Anda di halaman 1dari 8

Wound care: The role of advanced wound healing

technologies
Stephanie C. Wu, DPM, MD, PhD,a William Marston, MD,b and David G. Armstrong, DPM, MSc,c
North Chicago, Ill; Chapel Hill, NC; and Tucson, Ariz

Wound repair and regeneration is a highly complex combination of matrix destruction and reorganization. While major
hurdles remain, advances over the past generation have improved the clinician’s armamentarium in the medical and
surgical management of this problem. The purpose of this manuscript is to review the current literature regarding the
pragmatic use of three of the most commonly employed advanced therapies; namely, bioengineered tissue, negative
pressure wound therapy, and hyperbaric oxygen therapy with a focus on the near-term future of wound healing, including
stem cell therapy. ( J Vasc Surg 2010;52:59S-66S.)

Wound repair is an orchestra of highly integrated bio- BIOENGINEERED SKIN AND DERMAL
logical and molecular events of cell migration and prolifer- SUBSTITUTES OR EQUIVALENTS
ation, and of extracellular matrix deposition and remodel- Cell-based technologies to deliver exogenous
ing.1 Certain pathophysiologic and metabolic conditions growth factors to the wound bed. Autogenous and
can alter this normal course of events so that healing is non-autogenous skin grafts, commonly used for the heal-
impaired or delayed, resulting in chronic, nonhealing ing of skin ulcerations, may be associated with a number of
wounds.2,3 Diabetic foot ulcers (DFUs) readily become limitations such as risks of immune rejection, infection
chronic, and the factors that delay wound healing are transmission,6,7 and the creation of a donor site that is at
multiple and relate both to diabetes and to the effects of its risk of developing pain, scarring, infection, and/or delayed
complications.3 The costs associated with the healing of an healing.7 The need for a readily available, non-antigenic
ulcer have been noted to be as high as $45,000; however, tissue that possesses many of the histological and functional
these estimates do not include the deleterious effects on the characteristics of normal human skin spawned the evolu-
patient’s quality of life because of impaired mobility and tion of growing living human tissues for transplantation.
substantial loss of productivity.4 Clearly, the healing of Tissue engineering has revolutionized skin grafting from
ulcerations in a timely manner is of central importance in the initial autograft and allograft preparations to biosyn-
any plan for amputation prevention and limb preserva- thetic and tissue-engineered skin replacements and cell-
tion.5,6 The integration of technological advances with our based therapies. These advanced therapies include cultured
understanding of the complex cellular and biochemical autologous keratinocyte grafts, cultured allogeneic keratin-
mechanisms of wound healing has led to the development ocyte grafts, autologous/allogeneic composites, acellular
of various advanced wound healing modalities, such as collagen matrices, and cellular matrices. A number of prod-
bioengineered skin and tissue equivalents, negative pres- ucts are commercially available, and many others are in
sure wound therapy (NPWT), and hyperbaric oxygen ther- development. The ones that are currently available include
apy. This article examines the latest advances in wound human fibroblast-derived dermal substitute, human fibro-
healing using these three therapeutic categories and as- blast-derived temporary skin substitute, and allogeneic bi-
sesses their implications for clinical practice. layered cultured skin equivalent.
Human fibroblast-derived dermal substitute.
From the Rosalind Franklin University of Medicine and Science;a the
Human fibroblast-derived dermal substitute (Dermagraft,
Department of Surgery, Section of Vascular Surgery, University of North Advanced Biohealing Ltd, La Jolla, Calif) is composed of
Carolina School of Medicine;b and the Department of Surgery, Southern fibroblasts, extracellular matrix, and a bioabsorbable scaf-
Arizona Limb Salvage Alliance (SALSA), University of Arizona College of fold (Fig 1). It is produced by culturing human dermal
Medicine.c
Competition of interest: none.
fibroblast cells derived from newborn foreskin tissue onto a
This article is being co-published in the Journal of Vascular Surgery® and the bioabsorbable polyglactin mesh scaffold.8 As the fibroblasts
Journal of the American Podiatric Medical Association. proliferate to fill the interstices of this scaffold, they secrete
Reprint requests: Dr David G. Armstrong, Southern Arizona Limb Salvage human dermal collagen, matrix proteins, growth factors,
Alliance, University of Arizona College of Medicine, Department of
Surgery, Tucson, AZ 85724 (e-mail: armstrong@usa.net).
glycosaminoglycans, and cytokines to generate a three-
The editors and reviewers of this article have no relevant financial relationships dimensional, allogeneic, human dermal substitute contain-
to disclose per the JVS policy that requires reviewers to decline review of any ing metabolically active, living cells with a preferred, nearly
manuscript for which they may have a competition of interest. parallel alignment of the collagen fibers within human
0741-5214/$36.00
Copyright © 2010 by the Society for Vascular Surgery and the American
dermal substitute.8 The critical dependence of the thera-
Podiatric Medical Association. peutic properties of this living dermal implant on the recov-
doi:10.1016/j.jvs.2010.06.009 ery of protein synthesis, growth factor expression, and
59S
JOURNAL OF VASCULAR SURGERY
60S Wu et al September Supplement 2010

