious
anemia; cost
Bilayered substitute
Live epidermal
keratinocyte and dermal
fibroblast impregnated
into a type 1 bovine
collagen lattice
(Apligraf;
Organogenesis, Canton,
Mass.; formerly
Graftskin or Living Skin
Equivalent)
Noninfected partial- and full-thickness skin
ulcers secondary to venous insufficiency "1
mo duration that have not responded to
conventional treatment132 (PMA/2000); full-
thickness neuropathic diabetic foot ulcers
that have extended into the dermis but not
with tendon, muscle, capsule, or bone
exposure, of greater than 3-wk duration, that
have not adequately responded to
conventional ulcer therapy (PMA/1998)
Immediate availability; self-heal when
injured143; no clinical evidence of
rejection, sensitization, or
immunogenicity132,134,138,144; does not
contain blood vessels, sweat glands, hair
follicles, macrophages, Langerhans cells,
lymphocytes, melanocytes; no difference in
wound infection, cellulitis, or pain in
venous ulcer treatment132; contains stratum
corneum to prevent desiccation145; benefit
in large, deep, and chronic venous
ulcers132
10-day shelf life; studies performed
an average of 3.9 applications for
diabetic ulcers134 and 3.34
applications for venous ulcers132;
learning curve correlates with
outcome134; contraindications
include patients with known bovine
collagen allergies or who have
infected wounds
FDA, U.S. Food and Drug Administration; PMA, premarket approval; HDE, Humanitarian Device Exemption; 510K, device provides similar efficacy and safety to the device it replaces.
*Data from Eisenbud D, Huang NF, Luke S, Silberklang M. Skin substitutes and wound healing: Current status and challenges. Wounds 2004;16:2–17; Bello YM, Phillips TJ. Recent advances
in wound healing. JAMA. 2000;283:716–718; and Phillips TJ. New skin for old: Developments in biological skin substitutes. Arch Dermatol. 1998;134:344 –349.
Plastic and Reconstructive Surgery January Supplement 2011
54S
Another approach is to use acellular bovine
collagen matrices with shark-derived chondroitin
sulfate matrices bonded to a temporary Silastic
epidermis (Integra; Integra LifeSciences Corp.,
Plainsboro, N.J.).140 Along with serving as a con-
duit for growth factors and angiogenesis, these
acellular matrices serve as a transient biodegrad-
able scaffold as a new dermis is regenerated.143
Integra provides temporary covering when immedi-
ate grafting is not possible. Studies have shown that
Integra has similar graft take compared with non-
autograft controls in wounds with a large body sur-
face area, but with better patient satisfaction, less
hypertrophic scarring,135 and shorter hospital stays.156
Moiemen et al. demonstrated Integra’s use in recon-
structive surgery, especially in contracture and scar
repair, with patients reporting improvement in
range of motion, softness, and appearance.157
CONCLUSIONS
Wound healing has evolved much over the
past 40 years. Many therapies have been devised to
interrupt the vicious cycle of nonhealing wounds.
Exciting developments in the field of wound heal-
ing include the use of growth factors and skin
substitutes. Despite trials demonstrating the ben-
efits of the aforementioned treatments, many of
the modalities used in practice today, such as
dressings and antimicrobials, are based more on
clinical experience than evidence. High-quality
studies are lacking to evaluate such therapies.
Proof of patient benefit will provide the necessary
justification for use in clinical practice.
Robert S. Kirsner, M.D., Ph.D.
Department of Dermatology and Cutaneous Surgery
University of Miami Miller School of Medicine
1600 N.W. 10th Avenue
Rosenstiel Medical Science Building, Room 2023-A
Miami, Fla. 33136
rkirsner@med.miami.edu
REFERENCES
1. Centers for Medicare & Medicaid Services. Guidance to
surveyors for long term care facilities. Available at: http://
www.cms.hhs.gov. Accessed April 4, 2010.
2. Lin P, Phillips TJ. Approach to diagnosis of a patient with
a lower extremity ulcer. In: Falabella AF, Kirsner RS, eds.
Wound Healing. Boca Raton, Fla: Taylor & Francis Group;
2005:69–81.
3. Phillips T, Stanton B, Provan A, Lew R. A study of the impact
of leg ulcers on quality of life: Financial, social, and psy-
chologic implications. J Am Acad Dermatol. 1994;31:49–53.
4. Mustoe TA, O’Shaughnessy K, Kloeters O. Chronic wound
pathogenesis and current treatment strategies: A unifying
hypothesis. Plast Reconstr Surg. 2006;117:35S–41S.
5. Singer AJ, Clark RA. Cutaneous wound healing. N Engl
J Med. 1999;341:738–746.
6. Clark RA. Cutaneous tissue repair: Basic biologic consid-
erations. I. J Am Acad Dermatol. 1985;13:701–725.
7. Loots MA, Lamme EN, Zeegelaar J, Mekkes JR, Bos JD,
Middelkoop E. Differences in cellular infiltrate and extra-
cellular matrix of chronic diabetic and venous ulcers versus
acute wounds. J Invest Dermatol. 1998;111:850–857.
8. Wang XJ, Han G, Owens P, Siddiqui Y, Li AG. Role of TGF
beta-mediated inflammation in cutaneous wound healing.
J Investig Dermatol Symp Proc. 2006;11:112–117.
9. Trengove NJ, Bielefeldt-Ohmann H, Stacey MC. Mitogenic
activity and cytokine levels in non-healing and healing
chronic leg ulcers. Wound Repair Regen. 2000;8:13–25.
10. Wetzler C, Kampfer H, Stallmeyer B, Pfeilschifter J, Frank S.
Large and sustained induction of chemokines during im-
paired wound healing in the genetically diabetic mouse: Pro-
longed persistence of neutrophils and macrophages during
the late phase of repair. JInvestDermatol.2000;115:245–253.
11. Trengove NJ, Stacey MC, MacAuley S, et al. Analysis of the
acute and chronic wound environments: The role of pro-
teases and their inhibitors. Wound Repair Regen. 1999;7:442–
452.
