You are on page 1of 32

Advanced Drug Delivery Reviews 123 (2018) 33–64

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews

journal homepage: www.elsevier.com/locate/addr

Nanomedicine and advanced technologies for burns: Preventing


infection and facilitating wound healing
Mirza Ali Mofazzal Jahromi a,b,1, Parham Sahandi Zangabad c,d,e,l,1, Seyed Masoud Moosavi Basri e,f,g,
Keyvan Sahandi Zangabad e,h,i, Ameneh Ghamarypour e,j, Amir R. Aref k,
Mahdi Karimi d,l,m,n,⁎, Michael R. Hamblin n,o,p,⁎⁎
a
Department of Advanced Medical Sciences & Technologies, School of Medicine, Jahrom University of Medical Sciences (JUMS), Jahrom, Iran
b
Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences (JUMS), Jahrom, Iran
c
Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Science (TUOMS), Tabriz, Iran
d
Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
e
Bio-Nano-Interfaces: Convergence of Sciences (BNICS), Universal Scientific Education and Research Network (USERN), Tehran, Iran
f
Bioenvironmental Research Center, Sharif University of Technology, Tehran, Iran
g
Civil & Environmental Engineering Department, Shahid Beheshti University, Tehran, Iran
h
Department of Polymer Engineering, Sahand University of Technology, PO Box 51335-1996, Tabriz, Iran
i
Nanomedicine Research Association (NRA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
j
Department of Biology, Science and Research Branch, Islamic Azad university, Tehran, Iran
k
Department of Medical Oncology, Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
l
Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
m
Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
n
Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
o
Department of Dermatology, Harvard Medical School, Boston, USA
p
Harvard-MIT Division of Health Sciences and Technology, Cambridge, USA

a r t i c l e i n f o a b s t r a c t

Article history: According to the latest report from the World Health Organization, an estimated 265,000 deaths still occur every
Received 24 March 2017 year as a direct result of burn injuries. A widespread range of these deaths induced by burn wound happens in
Received in revised form 20 July 2017 low- and middle-income countries, where survivors face a lifetime of morbidity. Most of the deaths occur due
Accepted 1 August 2017
to infections when a high percentage of the external regions of the body area is affected. Microbial nutrient avail-
Available online 4 August 2017
ability, skin barrier disruption, and vascular supply destruction in burn injuries as well as systemic immunosup-
Keywords:
pression are important parameters that cause burns to be susceptible to infections. Topical antimicrobials and
Burn wound infection dressings are generally employed to inhibit burn infections followed by a burn wound therapy, because systemic
Wound healing antibiotics have problems in reaching the infected site, coupled with increasing microbial drug resistance. Nano-
Topical treatment technology has provided a range of molecular designed nanostructures (NS) that can be used in both therapeutic
Nanomedicine and diagnostic applications in burns. These NSs can be divided into organic and non-organic (such as polymeric
Nanoparticles nanoparticles (NPs) and silver NPs, respectively), and many have been designed to display multifunctional activ-
Stem cells ity. The present review covers the physiology of skin, burn classification, burn wound pathogenesis, animal

Abbreviations: 2ABI, 2-Aminobenzimidazole; Ang-2, Angiopoietin = 2; ASC, Adipose-derived stem cell; ATP, Adenosine triphosphate; AuNP, Gold nanoparticles; BALO, Bdellovibrio and
like organisms; BC, Bacterial cellulose; BLT, Blue light therapy; CNF, Carbon nanofibers; CNP, Carbon NP; CNT, Carbon nanotubes; CrHFC, Crypreserved human cultured fibroblasts; CRISPR,
clustered regularly interspaced palindromic repeats; CS, Chitosan; DC, Dendritic cell; DOTAP, N-(2,3-Dioleoyloxy-1-propyl) trimethylammonium methyl sulfate; ECM, Extracellular ma-
trix; EGF, Epidermal growth factor; EPS, Exopolysaccharide; ERK, Extracellular regulated kinase; FDA, US Food and drug administration; FGF, Fibroblast growth factor; FIR, Far infrared;
GMCSF, Granulocyte macrophage colony stimulating factor; GO, Graphene oxide; GQD, Graphene quantum dots; IGF, Insulin-C12like growth factor; iPSC, Induced pluripotent stem
cell; KGF, Keratinocye growth factor; IL, Interleukin; LED, Light emitting diode; LLLT, Low-level laser (light) therapy; LPL, Low power laser; mAb, Monoclonal antibody; MNP, Magnetic
NP; MRSA, Methicillin resistant Staphylococcus aureus; MSC, Mesenchymal stem cells; MSN, Mesoporous silica nanoparticle; NE, Nanoemulsion; NGF, Nerve growth factor; NHDF,
Normal human dermal fibroblasts; NHEK, Normal human epidermal keratinocytes; NIR, Near infrared; NO, Nitric oxide; NP, Nanoparticle; NS, Nanostructure; OP-GEL, Oxidized pectin gel-
atin; PcrV(A), P. aeruginosa V(A) antigen; PDGF, Platelet-derived growth factor; PDMS, polydimethylsiloxane; PDT, Photodynamic therapy; PEG, Polyethylene glycol; PGLA, Poly-lactic-co-
glycolic acid; Phyto-AuNP, Phytochemical decorated AuNP; PMN, Polymorphonuclear neutrophil; PS, Photosensitizer; Psl, Polysaccharide synthesis locus; PTT, Photothermal therapy;
PVPI, Povidone iodine complex; QD, Quantum dot; rhEGF, Recombinant human EGF; ROS, Reactive oxygen species; siRNA, Small interfering RNA; SLBN, Solid lipid-based NP; SSD,
Silver sulfadiazine; SWCNT, Single wall CNT; TGF, Transforming growth factor; TiO2 NP, Titanium dioxide NP; TLR, Toll-like receptor; TNF, Tumor necrosis factor; TTSS, Type III secretion
system; VEGF, Vascular endothelial growth factor.
⁎ Correspondence to: M. Karimi, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
⁎⁎ Correspondence to: M. R. Hamblin, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
E-mail addresses: m_karimy2006@yahoo.com (M. Karimi), hamblin@helix.mgh.harvard.edu (M.R. Hamblin).
1
These two authors contributed equally to this work.

http://dx.doi.org/10.1016/j.addr.2017.08.001
0169-409X/© 2017 Elsevier B.V. All rights reserved.
34 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

Stimulus-responsive drug delivery models of burn wound infection, and various topical therapeutic approaches designed to combat infection and
Growth factors stimulate healing. These include biological based approaches (e.g. immune-based antimicrobial molecules, ther-
Gene therapy apeutic microorganisms, antimicrobial agents, etc.), antimicrobial photo- and ultrasound-therapy, as well as
CRISPR
nanotechnology-based wound healing approaches as a revolutionizing area. Thus, we focus on organic and
3D printing
Cell-Imprinting
non-organic NSs designed to deliver growth factors to burned skin, and scaffolds, dressings, etc. for exogenous
stem cells to aid skin regeneration. Eventually, recent breakthroughs and technologies with substantial potentials
in tissue regeneration and skin wound therapy (that are as the basis of burn wound therapies) are briefly taken
into consideration including 3D-printing, cell-imprinted substrates, nano-architectured surfaces, and novel gene-
editing tools such as CRISPR-Cas.
© 2017 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
2. Skin structure and the burn wound healing process. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
2.1. Homeostasis phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.2. Inflammatory phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.3. Proliferation phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.4. Remodeling phase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3. Biological-based approaches for burn wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.1. Immune-based antimicrobial molecules. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.1.1. Antimicrobial peptides (AMPs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.1.2. Passive immunotherapy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.2. Reactive oxygen species and nitric oxide generators . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3.3. Antimicrobial agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3.3.1. Metallic elements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3.3.2. Biopolymeric agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
3.4. Therapeutic microorganisms. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
4. Antimicrobial light and ultrasound-based wound therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
4.1. Phototherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
4.2. Shockwave and ultrasound based-therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
5. Stem cell based-burn therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
6. Nanotechnology for treatment of burn infections and wound healing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
6.1. Therapy of burn wound infections using NSs with antimicrobial effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
6.1.1. Organic NPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
6.1.2. Inorganic NPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
6.2. Nanofibers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
6.3. 3D-scaffolds, films and wound dressings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
6.4. Smart stimulus-responsive NSs for burn wounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
6.5. Photothermal and photodynamic therapy for antimicrobial activity and wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . 50
6.6. NS-based cell therapy for wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
6.7. NSs for growth factor delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
6.8. NSs for gene delivery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
6.9. Combination therapies for wound healing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
6.10. New technologies in tissue regeneration and wound healing. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
6.10.1. Nanoscale architectured and textured substrates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
6.10.2. Cell-imprinted substrates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
6.10.3. 3D bio-printed scaffolds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
6.10.4. CRISPR-Cas9: new gene editing tool for reprogramming of stem cells. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7. Animal models for burn wounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.1. Gas flame burn model. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.1.1. Katakura burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.2. Burning ethanol bath burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.2.1. Stieritz-Holder burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.3. Pre-heated single metal plate/bar burn models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.3.1. Tavares burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 54
7.3.2. Orenstein burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.4. Boiling or hot water burn models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.4.1. Suzuki burn models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.4.2. Bahar burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.4.3. Bjornson burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.4.4. Mason and Walker burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.5. Pre-heated double brass blocks burned models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.5.1. Kaufman burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
7.5.2. Stevens burn model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
8. Recent clinical trial evaluations for wound healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
9. Concluding remarks and perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 35

Appendix A. Supplementary data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57


References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57

1. Introduction conducted to boost the antimicrobial activity of a broad range of topical-


ly applied therapeutic agents, facilitating disinfection in wound sites,
Burns are generally defined as skin wounds caused by thermal/heat as well as improving wound healing processes and tissue regeneration
exposure (e.g. fire, hot liquids, solids or gases), electricity, chemical ma- mechanisms [7,8]. Some conventional treatments are based on applica-
terials (e.g. strong bases or strong acids), radiation exposure (e.g. ultra- tion of topical antimicrobial substances such as combinations of
violet light, ionizing radiations such as X-ray, microwaves, etc.), and so topical antibiotics, povidone-iodine, silver-sulfadiazine, chlorhexidine,
forth. The ultimate therapeutic goal is to prevent and treat infection, mafenide acetate and such like. Newer avenues that are being explored
while encouraging satisfactory healing that preserves function [1]. In ac- are immune-based antimicrobial molecules (polypeptides like
cordance with the latest report from the World Health Organization, defensins) and therapeutic microorganisms (probiotics and bacterio-
about 265,000 deaths occur due to thermal burn wounds each year; phages). In addition, remarkable results have recently been reported
these fatalities are especially common in low-to-middle-income coun- for a range of light therapies using NIR, visible light (blue or green); ul-
tries, and particularly in the South-East Asia Region [2]. In India alone, traviolet light, and the combination of light with photosensitizing dyes
over 1,000,000 people are inflicted with moderate or severe burns [9]. Moreover, some other innovative approaches such as stem and
every year. N88% of these burns are caused by flames or contact with other types of cell therapy have led to significantly improved wound
hot or boiling liquids. Patients with severe burns over a significant pro- healing. In recent years, despite the noticeable progress, the relatively
portion of the body surface have received a potentially devastating inju- new areas of nanomedicine and nanotechnology in general, have been
ry, and need immediate skillful emergency care to minimize mortality, strongly pressed into service against burn infections. In this regard,
and will often require long-term rehabilitation and recovery to prevent nanostructures (NSs) have been developed as advanced delivery vehi-
the occurrence of crippling scars and other morbidity. The skin surface cles for: cell therapies, growth factors, gene therapy vectors, advanced
has a pivotal role in the maintenance of body fluid homeostasis, thermo- antimicrobial agents, and agents that will stimulate wound healing.
regulation, serves as a barrier against the outside world (in particular Moreover, a whole new arsenal of smart stimulus-responsive delivery
excluding pathogenic microorganisms), and regulates many metabolic vehicles and nano-robots have emerged using nanostructured plat-
processes [3]. The skin is also the body's largest and most active immune forms e.g. films, 3D-scaffolds, nanoparticles (NPs) (activated by e.g.
organ [4]. Thermal injury is a complex process that needs advanced light, enzymes, pH, ultrasound, etc.), photoactivated NS-based ap-
methods to prevent burn infection, and the application of effective anti- proaches. The most-often utilized NSs platforms for wound healing
microbial therapy if infection has taken place (as it most often does), to- and burn reconstruction can be categorized into nanofibers, NPs, films
gether with strategies to stimulate wound healing and prevent scarring. and 3D-scaffolds. Fig. 1 schematically illustrates several important con-
Initially burned patients should be provided with fluid resuscitation, ventional, developing or innovative approaches for wound healing and
pulmonary care, nutritional support, as well as antimicrobial control burn infection inhibition, which have all been under investigation in re-
as emergency measures [5]. The survival of thermally burned victims cent years.
may critically depend on exactly what is done, particularly to the burn Animal models are critically important for laboratory investigations
sites themselves [6]. Remarkable advances have been obtained in into burn wound healing, as in vitro studies can only go so far, and there
wound dressing platforms, advent of new antimicrobial agents, and rec- is a reluctance to test nanotechnology-based products directly on
ommendations have been made for early full thickness burn excision human burn victims. These models will allow better understanding of
and grafting. Nevertheless studies still are ongoing to improve the wound-associated processes and mechanisms, which then can lead to
wound healing process and its effectiveness. Most efforts have been substantial advancements in burn injury therapies.

Fig. 1. Schematic illustration of important conventional, developing or innovative approaches, (such as light therapy; therapeutic microorganisms e.g. bacteriophages and bacteria with
bactericidal effects; antibacterial NPs or NPs with therapeutic effects e.g. silver NPs and gold NPs; innovative nanocarriers e.g. smart nanocarriers and nanogels; novel scaffolds for
wound dressing, (stem) cell therapy and regenerative medicine, therapeutic (drug, growth factor, gene, etc.) delivery, and so forth; and innovative technologies e.g. 3D-printing, cell-
imprinting, and CRISPR-Cas9 gene editing), which have been the subject of various researches, conducted with the aim of wound healing and prevention of infection in burns.
36 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

Fig. 2. Schematic illustration of: a) the structure of skin, b) layers of skin including epidermis, dermis and hypodermis, and their constituent cells and sub-layers.

2. Skin structure and the burn wound healing process collagen protein, blood vessels (arterioles, venules, capillaries) sweat
glands, hair roots, nerve cells, lymphatic vessels and mesenchymal
An understanding of normal skin physiology, pathogenesis of burns, stem cells (MSCs) [15]. The primary function of the dermis is to provide
as well as appreciation of infections that develop in burns (with structural toughness to the skin. The differentiation of dermal MSCs cul-
different classes of microorganism arriving at different stages of burn tured in the laboratoryinto chondrocytes, osteocytes, adipocytes,
development) are all important for understanding the role of nanotech- smooth muscle cells, hematopoietic cells, neurons and glia have been
nology in burn wounds. reported [16]. The hypodermis or subcutaneous fat, is comprised of ad-
Generally, the skin is composed of three distinct layers (Fig. 2-a) in- ipocytes, macrophages, vasculature, nerves and fibroblasts, and its func-
cluding the epidermis, dermis and hypodermis [10]. The epidermis pre- tion is to support and anchor the dermal and epidermal layers [17]. Fig.
vents the entry of virulent microorganisms, as maintaining body 2 schematically illustrates the structure of skin (Fig. 2-a), and its differ-
hydration. The five layers (called stratums) consisting of: basale, ent constituent layers and cells (Fig. 2-b).
spinosum, granulosum, lucidum, and corneum form the epidermis Classification of the severity of burn injuries usually is evaluated
layers of skin, respectively [11]. These layers are made from diverse im- using two criteria; assessment of the depth of the thermal damage,
mune and non-immune cell types such as keratinocytes, melanocytes, and an estimation of percentage of total body surface area (TBSA) affect-
merkel cells, langerhans cells, CD8 + T cells, corneocytes and stem ed by a burn injury. In the first method burn wounds are classified into
cells [11–13]. The basal layer is the proliferative portion of the epidermis four degrees (Fig. 2-a). First degree burns that involve only the epider-
producing keratinocytes that can go on to differentiate into the spinous mis display redness, pain at the site of damage, and dryness. Second de-
layer, granular layer as well as outermost stratum corneum layers. The gree burns can affect either papillary layer (i.e. the upper dermis) or
skin of the palms and soles has an additional stratum lucidum or clear reticular layer (i.e. the deeper dermis) with a white or yellow color, blis-
layer. Stem cells residing in the basal epidermis are responsible for con- tering of the skin, and a moist appearance. In third-degree burns, there
tinually replacing keratinocytes that are normally lost by exfoliation, as is full thickness destruction of both epidermis and dermis, and a stiff or
well as for regenerating wounded skin [14]. The dermis layer is a layer leathery consistency. Scarring and contractures will develop after
between the hypodermis and epidermis, which is largely made of healing. Fourth-degree burns cause catastrophic damage in underlying
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 37

tissues, muscle, tendons, ligaments and even bone [18]. As mentioned, Table 1
burns are also assessed in terms of percentage of TBSA. The rule of nines Cellular and molecular events in normal wound healing.

in the affected area is a quick way to estimate the percent TBSA affected Phases
(Fig. 3) [3,5,19]. More precise assessment can be evaluated by Lund & Homeostasis Platelet aggregation, immunity activation, blood clotting and
Browder charts where diverse proportions of body parts in children and complement system induction
adults are accounted for; e.g. burn wounds N 10% in children, have the Inflammation Recruitment of neutrophils and macrophages as well as ECM
same severity as 15% for adults. This is important because hypovolemic molecule production by resident cells in the skin
Proliferation Re-epithelialization; expansion of keratinocytes, epithelial cells,
shock can be potentially life threatening. When about 25% of the body
stem cells and fibroblasts
area is affected by burns, loss of fluid from the damaged microvessels pro- Angiogenesis; activation of endothelial cells and creation of a site in
duces edema and hypoproteinemia. The volume of fluid replacement which these cells can proliferate in the wound
with isotonic crystalloid solution (Ringer's lactate) can be calculated by Granulation; the promotion of tissue granulation by fibroblasts,
the Parkland formula: V(mL) = 4 × body weight(kg) × %TBSA [7,20]. granulocytes and macrophages
Remodeling Production of collagen and elastin as well as fibroblasts maturing
The final goal in burn injury healing is the rapid replacement of nor- to myofibroblasts in the presence of T cells and macrophages
mal skin with minimal scarring. The healing of burn damage is a biolog-
ical process including four overlapping phases (Table 1), homeostasis
phase, inflammatory phase, proliferation phase and remodeling phase activate the inflammatory response and stimulate vascular endothelial
[21]. growth factor (VEGF) and IL-8 secretion to repair blood vessels, and
macrophages produce transforming growth factors (TGF-α, and TGF-
2.1. Homeostasis phase β), fibroblast growth factor (FGF), platelet-derived growth factor
(PDGF) and VEGF, to stimulate cell expansion and migration and the ex-
About 10-min after the thermal insult occurs, the initial phase com- tracellular matrix (ECM) generation molecules by resident cells of the
mences as an autonomic response to attempt to minimize the damage. skin. Adaptive immunity takes at least 4 days after the damage initiation
In this phase, several phenomena occur including platelet aggregation, involving lymphocytes (T-cells and B-cells) [22,23].
immune activation, blood clotting and complement system activation.
Here, the blood clot encompasses vitronectin, fibronectin, fibrin and 2.3. Proliferation phase
thrombospondins, which supply a scaffold-like matrix for the migration
of fibroblasts leukocytes, keratinocytes and endothelial cells as well as Following the inflammatory response, a coordinated proliferation
causing the accumulation of growth factors [15]. phase occurs, which includes three steps; re-epithelialization, angio-
genesis and formation of granulation tissue. The proliferation phase oc-
2.2. Inflammatory phase curs 3–10 days after the initiation of injury. Re-epithelialization is
induced by activation of cytokines and growth factors including
This occurs 1–3 days after the infliction of the burn wound. This insulin-like growth factors (IGF-1), nerve growth factor (NGF), epider-
phase can be divided into the early phase with neutrophils arriving in mal growth factor (EGF), and keratinocyte growth factor (KGF), which
the first days and a late phase; 3 days after the initiation of damage, cause expansion of keratinocytes, epithelial cells, stem cells and
when monocytes transform into macrophages. Neutrophils produce fibroblasts. Keratinocytes, fibroblasts and fibroblast-derived macro-
tumor necrosis factor (TNF-α), interleukin-1 (IL-1) and (IL-6) that phages immigrate into the wound and expand during the regeneration
of the burn-damaged tissue. Some fibroblasts differentiate into
myofibroblasts; then the fibroblasts and myofibroblasts manufacture
ECM, particularly collagen that produces the mature scar [15,24]. In
the proliferation phase, new blood vessels are created (angiogenesis).
In this phase, several growth factors are involved such as VEGF, PDGF,
FGF-β, granulocyte-macrophage colony-stimulating factor (GM-CSF)
and thrombin, which are the foremost activating molecules for endo-
thelial cell growth. The proteolytic enzymes secreted by the activated
endothelial cells secrete dissolve the basal lamina and create an envi-
ronment in which they can proliferate and immigrate into the wound
[15,25]. The final step of the proliferation phase is the production of
granulation tissue, which can be specified by the presence of many

Table 2
Topical methods for prevention and treatment of burn infection and
wound healing.

Biological-based approaches for burn wounds healing:


Immune-based antimicrobial molecules
Antimicrobial peptides (AMPs)
Passive immunotherapy
Reactive oxygen species and nitric oxide generators
Antimicrobial agents
Metallic elements
Biopolymeric agents
Therapeutic microorganisms
Antimicrobial light and ultrasound-based wound therapy:
Phototherapy
Shockwave and ultrasound based-therapy
Stem cell based-burn therapy
Fig. 3. Assessment of skin burn in terms of total body surface area (TBSA) percent, using
Burn wound infection therapy using nanostructures (NSs) and NPs
the rule of nines in applied area, thus to quickly estimate the affected TBSA percent.
38 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

fibroblasts, granulocytes and macrophages. The main cells in the granu- membrane, and can neutralize the biological activity of their endotoxins
lation phase are fibroblasts that produce collagen and other ECM mole- [36]. Moreover, they can adhere to membrane receptors of immune
cules. The ECM supplies an appropriate scaffold for cell adhesion, and cells such as toll-like receptors (TLRs) on antigen-presenting cell includ-
organizes the growth and differentiation of the cells including fibro- ing monocytes, macrophages, and dendritic cells, as well as non-
blasts. At the end of this phase, the differentiation of the fibroblasts immune cells such as epithelial cells and keratinocytes. Cathelicidins
into myofibroblasts occurs (forming a scar) or else undergo apoptosis and their derivative peptides such as LL-37 can be used topically to
[26,27]. treat burn wound infections [37,38]. Lactoferrin is an AMP, and binds
to iron in a similar manner to transferrin (serum carrier of iron). This
2.4. Remodeling phase peptide exists in neutrophil granules, milk, tears, saliva, sweat, nasal se-
cretions, and the genital tract. It exerts antibacterial and antifungal ac-
This final step of wound healing is called the remodeling phase. Dur- tivity through microbial membrane disruption as well as iron
ing this step, wound remodeling initiates 2–3 weeks after the burn dam- sequestration, thus repressing microorganism growth [39–41].
age, and continues for as long as a year or more; here the wound scar Synthetically-obtained AMPs have been also investigated for their
produces more collagen and elastin, and also fibroblasts mature into enhanced antimicrobial activity. In an investigation Zhong et al. devel-
myofibroblasts. Beside fibroblast transformation, death of keratinocytes oped β-sheet SAMPs including IK8D, IK8L, and IK8-2D to inhibit
and inflammatory cells (such as T cells and macrophages) play a vital multidrug-resistant P. aeruginosa, the most common Gram negative
role in the ending of the response to the injury [15,28]. bacteria isolated from burn wound infections of humans. These SAMPs
Topical methods have been conventionally used as treatments for were applied for treatment of P. aeruginosa-infected burn wounds in
burns. Many of these are traditional disinfectants and antiseptics such mice. Findings revealed that among the SAMPs, IK8L could significantly
as povidone-iodine, chlorhexidine, combinations of topical antibiotics, inhibit P. aeruginosa, and its successive administration did not cause
silver-sulfadiazine, and mafenide acetate. In the following sections, we drug resistance [31]. Recently, Mohamed et al. evaluated the antibacte-
will cover more innovative approaches such as immune-based rial activity of SAMPs for treatment of MRSA-induced infection, one of
antimicrobial molecules, nanotechnology based antimicrobial agents, the most virulent microorganisms in human wounds. Herein, the anti-
therapeutic microorganisms, and antimicrobial phototherapy and ultra- bacterial function of two topically applied SAMPs including WR12
sound therapy, and stem cell-based treatments for therapy of burn in- (consisting of a 12 residue peptide of arginine and tryptophan) and D-
fection and burn wound healing. Since many of these agents are IK8 (made of an 8 β-sheet peptide structure) against MRSA was evalu-
intrinsically cytotoxic towards bacteria, fungi and other microorgan- ated. These SAMPs were found to inhibit MRSA in both stationary and
isms, a clear concern that must be addressed is whether they will dam- intracellular bacterial growth phases compared to the control groups
age host cells in the burn site, or maybe adversely affect wound healing (i.e. linezolid and vancomycin) in vivo, as well eradication of S. aureus
Table 2. and S. epidermidis in vitro. The in-vivo evaluation indicated both the
SAMPs regulated the immune system and decreased generation of
3. Biological-based approaches for burn wound healing pro-inflammatory cytokines e.g. TNF-α and IL-6 [42]. In another study,
charge- and structure-manipulated SAMPs showed enhanced antimi-
Some conventional treatments are based on application of topical crobial activity and immunomodulatory ability. Herein, a pendant aro-
antimicrobial substances such as combinations of topical antibiotics, matic group was added to the SAMP sequence to achieve an enhanced
povidone-iodine, silver-sulfadiazine, chlorhexidine, mafenide acetate antibacterial activity against Gram negative bacteria e.g. E. coli. The
and such like. Newer avenues that are being explored are immune- SAMP with the addition of 6 charges and a naphthalene ring showed in-
based antimicrobial molecules (polypeptides like defensins) and thera- creased selectivity against both Gram negative- and Gram positive bac-
peutic microorganisms (probiotics and bacteriophages). teria e.g. E. coli and S. aureus. The regulation of macrophage
immunomodulatory function with and without a TLR agonist (i.e. lipo-
3.1. Immune-based antimicrobial molecules polysaccharide) as well as the generation of neutrophil chemoattractant
activity were also demonstrated [43].
3.1.1. Antimicrobial peptides (AMPs)
AMPs are naturally or synthetically generated cationic polypeptide 3.1.2. Passive immunotherapy
molecules, which include some specific amino acids that produce an This approach is based on humanized and chimerized forms of spe-
overall amphipathic conformation [29,30]. These molecules can be nat- cific monoclonal antibodies (mAb) generated by engineering transgenic
urally produced by many different organisms including bacteria, am- mice and cultured hybridoma cells. For example, antibodies against the
phibians, insects and mammals and an active research effort in Pseudomonas aeruginosa lipopolysaccharide O-side chain structure can
screening and discovery is still underway. AMPs are also produced by be produced in transgenic mice. Other examples of passive immuno-
a diverse range of cell types well found in the skin, mucosal surfaces therapeutics are flagellin-binding antibodies that recognize; bacterial
and immune cells. Synthetic approaches are also available for genera- flagellins. These structures are essential virulence factors responsible
tion of AMPs. AMPs as antimicrobial agents are able to destroy an for the rapid movement of bacteria and facilitate bacterial invasion
wide range of microorganisms. Their antimicrobial mechanism of action into tissue. High titer of mAbs such as anti-flagellin mAb could be
operates via attaching to and disrupting the negatively charged microbi- used as an effective molecular approach to diminish mortality and mor-
al cell membrane as well as suppressing the generation of important bidity induced through burn wounds with P. aeruginosa infection
molecules of microorganisms consisting of proteins and nucleic acids, [44–46]. Exopolysaccharide (EPS) of P. aeruginosa is biosynthesized by
thus inhibiting the microorganism growth. Hence, they can be applied the polysaccharide synthesis locus (Psl) and includes galactose- and
for the treatment of burn infections [29,31–33]. mannose-rich molecules. The psl gene cluster consists of 15 genes
Various natural AMPs such as indolicidin and ranalexin have been encoding proteins capable for EPS synthesis, which is considered to be
introduced as potential antimicrobial agents to treat infectious diseases an essential factor for bacterial biofilm formation [47]. Another ap-
[34,35]. Cathelicidins (a member of the AMP family) are found in lyso- proach uses antibodies that bind to P. aeruginosa V-antigen (PcrV).
somes of macrophages, polymorphonuclear leukocytes (PMNs), and PcrV is involved in the type III toxins secretion system (TTSS) that al-
keratinocytes. They are pivotal molecules for immune responses to mi- lows toxins to be released from the cells. PcrV is a structural protein
crobial infections. They have broad-spectrum antimicrobial effects and hydrophilic translocator of TTSS, which has a pivotal role in bacteri-
against fungi, viruses, and Gram-positive and Gram-negative bacteria. al injection into host cells in order to initiate an infection [48,49]. PcrG is
These small peptides kill virulent pathogens by destroying their cell also a cytoplasmic regulator that can interact with the intramolecular
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 39