The efficacy of human fibroblast-derived dermal substi-


tute in the healing of full thickness chronic diabetic wounds
has been confirmed in many studies.11,12 A randomized,
controlled, multicenter trial evaluated 314 patients with
chronic diabetic foot ulcers for complete wound closure by 12
weeks and found that 30.0% (39/130) of human fibroblast-
derived dermal substitute patients healed compared with
18.3% (21/115) of control, wet-to-dry dressing patients
(P ⫽ .023).11 In addition, the group of patients who
received human fibroblast-derived dermal substitute expe-
rienced significantly fewer ulcer-related adverse events.11
Another prospective, multicenter, randomized controlled
12-week study that enrolled 28 patients with chronic dia-
betic ulcers found significantly more patients in the human
fibroblast-derived dermis group healed versus the control
group (71.4% vs 14.3%; P ⫽ .003).12 Moreover, the study
Fig 1. Cultured human dermis in place on neuropathic wound
noted that patients who healed in the human fibroblast-
following transmetatarsal amputation.
derived dermis group achieved wound closure significantly
faster than the control group (P ⫽ .004); the study group
also exhibited a statistically significant higher percent of
angiogenesis has been demonstrated.9 Unlike human skin, wound closure by week 12 as compared with patients in the
human fibroblast-derived dermal substitute does not con- control arm (P ⫽ .002).12 Interestingly, the percentage of
tain macrophages, lymphocytes, blood vessels, or hair fol- patients who experienced an infection involving their study
licles. Human fibroblast-derived dermal substitute has wound was lower in the human fibroblast-derived dermis
good resistance to tearing and is packaged with bovine group than in the control group.
serum and a saline-based cryoprotectant that contains 10% One potential problem that can arise from the applica-
dimethyl sulfoxide (DMSO). Cryopreservation helps main- tion of a human dermal substitute onto an allogeneic host is
tain cell viability and provides off-the-shelf availability. the initiation of an immune response leading to its rejec-
However, because the packing medium may also contain tion.11 Human fibroblast-derived dermis has passed exten-
traces of bovine serum, human fibroblast-derived dermal sive safety testing, and to date, there has been no reported
substitute is contraindicated in patients with known hyper- case of rejection in clinical use. This may be secondary to
sensitivity to bovine products. Human fibroblast- the inherent properties of human dermal substitute since it
derived dermal substitute is also contraindicated for use in is derived from neonatal human tissue that has undeveloped
ulcers that have signs of clinical infection or in ulcers with human leukocyte antigen tissue markers.11 In addition,
sinus tracts. dermal-derived fibroblasts are relatively non-antigenic, do not
Laboratory studies suggest this bio-engineered dermal express human leukocyte antigen-DR markers,9 and are there-
substitute promotes the healing of chronic ulcerations via fore, not expected to cause an immune reaction.
two principal modes of action.9 First, it provides living, Risk factors related to improved healing with hu-
human dermal fibroblasts that deposit matrix proteins and man fibroblast-derived dermal substitute. Data from a
facilitate angiogenesis.10 It also provides a preformed col- Phase III randomized trial were analyzed to find risk factors
lagen matrix, receptors, and bound growth factors that related to ulcer healing.3 Of interest, age, race, diabetes
facilitate the migration and colonization of the host’s epi- type, ulcer duration prior to enrollment, and hours of
thelial cells, which promote wound closure.9 Previous stud- weight-bearing were not associated with healing rate. Ini-
ies have shown human fibroblast-derived dermal substitute tial ulcer area, gender, a history of ulcer infection, and
to be most effective in treating ulcers of greater than 6 change in hemoglobin A1c (Hgb A1c) were associated with
weeks duration.11 It may be possible that chronic ulcers are altered healing rates.
deficient in many of the factors necessary for healing and are An initial ulcer area greater than 2 cm2 was associated
most likely to benefit from human fibroblast-derived der- with a 1.5 times greater incidence of wound closure (P ⫽
mal substitute treatment. .02). Females were 2.0 times more likely to heal than males
Human fibroblast-derived dermal substitute is de- (P ⫽ .009). An episode of infection during the 12 weeks of
signed to assist in restoration of the dermal bed in an ulcer treatment was associated with a 3.4 times increased risk of
to improve the wound healing process and allow the pa- non-closure (P ⬍ .01).
tient’s own epithelial cells to migrate and close the wound. In another study, Browne et al reported on the impor-
It is approved by the United States Food and Drug Admin- tance of controlling bacterial load prior to the use of human
istration for the treatment of full-thickness, chronic (dura- fibroblast-derived dermal substitute. The authors found an
tion ⬎6 weeks) diabetic foot ulcers extending through the association between the bacterial load and healing rate both
dermis, but not involving tendon, muscle, joint capsule, or before and after human fibroblast-derived dermal substi-
bone. tute application. They recommended treatment of patients
JOURNAL OF VASCULAR SURGERY
Volume 52, Number 12S Wu et al 61S