12. Yager DR, Nwomeh BC. The proteolytic environment of
chronic wounds. Wound Repair Regen. 1999;7:433–441.
13. Tarnuzzer RW, Schultz GS. Biochemical analysis of acute
and chronic wound environments. Wound Repair Regen.
1996;4:321–325.
14. Grinnell F, Ho CH, Wysocki A. Degradation of fibronectin
and vitronectin in chronic wound fluid: Analysis by cell
blotting, immunoblotting, and cell adhesion assays. J Invest
Dermatol. 1992;98:410–416.
15. Stadelmann WK, Digenis AG, Tobin GR. Physiology and
healing dynamics of chronic cutaneous wounds. Am J Surg.
1998;176:26S–38S.
16. Bowler PG. Wound pathophysiology, infection and thera-
peutic options. Ann Med. 2002;34:419–427.
17. Hohn DC, MacKay RD, Halliday B, Hunt TK. Effect of O2
tension on microbicidal function of leukocytes in wounds
and in vitro. Surg Forum 1976;27:18–20.
18. Chang N, Goodson WH III, Gottrup F, Hunt TK. Direct
measurement of wound and tissue oxygen tension in post-
operative patients. Ann Surg. 1983;197:470–478.
19. Granger DN. Role of xanthine oxidase and granulocytes in
ischemia-reperfusion injury. Am J Physiol. 1988;255:H1269–
H1275.
20. Martin LK. Wound microbiology and the use of antibacte-
rial agents. In: Falabella AF, Kirsner RS, eds. Wound Healing.
Boca Raton, Fla: Taylor & Francis Group; 2005:83–101.
21. Ovington L. The art and science of wound dressings in the
twenty-first century. In: Falabella AF, Kirsner RS, eds. Wound
Healing. Boca Raton, Fla: Taylor & Francis Group; 2005:1–7.
22. Eriksson G, Eklund AE, Kallings LO. The clinical signifi-
cance of bacterial growth in venous leg ulcers. Scand J Infect
Dis. 1984;16:175–180.
23. Hutchinson JJ, Lawrence JC. Wound infection under oc-
clusive dressings. J Hosp Infect. 1991;17:83–94.
24. Pruitt BA Jr. The diagnosis and treatment of infection in the
burn patient. Burns Incl Therm Inj. 1984;11:79–91.
25. Krizek TJ, Robson MC. Evolution of quantitative bacteriol-
ogy in wound management. Am J Surg. 1975;130:579–584.
26. Viljanto J. Disinfection of surgical wounds without inhibi-
tion of normal wound healing. Arch Surg. 1980;115:253–256.
27. Hess CT, Kirsner RS. Orchestrating wound healing: Assess-
ing and preparing the wound bed. Adv Skin Wound Care
2003;16:246–257; quiz 258–259.
28. Elek SD. Experimental staphylococcal infections in the skin
of man. Ann N Y Acad Sci. 1956;65:85–90.
Volume 127, Number 1S Topical Wound Healing
55S
29. Nelson EA, Bradley MD. Dressings and topical agents for
arterial leg ulcers. Cochrane Database Syst Rev. 2007;1:
CD001836.
30. Palfreyman SJ, Nelson EA, Lochiel R, Michaels JA. Dressings
for healing venous leg ulcers. Cochrane Database Syst Rev.
2006;3:CD001103.
31. Cuzzell J. Choosing a wound dressing. Geriatr Nurs. 1997;
18:260–265.
32. Chaby G, Senet P, Vaneau M, et al. Dressings for acute and
chronic wounds: A systematic review. Arch Dermatol. 2007;
143:1297–1304.
33. Hanna JR, Giacopelli JA. A review of wound healing and
wound dressing products. J Foot Ankle Surg. 1997;36:2–14;
discussion 79.
34. Hess CT. Wound Care . 5th ed. Philadelphia: Lippincott
Williams & Wilkins; 2005.
35. Jones V, Grey JE, Harding KG. Wound dressings. BMJ. 2006;
332:777–780.
36. Falanga V. Occlusive wound dressings: Why, when, which?
Arch Dermatol. 1988;124:872–877.
37. Heyneman A, Beele H, Vanderwee K, Defloor T. A system-
atic review of the use of hydrocolloids in the treatment of
pressure ulcers. J Clin Nurs. 2008;17:1164–1173.
38. Winter GD. Formation of the scab and the rate of epithe-
lization of superficial wounds in the skin of the young
domestic pig. Nature 1962;193:293–294.
39. Hinman CD, Miabach H. Effect of air exposure and occlu-
sion on experimental human skin wounds. Nature 1963;
200:377–378.
40. Alper JC, Welch EA, Ginsberg M, Bogaars H, Maguire P.
Moist wound healing under a vapor permeable membrane.
J Am Acad Dermatol. 1983;8:347–353.
41. Eaglstein WH, Mertz PM. New methods for assessing epi-
dermal wound healing: The effects of triamcinolone ace-
tonide and polyethelene film occlusion. J Invest Dermatol.
1978;71:382–384.
42. Katz MH, Alvarez AF, Kirsner RS, Eaglstein WH, Falanga V.
Human wound fluid from acute wounds stimulates fibro-
blast and endothelial cell growth. J Am Acad Dermatol. 1991;
25:1054–1058.
43. Chen WY, Rogers AA, Lydon MJ. Characterization of bio-
logic properties of wound fluid collected during early stages
of wound healing. J Invest Dermatol. 1992;99:559–564.
44. Alper JC, Tibbetts LL, Sarazen AA Jr. The in vitro response
of fibroblasts to the fluid that accumulates under a vapor-
permeable membrane. J Invest Dermatol. 1985;84:513–515.
45. Eaglstein WH, Falanga V. Chronic wounds. Surg Clin North
Am. 1997;77:689–700.
46. Friedman SJ, Su WP. Management of leg ulcers with hydro-
colloid occlusive dressing. Arch Dermatol. 1984;120:1329–1336.