coiled-coil region of PcrV protein and regulates TTSS [50]. Passive mAb antimicrobial activity. Another metal element is ferric iron (Fe3 +),
treatment against PcrV, so-called PcrV immunization, enhances survival which is a vital element that has a critical role in bacterial growth and
rate in murine burn models infected with P. aeruginosa [51,52]. metabolic pathways [68]. The antimicrobial activity of Ga3+ maltolate
salts against P. aeruginosa infections in murine burn wounds, was
3.2. Reactive oxygen species and nitric oxide generators shown to induce inactivation of Fe3 +-dependent redox enzymes,
which had important roles in cell proliferation of bacteria. The results
Reactive oxygen species (ROS) as potent antimicrobial agents are showed substantial decreases in the colony forming units of
defined as very reactive molecules containing oxygen including hydrox- Acinetobacter baumannii and Staphylococcus aureus in the wound site
yl radicals(•OH), hydrogen peroxide (H2O2), superoxide (O− 2 ), and sin- [69].
glet oxygen(1O2), produced by oxygen reduction in inflammatory Halogen based compounds including: fluorine, chlorine, bromine
leukocytes particularly neutrophils at infectious damaged sites [53]. and iodine, have been used as antimicrobials. Hypochlorous acid is a re-
ROS produced by defending host cells are able to inhibit a diverse active chlorine compound that can damage DNA and inactivate oxida-
range of microorganisms. The activity of superoxide and hydrogen per- tive phosphorylation enzymes in microorganisms. HClO has a
oxide is less effective than both hydroxyl radical and single oxygen, be- potential virucidal effect and disturbs the nucleic acids and capsids of vi-
cause of their detoxification due to presence of endogenous anti- ruses, and can be also used for disinfection of contaminated burn
oxidants related to enzymatic and non-enzymatic mechanisms; while wounds [70]. Iodine molecules suppress essential enzymes of microor-
no enzyme can detoxify hydroxyl radical or single oxygen (although or- ganisms by attaching to the sulfur groups of their proteins [71]. Iodine
ganic antioxidant molecules can quench them), so they are extra lethal has effective antimicrobial activity against fungi and spores. Iodophors
for pathogenic microorganisms [54]. Unfortunately, ROS are able to in- are preparations that contain iodine in combination with a solubilizing
teract with host biomolecules and cause cellular and finally tissue dam- agent that can release free iodine molecules in situ. Iodine compounds
age. Pathogens can inhibit ROS by their own enzymes with anti-oxidant have been applied as antiseptics and surface disinfectants for treatment
activity such as catalase and superoxide dismutase. ROS have bactericid- of burn wound infections [72,73].
al and virucidal effects that are utilized by human cells against microor-
ganisms in burn wound infections [55,56]. Nitric oxide (NO) is a reactive 3.3.2. Biopolymeric agents
free radical with a short half-life. NO is made from L-arginine by the en- Other naturally occurring antimicrobial compounds can also interact
zyme NO synthase both constitutively and induced pathways of human with microbial cells and induce membrane disruption. Chitin is the sec-
immune cells [57]. Its antimicrobial activity is caused by damage to mi- ond most commonly-found biopolymer (behind cellulose) and is a lin-
crobial DNA, DNA repair enzymes and lipid damage to microorganisms. ear polysaccharide isolated from crustacean shells, fungal cells and
NO generation has been reported as one of the main processes occurring yeast. A common chitin derivative, chitosan, can be obtained by chemi-
in wounds and during skin reconstruction. Many recently produced top- cal extraction and hydrolysis of acetyl groups. The positively-charged
ical gels can deliver NO content for treatment of skin wounds. NO re- structure of chitosan (CS) is the base for its therapeutic properties
cruits hair follicle stem cells for the re-epithelialization of skin such as antimicrobial activity, fast blood coagulation and electrostatic
wounds. It also improves angiogenesis and accelerates collagen produc- immobilization of wound microorganisms [74,75]. XF porphyrins such
tion in the damaged site. Therefore NO has been utilized for burn wound as XF-73 and XF-70 are dicationic porphyrin compounds, originally de-
healing through several mechanisms [58,59]. veloped for photodynamic therapy [76]. However these agents were
also found to disrupt the bacterial membrane in many bacterial species.
3.3. Antimicrobial agents XF-70 and XF-73 have been reported as potential suppressors against
non-dividing or slow-growing forms of bacteria such as S. aureus. Addi-
3.3.1. Metallic elements tionally, these compounds have shown an effective inhibition activity
Several metallic elements have been tested as effective materials to against bacterial biofilms [77,78]. Polyphenolic compounds such as tan-
treat microbial infections [60]. Silver compounds have been utilized in nins can be purified from several plant species such as tea, vegetables,
burn infection treatment. Ionized silver (Ag+) compounds can react cereals, beverages, grapes, apples, pears and cherries. Plants containing
with thiol moieties of enzymes and proteins in the microbial cells, tannins have been used in medicine for their antimicrobial properties
thus inhibiting growth and metabolism of a wide range of microorgan- [79]. Essential oils such as Valencia orange oil and tea tree oil are sub-
isms. Moreover, silver ions are able to bind to cell walls and intracellular stances extracted from various plants and used in cosmetics, cuisine
vacuoles of bacteria, which inhibit microbial division. Silver also causes and medicine [80]. Many researchers have reported specific oils display
membrane damage and disrupts electrical conductivity and membrane antimicrobial activity, applicable as a topical therapy against infections
potential [61]. Silver nitrate was the original agent used for burns, which induced in burn wound [81]. Alginate is a derivative of Laminaria
is utilized to inhibit the growth of microorganisms. It is still utilized as a digitata brown seaweed [82]. Alginate-silver compounds and alginate
treatment for burns, but concerns about host toxicity have led to the oligomer-loaded with antibiotics have been utilized as antimicrobial
development of other silver derivatives [62]. Silver sulfadiazine (SSD) agents against various virulence microorganisms such as Enterococcus
is another derivative that can be applied to prevent against microbial faecalis, Enterococcus faecium, P. aeruginosa, E. coli, and so forth [83,84].
wound dressing and cell proliferating uses on the skin damaged Polyamines inhibit efflux pumps situated on microbial membranes
surface [63]. that function to pump out antibiotics, and which are a major cause of
Bismuth-thiols are other metal compounds that have antibacterial microbial multi-drug resistance. Infections in burns induced by drug-
activity against staphylococci causing wound damage [64]. Copper resistant bacteria e.g. S. aureus have been reported to be treated using
(Cu) as a metal element and its derivatives such as Cu-nicotinate com- preparations containing polyamine compounds [85]. Aerobic (and an-
plex have natural antimicrobial activity [65]. Cu ions bind to some aerobic microorganisms) quickly colonize skin wounds after burn dam-
vital groups of microorganisms and suppress their enzymes. It has age. Partly owing to the widespread application of antimicrobial
been reported that Cu salts can be used on infected skin wounds such compounds e.g. antibiotics, fungi and yeasts are observed to colonize
as burns [66]. Cu compounds have also been indicated to have antibac- burns particularly at later time points [86]. Mafenide acetate is a dose-
terial activity against Vibrio cholerae, Shigella flexneri, Escherichia coli, depended bactericidal and bacteriostatic agent that can inhibit Gram-
Salmonella typhi and Salmonella paratyphi [67]. Gallium (Ga) is a metal- negative and Gram-positive bacteria. Mafenide acetate is widely applied
lic element that is not found in its pure form in nature, while its ionic in burn injury treatment [87,88]. Firmocidin is a compound derived
Ga3+ form has been found in many minerals such as those containing from the normal commensal bacterial species that lives on the skin,
aluminum and zinc. Ga3 + compounds have been shown to have Staphylococcus epidermidis, and which can inhibit other pathogenic
40 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

bacteria such as Staphylococci and Streptococci spp. This bacterial agent inducing their death [105–107]. Recently, researchers have tested the
has been found to cause antimicrobial and immunomodulatory (i.e. activity of BALOs against E. coli as well as in burn wound infections
anti-inflammatory) activity in burns. Firmocidin can be utilized against caused by P. aeruginosa [108].
many burn pathogens such as S. aureus, group A and group B streptococ- Bacteriophages as viruses can enter their host bacterium, within
cus Streptococci [8]. Fusidic acid is a bacteriostatic agent derived from which they can replicate, effectively infecting the bacterial cell. Bacteri-
Fusidium coccineum fungi. It is capable of suppressing protein produc- ophage amplification occurs into the host bacterial cells, which enable
tion in Gram-positive bacteria such as Staphylococcus spp. and Strepto- bacteriophages to be widely used as gene carrier platforms. Different
coccus spp. in burn wound infections [89]. Usnic acid is an antibiotic types of phages have specific shape, a diverse loading capacity, a partic-
and anti-inflammatory molecule containing a dibenzofuran structure. ular mechanism and rate of molecular assembly [109]. Antibacterial
Thentimicrobial activity of this compound against Gram-positive bacte- phages have been developed due to their efficacy against antibiotic re-
ria has been reported. Hence, it has been used in treatment of burn sistant bacteria [110], however the high specificity of phages for their
wounds [8,90]. individual bacterial host, means that a cocktail of phages must be used
Nubiotics are novel antibiotics derived from proprietary protonated to obtaining any broad-spectrum activity. In a recent study, bacterio-
nucleic acid and applied against some of infectious pathogens. They phages were utilized for wound infection treatment in mice induced
were tested in a mouse model of burn wounds infected with a lethal by Klebsiella pneumonia suggesting that bacteriophage therapy could
dose of P. aeruginosa and gave a significant survival advantage [91]. be applied for K. pneumoniae induced wound infections [111]. In one
Biofilms are formed through complex multilayer communities com- study, phage therapy was tested in a preliminary clinical trial. Here, a
prised of bacterial cells embedded within a matrix with self-secreting bacteriophage cocktail (BFC-1) was employed to act against
capability, containing extracellular polymeric substance (EPS), proteins P. aeruginosa and S. aureus in colonized burn wounds. No adverse effects
and nucleic acids. They exist in an extensive span of natural and artificial or abnormality was detected due to the phage, opening the way for fur-
environments, which can supply a protective microenvironment for mi- ther investigation [112]. In another clinical trial, phage therapy gave
crobial cells. Biofilm-growing cells have been shown to express various fewer complications, temperature normalization, as well maintained
set of genes, thus are highly resistant to most antimicrobial treatments. the level of phagocytosis. Streptococcus and Staphylococcus strains
A multilayer biofilm model was described that could be used for testing were cultured less often in wound secretions [113]. Elsewhere, a
antimicrobial approaches (such as antibiotic ointments) against burn phage cocktail was used against K. pneumonia-mediated burn wound in-
wound bacterial isolates [92]. The compound 2-aminobenzimidazole fection, and its effectiveness for arresting infection processes without
(2-ABI) can be found in sea sponges. It can protect the sessile sea any likelihood of generating resistant strain mutants [114]. Multidrug-
sponges against the growth of marine biofilms. 2-ABI analogs exert resistant P. aeruginosa infection has been also illustrated to be controlled
anti-biofilm activity against biofilms of Gram-negative bacteria such as in mice through exploiting a lytic phage [115]. In Holguin's work,
P. aeruginosa [93]. Polyanionic polyphosphate materials can chelate me- phage Φ Pan70 showed a significant effect against P. aeruginosa in-
tallic elements including calcium, magnesium, manganese and iron. As a fections. Herein, all mice were rescued in the burned model, al-
result, they possess antimicrobial activity and are used to treat burn in- though the skin lesions remained present in several experimental
fections [8]. A biofilm destruction device was designed to generate groups. Thus, bacterial invasion into the blood was hindered by the
ozone and oxygen at a certain ratio. Ozone has a potent antimicrobial phages. However, some side-effects such as an intensified immuno-
activity and can be used in a controlled concentration to deactivate bac- logical response and mastocytosis in the skin was reported at the in-
teria and fungi in treatment of burns e.g. S. aureus and P. aeruginosa [94, oculation site [116].
95]. D-amino acids also show a dispersal activity against biofilms, and
can prevent important microorganisms such as S. aureus and 4. Antimicrobial light and ultrasound-based wound therapy
P. aeruginosa from forming biofilms. These D-amino acids are also gen-
erated by the bacteria themselves [96,97]. Dispersin B via an enzymatic 4.1. Phototherapy
anti-biofilm activity cleaves the poly-N-acetylglucosamine molecules,
one of the polysaccharide constituents of the biofilm produced by Antimicrobial phototherapy (PT) can utilize both visible and non-
S. aureus and E. coli, Therefore, this enzyme can be used to treat burn visible light (ultraviolet and near-infrared), to treat infectious diseases
wound biofilms [98,99]. in a minimally invasive manner. Light irradiation has been indicated
Various mechanisms for development of antimicrobial resistance to cause improvement in the healing process of burn wounds
have been detected in microorganisms [100]. These resistance mecha- [117–123]. PT consists of several therapeutic methods such as UV irradi-
nisms consist of chemical modification of the antimicrobial agent, ation (200–280 nm), blue light therapy (BLT) (400–470 nm) and low-
pumping out the drug from the cells, and modification of the microbial level laser therapy (LLLT) (390–1100 nm). Also, NP-based photodynam-
target. Antimicrobial resistance may be an inherent property of the or- ic therapy (PDT) and photothermal therapy (PTT) are promising varie-
ganism, e.g. related to type of the cell wall structure, rendering it resis- ties of phototherapy, which will be discussed in the following sections
tant, or it may be achieved via mutation in its microbial genome and by [124–128].
acquiring mobile DNA elements such as plasmids from other species Although UV light is supposed to have adverse effects on mammali-
[101,102]. an cells (e.g. by damaging their DNA) particularly upon prolonged or re-
peated exposure, several investigations have been conducted that
3.4. Therapeutic microorganisms showed that microorganisms are overall much more susceptible to UV
inactivation than mammalian cells. Therefore, it has been suggested
Therapeutic microorganisms exert antimicrobial activities against that UV light could be used as a viable approach for infection therapy
the growth, adhesion of various pathogens to host cells, as well as bio- [129,130]. UV irradiation damages microbial nucleic acids, and induces
film generation, and can be used for burn infection therapy [103]. Probi- repression of the microorganism replication [131]. Investigations have
otic bacteria include a group of Lactobacillus strains. L. acidophilus is also reported UV light for sterilization and wound care [132]. The chief
commercially available in fermented milk, yogurt and other foods. disadvantage of UV light is its poor penetration into tissue so it can
These bacteria can significantly inhibit Gram-positive and Gram- only be used for fairly superficial infections.
negative bacterial growth in burns [104]. Bdellovibrio bacteriovorus LLLT is a non-thermal and non-antimicrobial phototherapy that uti-
and Bdellovibrio like organisms (BALO) are naturally small and motive lizes laser or light irradiation [133]. LLLT has been tested in studies to
predatory bacteria belonging to the α- and δ-proteobacteria phylum, treat several disorders, and was shown to increase cellular proliferation,
which can only replicate inside Gram-negative bacteria, thereby modulate inflammation, and accelerate re-epithelization and repair of
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 41

the injured skin [125,134]. In one study by Yadav et al. [117] a super- closure (via contraction). They suggested that ultrasound would have
pulsed 904 nm laser (200 ns pulse width, 100 Hz, and mean output useful effects on the inflammatory and proliferative phases in burn
power equal to 0.7 mW) was used to increase ATP levels in burn treatment process; but could induce undesirable angiogenesis as well
wounds. The results indicated improved aerobic metabolism (by in- as inflammation in the remodeling phase [136].
creasing the activity of enzymes e.g. PFK, CS, G6PD, and CCO and higher
ATP levels, and lower NADP/NADPH ratio), decreased expression of GS1 5. Stem cell based-burn therapy
and up-regulation of GLUT1 and pAMPKα. These changes led to activa-
tion of cellular bioenergetics, regulation of the redox homeostasis and Considering their regenerative ability, stem cells have been regarded
reduced inflammatory response, thus explaining the improved wound as a versatile and efficacious therapy in biomedicine [137], and could
healing. provide new solutions for many diseases including, osteoporosis, osteo-
Blue light therapy (BLT) has shown impressive antimicrobial effects, arthritis, cartilage regeneration, and cardiovascular. In the case of
especially for treatment of infected skin wounds including burns. In an wound treatment, the combination of stem cells with nanostructures
investigation, Dai et al. [118] interrogated the mechanism of action of has opened up new horizons. Stem cells can be distinguished from
415 nm blue light in P. aeruginosa infected wounds. P. aeruginosa cells other kinds of cells by three main attributes: 1) renewability and ability
displayed 35-fold higher levels of killing by blue light, compared to to divide themselves for prolonged periods of time; 2) they are non-
the killing seen with keratinocytes. Results showed intracellular dam- specialized cells; and 3) they potentially can differentiate into miscella-
age to P. aeruginosa cells. Using an animal model where the infection neous specified cell types. Diverse stem cells derived from humans and
could be monitored using bioluminescence imaging, there was a re- animals have been reported including induced pluripotent stem cells
duced area-under-the-bioluminescence-time-curve after blue light ex- (iPSCs), non-embryonic “somatic” or “adult” stem cells, and embryonic
posure, improved survival rate of infected mice, and negligible stem cells. In this regard, stem cells have shown promising potential to
damage to the skin and keratinocytes of the mice caused by blue light. treat burn wounds.
Elsewhere, the antimicrobial activity of 415 nm blue light was exploited Stem cells can improve wound healing through differentiation into
against multidrug-resistant A. baumannii infection in mice with third- skin cells, by a pronounced anti-inflammatory effect and by stimulating
degree burn wounds. The multidrug-resistant bacteria showed higher angiogenesis. Foubert et al. [138], for instance, found acceleration of re-
blue light-susceptibility than human keratinocytes [123]. Also, other re- epithelialization in treated wounds by using adipose-derived stem cells
sults were reported including blue light-triggered ultrastructural bacte- (ASC). Subsequently, an increase in blood vessel density and deposition
rial cell damage, negligible damage to DNA of mouse skin, no resistance of ECM (collagen) was indicated. Directly injecting stem cells into the
to blue light inactivation, and highly attenuated bacterial burden in wound, or their intravenously administrating both led to comparable
mouse tissue samples. beneficial outcomes.
Furthermore, green light has also been the subject of some studies. Yang et al. [139], showed epidermal stem cells that overexpressed
Catão used green LED light to tackle skin burns in rats by exerting the EGF-like domain 7 (EGFL7-ESCs) could stimulate fibroblasts to pro-
anti-inflammatory effects. The results showed a substantial decrease liferate and migrate, and this effect could be used for enhanced healing
in inflammatory cells [119]. of wounds. Bliley et al. [140] employed adipose derived stem cells
Far-infrared (FIR, 3 μm–100 μm) light has also shown beneficial ef- (ASCs), and demonstrated an enhancements in the mRNA expression
fects on wound healing. In a research by Chiu et al., the exploitation of of collagen (type I and III), collagen deposition vascularity, which even-
FIR reduced NLRP3 inflammasome activity and IL-1β generation caused tually followed by adipogenesis. They suggested ASCs could have a vital
ASC ubiquitination (a regulatory role of inflammasomes), followed by role as an adjunctive therapy for full-thickness burn wounds.
enhanced transport of inflammasomes to autophagosomes. FIR irradia- Recently, in a research [141], bone marrow–derived mesenchymal
tion also lessened the infiltration of inflammatory cells, prevented loss stem cells (BM-MSCs) migration into the burn wound margins en-
of specific collagen fibers, and burn-triggered epidermal thickening, as hanced the re-epithelialization, as well, the mobilization of BM-MSCs
well enhanced autophagy. Hence, these results suggested FIR could pro- was regulated via CXCL12/CXCR4 signaling.
mote burn wound healing [120]. In another study, a single exposure of a In another study [142] allogeneic MSCs were shown to improve burn
low power laser (830 nm) equal to 3 J/cm2 was applied to thermally- wound healing via reducing unhealed areas, and enhancement of the
injured Swiss albino mice. Histological and immunohistochemical anal- dermal thickness, epidermal area and collagen content. In a study by
yses indicated improved tissue healing in the NIR-illuminated mice, and Chen et al. [143], adipogenic differentiated ASCs (using + 3-isobutyl-
regulation of several cellular markers [122]. 1-methylxanthine (IBMX) [D1–5] + INSULIN) enhanced early wound
Recently, low power laser (LPL) and LED have been employed to better compared to ASCs, as well as decreasing fibrosis and inflammato-
realize the molecular mechanisms related to epidermal healing in ry infiltration in the superficial dermis.
burn wounds. The results signified efficacy of 660 nm LPL and
850 nm LED to decrease oxidative stress levels and lower expression 6. Nanotechnology for treatment of burn infections and wound
of IL-6 and pERK1/2, leading to downregulated inflammatory re- healing
sponses, less dermal necrosis, and enhanced granulation tissue gen-
eration, subsequently all causing betterment of burn wound healing Treatment of burn wound infections and skin regeneration are com-
process [121]. plex processes, and have been researched for decades. Recently the rise
of antibiotic-resistant pathogenic microorganisms has correlated to
4.2. Shockwave and ultrasound based-therapy greater resistance to topical antimicrobial agents in burn injuries,
which is regarded as an important threat to public health. Hence, devel-
Therapeutic shockwaves and ultrasound have been indicated to oping new even more potential antimicrobial agents is a necessity.
have advantageous effects on wound treatment, thus to accelerate the Nanotechnology has introduced particles made of a wide variety of ma-
wound healing process in burns. Recently, Rünzler et al. indicated the terials, but what they have in common is a size range of 10–1000 nm
efficacy of shockwave treatment for wound healing by activating [144,145]. Nanotechnology has been called “a new paradigm” in medi-
purinergic ERK1/2 signaling pathways by inducing ATP release, and cine, and new therapeutic methods based on NSs have been developed
then enhancing cell proliferation [135]. In another study, Fantinati to tackle many diseases [146,147], particularly burn wounds and burn
et al. investigated chronic third-degree burn wounds and reported infections. Nanomedicine can lead to effective improvements in therapy
that low-intensity ultrasound could control the extent of tissue necrosis, for infected burns in the post-antibiotic era and better skin regeneration
enhance granulation tissue formation, as well as accelerate wound [148]. In this section, we review new nanotechnology-based approaches
42 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

for of burn infection treatment and healing of wounds. NSs have shown inflammatory cell functions. In addition, CS NPs are used as compo-
impressive antimicrobial activities against resistant strains of microor- nents of dressings and bandages designed for skin wounds to sup-
ganisms, and are also capable of accelerating regeneration of burn- port the mechanical strength of the wound, and increase the
injured skin [149,150]. NSs utilized in burn therapy can be classified as wound healing rate both in humans and animals [169,173]. In anoth-
two main groups: organic NPs and inorganic NPs; each of which con- er research, CS NPs were employed for the preparation of films in
tains diverse types of NPs utilized for burn infections infection and combination with alginate, and loaded with SSD cream to treat in-
wound healing [151,152]. Organic NSs include polymeric NPs, fected open wounds [174]. Curcumin NPs, are formed from curcumin
nanoemulsions, nanogels, liposomes, micelles and solid lipid-based (40–200 nm), which is a natural yellow pigment with a
NPs (SLBNs), while inorganic NSs contain nanocarbons, AuNPs, CuNPs, diarylheptanoid structure, an example of curcuminoid-related mole-
AgNPs, TiO2 NPs, magnetic NPs (MNPs) and quantum dots (QDs). cules extracted from turmeric, a spice prepared from the root of
Nanocarbons comprise fullerenes, carbon nanotubes (CNTs), graphene, Curcuma longa, which has been exploited in traditional medicine
nanodiamonds, etc. [150,153]. Furthermore, in recent years, NS-based for centuries [175]. Encapsulation of curcumin into NPs is an advan-
structures such as nanofibers (NFs), 3D-scaffolds and films have been tageous delivery method for this water-insoluble substance. In one
developed for wound therapy, which also will be discussed in the fol- study, curcumin NPs were found to have strong antibacterial activity
lowing sections. to suppress the growth of P. aeruginosa and S. aureus, and treat skin
infections in mouse wounds [176]. In other studies, it was found
6.1. Therapy of burn wound infections using NSs with antimicrobial effects that curcumin NPs could reduce inflammation and induce cell ex-
pansion that was useful in reconstruction of damaged tissue
6.1.1. Organic NPs [177–179].
Polymeric NPs can be fabricated through implementing various ma-
terials, and methods. They are conventionally synthesized via self- 6.1.1.1. Dendrimers. Dendrimers are a type of nano-scale (1–10 nm)
assembly of ingredients such as chitosan (CS), curcumin, poly lactic- polymeric macromolecule with a homogeneous and monodisperse
co-glycolic acid (PLGA), as well as having different shapes including structure, which have applications in both diagnosis and therapy.
dendrimers, hydrogels and Nanomicelles (NMs) [154]. Polymeric NPs Dendrimers possess a symmetric core, an inner shell and an outer
have been utilized in different biomedical applications [155,156]. They shell. Dendrimers can be prepared from phenyl acetylene subunits
are usually synthesized by self-assembly of charged polymers mediated [180,181]. Moreover, the surface functional groups of dendrimers can
by interactions between chains with cationic and anionic groups. This is act as antibacterial agents themselves (especially if cationic). In one
a relatively mild process and will not damage cargo components such as study, the interaction of the positively charged groups of a dendrimer
proteins that would be deactivated by high temperatures and organic (quaternary ammonium groups) and the negatively charged groups of
solvents. These methods use an emulsification process to synthesize the bacterial cell wall could lead disruption of the bacterial structure.
polymeric NPs. Such methods can be used as a strategy for production Moreover water-soluble dendrimers such as polyamidoamine -based
of others polymeric NPs incorporating therapeutic nucleic acids, pep- dendrimers can be loaded with water-insoluble antibiotics (e.g.
tides or proteins inside the NPs [157]. KGF is a potent growth factor in- nadifloxacin and prolifloxacin), which increases their bioavailability
volved in re-epithelization of skin wounds [158]. In two recent and enhances their antibacterial properties [181]. In another approach,
investigations it was shown that that KGF contained in self-assembled silver-dendrimeric NSs were found to have synergistic effects in terms
NPs could stimulate healing of skin wounds, giving increased re- of anti-inflammatory and antimicrobial activity; dendrimers were
epithelization, better tissue remodeling and skin regeneration [159, shown to be an ideal carrier to diminish inflammation, while
160]. PLGA is a FDA approved material used in the production of poly- disinfecting the wound and stimulate healing [182].
meric NPs (100–200 nm) due to its advantages including biodegradabil-
ity, biocompatibility and non-toxicity [161]. PLGA NPs are normally 6.1.1.2. Hydrogels. Hydrogels as polymeric networks are regarded as hy-
fabricated by emulsification of hydrophobic components, using an or- drophilic three-dimensional structures, which potentiate absorption of
ganic solvent and various surfactants [162]. In one study, PLGA was large amounts of aqueous fluid. Because they have high porosity and a
used to manufacture EGF-loaded NPs for wound healing. Full- soft consistency, they can be used for dressings and to help tissue regen-
thickness wounds treated with EGF-loaded PLGA-NPs gave the highest eration [183]. These materials are ideal for wound dressings, because
level of fibroblast proliferation and the fastest healing [163]. Re- they can protect the injured tissues make the patients more comfortable
searchers have reported that PLGA could provide a biocompatible sys- as they exert a cooling function, and their non-adhesive nature pre-
tem for delivery of growth factors. In one study, the delivery of the serves the wound tissue [184]. Recent advances have shown that hydro-
host defense peptide called LL37, incorporated into PLGA NPs reduced gel NPs combined with other components including CS, keratin, PVA,
the release of lactate and accelerated wound healing [164]. Studies PEG, AMPS, PEGDA, self-assembled peptides, dextran, hyaluronan, col-
have clearly indicated that PLGA- NPs loaded with LL-37 antimicrobial lagen, fibrin and antibiotics can induce skin regeneration in burns
peptide could be utilized as an effective agent against (polymicrobial) [185]. In one study, the antimicrobial activity and scar preventive
infected wounds [164,165]. CS NPs (150–300 nm) usually are made of (antifibrotic) properties of hydrogels were optimized for wound dress-
pristine soluble CS by using ionic gelation methods. CS can be employed ings using a coating comprised of a combination of PVP, PEG and CS
as a natural and safe biodegradable substance in the design of drug/gene coated onto a cotton fabric. A model drug, tetracycline hydrochloride
delivery systems [166]. Recently, CS due to considerable advantages has was loaded in the hydrogel structure. The Cumulative drug release
been utilized in burn wounds, and fabrication of haemostatic dressings, was indicated to be 80% over 48 h, followed by improved antimicrobial
bandages, etc. In addition, CS can also be used as a prophylactic agent effect of the dressings against both Gram-positive and Gram-negative
that prevents the progression of infections, and has also been described bacteria, and minimal scarring when the dressing was applied to
to produce significant acceleration of wound-healing [167–169]. Be- wounds [186]. Hydrogels also possess other features making them sub-
cause CS is a cationic polymer, CS NPs have been used as intrinsic anti- stantially invaluable for tissue regeneration and engineering. A 3D
microbial agents for prevention of microbial infections [170,171]. network-like matrix composed of hydrogel behaves more like natural
Many studies have reported that CS NPs and analogs could facilitate biological ECM to allow cell growth, compared to cells seeded on two-
wound treatment process by increasing the function of inflammatory dimensional substrates [187]. The nanoplatforms can be modulated to
cells in the wounds, and stimulating fibroblasts and osteolasts [172]. release various bioactive molecules such as antibacterial agents, growth
In two independent studies, it was shown that CS NPs could facilitate factors, and cytokinesin tissue damage site, as well to assist beneficial
coagulation by attaching to red blood cells, and improve implementation of exogenous epithelial cells, fibroblasts or stem cells
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 43

[188,189]. Recently, basic fibroblast growth factor (bFGF) transport by made from spherical solid lipids can also include hydrophilic molecules
gelatin-based hydrogels, and their subsequent release were reported such as PEG derivatives, and can be stabilized using surfactants. These
to help the healing of acute vocal fold damage without scarring. Using vehicles have been suggested to be as an alternative to, microparticles,
this bFGF hydrogel-based treatment enhanced tissue regeneration liposomes, and emulsions. It has been also shown that coating SLNs
[190]. In another study, ionic micellar NPs were designed using chitosan with hydrophilic agents enhances their plasma stability and improves
with hydrophobic modifications, combined with fatty acids, which un- the bioactivity of drug-loaded SLNs [204]. Several types of drugs can
dergo self-assembly in aqueous medium. Results indicated biocompati- be delivered to chosen organs with high specificity using ligand-
bility of the NPs, successful encapsulation of a poorly-soluble drug targeted or stimulus-responsive SLNs such as antibody-SLN conjugates,
(clarithromycin), and the ability to be administered both parenterally magnetic-SLN, pH sensitive SLN and cationic-SLN [205]. In one study,
and topically. Thus, this nanoplatform was suggested to be beneficial SSD-loaded-SLNs were tested for prolonged release of the cargo, and
for skin regeneration and wound healing [191]. to reduce the pain in severe skin wounds. In addition, the use of SLNs in-
creased cell migration and improved wound closure rates [206]. Anoth-
6.1.1.3. Nanoemulsions. Nanoemulsions (NEs) are made of oil-based and er study used a novel cell-based 3D–wound treatment approach,
aqueous solutions forming nano-sized particles in the range of showing that empty SLNs facilitated re-epithelialization with low cyto-
20–200 nm [192]. NMs are generally made of amphiphilic molecules toxicity and no irritation. Moreover these NPs could also encapsulate
that adopt a spherical form in aqueous solutions [193]; they have two and allow release of morphine in a wound dressing [207]. In another in-
parts including a hydrophobic core as well as a hydrophilic coating. vestigation, SLNs loaded with nitrofurazone acted as a delivery vehicle
The hydrophobic core can be efficiently loaded with insoluble agents, to get topical drug into the burn eschar, to improve the antimicrobial ac-
and the hydrophilic coating allows the NPs to be soluble in aqueous tivity of nitrofurazone for treatment of infection in burn patients [208].
media [194]. The aqueous phase of NE contains water and co- The second generation of SLNs, called nanostructured lipid carriers
surfactants e.g. glycerin and ethyleneglycol. NEs can be produced via (NLCs) are comprised of both solid and liquid components, and have a
mixing the constituents using ultrasonication and stirring. In one higher drug-loading capacity and better stability [209,210]. NLCs are
study, chlorhexidine acetate formulated in a nanoemulsion was used comprised of physiologically biocompatible, and biodegradable and
against a methicillin-resistant S. aurerus (MRSA) infection in a skin lipids and surfactants [211]. NLCs can be used to deliver highly lipophilic
burn wound model. Here, the nanoemulsion showed effective and drugs into targeted organs, and damaged tissues such as burn wounds
rapid activity against MRSA in-vitro and in-vivo and hindered formation [212]. Miconazole-encapsulated NLCs were recently used to enhance
of biofilm, disrupted MRSA cell walls, led to increased leakage of DNA, miconazole activity (a poorly-water soluble antifungal agent). It was re-
protein, Mg2+, K+ and alkaline phosphates out of the cells [195]. ported that the miconazole-encapsulated NLCs displayed enhanced an-
In another study, a CS film was impregnated with NEs encapsulating tifungal activity against Candida albicans yeast cells [213]. In recent
eucalyptus oil, produced by an emulsification method, which showed a studies, emulsified recombinant human (rh)EGF-loaded into NLCs by
good antibacterial effect against S. aureus. The mechanism of action of ultrasonication was indicated to improve wound healing, as well to
this eucalyptus oil-NE-CS film was attributed to cell membrane damage, stimulate fibroblast expansion and collagen production [214,215].
and wound healing efficacy to a non-irritant activity in the wounds These rhEGF-loaded NLCs are potential candidates for healing of chronic
[196]. NMs made via self-assembly have been recently used to encapsu- wounds and burns. In a study by Gainza et al., dressings were fabricated
late hydrophobic drugs to improve their solubility [197]. Recently, ionic in the form of fibrin-based solid scaffolds or semi-solid hydrogels, and
micelles have been fabricated using ionically-modified CS, and loaded loaded with lipid NP-encapsulated rhEGF. The growth factor underwent
with SSD as an antimicrobial. These micelles were also loaded with sustained release into wounded skin. The fibrin-based scaffolds were
platelet lysate and used for treatment of chronic burn wounds. suggested as the best approach for rhEGF delivery due to their superior
Unloaded CSs demonstrated good biocompatibility against fibroblasts. prolonged stability [216].
High concentrations of SSD could be obtained using the micelles as
well as reduction in the cytotoxicity and bio-incompatibility of SSD to- 6.1.2. Inorganic NPs
wards fibroblasts, and improvement in the activity PDGF-AB (platelet
derived growth factor (PDGF-AB) [198]. 6.1.2.1. Metallic NPs. Metallic NPs mostly consist of AuNPs, CuNPs, and
AgNPs have been employed in wound healing researches. Recently,
6.1.1.4. Liposomal NPs. Liposomal NPs consist of spherical vesicles made the therapeutic activity of phytochemical-decorated AuNPs (Phyto-
of one or more lamellar lipid-bilayers that spontaneously self-assemble AuNPs) was studied for treatment of surgical wounds and burn wounds
when amphipathic precursors are mixed. Unilamellar liposomes have a in a rat model. The Phyto-AuNPs were shown to act as an efficient anti-
single shell, while multilamellar liposomes have several concentraic oxidant that could stimulate high expression of the pro-angiogenic
shells. These bilayers can be produced from cholesterol and naturally agents (VEGF and Ang-2) thus improving skin regeneration. These
occurring phospholipids including phosphatidylcholine [199]. One Phyto-AuNPs were also suggested to be useful for healing of burn
study reported that dihydroquercetin formulated as a liposomal wounds [217]. In a similar study, cryopreserved human fibroblast cells
nanocomplex could improve endogenous antioxidant activity, reduce (CrHFC) cultured with AuNPs were used to treat third-degree skin
the necrotic area in burned skin, and finally induce better healing burns in white male rats. CrHFC-AuNPs facilitated skin repair and im-
[200]. Elsewhere, the use of other liposomal components e.g. DOTAP/ proved collagen production by the third week after treatment [218]. In-
cholesterol for encapsulating DNA ending the IGF-I and KGF genes into terestingly, a novel modifiable collagen-gelatin fleece also improved
dermis and epidermis was reported to be an effective method to im- burn wound healing without any added AuNPs [219]. In another
prove epithelial cell differentiation and expansion [201,202]. study, AuNPs-incorporated collagen scaffolds (AuNP-SCs) were de-
SLBNs have been tested as potential drug delivery systems (DDSs) signed and cross-liked with glutaraldehyde. Use of AuNPs, allowed the
that could be topically administered to the skin and eyes, or else deliv- scaffolds to have good biocompatibility, high mechanical strength, en-
ered by parenteral, oral, and inhalational routes. SLBNs can be classified hanced stability against enzymatic degradation, and reduced hydrolytic
into two main groups including solid lipid NPs (SLNs) and nanostruc- activity, compared to non-AuNP-incorporated scaffolds. Cross-linked
tured lipid carriers (NLCs). Various methods have been developed for AuNP-SCs inhibited inflammation and greatly improved the granulation
synthesis of these NPs including microemulsion, solvent emulsification, tissue generation, and produced more neovascularization. Wound clo-
evaporation, and high pressure homogenization [203]. sure was improved for both cross-linked AuNP-SCs and non-Au contain-
SLNs are solid colloidal drug delivery vehicles at room temperature, ing scaffolds and the AuNP-SCs were suggested as a potent skin
made of solid lipids, instead of the liquid lipids that are used in NEs. SLNs substitute for coverage of wounds [220]. Cu is considered to possess a
44 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

results showed AgNPs were localized in the cytoplasm of fibroblasts


with no cytotoxicity evident [229]. The presence of normal human epi-
dermal keratinocytes (NHEKs) and normal human dermal fibroblasts
(NHDFs) is essential for good wound healing, but because they synthe-
size interleukin 12 (IL-12), TNF-α, and VEGF, they can cause excessive
inflammation in the first two days. AgNPs could decrease the secretion
of VEGF and pro-inflammatory cytokines. The results indicated NHEKs
were more susceptible to AgNPs than NHDFs [230].