human skin cells.5 The lower dermal lattice combines bo-


vine type 1 collagen and cultured human neonatal dermal
fibroblasts,15 which produce additional matrix proteins and
organize the provided structural proteins. The upper epi-
dermal layer is formed by promoting human epidermal
keratinocytes to first multiply and then to differentiate to
replicate the architecture of the human epidermis5 and act
as a barrier to prevent water loss and infection.16 Allogeneic
bi-layered cultured skin equivalent has been shown to
produce all cytokines and growth factors that are produced
by the normal skin during the healing process.5 Unlike
Fig 2. Analysis of healing based on Hemoglobin A1C levels in
human skin, allogeneic bi-layered cultured skin equivalent
study of bioengineered tissue. does not contain melanocytes, Langerhans’ cells, macro-
phages, lymphocytes, or other structures such as blood
vessels, hair follicles, or sweat glands.
with combination antibiotics until the bacterial burden was Although the precise mode of action is not clear, allo-
reduced to less than106 colony forming units/gram prior geneic bi-layered cultured skin equivalent has been quoted
to the application of living skin substitutes or growth to behave similarly to a partial thickness autograft in that it
factors.13 provides immediate wound coverage15 and interacts with
The initial and terminal Hgb A1c levels did not inde- adjacent tissue after implantation.17 Similarities between
pendently relate to wound closure. To better determine the dermal layers of the host and graft may facilitate the cross-
relationship between glucose control and wound healing, over activity of mediators and response to signals from both
Marston and colleagues calculated the change in Hgb A1c the host and graft dermal cells.18 Allogeneic bi-layered
between initial and 12-week levels. Patients with a decrease cultured skin equivalent has also been noted to act through
in this value were expected to have better glucose control at least three modes of healing: secondary intention, persis-
on average than those with an increasing level. In the tent wound closure with underlying healing, and frank graft
control group, there was no significant difference in the rate take.15,16 It is believed that the allogeneic bi-layered cul-
of healing in those with improving compared with worsen- tured skin equivalent acts by both filling in the wound with
ing Hgb A1c. Further, the number healed in the human extracellular matrix and by inducing the expression and
fibroblast-derived dermal substitute treated group with production of numerous growth factors and mediators
worsening Hgb A1c was similar to the control rates such as interleukin transforming growth factors (TGF-␤),
(20.2%). However, the number of subjects that healed in granulocyte-macrophage colony-stimulating factor, platelet-
the human fibroblast-derived dermal substitute group with derived growth factor, basic fibroblast growth factor, and
an improving Hgb A1c (46.7%) was significantly higher cytokines that contribute to wound healing by the stimu-
than in the other three groups (P ⫽ .008).3 These data are lation of wound contraction and epithelization.5,16,17,19 A
detailed in Fig 2. Allenet et al assessed the cost-effectiveness study investigating the effects of dermal replacement ther-
of human fibroblast-derived dermis in the treatment of the apy on blood flow at the base of diabetic foot ulcers noted
diabetic foot ulcer compared with standard treatment.14 that blood flow increased by an average of 72% in the base
The authors developed a Markov model to simulate, over a of five of the seven ulcers studied.20 The changes in blood
52-week period, the health status of a cohort of 100 pa- flow observed may reflect angiogenesis in the newly formed
tients with a diabetic foot ulcer treated either with conven- granulation tissue and/or vasodilatation of existing vessels,
tional therapy or with human fibroblast-derived dermis.14 processes that are possibly enhanced by the allogeneic
They concluded that because human fibroblast-derived bi-layered cultured skin equivalent application.20
dermis heals more ulcers within 52 weeks, the average cost Allogeneic bi-layered cultured skin equivalent is pack-
per healed ulcer is lower (53,522 French francs [FF] vs aged in a sealed heavy-gauge polyethylene bag with a 10%
56,687 FF for standard treatment).14 The incremental cost- CO2/air atmosphere and an agarose nutrient medium. It is
effectiveness of human fibroblast-derived dermis equals contraindicated in patients with known allergies to bovine
38,784 FF, indicating the extra investment that the decision- collagen or hypersensitivity to the components of the ship-
maker has to accept for an additional ulcer healed with human ping medium. One of the previous inconveniences with
fibroblast-derived dermis compared with conventional allogeneic bi-layered cultured skin equivalent was its need
treatment.14 to be shipped overnight based on date of patient applica-
Allogeneic bi-layered cultured skin equivalent. tion. The newly improved shipping box with better insula-
Allogeneic bi-layered cultured skin equivalent (Apligraf; tion and temperature maintenance now affords allogeneic
Organogenesis Inc, Canton, Mass) is a living, biological bi-layered cultured skin equivalent a 10-day shelf life and
dressing developed from neonatal foreskin and consists of the clinician some flexibility on the date of application.
living cells and structural proteins. Like human skin, this Antiseptic agents such as Dakin’s solution, mafenide ace-
allogeneic bi-layered cultured skin equivalent has both an tate, scarlet red dressing, tincoban, zinc sulfate, povidone-
upper epidermal and a lower dermal layer and contains iodine solution, and chlorhexidine have been determined
JOURNAL OF VASCULAR SURGERY
62S Wu et al September Supplement 2010