47. Barnett A, Berkowitz RL, Mills R, Vistnes LM. Comparison
of synthetic adhesive moisture vapor permeable and fine
mesh gauze dressings for split-thickness skin graft donor
sites. Am J Surg. 1983;145:379–381.
48. Nemeth AJ, Eaglstein WH, Taylor JR, Peerson LJ, Falanga V.
Faster healing and less pain in skin biopsy sites treated with an
occlusive dressing. Arch Dermatol. 1991;127:1679–1683.
49. Eaglstein WH, Davis SC, Mehle AL, Mertz PM. Optimal use
of an occlusive dressing to enhance healing: Effect of de-
layed application and early removal on wound healing. Arch
Dermatol. 1988;124:392–395.
50. Hutchinson JJ, McGuckin M. Occlusive dressings: A micro-
biologic and clinical review. Am J Infect Control 1990;18:257–
268.
51. Rivera AE, Spencer JM. Clinical aspects of full-thickness
wound healing. Clin Dermatol. 2007;25:39–48.
52. De Coninck A, Draye JP, Van Strubarq A, et al. Healing of
full-thickness wounds in pigs: Effects of occlusive and non-
occlusive dressings associated with a gel vehicle. J Dermatol
Sci. 1996;13:202–211.
53. Dyson M, Young S, Pendle CL, Webster DF, Lang SM. Com-
parison of the effects of moist and dry conditions on dermal
repair. J Invest Dermatol. 1988;91:434–439.
54. Bucalo B, Eaglstein WH, Falanga V. Inhibition of cell pro-
liferation by chronic wound fluid. Wound Repair Regen. 1993;
1:181–186.
55. Phillips TJ, al-Amoudi HO, Leverkus M, Park HY. Effect of
chronic wound fluid on fibroblasts. J Wound Care 1998;7:
527–532.
56. Kirshen C, Woo K, Ayello EA, Sibbald RG. Debridement: A
vital component of wound bed preparation. Adv Skin Wound
Care 2006;19:506–519.
57. Kim YC, Shin JC, Park CI, Oh SH, Choi SM, Kim YS. Efficacy
of hydrocolloid occlusive dressing technique in decubitus ul-
cer treatment: A comparative study. Yonsei Med J. 1996;37:181–
185.
58. Ohlsson P, Larsson K, Lindholm C, Moller M. A cost-effec-
tiveness study of leg ulcer treatment in primary care: Com-
parison of saline-gauze and hydrocolloid treatment in a
prospective, randomized study. Scand J Prim Health Care
1994;12:295–299.
59. Phillips TJ, Palko MJ, Bhawan J. Histologic evaluation of
chronic human wounds treated with hydrocolloid and non-
hydrocolloid dressings. J Am Acad Dermatol. 1994;30:61–64.
60. American Diabetes Association. Consensus Development
Conference on Diabetic Foot Wound Care; April 7-8, 1999;
Boston, Mass.
61. Vermeulen H, Ubbink DT, Goossens A, de Vos R, Legemate
DA. Systematic review of dressings and topical agents for
surgical wounds healing by secondary intention. Br J Surg.
2005;92:665–672.
62. Bradley M, Cullum N, Nelson EA, Petticrew M, Sheldon T,
Torgerson D. Systematic reviews of wound care manage-
ment: (2). dressings and topical agents used in the healing
of chronic wounds. Health Technol Assess. 1999;3:1–35.
63. Singh A, Halder S, Menon GR, et al. Meta-analysis of ran-
domized controlled trials on hydrocolloid occlusive dress-
ing versus conventional gauze dressing in the healing of
chronic wounds. Asian J Surg. 2004;27:326–332.
64. Reddy M, Gill SS, Kalkar SR, Wu W, Anderson PJ, Rochon
PA. Treatment of pressure ulcers: A systematic review.
JAMA. 2008;300:2647–2662.
65. Edwards J, Stapley S. Debridement of diabetic foot ulcers.
Cochrane Database Syst Rev. 2010;1:CD003556.
66. Bouza C, Munoz A, Amate JM. Efficacy of modern dressings
in the treatment of leg ulcers: A systematic review. Wound
Repair Regen. 2005;13:218–229.
67. Palfreyman S, Nelson EA, Michaels JA. Dressings for venous
leg ulcers: Systematic review and meta-analysis. BMJ. 2007;
335:244.
68. Vermeulen H, van Hattem JM, Storm-Versloot MN, Ubbink
DT. Topical silver for treating infected wounds. Cochrane
Database Syst Rev. 2007;1:CD005486.
69. Sisco M, Mustoe TA. Animal models of ischemic wound heal-
ing: Toward an approximation of human chronic cutaneous
ulcers in rabbit and rat. Methods Mol Med. 2003;78:55–65.
70. Whitney J, Phillips L, Aslam R, et al. Guidelines for the treat-
ment of pressure ulcers. Wound Repair Regen. 2006;14:663–679.
71. Steed DL, Attinger C, Colaizzi T, et al. Guidelines for the
treatment of diabetic ulcers. Wound Repair Regen. 2006;14:
680–692.
Plastic and Reconstructive Surgery January Supplement 2011
56S
72. O’Donnell TF Jr, Lau J. A systematic review of randomized
controlled trials of wound dressings for chronic venous
ulcer. J Vasc Surg. 2006;44:1118–1125.
73. Hopf HW, Ueno C, Aslam R, et al. Guidelines for the treat-
ment of arterial insufficiency ulcers. Wound Repair Regen.
2006;14:693–710.
74. Robson MC, Cooper DM, Aslam R, et al. Guidelines for the
treatment of venous ulcers. Wound Repair Regen. 2006;14:
649–662.
75. Apelqvist J, Bakker K, van Houtum WH, Nabuurs-Franssen
MH, Schaper NC. International consensus and practical
guidelines on the management and the prevention of the
diabetic foot: International Working Group on the Diabetic
Foot. Diabetes Metab Res Rev. 2000;16(Suppl 1):S84–S92.