6.1.2.2. Carbon-based NPs. Carbon-based NPs include CNTs, fullerenes,


graphene, nano-diamonds and so forth. CNTs have recently been stud-
ied as novel drug and gene delivery platforms [231]. The molecular
structure of CNTs is extremely hydrophobic, and CNTs are highly
prone to aggregation. Nevertheless they can be attached to biomole-
cules by covalent and non-covalent bonds and used as delivery vehicles.
CNTs have also been explored as potential antimicrobial agents with an
intrinsic antibiotic function [232]. In one research, Cu/Zn bimetallic NPs
were combined with carbon nanofibers (CNFs) as a novel antimicrobial
agent for wound dressing and burn healing. This antibacterial platform
suppressed the growth of Gram-negative bacteria and Gram-positive
bacteria e.g. E. coli and methicillin-resistant MRSA, respectively [233].
In one study, SWCNTs showed effective bactericidal activity against bac-
Fig. 4. a) The platform based on GQDs/low level H2O2 used as antibacterial agent, b) The teria e.g. Salmonella enterica and E. coli. Here, SWCNTs caused the aggre-
GQD equipped bandage for disinfection of the wound site in vivo.
gation of bacterial cells and led to death of the microorganisms.
Reprinted with permission from ref. [239], copyright 2014, American Chemical Society.
Fullerenes have also shown an intrinsic antimicrobial function against
different bacteria such as E. coli, Salmonella and Streptococcus spp. This
broad-spectrum antimicrobial activity against bacteria, yeasts as well as antibacterial activity may be due to inhibition of energy metabolism
viruses [221]. Despite its beneficial properties, Cu can damage proteins and impairment of respiration by entry of the NPs into the cytoplasm
(especially lipoiproteins), can cause rapid oxidation reactions in unsat- [234].
urated lipids, therefore there is a possibility of Cu causing cytotoxicity Graphene is defined as a single monolayer of carbon atoms forming a
and tissue damage. Thus the use of innovative nanotechnology-based two-dimensional crystal, with derivatives such as graphite oxide (GO).
methods are essential to diminish the side effects of Cu, while still GO is hydrophilic (while graphene itself is highly hydrophobic) and
allowing it to act as a powerful antimicrobial agent. Recently, one can be produced by chemical modification of graphene. Graphene and
study showed that CuNPs were effective antimicrobial agents against GO NSs have been reported as appropriate carriers of drugs while show-
E. coli, S. aureus, Micrococcus luteus, K. pneumoniae, P. aeruginosa as ing antimicrobial activity against microbes such as E. coli., S. aureus,
well as fungi such as Aspergillus flavus, Aspergillus niger and Candida P. aeruginosa and C. albicans, besides can be exploited for fabrication of
albicans: all of these species are commonly found in burn wounds wound dressings and also for water disinfection. In one study, it was
[222]. A similar study examined Cu-encapsulated-CS-NPs for antimicro- reported that GO NPs could interact with bacteria, causing RNA efflux,
bial applications. As mentioned previously, CS is a natural antimicrobial and could disrupt cell membranes of Gram-negative bacteria and
polymer, that could exert synergistic activity between Cu and CS NPs Gram-positive bacteria [236]. The povidone-iodine complex (PVPI)
[223]. has a potent antiseptic activity against several microorganisms e.g.
For decades silver nitrate and SSD have been utilized as topical anti- E. coli. In one study, SWCNTs were coated with PVPI to produce water-
microbial agents in burn wound treatment. Recently nano-scale silver- soluble and stable structures with monolayer polymeric helical coil
containing platforms such as AgNPs, and Ag-containing hydrogels morphology. Eventually, a composite film composed of these NSs
have been applied in burn wound therapy. Two recent studies found served as a conductive bandage for skin wounds with improved flexibil-
that hydrogels containing AgNPs were a potent alternative to SSD for ity, besides demonstrating the slow-release of antiseptic iodine [237].
healing of burn wound infections, without causing cytotoxicity [224,
225]. In another study, AgNPs were used as an antimicrobial material 6.1.2.3. QDs. QDs are semiconductor nanocrystals with a three-
for healing of skin wounds, where also showed a good antimicrobial ef- dimensional structure and a tunable luminescence emission in any
fect and the capability to be used in wound dressings [226]. In a similar color, that are widely as fluorescent dyes and reporters in biomedicine
study, AgNPs loaded with additional components such as enoxaparin (a [238]. Graphene QDs (GQDs) were shown to have an intrinsic
low molecular weight heparin used as an anticoagulant) were used to peroxidase-like activity, whereby they functioned as a catalyst similar
induce anti-inflammatory and angiogenic activity for wound treatment. to an enzyme. The GQDs were able to generate hydroxyl radicals from
The enoxaparin-loaded AgNPs could shorten the healing time of the hydrogen peroxide. Therefore, since •OH has a potent antibacterial ac-
wounds [227]. In a similar study, cotton fabrics containing stable tivity, the reaction of H2O2 to produce •OH dramatically increased the
22 nm diameter AgNP suspensions were used as wound dressings and antibacterial function of H2O2 meaning that high concentrations H2O2
with effective antimicrobial and anti-inflammatory functions. Here, in the wound were not needed and host toxicity was reduced (Fig. 4)
250 ppm AgNP-containing cotton fabric demonstrated good anti- [239]. In another research, antibiotic NSs were fabricated comprised of
microbial activity compared to fabric containing less (60 and AgNPs combined with GQDs coated with PEGylation and produced by
125 ppm) AgNPs. Furthermore, using AgNPs led to same healing conse- a laser synthesis process. The antibacterial activity of Ag-GQD com-
quences in comparison with Dermazin® cream (SSD), and an anti- plexes was evaluated against bacteria including S. aureus and
inflammatory effect, the same as indomethacin drug with dose of P. aeruginosa. Ag-GQDs acted as an antimicrobial agent with synergistic
20 mL [228]. In another study, an AgNP-based dressing with 3D- activity of its two components [240].
cultured fibroblast cells was used for full-thickness skin damage in
burned patients. The results indicated that AgNPs reduced mitochondri- 6.1.2.4. Ceramic NPs. Ceramic NPs include oxides, carbides, phosphates,
al activity without activating cell death mechanisms, as well In vivo carbonates, calcium, and generally have porous structures such as
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 45

silicon oxide (silica) and aluminum oxide (alumina) [241,242]. They


have a wide range of industrial, diagnostic, and therapeutic applications pH and temperature dependent process, stimulated wound healing in
because of their high heat resistance and chemical inertness. Targeted rats [251]. In a similar research, films equipped with green TiO2 NPs, iso-
ceramic NPs have been explored as potential carriers for drugs, genes, lated from the plant Psidium guajava, exhibited an antibacterial and an-
proteins, imaging agents and photosensitizers in a broad span range of tioxidant mechanism against E.coli and S.aureus [252]. In another
biomedical applications. Ceramic NPs have also been utilized as drug de- investigation, the antimicrobial activity of TiO2 NPs and the related
livery platforms to enhance antimicrobial activity against pathogens mechanism of action against P. aeruginosa was studied [253].
[243,244]. In one study, the antibiotics gentamicin and rifamycin were
loaded into silica NPs immobilized inside collagen-hydrogels, and 6.2. Nanofibers
were used to treat wound infections and to evaluate their antibacterial
activity against P. aeruginosa and S. aureus wound pathogens. Results in- Nanofibers (NFs) are fabricated from natural and synthetic continu-
dicated the gentamicin-loaded silica NPs embedded in collagen ous polymer chains. Nanoscale biodegradable NFs can enhance cellular
hydrogels could significantly improve wound healing and demonstrat- and drug interactions with scaffolds used in tissue regenerating and en-
ed a long-lasting antibacterial activity, with low cytotoxicity toward fi- gineering as well anti-microbial drug delivery applications. NFs with a
broblast cells. On the other hand, the rifamycin-loaded silica NPs in diameter near 5 nm, can emulate collagen in size and the fibril structure
collagen hydrogels showed no bactericidal activity, because of intense that exists in the ECM. Hence, these NFs have been applied as beneficial
absorption of rifamycin onto the collagen fibers and no subsequent re- nano-textured materials and scaffolds in burn wound care. NFs can be
lease [245]. As mentioned above NO is an antimicrobial agent that can fabricated with diverse pore sizes, diameters and density [254,255].
prevent infections developing in burn wound sites [246]. Recent studies Electrospinning is a well-known technique to produce electrospun
have assessed the potential of NO-loaded silica NPs to act as an antimi- biodegradable NFs [256]. In this technique, a charged liquid jet of sub-
crobial agents against various microorganisms in burn infections. Deliv- strate is injected into a quickly rotating collector. Subsequently, the
ery of high doses of NO-loaded silica NPs has been shown to inhibit NFs is fabricated taking advantage of an electrostatic charge imbalance
fibroblast proliferation compared to clinically relevant doses of NO. Fur- that overcomes the surface tension of the solution and the solvent is
thermore, these NO-NPs have been shown to have antimicrobial activity evaporated [257]. Regarding the high area to volume ratio of
against biofilms [246,247]. TiO2 NPs show antibacterial activity upon UV electrospun NFs, they can increase transfer of various molecules e.g. an-
irradiation due to ROS generation by the process known as tibacterial agents, anticancer drugs, nucleic acids, and diverse growth
photocatalysis. The aggregates/agglomerates of TiO2 NPs N 100 nm in di- factors, as well improve cellular attachment to tissues. Thus, electrospun
ameter could not photo-inactivate E. coli and Saccharomyces cerevisiae, NFs have been extensively employed in the form of NFs scaffolds used in
but conversely were able to act as a “sunscreen” and protect the micro- tissue regeneration and tissue engineering [258]. In a study, Jayesh et al.
bial cells from inactivation by UV light alone [248,249]. In one study, loaded the iron chelator 2,3-dihydroxybenzoic acid (DHBA) within poly
ZnO and TiO2 NPs demonstrated a strong inhibition against some spe- D, L-lactide (PDLLA)-poly ethylene oxide (PEO) NFs (called DF) created
cies of bacteria including MRSA biofilms [250]. In another study, TiO2 by electrospinning. They used DF for therapy of chronic infections in-
NPs were applied as an antibacterial agent against Gram-negative and duced by P. aeruginosa biofilms in wounds. Thus it was found that DF
Gram-positive bacteria to improve wound treatment. Furthermore, could decrease biofilm creation by nearly 75% in 8 h and could be incor-
the “green-synthesized” TiO2 NPs extracted from Moringa oleifera in a porated into a wound dressing to cure P. aeruginosa chronic infections.

Fig. 5. Curves showing morphological measurements evaluations from mice treated with SHAM group, GEL (hydrogel), GEL/ASCs at the burn wound site: a) wound size measurement; 1, 2,
and 3 weeks after burn injury initiation compared to initial injury. Here, no effect of the gel wound dressing and wound closure was reported, b) and c) wound thickness (mm) after 1 and
2 week, and area occupied by nuclei, respectively, indicative of granulation tissue, which was facilitated by ASC-containing gels, c) Quantitative measurement of blood vessels in
regenerating regions 1 week after the burn initiation. SHAM group (untreated), GEL group (treated only with P-fibrin gel), and GEL/ASCs group (GEL group integrated with adipose-
derived stem cells).
Reprinted with permission from ref. [267], copyright 2016, John Wiley & Sons, Inc.
46 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

Fig. 6. Schematic of the synthesis procedure of QCSP/PEGS-FA hydrogels.


Reprinted with permission from ref. [277], copyright 2017, Elsevier.

Here, DHBA was released from DF for 4 h, comparable to the time need- NFs can be also fabricated by thermally induced phase separation
ed for bacterial growth inhibition [259]. (TIPS). In a study Li et al. generated a silk fibroin-PLGA NF scaffold (to
Copper can be incorporated in the regeneration and healing of dam- mimic the ECM) by the TIPS method for a wound dressing. This struc-
aged tissue. Cu can increase integrin expression (as an adhesive mole- ture showed the highest equilibrium water uptake rate of 72.2% and a
cule), collagen fabrication and fibrinogen stabilization [260]. More water vapor transmission rate of 5591.88 g/m2 per day. Furthermore,
recently, a copper containing NF structure (326 ± 149 nm in diameter) the optimum properties of silk fibroin-PLGA NF scaffolds including
fabricated via electrospinning of PDLLA and poly-ethylene oxide PEO water vapor permeability, water uptake rate/loss rate (to absorb
was shown to inhibit S. aureus and P. aeruginosa biofilm formation, by wound skin humidity for suppression of wound infection and protec-
41% and 50%, respectively [261]. tion of damaged skin humidity) were achieved at the weight ratio of

Fig. 7. a) Schematic illustrating the fabrication process of the biocomposite wound dressing, b) microscopic images of 8 mm full thickness wounds of C57BL/6J mice on day 21 after the burn
injury initiation: (i) Spontaneous healing, (ii) OP-Gel, (iii) OP-Gel-NS, (iv) OP-Gel-Cipro, and (v) Bactigras, c) Histological analysis of the excised tissue at wound site: (i) intact tissue at first
day, treated tissue after 3 weeks under different conditions of:(ii) Spontaneous healing, (iii) using OP-Gel, (iv) using OP-Gel- NS, (v) using OP-Gel-Cipro and (vi) using Bactigras
(commercially available dressing).
Reprinted with permission from ref. [278], copyright 2016, Elsevier.
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 47

8/8. The scaffolds were also reported to show sustained release of early mononuclear cell recruitment, but did not show any negative ef-
curcumin (as a model drug) [262]. fects on wound closure. Fig. 5 shows the measurements of wound size
Shape memory polymer–based NFs can be designed to control drug 1, 2, and 3 weeks after initiation of burn injury (Fig. 5-a), the wound
release, and as smart wound healing dressings are able to be applied for thickness 1 and 2 weeks after initiation of burn injury (Fig. 5-b), the
chronic wound healing and regeneration of damaged skin [263,264]. In area occupied by nuclei 1 week after the injury initiation (Fig. 5-c) and
a study, Tan et al. generated a multifunctional and smart electrospun- of blood vessel quantification in regenerating areas of the wound site
based NF for wound repair. They fabricated the NF from chitosan, gelatin 1 week after the initiation of the burn injury (Fig. 5-d) [267].
and shape-memory polyurethane, loaded with silver nitrate (AgNO3). Bacterial cellulose (BC) is defined as a natural polysaccharide with
The antibacterial activity of the NF was evaluated against bacteria in- well-known beneficial properties such as biocompatibility, biodegrad-
cluding E. coli, P. aeruginosa and S. aureus. The results demonstrated ability, mechanical strength, nanofibrillar structure, moldability, water
higher antibacterial activity against E. coli and P. aeruginosa compared absorbency, porosity, and noticeable biological affinity in general. The
to S. aureus [265]. ability of BCs to play a role in the fabrication and construction of 3D-
In a recent research, dendrimers (as branched monodisperse nano- scaffolds applied for tissue regeneration and healing has been reported
scale molecules) were incorporated in a NF structure due to properties [268,269].
such as biocompatibility, flexibility, solubility, chemical stability, etc. In the literature, various reports of a diverse assortment of films/
Here, Dongargaonkar et al. designed electrospun blends, comprised of membranes acting as wound healing dressings have been published.
gelatin and highly branched star-shaped polyamidoamine dendrimer For example Cu2 +-loaded bioactive glass/eggshell films [270], NO-
covalently conjugated to gelatin. Silver acetate (0.83, 1.65, and 3.30% releasing CS films [271], and Ag/ZnO-microporous CS composite dress-
w/w) as an antibacterial agent was loaded into the constructed NFs. Sil- ings [272]. All these have resulted in substantial improvements in anti-
ver was shown to be significantly released at 48 h. antibacterial activity bacterial activity, angiogenesis, re-epithelialization, skin reconstruction,
of the NF was evaluated on S. aureus and P. aeruginosa at 37 °C for 4, 24, blood clotting ability, and so forth leading to improved wound healing.
and 48 h. In control groups including a blend of gelatin and silver ace- Particularly concentrating on burn wounds, miscellaneous dressings
tate, S. aureus and P. aeruginosa showed rapid growth for 4 and 24 h have been reported such as Ag+ sulfadiazine-containing nanosheet-
while in active groups, S. aureus and P. aeruginosa growth was complete- based ultra-thin films [273], NO-releasing polymeric films [274], tyro-
ly inhibited by silver acetate released from the constructed NF in 4 h. thricin (cyclic polypeptide antibiotic)-containing CS-based film-
Thus, dendrimers covalently associated to gelatin both increased load- forming gel layers [275] with particular advantages including insignifi-
ing capacity as well bestowed immense flexibility to NFs as a candidate cant cytotoxicity, high activity against burn-wound bacterial pathogens,
for dressing materials [266]. blocking bacterial invasion, inhibition of inflammation, reduced scar-
ring, enhanced epithelialization and formation of granulation tissues,
6.3. 3D-scaffolds, films and wound dressings and so forth. Namazi et al. fabricated a carboxymethylcellulose hydrogel
that incorporated MCM-41 mesoporous silica nanoparticles (MSN)
New platforms including 3D-scaffolds, engineered films and hi-tech nanocarriers, loaded with tetracycline and methylene blue. The MCNs
wound dressings are regarded as promising materials for tissue engi- helped control the hydrogel swelling and provided controlled drug re-
neering, and improved results have been reported in wound healing lease, potentiating the antimicrobial effects against S. aureus. These ben-
studies. For example, ASC-containing PEGylated fibrin-based gel scaf- efits suggested that the time intervals between changing the
folds used as a construct for wound coverage were shown to improve antimicrobial bandage could be elongated [276]. Zhao et al. also report-
burn wound healing. Using this scaffold, ASCs were still present at the ed an electroactive injectable hydrogel wound dressing for cutaneous
wound site 14 days after wound initiation. The scaffold enhanced the wounds with properties such as antibacterial activity, antioxidant,
vascularization and attracted mannose receptor-expressing cells into self-healing, improved hemostasis and adhesiveness. Fig. 6 illustrates
the wound, improved the generation of granulation tissue as well as the synthesis process of these hydrogels [277].

Fig. 8. a) Scanning electron microscopy of RADA16, (b) RADA16-FPG and (c) RADA16-RGD. (d) Cell viability result for the implemented keratinocyte (i.e. HaCaT) and fibroblast (i.e. NIH/
3T3) using the three 1% w/v RADA16 NF scaffolds and cell control after 0 h and 3 days of incubation.
Reprinted from ref. [288], copyright 2014, Nature (licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License).
48 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

Fig. 9. a) Fluorescence micrographs of cells showing clustered growth after days 3 on HEMA (left; scaffolds coated with fibronectin were seeded with NIH/3T3 mouse fibroblasts); with
homogenous growth of the cells cultured on pH-sensitive DMAEMA/HEMA scaffolds (right) [299], b) percentage swelling of zinc ion containing hydrogels; c) TA release from the
hydrogels, at various pH values Reprinted with permission from ref. [300], copyright 2016, American Chemical Society; d) Scheme illustrating the synthesis of enzyme-sensitive self-as-
sembled polymeric vehicles (from PEG-b-PP and PEG-b-PC) for selective delivery of antibiotics. They were designed to be responsive to penicillin G amidase (side chain cleavage) and to β-
lactamase (microstructural transformation). This stimulus-responsive nanoplatform resulted in the constant cargo liberation and bioactivity of the encapsulated antimicrobial agent.
Reprinted with permission from ref. [302], copyright 2015, John Wiley & Sons, Inc.

Recently, a wound-dressing platform was developed based upon ox- mechanically strong core and used collagen as a biocompatible shell.
idized pectin-gelatin nano-hydrocolloids (OP-Gel). These were pre- In this NF scaffold, vitamin A palmitate was encapsulated in the PCL
pared by dip coating technique, consisting the entrapment of silver core as a pharmaceutical to accelerate wound healing, and Ag-NPs as
NPs (NS-OP-Gel) or ciprofloxacin (Cipro-OP-Gel) inside the hydrogel an antibacterial material was integrated in the collagen shell. The en-
(Fig. 7-a). The results (compared to commercially available Bactigras® capsulated Ag-NPs demonstrated antibacterial activity against
chlorhexidine dressing) indicated high antimicrobial activity, enhanced S. aureus, and nearly 70% of loaded vitamin A palmitate was released
neovascularization, stimulated cell migration, better-organized deposi- after 6 days. In addition, L929 cells (mouse fibroblasts) attached well
tion of collagen, and good hydrophilicity for both NS-OP-Gel and Cipro- on the PCL-collagen core-shell NFs after 5 days [283].
OP-Gel dressings. The NS-OP-Gel showed enhanced antibacterial func- Three-dimensional (3D) NFs scaffolds are considered to be a prom-
tion (at very low loading of 3.75 mg per cm2) and Cipro-Op-Gel (at 1% ising approach in tissue engineering to emulate ECM [284,285]. In one
drug loading) against E. coli and S. aureus. NIH3T3 fibroblast prolifera- study, Park et al. designed a 3D electrospun-based silk-fibroin NF scaf-
tion was inhibited most by Bactigras® slightly by NS-OP-Gel and if not fold using freeze drying, and salt-leaching followed by electrospinning
all by Cipro-OP-Gel. Fig. 7-b shows the microscopic images of full thick- with NaCl. Moreover, human skin keratinocytes (HaCAT) and fibro-
ness wounds of C57BL/6J mice on day 21 after the burn injury. The his- blasts (NIH 3T3) were co-cultured in 3D silk-fibroin NF scaffolds to
tological analysis of the excised tissue at wound site is also shown in Fig. mimic a bilayered skin structure in vitro. Their finding showed im-
7-c. Healing rate was enhanced by NS-OP-Gel, while there were similar proved fibroblast proliferation in the internal layer of electrospun/
rates for Cipro-OP-Gel and Bactrigas® [278]. NaCl based-scaffold compared to freeze dried and salt-leached 3D scaf-
Microspheres can be also employed to prepare wound dressings. folds. Better keratinocyte differentiation was demonstrated in the exter-
Romic et al. showed the ability of CS/Pluronic® F127 microspheres load- nal layer of salt-leached 3D scaffolds than that prepared by freeze dried
ed with melatonin to serve as wound healing dressings A good and electrospun with NaCl methods [286].
sustained release of melatonin, showed an antibacterial effect against Self-assembled NFs produced from self-assembled peptides (SAP)
S. aureus and clinical isolates of MRSA, along with high biocompatibility can be employed to fabricate self-assembled hydrogels as scaffolds, ap-
with keratinocytes (e.g. HaCaT) and fibroblasts (e.g. BJ) [279]. plied for the proliferation and regeneration of skin cells in wound
Nanofibers have also been investigated for wound therapy as dress- healing [287]. In one report, Bradshaw et al. generated hydrogel NF scaf-
ings or membranes, and so forth. In a recent study, N,N,N-trimethyl CS folds from RADA16 (1% w/v) self-assembled peptide. They studied the
fibers were used as wound dressings and showed good antibacterial ac- effect of three peptides including RADA16, RADA16 containing a fibro-
tivity against S. aureus (N 99%) and E. coli (N 63%), while showing a lack of nectin motif (RADA16-GG-RGDS) and RADA16 containing a collagen
toxicity to mouse embryonic fibroblast cells at low concentrations. In- type I motif (RADA16-GG-FPGERGVEGPGP) on the migration and ex-
vivo tests revealed improved re-epithelialization and better wound con- pansion of immortalized human keratinocytes as well as mouse embry-
traction [280]. Elsewhere, electrospun SF nanofiber membranes were onic fibroblasts. Two glycine residues (GG) were also added to ensure
designed and loaded with an antimicrobial peptide immobilized via the motif flexibility. RADA16-FPG effectively inhibited fibroblast prolif-
EDC/NHS and thiol-maleimide click chemistry conjugation. This plat- eration and mildly suppressed keratinocyte expansion (Fig. 8), com-
form illustrated a long-lasting antibacterial activity. Beneficial effects pared with RADA16 and RADA16-RGD (Arg-Gly-Asp) scaffolds, at 72 h
on monocytes, keratinocytes and fibroblasts were reported, such as en- [288].
hanced proliferation and/or differentiation of various cells (HaCaT cells
and NHDF cells) and improved cell-cell attachment, together with inhi- 6.4. Smart stimulus-responsive NSs for burn wounds
bition of the LPS-triggered expression of TNF-α from monocytes e.g.
Raw264.7 cells [281]. Multifunctional core-shell NFs can be utilized for Stimuli-responsive smart materials are starting to open new hori-
wound dressings [282]. Recently, Wei et al. used such structures to ac- zons in nanomedicine [289–295], and their abilities are expected to po-
celerate wound healing. They applied polycaprolactone (PCL) as a tentiate development of effective therapies for burn wound injuries and
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 49

Fig. 10. a) Images of the infected wounds with different treatments: (i, ii) Untreated wounds on control at postoperative day 0 and 7. (iii, iv) Infected untreated wounds on postoperative
day 0 and 7. (v, vi) Infected wounds treated ultrasound-activated TiO2 (UIT) on postoperative day 0 and 7, b) Effect of ultrasound-activated TiO2 on the treatment of the infected wound in
regard to wound size percentage according to the initial wound size (day 0) Reprinted with permission from ref. [304] copyright 2016, John Wiley & Sons, Inc., c) Scheme of the smart
antimicrobial system. Here, the exposure to lysozyme led to hydrolysis of NAc-CTS to give COS. The generated COS served as a substrate for CDH that produces the antimicrobial H2O2.
Reprinted with permission from ref. [305], copyright 2017, John Wiley & Sons, Inc.

infections. This aim is possible due to the ability of so-called “smart can provide on-demand and appropriately-controlled liberation of mis-
nanomaterials” to respond to cues in their microenvironment, so they cellaneous therapeutic agents [296–298].
can provide “on-demand” interaction with cells (e.g. gene expression Stimulus-responsive NSs in the form of 3D-scaffolds are able to un-
only in specific cells and only when desired). Smart nanomaterials can dergo on-demand morphological deformation such as volumetric
control their own behavior and fate, resulting in improved signaling swelling in response to environmental changes e.g. changes in the cellu-
and treatment mechanisms in burn wounds and infections, and mini- lar environment and in the surrounding medium. This could lead to im-
mize side-effects. In addition, stimulus-responsive smart nanomaterials proved healing of burns and chronic wounds. For example, You et al.