to be cytotoxic to allogeneic bi-layered cultured skin equiv-


alent and should not be used immediately prior to its
application.
Apligraf is the first bi-layered living skin equivalent
approved in the United States for the treatment of chronic
venous and full-thickness neuropathic diabetic foot ulcers
of greater than three weeks’ duration that extend through
the dermis but without tendon, muscle, capsule, or bone
exposure.21
In a large, multicenter, randomized, prospective clini-
cal trial, allogeneic bi-layered cultured skin equivalent was
shown to heal non-infected, non-ischemic chronic plantar
diabetic foot ulcers faster and in more patients than con-
ventional therapy.5 By 12 weeks of treatment, 56% (63/
112) of chronic diabetic foot ulcers treated with allogeneic
bi-layered cultured skin equivalent were 100% closed, com-
pared with 39% (36/96) of ulcers treated with conven-
tional therapy (debridement plus saline dressings alone and
total off-loading [P ⫽ .0026]).5 The median time to
wound closure was 65 days for diabetic foot ulcers treated
with allogeneic bi-layered cultured skin equivalent versus
90 days for ulcers treated with conventional therapy (P ⫽
.0026).5 A similar study conducted in the European Union
and Australia reported comparable results.22 The incidence
of adverse reactions was similar between the two groups,
with the exception of osteomyelitis and lower-limb ampu-
tations, both of which were noted to be less frequent in the Fig 3. Negative pressure wound therapy foam in place with con-
tinuous streaming infusion (⬙ChemoVAC⬙) modification.
allogeneic bi-layered cultured skin equivalent group.5,23 Allo-
geneic bi-layered cultured skin equivalent is well-tolerated,
appears to be immunologically inert, with no clinical evidence patient groups with chronic wounds, health care providers
of rejection by any patient.18,24 and coverage decision makers should take not only the high
Because the allogeneic bi-layered cultured skin is made cost of the biotechnology product but the total cost of care
up of viable human cells, it cannot be terminally steril- into account when deciding about the appropriate alloca-
ized.25 Safety concerns, which have been addressed, in- tion of their financial resources.28
clude the risk of possible transmission of infection, immu- Negative pressure wound therapy. Since its intro-
nogenicity, immunological graft rejection, and tumor duction in the United States in 1997, negative pressure
formation.25 The maternal blood of the neonatal donor wound therapy (NPWT) or vacuum-assisted closure has
and the working cell banks are thoroughly screened for emerged as a commonly employed option in the treatment
infectious agents, pathogens, and other contaminants.25 of complex wounds (Fig 3). The incorporation of NPWT
Allogeneic bi-layered cultured skin equivalents are of into wound treatment regimens has been advocated by
considerable cost and should therefore be reserved for many clinicians, and NPWT has been noted to help de-
chronic foot ulcers that have failed to respond to the crease the number of dressing changes, reduce the time
currently available standard care.26 Redekop et al found between debridement and definitive closure, and reduce
that treatment with allogeneic bi-layered cultured skin costs associated with a protracted course of hospital
equivalent plus good wound care resulted in a 12% reduc- stay.29,30
tion in costs over the first year of treatment compared with Armstrong and colleagues evaluated the efficacy of
good wound care alone.27 This benefit was realized after 5 NPWT to heal 31 indolent diabetic foot wounds immedi-
months, the crossover point of the two cost curves. Alloge- ately after wide surgical debridement. A cessation of ther-
neic bi-layered cultured skin equivalent use increased the apy protocol was utilized where NPWT was discontinued
amount of ulcer-free time by 1.53 months (7.78 vs 6.25), when the wound bed approached 100% coverage with
reduced the risk of amputation (6.3% vs 17.1%),27 and granulation tissue and no exposed tendon, joint capsule, or
subsequently was cost-effective in the long-term. Langer bone. They noted that 90.3% of the wounds treated with
and Rogowski assessed the cost-effectiveness of growth NPWT healed at the level of debridement without the need
factors and tissue-engineered artificial skin for treating for further bony resection in a mean 8.1 ⫾ 5.5 weeks.30 In
chronic wounds based on a review of 11 qualifying eco- a randomized trial, Eginton et al compared the wound
nomic evaluations.28 The authors noted that, although healing efficacy of NPWT and conventional moist dressings
some growth factors and tissue-engineered artificial skin to treat large diabetic foot ulcers, and found NPWT de-
products had favorable cost-effectiveness ratios in selected creased wound volume and depth significantly more than
JOURNAL OF VASCULAR SURGERY
Volume 52, Number 12S Wu et al 63S