76. Edwards JV, Bopp AF, Batiste S, et al. Inhibition of elastase
by a synthetic cotton-bound serine protease inhibitor: In
vitro kinetics and inhibitor release. Wound Repair Regen.
1999;7:106–118.
77. Jorgensen B, Price P, Andersen KE, et al. The silver-releas-
ing foam dressing, Contreet Foam, promotes faster healing
of critically colonised venous leg ulcers: A randomised,
controlled trial. Int Wound J. 2005;2:64–73.
78. Purna SK, Babu M. Collagen based dressings: A review.
Burns 2000;26:54–62.
79. Cullen B, Watt PW, Lundqvist C, et al. The role of oxidised
regenerated cellulose/collagen in chronic wound repair
and its potential mechanism of action. Int J Biochem Cell Biol.
2002;34:1544–1556.
80. Lobmann R, Zemlin C, Motzkau M, Reschke K, Lehnert H.
Expression of matrix metalloproteinases and growth factors
in diabetic foot wounds treated with a protease absorbent
dressing. J Diabetes Complications 2006;20:329–335.
81. Veves A, Sheehan P, Pham HT. A randomized, controlled
trial of Promogran (a collagen/oxidized regenerated cel-
lulose dressing) vs standard treatment in the management
of diabetic foot ulcers. Arch Surg. 2002;137:822–827.
82. Vin F, Teot L, Meaume S. The healing properties of Pro-
mogran in venous leg ulcers. J Wound Care 2002;11:335–341.
83. Niezgoda JA, Van Gils CC, Frykberg RG, Hodde JP. Ran-
domized clinical trial comparing OASIS Wound Matrix to
Regranex Gel for diabetic ulcers. Adv Skin Wound Care 2005;
18:258–266.
84. Mostow EN, Haraway GD, Dalsing M, Hodde JP, King D;
OASIS Venus Ulcer Study Group. Effectiveness of an ex-
tracellular matrix graft (OASIS wound matrix) in the treat-
ment of chronic leg ulcers: A randomized clinical trial. J Vasc
Surg. 2005;41:837–843.
85. Romanelli M, Dini V, Bertone MS. Randomized comparison
of OASIS wound matrix versus moist wound dressing in the
treatment of difficult-to-heal wounds of mixed arterial/
venous etiology. Adv Skin Wound Care 2010;23:34–38.
86. Mustoe T. Understanding chronic wounds: A unifying hy-
pothesis on their pathogenesis and implications for ther-
apy. Am J Surg. 2004;187:65S–70S.
87. McDonnell G, Russell AD. Antiseptics and disinfectants:
Activity, action, and resistance. Clin Microbiol Rev. 1999;12:
147–179.
88. Drosou A, Falabella A, Kirsner RS. Antiseptic on wounds:
Area of controversy. Wounds 2003;15:149–166.
89. 56 Federal Register 33644 at 33662.
90. Howell JM, Stair TO, Howell AW, Mundt DJ, Falcone A,
Peters SR. The effect of scrubbing and irrigation with nor-
mal saline, povidone iodine, and cefazolin on wound bac-
terial counts in a guinea pig model. Am J Emerg Med. 1993;
11:134–138.
91. Lammers RL, Fourre M, Callaham ML, Boone T. Effect of
povidone-iodine and saline soaking on bacterial counts in
acute, traumatic, contaminated wounds. Ann Emerg Med.
1990;19:709–714.
92. Kucan JO, Robson MC, Heggers JP, Ko F. Comparison of
silver sulfadiazine, povidone-iodine and physiologic saline
in the treatment of chronic pressure ulcers. J Am Geriatr Soc.
1981;29:232–235.
93. Houang ET, Gilmore OJ, Reid C, Shaw EJ. Absence of
bacterial resistance to povidone iodine. J Clin Pathol. 1976;
29:752–755.
94. Landis SJ. Chronic wound infection and antimicrobial use.
Adv Skin Wound Care 2008;21:531–42.
95. Orsted H, Sibbald RG. Topical treatment of wound infec-
tion. In: Falabella AF, Kirsner RS, eds. Wound Healing. Boca
Raton, Fla: Taylor & Francis Group; 2005:599–615.
96. O’Meara S, Al-Kurdi D, Ologun Y, Ovington LG. Antibiotics
and antiseptics for venous leg ulcers. Cochrane Database Syst
Rev. 2010;1:CD003557.
97. Hansson C. The effects of cadexomer iodine paste in the
treatment of venous leg ulcers compared with hydrocolloid
dressing and paraffin gauze dressing: Cadexomer iodine
study group. Int J Dermatol. 1998;37:390–396.
98. Lamme EN, Gustafsson TO, Middelkoop E. Cadexomer-
iodine ointment shows stimulation of epidermal regener-
ation in experimental full-thickness wounds. Arch Dermatol
Res. 1998;290:18–24.
99. Lau WY, Wong SH. Randomized, prospective trial of topical
hydrogen peroxide in appendectomy wound infection:
High risk factors. Am J Surg. 1981;142:393–397.
100. Gruber RP, Vistnes L, Pardoe R. The effect of commonly
used antiseptics on wound healing. Plast Reconstr Surg. 1975;
55:472–476.
101. Smith RG. A critical discussion of the use of antiseptics in
acute traumatic wounds. J Am Podiatr Med Assoc. 2005;95:
148–153.
102. Klasen HJ. A historical review of the use of silver in the
treatment of burns: II. renewed interest for silver. Burns
2000;26:131–138.
103. Bader KF. Organ deposition of silver following silver nitrate
therapy of burns. Plast Reconstr Surg. 1966;37:550–551.
104. Storm-Versloot MN, Vos CG, Ubbink DT, Vermeulen H.
Topical silver for preventing wound infection. Cochrane Da-
tabase Syst Rev. 2010;3:CD006478.
105. Wasiak J, Cleland H, Campbell F. Dressings for superficial
and partial thickness burns. Cochrane Database Syst Rev. 2008;
4:CD002106.