Fig. 11. a) Scheme of a PEG-MoS2 nanoplatform to destroy bacteria via a synergistic combination of peroxidase catalysis and photothermal activation: (i) Capture of the nanoplatform by
bacteria; (ii) catalytic activity of the nanoplatform decomposes low-concentration H2O2, leading to generation of OH that damages cell wall integrity; 808 nm laser treatment induces
hyperthermia, causing accelerated GSH oxidation, b) Relative bacterial viability for B. subtilis, obtained after incubation with PBS, MoS2 (100 μg mL−1), H2O2 (100 μM), or MoS2
+ H2O2 for 20 min, with or without the laser treatment, c) GSH loss after heating by water bath or 808 nm NIR-irradiation compared to control (non-treated GSH) for 20 min at 50 °C.
Reprinted with permission from ref. [315], copyright 2016, American Chemical Society, d) Photomicrographs indicating PDT-triggered healing: (i) normal skin, (ii) wound without infec-
tion, (iii) infected wounds, (iv) infected wounds with 200 μM pl–cp6 treatment, and (v) infected wounds with 200 μM pl–cp6 treatment plus with exposure to the light dose of 60 J/cm2.
Reprinted with permission from ref. [316], copyright 2013, Springer.
50 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

hypothesized that local pH changes (e.g. when hypoxia occurs) could Elsewhere, the use of a nanofibrous matrix containing a MMP-
change the ability of a pH-dependent scaffold to transport vital mole- cleavable linker and decorated with siRNA encoding the silencing of
cules (e. g. oxygen, glucose and other nutrients), as well as affecting the MMP2-gene was tested in a wound healing model [307]. In another
the local cell density growing on the scaffold. Here, acidic pH-induced study, MMP secretion was shown to have a key role in re-
swelling of dimethylaminoethyl methacrylate (DMAEMA)/2- epithelialization and acceleration of healing as well as extracellular-
hydroxyethyl methacrylate (HEMA) scaffolds enhanced the penetration induced regulation of kinase signaling in the wound healing process
of oxygen and increased cell infiltration, compared to non-pH sensitive [308]. In a recent study elevated lysozyme activity was exploited as a
scaffolds. Furthermore, other results showed improved formation of biomarker of infection in order to activate a stimulus-responsive anti-
granulation tissue, increased expression of tissue-regenerating factors microbial platform. N-acetyl chitosan (degree of N-acetylation: 40%)
and pro-healing genes, more angiogenesis and better tissue remodeling was hydrolyzed by lysozyme in artificial wound fluid to give N-
were reported. Mechanical stretching of the scaffold was also suggested acetylated chito oligosaccharides (COS). These COS were shown to
as a probable factor that could induce tissue regeneration (Fig. 9-a) serve as substrates for added cellobiose dehydrogenase (CDH) leading
[299]. In another study, a tannic acid (TA) loaded hydrogel designed to the production of 1 mM hydrogen peroxide (H2O2) after 24 h incuba-
by Voelcker et al. was employed in which carboxylated agarose was tion. This was a sufficient concentration to induce an antimicrobial ef-
cross-linked with zinc ions to allow pH-dependent liberation of the fect against S. aureus and E. coli (Fig. 10-c) [305].
cargo [300]. The results showed sustained TA release at acidic pH values
along with significant swelling (with a highly enhanced compressive 6.5. Photothermal and photodynamic therapy for antimicrobial activity and
modulus of 0.947 MPa), and wioth insignificant release at alkaline and wound healing
neutral pH values (Fig. 9-b and -c). No cytotoxicity was seen towards
3T3 fibroblasts, while good anti-bacterial and anti-inflammatory effects NPs can be activated by incident light taking advantage of either
were observed (the latter due to TA-induced inhibition of NO genera- photothermal or photodynamic therapy-based mechanisms, and can
tion in activated human macrophages). Grützner et al. [301] studied kill microbial cells in wound sites and improve wound healing. The prin-
the effect of enzyme-sensitive antimicrobial nanoplatforms loaded ciple of photothermal therapy (PTT) is conversion of light to a localized
with polyhexanide biguanide (cationic polymer) and octenidine (cat- heating effect due to photo-excitation of metallic NPs (AuNPs) or inor-
ionic surfactant) for prophylaxis against infection in cutaneous burns. ganic NPs (graphene, magnetic NPs. MoS2) [309–313]. Various mate-
The results showed time-dependent cellular uptake, along with no rials can be utilized to obtain the PTT-activated antibacterial effect
major effect on expression of cytokines and pro-inflammatory mole- such as photothermal NPs or agents such as molybdenum disulfide
cules by endothelial cells, insignificant cytotoxicity compared to solu- (MoS2), GO, and polypyrrole NPs (PPy) [127,314,315].
tions of antiseptic agents, and no inhibition of angiogenesis. In one study, a 1064 nm NIR laser was used for surface photothermal
Elsewhere, an antimicrobial enzyme-responsive antibiotic-loaded activation of GO, which led to highly efficacious antibacterial activity in
nanocarrier was reported that could respond to microbial drug- a non-invasive and inexpensive approach. This nanoplatform was ap-
resistance enzymes such as penicillin G amidase and β-lactamase. (Fig. plied against pathogens including P. aeruginosa and S. aureus [127]. Else-
9-d) shows enhanced wound healing, bacterial-selective delivery, high where, an NIR-activated nanoplatform was designed for combination
structural stability with modular design, and decreased side-effects therapy, comprised of a photothermal agent (PPy) and an antibiotic
using this system [302]. (Van), both incorporated into hollow microspheres made of PLGA.
Temperature-responsive NSs have also been investigated for disin- When administered into the wound site, NIR light photothermally trig-
fection purposes. A smart-stimulus-responsive nanocarrier equipped gered both localized hyperthermia and antibiotic drug release, resulting
with the thermo-sensitive moiety, poly-N-isopropylacrylamide, in activation of a synergistic antibacterial activity [314].
showed triggered release of bacteriophages targeted against specific Recently, PEG-functionalized MoS2 nanoparticles were synthesized,
bacterial infections. This innovative concept could be used for therapy and exploited for simultaneous NIR-induced photothermal and
of infected burns [303]. peroxidase-like catalytic activity. Fig. 11-a shows the mechanism of ac-
Ultrasound irradiation has been used to activate NPs in infected tion of this multifunctional nanosystem. Peroxidase activity allows con-
wounds. Osumi et al. used ultrasound (1.0 MHz, intensity of version of H2O2 to hydroxyl radical (•OH) that has strong antibacterial
0.4 W cm−2) to activate TiO2 NPs, which produced ROS. They claimed activity. The synergistic effect of the catalysis and 808 nm-irradiation-
that ROS at physiological level could improve cell response and angio- triggered PTT of PEG-MoS2 NPs led to effective eradication of Gram-
genesis, whereas higher levels could carry out disinfection; therefore negative ampicillin resistant E. coli and Gram-positive endospore-
enhancing healing of the infected sites. Ultrasound therapy could en- forming B. subtilis (Fig. 11-b) in wounds. The photothermal effect was
hance the penetration of NP into the tissues to treat deeper infected also reported to cause glutathione (GSH) oxidation, inducing even
sites. Fig. 10 shows (a) images of the infected wounds treated with or more effective bacterial death (Fig. 11-c) [315].
without using ultrasound-activated TiO2 NPs, (b) curves illustrating rel- The concept of PDT is that photon absorption can excite the ground
ative wound size percentage calculated compared to the initial wound state of a photo-sensitizing agent (incorporated for instance in NPs) to a
size [304]. long-lived triplet state, leading to an energy transfer reaction with mo-
The family of matrix metallo-proteinase (MMP) enzymes, which lecular oxygen, thus generating singlet oxygen as reactive species. Elec-
mediate the balance between the generation and degradation of ECM, tron transfer reactions can also take place producing other ROS e.g.
has an inevitable function in the healing of wounds, as well as in the re- superoxide radicals, free hydroxyl radicals and hydrogen peroxide
modeling of epidermal layers. In one study Kim et al. [306] showed the [317,318]. PDT can be used to kill various microorganisms (bacteria, vi-
MMP2-responsiveness of a 4-arm PEG-conjugated siRNA molecule that ruses, yeasts, fungi, parasites and cancer cells) [319,320]. PDT has been
underwent clustering leading to formation of nanoscale particles con- reported to treat dermatological diseases and infections [321]. Two re-
taining MMP-cleavable linkers. These NPs were resistant to RNase hy- cent studies suggested that PDT could be used as an effective antimicro-
drolysis (compared with free siRNA) and could mediate gene bial method for burn infections [322,323].
transfection, leading to enhanced wound healing. There was low cellu- Nafee et al. reported a polymeric nanoplatform encapsulating
lar uptake of the clusters in the absence of MMP, while in cells that the naturally-occurring photosensitizer, hypericin, as an antibiotic-free
expressed MMP, the clusters dissociated and endocytosis occurred. agent for PDT-based antimicrobial therapy. They tested clinical
Wound closure was enhanced, while decreased MMP2 expression was isolates of MRSA biofilms and planktonic cells. Excitation of the
caused by gene silencing together with higher cytokeratin expression photoactivatable 45 nm-diameter NPs generatied ROS and gave a higher
in the healed wound. antibacterial effect against biofilms than planktonic cells, and in-vivo
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 51

Fig. 12. a) Scheme illustrating the preparation of hydrogel sheet loaded with hEGF, used for healing of full thickness wounds, b) Wound closure-time curve in mice; and c) Macroscopic
images of wound sites, obtained for different samples compared to non-treated controls.
Reprinted with permission from ref. [328], copyright 2016, Elsevier.

tests showed accelerated healing, and improved epithelialization, kera- wound site using the scaffold produced improved skin angiogenesis
tinization and generation of collagen fibers [128]. In a recent study, (in a dose-related manner) and better wound healing. Furthermore,
poly-cationic poly-lysine was conjugated to chlorin p6 (photosensitiz- the PEG-fibrin scaffolds could inhibit skin contraction after the meshing
er) was reported for topical PDT of mice with wounds infected with of autografts. In another study, Bio-NSs based on BC and MNPs were in-
P. aeruginosa. To this end, hyper-inflammatory response of bacteria vestigated an as effective tool for healing of chronic wounds. The mor-
was investigated. Fig. 11 shows histological analysis including un- phology, viability and expansion of human ASCs, and the low cell
wounded tissue (Fig. 11-d(i)) and wounded tissue with different treat- toxicity of the scaffolds indicated BC-MNPs as a smart NS for chronic
ments (Fig. 11-d(ii–v)). Complete re-epithelialization, and no necrotic wound healing [269].
or inflammatory areas were reported in the uninfected wounds, while
the non-treated infected wound and infected wound treated only with 6.7. NSs for growth factor delivery
drug-conjugate (no light) and slow healing, and disorganized collagen
(due to excessive production of TNF-α). Light irradiation with doses of Growth factors can be exogenously delivered into wounds so as to
60 and 120 J/cm2 onto the drug-treated infected wound area decreased activate healing process. Basic fibroblast growth factors (bFGFs) can in-
the bacterial load, led to 5-days earlier healing, and the wounds showed duce fibroblasts to proliferate and migrate, endothelial cells to prolifer-
parallel collagen fibers, no edema, shortened inflammatory phase, ate, as well to carry out angiogenesis. In research conducted by Butko
fewer inflammatory cells, and faster re-epithelialization. Significant re- et al. [327] bFGF was loaded into NPs made from succinylated-CS. The
ductions in TNF-α and cytokines IL-6 were reported at 96 h post- results showed a burst release profile, while co-encapsulation of bFGF
treatment. Culture supernatants showed decreased bacterial protease together with heparin led to the maximum release (18%).
activity (~50%) indicative of inactivation of extracellular virulence fac- In another research [328], a hydrogel-sheet based on thiolated hep-
tors [316]. arin attached to PEG and loaded with human EGF was used for wound
healing (Fig. 12-a). The profile showed a sustained release pattern. Ap-
6.6. NS-based cell therapy for wound healing plying the hydrogel sheet onto deep wounds on the back of mice im-
proved the wound closure rate (Fig. 12-b and -c), while stimulating
Stem cell therapy is an example of advanced tissue-engineering granulation tissue formation and better epithelialization and capillary
techniques, which can be potentiated by different nanotechnology- formation.
based platforms, such as 3D cell-scaffold biostructures and hydrogels
to improve wound healing. Kamp et al. [324] interrogated the capability 6.8. NSs for gene delivery
of MSCs for therapy of chronic wounds or burns, that were combined
with an MSC chemoattractant, hepatocyte growth factor (HGF) loaded Gene therapy is regarded as a novel and powerful technique for
in collagen and silk fibroin scaffolds. Here, the release of active HGF treatment of wounds and skin damages. In one study, Du et al. reported
from the scaffolds was indicated to induce direct cell migration, leading the rapid closure of burn wounds and improved healing stimulated by
to endogenous recruitment of MSCs from the local environment in the the gene transfection of DNA coding for hypoxia inducible factor-1
scaffolds (HIF-1α) to induce angiogenesis, integrated with intravenous infusion
In one study Chung et al. [325] developed an engineered 3D-scaffold of HIF-1-activated angiogenic cells cultured with the hydroxylase inhib-
made of PEGylated fibrin gel that was assessed for delivery of ASCs into itor, dimethyloxalylglycine [329]. Moreover, because highly efficient de-
the burn injury site. ASC delivery led to enhanced vascularization and livery of genes into cells and tissues is regarded as the path to success for
more cells expressing the mannose receptor, as well as early recruit- gene therapy, NSs can function as a non-viral platform for efficacious
ment of mononuclear cells with improved granulation tissue genera- delivery of genes. For example in one study, gene-silencing using Flight-
tion. No negative influence on connective tissue extension or wound less I (Flii) siRNA loaded into CS-coated porous silicon NPs was used in a
closure kinetics was reported. cutaneous wound site for down-regulation of Flii (a cytoskeletal actin
Elsewhere, Burmeister et al. [326] reported a platform comprised of remodelling protein, which negatively affects wound healing). The ad-
ASCs and PEG-fibrin hydrogels as a scaffold, which was tested in a por- dition of this nanoplatform to keratinocytes caused a reduction in Flii
cine model of debrided burn wounds. The delivery of ASCs into the gene-expression and lower Flii protein production. Cell migration and
52 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

proliferation were enhanced, and were followed by better healing in such advances, nano-scale architectured substrates, cell-imprinted sub-
acute excisional wounds [330]. Peng et al. prepared pDNA-VEGF165/ strates and 3D-printed scaffolds are of particular interest. Furthermore,
β-cyclodextrin- linked polyethylenimine (CYD-PEI) polyplex NPs em- from a biotechnology aspect, some revolutionary techniques have
bedded in a gelatin/β -TCP scaffold. This 3D transfection platform was emerged, including CRISPR-Cas9 gene editing, which will change the fu-
utilized for manipulation of ESCs in wound site. The results indicated ture of genetic engineering, tissue regeneration and may lead to new
prolonged VEGF expression in the ESCs and topical administration led therapies for serious diseases such as cancer, HIV, and genetic defects.
to angiogenesis, accelerated re-epithelialization, and increased dermal Hence, these newly emerged technologies are briefly discussed below:
collagen and hair follicles [331].

6.9. Combination therapies for wound healing 6.10.1. Nanoscale architectured and textured substrates
Cells live in an environment with features on both the micrometer
Various approaches have been developed for wound treatment, and and nanometer scales where they govern various functional behaviors.
recently it has been realized that these approaches will probably need to Nanoscale-architectured structures employed in implantable biomate-
be used in combination if a substantial advance in potentiating the rials can be used to mimic bio-physicochemical environments and cel-
healing process is to be achieved. In recent investigations, different lular niches to control cellular behavior [351]. More recently,
combination wound therapies have been reported including combina- advances in nanotechnology have provided new methods to
tions of already approved clinical modalities, negative-pressure create substrates equipped with surface nanotopography, emulating
wound therapy combined with pulsed radiofrequency energy [332]; ECM architecture or providing suitable cell microenvironments.
and a combination of photobiomodulation with antidiabetic medication Nanotopography or nano-architectures have been shown to have a
[333]. The combination of topical negative pressure and silver- vital function in inducing or modulating cellular activity, motility, viabil-
impregnated foam has also shown a synergistic inactivation against bac- ity, morphology, adhesion, differentiation, expansion and migration
terial biofilms in the wound site [334]. [352,353]. In the body environment, stem cells of various lineages e.g.
Integration of stem cells with other potent therapeutics have dem- human embryonic stem cells (hESCs) and hMSCs have the potential to
onstrated significant effects such as improved wound closure and an- differentiate into diverse cell types and can be applied in regeneration
giogenesis, enhanced cell migration and proliferation, as well as and tissue engineering. Stem cells can be influenced by employing tai-
negligible toxicity or inflammation in wounds [335–340]. Such investi- lored biochemical micro- and nano-architectures consisting of both
gations have designed stem cell-integrated combination therapies, solid materials and soluble molecules. The use of nano-topographies
which can also possibly be considered for burn wounds. and nano-architectures as advanced implantable platforms that mimic
External stimuli have been also utilized in combination wound treat- the biophysical properties of the cellular microenvironment can accu-
ments. Some examples are the combined use of pulsed wave low-level rately guide stem cells in their self-renewal, proliferation and differenti-
laser therapy and stem cells [340]; the combined administration of ation processes [354–356]. In the recent literature, a wide range of
LLLT/light-emitting diode (LED) [341]; and the concurrent exploitation nano-topographic geometries and arrays including nanopits, nanoposts,
of therapeutic high-frequency ultrasound and microcurrent electrical nanogratings, pillars, grooves, ridges, nanotubes, nanopores, nanowires,
stimulation [342]. and so forth, have all been reported to have tissue engineering applica-
The introduction of nanotechnology to wound therapy has been in- tions [357–362].
vestigated via diverse routes. Some recent examples are: a complex of The use of nano-architectured substrates that incorporate stem cells
different GF combinations and hyaluronic acid (HA) loaded into lipo- in cell-based therapy approaches is of high interest. Recently, the effect
somes [343]; and the synchronous use of an ointment containing ther- of controllable biophysical signals on the mechanosensitivity of cells has
apeutic NP and wound healing medicines, which have been reported gained substantial attention. In one study, Chen et al. designed patterns
to have beneficial results for skin wound healing [344]. In another inves- with controllable nano-roughness on glass surfaces as a culture sub-
tigation, the targeting ability of magnetic NPs was combined with an an- strate for hESCs and NIH/3T3 fibroblasts. Their finding revealed that
giogenic gene therapy using a gene transfer technique to improve the nano-topography created a potent regulatory environment for
angiogenesis and wound healing [345]. In another study on wound hESC responses related to cell signaling and functions such as cell mor-
treatment, a “lipoproteoplex” (i.e. liposome and protein hybrid) NP phology, adhesion, self-renewal and proliferation, which were cell-type
preparation was developed for efficient siRNA delivery as a gene thera- specific. Here, the topological sensing ability of hESCs resulted in
py approach for wound therapy [346]. Hydrogels have also been attrac-
tive components in combination therapies; some recent examples are
as follows: combination of LLLT and policaju (derived from Anacardium
occidentale L gum)/chitosan hybrid hydrogels [347]; the integration of
hydrogels and stem cells [337], the combination of hydrogels and
platelet-rich plasma [348,349], and so forth.
Several combination therapies aimed at wound healing have been
studied in different phases of clinical trials, such as a clinical trial recent-
ly conducted to investigate the effect of simultaneously using gelatin
sheet and platelet-rich plasma release [350].

6.10. New technologies in tissue regeneration and wound healing

In the era of translational medicine with a range of bench-to-bedside


efforts being conducted to transfer preclinical studies to clinical prod-
ucts, well-designed efforts are absolutely required in order to eliminate
previous deficiencies and shortcomings, especially in the fabrication
methods of therapeutic products, such as complicated design methods
and the high-cost of processes and materials, but also to produce user- Fig. 13. Schematic of 3D-printing technology developed for the fabrication of 3D-scaffolds
friendly and low cost technologies. In this regard, some innovative ap- and dressings, within which cell-matrix components can be implanted, with applications
proaches have recently been introduced in tissue engineering. Among in burn wound healing and tissue regeneration.
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 53

feedback regulation, associated with the mechanosensory integrin- myofibroblasts toward surface ridges larger than 1 μm was also report-
mediated adhesion to the cell-matrix, E-cadherin, and myosin II [363]. ed; however, directional guidance on submicron/nano-topographic or
In a similar study, Yim et al. observed that the nano-topography and planar surfaces was lacking [370].
stiffness of substrates could modulate the behavior of hMSCs by altering
the formation of activated integrin clusters and assembly of focal adhe- 6.10.2. Cell-imprinted substrates
sion complexes. This, consequently, regulated the organization of the Cell-imprinted biomaterials are able to mimic the natural environ-
cell cytoskeleton of hMSCs cultured on a substrate with 350 nm grating ment of stem cells, and induce stem cell differentiation and expansion
nano-topographies on the surface of tissue-culture grade polystyrene for wound healing and artificial tissue applications [371,372]. In this re-
and polydimethylsiloxane (PDMS) substrates. Here, by employing the gard, various cell types and morphologies can be imprinted to design a
nano-topographies, the expression of various integrin molecules includ- proper cell niche micro-/nano-environment. The cost-effectiveness and
ing alpha, alpha 2, alpha V, beta2, beta 3 and beta 4 was decreased in high efficiency of this approach is likely to have many advantages in tis-
comparison with the control group (i.e. non-patterned substrates). sue engineering and regeneration as well as wound therapies, using var-
Moreover, the mechanical properties (e.g. Young's modulus and appar- ious cell casting processes on surfaces [373]. In one study, cell-
ent viscosity) and expression of cytoskeleton and focal adhesion com- imprinted substrates were reported to function as an artificial NS for
ponents were also changed on the patterned substrates. The grating skin repair. Herein, PDMS casting based-cell-imprinted micro-/nano-
nano-topography induced alignment of the actin cytoskeleton of the environments were created using human keratinocyte patterned tem-
elongated hMSCs whereas the non-patterned substrate led to a dense plates. The keratinocyte-cell-imprinted biomimetic substrates could
and random spreading of the actin cytoskeleton network [364]. mimic the surface morphology of the cell membrane, which created a
The differentiation of fibroblasts into myofibroblasts is an important suitable micro-/nano-environment for topographical-induced cell dif-
stage in wound healing because of the important role of myofibroblasts ferentiation. Moreover, seeded ASCs on the cell-imprinted surfaces
in restoring skin homeostasis and modulating tissue physiology in the were stimulated to adopt keratinocyte morphology and properties. Mo-
wound healing process [365,366]. Keratinocyte behaviors can be influ- lecular dynamics simulation of the stem cell nucleus interestingly re-
enced by modification of nanoscale topographies, which may cause im- vealed that changing the shape of stem cells into that resembling
provements in wound healing [367,368]. Parkinson et al. in a study, mature cells could affect the deformation of the nucleus and subsequent
evaluated the effect of nano-topography on keratinocytes including gene expression. The results demonstrated the cell-imprinted substrate
their morphology, proliferation and migration for treatment of burn could be utilized as an effective strategy to differentiate stem cells to
wounds. Herein, nanoporous anodic aluminum oxide (AAO) mem- skin cells for wound healing [374]. More recently, a similar research re-
branes were employed with a pore size of 300 nm, which increased ported cell-imprinted surfaces as a smart nano-environment, construct-
keratinocyte expansion and migration and could cover a partial thick- ed by utilizing templates so that different cell types, including
ness burn wound. A burn injury dressing incorporating a nano- chondrocytes, tenocytes and semifibroblasts had potentials to differen-
topographical AAO membrane enhanced skin healing with a significant- tiate, re-differentiate and trans-differentiate from the seeded ASCs.
ly reduction in tissue granulation and maturation of epidermal layers, Moreover, immunofluorescent staining of proteins such as collagen
compared with a control group [369]. In a similar study, Pot et al. report- type II in chondrocytes revealed that ASCs, semifibroblasts and
ed nano-topography could regulate keratocytes, fibroblasts and tenocytes were able to attain the chondrocyte phenotype after
myofibroblasts, which have a major role in corneal wound healing. 2 weeks of being implanted on chondrocyte-imprinted substrates.
These cells were seeded onto planar polyurethane surfaces. After 24 h, These results indicated that cell-Imprinted substrates could be applied
the keratocytes, fibroblasts and myofibroblasts were stained and im- in skin regeneration and cell-based therapy [375].
aged for cell shape analysis as well as for studying migration rates.
Here, the elongation of keratocytes, fibroblasts and myofibroblasts 6.10.3. 3D bio-printed scaffolds
were reported to be N1000 nm on a pitch (i.e. groove + ridge) pat- 3D printing is an advanced technology that has been progressing
terned surfaces where cells responded to topographic features that rapidly, and demonstrating revolutionary effects in biomedical research
had a lower limit of size with a transition zone of 800 to 1200-nm and therapies. In 3D printing, complex biomolecules and even living
pitch size. Moreover, using this nano-topography, while keratocytes components such as (stem) cells, proteins (e.g. fibrin, collagen, growth
remained motionless, the migration of fibroblasts as well as factors, etc.), and therapeutic molecules can be precisely patterned

Fig. 14. Schematic representing the concept of using gene editing tools such as CRISPR-Cas for reprogramming of stem cells (e.g. iPSCs) in wound healing applications.
54 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

and assembled through layer-by-layer deposition on scaffolds e.g. 7. Animal models for burn wounds
hydrogels, with the aid of computer-aided designs and manufacturing
(e.g. CAD and CAM) [373]. This technology can be used in several health Animal models of burn wounds are widely used in the laboratory to
care applications, including tissue regeneration; prosthetics fabrication, evaluate different drugs and treatments that are being developed for
implant creation, anatomical model fabrication and drug delivery inves- treatment of human burn victims [388].
tigations [376,377]. The potential of 3D printing and its ability to con- In wound therapy research, regarding various wound models, differ-
struct complex and elaborate patterns and architectures can be ent aspects should be considered including incision or excision models,
utilized in the study of wound healing therapies to manufacture 3D- acute, chronic or diabetic wounds, administration methods for therapies
scaffolds, dressings, etc. or to create powerful platforms as skin models (e.g. topically, intravenously, intradermally, and spray), in-vitro models
to investigate and find treatments for skin diseases such as burn (e.g. EpiDermFT as an epidermal full-thickness model, and human/
wounds (Fig. 13). Bio-printing based formation of cell-matrix compo- mouse fibroblast and keratinocyte models as popular models), in-vivo
nents has been reported for the regeneration of skin tissue and forma- models (e.g. rat and mouse that are cost-effective and user-friendly, as
tion of stem cell-scaffold complexes to thoroughly cover the full- well as pig and rabbit), and so forth [154].
thickness skin damage in burn wounds [373]. As an example of recent Burn wounds are usually created on the shaved backs of animal
pioneering studies, Lee et al. produced eight collagen layers as human- models such as mice, rats, rabbits and pigs, with a range from 5% to
like skin using a 3D printing platform. The structure of the 3D printed 30% TBSA for 3 to 30 s to produce partial to full thickness thermal dam-
platform consisted of implanted cells within a multi-layered collagen age. As an advantage, anatomically and physiologically the skin struc-
construction on poly-d-lysine-coated glass petri dishes previously neb- ture of the guinea pig is more analogous to human skin compared
ulized by sodium bicarbonate steam. The eight layers of collagen with mouse and rat. The epidermal thickness of pig is 30 to 140 μm sim-
consisted of six collagen layers sandwiching three layers of fibroblasts ilar to the human epidermal thickness which is 50 to 120 μm [389,390].
and two additional collagen layers that included keratinocytes [378]. Thus guinea pig burn models are better able to emulate human thermal
As 3D-printable platforms have been dramatically progressing in re- wounds. However, mice and rats are less expensive than guinea pigs.
cent years, unprecedented concepts have been also introduced to this Studies have revealed that inoculating infectious microorganisms topi-
area such as “4D-printing”. In this regard, the introduction of smart ma- cally is more clinically relevant than injecting them under the burn
terials into 3D-printable platforms has been called as “4D-printing”, wound [391]. Investigations have also shown that acute infections can
which means the shape, performance, functionality and properties of be induced in animal models e.g. mouse, rat and pig by invasive micro-
printed smart materials can be altered over time (as the 4th dimension) organisms such as P. aeruginosa, whereas chronic infections can be cre-
and in response to different stimuli. 4D-printed structures can be de- ated using less virulent microorganisms such as S. aureus [7,392]. Here
signed to be self-assembled, multi-functional, self-repairable, etc. In we briefly discuss important animal models for burn wounds, which
this regard, 4D-bioprintable platforms will likely become an important are classified as: gas flame burn model, burning ethanol bath burn
area of research in tissue regeneration and engineering, particularly model, pre-heated single metal plate/bar boiling or hot water burn
for printable smart scaffolds, skin regeneration and artificial skin, with models, and pre-heated double brass block burn model [391].
applications in (burn) wound healing and regeneration [379,380].
From the bio-inspired aspect, 4D-bio-pintable platforms are printed ob- 7.1. Gas flame burn model
jects that can show smart stimuli-responsive behavior when for in-
stance, external stimuli are applied, or when cell-fusion or post- 7.1.1. Katakura burn model
printing self-assembly are employed [381]. In this model, a 2.5 × 3.5 cm window equipped template is placed on
the shaved back of mice, and subsequently, the exposed site on the skin
is heated for 9 s using gas flame. This flame-induced burn model can
6.10.4. CRISPR-Cas9: new gene editing tool for reprogramming of stem cells create a third degree burn with a nearly 15% TBSA on 26 g SCID-bg
Gene editing through the clustered regularly interspaced palin- mice. Subsequently, gas flame burned immune-deficient mice are incul-
dromic repeats (CRISPR)-Cas system has been introduced for the mod- cated with a 2 x 103CFU MRSA and various numbers of CFU E. faecalis to
ification of the genome in many microorganisms and eukaryotic cells test the influence of an antimicrobial agent on antibiotic-resistant burn
[382]. Originally discovered as a form of bacterial immunity to viruses, infections [393].
type 2 known as CRISPR-Cas9 system is a simple and efficient tool
employed for manipulation of heterogenous and endogenous gene ex- 7.2. Burning ethanol bath burn model
pression. This breakthrough approach can be applied for somatic and
embryonic gene therapy, genomic screening and the generation of ani- 7.2.1. Stieritz-Holder burn model
mal models that have been exploited in all biomedical-related re- These authors applied an ethanol bath to make a burn wound animal
searches, ranges from basic research to gene therapy [383,384]. model. In this model, anesthetized and back-shaved female BALB/c mice
CRISPR/Cas9 system can be applied to reprogram primary fibroblasts weighing 22–24 g are used. To generate a nearly 30% of TBSA, back-
so as to produce iPSCs. iPSCs themselves have the potential to differen- shaved mice are exposed to an asbestos sheet containing a window,
tiate into other cell types such as keratinocytes and MSCs, which are the firmly pressed on the back. Subsequently, ethanol is evenly sprayed
most important cells for wound regeneration [385,386]. More recently, on the region specified by the window, and then ignited and allowed
a CRISPR-Cas system was reported, which could be applied in gene ther- to burn for 10 s. The bacterial infection models are created via inoculat-
apy for skin diseases and wound healing. Herein, the designed gene ing 100 CFU of P. aeruginosa and K. pneumoniae into the burned subcu-
therapy system enhanced skin growth factors expression such as EGF, taneous area. Here, the infections can rapidly lead to death depending
TGF-β, PDGF and FGF [387]. Hence, CRISPR-Cas9 gene editing technolo- on the virulence of the bacterial strain chosen. This model has been
gy can be a highly potent tool in cell and tissue engineering as well as employed to investigate the antibacterial function of polyclonal immu-
regenerative medicine. In addition, it suggests new horizons in wound noglobulin antibodies for burn wound healing [391,394].
healing and infection prevention, which requires more research to be
conducted to fully realize the potential. Fig. 14 illustrates the concept 7.3. Pre-heated single metal plate/bar burn models
of exploiting the gene editing tool, CRISPR-Cas, for reprogramming of
stem cells (e.g. iPSCs), where reprogrammed (i.e. differentiated) cells 7.3.1. Tavares burn model
can be integrated in fabricated 3D–scaffolds, for wound healing In this model, a pre-heated aluminum rod (51 g in weight and
applications. 10 mm in diameter) is utilized. To make a second degree burn wound,
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 55

Fig. 15. I–II) In Stevens burn model, a deep burn wound is generated using two blocks of brass with pre-heating of 92–95 °C on a shaved mouse back. III) Square show burned region. IV)
Bacterial suspension in PBS was used on the surface of burn.
Reprinted from ref. [391], copyright 2011, Taylor and Francis Group (open access, licensed under a Creative Commons Attribution-NonCommercial 3.0 Unported License.)