moist gauze dressings (59% vs 0% and 49% vs 8%, respec- plex wounds. Abidia and colleagues randomly assigned 18
tively).31 patients with ischemic non-healing diabetic limb ulcers to
Recently published trials have further demonstrated the 100% oxygen or air at 2.4 atmospheres for 90 minutes in a
wound-healing efficacy of NPWT. Blume and coworkers hyperbaric chamber daily.37 In this double-blind study,
compared the safety and clinical efficacy of NPWT with oxygen and control groups received 30 sessions after which
advanced moist wound therapy (AMWT) to treat diabetic the outcome was measured. In the oxygen group, five of
foot ulcers in a multicenter, randomized, controlled trial. eight ulcers were closed completely, compared to one of
They enrolled 342 subjects, 79% male, with a mean age of the eight control ulcers (P ⫽ .027).
58 years, who were randomized to receive either NPWT or In another prospective study of diabetic foot ulcers,
AMWT and standard off-loading therapy as needed.32 The Kalani et al enrolled 38 patients with non-healing foot
authors noted that a greater proportion of foot ulcers ulcers and basal transcutaneous oxygen levels in the foot
achieved complete ulcer closure with NPWT (73/169; below 40 mm Hg.38 Seventeen patients were treated with
43.2%) than AMWT (48/166; 28.9%) within the 112-day HBO for 40 to 60 sessions, and 21 were treated with
active treatment phase (P ⫽ .007). The Kaplan-Meier standard diabetic ulcer care. Patients were followed for 3
median estimate for 100% ulcer closure was 96 days (95% years. At final follow-up, 76% of patients treated with HBO
confidence interval: 75.0, 114.0) for NPWT and not deter- had healed their DFU, and only 12% required limb ampu-
minable for AMWT (P ⫽ .001). The authors noted no tation. In the standard care group, 48% had healed their
significant difference between the groups in treatment- foot ulcer at 3 years, and 33% required limb amputation.
related complications at 6 months. In 2004, the Cochrane Collaborative reviewed hyper-
Similarly, a 16-week, 18-center, randomized clinical baric oxygen therapy for chronic wounds and concluded
trial conducted by Armstrong and Lavery, involving 162 that HBO reduces the risk of amputation for patients with
diabetic patients with larger and more complex wounds DFU and increases the chance of healing at 1 year.39
than those from previous randomized trials, found that However, it was noted that these recommendations were
NPWT healed more wounds after partial foot amputation based on small, underpowered studies and that further
versus the standard of care (43 [56%] vs 33 [39%]; P ⫽ randomized studies were greatly needed to clarify the ben-
.040). The authors noted that NPWT produced faster efits of this costly therapy. Addressing some of the method-
wound-healing rates (P ⫽ .005) and faster granulation ologic concerns of previous studies, a rather robust double-
tissue formation rates, versus standard of care, based on the blind randomized controlled study conducted by Londahl
time needed to complete closure (P ⫽ .002).33 Resource and coworkers42 suggested improved healing in the active
utilization for patients treated with NPWT also was evalu- HBO group compared with placebo at 1 year in patients
ated in this same study population. Apelqvist34 and co- suffering from complex diabetic foot wounds (52% vs 29%;
workers reported that patients randomized to the NPWT P ⫽ .03). This study used an intent-to-treat analysis for
group required fewer surgical procedures (including de- patients with chronic (⬎3 month duration) neuropathic
bridement) than the control group (43 vs 120; P ⬍ .001), and neuroischemic wounds. All received treatment in a
fewer average number of dressing changes (41 [range, multiplace chamber receiving either active HBO or placebo
6-140] for NPWT versus 118.0 [range, 12-226] for con- (compressed air).
trol MWT [P ⬍ .0001]), and fewer outpatient treatment Further work has been conducted to identify better
visits (4 [range, 0-47] in the NPWT versus 11 [range, decision-making parameters for the use of HBO. Works
0-106] for control, [P ⬍ .05]). This yielded a cost savings have focused on the use of transcutaneous oxygen tension
in excess of $12,800 compared with standard therapy. (TcPO2) as a predictive modality in selecting patients for
Combined with the clinical data, these analyses provide HBO treatment. Specifically, Grolman et al measured
compelling evidence that appropriate use of NPWT is TcPO2 in the ischemic limb of 36 patients breathing room
efficacious and cost-effective in achieving healing of prop- air, followed by 100% O2. They found an increase of
erly selected wounds, both on an inpatient and outpatient TcPO2 in the ischemic foot of ⬎10 torr was associated
basis. with a healing rate of 70%, compared with a healing rate of
Hyperbaric oxygen therapy. Hyperbaric oxygen 11% in those with an increase of ⬍10 torr.40 In a larger
(HBO) has long been considered as a potential treatment study of ischemic and non-ischemic DFUs, Fife and col-
modality for complicated or recalcitrant ulcers. Oxygen has leagues evaluated the use of TcPO2 in 1144 patients. In
been reported to stimulate angiogenesis, enhance fibroblast this study, predictive values were relatively poor, with some
and leukocyte function, and normalize cutaneous micro- patients healing despite low initial and in-chamber
vascular reflexes.35,36 Clinically, HBO has been demon- TcPO2.41
strated to improve transcutaneous pO2 in the limbs of While data, particularly recently, have pointed toward
some patients with ischemic ulcers. Significant side ef- HBO therapy as having some degree of potential benefit, its
fects of treatment are uncommon, but may be severe, cost is high. Patients often travel long distances for daily
including barotraumatic otitis, hyperoxic seizures, and treatments at great cost to themselves and their families.
pneumothorax. Although reported protocols for treatment of ischemic
For the treatment of DFU, one randomized study was limb ulcers vary significantly, most involve a total cost of
particularly influential in supporting use of HBO for com- $15,000 to $40,000. In an assessment for the Canadian
JOURNAL OF VASCULAR SURGERY
64S Wu et al September Supplement 2010

government, Chuck et al from the University of Alberta


compared HBO and standard care for DFUs using a deci-
sion model and available outcomes data. They found that
the average yearly cost for HBO treated ulcers was $40,695
(Canadian) compared with $49,786 (Canadian) for stan-
dard care. They concluded that adjunctive HBO is cost-
effective compared with standard care alone.43
Currently, HBO is an option that may improve limb
preservation in a limited group of patients with complicated
ischemic and diabetic limb ulcers. Other HBO indications
reimbursed by most insurers include the treatment of inha-
lation injury, necrotizing wound infections, crush injury,
and osteoradionecrosis. Until larger randomized studies
are performed, it cannot be recommended as a primary
treatment for patients with uncomplicated diabetic or isch-
emic ulcers. But in selected more complicated cases, HBO
may have a role.