106. Hendry AT, Stewart IO. Silver-resistant Enterobacteriaceae
from hospital patients. Can J Microbiol. 1979;25:915–921.
107. Kramer SA. Effect of povidone-iodine on wound healing: A
review. J Vasc Nurs. 1999;17:17–23.
108. Moyer CA, Brentano L, Gravens DL, Margraf HW, Monafo
WW Jr. Treatment of large human burns with 0.5 per cent
silver nitrate solution. Arch Surg. 1965;90:812–867.
109. Atiyeh BS, Costagliola M, Hayek SN, Dibo SA. Effect of silver
on burn wound infection control and healing: Review of the
literature. Burns 2007;33:139–148.
110. Fox CL Jr. Silver sulfadiazine: A new topical therapy for
Pseudomonas in burns. Therapy of Pseudomonas infection in
burns. Arch Surg. 1968;96:184–188.
111. Lowbury EJ, Babb JR, Bridges K, Jackson DM. Topical che-
moprophylaxis with silver sulphadiazine and silver nitrate
chlorhexidine creams: Emergence of sulphonamide-resis-
tant Gram-negative bacilli. BMJ. 1976;1:493–496.
112. Poon VK, Burd A. In vitro cytotoxity of silver: Implication
for clinical wound care. Burns 2004;30:140–147.
Volume 127, Number 1S Topical Wound Healing
57S
113. Stern HS. Silver sulphadiazine and the healing of partial
thickness burns: A prospective clinical trial. Br J Plast Surg.
1989;42:581–585.
114. O’Meara SM, Cullum NA, Majid M, Sheldon TA. Systematic
review of antimicrobial agents used for chronic wounds. Br
J Surg. 2001;88:4–21.
115. Bergin SM, Wraight P. Silver based wound dressings and
topical agents for treating diabetic foot ulcers. Cochrane
Database Syst Rev. 2006;1:CD005082.
116. Cohen S. Isolation of a mouse submaxillary gland protein
accelerating incisor eruption and eyelid opening in the
new-born animal. J Biol Chem. 1962;237:1555–1562.
117. Doughty D, Sparks-Defriese B. Wound-healing physiology.
In: Bryant RA, Nix DP, eds. Acute & Chronic Wounds: Current
Management Concepts. 3rd ed. St. Louis: Mosby/Elsevier;
2007:56–81.
118. Higley HR, Ksander GA, Gerhardt CO, Falanga V. Extrav-
asation of macromolecules and possible trapping of trans-
forming growth factor-beta in venous ulceration. Br J Der-
matol. 1995;132:79–85.
119. Harding KG, Morris HL, Patel GK. Science, medicine and the
future: Healing chronic wounds. BMJ. 2002;324:160–163.
120. Hammacher A, Mellstrom K, Heldin CH, Westermark B.
Isoform-specific induction of actin reorganization by plate-
let-derived growth factor suggests that the functionally ac-
tive receptor is a dimer. EMBO J. 1989;8:2489–2495.
121. Smiell JM, Wieman TJ, Steed DL, Perry BH, Sampson AR,
Schwab BH. Efficacy and safety of becaplermin (recombi-
nant human platelet-derived growth factor-BB) in patients
with nonhealing, lower extremity diabetic ulcers: A com-
bined analysis of four randomized studies. Wound Repair
Regen. 1999;7:335–346.
122. U.S. Food and Drug Administration. FDA announces new
labeling changes for Regranex: Product to carry boxed
warning. Available at: http://www.fda.gov/NewsEvents/
Newsroom/PressAnnouncements/2008/ucm116909.htm.
Accessed March 18, 2010.
123. Jarvis CI. Becaplermin (Regranex) for diabetic foot ulcers.
Am Fam Physician 2008;78:255–256.
124. Steed DL. Clinical evaluation of recombinant human plate-
let-derived growth factor for the treatment of lower extrem-
ity diabetic ulcers. Diabetic Ulcer Study Group. J Vasc Surg.
1995;21:71–78; discussion 79–81.
125. Wieman TJ, Smiell JM, Su Y. Efficacy and safety of a topical gel
formulation of recombinant human platelet-derived growth
factor-BB (becaplermin) in patients with chronic neuropathic
diabetic ulcers: A phase III randomized placebo-controlled
double-blind study. Diabetes Care 1998;21:822–827.
126. Li W. Personal communication, 2008.
127. Brown GL, Nanney LB, Griffen J, et al. Enhancement of
wound healing by topical treatment with epidermal growth
factor. N Engl J Med. 1989;321:76–79.
128. Kaplan G, Walsh G, Guido LS, et al. Novel responses of
human skin to intradermal recombinant granulocyte/mac-
rophage-colony-stimulating factor: Langerhans cell recruit-
ment, keratinocyte growth, and enhanced wound healing.
J Exp Med. 1992;175:1717–1728.
129. Janis JE, Kwon RK, Lalonde DH. A practical guide to wound
healing. Plast Reconstr Surg. 2010;125:230e–244e.
130. Limova M. Cultured skin substitutes. In: Falabella AF,
Kirsner RS, eds. Wound Healing. Boca Raton, Fla: Taylor &
Francis Group; 2005:545–559.
131. Eisenbud D, Huang NF, Luke S, Silberklang M. Skin sub-
stitutes and wound healing: Current status and challenges.
Wounds 2004;16:2–17.
132. Falanga V, Margolis D, Alvarez O, et al. Rapid healing of
venous ulcers and lack of clinical rejection with an alloge-
neic cultured human skin equivalent. Human Skin Equiv-
alent Investigators Group. Arch Dermatol. 1998;134:293–300.
133. Marston WA, Hanft J, Norwood P, Pollak R; Dermagraft
Diabetic Foot Ulcer Study Group. The efficacy and safety of
Dermagraft in improving the healing of chronic diabetic
foot ulcers: Results of a prospective randomized trial. Dia-
betes Care 2003;26:1701–1705.