it is heated to about 98 to 100 °C for 10 min. Subsequently, the heated 2nd degree) and full-thickness (as 3rd degree) burn model, back-
aluminum rod is pressed on the shaved back of male rats for 15 s. shaved rats are exposed 3 and 10 s to boiling water, respectively.
Every seven-days for 4 weeks, the anesthetized burned animal is evalu- The upper restriction on wound burn size, permitting by Mason and
ated for bleeding and infection. After 4 weeks, re-epithelialization cis Walker model is about 30% of TBSA. This model has been broadly
usually completed through fibroblast expansion and new collagen syn- employed in studies conducted on the suppression and therapy of
thesis [395]. burn wound infections, for example: bacterial translocation, testing var-
ious antimicrobial agents, gene therapy, wound dressing for inhibiting
7.3.2. Orenstein burn model burn infections such as fungal (e.g. candidiasis) infections [391,400].
In this model, to create a third degree burn, a pre-heated 1 × 1
× 3 cm plate is used, with a 20 cm handle. The temperature of the cop- 7.5. Pre-heated double brass blocks burned models
per plate is increased to 150 °C where the plate is pressed on the back-
shaved guinea pigs for 10 s. The burned area is inoculated after 15 min 7.5.1. Kaufman burn model
by 1 mL suspension containing 108 S. aureus. This model can be In this model, a cylindrical aluminum rod is utilized weighing at least
employed for the investigation of prevention and treatment of infected 500 g, with diameter and length of the handle of 3.78 cm and 24 cm, re-
burns [396]. spectively. The heating of the equipment is done while submerged in
water for 120 min, and then to create a deep partial skin thickness ther-
7.4. Boiling or hot water burn models mal wound at 75 °C on shaved back of an anesthetized guinea-pig. The
model can be applied to investigate epithelialization, contraction and
7.4.1. Suzuki burn models scar formation of burn wounds [401]
In this model, male Wistar rats weighing 450–550 g are used and
their skin is brought into contact with a glass chamber device through 7.5.2. Stevens burn model
which water circulates for 1 min at 35–60 °C over a seven-day period. This model uses two pre-heated brass blocks applied to the back of
The important variable is how firmly the device is pressed onto the shaved mice for between 3 and 10 s depending on the depth of burn re-
skin (a common applied pressure is 10 g/cm2). This model benefits quired. In this model, pre-heated double brass blocks (heated to 92–95
from the ability to vary exposure time and temperature [391,397]. °C) are placed onto either side of a raised skin fold on the back of the
shaved mice for between 5 s depending on the depth of burn required,
7.4.2. Bahar burn model resulting in a 1.8 cm2 burned area being created with nearly 5% TBSA (as
This burn model involves the application of a 2.5 cm2 thick piece of an example see Fig. 15). Subsequently, to generate a bacterial infection
absorbent lint cloth submerged in boiling water. This cloth is then ap- model, various numbers of P. aeruginosa CFU are inoculated onto the
plied to the back of male Wistar rats weighing 250–300 g for varying burned region or else are intradermally injected into the eschars. Bacte-
times (2 or 10 s) [398]. ria can be recovered from the unburned muscle by 1 day for some bac-
teria. In this burn model, survival at 10 d has been reported to be 60%
7.4.3. Bjornson burn model with P. aeruginosa infection [391,402].
Here, female or male guinea pigs are utilized weighing 300–350 g. as
a burn model. In this model, 60 cm2 area is submerged in water at 99 °C 8. Recent clinical trial evaluations for wound healing
for 13 s to make an about 15% TBSA. To create second thermal wound,
after 1 h, the guinea pigs are again exposed in the template. Subsequent- An essential step in using the various wound healing approaches
ly, the animals are inoculated with 5 × 105CFU of P. aeruginosa, S. aureus discussed in the above-mentioned sections, is their translation into clin-
and C. albicans under the burn area [399]. ical applications and bench-to-bedside evaluation. In this regard, in this
section, some of recent clinical trials in different stages of completion
7.4.4. Mason and Walker burn model are summarized.
These workers developed the first animal burn model in rats using Essentially, clinical trials to understand the efficacy of (stem) cell
boiling water, which is a standard boiling water burn model used in di- therapy on the healing process of burn wounds are still needed so as
verse investigations. In this model, a template is employed in the form to confirm their efficacy and wound-healing properties. In April 2017,
of a thin metal half-cylinder that fits over the back of the rat. It has a Cytori Therapeutics Inc. obtained FDA approval for a pilot clinical trial
hole of calculated dimensions in the central region of the half- of their product “Cytori Cell TherapyTM” using for thermal-burn injured
cylinder. The size of the hole is determined to correspond with the patients. Here, the safety and feasibility of the product along with stan-
size of the burn in the rat. The animal is anesthetized, then shaved on dard care for patients with 20% to 50% TBSA injuries was assessed [403].
the back, followed by placing supine in the template. The exposed re- In March 2017, a Phase I clinical trial in 2nd degree burn wound patients
gion is submerged into boiling water. To create a partial thickness (as with b 20% TBSA was verified to be conducted by University of Miami.
56 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

The purpose of this investigation was to evaluate the safety of allogeneic bedrock of burns care, alternative approaches including immune-
MSC based treatment, and then to find the maximum safe dose to be based antimicrobial molecules, therapeutic microorganisms, antimicro-
used in the Phase II of this study [404]. bial phototherapy (PTT, PDT, blue and green light or NIR based thera-
Among currently used approaches for wound therapy, skin allografts pies), and different kinds of cell therapy have been explored.
have been suggested for dermo-protection and enhancing re- Nonetheless, antimicrobial resistance developing in a wide range of mi-
epithelialization in burn injuries [405]. croorganisms has created vast problems for topical treatment of burned
The wound closure effect of “nitrosating substances” e.g. nitroso- infections in recent years. Since antibiotic-resistant microorganisms are
thiols or RSNOs have been suggested due to their inflammation modu- now the main threat to public health, new advanced approaches such as
lation, cytokine generation and vascular improvement functions. nanotechnology are being investigated that may be able to offer novel
Employing nanotechnology can enhance the effect of such products. therapeutic strategies in the post-antibiotic era.
For example, recently an S-nitroso-N-acetyl cysteine containing Up until the present the most important problem in burn wounds
hydrogel-based NP platform (NAC-SNO-np) was developed, with en- has been chronic infections caused by some microorganisms such as
couraging results in translational and bench-to-bedside studies. This P. aeruginosa, that are especially serious in deep burns with large areas
nanoplatform could release NO in a timely manner, besides facilitating involved. This is due to the intrinsic and acquired resistance of bacteria
nitrosation. NAC-SNO-np also has been licensed by the company Nano to antibiotics resulting in high mortality in burn wound patients.
Biomed Inc. [406,407]. In another approach in 2007, Phase I and II clin- A wide range of sophisticated nanostructures are now being ex-
ical trials were designed and funded by National Institute of Health plored in burn wounds, which may be able to overcome the problem
(NIH) to support filing with the FDA and marketing potential of a NO- of infection, while at the same time stimulating skin regeneration.
generating topical gel for burn wound healing [408]. Hence, in this review we have focused on both organic and non-
Ag NPs provide great promise in clinical applications of wound organic NSs including polymeric NPs, NEs, SLNs, hydrogels, ceramic
healing therapies. This is due to their intrinsic antimicrobial activity NPs, metal NPs, TiO2 NPs, MNPs, carbon-based NPs, QDs and NFs all of
along with anti-inflammatory and wound healing effects resulting in which could be possibly utilized as potential antimicrobial agents in
scar reduction, suppression of local and systemic inflammation, modu- burns. In addition, other NS-based platforms may be more directed to-
lation of cytokine levels, increasing wound closure via proliferation wards improving burn wound healing such as 3D-scaffolds, wound
and migration of keratinocytes, and enhancing wound contraction via dressings and films, strategies to deliver growth factors, and stimuli-
myoblast generation by fibroblast differentiation. Furthermore, the ef- responsive NSs. These various platforms can be effectively utilized for
fect of Ag NPs as a UV and radio-therapy sensitizing agent and their ef- wound therapy applications such as PDT, PTT, stimulus-sensitive drug
fect on the immune system can be also considered [409]. delivery, cell therapy, growth factor delivery, and gene therapy.
Growth factors can influence wound healing, and clinical evaluation NSs can be incorporated into dressings that are applied to the
of their exogeneous application is required. PDGF, a growth factor and burned area, especially for inhibition of microbial proliferation, modula-
cytokine, can facilitate the transition from a chronic wound to short- tion of immune response, and acceleration of wound healing. Studies
term healing by stimulation of granulation tissue formation, and has have shown that NSs either used alone or as multifunctional targeted
been tested in clinical trials [405]. Delivery of PDGF as a topical agent and smart agents can be effective strategies to inhibit microbial growth
to burn wounds may be achieved by nano-platform approaches. In in the burn wound microenvironment. NSs can also be implemented to
2010, a clinical study was reported to evaluate healing of second degree design stem cell–based scaffolds for skin regeneration and remodeling.
burn wounds using the growth factor, rhEGF. In this study, 80 elderly The unique properties of stem cells combined with nano-structured
patients were studied from 2003 to 2008, and the results indicated scaffolds may give rise to insights that may lead to substantial advance-
that rhEGF enhanced burn wound healing [410]. In a recent first-in- ments in burn therapy. The advanced properties of NSs suggest they can
man clinical trial, a collagen/gelatin scaffold was developed capable of be a technological step forward to prevent infections in thermal wounds
sustained release of FGFs. The results of this clinical trial indicated the and improved healing of damaged tissues compared with conventional
efficacy and safety of the scaffold applied on patients [411]. topical treatments.
A phase I clinical trial carried out by Stanford University Medical Burn wound healing can also be improved by using scaffolds
Center (CA, USA) used a skin-based gene therapy for wound healing, possessing innovative features that have been fabricated via novel and
and was hailed as the first safe and effective skin-based gene therapy. user-friendly techniques. Materials such as electrospun NFs, smart
This study aimed at grafting genetically altered skin onto chronic stimuli-responsive and multifunctional 3D-scaffolds with favorable
wounds of epidermolysis bullosa patients [412]. properties including the ability to control proliferation, migration and
In 2012, a randomized controlled trial was carried out where the an- differentiation of various cell types, loading and on-demand release of
algesia effect of a combined donor compound that could release nitrous antimicrobial therapeutics, and so forth, will lead to more effective
oxide (N2O)/oxygen was investigated to reduce pain associated with wound therapies and suppression of burn infection. More importantly,
changing burn dressings [413]. recently developed technologies including 3D/4D-printing, use of cell-
In a recently conducted phase 1 clinical trial, the combination of imprinted substrates, and nano-scale architectured surfaces have
high-voltage, pulsed-current electric therapy and silver-collagen dress- emerged with beneficial properties and features. In addition, some
ing were shown to be efficacious for chronic full-thickness wounds biotechnology-related advances such as powerful gene-editing tech-
[414]. niques including CRISPR-Cas-based systems will create a milestone in
In addition to the aforementioned case studies, the literature also the therapy of various diseases, particularly with major effects on treat-
contains examples, where the effects of keratinocytes and stem cells, ment of (burn) wound healing and suppression of pathogens and infec-
pirfenidone NPs, hyperbaric oxygen, opioids, and so forth, on burn tions, which are expected to become reality in the coming years.
wound healing have been explored [415]. Considering the immense research effort that is currently being
invested in nanomedicine applications, future progress in burn treat-
9. Concluding remarks and perspectives ment is expected to result from these above-mentioned innovative
approaches.
Burns pose a highly complex problem in medical care. The high sus-
ceptibility to microbial infection coupled with the need to achieve rapid Acknowledgment
and satisfactory wound healing, while at the same time avoiding unde-
sirable scarring taxes the resources of even specialized burns units. Michael R. Hamblin was supported by US NIH Grants R01AI050875
While topical application of antimicrobial agents has formed the and R21AI121700
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 57

Appendix A. Supplementary data are inhibitory on the production of antimicrobial peptides by epidermal
keratinocytes, PLoS One 9 (2014), e82926.
[33] L.U. Lahoda, S.C. Wang, P.M. Vogt, A mixture of antimicrobial peptides and fi-
Supplementary data to this article can be found online at http://dx. brin glue in treatment of partial-thickness burn wounds, Chirurg 77 (2006)
doi.org/10.1016/j.addr.2017.08.001. 251–256.
[34] M. Jaskiewicz, M. Orlowska, G. Olizarowicz, D. Migon, D. Grzywacz, W. Kamysz,
Rapid screening of antimicrobial synthetic peptides, Int. J. Pept. Res. Ther. 22
(2016) 155–161.
References [35] H.M. Jindal, C.F. Le, M.Y. Mohd Yusof, R.D. Velayuthan, V.S. Lee, S.M. Zain, D.M. Isa,
S.D. Sekaran, Antimicrobial activity of novel synthetic peptides derived from
[1] M. Peck, J. Molnar, D. Swart, A global plan for burn prevention and care, Bull. World indolicidin and ranalexin against Streptococcus pneumoniae, PLoS One 10 (2015),
Health Organ. 87 (2009) 802–803. e0128532.
[2] http://www.who.int. [36] J. Agier, E. Brzezinska-Blaszczyk, Cathelicidins and defensins regulate mast cell an-
[3] D. Church, S. Elsayed, O. Reid, B. Winston, R. Lindsay, Burn wound infections, Clin. timicrobial activity, Postepy Hig. Med. Dosw. 70 (2016) 618–636.
Microbiol. Rev. 19 (2006) 403–434. [37] A. Fabisiak, N. Murawska, J. Fichna, LL-37: cathelicidin-related antimicrobial
[4] J.K. Salmon, C.A. Armstrong, J.C. Ansel, The skin as an immune organ, West. J. Med. peptide with pleiotropic activity, Pharmacol. Rep. 68 (4) (2016 Aug) 802–808,
160 (1994) 146–152. http://dx.doi.org/10.1016/j.pharep.2016.03.015.
[5] M.P. Rowan, L.C. Cancio, E.A. Elster, D.M. Burmeister, L.F. Rose, S. Natesan, R.K. [38] J. Agier, M. Efenberger, E. Brzezinska-Blaszczyk, Cathelicidin impact on inflamma-
Chan, R.J. Christy, K.K. Chung, Burn wound healing and treatment: review and ad- tory cells, Cent. Eur. J. Immunol. 40 (2015) 225–235.
vancements, Crit. Care 19 (2015) 243. [39] J.K. Actor, Lactoferrin: a modulator for immunity against tuberculosis related gran-
[6] O. Kallinen, V. Koljonen, E. Tukiainen, T. Randell, H. Kirves, Prehospital care of burn ulomatous pathology, Mediat. Inflamm. 2015 (2015) 409596.
patients and trajectories on survival, Prehosp. Emerg. Care 20 (2016) 97–105. [40] J. Schauber, R.L. Gallo, Antimicrobial peptides and the skin immune defense sys-
[7] T. Dai, Y.Y. Huang, S.K. Sharma, J.T. Hashmi, D.B. Kurup, M.R. Hamblin, Topical an- tem, J. Allergy Clin. Immunol. 122 (2008) 261–266.
timicrobials for burn wound infections, Recent Pat. Anti-Infect. Drug Discov. 5 [41] B. Lonnerdal, S. Iyer, Lactoferrin: molecular structure and biological function, Annu.
(2010) 124–151. Rev. Nutr. 15 (1995) 93–110.
[8] M. Sevgi, A. Toklu, D. Vecchio, M.R. Hamblin, Topical antimicrobials for burn infec- [42] M.F. Mohamed, A. Abdelkhalek, M.N. Seleem, Evaluation of short synthetic antimi-
tions — an update, Recent Pat. Anti-Infect. Drug Discov. 8 (2013) 161–197. crobial peptides for treatment of drug-resistant and intracellular Staphylococcus
[9] R. Yin, T. Dai, P. Avci, A.E. Jorge, W.C. de Melo, D. Vecchio, Y.Y. Huang, A. Gupta, M.R. aureus, Sci Rep 6 (2016) 29707.
Hamblin, Light based anti-infectives: ultraviolet C irradiation, photodynamic ther- [43] H.D. Thaker, A. Som, F. Ayaz, D. Lui, W. Pan, R.W. Scott, J. Anguita, G.N. Tew, Syn-
apy, blue light, and beyond, Curr. Opin. Pharmacol. 13 (5) (2013 Oct) 731–762, thetic mimics of antimicrobial peptides with immunomodulatory responses, J.
http://dx.doi.org/10.1016/j.coph.2013.08.009. Am. Chem. Soc. 134 (2012) 11088–11091.
[10] C. Marquart-Elbaz, D. Lipsker, H. Sick, E. Grosshans, B. Cribier, Does subcutaneous [44] S. Hemachandra, K. Kamboj, J. Copfer, G. Pier, L.L. Green, J.R. Schreiber, Human
cellular tissue exist? Ann. Dermatol. Venereol. (2001) 1201–1205. monoclonal antibodies against Pseudomonas aeruginosa lipopolysaccharide de-
[11] E. Fuchs, Skin stem cells: rising to the surface, J. Cell Biol. 180 (2008) 273–284. rived from transgenic mice containing megabase human immunoglobulin loci
[12] M. Merad, F. Ginhoux, M. Collin, Origin, homeostasis and function of Langerhans are opsonic and protective against fatal Pseudomonas sepsis, Infect. Immun. 69
cells and other langerin-expressing dendritic cells, Nat. Rev. Immunol. 8 (2008) (2001) 2223–2229.
935–947. [45] Z. Lai, R. Kimmel, S. Petersen, S. Thomas, G. Pier, B. Bezabeh, R. Luo, J.R. Schreiber,
[13] F.O. Nestle, P. Di Meglio, J.Z. Qin, B.J. Nickoloff, Skin immune sentinels in health and Multi-valent human monoclonal antibody preparation against Pseudomonas
disease, Nat. Rev. Immunol. 9 (2009) 679–691. aeruginosa derived from transgenic mice containing human immunoglobulin loci
[14] C. Blanpain, E. Fuchs, Epidermal stem cells of the skin, Annu. Rev. Cell Dev. Biol. 22 is protective against fatal pseudomonas sepsis caused by multiple serotypes, Vac-
(2006) 339–373. cine 23 (2005) 3264–3271.
[15] G.C. Gurtner, S. Werner, Y. Barrandon, M.T. Longaker, Wound repair and regenera- [46] Y. Barnea, Y. Carmeli, L.F. Neville, H. Kahel-Reifer, R. Eren, S. Dagan, S. Navon-
tion, Nature 453 (2008) 314–321. Venezia, Therapy with anti-flagellin A monoclonal antibody limits Pseudomonas
[16] K. Sellheyer, D. Krahl, Skin mesenchymal stem cells: prospects for clinical derma- aeruginosa invasiveness in a mouse burn wound sepsis model, Burns 35 (2009)
tology, J. Am. Acad. Dermatol. 63 (2010) 859–865. 390–396.
[17] E. Bellas, M. Seiberg, J. Garlick, D.L. Kaplan, In vitro 3D full-thickness skin- [47] L. Ma, H. Lu, A. Sprinkle, M.R. Parsek, D.J. Wozniak, Pseudomonas aeruginosa Psl is a
equivalent tissue model using silk and collagen biomaterials, Macromol. Biosci. galactose- and mannose-rich exopolysaccharide, J. Bacteriol. 189 (2007)
12 (2012) 1627–1636. 8353–8356.
[18] S. Guo, L.A. Dipietro, Factors affecting wound healing, J. Dent. Res. 89 (2010) [48] T. Sawa, E. Ito, V.H. Nguyen, M. Haight, Anti-PcrV antibody strategies against
219–229. virulent Pseudomonas aeruginosa, Hum. Vaccin. Immunother. 10 (2014)
[19] S. Novelli, P. Garcia-Muret, A. Mozos, J. Sierra, J. Briones, Total body-surface area as 2843–2852.
a new prognostic variable in mycosis fungoides and Sezary syndrome, Leuk. Lym- [49] P. Warrener, R. Varkey, J.C. Bonnell, A. DiGiandomenico, M. Camara, K. Cook, L.
phoma 57 (2016) 1060–1066. Peng, J. Zha, P. Chowdury, B. Sellman, C.K. Stover, A novel anti-PcrV antibody pro-
[20] M. Staley, R. Richard, Management of the acute burn wound: an overview, Adv. viding enhanced protection against Pseudomonas aeruginosa in multiple animal in-
Wound Care 10 (1997) 39–44. fection models, Antimicrob. Agents Chemother. 58 (2014) 4384–4391.
[21] B. Mahdavian Delavary, W.M. van der Veer, M. van Egmond, F.B. Niessen, R.H. [50] A. Basu, U. Das, S. Dey, S. Datta, PcrG protects the two long helical oligomerization
Beelen, Macrophages in skin injury and repair, Immunobiology 216 (2011) domains of PcrV, by an interaction mediated by the intramolecular coiled-coil re-
753–762. gion of PcrG, BMC Struct. Biol. 14 (2014) 5.
[22] N. Strbo, N. Yin, O. Stojadinovic, Innate and adaptive immune responses in wound [51] I.A. Holder, A.N. Neely, D.W. Frank, PcrV immunization enhances survival of burned
epithelialization, Adv. Wound Care 3 (2014) 492–501. Pseudomonas aeruginosa-infected mice, Infect. Immun. 69 (2001) 5908–5910.
[23] J. Li, J. Chen, R. Kirsner, Pathophysiology of acute wound healing, Clin. Dermatol. 25 [52] A.N. Neely, I.A. Holder, J.P. Wiener-Kronish, T. Sawa, Passive anti-PcrV treatment
(2007) 9–18. protects burned mice against Pseudomonas aeruginosa challenge, Burns 31
[24] I. Pastar, O. Stojadinovic, N.C. Yin, H. Ramirez, A.G. Nusbaum, A. Sawaya, S.B. Patel, (2005) 153–158.
L. Khalid, R.R. Isseroff, M. Tomic-Canic, Epithelialization in wound healing: a com- [53] M. Mittal, M.R. Siddiqui, K. Tran, S.P. Reddy, A.B. Malik, Reactive oxygen species in
prehensive review, Adv. Wound Care 3 (2014) 445–464. inflammation and tissue injury, Antioxid. Redox Signal. 20 (2014) 1126–1167.
[25] M.G. Tonnesen, X. Feng, R.A. Clark, Angiogenesis in wound healing, J. Investig. [54] F. Vatansever, W.C. de Melo, P. Avci, D. Vecchio, M. Sadasivam, A. Gupta, R.
Dermatol. Symp. Proc. 5 (2000) 40–46. Chandran, M. Karimi, N.A. Parizotto, R. Yin, G.P. Tegos, M.R. Hamblin, Antimicrobial
[26] J.M. Reinke, H. Sorg, Wound repair and regeneration, Eur. Surg. Res. 49 (2012) strategies centered around reactive oxygen species—bactericidal antibiotics, pho-
35–43. todynamic therapy, and beyond, FEMS Microbiol. Rev. 37 (2013) 955–989.
[27] T.N. Demidova-Rice, M.R. Hamblin, I.M. Herman, Acute and impaired wound [55] M. Terashvili, P.F. Pratt, D. Gebremedhin, J. Narayanan, D.R. Harder, Reactive oxy-
healing: pathophysiology and current methods for drug delivery, part 2: role of gen species cerebral autoregulation in health and disease, Pediatr. Clin. N. Am. 53
growth factors in normal and pathological wound healing: therapeutic potential (2006) 1029–1037 (xi).
and methods of delivery, Adv. Skin Wound Care 25 (2012) 349–370. [56] A.A. Alfadda, R.M. Sallam, Reactive oxygen species in health and disease, Biomed.
[28] B. Hinz, Formation and function of the myofibroblast during tissue repair, J. Res. Int. 2012 (2012).
Investig. Dermatol. 127 (2007) 526–537. [57] D.O. Schairer, J.S. Chouake, J.D. Nosanchuk, A.J. Friedman, The potential of nitric
[29] A.A. Bahar, D. Ren, Antimicrobial peptides, Pharmaceuticals (Basel) 6 (2013) oxide releasing therapies as antimicrobial agents, Virulence 3 (2012) 271–279.
1543–1575. [58] H. Zhu, B. Ka, F. Murad, Nitric oxide accelerates the recovery from burn wounds,
[30] M. Xiong, M. Chen, J. Zhang, Rational evolution of antimicrobial peptides contain- World J. Surg. 31 (2007) 624–631.
ing unnatural amino acids to combat burn wound infections, Chem. Biol. Drug [59] H. Zhu, X. Wei, K. Bian, F. Murad, Effects of nitric oxide on skin burn wound healing,
Des. 88 (3) (2016 Sep) 404–410, http://dx.doi.org/10.1111/cbdd.12768. J. Burn Care Res. 29 (2008) 804–814.
[31] G. Zhong, J. Cheng, Z.C. Liang, L. Xu, W. Lou, C. Bao, Z.Y. Ong, H. Dong, Y.Y. Yang, W. [60] J.A. Lemire, J.J. Harrison, R.J. Turner, Antimicrobial activity of metals: mechanisms,
Fan, Short synthetic beta-sheet antimicrobial peptides for the treatment of molecular targets and applications, Nat. Rev. Microbiol. 11 (2013) 371–384.
multidrug-resistant Pseudomonas aeruginosa burn wound infections, Adv. Healthc. [61] A. Heyneman, H. Hoeksema, D. Vandekerckhove, A. Pirayesh, S. Monstrey, The role
Mater. 6 (2017). of silver sulphadiazine in the conservative treatment of partial thickness burn
[32] S. Yoshida, J.O. Lee, K. Nakamura, S. Suzuki, D.N. Hendon, M. Kobayashi, F. Suzuki, wounds: a systematic review, Burns. 42 (7) (2016 Nov) 1377–1386, http://dx.
Lineage — CD34+CD31+ cells that appear in association with severe burn injury doi.org/10.1016/j.burns.2016.03.029.
58 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