WOUND TREATMENTS ON THE HORIZON


Stem cell therapy. Marrow cells have been shown to
play an important role in the healing of cutaneous wounds.
Subsequent to dermal wounding, marrow-derived mesen-
chymal stem cells (MSC) are mobilized into the peripheral
circulation and engraft near adnexal structures in the skin.
These cells eventually differentiate into skin cells.44
Deng et al transplanted fluorescent-labeled bone mar-
row MSC into lethally irradiated mice and found that
labeled cells gave rise to stem cells and committed cells in
the skin.45 Another study found that 15% to 20% of the
dermal fibroblasts originated from the bone marrow in
a murine wound model.46 Animal studies have shown
wound healing significantly improved after injection of
Fig 4. Harvest of (A) and seeding (B) marrow-derived stem cells
MSC into the wound.47 Bauer et al found that although
on acellular matrix.
extremity ischemia is a powerful stimulant for marrow stem
cell recruitment, fewer progenitor cells were able to migrate
to the ischemic wound.48 This may be a result of macro- progenitor cells that can engraft into wound and accelerate
and microvascular disease, obstructing vascular conduits healing.
for mobilization of MSC. Local application, topically or via Other critical considerations. Despite the plethora
injection, of MSC would place these progenitor cells at the of advances, results from a myriad of published and unpub-
site of injury, assisting in homing and delivery. Human lished industry-sponsored, randomized trials that evaluated
studies with recent small case-series transplanting autolo- the efficacy of these advanced wound healing agents have
gous cultured marrow derived MSC and pure marrow been less than ideal and are difficult to place in perspec-
aspirate (Fig 4) have shown rapid improvement in granula- tive.5,51-53 For example, published healing rates for DFUs
tion and healing in chronic wounds.44,49,50 treated by TCCs (total contact casts) are noted to be 80% to
Badiavas and Falanga44 harvested bone marrow aspi- 90% compared with only 45% to 55% for biologic tis-
rate from the ileum in three patients with chronic wounds sues.54,55 Most studies either offered shoes or sandals to
unresponsive to aggressive therapy (abdominal dehiscence, study participants or allowed individual centers to select the
lower extremity arterial ulcer, and lower extremity venous type of pressure relief;55 however, not one of these studies
ulcer) and injected the marrow aspirate into the peri- employed irremovable offloading.
wound tissue and applied the aspirate topically. The re- Such considerations emphasize the importance of using
maining aspirate was cultured and expanded in vitro and advanced wound healing modalities as adjuvant therapies
later applied topically to the wounds in a repeat treatment. that work synergistically with standard wound care regi-
The authors noted that, after each treatment with cultured mens such as routine debridement, pressure mitigation,
MSC, the wounds experienced a burst of granulation tis- infection control, and provision of adequate vascularity to
sue. Two patients healed rapidly by secondary intention; the affected area. Without adhering to these important
one patient healed with the use of a skin substitute (cul- principles, the addition of any adjunctive modality is un-
tured fetal foreskin). They concluded that their study sup- likely to result in improved healing rates.1,13 Wound bed
ported the hypothesis that bone marrow aspirate contained preparation encompasses the removal of necrotic tissue,
JOURNAL OF VASCULAR SURGERY
Volume 52, Number 12S Wu et al 65S