134. Veves A, Falanga V, Armstrong DG, Sabolinski ML; Apligraf
Diabetic Foot Ulcer Study. Graftskin, a human skin equiv-
alent, is effective in the management of noninfected neu-
ropathic diabetic foot ulcers: A prospective randomized
multicenter clinical trial. Diabetes Care 2001;24:290–295.
135. Heimbach D, Luterman A, Burke J, et al. Artificial dermis
for major burns: A multi-center randomized clinical trial.
Ann Surg. 1988;208:313–320.
136. Purdue GF, Hunt JL, Still JM Jr, et al. A multicenter clinical
trial of a biosynthetic skin replacement, Dermagraft-TC,
compared with cryopreserved human cadaver skin for tem-
porary coverage of excised burn wounds. J Burn Care Rehabil.
1997;18:52–57.
137. Hansbrough JF, Mozingo DW, Kealey GP, Davis M, Gidner
A, Gentzkow GD. Clinical trials of a biosynthetic temporary
skin replacement, Dermagraft-Transitional Covering, com-
pared with cryopreserved human cadaver skin for tempo-
rary coverage of excised burn wounds. J Burn Care Rehabil.
1997;18:43–51.
138. Phillips TJ. New skin for old: Developments in biological
skin substitutes. Arch Dermatol. 1998;134:344–349.
139. Wainwright DJ. Use of an acellular allograft dermal matrix
(AlloDerm) in the management of full-thickness burns.
Burns 1995;21:243–248.
140. Burke JF, Yannas IV, Quinby WC Jr, Bondoc CC, Jung WK.
Successful use of a physiologically acceptable artificial skin
in the treatment of extensive burn injury. Ann Surg. 1981;
194:413–428.
141. Stern R, McPherson M, Longaker MT. Histologic study of
artificial skin used in the treatment of full-thickness thermal
injury. J Burn Care Rehabil. 1990;11:7–13.
142. Michaeli D, McPherson M. Immunologic study of artificial
skin used in the treatment of thermal injuries. J Burn Care
Rehabil. 1990;11:21–26.
143. Jimenez PA, Jimenez SE. Tissue and cellular approaches to
wound repair. Am J Surg. 2004;187:56S–64S.
144. Eaglstein WH, Alvarez OM, Auletta M, et al. Acute exci-
sional wounds treated with a tissue-engineered skin (Apli-
graf). Dermatol Surg. 1999;25:195–201.
145. Wilkins LM, Watson SR, Prosky SJ, Meunier SF, Parenteau
NL. Development of a bilayered living skin construct for
clinical applications. Biotechnol Bioeng. 1994;43:747–756.
146. Cooper ML, Hansbrough JF, Spielvogel RL, Cohen R, Bartel
RL, Naughton G. In vivo optimization of a living dermal
substitute employing cultured human fibroblasts on a bio-
degradable polyglycolic acid or polyglactin mesh. Biomate-
rials 1991;12:243–248.
147. Herndon DN. Perspectives in the use of allograft. J Burn Care
Rehabil. 1997;18:S6.
148. Naughton G, Mansbridge J, Gentzkow G. A metabolically
active human dermal replacement for the treatment of
diabetic foot ulcers. Artif Organs 1997;21:1203–1210.
149. Bello YM, Phillips TJ. Recent advances in wound healing.
JAMA. 2000;283:716–718.
150. Sabolinski ML, Alvarez O, Auletta M, Mulder G, Parenteau
NL. Cultured skin as a ‘smart material’ for healing wounds:
Experience in venous ulcers. Biomaterials 1996;17:311–320.
Plastic and Reconstructive Surgery January Supplement 2011
58S
151. Broughton G II, Janis JE, Attinger CE. Wound healing: An
overview. Plast Reconstr Surg. 2006;117:1S–32S.
152. Jones JE, Nelson EA. Skin grafting for venous leg ulcers.
Cochrane Database Syst Rev. 2007;2:CD001737.
153. Buinewicz B, Rosen B. Acellular cadaveric dermis (Allo-
Derm): A new alternative for abdominal hernia repair. Ann
Plast Surg. 2004;52:188–194.
154. Spear SL, Parikh PM, Reisin E, Menon NG. Acellular der-
mis-assisted breast reconstruction. Aesthetic Plast Surg. 2008;
32:418–425.
155. Gamboa-Bobadilla GM. Implant breast reconstruction us-
ing acellular dermal matrix. Ann Plast Surg. 2006;56:22–25.
156. Ryan CM, Schoenfeld DA, Malloy M, Schulz JT III, Sheridan
RL, Tompkins RG. Use of Integra artificial skin is associated
with decreased length of stay for severely injured adult burn
survivors. J Burn Care Rehabil. 2002;23:311–317.
157. Moiemen NS, Staiano JJ, Ojeh NO, Thway Y, Frame JD.
Reconstructive surgery with a dermal regeneration tem-
plate: Clinical and histologic study. Plast Reconstr Surg. 2001;
108:93–103.
Volume 127, Number 1S Topical Wound Healing
59S
... Para que uma ferida tenha uma boa e completa evolução, é necessária uma boa limpeza com solução fisiológica, associada ao processo de debridamento. O ato de debridar o tecido tem a intenção de remover microrganismos e revitalizar e expor o tecido saudável (DAVIDSON, 2015;FAN et al., 2011). Neste caso, o animal apresentava presença de exsudatos e início de áreas necróticas, dessa forma, optou-se pelo debridamento autolítico, com o uso de adesivos de hidrocolóides, na intenção de manter um meio úmido e em temperatura de 37 graus para realizar a lise e fagocitose do tecido necrótico (FAN et al., 2011). ...
... O ato de debridar o tecido tem a intenção de remover microrganismos e revitalizar e expor o tecido saudável (DAVIDSON, 2015;FAN et al., 2011). Neste caso, o animal apresentava presença de exsudatos e início de áreas necróticas, dessa forma, optou-se pelo debridamento autolítico, com o uso de adesivos de hidrocolóides, na intenção de manter um meio úmido e em temperatura de 37 graus para realizar a lise e fagocitose do tecido necrótico (FAN et al., 2011). ...