[62] S. Ganatra, D. Cohen, Silver nitrate burn of the lower lip: a case report, Gen. Dent. [90] P.S. Nunes, R.L. Albuquerque Jr., D.R. Cavalcante, M.D. Dantas, J.C. Cardoso, M.S.
64 (2016) 75–77. Bezerra, J.C. Souza, M.R. Serafini, L.J. Quitans Jr., L.R. Bonjardim, A.A. Araujo,
[63] M. Mohseni, A. Shamloo, Z. Aghababaei, M. Vossoughi, H. Moravvej, Antimicrobial Collagen-based films containing liposome-loaded usnic acid as dressing for dermal
wound dressing containing silver sulfadiazine with high biocompatibility: in vitro burn healing, J Biomed Biotechnol 2011 (2011) 761593.
study, Artif Organs. 40 (8) (2016 Aug) 765–773, http://dx.doi.org/10.1111/aor. [91] R.M. Dale, G. Schnell, J.P. Wong, Therapeutic efficacy of “nubiotics” against burn
12682. wound infection by Pseudomonas aeruginosa, Antimicrob. Agents Chemother. 48
[64] P. Domenico, L. Baldassarri, P.E. Schoch, K. Kaehler, M. Sasatsu, B.A. Cunha, Activi- (2004) 2918–2923.
ties of bismuth thiols against staphylococci and staphylococcal biofilms, [92] A.A. Hammond, K.G. Miller, C.J. Kruczek, J. Dertien, J.A. Colmer-Hamood, J.A.
Antimicrob. Agents Chemother. 45 (2001) 1417–1421. Griswold, A.R. Horswill, A.N. Hamood, An in vitro biofilm model to examine the ef-
[65] M.A. Nassar, H.M. Eldien, H.S. Tawab, T.H. Saleem, H.M. Omar, A.Y. Nassar, M.R. fect of antibiotic ointments on biofilms produced by burn wound bacterial isolates,
Hussein, Time-dependent morphological and biochemical changes following cuta- Burns 37 (2011) 312–321.
neous thermal burn injury and their modulation by copper nicotinate complex: an [93] R. Frei, A.S. Breitbach, H.E. Blackwell, 2-Aminobenzimidazole derivatives strongly
animal model, Ultrastruct. Pathol. 36 (2012) 343–355. inhibit and disperse Pseudomonas aeruginosa biofilms, Angew. Chem. 51 (2012)
[66] V.K. Champagne, D.J. Helfritch, A demonstration of the antimicrobial effectiveness 5226–5229.
of various copper surfaces, J. Biol. Eng. 7 (2013) 1. [94] D. Bialoszewski, A. Pietruczuk-Padzik, A. Kalicinska, E. Bocian, M. Czajkowska, B.
[67] V.B. Sudha, S. Ganesan, G.P. Pazhani, T. Ramamurthy, G.B. Nair, P. Bukowska, S. Tyski, Activity of ozonated water and ozone against Staphylococcus
Venkatasubramanian, Storing drinking-water in copper pots kills contaminating aureus and Pseudomonas aeruginosa biofilms, Med. Sci. Monit. 17 (2011)
diarrhoeagenic bacteria, J. Health Popul. Nutr. 30 (2012) 17–21. BR339–344.
[68] M.V. Dzyuba, A.V. Mardanov, A.V. Beletskii, T.V. Kolganova, M.V. Sukhacheva, A.A. [95] G.V. Sunnen, Method and apparatus for the deactivation of bacterial and fungal
Shelenkov, V.M. Gorlenko, B.B. Kuznetsov, K.G. Skryabin, Reconstruction of iron toxins in wounds, and for the disruption of wound biofilms, Google Patents, 2010.
metabolism pathways of bacteria Magnetospirillum aberrantis SpK spp. based on [96] S.A. Leiman, J.M. May, M.D. Lebar, D. Kahne, R. Kolter, R. Losick, D-amino acids in-
sequenced genome analysis, Dokl. Biol. Sci. 444 (2012) 202–205. directly inhibit biofilm formation in Bacillus subtilis by interfering with protein syn-
[69] K. DeLeon, F. Balldin, C. Watters, A. Hamood, J. Griswold, S. Sreedharan, K.P. thesis, J. Bacteriol. 195 (2013) 5391–5395.
Rumbaugh, Gallium maltolate treatment eradicates Pseudomonas aeruginosa infec- [97] H.S. Joo, M. Otto, Molecular basis of in vivo biofilm formation by bacterial patho-
tion in thermally injured mice, Antimicrob. Agents Chemother. 53 (2009) gens, Chem. Biol. 19 (2012) 1503–1513.
1331–1337. [98] S.G. Manuel, C. Ragunath, H.B. Sait, E.A. Izano, J.B. Kaplan, N. Ramasubbu, Role of
[70] H.S. Mangat, T.L. Stewart, L. Dibden, E.E. Tredget, Complications of chlorine inhala- active-site residues of dispersin B, a biofilm-releasing beta-hexosaminidase from
tion in a pediatric chemical burn patient: a case report, J. Burn Care Res. 33 (2012) a periodontal pathogen, in substrate hydrolysis, FEBS J. 274 (2007) 5987–5999.
e216–221. [99] G. Donelli, I. Francolini, D. Romoli, E. Guaglianone, A. Piozzi, C. Ragunath, J.B.
[71] E.B. Yuksel, A.M. Yildirim, A. Bal, T. Kuloglu, The effect of different topical agents Kaplan, Synergistic activity of dispersin B and cefamandole nafate in inhibition of
(silver sulfadiazine, povidone-iodine, and sodium chloride 0.9%) on burn injuries staphylococcal biofilm growth on polyurethanes, Antimicrob. Agents Chemother.
in rats, Plast. Surg. Int. 2014 (2014) 907082. 51 (2007) 2733–2740.
[72] Y.S. Lau, P. Brooks, Innovative use of povidone-iodine to guide burn wound de- [100] G.M. Soares, L.C. Figueiredo, M. Faveri, S.C. Cortelli, P.M. Duarte, M. Feres, Mecha-
bridement and predict the success of biobrane as a definitive treatment for nisms of action of systemic antibiotics used in periodontal treatment and mecha-
burns, Adv. Skin Wound Care 27 (2014) 111–113. nisms of bacterial resistance to these drugs, J. Appl. Oral Sci. 20 (2012) 295–309.
[73] G.T. Rodeheaver, C.B. Wheeler, J.A. Vensko, M.T. Edgerton, R.F. Edlich, Extinguishing [101] H. Heidari, M. Emaneini, H. Dabiri, F. Jabalameli, Virulence factors, antimicrobial re-
the flaming burn victim, JACEP 8 (1979) 307–311. sistance pattern and molecular analysis of Enterococcal strains isolated from burn
[74] I. Younes, M. Rinaudo, Chitin and chitosan preparation from marine sources. Struc- patients, Microb. Pathog. 90 (2016) 93–97.
ture, properties and applications, Mar. Drugs 13 (2015) 1133–1174. [102] S. Ohadian Moghadam, M.R. Pourmand, F. Aminharati, Biofilm formation and anti-
[75] A.S. Moghaddam, A. Raji, J. Movaffagh, A.T. Yazdi, M. Mahmoudi, Effects of autolo- microbial resistance in methicillin-resistant Staphylococcus aureus isolated from
gous keratinocyte cell spray with and without chitosan on third degree burn burn patients, Iran, J. Infect. Dev. Countries 8 (2014) 1511–1517.
healing: an animal experiment, Wounds 26 (2014) 109–117. [103] E.J. Gudina, J.A. Teixeira, L.R. Rodrigues, Biosurfactants produced by marine micro-
[76] T. Maisch, C. Bosl, R.M. Szeimies, N. Lehn, C. Abels, Photodynamic effects of novel organisms with therapeutic applications, Mar. Drugs 14 (2016).
XF porphyrin derivatives on prokaryotic and eukaryotic cells, Antimicrob. Agents [104] M. Jebur, Therapeutic efficacy of Lactobacillus acidophilus against bacterial isolates
Chemother. 49 (2005) 1542–1552. from burn wounds, N. Am. J. Med. Sci. 2 (2010) 586–591.
[77] N. Ooi, K. Miller, C. Randall, W. Rhys-Williams, W. Love, I. Chopra, XF-70 and XF-73, [105] R.E. Sockett, Predatory lifestyle of Bdellovibrio bacteriovorus, Annu. Rev. Microbiol.
novel antibacterial agents active against slow-growing and non-dividing cultures 63 (2009) 523–539.
of Staphylococcus aureus including biofilms, J. Antimicrob. Chemother. (2009), [106] Z. Pasternak, M. Njagi, Y. Shani, R. Chanyi, O. Rotem, M.N. Lurie-Weinberger, S.
dkp409. Koval, S. Pietrokovski, U. Gophna, E. Jurkevitch, In and out: an analysis of epibiotic
[78] N. Ooi, K. Miller, C. Randall, W. Rhys-Williams, W. Love, I. Chopra, XF-70 and XF-73, vs periplasmic bacterial predators, ISME J. 8 (2014) 625–635.
novel antibacterial agents active against slow-growing and non-dividing cultures [107] V. Iebba, V. Totino, F. Santangelo, A. Gagliardi, L. Ciotoli, A. Virga, C. Ambrosi, M.
of Staphylococcus aureus including biofilms, J. Antimicrob. Chemother. 65 (2010) Pompili, R.V. De Biase, L. Selan, M. Artini, F. Pantanella, F. Mura, C. Passariello, M.
72–78. Nicoletti, L. Nencioni, M. Trancassini, S. Quattrucci, S. Schippa, Bdellovibrio
[79] K.B. Pandey, S.I. Rizvi, Plant polyphenols as dietary antioxidants in human health bacteriovorus directly attacks Pseudomonas aeruginosa and Staphylococcus aureus
and disease, Oxidative Med. Cell. Longev. 2 (2009) 270–278. cystic fibrosis isolates, Front. Microbiol. 5 (2014) 280.
[80] The home reference to holistic health & healing: easy-to-use natural remedies, [108] K. Harini, V. Ajila, S. Hegde, Bdellovibrio bacteriovorus: a future antimicrobial agent?
herbs, flower essences, essential oils, supplements, and therapeutic practices for J. Indian Soc. Periodontol. 17 (2013) 823–825.
health, happiness, and well-being, Libr. J. 140 (2015) 106–107. [109] M. Karimi, H. Mirshekari, S.M. Moosavi Basri, S. Bahrami, M. Moghoofei, M.R.
[81] M. Sienkiewicz, M. Łysakowska, M. Pastuszka, W. Bienias, E. Kowalczyk, The poten- Hamblin, Bacteriophages and phage-inspired nanocarriers for targeted delivery
tial of use basil and rosemary essential oils as effective antibacterial agents, Mole- of therapeutic cargos, Adv. Drug Deliv. Rev. 106 (Pt A) (2016 Nov 15) 45–62,
cules 18 (2013) 9334–9351. http://dx.doi.org/10.1016/j.addr.2016.03.003.
[82] M. Fertah, A. Belfkira, Brouillette, Extraction and characterization of sodium [110] D. Hoff-Lenczewska, M. Kawecki, J. Glik, A. Klama-Baryla, M. Nowak, The potential
alginate from Moroccan Laminaria digitata brown seaweed, Arab. J. Chem. 10 (Sup- of bacteriophages in the treatment of burn wounds, Pol. Przegl. Chir. 85 (2013)
plement 2) (2017) S3707–S3714. 615–618.
[83] J.G. Thomas, W. Slone, S. Linton, T. Okel, L. Corum, S.L. Percival, In vitro antimicro- [111] S. Kumari, K. Harjai, S. Chhibber, Bacteriophage versus antimicrobial agents for the
bial efficacy of a silver alginate dressing on burn wound isolates, J. Wound Care 20 treatment of murine burn wound infection caused by Klebsiella pneumoniae B5055,
(2011) 124,126–128. J. Med. Microbiol. 60 (2011) 205–210.
[84] M.I. Brachkova, P. Marques, J. Rocha, B. Sepodes, M.A. Duarte, J.F. Pinto, Alginate [112] T. Rose, G. Verbeken, D. De Vos, M. Merabishvili, M. Vaneechoutte, R. Lavigne, S.
films containing Lactobacillus plantarum as wound dressing for prevention of Jennes, M. Zizi, J.P. Pirnay, Experimental phage therapy of burn wound infection:
burn infection, J. Hosp. Infect. 79 (2011) 375–377. difficult first steps, Int. J. Burns Trauma 4 (2014) 66–73.
[85] Q.C. Truong-Bolduc, R.A. Villet, Z.A. Estabrooks, D.C. Hooper, Native efflux pumps [113] E. Lazareva, S. Smirnov, V. Khvatov, T. Spiridonova, E. Bitkova, O. Darbeeva, L.
contribute resistance to antimicrobials of skin and the ability of Staphylococcus au- Maĭskaia, R. Parfeniuk, D. Men'shikov, Efficacy of bacteriophages in complex treat-
reus to colonize skin, J Infect Dis 209 (2014) 1485–1493. ment of patients with burn wounds, Antibiot. Khimioter. 46 (2000) 10–14.
[86] P.G. Bowler, B.I. Duerden, D.G. Armstrong, Wound microbiology and associated ap- [114] P. Chadha, O.P. Katare, S. Chhibber, In vivo efficacy of single phage versus phage
proaches to wound management, Clin. Microbiol. Rev. 14 (2001) 244–269. cocktail in resolving burn wound infection in BALB/c mice, Microb. Pathog. 99
[87] P.A. Falcone, H.N. Harrison, G.O. Sowemimo, G.P. Reading, Mafenide acetate con- (2016) 68–77.
centrations and bacteriostasis in experimental burn wounds treated with a [115] Z. Golkar, O. Bagasra, N. Jamil, Experimental phage therapy on multiple drug resis-
three-layered laminated mafenide-saline dressing, Ann. Plast. Surg. 5 (1980) tant Pseudomonas aeruginosa infection in mice, J. Antivirals Antiretrovirals 2013
266–269. (2013).
[88] M. Zahmatkesh, M.J. Manesh, R. Babashahabi, Effect of Olea ointment and acetate [116] A.V. Holguín, G. Rangel, V. Clavijo, C. Prada, M. Mantilla, M.C. Gomez, E. Kutter, C.
mafenide on burn wounds — a randomized clinical trial, Iran. J. Nurs. midwifery Taylor, P.C. Fineran, A.F.G. Barrios, Phage ΦPan70, a putative temperate phage, con-
Res. 20 (2015) 599–603. trols Pseudomonas aeruginosa in planktonic, biofilm and burn mouse model assays,
[89] E. Ulkur, O. Oncul, H. Karagoz, E. Yeniz, B. Celikoz, Comparison of silver-coated Viruses 7 (2015) 4602–4623.
dressing (Acticoat), chlorhexidine acetate 0.5% (Bactigrass), and fusidic acid 2% [117] A. Yadav, A. Gupta, G.K. Keshri, S. Verma, S.K. Sharma, S.B. Singh,
(Fucidin) for topical antibacterial effect in methicillin-resistant Staphylococci- Photobiomodulatory effects of superpulsed 904 nm laser therapy on bioenergetics
contaminated, full-skin thickness rat burn wounds, Burns 31 (2005) 874–877. status in burn wound healing, J. Photochem. Photobiol. B Biol. 162 (2016) 77–85.
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 59

[118] T. Dai, A. Gupta, Y.-Y. Huang, R. Yin, C.K. Murray, M.S. Vrahas, M.E. Sherwood, G.P. [142] A.J.P. Clover, A.H.S. Kumar, M. Isakson, D. Whelan, A. Stocca, B.M. Gleeson, N.M.
Tegos, M.R. Hamblin, Blue light rescues mice from potentially fatal Pseudomonas Caplice, Allogeneic mesenchymal stem cells, but not culture modified monocytes,
aeruginosa burn infection: efficacy, safety, and mechanism of action, Antimicrob. improve burn wound healing, Burns 41 (2015) 548–557.
Agents Chemother. 57 (2013) 1238–1245. [143] Y.-W. Chen, T.T. Scutaru, N. Ghetu, E. Carasevici, C.D. Lupascu, D. Ferariu, D. Pieptu,
[119] M.H.C.V. Catão, R.O. Costa, C.F.W. Nonaka, R.L.C.A. Junior, I.R.R.S. Costa, Green LED C.-G. Coman, M. Danciu, The effects of adipose-derived stem cell–differentiated ad-
light has anti-inflammatory effects on burns in rats, Burns 42 (2016) 392–396. ipocytes on skin burn wound healing in rats, J. Burn Care Res. 38 (2017) 1–10.
[120] H.-W. Chiu, C.-H. Chen, J.-N. Chang, C.-H. Chen, Y.-H. Hsu, Far-infrared promotes [144] I. Kranz, J.B. Gonzalez, I. Dorfel, M. Gemeinert, M. Griepentrog, D. Klaffke, C. Knabe,
burn wound healing by suppressing NLRP3 inflammasome caused by enhanced W. Osterle, U. Gross, Biological response to micron- and nanometer-sized particles
autophagy, J. Mol. Med. 94 (2016) 809–819. known as potential wear products from artificial hip joints: part II: reaction of mu-
[121] P.C.L. Silveira, K.B. Ferreira, F.R. da Rocha, B.L.S. Pieri, G.S. Pedroso, C.T. De Souza, rine macrophages to corundum particles of different size distributions, J. Biomed.
R.T. Nesi, R.A. Pinho, Effect of low-power laser (LPL) and light-emitting diode Mater. Res. A 89 (2009) 390–401.
(LED) on inflammatory response in burn wound healing, Inflammation 39 [145] H. Malekzad, H. Mirshekari, P. Sahandi Zangabad, S.M. Moosavi Basri, F. Baniasadi,
(2016) 1395–1404. M. Sharifi Aghdam, M. Karimi, M.R. Hamblin, Plant protein-based hydrophobic fine
[122] B. Rathnakar, V. Prabhu, B.S.S. Rao, S. Chandra, S. Rai, K.K. Mahato, Regulation of and ultrafine carrier particles in drug delivery systems, Crit. Rev. Biotechnol.
cellular marker modulated upon irradiation of low power laser light in burn in- (2017) 1–21.
jured mice, Proc. SPIE 10013, SPIE BioPhotonics Australasia, 100132Z, International [146] M. Karimi, H. Zare, A. Bakhshian Nik, N. Yazdani, M. Hamrang, E. Mohamed, P.
Society for Optics and Photonics, 9 December 2016 http://dx.doi.org/10.1117/12. Sahandi Zangabad, S.M. Moosavi Basri, L. Bakhtiari, M.R. Hamblin, Nanotechnology
2242919. in diagnosis and treatment of coronary artery disease, Nanomedicine 11 (2016)
[123] Y. Zhang, Y. Zhu, A. Gupta, Y. Huang, C.K. Murray, M.S. Vrahas, M.E. Sherwood, D.G. 513–530.
Baer, M.R. Hamblin, T. Dai, Antimicrobial blue light therapy for multidrug-resistant [147] M. Karimi, S. Bahrami, S.B. Ravari, P.S. Zangabad, H. Mirshekari, M. Bozorgomid, S.
Acinetobacter baumannii infection in a mouse burn model: implications for prophy- Shahreza, M. Sori, M.R. Hamblin, Albumin nanostructures as advanced drug deliv-
laxis and treatment of combat-related wound infections, J Infect Dis 209 (2013) ery systems, Expert Opin. Drug Deliv. 13 (2016) 1609–1623.
1963–1971. [148] X. Zhu, A.F. Radovic-Moreno, J. Wu, R. Langer, J. Shi, Nanomedicine in the manage-
[124] C. Bay, K. Togsverd-Bo, C.M. Lerche, M. Haedersdal, Skin tumor development after ment of microbial infection — overview and perspectives, Nano Today 9 (2014)
UV irradiation and photodynamic therapy is unaffected by short-term pretreat- 478–498.
ment with 5-fluorouracil, imiquimod and calcipotriol. An experimental hairless [149] F. Jia, X. Liu, L. Li, S. Mallapragada, B. Narasimhan, Q. Wang, Multifunctional nano-
mouse study, J. Photochem. Photobiol. B Biol. 154 (2016) 34–39. particles for targeted delivery of immune activating and cancer therapeutic agents,
[125] P. Avci, A. Gupta, M. Sadasivam, D. Vecchio, Z. Pam, N. Pam, M.R. Hamblin, Low- J. Control. Release 172 (2013) 1020–1034.
level laser (light) therapy (LLLT) in skin: stimulating, healing, restoring, Semin. [150] I. Tocco, B. Zavan, F. Bassetto, V. Vindigni, Nanotechnology-based therapies for skin
Cutan. Med. Surg. 32 (2013) 41–52. wound regeneration, J. Nanomater. 2012 (2012) 4.
[126] M.H. Gold, A. Andriessen, J. Biron, H. Andriessen, Clinical efficacy of self-applied [151] D.M. Smith, J.K. Simon, J.R. Baker Jr., Applications of nanotechnology for immunol-
blue light therapy for mild-to-moderate facial acne, J. Clin. Aesthet. Dermatol. 2 ogy, Nat. Rev. Immunol. 13 (2013) 592–605.
(2009) 44–50. [152] C.S. Yah, G.S. Simate, Nanoparticles as potential new generation broad spectrum
[127] M.S. Khan, H.N. Abdelhamid, H.-F. Wu, Near infrared (NIR) laser mediated antimicrobial agents, Daru 23 (2015) 43.
surface activation of graphene oxide nanoflakes for efficient antibacterial, [153] L. Zhao, A. Seth, N. Wibowo, C.X. Zhao, N. Mitter, C. Yu, A.P. Middelberg, Nanopar-
antifungal and wound healing treatment, Colloids Surf. B: Biointerfaces 127 ticle vaccines, Vaccine 32 (2014) 327–337.
(2015) 281–291. [154] I. Kalashnikova, S. Das, S. Seal, Nanomaterials for wound healing: scope and ad-
[128] N. Nafee, A. Youssef, H. El-Gowelli, H. Asem, S. Kandil, Antibiotic-free vancement, Nanomedicine (London) 10 (2015) 2593–2612.
nanotherapeutics: hypericin nanoparticles thereof for improved in vitro and [155] I.U. Khan, C.A. Serra, N. Anton, T.F. Vandamme, Production of nanoparticle drug de-
in vivo antimicrobial photodynamic therapy and wound healing, Int. J. Pharm. livery systems with microfluidics tools, Expert Opin. Drug Deliv. 12 (2015)
454 (2013) 249–258. 547–562.
[129] V.G. Garcia, M.A. de Lima, T. Okamoto, L.A. Milanezi, E.C.G. Júnior, L.A. Fernandes, [156] M. Fathi, P.S. Zangabad, A. Aghanejad, J. Barar, H. Erfan-Niya, Y. Omidi, Folate-
J.M. de Almeida, L.H. Theodoro, Effect of photodynamic therapy on the healing of conjugated thermosensitive O-maleoyl modified chitosan micellar nanopar-
cutaneous third-degree-burn: histological study in rats, Lasers Med. Sci. 25 ticles for targeted delivery of erlotinib, Carbohydr. Polym. 172 (2017)
(2010) 221–228. 130–141.
[130] N.A. Aleem, M. Aslam, M.F. Zahid, A.J. Rahman, F.U. Rehman, Treatment of burn [157] M. Tokuda, M. Yamane, S.C. Thickett, H. Minami, P.B. Zetterlund, Synthesis of poly-
wound infection using ultraviolet light: a case report, J. Am. Coll. Clin. Wound meric nanoparticles containing reduced graphene oxide nanosheets stabilized by
Specialists 5 (2013) 19–22. poly(ionic liquid) using miniemulsion polymerization, Soft Matter 12 (2016)
[131] Z. Bohrerova, K.G. Linden, Assessment of DNA damage and repair in Mycobacterium 3955–3962.
terrae after exposure to UV irradiation, J. Appl. Microbiol. 101 (2006) 995–1001. [158] J.C. Gardner, H. Wu, J.G. Noel, B.J. Ramser, L. Pitstick, A. Saito, N.M. Nikolaidis,
[132] A. Gupta, P. Avci, T. Dai, Y.Y. Huang, M.R. Hamblin, Ultraviolet radiation in wound F.X. McCormack, Keratinocyte growth factor supports pulmonary innate im-
care: sterilization and stimulation, Adv. Wound Care 2 (2013) 422–437. mune defense through maintenance of alveolar antimicrobial protein levels
[133] C. Ferraresi, M.R. Hamblin, N.A. Parizotto, Low-level laser (light) therapy (LLLT) on and macrophage function, Am. J. Phys. Lung Cell. Mol. Phys. 310 (2016)
muscle tissue: performance, fatigue and repair benefited by the power of light, L868–879.
Photon. Lasers Med. 1 (2012) 267–286. [159] Z.G. Feng, S.F. Pang, D.J. Guo, Y.T. Yang, B. Liu, J.W. Wang, K.Q. Zheng, Y. Lin, Recom-
[134] E.J. Brisbois, J. Bayliss, J. Wu, T.C. Major, C. Xi, S.C. Wang, R.H. Bartlett, H. Handa, binant keratinocyte growth factor 1 in tobacco potentially promotes wound
M.E. Meyerhoff, Optimized polymeric film-based nitric oxide delivery inhibits healing in diabetic rats, Biomed. Res. Int. 2014 (2014) 579632.
bacterial growth in a mouse burn wound model, Acta. Biomater. 10 (2014) [160] P. Koria, H. Yagi, Y. Kitagawa, Z. Megeed, Y. Nahmias, R. Sheridan, M.L. Yarmush,
4136–4142. Self-assembling elastin-like peptides growth factor chimeric nanoparticles for the
[135] D. Rünzler, A.M. Weihs, H. Redl, A.H. Teuschl, C. Fuchs, Shockwave Treatment treatment of chronic wounds, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 1034–1039.
Enhances Proliferation and Improves Wound Healing via Purinergic Signaling [161] K.K. Chereddy, G. Vandermeulen, V. Preat, PLGA based drug delivery systems:
Induced ERK1/2 Pathway, 2016. promising carriers for wound healing activity, Wound Repair Regen. 24 (2016)
[136] M.S. Fantinati, D.E.O. Mendonça, A.M.M. Fantinati, B.F.D. Santos, J.C.O. Reis, C.L. 223–236.
Afonso, M.C. Vinaud, L. Júnior, R. de Souza, Low intensity ultrasound therapy in- [162] Y. Zhang, C. Wischke, S. Mittal, A. Mitra, S.P. Schwendeman, Design of controlled
duces angiogenesis and persistent inflammation in the chronic phase of the release PLGA microspheres for hydrophobic fenretinide, Mol. Pharm. 13 (8)
healing process of third degree burn wounds experimentally induced in diabetic (2016 Aug 1) 2622–2630, http://dx.doi.org/10.1021/acs.molpharmaceut.5b00961.
and non-diabetic rats, Acta Cir. Bras. 31 (2016) 463–471. [163] E. Yuksel, A. Karakecili, T.T. Demirtas, M. Gumusderelioglu, Preparation of bioactive
[137] M. Karimi, S. Bahrami, H. Mirshekari, S.M.M. Basri, A.B. Nik, A.R. Aref, M. Akbari, and antimicrobial PLGA membranes by magainin II/EGF functionalization, Int. J.
M.R. Hamblin, Microfluidic systems for stem cell-based neural tissue engineering, Biol. Macromol. 86 (2016) 162–168.
Lab Chip 16 (2016) 2551–2571. [164] K.K. Chereddy, C.H. Her, M. Comune, C. Moia, A. Lopes, P.E. Porporato, J. Vanacker,
[138] P. Foubert, M. Doyle-Eisele, A. Gonzalez, F. Berard, W. Weber, D. Zafra, Z. Alfonso, S. M.C. Lam, L. Steinstraesser, P. Sonveaux, H. Zhu, L.S. Ferreira, G. Vandermeulen, V.
Zhao, M. Tenenhaus, J.K. Fraser, Development of a combined radiation and full Preat, PLGA nanoparticles loaded with host defense peptide LL37 promote
thickness burn injury minipig model to study the effects of uncultured adipose- wound healing, J. Control. Release 194 (2014) 138–147.
derived regenerative cell therapy in wound healing, Int. J. Radiat. Biol. (2016) [165] A.J. Duplantier, M.L. van Hoek, The human cathelicidin antimicrobial peptide LL-37
1–11. as a potential treatment for polymicrobial infected wounds, Front. Immunol. 4
[139] R.-H. Yang, S.-H. Qi, S.-B. Ruan, Z.-P. Lin, Y. Lin, F.-G. Zhang, X.-D. Chen, J.-L. Xie, (2013) 143.
EGFL7-overexpressing epidermal stem cells promotes fibroblast proliferation and [166] M. Mofazzal Jahromi, M. Karimi, K. Azadmanesh, H. Naderi Manesh, Z. Hassan, S.
migration via mediating cell adhesion and strengthening cytoskeleton, Mol. Cell. Moazzeni, The effect of chitosan-tripolyphosphate nanoparticles on maturation
Biochem. 423 (2016) 1–8. and function of dendritic cells, Comp. Clin. Pathol. (2013) 1–7.
[140] J.M. Bliley, A. Argenta, L. Satish, M.M. McLaughlin, A. Dees, C. Tompkins-Rhoades, [167] A. Shrestha, M.R. Hamblin, A. Kishen, Characterization of a conjugate between rose
K.G. Marra, J.P. Rubin, Administration of adipose-derived stem cells enhances vas- bengal and chitosan for targeted antibiofilm and tissue stabilization effects as a po-
cularity, induces collagen deposition, and dermal adipogenesis in burn wounds, tential treatment of infected dentin, Antimicrob. Agents Chemother. 56 (2012)
Burns 42 (2016) 1212–1222. 4876–4884.
[141] C. Hu, X. Yong, C. Li, M. Lü, D. Liu, L. Chen, J. Hu, M. Teng, D. Zhang, Y. Fan, G. Liang, [168] A. Shrestha, M.R. Hamblin, A. Kishen, Photoactivated rose bengal functionalized
CXCL12/CXCR4 axis promotes mesenchymal stem cell mobilization to burn chitosan nanoparticles produce antibacterial/biofilm activity and stabilize dentin-
wounds and contributes to wound repair, J. Surg. Res. 183 (2013) 427–434. collagen, Nanomedicine 10 (2014) 491–501.
60 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