formation of granulation tissue, and elimination of wound 19. Brem H, Balledux J, Bloom T, Kerstein MD, Hollier L. Healing of
exudate.56 Brem et al found that two separate clinical and diabetic foot ulcers and pressure ulcers with human skin equivalent: a
new paradigm in wound healing. Arch Surg 2000;135:627-34.
specialty sites were able to achieve a frequency of complete
20. Newton DJ, Khan F, Belch JJ, Mitchell MR, Leese GP. Blood flow
wound closure of greater than 70% within 6 months via changes in diabetic foot ulcers treated with dermal replacement therapy.
optimal wound preparation.56 Care for a patient with dia- J Foot Ankle Surg 2002;41:233-7.
betic foot ulceration is complex, necessitating collaboration 21. Introduction. Healing chronic wounds: technologic solutions for today
of a multidisciplinary team to achieve treatment goals; such and tomorrow. Adv Skin Wound Care 2000;13(2 Suppl):4-5.
22. Edmonds M; European and Australian Apligraf Diabetic Foot Ulcer
a team must accurately assess and manage current wound
Study Group. Apligraf in the treatment of neuropathic diabetic foot
and patient conditions, optimally manage subsequent com- ulcers. Int J Low Extrem Wounds 2009;8:11-8.
plications, and aim to promote shorter healing time. Ad- 23. Steinberg JS, Edmonds M, Hurley DP Jr, King WN. Confirmatory data
vanced wound healing modalities, when used appropri- from EU study supports Apligraf for the treatment of neuropathic
ately, may serve as useful components of the wound diabetic foot ulcers. J Am Podiatr Med Assoc 2010;100:73-7.
24. Molecule of the month: Apligraf living human skin equivalent. Drug
management algorithm.
News Perspect 1998;11:43.
25. Dolynchuk K, Hull P, Guenther L, Sibbald RG, Brassard A, Cooling M,
et al. The role of Apligraf in the treatment of venous leg ulcers. Ostomy
REFERENCES
Wound Manage 1999;45:34-43.
1. Falanga V. Classifications for wound bed preparation and stimulation of 26. Fagrell B, Jorneskog G, Intaglietta M. Disturbed microvascular reactiv-
chronic wounds. Wound Repair Regen 2000;8:347-52. ity and shunting–a major cause for diabetic complications. Vasc Med
2. Mustoe TA, O’Shaughnessy K, Kloeters O. Chronic wound pathogen- 1999;4:125-7.
esis and current treatment strategies: a unifying hypothesis. Plast Re- 27. Redekop WK, McDonnell J, Verboom P, Lovas K, Kalo Z. The cost
constr Surg 2006;117(7 Suppl):35S-41S. effectiveness of Apligraf treatment of diabetic foot ulcers. Pharmaco-
3. Marston WA; Dermagraft Diabetic Foot Ulcer Study Group. Risk economics 2003;21:1171-83.
factors associated with healing chronic diabetic foot ulcers: the impor- 28. Langer A, Rogowski W. Systematic review of economic evaluations of
tance of hyperglycemia. Ostomy Wound Manage 2006;52:26-8, 30, 32 human cell-derived wound care products for the treatment of venous
passim. leg and diabetic foot ulcers. BMC Health Serv Res 2009;9:115.
4. Paquette D, Falanga V. Leg ulcers. Clin Geriatr Med 2002;18:77-88, vi. 29. McCallon SK, Knight CA, Valiulus JP, Cunningham MW, McCulloch
5. Veves A, Falanga V, Armstrong DG, Sabolinski ML; Apligraf Diabetic JM, Farinas LP. Vacuum-assisted closure versus saline-moistened gauze
Foot Ulcer Study. Graftskin, a human skin equivalent, is effective in the in the healing of postoperative diabetic foot wounds. Ostomy Wound
management of noninfected neuropathic diabetic foot ulcers: a prospec- Manage 2000;46:28-32, 34.
tive randomized multicenter clinical trial. Diabetes Care 2001;24: 30. Armstrong DG, Lavery LA, Abu-Rumman P, Espensen EH, Vazquez
290-5. JR, Nixon BP, et al. Outcomes of subatmospheric pressure dressing
6. Lee KH. Tissue-engineered human living skin substitutes: development therapy on wounds of the diabetic foot. Ostomy Wound Manage
and clinical application. Yonsei Med J 2000;41:774-9. 2002;48:64-8.
7. Bello YM, Falabella AF, Eaglstein WH. Tissue-engineered skin. Current 31. Eginton MT, Brown KR, Seabrook GR, Towne JB, Cambria RA. A
status in wound healing. Am J Clin Dermatol 2001;2:305-13.
prospective randomized evaluation of negative-pressure wound dress-
8. Marston WA. Dermagraft, a bioengineered human dermal equivalent
ings for diabetic foot wounds. Ann Vasc Surg 2003;17:645-9.
for the treatment of chronic nonhealing diabetic foot ulcer. Expert Rev
32. Blume PA, Walters J, Payne W, Ayala J, Lantis J. Comparison of
Med Devices 2004;1:21-31.
negative pressure wound therapy using vacuum-assisted closure with
9. Mansbridge J, Liu K, Patch R, Symons K, Pinney E. Three-dimensional
advanced moist wound therapy in the treatment of diabetic foot ulcers:
fibroblast culture implant for the treatment of diabetic foot ulcers:
a multicenter randomized controlled trial. Diabetes Care 2008;31:
metabolic activity and therapeutic range. Tissue Eng 1998;4:403-14.
631-6.
10. Mansbridge J. Skin substitutes to enhance wound healing. Expert Opin
33. Armstrong DG, Lavery LA. Negative pressure wound therapy after
Investig Drugs 1998;7:803-9.
partial diabetic foot amputation: a multicentre, randomised controlled
11. Marston WA, Hanft J, Norwood P, Pollak R; Dermagraft Diabetic Foot
trial. Lancet 2005;366:1704-10.
Ulcer Study Group. The efficacy and safety of Dermagraft in improving
34. Apelqvist J, Armstrong DG, Lavery LA, Boulton AJM. Resource utili-
the healing of chronic diabetic foot ulcers: results of a prospective
randomized trial. Diabetes Care 2003;26:1701-5. zation and economic costs of care based on a randomized trial of
12. Hanft JR, Surprenant MS. Healing of chronic foot ulcers in diabetic vacuum-assisted closure therapy in the treatment of diabetic foot
patients treated with a human fibroblast-derived dermis. J Foot Ankle wounds. Am J Surg 2008;195:782-8.
Surg 2002;41:291-9. 35. Kuffler DP. Hyperbaric oxygen therapy: an overview. J Wound Care
13. Browne AC, Vearncombe M, Sibbald RG. High bacterial load in 2010;19:77-9.
asymptomatic diabetic patients with neurotrophic ulcers retards wound 36. Goldman RJ. Hyperbaric oxygen therapy for wound healing and limb
healing after application of Dermagraft. Ostomy Wound Manage 2001; salvage: a systematic review. PM R 2009;1:471-89.
47:44-9. 37. Abidia A, Laden G, Kuhan G, Johnson BF, Wilkinson AR, Renwick PM,
14. Allenet B, Parée F, Lebrun T, Carr L, Posnett J, Martini J, et al. et al. The role of hyperbaric oxygen therapy in ischaemic diabetic lower
Cost-effectiveness modeling of Dermagraft for the treatment of diabetic extremity ulcers: a double-blind randomised-controlled trial. Eur J Vasc
foot ulcers in the French context. Diabetes Metab 2000;26:125-32. Endovasc Surg 2003;25:513-8.
15. Zaulyanov L, Kirsner RS. A review of a bi-layered living cell treatment 38. Kalani M, Jorneskog G, Naderi N, Lind F, Brismar K. Hyperbaric
(Apligraf) in the treatment of venous leg ulcers and diabetic foot ulcers. oxygen (HBO) therapy in treatment of diabetic foot ulcers. Long-term
Clin Interv Aging 2007;2:93-8. follow-up. J Diabetes Complications 2002;16:153-8.
16. De SK, Reis ED, Kerstein MD. Wound treatment with human skin 39. Kranke P, Bennett M, Roeckl-Wiedmann I, Debus S. Hyperbaric
equivalent. J Am Podiatr Med Assoc 2002;92(1):19-23. oxygen therapy for chronic wounds. Cochrane Database Syst Rev
17. Trent JF, Kirsner RS. Tissue engineered skin: Apligraf, a bi-layered 2004;(2):CD004123.
living skin equivalent. Int J Clin Pract 1998;52:408-13. 40. Grolman RE, Wilkerson DK, Taylor J, Allinson P, Zatina MA. Trans-
18. Donohue KG, Carson P, Iriondo M, Zhou L, Saap L, Gibson K, et al. cutaneous oxygen measurements predict a beneficial response to hyper-
Safety and efficacy of a bilayered skin construct in full-thickness surgical baric oxygen therapy in patients with nonhealing wounds and critical
wounds. J Dermatol 2005;32:626-31. limb ischemia. Am Surg 2001;67:1072-9; discussion 1080.
JOURNAL OF VASCULAR SURGERY
66S Wu et al September Supplement 2010