... Seu uso demonstrou bons resultados em feridas inflamatórias, pois auxilia a reduzir a secreção e a reparação das feridas (FAN et al., 2011), pois em contato com as secreções o hidrocolóide absorve o exsudato e forma uma camada de gel (DINIZ, 2021), fato este que era observado nas trocas do curativo do paciente relatado, visto que visualizava um conteúdo viscoso semelhante ao gel. ...
Article
A Síndrome Familiar do Shar-Pei Chinês (SFSPC) é uma doença hereditária que afeta a pele e os tecidos musculoesqueléticos. Este relato de caso descreve o uso de adesivo à base de carboximetilcelulose para a cicatrização de uma ferida no membro pélvico de um cão suspeito de ter SFSPC. Um cão Shar-Pei Chinês, suspeito de SFSPC, apresentou uma ferida profunda e extensa no membro pélvico. O tratamento convencional incluiu limpeza da ferida, terapia com antibióticos e curativos regulares. Além disso, um adesivo à base de carboximetilcelulose foi aplicado na ferida. Durante o período de tratamento, parâmetros de cicatrização como tempo de fechamento da ferida, granulação tecidual e resposta inflamatória foram avaliados. A aplicação do adesivo à base de carboximetilcelulose resultou em uma cicatrização acelerada da ferida, com fechamento mais rápido e melhora significativa na granulação tecidual. Além disso, observou-se uma redução na resposta inflamatória local. Este relato de caso sugere que o adesivo à base de carboximetilcelulose pode ser uma opção terapêutica eficaz para a cicatrização de feridas nos membros pélvicos de cães suspeitos de SFSPC. Estudos adicionais são necessários para avaliar a eficácia e segurança deste adesivo, proporcionando assim evidências científicas mais robustas. Palavras-chave: Cães. Afecções de Pele Recorrentes. Síndrome Hereditária. Carboximetilcelulose. AbstractThe Familial Chinese Shar-Pei Syndrome (FCSPS) is a hereditary disease that affects the skin and musculoskeletal tissue. This case report describes the use of a carboxymethylcellulose-based adhesive for the healing of a wound on the pelvic limb of a dog suspected of having FCSPS. A Chinese Shar-Pei dog, suspected of FCSPS, presented with a deep and extensive wound on the pelvic limb. The conventional treatment included wound cleaning, antibiotic therapy, and regular dressings. Additionally, a carboxymethylcellulose-based adhesive was applied to the wound. During the treatment period, healing parameters such as wound closure time, tissue granulation, and inflammatory response were evaluated. The application of the carboxymethylcellulose-based adhesive resulted in accelerated wound healing, with faster closure and significant improvement in tissue granulation. Furthermore, a reduction in the local inflammatory response was observed. This case report suggests that the carboxymethylcellulose-based adhesive may be an effective therapeutic option for wound healing in the pelvic limbs of dogs suspected of FCSPS. Further studies are needed to evaluate the efficacy and safety of this adhesive, thus providing more robust scientific evidence. Keywords: Dogs. Recurrent Skin Issues. Hereditary Syndrome. Carboxymethylcellulose.
... The Acacia catechu shrub has been traditionally used for the treatment of several medical conditions, such as asthma, bronchitis, cancer, chest pain, diarrhoea, mouth sores, sore throats, ulceration, vitiligo, and wound healing. Additionally, this shrub has shown antifungal, antiviral, spasmolytic, and hypoglycemic properties [23,24]. ...
... Hence, the possible wound-healing power of test extracts can be explained on the basis of their anti-oxidative and anti-bacterial activity of the same [57]. The existing body of literature has a multitude of publications discussing natural compounds that possess the capacity to facilitate the process of wound healing [20,24,45,50]. These compounds have shown promise as viable treatments throughout the many stages of wound healing [1,41]. ...
... The excessive generation of oxidative stress subsequently induces cytotoxic effects and delays the process of wound healing [27,49]. Consequently, the inhibition of oxidative stress might potentially serve as a significant approach in facilitating the healing of chronic wounds [22,24]. Some earlier reports also emphasized the potential antioxidative potential of test plant samples promoted wound healing either via scavenging free radicals or via inhibiting the generation of more free radicals at wound sites [26,32,37,58]. ...
Article
Full-text available
Objective: The objective of this work is to elucidate the wound healing capabilities of various extracts derived from the bark of Acacia catechu, and to explore their potential therapeutic effects. Furthermore, the examination of several seasons has been conducted to assess their influence on the examined parameters. Methods: In this study, albino mice were used to assess acute dermal toxicity, excision wound healing, and histological changes. The wounds were monitored, and the area of the wound was measured at the 0th, 4th, 8th, 12th, 16th, and 21st days as compared to the control animals. Results: The test extracts showed considerable protection and wound healing capabilities in acute dermal toxicity, excision wound, and histopathological studies. Among the tested extracts, the ethanolic extract showed the highest wound healing (46.68%, p<0.001), followed by the methanolic (38.50%, p<0.001), acetone (33.87%, p<0.05), aqueous (32.04%, p<0.001), chloroform (29.83%, p<0.05), and benzene extracts (28.60%, p<0.05) at the 21st d of wound healing. However, the samples gathered throughout the winter, summer, and rainy seasons did not show a statistically significant difference (p>0.05) in the present research work. Conclusion: This study helps to provide preliminary data on the concentration range of different extracts collected in different seasons. It is anticipated that this evaluation represents significant protective potential of indigenous flora for medicinal applications.
... A pesquisa possibilitou elencar 16 tipos de produtos ou coberturas associadas ou não à Terapia por Pressão Negativa (TPN) também citada como vácuo utilizadas no tratamento de lesões em recém-nascidos. O Quadro 2 descreve os tipos de curativos, sua classificação conforme, modelo proposto por Fan et al., (11) além de apresentar o número de estudos que relataram seu uso e os tipos de feridas tratadas na população neonatal. (11) . ...