[169] T. Dai, M. Tanaka, Y.Y. Huang, M.R. Hamblin, Chitosan preparations for wounds and [196] S. Sugumar, A. Mukherjee, N. Chandrasekaran, Eucalyptus oil nanoemulsion-
burns: antimicrobial and wound-healing effects, Expert Rev. Anti-Infect. Ther. 9 impregnated chitosan film: antibacterial effects against a clinical pathogen, Staph-
(2011) 857–879. ylococcus aureus, in vitro, Int. J. Nanomedicine 10 (Suppl. 1) (2015) 67–75.
[170] T. Dai, G.P. Tegos, M. Burkatovskaya, A.P. Castano, M.R. Hamblin, Chitosan acetate [197] Z.L. Tyrrell, Y. Shen, M. Radosz, Fabrication of micellar nanoparticles for drug deliv-
bandage as a topical antimicrobial dressing for infected burns, Antimicrob. Agents ery through the self-assembly of block copolymers, Prog. Polym. Sci. 35 (2010)
Chemother. 53 (2009) 393–400. 1128–1143.
[171] M. Burkatovskaya, G.P. Tegos, E. Swietlik, T.N. Demidova, A.P. Castano, M.R. [198] E. Dellera, M.C. Bonferoni, G. Sandri, S. Rossi, F. Ferrari, C. Del Fante, C. Perotti, P.
Hamblin, Use of chitosan bandage to prevent fatal infections developing from Grisoli, C. Caramella, Development of chitosan oleate ionic micelles loaded with sil-
highly contaminated wounds in mice, Biomaterials 27 (2006) 4157–4164. ver sulfadiazine to be associated with platelet lysate for application in wound
[172] R.M. Baxter, T. Dai, J. Kimball, E. Wang, M.R. Hamblin, W.P. Wiesmann, S.J. healing, Eur. J. Pharm. Biopharm. 88 (2014) 643–650.
McCarthy, S.M. Baker, Chitosan dressing promotes healing in third degree burns [199] A. Akbarzadeh, R. Rezaei-Sadabady, S. Davaran, S.W. Joo, N. Zarghami, Y.
in mice: gene expression analysis shows biphasic effects for rapid tissue regener- Hanifehpour, M. Samiei, M. Kouhi, K. Nejati-Koshki, Liposome: classification, prep-
ation and decreased fibrotic signaling, J. Biomed. Mater. Res. A 101 (2013) aration, and applications, Nanoscale Res. Lett. 8 (2013) 102.
340–348. [200] A.A. Naumov, Y.V. Shatalin, M.M. Potselueva, Effects of a nanocomplex containing
[173] M. Karimi, P. Avci, M. Ahi, T. Gazori, M.R. Hamblin, H. Naderi-Manesh, Evaluation of antioxidant, lipid, and amino acid on thermal burn wound surface, Bull. Exp. Biol.
chitosan-tripolyphosphate nanoparticles as a p-shRNA delivery vector: formula- Med. 149 (2010) 62–66.
tion, optimization and cellular uptake study, J. Nanopharm. Drug Deliv. 1 (2013) [201] M.G. Jeschke, G. Sandmann, C.C. Finnerty, D.N. Herndon, C.T. Pereira, T. Schubert, D.
266–278. Klein, The structure and composition of liposomes can affect skin regeneration,
[174] L. Huang, T. Dai, Y. Xuan, G.P. Tegos, M.R. Hamblin, Synergistic combination of chi- morphology and growth factor expression in acute wounds, Gene Ther. 12
tosan acetate with nanoparticle silver as a topical antimicrobial: efficacy against (2005) 1718–1724.
bacterial burn infections, Antimicrob. Agents Chemother. 55 (2011) 3432–3438. [202] M.G. Jeschke, G. Richter, F. Hofstadter, D.N. Herndon, J.R. Perez-Polo, K.W. Jauch,
[175] P. Anand, S.G. Thomas, A.B. Kunnumakkara, C. Sundaram, K.B. Harikumar, B. Sung, Non-viral liposomal keratinocyte growth factor (KGF) cDNA gene transfer im-
S.T. Tharakan, K. Misra, I.K. Priyadarsini, K.N. Rajasekharan, B.B. Aggarwal, Biologi- proves dermal and epidermal regeneration through stimulation of epithelial and
cal activities of curcumin and its analogues (congeners) made by man and mother mesenchymal factors, Gene Ther. 9 (2002) 1065–1074.
nature, Biochem. Pharmacol. 76 (2008) 1590–1611. [203] G. Yoon, J.W. Park, I.-S. Yoon, Solid lipid nanoparticles (SLNs) and nanostructured
[176] M.A. Mofazzal-Jahromi, Reza Mirnejad, Curcumin-loaded chitosan lipid carriers (NLCs): recent advances in drug delivery, J. Pharm. Investig. 43
tripolyphosphate nanoparticles as a safe, natural and effective antibiotic inhibits (2013) 353–362.
the infection of Staphylococcus aureus and Pseudomonas aeruginosa in vivo, Iran. [204] M. Uner, G. Yener, Importance of solid lipid nanoparticles (SLN) in various admin-
J. Biotechnol. 12 (2014) 1–8. istration routes and future perspectives, Int. J. Nanomedicine 2 (2007) 289–300.
[177] M. Xie, D. Fan, Z. Zhao, Z. Li, G. Li, Y. Chen, X. He, A. Chen, J. Li, X. Lin, M. Zhi, Y. Li, P. [205] E. Rostami, S. Kashanian, A.H. Azandaryani, H. Faramarzi, J.E. Dolatabadi, K.
Lan, Nano-curcumin prepared via supercritical: improved anti-bacterial, anti- Omidfar, Drug targeting using solid lipid nanoparticles, Chem. Phys. Lipids 181
oxidant and anti-cancer efficacy, Int. J. Pharm. 496 (2015) 732–740. (2014) 56–61.
[178] Y.X. Xu, K.R. Pindolia, N. Janakiraman, C.J. Noth, R.A. Chapman, S.C. Gautam, [206] G. Sandri, M.C. Bonferoni, F. D'Autilia, S. Rossi, F. Ferrari, P. Grisoli, M. Sorrenti, L.
Curcumin, a compound with anti-inflammatory and anti-oxidant properties, Catenacci, C. Del Fante, C. Perotti, C. Caramella, Wound dressings based on silver
down-regulates chemokine expression in bone marrow stromal cells, Exp. sulfadiazine solid lipid nanoparticles for tissue repairing, Eur. J. Pharm. Biopharm.
Hematol. 25 (1997) 413–422. 84 (2013) 84–90.
[179] R.K. Bhawana, H.S. Basniwal, V.K. Buttar, N. Jain, Curcumin nanoparticles: prepara- [207] S. Kuchler, N.B. Wolf, S. Heilmann, G. Weindl, J. Helfmann, M.M. Yahya, C. Stein, M.
tion, characterization, and antimicrobial study, J. Agric. Food Chem. 59 (2011) Schafer-Korting, 3D-wound healing model: influence of morphine and solid lipid
2056–2061. nanoparticles, J. Biotechnol. 148 (2010) 24–30.
[180] E. Abbasi, S.F. Aval, A. Akbarzadeh, M. Milani, H.T. Nasrabadi, S.W. Joo, Y. [208] B. Zadeh, N. Barati, M. Hassani, F. Rahim, Development of solid lipid nanoparticles
Hanifehpour, K. Nejati-Koshki, R. Pashaei-Asl, Dendrimers: synthesis, applications, as Eschar delivery system for nitrofurazone using Taguchi design approach, Int. J.
and properties, Nanoscale Res. Lett. 9 (2014) 247. Res. Pharm. Sci. 1 (2010) 466–472.
[181] M. Kalomiraki, K. Thermos, N.A. Chaniotakis, Dendrimers as tunable vectors of drug [209] R.H. Müller, U. Alexiev, P. Sinambela, C.M. Keck, Nanostructured lipid carriers
delivery systems and biomedical and ocular applications, Int. J. Nanomedicine 11 (NLC): the second generation of solid lipid nanoparticles, in: N. Dragicevic, I.H.
(2016) 1–12. Maibach (Eds.), Percutaneous Penetration Enhancers Chemical Methods in Pene-
[182] X. Liu, W. Hao, C.N. Lok, Y.C. Wang, R. Zhang, K.K. Wong, Dendrimer encapsulation tration Enhancement: Nanocarriers, Springer Berlin Heidelberg, Berlin, Heidelberg
enhances anti-inflammatory efficacy of silver nanoparticles, J. Pediatr. Surg. 49 2016, pp. 161–185.
(2014) 1846–1851. [210] C.L. Fang, S.A. Al-Suwayeh, J.Y. Fang, Nanostructured lipid carriers (NLCs) for drug
[183] E. Caló, V.V. Khutoryanskiy, Biomedical applications of hydrogels: a review of pat- delivery and targeting, Recent Pat. Nanotechnol. 7 (2013) 41–55.
ents and commercial products, Eur. Polym. J. 65 (2015) 252–267. [211] M.A. Iqbal, S. Md, J.K. Sahni, S. Baboota, S. Dang, J. Ali, Nanostructured lipid carriers
[184] M. Hajimiri, S. Shahverdi, M.A. Esfandiari, B. Larijani, F. Atyabi, A. Rajabiani, A.R. system: recent advances in drug delivery, J. Drug Target. 20 (2012) 813–830.
Dehpour, M. Amini, R. Dinarvand, Preparation of hydrogel embedded polymer- [212] S. Khan, S. Baboota, J. Ali, S. Khan, R.S. Narang, J.K. Narang, Nanostructured lipid car-
growth factor conjugated nanoparticles as a diabetic wound dressing, Drug Dev. riers: an emerging platform for improving oral bioavailability of lipophilic drugs,
Ind. Pharm. 42 (2016) 707–719. Int. J. Pharm. Investig. 5 (2015) 182–191.
[185] M. Madaghiele, C. Demitri, A. Sannino, L. Ambrosio, Polymeric hydrogels for burn [213] A.I. Mendes, A.C. Silva, J.A. Catita, F. Cerqueira, C. Gabriel, C.M. Lopes, Miconazole-
wound care: advanced skin wound dressings and regenerative templates, Burns loaded nanostructured lipid carriers (NLC) for local delivery to the oral mucosa:
Trauma 2 (2015) 153. improving antifungal activity, Colloids Surf. B: Biointerfaces 111 (2013) 755–763.
[186] S. Anjum, A. Arora, M.S. Alam, B. Gupta, Development of antimicrobial and scar pre- [214] G. Gainza, D.C. Bonafonte, B. Moreno, J.J. Aguirre, F.B. Gutierrez, S. Villullas, J.L.
ventive chitosan hydrogel wound dressings, Int. J. Pharm. 508 (2016) 92–101. Pedraz, M. Igartua, R.M. Hernandez, The topical administration of rhEGF-loaded
[187] E.M. Ahmed, Hydrogel: preparation, characterization, and applications: a review, J. nanostructured lipid carriers (rhEGF-NLC) improves healing in a porcine full-
Adv. Res. 6 (2015) 105–121. thickness excisional wound model, J. Control. Release 197 (2015) 41–47.
[188] J. Parker, N. Mitrousis, M.S. Shoichet, Hydrogel for simultaneous tunable growth [215] G. Gainza, M. Pastor, J.J. Aguirre, S. Villullas, J.L. Pedraz, R.M. Hernandez, M. Igartua,
factor delivery and enhanced viability of encapsulated cells in vitro, A novel strategy for the treatment of chronic wounds based on the topical ad-
Biomacromolecules 17 (2016) 476–484. ministration of rhEGF-loaded lipid nanoparticles: in vitro bioactivity and
[189] A. Skardal, S.V. Murphy, K. Crowell, D. Mack, A. Atala, S. Soker, A tunable hydrogel in vivo effectiveness in healing-impaired db/db mice, J. Control. Release 185
system for long-term release of cell-secreted cytokines and bioprinted in situ (2014) 51–61.
wound cell delivery, J Biomed Mater Res B Appl Biomater (2016). [216] G. Gainza, W.S. Chu, R.H. Guy, J.L. Pedraz, R.M. Hernandez, B. Delgado-Charro, M.
[190] T. Kobayashi, M. Mizuta, N. Hiwatashi, Y. Kishimoto, T. Nakamura, S.I. Kanemaru, S. Igartua, Development and in vitro evaluation of lipid nanoparticle-based dressings
Hirano, Drug delivery system of basic fibroblast growth factor using gelatin hydro- for topical treatment of chronic wounds, Int. J. Pharm. 490 (2015) 404–411.
gel for restoration of acute vocal fold scar, Auris Nasus Larynx. 44 (1) (2017 Feb) [217] J. Lee, J. Kim, J. Go, J.H. Lee, D.W. Han, D. Hwang, J. Lee, Transdermal treatment of
86–92, http://dx.doi.org/10.1016/j.anl.2016.04.005. the surgical and burned wound skin via phytochemical-capped gold nanoparticles,
[191] M.C. Bonferoni, G. Sandri, E. Dellera, S. Rossi, F. Ferrari, M. Mori, C. Caramella, Ionic Colloids Surf. B: Biointerfaces 135 (2015) 166–174.
polymeric micelles based on chitosan and fatty acids and intended for wound [218] N. Volkova, M. Yukhta, O. Pavlovich, A. Goltsev, Application of cryopreserved fibro-
healing. Comparison of linoleic and oleic acid, Eur. J. Pharm. Biopharm. 87 (2014) blast culture with Au nanoparticles to treat burns, Nanoscale Res. Lett. 11 (2016) 22.
101–106. [219] W. Petersen, A. Rahmanian-Schwarz, J.-O. Werner, J. Schiefer, J. Rothenberger, G.
[192] M. Jaiswal, R. Dudhe, P. Sharma, Nanoemulsion: an advanced mode of drug deliv- Hübner, H.-E. Schaller, M. Held, The use of collagen-based matrices in the treat-
ery system, 3 Biotech 5 (2015) 123–127. ment of full-thickness wounds, Burns. 42 (6) (2016 Sep) 1257–1264, http://dx.
[193] E. Blanco, C.W. Kessinger, B.D. Sumer, J. Gao, Multifunctional micellar doi.org/10.1016/j.burns.2016.03.017.
nanomedicine for cancer therapy, Exp. Biol. Med. 234 (2009) 123–131. [220] O. Akturk, K. Kismet, A.C. Yasti, S. Kuru, M.E. Duymus, F. Kaya, M. Caydere, S.
[194] M. Jabbarzadegan, H. Rajayi, M.A. Mofazzal Jahromi, H. Yeganeh, M. Yousefi, Z. Hucumenoglu, D. Keskin, Collagen/gold nanoparticle nanocomposites: a potential
Muhammad Hassan, J. Majidi, Application of arteether-loaded polyurethane skin wound healing biomaterial, J. Biomater. Appl. 31 (2) (2016 Aug) 283–301,
nanomicelles to induce immune response in breast cancer model, Artif. Cells, http://dx.doi.org/10.1177/0885328216644536.
Nanomed. Biotechnol. (2016) 1–9. [221] R. Gyawali, S.A. Ibrahim, S.H. Abu Hasfa, S.Q. Smqadri, Y. Haik, Antimicrobial activ-
[195] Z. Song, H. Sun, Y. Yang, H. Jing, L. Yang, Y. Tong, C. Wei, Z. Wang, Q. Zou, H. Zeng, ity of copper alone and in combination with lactic acid against Escherichia coli
Enhanced efficacy and anti-biofilm activity of novel nanoemulsions against skin O157:H7 in laboratory medium and on the surface of lettuce and tomatoes, J.
burn wound multi-drug resistant MRSA infections, Nanomedicine (2016). Pathog. 2011 (2011) 650968.
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 61

[222] J. Ramyadevi, K. Jeyasubramanian, A. Marikani, G. Rajakumar, A.A. Rahuman, Syn- [251] V. Sivaranjani, P. Philominathan, Synthesize of titanium dioxide nanoparticles
thesis and antimicrobial activity of copper nanoparticles, Mater. Lett. 71 (2012) using Moringa oleifera leaves and evaluation of wound healing activity, Wound
114–116. Med. 12 (2016) 1–5.
[223] M.S. Usman, M.E. El Zowalaty, K. Shameli, N. Zainuddin, M. Salama, N.A. Ibrahim, [252] T. Santhoshkumar, A.A. Rahuman, C. Jayaseelan, G. Rajakumar, S. Marimuthu, A.V.
Synthesis, characterization, and antimicrobial properties of copper nanoparticles, Kirthi, K. Velayutham, J. Thomas, J. Venkatesan, S.K. Kim, Green synthesis of titani-
Int. J. Nanomedicine 8 (2013) 4467–4479. um dioxide nanoparticles using Psidium guajava extract and its antibacterial and
[224] A. Adhya, J. Bain, O. Ray, A. Hazra, S. Adhikari, G. Dutta, S. Ray, B.K. Majumdar, antioxidant properties, Asian Pac J Trop Med 7 (2014) 968–976.
Healing of burn wounds by topical treatment: a randomized controlled compari- [253] A. Kubacka, M.S. Diez, D. Rojo, R. Bargiela, S. Ciordia, I. Zapico, J.P. Albar, C. Barbas,
son between silver sulfadiazine and nano-crystalline silver, J. Basic Clin. Pharm. 6 V.A. Martins dos Santos, M. Fernandez-Garcia, M. Ferrer, Understanding the anti-
(2014) 29–34. microbial mechanism of TiO(2)-based nanocomposite films in a pathogenic bacte-
[225] B. Boonkaew, P. Suwanpreuksa, L. Cuttle, P.M. Barber, P. Supaphol, Hydrogels con- rium, Sci Rep 4 (2014) 4134.
taining silver nanoparticles for burn wounds show antimicrobial activity without [254] M. Hromadka, J.B. Collins, C. Reed, L. Han, K.K. Kolappa, B.A. Cairns, T. Andrady, J.A.
cytotoxicity, J. Appl. Polym. Sci. 131 (2014). van Aalst, Nanofiber applications for burn care, J. Burn Care Res. 29 (2008)
[226] M. Konop, T. Damps, A. Misicka, L. Rudnicka, Certain aspects of silver and silver 695–703.
nanoparticles in wound care: a minireview, J. Nanomater. 2016 (2016) 47. [255] S. Sett, M. Lee, M. Weith, B. Pourdeyhimi, A. Yarin, Biodegradable and biocompat-
[227] P. Marcato, L. De Paula, P. Melo, I. Ferreira, A. Almeida, A. Torsoni, O. Alves, In vivo ible soy protein/polymer/adhesive sticky nano-textured interfacial membranes
evaluation of complex biogenic silver nanoparticle and enoxaparin in wound for prevention of esca fungi invasion into pruning cuts and wounds of vines, J.
healing, J. Nanomater. 2015 (2015) 91. Mater. Chem. B 3 (2015) 2147–2162.
[228] A. Hebeish, M.H. El-Rafie, M.A. El-Sheikh, A.A. Seleem, M.E. El-Naggar, Antimicrobi- [256] N. Bhardwaj, S.C. Kundu, Electrospinning: a fascinating fiber fabrication technique,
al wound dressing and anti-inflammatory efficacy of silver nanoparticles, Int. J. Biotechnol. Adv. 28 (2010) 325–347.
Biol. Macromol. 65 (2014) 509–515. [257] M.K. Leach, Z.Q. Feng, S.J. Tuck, J.M. Corey, Electrospinning fundamentals: optimiz-
[229] C. Rigo, L. Ferroni, I. Tocco, M. Roman, I. Munivrana, C. Gardin, W.R. Cairns, V. ing solution and apparatus parameters, J. Vis. Exp. 21 (47) (2011 Jan) http://dx.doi.
Vindigni, B. Azzena, C. Barbante, B. Zavan, Active silver nanoparticles for wound org/10.3791/2494 pii: 2494.
healing, Int. J. Mol. Sci. 14 (2013) 4817–4840. [258] X. Hu, S. Liu, G. Zhou, Y. Huang, Z. Xie, X. Jing, Electrospinning of polymeric nano-
[230] J. Frankova, V. Pivodova, H. Vagnerova, J. Juranova, J. Ulrichova, Effects of sil- fibers for drug delivery applications, J. Control. Release 185 (2014) 12–21.
ver nanoparticles on primary cell cultures of fibroblasts and keratinocytes in [259] J.J. Ahire, L.M. Dicks, 2,3-dihydroxybenzoic acid-containing nanofiber wound
a wound-healing model, J. Appl. Biomater. Funct. Mater. 14 (2016) dressings inhibit biofilm formation by Pseudomonas aeruginosa, Antimicrob.
e137–142. Agents Chemother. 58 (2014) 2098–2104.
[231] M. Karimi, N. Solati, A. Ghasemi, M.A. Estiar, M. Hashemkhani, P. Kiani, E. [260] G. Borkow, J. Gabbay, R. Dardik, A.I. Eidelman, Y. Lavie, Y. Grunfeld, S. Ikher, M. Huszar,
Mohamed, A. Saeidi, M. Taheri, P. Avci, A.R. Aref, M. Amiri, F. Baniasadi, M.R. R.C. Zatcoff, M. Marikovsky, Molecular mechanisms of enhanced wound healing by
Hamblin, Carbon nanotubes part II: a remarkable carrier for drug and gene deliv- copper oxide-impregnated dressings, Wound Repair Regen. 18 (2010) 266–275.
ery, Expert Opin. Drug Deliv. 12 (2015) 1089–1105. [261] J.J. Ahire, M. Hattingh, D.P. Neveling, L.M. Dicks, Copper-containing anti-
[232] M. Karimi, N. Solati, M. Amiri, H. Mirshekari, E. Mohamed, M. Taheri, M. biofilm nanofiber scaffolds as a wound dressing material, PLoS One 11 (2016),
Hashemkhani, A. Saeidi, M.A. Estiar, P. Kiani, A. Ghasemi, S.M. Basri, A.R. e0152755.
Aref, M.R. Hamblin, Carbon nanotubes part I: preparation of a novel and [262] Y. Liu, Q. Sun, S. Wang, R. Long, J. Fan, A. Chen, W. Wu, Studies of silk fibroin/poly
versatile drug-delivery vehicle, Expert Opin. Drug Deliv. 12 (2015) (Lactic-Co-Glycolic acid) scaffold, prepared by thermally induced phase separation,
1071–1087. as a possible wound dressing, Sci. Adv. Mater. 8 (2016) 1045–1052.
[233] M. Ashfaq, N. Verma, S. Khan, Copper/zinc bimetal nanoparticles-dispersed carbon [263] L. Weng, J. Xie, Smart electrospun nanofibers for controlled drug release: recent ad-
nanofibers: a novel potential antibiotic material, Mater Sci Eng C Mater Biol Appl vances and new perspectives, Curr. Pharm. Des. 21 (2015) 1944–1959.
59 (2016) 938–947. [264] C. Dwivedi, H. Pandey, A.C. Pandey, P.W. Ramteke, Nanofibre based smart pharma-
[234] S. Maleki Dizaj, A. Mennati, S. Jafari, K. Khezri, K. Adibkia, Antimicrobial activity of ceutical scaffolds for wound repair and regenerations, Curr. Pharm. Des. 22 (2016)
carbon-based nanoparticles, Adv. Pharm. Bull. 5 (2015) 19–23. 1460–1471.
[236] H. Ji, H. Sun, X. Qu, Antibacterial applications of graphene-based nanomaterials: re- [265] L. Tan, J. Hu, H. Huang, J. Han, H. Hu, Study of multi-functional electrospun compos-
cent achievements and challenges, Adv. Drug Deliv. Rev. 105 (Pt B) (2016 Oct 1) ite nanofibrous mats for smart wound healing, Int. J. Biol. Macromol. 79 (2015)
176–189, http://dx.doi.org/10.1016/j.addr.2016.04.009. 469–476.
[237] T.J. Simmons, S.-H. Lee, T.-J. Park, D. Hashim, P. Ajayan, R. Linhardt, Antiseptic single [266] A.A. Dongargaonkar, G.L. Bowlin, H. Yang, Electrospun blends of gelatin and
wall carbon nanotube bandages, Carbon 47 (2009) 1561–1564. gelatin-dendrimer conjugates as a wound-dressing and drug-delivery platform,
[238] S. Ornes, Core concept: quantum dots, Proc. Natl. Acad. Sci. 113 (2016) 2796–2797. Biomacromolecules 14 (2013) 4038–4045.
[239] H. Sun, N. Gao, K. Dong, J. Ren, X. Qu, Graphene quantum dots-band-aids used for [267] E. Chung, V.Y. Rybalko, P.L. Hsieh, S.L. Leal, M.A. Samano, A.N. Willauer, R.S.
wound disinfection, ACS Nano 8 (2014) 6202–6210. Stowers, S. Natesan, D.O. Zamora, R.J. Christy, Fibrin-based stem cell containing
[240] K. Habiba, D.P. Bracho-Rincon, J.A. Gonzalez-Feliciano, J.C. Villalobos-Santos, V.I. scaffold improves the dynamics of burn wound healing, Wound Repair Regen.
Makarov, D. Ortiz, J.A. Avalos, C.I. Gonzalez, B.R. Weiner, G. Morell, Synergistic an- 24 (2016) 810–819.
tibacterial activity of PEGylated silver–graphene quantum dots nanocomposites, [268] W. Hu, S. Chen, J. Yang, Z. Li, H. Wang, Functionalized bacterial cellulose derivatives
Appl. Mater. Today 1 (2015) 80–87. and nanocomposites, Carbohydr. Polym. 101 (2014) 1043–1060.
[241] H.K. Park, K.Y. Park, Control of particle morphology and size in vapor-phase syn- [269] B. Galateanu, M.C. Bunea, P. Stanescu, E. Vasile, A. Casarica, H. Iovu, A. Hermenean,
thesis of titania, silica and alumina nanoparticles, KONA Powder Part. J. 32 C. Zaharia, M. Costache, In vitro studies of bacterial cellulose and magnetic nano-
(2015) 85–101. particles smart nanocomposites for efficient chronic wounds healing, Stem Cells
[242] Y. Wang, Q. Zhao, N. Han, L. Bai, J. Li, J. Liu, E. Che, L. Hu, Q. Zhang, T. Jiang, S. Wang, Int. 2015 (2015) 195096.
Mesoporous silica nanoparticles in drug delivery and biomedical applications, [270] J. Li, D. Zhai, F. Lv, Q. Yu, H. Ma, J. Yin, Z. Yi, M. Liu, J. Chang, C. Wu, Preparation of
Nanomedicine 11 (2015) 313–327. copper-containing bioactive glass/eggshell membrane nanocomposites for im-
[243] S.C. Thomas, Harshita, P.K. Mishra, S. Talegaonkar, Ceramic nanoparticles: fabrica- proving angiogenesis, antibacterial activity and wound healing, Acta Biomater. 36
tion methods and applications in drug delivery, Curr. Pharm. Des. 21 (2015) (2016) 254–266.
6165–6188. [271] J.O. Kim, J.-K. Noh, R.K. Thapa, N. Hasan, M. Choi, J.H. Kim, J.-H. Lee, S.K. Ku, J.-W.
[244] J.M. Rosenholm, C. Sahlgren, M. Linden, Multifunctional mesoporous silica nano- Yoo, Nitric oxide-releasing chitosan film for enhanced antibacterial and in vivo
particles for combined therapeutic, diagnostic and targeted action in cancer treat- wound-healing efficacy, Int. J. Biol. Macromol. 79 (2015) 217–225.
ment, Curr. Drug Targets 12 (2011) 1166–1186. [272] Z. Lu, J. Gao, Q. He, J. Wu, D. Liang, H. Yang, R. Chen, Enhanced antibacterial and
[245] G.S. Alvarez, C. Hélary, A.M. Mebert, X. Wang, T. Coradin, M.F. Desimone, wound healing activities of microporous chitosan-Ag/ZnO composite dressing,
Antibiotic-loaded silica nanoparticle–collagen composite hydrogels with Carbohydr. Polym. 156 (2017) 460–469.
prolonged antimicrobial activity for wound infection prevention, J. Mater. Chem. [273] K. Ito, A. Saito, T. Fujie, K. Nishiwaki, H. Miyazaki, M. Kinoshita, D. Saitoh, S.
B 2 (2014) 4660–4670. Ohtsubo, S. Takeoka, Sustainable antimicrobial effect of silver sulfadiazine-loaded
[246] J.L. Dunn, R.A. Hunter, K. Gast, R. Maile, B.A. Cairns, M.H. Schoenfisch, Direct detec- nanosheets on infection in a mouse model of partial-thickness burn injury, Acta
tion of blood nitric oxide reveals a burn-dependent decrease of nitric oxide in re- Biomater. 24 (2015) 87–95.
sponse to Pseudomonas aeruginosa infection, Burns. 42 (7) (2016 Nov) [274] H. Nurhasni, J. Cao, M. Choi, I. Kim, B.L. Lee, Y. Jung, J.W. Yoo, Nitric oxide-releasing
1522–1527, http://dx.doi.org/10.1016/j.burns.2016.05.005. poly(lactic-co-glycolic acid)-polyethylenimine nanoparticles for prolonged nitric
[247] E.M. Hetrick, J.H. Shin, H.S. Paul, M.H. Schoenfisch, Anti-biofilm efficacy of nitric oxide release, antibacterial efficacy, and in vivo wound healing activity, Int. J.
oxide-releasing silica nanoparticles, Biomaterials 30 (2009) 2782–2789. Nanomedicine. 10 (2015 Apr 22) 3065–3080, http://dx.doi.org/10.2147/IJN.
[248] I. Yamada, K. Nomura, H. Iwahashi, M. Horie, The effect of titanium dioxide (TiO2) S82199 eCollection 2015.
nano-objects, and their aggregates and agglomerates greater than 100 nm (NOAA) [275] J.-Y. Kim, J.-H. Jun, S.-J. Kim, K.-M. Hwang, S.R. Choi, S.D. Han, M.-W. Son, E.-S. Park,
on microbes under UV irradiation, Chemosphere 143 (2016) 123–127. Wound healing efficacy of a chitosan-based film-forming gel containing tyrothricin
[249] N. von Goetz, C. Lorenz, L. Windler, B. Nowack, M. Heuberger, K. Hungerbuhler, Mi- in various rat wound models, Arch. Pharm. Res. 38 (2015) 229–238.
gration of Ag- and TiO2-(Nano)particles from textiles into artificial sweat under [276] H. Namazi, R. Rakhshaei, H. Hamishehkar, H.S. Kafil, Antibiotic loaded
physical stress: experiments and exposure modeling, Environ. Sci. Technol. 47 carboxymethylcellulose/MCM-41 nanocomposite hydrogel films as potential
(2013) 9979–9987. wound dressing, Int. J. Biol. Macromol. 85 (2016) 327–334.
[250] A. Jesline, N.P. John, P. Narayanan, C. Vani, S. Murugan, Antimicrobial activity of [277] X. Zhao, H. Wu, B. Guo, R. Dong, Y. Qiu, P.X. Ma, Antibacterial anti-oxidant
zinc and titanium dioxide nanoparticles against biofilm-producing methicillin- electroactive injectable hydrogel as self-healing wound dressing with
resistant Staphylococcus aureus, Appl. Nanosci. 5 (2015) 157–162.
62 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

hemostasis and adhesiveness for cutaneous wound healing, Biomaterials 122 [303] H. Hathaway, D.R. Alves, J. Bean, P.P. Esteban, K. Ouadi, J. Mark Sutton, A.T.A.
(2017) 34–47. Jenkins, Poly(N-isopropylacrylamide-co-allylamine) (PNIPAM-co-ALA) nano-
[278] M. Tummalapalli, M. Berthet, B. Verrier, B. Deopura, M. Alam, B. Gupta, Drug loaded spheres for the thermally triggered release of bacteriophage K, Eur. J. Pharm.
composite oxidized pectin and gelatin networks for accelerated wound healing, Biopharm. 96 (2015) 437–441.
Int. J. Pharm. 505 (2016) 234–245. [304] K. Osumi, S. Matsuda, N. Fujimura, K. Matsubara, M. Kitago, O. Itano, C. Ogino, N.
[279] M.D. Romić, M.Š. Klarić, J. Lovrić, I. Pepić, B. Cetina-Čižmek, J. Filipović-Grčić, A. Shimizu, H. Obara, Y. Kitagawa, Acceleration of wound healing by ultrasound acti-
Hafner, Melatonin-loaded chitosan/Pluronic® F127 microspheres as in situ vation of TiO2 in Escherichia coli-infected wounds in mice, J Biomed Mater Res B
forming hydrogel: an innovative antimicrobial wound dressing, Eur. J. Pharm. Appl Biomater (2016).
Biopharm. 107 (2016) 67–79. [305] C. Öhlknecht, G. Tegl, B. Beer, C. Sygmund, R. Ludwig, G.M. Guebitz, Cellobiose de-
[280] Z. Zhou, D. Yan, X. Cheng, M. Kong, Y. Liu, C. Feng, X. Chen, Biomaterials based on N, hydrogenase and chitosan-based lysozyme responsive materials for antimicrobial
N, N-trimethyl chitosan fibers in wound dressing applications, Int. J. Biol. wound treatment, Biotechnol. Bioeng. 114 (2017) 416–422.
Macromol. 89 (2016) 471–476. [306] H.S. Kim, Y.J. Son, H.S. Yoo, Clustering siRNA conjugates for MMP-responsive ther-
[281] D.W. Song, S.H. Kim, H.H. Kim, K.H. Lee, C.S. Ki, Y.H. Park, Multi-biofunction of an- apeutics in chronic wounds of diabetic animals, Nano 8 (2016) 13236–13244.
timicrobial peptide-immobilized silk fibroin nanofiber membrane: implications for [307] H.S. Kim, H.S. Yoo, Matrix metalloproteinase-inspired suicidal treatments of diabetic
wound healing, Acta Biomater. 39 (2016) 146–155. ulcers with siRNA-decorated nanofibrous meshes, Gene Ther. 20 (2013) 378–385.
[282] M. Maleki, M. Latifi, M. Amani-Tehran, S. Mathur, Electrospun core–shell nanofi- [308] L.J. Stevens, A. Page-McCaw, A secreted MMP is required for reepithelialization
bers for drug encapsulation and sustained release, Polym. Eng. Sci. 53 (2013) during wound healing, Mol. Biol. Cell 23 (2012) 1068–1079.
1770–1779. [309] J. Zhang, P. Du, D. Xu, Y. Li, W. Peng, G. Zhang, F. Zhang, X. Fan, Near-infrared re-
[283] Q. Wei, F. Xu, X. Xu, X. Geng, L. Ye, A. Zhang, Z. Feng, The multifunctional wound sponsive MoS2/Poly(N-isopropylacrylamide) hydrogels for remote light-
dressing with core–shell structured fibers prepared by coaxial electrospinning, controlled microvalves, Ind. Eng. Chem. Res. 55 (2016) 4526–4531.
Front. Mater. Sci. 10 (2016) 113–121. [310] M. Zhang, Y. Cao, L. Wang, Y. Ma, X. Tu, Z. Zhang, Manganese doped iron oxide
[284] T. Lu, Y. Li, T. Chen, Techniques for fabrication and construction of three-dimensional theranostic nanoparticles for combined T 1 magnetic resonance imaging and
scaffolds for tissue engineering, Int. J. Nanomedicine 8 (2013) 337–350. photothermal therapy, ACS Appl. Mater. Interfaces 7 (2015) 4650–4658.
[285] J. Hu, P.X. Ma, Nano-fibrous tissue engineering scaffolds capable of growth factor [311] H. Kim, J. Kim, M. Lee, H.C. Choi, W.J. Kim, Stimuli-Regulated Enzymatically Degrad-
delivery, Pharm. Res. 28 (2011) 1273–1281. able Smart Graphene-Oxide Polymer NanocarrierFacilitating Photothermal Gene
[286] Y.R. Park, H.W. Ju, J.M. Lee, D.K. Kim, O.J. Lee, B.M. Moon, H.J. Park, J.Y. Jeong, Y.K. Delivery, Adv. Healthc. Mater. 5 (15) (2016 Aug) 1918–1930, http://dx.doi.org/
Yeon, C.H. Park, Three-dimensional electrospun silk-fibroin nanofiber for skin tis- 10.1002/adhm.201600246 Epub 2016 May 17.
sue engineering, Int. J. Biol. Macromol. 93 (2016) 1567–1574. [312] X. Huang, P.K. Jain, I.H. El-Sayed, M.A. El-Sayed, Plasmonic photothermal therapy
[287] A. Schneider, J.A. Garlick, C. Egles, Self-assembling peptide nanofiber scaffolds ac- (PPTT) using gold nanoparticles, Lasers Med. Sci. 23 (2008) 217–228.
celerate wound healing, PLoS One 3 (2008), e1410. [313] Z. Bao, X. Liu, Y. Liu, H. Liu, K. Zhao, Near-infrared light-responsive inorganic
[288] M. Bradshaw, D. Ho, M.W. Fear, F. Gelain, F.M. Wood, K.S. Iyer, Designer self- nanomaterials for photothermal therapy, Asian J. Pharm. Sci. 11 (2016) 349–364.
assembling hydrogel scaffolds can impact skin cell proliferation and migration, [314] W.-L. Chiang, T.-T. Lin, R. Sureshbabu, W.-T. Chia, H.-C. Hsiao, H.-Y. Liu, C.-M. Yang,
Sci Rep 4 (2014) 6903. H.-W. Sung, A rapid drug release system with a NIR light-activated molecular
[289] M. Karimi, H. Mirshekari, M. Aliakbari, P. Sahandi-Zangabad, Smart mesoporous switch for dual-modality photothermal/antibiotic treatments of subcutaneous ab-
silica nanoparticles for controlled-release drug delivery, Nanotechnol. Rev. (2016 scesses, J. Control. Release 199 (2015) 53–62.
Jan 21) http://dx.doi.org/10.1515/ntrev-2015-0057. [315] W. Yin, J. Yu, F. Lv, L. Yan, L.R. Zheng, Z. Gu, Y. Zhao, Functionalized Nano-MoS2
[290] M. Karimi, P. Sahandi Zangabad, A. Ghasemi, M. Amiri, M. Bahrami, H. with peroxidase catalytic and near-infrared photothermal activities for safe and
Malekzad, H. Ghahramanzadeh Asl, Z. Mahdieh, M. Bozorgomid, A. Ghasemi, synergetic wound antibacterial applications, ACS Nano 10 (2016) 11000–11011.
M.R. Rahmani Taji Boyuk, M.R. Hamblin, Temperature-responsive smart [316] K. Sahu, M. Sharma, H. Bansal, A. Dube, P.K. Gupta, Topical photodynamic treat-
nanocarriers for delivery of therapeutic agents: applications and recent ment with poly-l-lysine–chlorin p6 conjugate improves wound healing by reduc-
advances, ACS Appl. Mater. Interfaces 8 (33) (2016 Aug 24) 21107–21133, ing hyperinflammatory response in Pseudomonas aeruginosa-infected wounds of
http://dx.doi.org/10.1021/acsami.6b00371. mice, Lasers Med. Sci. 28 (2013) 465–471.
[291] M. Karimi, M. Eslami, P. Sahandi-Zangabad, F. Mirab, N. Farajisafiloo, Z. Shafaei, D. [317] S.S. Lucky, N. Muhammad Idris, Z. Li, K. Huang, K.C. Soo, Y. Zhang, Titania coated
Ghosh, M. Bozorgomid, F. Dashkhaneh, M.R. Hamblin, pH-sensitive stimulus- upconversion nanoparticles for near-infrared light triggered photodynamic thera-
responsive nanocarriers for targeted delivery of therapeutic agents, Wiley py, ACS Nano 9 (2015) 191–205.
Interdiscip. Rev. Nanomed. Nanobiotechnol. 8 (5) (2016 Sep) 696–716, http://dx. [318] G.B. Kharkwal, S.K. Sharma, Y.Y. Huang, T. Dai, M.R. Hamblin, Photodynamic ther-
doi.org/10.1002/wnan.1389. apy for infections: clinical applications, Lasers Surg. Med. 43 (2011) 755–767.
[292] S. Davaran, A. Ghamkhari, E. Alizadeh, B. Massoumi, M. Jaymand, Novel dual [319] T. Dai, Y.Y. Huang, M.R. Hamblin, Photodynamic therapy for localized infections–
stimuli-responsive ABC triblock copolymer: RAFT synthesis,“schizophrenic” mi- state of the art, Photodiagnosis Photodyn. Ther. 6 (3-4) (2009 Sep-Dec)
cellization, and its performance as an anticancer drug delivery nanosystem, J. Col- 170–188, http://dx.doi.org/10.1016/j.pdpdt.2009.10.008.
loid Interface Sci. 488 (2017) 282–293. [320] M.R. Hamblin, L.Y. Chiang, S. Lakshmanan, Y.Y. Huang, M. Garcia-Diaz, M. Karimi,
[293] B. Massoumi, N. Poorgholy, M. Jaymand, Multistimuli responsive polymeric nanosys- A.N. de Souza Rastelli, R. Chandran, Nanotechnology for photodynamic therapy:
tems for theranostic applications, Int. J. Polym. Mater. Polym. Biomater. 66 (2017) a perspective from the laboratory of Dr. Michael R. Hamblin in the Wellman Center
38–47. for Photomedicine at Massachusetts General Hospital and Harvard Medical School,
[294] M. Fathi, A.A. Entezami, R. Pashaei-Asl, Swelling/deswelling, thermal, and rheolog- Nanotechnol. Rev. 4 (2015) 359–372.
ical behavior of PVA-g-NIPAAm nanohydrogels prepared by a facile free-radical po- [321] P. Deyhimi, H. Khademi, R. Birang, M. Akhoondzadeh, Histological evaluation of
lymerization method, J. Polym. Res. 20 (2013) 125. wound healing process after photodynamic therapy of rat oral mucosal ulcer, J.
[295] M. Karimi, P. Sahandi Zangabad, S. Baghaee-Ravari, M. Ghazadeh, H. Mirshekari, Dent. 17 (2016) 43–48.
M.R. Hamblin, Smart nanostructures for cargo delivery: uncaging and activating [322] F.F. Sperandio, Y.-Y. Huang, M.R. Hamblin, Antimicrobial photodynamic therapy to
by light, J. Am. Chem. Soc. 139 (13) (2017 Apr 5) 4584–4610, http://dx.doi.org/ kill gram-negative bacteria, Recent Pat. Anti-Infect. Drug Discov. 8 (2013) 108–120.
10.1021/jacs.6b08313. [323] S. Hettiaratchy, J. Clarke, J. Taubel, C. Besa, Burns after photodynamic therapy, BMJ
[296] M. Karimi, A. Ghasemi, P. Sahandi Zangabad, R. Rahighi, S.M. Moosavi Basri, H. 320 (2000) 1245.
Mirshekari, M. Amiri, Z. Shafaei Pishabad, A. Aslani, M. Bozorgomid, D. Ghosh, A. [324] J. van de Kamp, V. Paefgen, M. Wöltje, M. Böbel, J. Jaekel, B. Rath, N. Labude, R.
Beyzavi, A. Vaseghi, A.R. Aref, L. Haghani, S. Bahrami, M.R. Hamblin, Smart micro/ Knüchel, W. Jahnen-Dechent, S. Neuss, Mesenchymal stem cells can be recruited
nanoparticles in stimulus-responsive drug/gene delivery systems, Chem. Soc. to wounded tissue via hepatocyte growth factor-loaded biomaterials, J. Tissue
Rev. 45 (5) (2016 Mar 7) 1457–1501, http://dx.doi.org/10.1039/c5cs00798d. Eng. Regen. Med. (2016 Sep 19) http://dx.doi.org/10.1002/term.2201.
[297] P. Sahandi Zangabad, M. Karimi, F. Mehdizadeh, H. Malekzad, A. Ghasemi, S. [325] V. Bayati, M.R. Abbaspour, N. Neisi, M. Hashemitabar, Skin-derived precursors pos-
Bahrami, H. Zare, M. Moghoofei, A. Hekmatmanesh, M.R. Hamblin, Nanocaged sess the ability of differentiation into the epidermal progeny and accelerate burn
platforms: modification, drug delivery and nanotoxicity. Opening synthetic cages wound healing, M. Cell Biol. Int. 41 (2) (2017 Feb) 187–196, http://dx.doi.org/
to release the tiger, Nano 9 (2017) 1356–1392. 10.1002/cbin.10717.
[298] A. Bansal, Y. Zhang, Photocontrolled nanoparticle delivery systems for biomedical [326] D.M. Burmeister, R. Stone, N.L. Wrice, S.C. Becerra, S. Natesan, R.J. Christy, Fibrin
applications, Acc. Chem. Res. 47 (10) (2014 Oct 21) 3052–3060, http://dx.doi.org/ hydrogels prevent contraction and deliver adipose stem cells to debrided
10.1021/ar500217w. deep partial thickness burns for accelerated angiogenesis, FASEB J. 30 (2016)
[299] J.-O. You, M. Rafat, D. Almeda, N. Maldonado, P. Guo, C.S. Nabzdyk, M. Chun, F.W. 1300.1307.
LoGerfo, J.W. Hutchinson, L.K. Pradhan-Nabzdyk, pH-responsive scaffolds generate [327] A. Butko, G. Bonat Celli, A. Paulson, A. Ghanem, Entrapment of basic fibroblast
a pro-healing response, Biomaterials 57 (2015) 22–32. growth factor (bFGF) in a succinylated chitosan nanoparticle delivery system
[300] N. Ninan, A. Forget, V.P. Shastri, N.H. Voelcker, A. Blencowe, Antibacterial and anti- and release profile, J. Biomater. Sci. Polym. Ed. (2016) 1–13.
inflammatory pH-responsive tannic acid-carboxylated agarose composite [328] M. Goh, Y. Hwang, G. Tae, Epidermal growth factor loaded heparin-based hydrogel
hydrogels for wound healing, ACS Appl. Mater. Interfaces 8 (2016) 28511–28521. sheet for skin wound healing, Carbohydr. Polym. 147 (2016) 251–260.
[301] V. Grützner, R.E. Unger, G. Baier, L. Choritz, C. Freese, T. Böse, K. Landfester, C.J. [329] J. Du, L. Liu, F. Lay, Q. Wang, C. Dou, X. Zhang, S.M. Hosseini, A. Simon, D.J. Rees, A.K.
Kirkpatrick, Enzyme-responsive nanocomposites for wound infection prophylaxis Ahmed, R. Sebastian, K. Sarkar, S. Milner, G.P. Marti, G.L. Semenza, J.W. Harmon,
in burn management: in vitro evaluation of their compatibility with healing pro- Combination of HIF-1[alpha] gene transfection and HIF-1-activated bone
cesses, Int. J. Nanomedicine 10 (2015) 4111. marrow-derived angiogenic cell infusion improves burn wound healing in aged
[302] Y. Li, G. Liu, X. Wang, J. Hu, S. Liu, Enzyme-responsive polymeric vesicles for mice, Gene Ther. 20 (2013) 1070–1076.
bacterial-strain-selective delivery of antimicrobial agents, Angew. Chem. 128 [330] C.T. Turner, M. Hasanzadeh Kafshgari, E. Melville, B. Delalat, F.J. Harding, E. Mäkilä,
(2016) 1792–1796. J.J. Salonen, A.J. Cowin, N.H. Voelcker, Delivery of flightless I siRNA from porous
M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64 63