41. Fife CE, Buyukcakir C, Otto GH, Sheffield PJ, Warriner RA, Love TL, 49. Badiavas EV, Ford D, Liu P, Kouttab N, Morgan J, Richards A, et al.
et al. The predictive value of transcutaneous oxygen tension measure- Long-term bone marrow culture and its clinical potential in chronic
ment in diabetic lower extremity ulcers treated with hyperbaric oxygen wound healing. Wound Repair Regen 2007;15:856-65.
therapy: a retrospective analysis of 1,144 patients. Wound Repair Regen 50. Falanga V, Iwamoto S, Chartier M, Yufit T, Butmarc J, Kouttab N, et al.
2002;10:198-207. Autologous bone marrow-derived cultured mesenchymal stem cells
42. Londahl M, Katzman P, Nilsson A, Hammarlund C. Hyperbaric oxy- delivered in a fibrin spray accelerate healing in murine and human
gen therapy facilitates healing of chronic foot ulcers in patients with cutaneous wounds. Tissue Eng 2007;13:1299-312.
diabetes. Diabetes Care 2010;33:998-1003. 51. Steed DL. Clinical evaluation of recombinant human platelet-derived
43. Chuck AW, Hailey D, Jacobs P, Perry DC. Cost-effectiveness and growth factor for the treatment of lower extremity diabetic ulcers.
budget impact of adjunctive hyperbaric oxygen therapy for diabetic foot Diabetic Ulcer Study Group. J Vasc Surg 1995;21:71-8.
ulcers. Int J Technol Assess Health Care 2008;24:178-83.
52. Veves A, Sheehan P, Pham HT. A randomized, controlled trial of
44. Badiavas EV, Falanga V. Treatment of chronic wounds with bone
Promogran (a collagen/oxidized regenerated cellulose dressing) vs
marrow-derived cells. Arch Dermatol 2003;139:510-6.
standard treatment in the management of diabetic foot ulcers. Arch
45. Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell ED.
Surg 2002;137:822-7.
Mesenchymal stem cells spontaneously express neural proteins in
53. Gentzkow GD, Iwasaki SD, Hershon KS. Use of Dermagraft, a cultured
culture and are neurogenic after transplantation. Stem Cells 2006;24:
1054-64. human dermis, to treat diabetic foot ulcers. Diabetes Care 1996;19:
46. Fathke C, Wilson L, Hutter J, Kapoor V, Smith A, Hocking A, et al. 350-4.
Contribution of bone marrow-derived cells to skin: collagen deposition 54. Cavanagh PR, Lipsky BA, Bradbury AW, Botek G. Treatment for
and wound repair. Stem Cells 2004;22:812-22. diabetic foot ulcers. Lancet 2005;366:1725-35.
47. McFarlin K, Gao X, Liu YB, Dulchavsky DS, Kwon D, Arbab AS, et al. 55. Armstrong DG, Boulton AJ. Pressure offloading and “advanced”
Bone marrow-derived mesenchymal stromal cells accelerate wound wound healing: isn’t it finally time for an arranged marriage? Int J Low
healing in the rat. Wound Repair Regen 2006;14:471-8. Extrem Wounds 2004;3:184-7.
48. Bauer SM, Goldstein LJ, Bauer RJ, Chen H, Putt M, Velazquez OC. 56. Brem H, Balledux J, Sukkarieh T, Carson P, Falanga V. Healing of
The bone marrow-derived endothelial progenitor cell response is im- venous ulcers of long duration with a bilayered living skin substitute:
paired in delayed wound healing from ischemia. J Vasc Surg 2006;43: results from a general surgery and dermatology department. Dermatol
134-41. Surg 2001;27:915-9.

Anda mungkin juga menyukai