... O Quadro 2 descreve os tipos de curativos, sua classificação conforme, modelo proposto por Fan et al., (11) além de apresentar o número de estudos que relataram seu uso e os tipos de feridas tratadas na população neonatal. (11) . Bahia, Brasil, 2023. ...
Article
Full-text available
Introduction: Given the particularities and challenges in the management and treatment of wounds in newborns, it becomes important for nurses to know, indicate and evaluate the efficacy and safety of dressings and products, even in the analysis of products for the neonatal population. Objective: To identify in the literature the main dressings and products used to treat wounds in newborns. Methodology: This is an integrative literature review, with articles published between 2018 and 2022, which addressed the topics of dressings and products used in the treatment of wounds in newborns. Data collection was carried out through consultation in Biblioteca Virtual de Saúde and in the Scientific Electronic Library Online. The “newborn”, “wounds” and “bandage” descriptors were used. The analysis was carried out according to the PICO strategy and structured following PRISMA. Results: Given the findings, it was possible to identify 13 articles that addressed products and dressings intended for treating wounds in newborns. Fourteen types of dressings/products and devices that use negative pressure therapy were found. Final considerations: The literature indicates the use of products and dressings with passive, active and biological principles for the treatment of wounds in newborns, including premature neonates, and it is worth highlighting the importance of considering the safe use of dressings that are suitable for this population.
... In general, primary care is based on cleaning and debridement to remove foreign bodies and devitalized tissue; reducing odor, pain, and inflammation; preventing microbial colonization and infection; and inducing epithelialization and granular tissue formation. Dressing is one of the most used strategies for the treatment of wounds and aims at favoring the wound healing process, as well as protecting against new external aggressions [38]. Data from the literature show that dressings incorporated with natural products, such as honey [39], and those obtained from plants [24] have the potential for treating wounds. ...
Article
Full-text available
Wounds of an acute or chronic etiology affect millions of people worldwide, with increasing prevalence every year. Microbial infections are one of the main causes that impair the wound healing process, and Staphylococcus aureus, a commensal member of the skin microbiota, is one of the main causative agents of wound infections. Crucially, a high proportion of these infections are caused by methicillin-resistant Staphylococcus aureus, which, in addition to β-lactams, has acquired resistance to almost all the antibacterial agents used to treat it, limiting therapeutic options. Studies on the antimicrobial and healing activities of extracts, essential oils, or metabolites obtained from native plants have been reported in many countries that have a diverse flora and traditions with the use of medicinal plants for the treatment of wound infections. Due to their great chemical diversity, plants have proven to be promising sources of bioactive molecules for the discovery and development of new drugs or strategies for the treatment of wounds. This review highlights the main herbal preparations that have antimicrobial and healing activities with potential for the treatment of wound infections caused by Staphylococcus aureus.
... However, management of PU should follow a comprehensive approach to include both the wound locally through proper non-operative wound care and surgery and systemically to incorporate pressure redistribution therapy, nutrition optimization, ulceration precautions, and patient education to manage the ulcer and prevent new ulcers or recurrence. The field of pressure ulcer wound healing has huge potentials through the use of topical products such as Cytokine growth factor therapy [3], and cell-based therapies and the improvements in pressure-relieving surface technology (e.g., fluid immersion technology) [4]. ...
... Acute wounds usually respond well to the available treatments; they have a good granulation and regeneration, and many of these wounds can be healed right away. However, chronic wound healing usually takes longer than three months, and many chronic wounds can be associated with other underlying conditions -diabetes, immunodeficiency, and/or vascular disease [6][7][8]. ...
Chapter
Currently, the therapeutic effect of appropriate wound dressings in the clinical management of wounds is widely accepted. Indeed, it is well established that no wound will heal if the factors that inhibit tissue repair in each stage are not addressed by selecting the appropriate dressing. To do this, clinicians need a good knowledge of the differences between products based on their physicochemical properties and their effective mechanism of action. However, traditional dressing classifications based on product ingredients or guided by TIME concepts are only partially applicable and there is clearly a need to classify dressings based on their function. To date no successful attempt has been made to implement this new classification. This article would like ti present a classification of wound dressings based on their functions.
Chapter
The wound care chapter is a comprehensive overview of the important concepts of wound healing, wound characterization, and wound care techniques. Additionally, an illustrated suturing guide is provided with step-by-step instructions.
Article
In this lecture, evidence is presented to support the following hypothesis regarding the roles of xanthine oxidase-derived oxidants and granulocytes in ischemia-reperfusion-induced microvascular injury. During the ischemic period, ATP is catabolized to yield hypoxanthine. The hypoxic stress also triggers the conversion of NAD-reducing xanthine dehydrogenase to the oxygen radical-producing xanthine oxidase. During reperfusion, molecular oxygen is reintroduced into the tissue where it reacts with hypoxanthine and xanthine oxidase to produce a burst of superoxide anion and hydrogen peroxide. In the presence of iron, superoxide anion and hydrogen peroxide react via the Haber-Weiss reaction to form hydroxyl radicals. This highly reactive and cytotoxic radical then initiates lipid peroxidation of cell membrane components and the subsequent release of substances that attract, activate, and promote the adherence of granulocytes to microvascular endothelium. The adherent granulocytes then cause further endothelial cell injury via the release of superoxide and various proteases.
Article
For more than a century, the practice of dressing a wound has had as one of its major rationales the prevention of infection. Dressings can theoretically protect the wound from gross microbial contamination and help to prevent the spread of infection to other patients on the surgical ward. In this setting, one thinks of dressings as a barrier between the wound and the outside world. It is a relatively novel idea that the composition and properties of the dressing itself can play a major role in modifying the wound microenvironment. In the span of a decade, three key observations have helped physicians think of dressings as pharmacologic agents. Odland1 first observed in 1958 that a blister healed faster if left unbroken. Then, in a 1962 landmark article, Winter2 showed that occlusion of wounds with a polyethylene film (Polythene) more than doubled wound epithelialization in domestic pigs. Finally, Hinman