silicon nanoparticles improves wound healing in mice, ACS Biomater. Sci. Eng. 2 [353] J.M. Lopacinska, C. Gradinaru, R. Wierzbicki, C. Kobler, M.S. Schmidt, M.T. Madsen,
(12) (2016) 2339–2346, http://dx.doi.org/10.1021/acsbiomaterials.6b00550. M. Skolimowski, M. Dufva, H. Flyvbjerg, K. Molhave, Cell motility, morphology, vi-
[331] L.-H. Peng, W. Wei, X.-T. Qi, Y.-H. Shan, F.-J. Zhang, X. Chen, Q.-Y. Zhu, L. Yu, W.-Q. ability and proliferation in response to nanotopography on silicon black, Nano 4
Liang, J.-Q. Gao, Epidermal stem cells manipulated by pDNA-VEGF165/CYD-PEI (2012) 3739–3745.
nanoparticles loaded gelatin/β-TCP matrix as a therapeutic agent and gene deliv- [354] J. Kim, H.N. Kim, K.T. Lim, Y. Kim, H. Seonwoo, S.H. Park, H.J. Lim, D.H. Kim, K.Y. Suh,
ery vehicle for wound healing, Mol. Pharm. 10 (2013) 3090–3102. P.H. Choung, Y.H. Choung, J.H. Chung, Designing nanotopographical density of ex-
[332] B. Chen, H.-K. Kao, Z. Dong, Z. Jiang, L. Guo, Complementary effects of negative- tracellular matrix for controlled morphology and function of human mesenchymal
pressure wound therapy and pulsed radiofrequency energy on cutaneous wound stem cells, Sci Rep 3 (2013) 3552.
healing in diabetic mice, Plast. Reconstr. Surg. 139 (2017) 105–117. [355] J. Guasch, J. Diemer, H. Riahinezhad, S. Neubauer, H. Kessler, J.P. Spatz, Synthesis of
[333] M. Asghari, A. Kanonisabet, M. Safakhah, Z. Azimzadeh, A. Mostafavinia, S. Taheri, binary nanopatterns on hydrogels for initiating cellular responses, Chem. Mater. 28
A. Amini, S.K. Ghorishi, R. JalaliFiroozkohi, S. Bayat, M. Bayat, The effect of com- (2016) 1806–1815.
bined photobiomodulation and metformin on open skin wound healing in a [356] B.K. Teo, S. Ankam, L.Y. Chan, E.K. Yim, Nanotopography/mechanical induction of
non-genetic model of type II diabetes, J. Photochem. Photobiol. B Biol. 169 stem-cell differentiation, Methods Cell Biol. 98 (2010) 241–294.
(2017) 63–69. [357] O. Mashinchian, L.A. Turner, M.J. Dalby, S. Laurent, M.A. Shokrgozar, S. Bonakdar, M.
[334] P.M.D.S. Valente, A. Deva, Q. Ngo, K. Vickery, The increased killing of biofilms Imani, M. Mahmoudi, Regulation of stem cell fate by nanomaterial substrates,
in vitro by combining topical silver dressings with topical negative pressure in Nanomedicine (London) 10 (2015) 829–847.
chronic wounds, Int. Wound J. 13 (2016) 130–136. [358] F.J. Harding, S. Surdo, B. Delalat, C. Cozzi, R. Elnathan, S. Gronthos, N.H. Voelcker, G.
[335] L. Larsen, C.N. Tchanque-Fossuo, F. Gorouhi, D. Bodreault, C. Nguyen, R.W. Barillaro, Ordered silicon pillar arrays prepared by electrochemical
Crawford, S.E. Dahle, T. Whetzel, R.R. Isseroff, Combination therapy of autologous micromachining: substrates for high-efficiency cell transfection, ACS Appl. Mater.
adipose mesenchymal stem cell-enriched, high density lipoaspirate and topical Interfaces 8 (2016) 29197–29202.
timolol for healing chronic wounds, J. Tissue Eng. Regen. Med. (2016 Dec 12) [359] D. Brodoceanu, H.Z. Alhmoud, R. Elnathan, B. Delalat, N.H. Voelcker, T. Kraus, Fab-
http://dx.doi.org/10.1002/term.2390. rication of silicon nanowire arrays by near-field laser ablation and metal-assisted
[336] E. Seo, J.S. Lim, J.-B. Jun, W. Choi, I.-S. Hong, H.-S. Jun, Exendin-4 in combination chemical etching, Nanotechnology 27 (2016), 075301.
with adipose-derived stem cells promotes angiogenesis and improves diabetic [360] R. Elnathan, B. Delalat, D. Brodoceanu, H. Alhmoud, F.J. Harding, K. Buehler, A.
wound healing, J. Transl. Med. 15 (2017) 35. Nelson, L. Isa, T. Kraus, N.H. Voelcker, Maximizing transfection efficiency of ver-
[337] L. Sun, Y. Huang, Z. Bian, J. Petrosino, Z. Fan, Y. Wang, K.H. Park, T. Yue, M. Schmidt, tically aligned silicon nanowire arrays, Adv. Funct. Mater. 25 (2015)
S. Galster, J. Ma, H. Zhu, M. Zhang, Sundew-inspired adhesive hydrogels combined 7215–7225.
with adipose-derived stem cells for wound healing, ACS Appl. Mater. Interfaces 8 [361] D. Brodoceanu, R. Elnathan, B. Prieto-Simón, B. Delalat, T. Guinan, E. Kroner, N.H.
(2016) 2423–2434. Voelcker, T. Kraus, Dense arrays of uniform submicron pores in silicon and their
[338] L. Zhang, P. Xu, X. Wang, M. Zhang, Y. Yan, Y. Chen, L. Zhang, L. Zhang, Activin B applications, ACS Appl. Mater. Interfaces 7 (2015) 1160–1169.
regulates adipose-derived mesenchymal stem cells to promote skin wound [362] R. Elnathan, L. Isa, D. Brodoceanu, A. Nelson, F.J. Harding, B. Delalat, T. Kraus, N.H.
healing via activation of the MAPK signaling pathway, Int. J. Biochem. Cell Biol. Voelcker, Versatile particle-based route to engineer vertically aligned silicon nano-
87 (2017) 69–76. wire arrays and nanoscale pores, ACS Appl. Mater. Interfaces 7 (2015)
[339] S. Yoshida, H. Yoshimoto, A. Hirano, S. Akita, Wound healing and angiogenesis 23717–23724.
through combined use of a vascularized tissue flap and adipose-derived stem [363] W. Chen, L.G. Villa-Diaz, Y. Sun, S. Weng, J.K. Kim, R.H. Lam, L. Han, R. Fan, P.H.
cells in a rat hindlimb irradiated ischemia model, Plast. Reconstr. Surg. 137 Krebsbach, J. Fu, Nanotopography influences adhesion, spreading, and self-
(2016) 1486–1497. renewal of human embryonic stem cells, ACS Nano 6 (2012) 4094–4103.
[340] R. Pouriran, A. Piryaei, A. Mostafavinia, S. Zandpazandi, F. Hendudari, A. Amini, M. [364] E.K. Yim, E.M. Darling, K. Kulangara, F. Guilak, K.W. Leong, Nanotopography-
Bayat, The effect of combined pulsed wave low-level laser therapy and human induced changes in focal adhesions, cytoskeletal organization, and mechanical
bone marrow mesenchymal stem cell-conditioned medium on open skin wound properties of human mesenchymal stem cells, Biomaterials 31 (2010) 1299–1306.
healing in diabetic rats, Photomed. Laser Surg. 34 (2016) 345–354. [365] I.A. Darby, B. Laverdet, F. Bonte, A. Desmouliere, Fibroblasts and myofibroblasts in
[341] M. Figurova, V. Ledecky, M. Karasova, M. Hluchy, A. Trbolova, I. Capik, S. Hornak, P. wound healing, Clin. Cosmet. Investig. Dermatol. 7 (2014) 301–311.
Reichel, J.M. Bjordal, P. Gal, Histological assessment of a combined low-level laser/ [366] J.L. Funderburgh, M.L. Funderburgh, M.M. Mann, L. Corpuz, M.R. Roth, Proteoglycan
light-emitting diode therapy (685 nm/470 nm) for sutured skin incisions in a por- expression during transforming growth factor beta-induced keratocyte-
cine model: a short report, Photomed. Laser Surg. 34 (2016) 53–55. myofibroblast transdifferentiation, J. Biol. Chem. 276 (2001) 44173–44178.
[342] R.I. Korelo, M. Kryczyk, C. Garcia, K. Naliwaiko, L.C. Fernandes, Wound healing [367] L.G. Parkinson, N.L. Giles, K.F. Adcroft, M.W. Fear, F.M. Wood, G.E. Poinern, The po-
treatment by high frequency ultrasound, microcurrent, and combined thera- tential of nanoporous anodic aluminium oxide membranes to influence skin
py modifies the immune response in rats, Braz. J. Phys. Ther. 20 (2016) wound repair, Tissue Eng. A 15 (2009) 3753–3763.
133–141. [368] D. Franco, F. Milde, M. Klingauf, F. Orsenigo, E. Dejana, D. Poulikakos, M. Cecchini, P.
[343] J.U. Choi, S.W. Lee, R. Pangeni, Y. Byun, I.-S. Yoon, J.W. Park, Preparation and in vivo Koumoutsakos, A. Ferrari, V. Kurtcuoglu, Accelerated endothelial wound healing
evaluation of cationic elastic liposomes comprising highly skin-permeable growth on microstructured substrates under flow, Biomaterials 34 (2013) 1488–1497.
factors combined with hyaluronic acid for enhanced diabetic wound-healing ther- [369] L.G. Parkinson, S.M. Rea, A.W. Stevenson, F.M. Wood, M.W. Fear, The effect of nano-
apy, Acta Biomater. 57 (2017) 197–215. scale topography on keratinocyte phenotype and wound healing following burn
[344] N. Nagai, F. Ogata, S. Deguchi, A. Ueno, N. Kawasaki, Y. Ito, Combination ointment injury, Tissue Eng. A 18 (2012) 703–714.
containing solid tranilast nanoparticles and dissolved sericin is efficacious for [370] S.A. Pot, S.J. Liliensiek, K.E. Myrna, E. Bentley, J.V. Jester, P.F. Nealey, C.J. Murphy,
treating skin wound-healing deficits and redness in diabetic rats, Biol. Pharm. Nanoscale topography-induced modulation of fundamental cell behaviors of rabbit
Bull. 40 (2017) 444–450. corneal keratocytes, fibroblasts, and myofibroblasts, Invest. Ophthalmol. Vis. Sci.
[345] Y. Heun, K. Pogoda, M. Anton, J. Pircher, A. Pfeifer, M. Woernle, A. Ribeiro, P. 51 (2010) 1373–1381.
Kameritsch, O. Mykhaylyk, C. Plank, F. Kroetz, U. Pohl, H. Mannell, HIF-1α depen- [371] M. Mahmoudi, S. Bonakdar, M.A. Shokrgozar, H. Aghaverdi, R. Hartmann, A. Pick, G.
dent wound healing angiogenesis in vivo can be controlled by site-specific Witte, W.J. Parak, Cell-imprinted substrates direct the fate of stem cells, ACS Nano
lentiviral magnetic targeting of SHP-2, Mol. Ther. 25 (7) (2017 Jul 5) 1616–1627, 7 (2013) 8379–8384.
http://dx.doi.org/10.1016/j.ymthe.2017.04.007. [372] L.M. Murray, V. Nock, J.J. Evans, M.M. Alkaisi, Bioimprinted polymer platforms for
[346] P.S. Rabbani, A. Zhou, Z.M. Borab, J.A. Frezzo, N. Srivastava, H.T. More, W.J. Rifkin, cell culture using soft lithography, J. Nanobiotechnol. 12 (2014) 60.
J.A. David, S.J. Berens, R. Chen, S. Hameedi, M.H. Junejo, C. Kim, R.A. Sartor, C.F. [373] S. Tavakol, S. Jalili-Firoozinezhad, O. Mashinchian, M. Mahmoudi, Chapter 26 —
Liu, P.B. Saadeh, J.K. Montclare, D.J. Ceradini, Novel lipoproteoplex delivers Keap1 bioinspired nanotechnologies for skin regeneration A2 — Hamblin, Michael R, in:
siRNA based gene therapy to accelerate diabetic wound healing, Biomaterials P. Avci, T.W. Prow (Eds.), Nanoscience in Dermatology, Academic Press, Boston
132 (2017) 1–15. 2016, pp. 337–352.
[347] A.C. Aragão-Neto, P.A.G. Soares, M.H.M. Lima-Ribeiro, E.J.A. Carvalho, M.T.S. Correia, [374] O. Mashinchian, S. Bonakdar, H. Taghinejad, V. Satarifard, M. Heidari, M. Majidi, S.
M.G. Carneiro-da-Cunha, Combined therapy using low level laser and chitosan- Sharifi, A. Peirovi, S. Saffar, M. Taghinejad, Cell-imprinted substrates act as an arti-
policaju hydrogel for wound healing, Int. J. Biol. Macromol. 95 (2017) 268–272. ficial niche for skin regeneration, ACS Appl. Mater. Interfaces 6 (2014)
[348] Y.G. Park, I.H. Lee, E.S. Park, J.Y. Kim, Hydrogel and platelet-rich plasma combined 13280–13292.
treatment to accelerate wound healing in a nude mouse model, Arch. Plast. Surg. [375] S. Bonakdar, M. Mahmoudi, L. Montazeri, M. Taghipoor, A. Bertsch, M.A.
44 (2017) 194–201. Shokrgozar, S. Sharifi, M. Majidi, O. Mashinchian, M. Hamrang Sekachaei, P.
[349] N. Morimoto, N. Kakudo, T. Ogura, T. Hara, M. Matsui, M. Yamamoto, Y. Tabata, K. Zolfaghari, P. Renaud, Cell-imprinted substrates modulate differentiation,
Kusumoto, Easy-to-use preservation and application of platelet-rich plasma in redifferentiation, and transdifferentiation, ACS Appl. Mater. Interfaces 8 (2016)
combination wound therapy with a gelatin sheet and freeze-dried platelet-rich 13777–13784.
plasma: a case report, Eplasty 16 (2016), e22. [376] C.L. Ventola, Medical applications for 3D printing: current and projected uses, P T
[350] N. Morimoto, N. Kakudo, M. Matsui, T. Ogura, T. Hara, K. Suzuki, M. Yamamoto, Y. 39 (2014) 704–711.
Tabata, K. Kusumoto, Exploratory clinical trial of combination wound therapy with [377] H. Dodziuk, Applications of 3D printing in healthcare, Kardiochir. Torakochirurgia
a gelatin sheet and platelet-rich plasma in patients with chronic skin ulcers: study Pol. 13 (2016) 283–293.
protocol, BMJ Open 5 (2015). [378] V. Lee, G. Singh, J.P. Trasatti, C. Bjornsson, X. Xu, T.N. Tran, S.S. Yoo, G. Dai, P.
[351] E. Lamers, R. van Horssen, J. te Riet, F.C. van Delft, R. Luttge, X.F. Walboomers, J.A. Karande, Design and fabrication of human skin by three-dimensional bioprinting,
Jansen, The influence of nanoscale topographical cues on initial osteoblast mor- Tissue Eng. Part C Methods 20 (2014) 473–484.
phology and migration, Eur. Cell. Mater. 20 (2010) 329–343. [379] Z.X. Khoo, J.E.M. Teoh, Y. Liu, C.K. Chua, S. Yang, J. An, K.F. Leong, W.Y. Yeong, 3D
[352] D.-H. Kim, P.P. Provenzano, C.L. Smith, A. Levchenko, Matrix nanotopography as a printing of smart materials: a review on recent progresses in 4D printing, Virtual
regulator of cell function, J. Cell Biol. 197 (2012) 351–360. Phys. Prototyp. 10 (2015) 103–122.
64 M.A. Mofazzal Jahromi et al. / Advanced Drug Delivery Reviews 123 (2018) 33–64

[380] F. Momeni, S.M. Mehdi Hassani.N, X. Liu, J. Ni, A review of 4D printing, Mater. Des. [399] A.B. Bjornson, H.S. Bjornson, N.A. Lincoln, W.A. Altemeier, Relative roles of burn in-
122 (2017) 42–79. jury, wound colonization, and wound infection in induction of alterations of com-
[381] B. Gao, Q. Yang, X. Zhao, G. Jin, Y. Ma, F. Xu, 4D bioprinting for biomedical applica- plement function in a guinea pig model of burn injury, J. Trauma 24 (1984)
tions, Trends Biotechnol. 34 (2016) 746–756. 106–115.
[382] J.D. Sander, J.K. Joung, CRISPR-Cas systems for editing, regulating and targeting ge- [400] H.L. Walker, A.D. Mason Jr., A standard animal burn, J. Trauma 8 (1968)
nomes, Nat. Biotechnol. 32 (2014) 347–355. 1049–1051.
[383] X. Yang, Applications of CRISPR-Cas9 mediated genome engineering, Mil. Med. Res. [401] T. Kaufman, S.N. Lusthaus, U. Sagher, M.R. Wexler, Deep partial skin thickness
2 (2015) 11. burns: a reproducible animal model to study burn wound healing, Burns 16
[384] W. Jiang, L.A. Marraffini, CRISPR-Cas: new tools for genetic manipulations from (1990) 13–16.
bacterial immunity systems, Annu. Rev. Microbiol. 69 (2015) 209–228. [402] E.J. Stevens, C.M. Ryan, J.S. Friedberg, R.L. Barnhill, M.L. Yarmush, R.G. Tompkins, A
[385] B.R. Webber, M.J. Osborn, A.N. McElroy, K. Twaroski, C.L. Lonetree, A.P. DeFeo, L. quantitative model of invasive Pseudomonas infection in burn injury, J. Burn Care
Xia, C. Eide, C.J. Lees, R.T. McElmurry, M.J. Riddle, C.J. Kim, D.D. Patel, B.R. Blazar, Rehabil. 15 (1994) 232–235.
J. Tolar, CRISPR/Cas9-based genetic correction for recessive dystrophic [403] http://ir.cytori.com/investor-relations/news/news-details/2017/Cytori-Receives-
epidermolysis bullosa, NPJ Regen. Med. 1 (2016). US-FDA-Approval-for-Burn-Clinical-Trial-Related-to-BARDA-Contract/default.
[386] M. Chen, M. Przyborowski, F. Berthiaume, Stem cells for skin tissue engineering aspx.
and wound healing, Crit. Rev. Biomed. Eng. 37 (2009) 399–421. [404] https://clinicaltrials.gov/ct2/show/NCT02104713.
[387] E. Gorell, N. Nguyen, A. Lane, Z. Siprashvili, Gene therapy for skin diseases, Cold [405] F. Haubner, E. Ohmann, F. Pohl, J. Strutz, H.G. Gassner, Wound healing after radia-
Spring Harb. Perspect. Med. 4 (2014) a015149. tion therapy: review of the literature, Radiat. Oncol. 7 (2012) 162.
[388] V. Nomellini, C.R. Gomez, R.L. Gamelli, E.J. Kovacs, Aging and animal models of sys- [406] A. Landriscina, T. Musaev, J. Rosen, A. Ray, P. Nacharaju, J.D. Nosanchuk, A.J.
temic insult: trauma, burn, and sepsis, Shock 31 (2009) 11–20. Friedman, N-acetylcysteine S-nitrosothiol nanoparticles prevent wound expansion
[389] S. Domergue, C. Jorgensen, D. Noel, Advances in research in animal models of burn- and accelerate wound closure in a murine burn model, J. Drugs Dermatol. 14
related hypertrophic scarring, J. Burn Care Res. 36 (2015) e259–266. (2015) 726–732.
[390] T.P. Sullivan, W.H. Eaglstein, S.C. Davis, P. Mertz, The pig as a model for human [407] http://www.internano.org/node/4211.
wound healing, Wound Repair Regen. 9 (2001) 66–76. [408] http://grantome.com/grant/NIH/R43-GM081967-01.
[391] T. Dai, G.B. Kharkwal, M. Tanaka, Y.Y. Huang, V.J. Bil de Arce, M.R. Hamblin, Animal [409] M. Yamada, M. Foote, T.W. Prow, Therapeutic gold, silver, and platinum nanoparti-
models of external traumatic wound infections, Virulence 2 (2011) 296–315. cles, Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 7 (2015) 428–445.
[392] X. Ragas, D. Sánchez-García, R. Ruiz-González, T. Dai, M. Agut, M.R. Hamblin, S. [410] P. Guo, The emerging field of RNA nanotechnology, Nat. Nanotechnol. 5 (2010)
Nonell, Cationic porphycenes as potential photosensitizers for antimicrobial pho- 833–842.
todynamic therapy, J. Med. Chem. 53 (2010) 7796–7803. [411] N. Morimoto, K. Yoshimura, M. Niimi, T. Ito, R. Aya, J. Fujitaka, H. Tada, S.
[393] T. Katakura, T. Yoshida, M. Kobayashi, D.N. Herndon, F. Suzuki, Immunological con- Teramukai, T. Murayama, C. Toyooka, K. Miura, S. Takemoto, N. Kanda, K. Kawai,
trol of methicillin-resistant Staphylococcus aureus (MRSA) infection in an immuno- M. Yokode, A. Shimizu, S. Suzuki, Novel collagen/gelatin scaffold with sustained re-
deficient murine model of thermal injuries, Clin. Exp. Immunol. 142 (2005) lease of basic fibroblast growth factor: clinical trial for chronic skin ulcers, Tissue
419–425. Eng. A 19 (2013) 1931–1940.
[394] D.D. Stieritz, I.A. Holder, Experimental studies of the pathogenesis of infections due [412] https://www.regmednet.com/channels/544-spotlight-on-wound-healing/posts/
to Pseudomonas aeruginosa: description of a burned mouse model, J Infect Dis 131 13238-skin-based-gene-therapy-safe-and-effective-in-phase-1-clinical-trial-for-
(1975) 688–691. epidermolysis-bullosa-wounds.
[395] S. Tavares Pereira Ddos, M.H. Lima-Ribeiro, N.T. de Pontes-Filho, A.M. Carneiro- [413] L. Yuxiang, T. Lu, Y. Jianqiang, D. Xiuying, Z. Wanfang, Z. Wannian, H. Xiaoyan, X.
Leao, M.T. Correia, Development of animal model for studying deep second- Shichu, N. Wen, M. Xiuqiang, Analgesia effect of a fixed nitrous oxide/oxygen mix-
degree thermal burns, J Biomed Biotechnol 2012 (2012) 460841. ture on burn dressing pain: study protocol for a randomized controlled trial, Trials
[396] A. Orenstein, D. Klein, J. Kopolovic, E. Winkler, Z. Malik, N. Keller, Y. Nitzan, The use 13 (2012) 67.
of porphyrins for eradication of Staphylococcus aureus in burn wound infections, [414] K. Zhou, K. Krug, J. Stachura, P. Niewczyk, M. Ross, J. Tutuska, G. Ford, Silver-
FEMS Immunol. Med. Microbiol. 19 (1997) 307–314. collagen dressing and high-voltage, pulsed-current therapy for the treatment of
[397] T. Suzuki, T. Hirayama, K. Aihara, Y. Hirohata, Experimental studies of moderate chronic full-thickness wounds: a case series, Ostomy Wound Manage 62 (2016)
temperature burns, Burns 17 (1991) 443–451. 36–44.
[398] T. Bahar, B. Bilezikci, T. Maral, H. Borman, A modified partial-thickness burn model [415] F.W. Endorf, D. Ahrenholz, Burn management, Curr. Opin. Crit. Care 17 (6) (2011
in rats, Burns 33 (2007) S52–S53. Dec) 601–605, http://dx.doi.org/10.1097/MCC.0b013e32834c563f.

You might also like